You are on page 1of 1329

 i

Schottenfeld and Fraumeni


Cancer Epidemiology and Prevention
ii
 iii

Schottenfeld and Fraumeni


Cancer Epidemiology and
Prevention
Fourth Edition

Lead Editor CHRISTOPHER HAIMAN, SCD


Department of Preventive Medicine
MICHAEL J. THUN, MD, MS Keck School of Medicine, University
Epidemiology and Surveillance Research (Retired) of Southern California
American Cancer Society Los Angeles, California
Atlanta, Georgia

Co-​Editors DAVID SCHOTTENFELD, MD, MSC


Department of Epidemiology (Retired)
MARTHA S. LINET, MD, MPH University of Michigan School of Public Health
Division of Cancer Epidemiology and Genetics Ann Arbor, Michigan
National Cancer Institute
Bethesda, Maryland Project Manager

ANNELIE M. LANDGREN, MPH, PMP


JAMES R. CERHAN, MD, PHD
Department of Health Sciences Research
Mayo Clinic
Rochester, Minnesota

1
iv

1
Oxford University Press is a department of the University of Oxford. It furthers
the University’s objective of excellence in research, scholarship, and education
by publishing worldwide. Oxford is a registered trade mark of Oxford University
Press in the UK and certain other countries.

Published in the United States of America by Oxford University Press


198 Madison Avenue, New York, NY 10016, United States of America.

© Oxford University Press 2018

Third Edition published 2006


Second edition published 1996
First edition published 1982

All rights reserved. No part of this publication may be reproduced, stored in


a retrieval system, or transmitted, in any form or by any means, without the
prior permission in writing of Oxford University Press, or as expressly permitted
by law, by license, or under terms agreed with the appropriate reproduction
rights organization. Inquiries concerning reproduction outside the scope of the
above should be sent to the Rights Department, Oxford University Press, at the
address above.

You must not circulate this work in any other form


and you must impose this same condition on any acquirer.

Library of Congress Cataloging-in-Publication Data


Names: Thun, Michael J., editor. | Linet, Martha S., editor. |
Cerhan, James R., editor. | Haiman, Christopher, editor. | Schottenfeld, David, editor.
Title: Schottenfeld and Fraumeni Cancer Epidemiology and Prevention / lead editor,
Michael J. Thun ; co-editors, Martha S. Linet, James R. Cerhan,
Christopher Haiman, David Schottenfeld ; project manager, Annelie M. Landgren.
Other titles: Cancer epidemiology and prevention
Description: Fourth edition. | New York, NY : Oxford University Press, [2018] |
Preceded by Cancer epidemiology and prevention / edited by David Schottenfeld,
Joseph F. Fraumeni Jr. 3rd ed. 2006. | Includes bibliographical references and index.
Identifiers: LCCN 2017038170 | ISBN 9780190238667 (hardcover : alk. paper)
Subjects: | MESH: Neoplasms—epidemiology | Neoplasms—prevention & control
Classification: LCC RA645.C3 | NLM QZ 220.1 |
DDC 614.5/999—dc23
LC record available at https://lccn.loc.gov/2017038170

9 8 7 6 5 4 3 2 1
Printed by Sheridan Books, Inc., United States of America
 v

Contents

Acknowledgments  ix
Contributors  xi
Preface  xix

1. Introduction  1
Michael J. Thun, Martha S. Linet, James R. Cerhan, Christopher A. Haiman,
and David Schottenfeld

I  BASIC CONCEPTS
2. Biology of Neoplasia  9
Michael Dean and Karobi Moitra
3. Morphological and Molecular Classification of Human Cancer  19
Mark E. Sherman, Melissa A. Troester, Katherine A. Hoadley,
and William F. Anderson
4. Genomic Landscape of Cancer: Insights for Epidemiologists  43
Christopher J. Maher and Elaine R. Mardis
5. Genetic Epidemiology of Cancer  53
Kathryn L. Penney, Kyriaki Michailidou, Deanna Alexis Carere, Chenan Zhang,
Brandon Pierce, Sara Lindström, and Peter Kraft
6. Application of Biomarkers in Cancer Epidemiology  77
Roel Vermeulen, Douglas A. Bell, Dean P. Jones, Montserrat Garcia-​Closas,
Avrum Spira, Teresa W. Wang, Martyn T. Smith, Qing Lan, and Nathaniel Rothman
7. Causal Inference in Cancer Epidemiology  97
Steven N. Goodman and Jonathan M. Samet

II  THE MAGNITUDE OF CANCER


8. Patterns of Cancer Incidence, Mortality, and Survival  107
Ahmedin Jemal, D. Maxwell Parkin, and Freddie Bray
9. Socioeconomic Disparities in Cancer Incidence and Mortality  141
Candyce Kroenke and Ichiro Kawachi
10. The Economic Burden of Cancer in the United States  169
K. Robin Yabroff, Gery P. Guy Jr., Matthew P. Banegas, and Donatus U. Ekwueme
vi

vi Contents

III  THE CAUSES OF CANCER


11. Tobacco  185
Michael J. Thun and Neal D. Freedman
12. Alcohol and Cancer Risk  213
Susan M. Gapstur and Philip John Brooks
13. Ionizing Radiation  227
Amy Berrington de González, André Bouville, Preetha Rajaraman,
and Mary Schubauer-​Berigan
14. Ultraviolet Radiation  249
Adèle C. Green and David C. Whiteman
15. Electromagnetic Fields  259
Maria Feychting and Joachim Schüz
16. Occupational Cancer  275
Kyle Steenland, Shelia Hoar Zahm, and A. Blair
17. Air Pollution  291
Jonathan M. Samet and Aaron J. Cohen
18. Water Contaminants  305
Kenneth P. Cantor, Craig M. Steinmaus, Mary H. Ward,
and Laura E. Beane Freeman
19. Diet and Nutrition  329
Marjorie L. McCullough and Walter C. Willett
20. Obesity and Body Composition  351
NaNa Keum, Mingyang Song, Edward L. Giovannucci, and A. Heather Eliassen
21. Physical Activity, Sedentary Behaviors, and Risk of Cancer  377
Steven C. Moore, Charles E. Matthews, Sarah Keadle, Alpa V. Patel,
and I-​Min Lee
22. Hormones and Cancer  395
Robert N. Hoover, Amanda Black, and Rebecca Troisi
23. Pharmaceutical Drugs Other Than Hormones  411
Marie C. Bradley, Michael A. O’Rorke, Janine A. Cooper, Søren Friis,
and Laurel A. Habel
24. Infectious Agents  433
Silvia Franceschi, Hashem B. El-​Serag, David Forman, Robert Newton,
and Martyn Plummer
25. Immunologic Factors  461
Eric A. Engels and Allan Hildesheim

IV  CANCERS BY TISSUE OF ORIGIN


26. Nasopharyngeal Cancer  489
Ellen T. Chang and Allan Hildesheim
27. Cancer of the Larynx  505
Andrew F. Olshan and Mia Hashibe
28. Lung Cancer  519
Michael J. Thun, S. Jane Henley, and William D. Travis
29. Oral Cavity, Oropharynx, Lip, and Salivary Glands  543
Mia Hashibe, Erich M. Sturgis, Jacques Ferlay, and Deborah M. Winn
30. Esophageal Cancer  579
William J. Blot and Robert E. Tarone
 vi

Contents vii
31. Stomach Cancer  593
Catherine de Martel and Julie Parsonnet
32. Cancer of the Pancreas  611
Samuel O. Antwi, Rick J. Jansen, and Gloria M. Petersen
33. Liver Cancer  635
W. Thomas London, Jessica L. Petrick, and Katherine A. McGlynn
34. Biliary Tract Cancer  661
Jill Koshiol, Catterina Ferreccio, Susan S. Devesa, Juan Carlos Roa,
and Joseph F. Fraumeni, Jr.
35. Small Intestine Cancer  671
Jennifer L. Beebe-​Dimmer, Fawn D. Vigneau, and David Schottenfeld
36. Cancers of the Colon and Rectum  681
Kana Wu, NaNa Keum, Reiko Nishihara, and Edward L.Giovannucci
37. Anal Cancer  707
Andrew E. Grulich, Fengyi Jin, and I. Mary Poynten
38. Leukemias  715
Martha S. Linet, Lindsay M. Morton, Susan S. Devesa, and Graça M. Dores
39. Hodgkin Lymphoma  745
Henrik Hjalgrim, Ellen T. Chang, and Sally L. Glaser
40. The Non-​Hodgkin Lymphomas  767
James R. Cerhan, Claire M. Vajdic, and John J. Spinelli
41. Multiple Myeloma  797
Mark P. Purdue, Jonathan N. Hofmann, Elizabeth E. Brown, and Celine M. Vachon
42. Bone Cancers  815
Lisa Mirabello, Rochelle E. Curtis, and Sharon A. Savage
43. Soft Tissue Sarcoma  829
Marianne Berwick and Charles Wiggins
44. Thyroid Cancer  839
Cari M. Kitahara, Arthur B. Schneider, and Alina V. Brenner
45. Breast Cancer  861
Louise A. Brinton, Mia M. Gaudet, and Gretchen L. Gierach
46. Ovarian Cancer  889
Shelley S. Tworoger, Amy L. Shafrir, and Susan E. Hankinson
47. Endometrial Cancer  909
Linda S. Cook, Angela L. W. Meisner, and Noel S. Weiss
48. Cervical Cancer  925
Rolando Herrero and Raul Murillo
49. Vulvar and Vaginal Cancers  947
Margaret M. Madeleine and Lisa G. Johnson
50. Choriocarcinoma  953
Julie R. Palmer
51. Renal Cancer  961
Wong-​Ho Chow, Ghislaine Scelo, and Robert E. Tarone
52. Bladder Cancer  977
Debra T. Silverman, Stella Koutros, Jonine D. Figueroa, Ludmila Prokunina-​Olsson,
and Nathaniel Rothman
53. Prostate Cancer  997
Catherine M. Tangen, Marian L. Neuhouser, and Janet L. Stanford
vi

viii Contents
54. Testicular Cancer  1019
Katherine A. McGlynn, Ewa Rajpert-​De Meyts, and Andreas Stang
55. Penile Cancer  1029
Morten Frisch
56. Nervous System  1039
E. Susan Amirian, Quinn T. Ostrom, Yanhong Liu, Jill Barnholtz-​Sloan,
and Melissa L. Bondy
57. Melanoma  1061
Bruce K. Armstrong, Claire M. Vajdic, and Anne E. Cust
58. Keratinocyte Cancers  1089
Anala Gossai, Dorothea T. Barton, Judy R. Rees, Heather H. Nelson,
and Margaret R. Karagas
59. Childhood Cancers  1119
Eve Roman, Tracy Lightfoot, Susan Picton, and Sally Kinsey
60. Multiple Primary Cancers  1155
Lindsay M. Morton, Sharon A. Savage, and Smita Bhatia

V  CANCER PREVENTION AND CONTROL


61. Framework for Understanding Cancer Prevention  1193
Michael J. Thun, Christopher P. Wild, and Graham Colditz
62. Primary Prevention of Cancer  1205
62.1. Tobacco Control  1207
Jeffrey Drope, Clifford E. Douglas, and Brian D. Carter
62.2. Prevention of Obesity and Physical Inactivity  1211
Ambika Satija and Frank B. Hu
62.3. Prevention of Infection-​Related Cancers  1217
Marc Bulterys, Julia Brotherton, and Ding-​Shinn Chen
62.4. Protection from Ultraviolet Radiation  1221
Robyn M. Lucas, Rachel E. Neale, Peter Gies, and Terry Slevin
62.5. Preventive Therapy  1229
Jack Cuzick
62.6. Regulation  1239
Jonathan M. Samet and Lynn Goldman
63. Cancer Screening  1255
Jennifer M. Croswell, Russell P. Harris, and Barnett S. Kramer

Index 1271
 ix

Acknowledgments

We are indebted to the more than 190 chapter authors who generously contributed their time,
labor, and expertise to produce this comprehensively updated fourth edition. The multi-​authored
text reflects the increasingly interdisciplinary and collaborative nature of the field; it provides a
resource for researchers seeking to harness the unprecedented advances in genetic and molecu-
lar research into large-​scale population studies of cancer etiology, and ultimately into effective
preventive interventions. We owe special thanks to Ms. Annelie Landgren, whose energy, enthu-
siasm, and organizational expertise as project manager have been invaluable in bringing this
text to completion. We also thank Dr. Stephen Chanock for his early and unfailing encourage-
ment and for supporting the critical infrastructure necessary for such a collaborative enterprise.
This book would not have been possible without the generous forbearance of our spouses and
families. Finally, Michael Thun thanks Dr. Lynne Moody for her insights as a sounding board
throughout this process.

ix
x
 xi

Contributors

E. Susan Amirian, PhD Jennifer L. Beebe-​Dimmer, PhD, MPH


Dan L Duncan Cancer Center Wayne State University School of Medicine
Baylor College of Medicine Karmanos Cancer Institute
Houston, Texas Detroit, Michigan
William F. Anderson, MD, MPH Douglas A. Bell, PhD
Division of Cancer Epidemiology and Genetics Environmental Epigenomics, Immunity, Inflammation and
National Cancer Institute Disease Laboratory
Bethesda, Maryland National Institute of Environmental Health Sciences
Research Triangle Park, North Carolina
Samuel O. Antwi, PhD
Department of Health Sciences Research Amy Berrington de González, DPhil
Mayo Clinic College of Medicine Division of Cancer Epidemiology and Genetics
Rochester, Minnesota National Cancer Institute
Bethesda, Maryland
Bruce K. Armstrong, MD, PhD*
School of Public Health Marianne Berwick, PhD
The University of Sydney Department of Internal Medicine
Sydney, New South Wales, Australia University of New Mexico
Albuquerque, New Mexico
Matthew P. Banegas, PhD, MPH
Center for Health Research Smita Bhatia, MD, MPH
Kaiser Permanente Institute of Cancer Outcomes and Survivorship
Portland, Oregon University of Alabama at Birmingham, School of Medicine
Birmingham, Alabama
Jill Barnholtz-​Sloan, PhD
Case Comprehensive Cancer Center Amanda Black, PhD, MPH
Case Western Reserve University School of Medicine Division of Cancer Epidemiology and Genetics
Cleveland, Ohio National Cancer Institute
Bethesda, Maryland
Dorothea T. Barton, MD
Department of Surgery A. Blair, PhD
Dartmouth-​Hitchcock Medical Center Division of Cancer Epidemiology and Genetics
Lebanon, New Hampshire National Cancer Institute
Bethesda, Maryland
Laura E. Beane Freeman, PhD
Division of Cancer Epidemiology and Genetics William J. Blot, PhD
National Cancer Institute Vanderbilt-​Ingram Cancer Center
Bethesda, Maryland Nashville, Tennessee

* Retired

xi
xi

xii Contributors
Melissa L. Bondy, PhD James R. Cerhan, MD, PhD, (Editor)
Department of Medicine, Section of Epidemiology and Population Sciences Department of Health Sciences Research
Baylor College of Medicine Mayo Clinic
Houston, Texas Rochester, Minnesota
AndrÉ Bouville, PhD* Ellen T. Chang, ScD
Division of Cancer Epidemiology and Genetics Center for Health Sciences Exponent Inc.
National Cancer Institute Menlo Park, California
Bethesda, Maryland
Ding-​Shinn Chen, MD
Marie C. Bradley, PhD, MScPH Hepatitis Research Center
Division of Cancer Control and Population Sciences National Taiwan University Hospital
National Cancer Institute Taipei, Taiwan
Bethesda, Maryland
Wong-​Ho Chow, PhD
Freddie Bray, PhD Department of Epidemiology
Section of Cancer Surveillance The University of Texas
International Agency for Research on Cancer MD Anderson Cancer Center
Lyon, France Houston, Texas
Alina V. Brenner, MD, PhD Aaron J. Cohen, MPH, DSc‡
Division of Cancer Epidemiology and Genetics Health Effects Institute
National Cancer Institute Boston, Massachusetts
Bethesda, Maryland
Graham Colditz, MD, DrPH
Louise A. Brinton, PhD Division of Public Health Services
Division of Cancer Epidemiology and Genetics Washington University
National Cancer Institute St. Louis, Missouri
Bethesda, Maryland
Linda S. Cook, PhD
Philip John Brooks, PhD Department of Internal Medicine
Laboratory of Neurogenetics University of New Mexico
National Institute on Alcohol Abuse and Alcoholism, NIH Albuquerque, New Mexico
Bethesda, Maryland
Janine A. Cooper, PhD
Julia Brotherton, MD, PhD School of Pharmacy
National HPV Vaccination Program Register Queen’s University Belfast
Victorian Cytology Service Belfast, Northern Ireland
East Melbourne, Victoria, Australia
Jennifer M. Croswell, MD, MPH
Elizabeth E. Brown, PhD, MPH Patient-​Centered Outcomes Research Institute
Department of Pathology Washington, DC
University of Alabama at Birmingham
Rochelle E. Curtis, MA
Birmingham, Alabama
Division of Cancer Epidemiology and Genetics
Marc Bulterys, MD, PhD National Cancer Institute
HIV/​Hepatitis Department Bethesda, Maryland
World Health Organization
Anne E. Cust, PhD
Geneva, Switzerland
School of Public Health and Melanoma Institute Australia
Kenneth P. Cantor, PhD, MPH* The University of Sydney
Division of Cancer Epidemiology and Genetics Sydney, New South Wales, Australia
National Cancer Institute
Jack Cuzick, PhD
Bethesda, Maryland
Wolfson Institute of Preventive Medicine
Deanna Alexis Carere, ScD, CGC Queen Mary University of London
Department of Pathology and Molecular Medicine London, United Kingdom
McMaster University
Catherine de Martel, MD, PhD
Hamilton, Ontario, Canada
Infections and Cancer Epidemiology Group
Brian D. Carter, MPH International Agency for Research on Cancer
Epidemiology Research Program Lyon, France
American Cancer Society
Atlanta, Georgia

* Retired

Consultant/​Contractor
 xi

Contributors xiii
Michael Dean, PhD David Forman, PhD
Division of Cancer Epidemiology and Genetics International Agency for Research on Cancer
National Cancer Institute Lyon, France
Bethesda, Maryland
Silvia Franceschi, MD
Susan S. Devesa, PhD* International Agency for Research on Cancer
Division of Cancer Epidemiology and Genetics Lyon, France
National Cancer Institute
Joseph F. Fraumeni, Jr., MD
Bethesda, Maryland
Division of Cancer Epidemiology and Genetics
Graça M. Dores, MD§ National Cancer Institute
Division of Cancer Epidemiology and Genetics Bethesda, Maryland
National Cancer Institute
Neal D. Freedman, PhD
Bethesda, Maryland
Division of Cancer Epidemiology and Genetics
Clifford E. Douglas, JD National Cancer Institute
Center for Tobacco Control Bethesda, Maryland
American Cancer Society
Søren Friis, MD
Atlanta, Georgia
Statistics and Pharmacoepidemiology
Jeffrey Drope, PhD Danish Cancer Society Research Center
Economic & Health Policy Research Copenhagen, Denmark
American Cancer Society
Morten Frisch, MD, PhD, DrSci(Med)
Atlanta, Georgia
Department of Epidemiology Research
Donatus U. Ekwueme, PhD, MS Statens Serum Institut
National Center for Chronic Disease Prevention and Health Promotion Copenhagen, Denmark
Centers for Disease Control and Prevention
Susan M. Gapstur, PhD, MPH
Atlanta, Georgia
Epidemiology Research Program
A. Heather Eliassen, ScD American Cancer Society
Brigham & Women’s Hospital and Harvard Medical School Atlanta, Georgia
Harvard TH Chan School of Public Health
Montserrat Garcia-​Closas, MD, DrPH
Boston, Massachusetts
Division of Cancer Epidemiology and Genetics
Hashem B. El-​Serag, MD, MPH National Cancer Institute
Gastroenterology and Hepatology Bethesda, Maryland
Baylor College of Medicine
Mia M. Gaudet, PhD
Houston, Texas
Epidemiology Research Program
Eric A. Engels, MD American Cancer Society
Division of Cancer Epidemiology and Genetics Atlanta, Georgia
National Cancer Institute
Gretchen L. Gierach, PhD
Bethesda, Maryland
Division of Cancer Epidemiology and Genetics
Jacques Ferlay, MSc National Cancer Institute
Section of Cancer Surveillance Bethesda, Maryland
International Agency for Research on Cancer
Peter Gies, PhD
Lyon, France
Australian Radiation Protection and Nuclear Safety Agency
Catterina Ferreccio, MD, MPH Melbourne, Victoria, Australia
Division of Public Health and Family Medicine
Edward L. Giovannucci, MD, ScD
School of Medicine, Pontificia Universidad Católica de Chile
Departments of Nutrition and Epidemiology
Santiago, Chile
Harvard TH Chan School of Public Health
Maria Feychting, PhD Boston, Massachusetts
Institute of Environmental Medicine
Sally L. Glaser, PhD
Karolinska Institutet
Cancer Prevention Institute of California
Stockholm, Sweden
Fremont, California
Jonine D. Figueroa, PhD
Lynn Goldman, MD, MS, MPH
Usher Institute of Population Health Sciences and Informatics,
Milken Institute School of Public Health
CRUK Edinburgh Centre
George Washington University
University of Edinburgh
Washington, DC
Edinburg, United Kingdom

* Retired
§
Adjunct
vxi

xiv Contributors
Steven N. Goodman, MD, PhD Henrik Hjalgrim, MD, PhD, DrSci(med)
Department of Medicine, Clinical and Translational Research Department of Epidemiology Research
Stanford University School of Medicine Statens Serum Institut
Stanford, California Copenhagen, Denmark
Anala Gossai, MPH, PhD Katherine A. Hoadley, PhD
Geisel School of Medicine Department of Genetics, Lineberger Comprehensive Cancer Center
Dartmouth College University of North Carolina at Chapel Hill
Hanover, New Hampshire Chapel Hill, North Carolina
Adèle C. Green, MD, PhD Jonathan N. Hofmann, PhD
Population Health Division Division of Cancer Epidemiology and Genetics
QIMR Berghofer Medical Research Institute National Cancer Institute
Brisbane, Queensland, Australia Bethesda, Maryland
Andrew E. Grulich, PhD Robert N. Hoover, MD, ScD
Kirby Institute Division of Cancer Epidemiology and Genetics
The University of New South Wales National Cancer Institute
Sydney, New South Wales, Australia Bethesda, Maryland
Gery P. Guy Jr, PhD, MPH Frank B. Hu, MD, PhD
National Center for Chronic Disease Prevention and Health Departments of Nutrition and Epidemiology
Promotion Harvard TH Chan School of Public Health
Centers for Disease Control and Prevention Boston, Massachusetts
Atlanta, Georgia
Rick J. Jansen, MS, PhD
Laurel A. Habel, PhD, MPH Department of Public Health
Division of Research North Dakota State University
Kaiser Permanente Northern California Fargo, North Dakota
Oakland, California
Ahmedin Jemal, DVM, PhD
Christopher A. Haiman, ScD, (Editor) Surveillance and Health Services Research Program
Department of Preventive Medicine American Cancer Society
Keck School of Medicine of University of Southern California Atlanta, Georgia
Los Angeles, California
Fengyi Jin, PhD
Susan E. Hankinson, ScD Kirby Institute
Department of Biostatistics and Epidemiology The University of New South Wales
University of Massachusetts Sydney, New South Wales, Australia
Amherst, Massachusetts
Lisa G. Johnson, PhD, MPH
Russell P. Harris, MD, MPH§ Division of Public Health Sciences
Lineberger Comprehensive Cancer Center Fred Hutchinson Cancer Research Center
University of North Carolina School of Medicine Seattle, Washington
Chapel Hill, North Carolina
Dean P. Jones, PhD
Mia Hashibe, PhD Department of Medicine
Department of Family and Preventive Medicine Emory University
Huntsman Cancer Institute, University of Utah School of Medicine Atlanta, Georgia
Salt Lake City, Utah
Margaret R. Karagas, PhD
S. Jane Henley, MSPH Department of Epidemiology
Division of Cancer Prevention and Control Geisel School of Medicine at Dartmouth
US Centers for Disease Control and Prevention Hanover, New Hampshire
Atlanta, Georgia
Ichiro Kawachi, MD, PhD
Rolando Herrero, MD, PhD Department of Social and Behavioral Sciences
Prevention and Implementation Group Harvard School of Public Health
International Agency for Research on Cancer Boston, Massachusetts
Lyon, France
Sarah Keadle, PhD, MPH
Allan Hildesheim, PhD Kinesiology Department
Division of Cancer Epidemiology and Genetics California Polytechnic State University
National Cancer Institute San Luis Obispo, California
Bethesda, Maryland

§
adjunct
 xv

Contributors xv
NaNa Keum, ScD Yanhong Liu, PhD
Department of Nutrition Department of Medicine, Section of Epidemiology
Harvard TH Chan School of Public Health and Population Sciences
Boston, Massachusetts Baylor College of Medicine
Houston, Texas
Sally Kinsey, MD, FRCP
Department of Pediatric Hematology W. Thomas London, MD*
Leeds Teaching Hospitals NHS Trust Fox Chase Cancer Center
Leeds, United Kingdom Philadelphia, Pennsylvania
Cari M. Kitahara, PhD Robyn M. Lucas, MBChB, MPH&TM, PhD
Division of Cancer Epidemiology and Genetics Radiation Health Services Branch
National Cancer Institute The Australian National University
Bethesda, Maryland Canberra, The Australian Capital Territory, Australia
Jill Koshiol, PhD Margaret M. Madeleine, PhD
Division of Cancer Epidemiology and Genetics Division of Public Health Sciences
National Cancer Institute Fred Hutchinson Cancer Research Center
Bethesda, Maryland Seattle, Washington
Stella Koutros, PhD Christopher J. Maher, PhD
Division of Cancer Epidemiology and Genetics McDonnell Genome Institute
National Cancer Institute Washington University School of Medicine
Bethesda, Maryland St. Louis, Missouri
Peter Kraft, PhD Elaine R. Mardis, PhD
Departments of Epidemiology and Biostatistics McDonnell Genome Institute
Harvard TH Chan School of Public Health Washington University School of Medicine
Boston, Massachusetts St. Louis, Missouri
Barnett S. Kramer, MD, MPH Charles E. Matthews, PhD
Division of Cancer Prevention Division of Cancer Epidemiology and Genetics
National Cancer Institute National Cancer Institute
Bethesda, Maryland Bethesda, Maryland
Candyce Kroenke, ScD, MPH Marjorie L. McCullough, ScD, RD
Division of Research Epidemiology Research Program
Kaiser Permanente Northern California American Cancer Society
Oakland, California Atlanta, Georgia
Qing Lan, MD, MPH Katherine A. McGlynn, PhD
Division of Cancer Epidemiology and Genetics Division of Cancer Epidemiology and Genetics
National Cancer Institute National Cancer Institute
Bethesda, Maryland Bethesda, Maryland
I-​Min Lee, MBBS, ScD Angela L. W. Meisner, MPH
Brigham and Women’s Hospital New Mexico Tumor Registry
Harvard Medical School University of New Mexico
Boston, Massachusetts Albuquerque, New Mexico
Tracy Lightfoot, PhD Kyriaki Michailidou, PhD
Department of Health Sciences Department of Electron Microscopy/​Molecular Pathology
University of York The Cyprus Institute of Neurology and Genetics
York, United Kingdom Nicosia, Cyprus
Sara Lindström, PhD Lisa Mirabello, PhD
Department of Epidemiology Division of Cancer Epidemiology and Genetics
University of Washington National Cancer Institute
Seattle, Washington Bethesda, Maryland
Martha S. Linet, MD, MPH, (Editor) Karobi Moitra, PhD
Division of Cancer Epidemiology and Genetics Trinity Washington University
National Cancer Institute Washington, DC
Bethesda, Maryland

* Retired
xvi

xvi Contributors
Steven C. Moore, PhD Alpa V. Patel, PhD
Division of Cancer Epidemiology and Genetics Epidemiology Research Program
National Cancer Institute American Cancer Society
Bethesda, Maryland Atlanta, Georgia
Lindsay M. Morton, PhD Kathryn L. Penney, ScD
Division of Cancer Epidemiology and Genetics ScD Department of Medicine
National Cancer Institute Brigham and Women’s Hospital /​Harvard Medical School
Bethesda, Maryland Boston, Massachusetts
Raul Murillo, MD Gloria M. Petersen, PhD
Prevention and Implementation Group Department of Health Sciences Research
International Agency for Research on Cancer Mayo Clinic College of Medicine
Lyon, France Rochester, Minnesota
Rachel E. Neale, PhD Jessica L. Petrick, PhD
Population Health Division Division of Cancer Epidemiology and Genetics
QIMR Berghofer Medical Research Institute National Cancer Institute
Brisbane, Queensland, Australia Bethesda, Maryland
Heather H. Nelson, MPH, PhD Susan Picton, BMBS, FRCPCH
Division of Epidemiology, Masonic Cancer Center Department of Pediatric Oncology
University of Minnesota, Twin Cities Leeds Teaching Hospitals NHS Trust
Minneapolis, Minnesota Leeds, United Kingdom
Marian L. Neuhouser, PhD Brandon Pierce, PhD
Division of Public Health Sciences Departments of Public Health Sciences and Human Genetics
Fred Hutchinson Cancer Research Center University of Chicago
Seattle, Washington Chicago, Illinois
Robert Newton, PhD Martyn Plummer, PhD
MRC/​UVRI Uganda Research Unit International Agency for Research on Cancer
Entebbe, Uganda Lyon, France
Reiko Nishihara, PhD I. Mary Poynten, PhD Kirby Institute
Department of Pathology The University of New South Wales
Brigham and Women’s Hospital Sydney, New South Wales, Australia
Boston, Massachusetts
Ludmila Prokunina-​Olsson, PhD
Michael A. O’Rorke, PhD Division of Cancer Epidemiology and Genetics
School of Medicine Dentistry and Biomedical Sciences National Cancer Institute
Queen’s University Belfast Bethesda, Maryland
Belfast, Northern Ireland
Mark P. Purdue, PhD
Andrew F. Olshan, PhD Division of Cancer Epidemiology and Genetics
Department of Epidemiology National Cancer Institute
Gillings School of Global Public Health Bethesda, Maryland
University of North Carolina
Preetha Rajaraman, PhD
Chapel Hill, North Carolina
Division of Cancer Epidemiology and Genetics
Quinn T. Ostrom, MA, MPH National Cancer Institute
Case Comprehensive Cancer Center Bethesda, Maryland
Case Western Reserve University School of Medicine
Ewa Rajpert-​De Meyts, MD, PhD
Cleveland, Ohio
Department of Growth & Reproduction
Julie R. Palmer, ScD Copenhagen University Hospital Rigshospitalet
Slone Epidemiology Center at Boston University Copenhagen, Denmark
Boston, Massachusetts
Judy R. Rees, BM, BCh, PhD
D. Maxwell Parkin, MD, DSc Department of Epidemiology
Nuffield Department of Public Health University of Oxford Geisel School of Medicine at Dartmouth
Oxford, United Kingdom Hanover, New Hampshire
Julie Parsonnet, MD Juan Carlos Roa, MD, Msc
Department of Medicine Department of Pathology
Stanford University School of Medicine School of Medicine, Pontificia Universidad Católica de Chile
Stanford, California Santiago, Chile
 xvi

Contributors xvii
Eve Roman, PhD Martyn T. Smith, PhD
Department of Health Sciences School of Public Health
University of York University of California at Berkeley
York, United Kingdom Berkeley, California
Nathaniel Rothman, MD, MPH Mingyang Song, MD, ScD
Division of Cancer Epidemiology and Genetics Clinical and Translational Epidemiology Unit and Division
National Cancer Institute of Gastroenterology
Bethesda, Maryland Massachusetts General Hospital and Harvard Medical School
Boston, Massachusetts
Jonathan M. Samet, MD
Department of Preventive Medicine John J. Spinelli, PhD Cancer Control Research
Keck School of Medicine of University of Southern California British Columbia Cancer Agency
Los Angeles, California Vancouver, British Columbia, Canada
Ambika Satija, ScD Avrum Spira, MD, MSc
Department of Nutrition Division of Computational Biomedicine
Harvard TH Chan School of Public Health Boston University School of Medicine
Boston, Massachusetts Boston, Massachusetts
Sharon A. Savage, MD Janet L. Stanford, PhD
Division of Cancer Epidemiology and Genetics Division of Public Health Sciences
National Cancer Institute Fred Hutchinson Cancer Research Center
Bethesda, Maryland Seattle, Washington
Ghislaine Scelo, PhD Andreas Stang, MD, MPH
Genetic Epidemiology Group Institute of Medical Informatics, Biometry and Epidemiology
International Agency for Research on Cancer University Hospital Essen
Lyon, France Essen, Germany
Arthur B. Schneider, MD, PhD* Kyle Steenland, PhD
Section of Endocrinology, Diabetes and Metabolism Rollins School of Public Health Emory University
University of Illinois at Chicago College of Medicine Atlanta, Georgia
Chicago, Illinois
Craig M. Steinmaus, MD, MPH
David Schottenfeld, MD, MSc (Editor)* School of Public Health
Department of Epidemiology University of California Berkeley
University of Michigan School of Public Health Berkeley, California
Ann Arbor, Michigan
Erich M. Sturgis, MD, MPH
Mary Schubauer-​Berigan, PhD Department of Head and Neck Surgery
Division of Surveillance Hazard Evaluation and Field Studies The University of Texas MD Anderson Cancer Center
Centers for Disease Control and Prevention Houston, Texas
Atlanta, Georgia
Catherine M. Tangen, DrPH
Joachim Schüz, PhD Division of Public Health Sciences
Section of Environment and Radiation Fred Hutchinson Cancer Research Center
International Agency for Research on Cancer Seattle, Washington
Lyon, France
Robert E. Tarone, PhD*
Amy L. Shafrir, ScD International Epidemiology Institute
Division of Adolescent and Young Adult Medicine Rockville, Maryland
Boston Children’s Hospital
Michael J. Thun, MD, MS (Editor)*
Boston, Massachusetts
Epidemiology and Surveillance Research
Mark E. Sherman, MD American Cancer Society
Health Sciences Research Atlanta, Georgia
Mayo Clinic College of Medicine
William D. Travis, MD
Jacksonville, Florida
Department of Pathology
Debra T. Silverman, ScD, ScM Memorial Sloan Kettering Cancer Center
Division of Cancer Epidemiology and Genetics New York, New York
National Cancer Institute
Melissa A. Troester, PhD
Bethesda, Maryland
Department of Epidemiology
Terry Slevin, MPH Lineberger Comprehensive Cancer Center
Cancer Council Western Australia University of North Carolina at Chapel Hill
Perth, Western Australia, Australia Chapel Hill, North Carolina

* Retired
xvii

xviii Contributors
Rebecca Troisi, ScD, MA David C. Whiteman, MD, PhD
Division of Cancer Epidemiology and Genetics Population Health Division
National Cancer Institute QIMR Berghofer Medical Research Institute
Bethesda, Maryland Brisbane, Queensland, Australia
Shelley S. Tworoger, PhD Charles Wiggins, PhD, MPH
Harvard Medical School and the Brigham and Women’s Hospital Department of Internal Medicine
Harvard TH Chan School of Public Health University of New Mexico
Boston, Massachusetts Albuquerque, New Mexico
Celine M. Vachon, PhD Christopher P. Wild, PhD
Department of Health Sciences Research Director’s Office
Mayo Clinic International Agency for Research on Cancer
Rochester, Minnesota Lyon, France
Claire M. Vajdic, PhD Walter C. Willett, MD, DrPH
Centre for Big Data Research in Health Department of Nutrition
University of New South Wales Harvard TH Chan School of Public Health
Sydney, New South Wales, Australia Boston, Massachusetts
Roel Vermeulen, PhD Deborah M. Winn, PhD
Institute for Risk Assessment Sciences Division of Cancer Control and Population Sciences
Utrecht University National Cancer Institute
Utrecht, The Netherlands Bethesda, Maryland
Fawn D. Vigneau, JD, MPH Kana Wu, MD, PhD
Wayne State University School of Medicine Department of Nutrition
Karmanos Cancer Institute Harvard TH Chan School of Public Health
Detroit, Michigan Boston, Massachusetts
Teresa W. Wang, PhD K. Robin Yabroff, PhD
Division of Computational Biomedicine Division of Cancer Control and Population Sciences
Boston University School of Medicine National Cancer Institute
Boston, Massachusetts Bethesda, Maryland
Mary H. Ward, PhD Shelia Hoar Zahm, ScD‡
Division of Cancer Epidemiology and Genetics Division of Cancer Epidemiology and Genetics
National Cancer Institute National Cancer Institute
Bethesda, Maryland Bethesda, Maryland
Noel S. Weiss, MD, DrPH Chenan Zhang, PhD
Department of Epidemiology Department of Epidemiology and Biostatistics
University of Washington University of California, San Francisco
Seattle, Washington San Francisco, California


Consultant/​Contractor
x i

Preface

The Schottenfeld and Fraumeni text on Cancer Epidemiology and Prevention has served as the
premier reference text for population research on the causes and prevention of cancers since the
publication of the first edition in 1982 (Schottenfeld and Fraumeni, 1982). It is written for col-
leagues pursuing careers in research in cancer epidemiology and, more broadly, in preventive
oncology. The founding editors, Dr. David Schottenfeld, now emeritus professor of epidemiol-
ogy at the University of Michigan, and Dr. Joseph Fraumeni, recently retired as the director of the
Division of Cancer Epidemiology and Genetics at the National Cancer Institute (NCI), updated
their landmark text in 1996 and 2006 (Schottenfeld and Fraumeni, 1996, 2006).
The current edition again provides a comprehensive update of research advances in cancer
epidemiology, prevention, and related fields in the past 10–​15 years, and honors the founding
editors in the title. The new editorial team is led by Dr. Michael Thun (editor-​in-​chief), formerly
with the American Cancer Society, and includes four senior co-​editors: Drs. Martha Linet from
NCI, James Cerhan from the Mayo Clinic, Christopher Haiman from the University of Southern
California, and David Schottenfeld. We are also deeply indebted to the internationally recog-
nized experts who authored the 63 chapters. Without their generous effort and commitment, this
updated synthesis would not be possible.

xix
x
 1

1 Introduction

MICHAEL J. THUN, MARTHA S. LINET, JAMES R. CERHAN,


CHRISTOPHER A. HAIMAN, AND DAVID SCHOTTENFELD

I n this introduction, we provide an overview of the text and highlight


cross-​cutting developments and new opportunities that are trans-
forming our understanding of the causes and prevention of cancer. As
costs. In 2012, these countries accounted for over half (57%) of all
incident cancers; this is projected to increase to nearly two-​thirds
(65%) by 2035. Much of the increase will result from the growth and
in previous editions, the text is grouped into five major parts: “Basic aging of populations, since LMICs currently comprise about 80% of
Concepts,” “The Magnitude of Cancer,” “The Causes of Cancer,” the world’s population, and large numbers of young adults are now
“Cancers by Tissue of Origin,” and “Cancer Prevention and Control.” surviving to older ages, when cancer becomes more common. In
Part I first describes research advances in understanding “the biol- addition to the effect of demographic changes, cancer incidence and
ogy of neoplasia,” including the progressive disruption of genetic and mortality rates are increasing in LMICs because of the widespread
epigenetic controls that regulate cell growth, division, and survival adoption of Western patterns of diet, physical inactivity, excess body
(Chapter 2). Advances in high-​throughput technologies have greatly fat, delayed reproduction, and tobacco smoking, especially of manu-
expanded the ability to identify germline and somatic mutations and factured cigarettes. As countries advance economically, the incidence
to relate these to etiology, prognosis, and treatment. Tumor classifi- rates of cancers traditionally associated with Westernization (e.g.,
cation is also changing for certain cancers, as data on the molecular breast, colorectum, lung, and prostate) increase more rapidly than the
features and lineage of the neoplastic cells is combined with infor- decrease in cancers caused partly or wholly by infectious agents (e.g.,
mation on the primary anatomic location and the morphologic, histo- stomach, liver, uterine cervix). Survival after a diagnosis of cancer is
pathologic and clinical characteristics of the tumor (Chapter 3). The also lower in LMICs than in high-​resource countries, because of later
“landscape” of genomic and epigenomic alterations in tumor tissue stage at diagnosis, a higher proportion of tumors diagnosed clinically
has been cataloged for multiple human cancers (Chapter 4), revealing rather than incidentally, and limited access to standard and state-​of-​
both the singularity of individual cancer genomes and the commonal- the-​art treatment protocols.
ity of genetic alterations that drive cancer in different tissues. Chapter In economically developed countries, the incidence rates of most
5 describes advances in research on inherited genomic variants that cancers are either stabilizing at a high level or decreasing, depend-
affect cancer risk. Genome-​wide association studies (GWAS) have ing on the temporal trends of underlying risk factors and utilization of
identified more than 700 germline loci associated with increased or cancer screening. Despite the decreasing rates, the disease burden, or
decreased risk for various types of cancer, although the risk estimates number of cancer cases and deaths, continues to increase. The increas-
for almost all are small to modest. Innovations in genomics and other ing burden results from the aging and growth of populations, and the
“OMIC” technologies are identifying biomarkers that reflect internal decline in competing causes of death from circulatory and infectious
exposures, biological processes, and intermediate outcomes in large diseases. Mortality rates are decreasing more rapidly than incidence
population studies (Chapter 6). While research in many of these areas rates for many cancer sites due to a combination of prevention, early
is still in its infancy, mechanistic and molecular insights are extending detection, and improvements in treatment.
the traditional criteria for inferring causation in epidemiologic studies Part III, “The Causes of Cancer,” discusses 15 broad categories
of cancer (Chapter 7). of exposure that affect cancer risk. These include exposures that are
Part 2 of the book discusses the global public health impact of can- typically considered “environmental” by the public (chemical carcino-
cer and its relationship to demographic trends, changing risk factors, gens, ionizing radiation, occupational exposures, pollutants in air and
socioeconomic disparities, and economic development. It considers drinking water), as well as exposures that are less widely recognized as
the direct and indirect costs of cancer in the United States to illustrate carcinogenic (infectious agents, metabolic factors, body composition,
the economic burden in a high income country. Parts 3–​5 of the book reproductive and other hormones, pharmaceutical drugs, and immu-
discuss the growing list of exposures known to affect cancer risk, the nological conditions). All of these exposures are “environmental” in
epidemiology of over 30 types of cancer by tissue of origin, and the the sense that they are acquired after conception rather than inherited.
encouraging progress in cancer prevention and control. Major devel- Some are genotoxic and damage the structure of DNA or alter DNA
opments in these areas are discussed below, beginning with those that repair; others modify gene expression, induce oxidative stress and/​
affect the public health impact of cancer. or chronic inflammation, suppress host immunity, immortalize cells,
modulate receptors, and/​or alter cell proliferation, cell death, or nutri-
ent supply (Smith et al., 2016).
MAJOR NEW DEVELOPMENTS Although some exposures are conventionally perceived as “life-
style choices”, they are by no means entirely voluntary. For example,
behavioral risk factors such as tobacco smoking, energy imbal-
Global Trends in Cancer Risk and Burden
ance, and physical inactivity are strongly influenced by factors in
Part II, “The Magnitude of Cancer,” provides a global public health the social, economic, and cultural environment, beginning in early
perspective on cancer. The human and economic costs of cancer are childhood. Physiologic addiction is a major driver of tobacco use at
increasing worldwide (http://​globocan.iarc.fr). The World Health all ages.
Organization (WHO) estimates that 14 million new cases and 8.2 mil- Part IV of the book describes “Cancers by Tissue of Origin” for
lion deaths from cancer occurred in 2012. This burden is projected to 33 anatomic sites, multiple primary tumors, and cancers in children.
increase to 24 million cases and 13 million deaths annually by 2035 Rapid advances in discovering the molecular events that drive certain
(Ferlay et al., 2013). Chapter 8 decribes the disproportionate increase forms of cancer are transforming clinical diagnoses and treatment, and
in the cancer burden in low-​and middle-​income countries (LMICs), affecting tumor classification. This will influence future endpoints in
which can least afford additional health-​related, social and financial etiologic studies and population-​based cancer surveillance.

1
2

2 Introduction
Part V, “Cancer Prevention and Control,” discusses the impact of (“genome-​wide significance”), large sample size, and replication
interventions that effectively reduce carcinogenic exposures or disrupt in more than one study to exclude chance associations. Most of the
the multistage progression of tumors. It focuses on interventions that associations identified through GWAS are modest (per allele ORs:
demonstrably reduce cancer risk in the general population, rather than 1.5–​2.0) or weak (ORs <1.5), but in aggregate these loci can distin-
in special circumstances or high-​risk subgroups. Examples of these guish a wide range of risk in the population, thus providing oppor-
are discussed in Chapters 61–​63. In all cases, the design and imple- tunities for targeted screening and prevention. While our current
mentation of preventive measures require translational research to knowledge regarding germline risk comes from studies in popula-
ensure safety, optimize feasibility and impact, and critically evaluate tions of European ancestry, the identification of population-​specific
all stages of the process. risk loci highlights the importance of conducting GWAS in diverse
racial and ethnic populations.
Statistical modeling suggests that, for many cancers, additional
Cancer Prevention and Control variants remain to be identified, yet the search for variants with smaller
A growing number of population-​level preventive interventions are effect sizes, as well as less common variants, drives the need for
proving to be highly effective, as confirmed by the decreases in inci- even larger studies. With the recent development of next-​generation
dence as well as mortality rates from certain cancers (Chapters 61–​63). sequence technology, it is now practical to sequence whole exomes
Tobacco control has reduced the age-​standardized incidence rate of (coding regions plus regulatory regions) and whole genomes in
lung cancer by up to 40% among men in high-​and middle-​income population-​and family-​based studies in the search for heritability not
countries. Increased screening for colorectal cancer and removal of identified through common variation in GWAS.
precursor lesions is credited for the 30% decrease in the incidence An important limitation of GWAS is biological interpretation,
rates at this site in the United States. Universal neonatal vaccination as the vast majority of risk variants revealed through GWAS are in
against hepatitis B virus (HBV) has markedly decreased the preva- non-​coding genetic sequences. Functional analyses are underway to
lence of chronic HBV infection and liver cancer at younger ages in address this issue. The process is time-​consuming, however, since
high-​risk areas of East Asia and will yield maximal benefits against it incorporates new bioinformatics tools and a comparison of gene
cancer in the future. The development of safe and effective vaccines expression in tumor and normal tissue to localize the functional SNP
against human papillomavirus (HPV) and less expensive and less oner- and ultimately the affected gene.
ous screening tests for cervical cancer have greatly expanded opportu- There has as yet been little progress in identifying interactions
nities to prevent HPV-​related cancers among women in many LMICs. between inherited germline loci identified through GWAS and acquired
Increased funding is becoming available for application research and “environmental” risk factors. Candidate gene studies have docu-
cancer preventive services in LMICs. Cancer prevention presents both mented gene–​environment interactions between tobacco smoking and
opportunities and challenges, as discussed in Part V of the text. The the slow NAT-​2 acetylation phenotype for bladder cancer (Chapter 52)
best practices developed for tobacco control provide an encourag- and between alcohol consumption and slow ADH1B metabolizers for
ing model of how health-​related policies can address the behavioral esophageal cancer (Chapter  30). However, much larger GWAS with
causes of cancer. However, these must be tailored to fit the particular more precise measures of exposure and risk will be needed to assess
social, economic, and other considerations that affect the exposure other, subtler gene–​environment interactions.
(Chapter 61).
Somatic Genomic Alterations
Most of the somatic genomic alterations, including mutations,
indels, copy number alterations, and chromosomal rearrangements,
Advances in Genomics and other OMICs that drive neoplastic progression in tumor tissue are acquired rather
Technological advances in high-​ throughput genotyping/​ sequenc- than inherited. As mentioned, the Human Cancer Genome Project
ing and gene expression arrays have transformed research on both and other international laboratory and clinical collaborations have
inherited (germline) susceptibility variants and the largely acquired characterized so-​called driver mutations (i.e., those which confer
(somatic) mutations in tumor tissue. Epidemiologic studies of cancer growth advantage to a mutated cell line) for multiple types of human
genetics have focused mainly on germline variants associated with cancer (Chapter 4). These mutations represent the events involved in
cancer risk and etiology, whereas clinical and basic researchers have the multistage development of particular forms of cancer (Armitage
characterized the landscape of somatic alterations in tumor cells that and Doll, 2004; Hornsby et al., 2007; Wu et al., 2016). It is notewor-
drive the development and progression of cancer. thy that discoveries in somatic mutations over the past three decades
provide strong support for the theory of multistage carcinogenesis
Germline Susceptibility Variants that was proposed by Armitage and Doll, 10 years before elucidation
The tools to identify inherited genetic susceptibility variants have of the structure of DNA, and over 30 years before the identification
advanced enormously since publication of the previous edition of of the first proto-​oncogenes and tumor suppression genes (Armitage
this text in 2006. At that time, studies involved either high-​risk fami- and Doll, 1954). Sequencing studies have also implicated epigenetic
lies or the evaluation of a small number of pre-​specified “candidate modification as a major source of alterations in cancer (Chapters 2
genes” in case-control studies of sporadic cancers in the general pop- and 4).
ulation. The candidate gene approach was largely unsuccessful in Variable combinations of genetic and epigenetic abnormalities
identifying robust associations for several reasons, including small account for the phenotypic heterogeneity within and among cancers.
sample size, limited statistical power, failure to account for multi- Molecular characterization of tumors is increasingly used to predict
ple testing (generating negative and false positive results, respec- prognosis and to guide the use of targeted therapies for individual can-
tively), and limited biologic knowledge to inform the selection of cer patients. These markers are only beginning to be evaluated and
candidate genes. Following the completion of the Human Genome integrated into large-​scale epidemiologic studies, yet they are already
Project in 2003, genome-​wide maps of single nucleotide polymor- changing the taxonomy of some types of cancers and are likely to pro-
phisms (SNPs) became available. Advances in high-​ throughput foundly affect future etiologic studies (Chapter  3). There has been
genotyping technology, combined with knowledge about the struc- some progress in efforts to link specific classes of somatic muta-
ture of genetic linkage disequilibrium, created opportunities to con- tions, such as the mutational signatures of ultraviolet (UV) radiation,
duct exploratory (hypotheis-​free or “agnostic”) surveys across the tobacco smoke, and oncogenic viruses, to established carcinogenic
entire genome. Over the past decade, GWAS have robustly identi- exposures (Chapters  2 and 4). These molecular signatures may, in
fied more than 700 common (i.e., minor allele frequency >5%) sus- the future, identify the causal exposure(s) for cancer in individuals as
ceptibility loci associated with cancer risk, as discussed for specific well as populations. Hopefully this goal will motivate interdisciplinary
sites in Part IV, “Cancers by Tissue of Origin.” Because GWAS test collaborations between epidemiologists, cancer prevention scientists,
millions of alleles across the genome, they require stringent criteria geneticists, cancer biologists, and clinicians.
 3

Introduction 3
Other OMICs (Chapter 17), the combustion of coal as household fuel (Chapters 16,
While genomic research is the poster child for the value of agnos- 17, and 28), diesel engine exhaust (Chapter  17), the consumption
tic, comprehensive explorations of germline variants associated with of red and processed meat (Chapter 19), and to a lesser extent, UV-​
cancer, other areas of OMIC research are moving toward this goal emitting tanning beds (Chapter 14). The search for other modifiable
(Chapter  6). The development of technologies to screen many thou- causes of cancer continues. New associations have been reported with
sands of analytes related to gene expression (e.g., RNAseq), epi- shift work, sedentary behavior (as distinct from physical inactivity),
genetics (methylation; ChIP-​Seq), metabolomics, and the microbiome computed tomography scans during childhood, sun-​sensitizing phar-
will open new opportunities to identify the connections between expo- maceutical drugs, and others. While the studies may be methodologi-
sures and the biologic effects that mediate carcinogenesis. Various cally strong, the evidence for causality is not yet considered definitive.
OMIC technologies are at different stages of development. One of the There is a continuing need to monitor both the immediate and long-​
more advanced efforts along these lines is the identification of hor- term effects of more recently implemented medical technologies
mone metabolites that influence breast cancer risk, illustrating the and newly developed drugs, products such as cellular phones and e-​
potential of these new technologies (Chapter 22). cigarettes, nuclear accidents, exposures occurring in war zones, and
other exposures potentially related to cancer.
Technological advancements in biomarker studies will allow more
OUTCOMES AND EXPOSURES comprehensive examination of an individual’s metabolome, microbi-
ome, genome, epigenome, and exposome. The use of specific biomark-
ers to assess internal exposures and identify children, adolescents, and
Changing Taxonomy of Cancer other subsets of individuals who may be particularly susceptible to the
Accurate and reproducible classification of neoplastic diseases is factor being investigated (for example, dietary exposure, pesticide act-
essential for advances in diagnosis and treatment, for quantifying ing as a hormonal disruptor, or medication) could increase our ability
geographic, temporal, and demographic variations in incidence, and to detect complex exposure–​disease relationships.
for identifying etiologic relationships and mechanisms. Tumor clas-
sification has historically been based on the primary anatomic site and
morphology for most solid tumors and on histologic characteristics for
ESTIMATES OF ATTRIBUTABLE FRACTION
leukemias. Classification systems have evolved to incorporate infor-
Epidemiologists have long debated the fraction of cancer cases or
mation on morphology, genetics, cell lineage, developmental char-
deaths that could be avoided by preventive interventions (Chapter 61).
acteristics, and an array of molecular, clinical, and etiologic factors
Estimates of the percent of cancer deaths that could theoretically be
(ICD-​O-​3, 2013) (Fritz et al., 2000).
avoided, if the exposures were eliminated, range from 50% to 80%,
While the refinement of cancer endpoints based on molecular or
although the potential for primary prevention differs for incidence and
other characteristics will potentially increase the ability of etiologic
mortality, by geographic region, gender, and attained age (Whiteman
studies to detect associations with specific tumor subtypes, it also
and Wilson, 2016). About half of the deaths that could be avoided in
poses serious challenges. Very large studies will be needed for both
principle relate to 11 potentially avoidable risk factors, including the
discovery and replication. Even well-​established tumor markers are
behavioral risk factors discussed above. The attributable fraction esti-
not measured uniformly in all patients. Newer classification systems
mates are predicated on the idea that cancer risk is largely acquired,
based on molecular features have generally been evaluated in only a
rather than inherited. Inherited factors do contribute to the variation in
few hundred sporadic cancers, with little consideration of patient or
risk among individuals, but they cannot account for the large tempo-
population characteristics. Clonal heterogeneity within tumors and
ral changes in risk within countries, or the differences in risk among
changes in tumor pathology during treatment further complicate clas-
migrants who move from one country to another.
sification (Norum et al., 2014). While the use of automated algorithms
Chapter 19, “Diet and Nutrition,” provides the first estimates of the
and computer-​based image analysis is increasing among pathologists,
fraction of all cancers attributable to diet, in combination with or sepa-
these methods and the assessment of reproducibility and validity may
rate from overweight and physical inactivity. The authors estimated
not be reported to clinicians, epidemiologists, and others using the data.
the total as about 20% overall, which is weaker than previously esti-
Population-​based cancer registries are already challenged by efforts to
mated. The proportion contributed by dietary composition indepen-
keep abreast of changing tumor classifications, especially when the
dent of adiposity is less clear because some dietary factors have yet to
new criteria are not uniformly applied in a standardized manner.
be identified or established with sufficient certainty, and associations
are likely underestimated because of measurement error or misspecifi-
cation of temporal relationships. The authors estimate an etiologic
Exposures and Exposure Measurement contribution of 5%–​12% for dietary composition alone, but suggest
More than 100 different agents and exposures are now designated as that this could be appreciably higher when considering nutrient and
causally related to cancer in humans (Group I) by the International genetic interactions.
Agency for Research on Cancer (IARC). Variations in the prevalence
and intensity of these exposures account for the striking geographic
and temporal variations in the occurrence of many types of cancer. ONGOING CHALLENGES
While many exposures such as tobacco, alcohol, and numerous indus-
trial chemicals have long been classified as human carcinogens, expo-
sure patterns change, new agents are introduced, the ability to measure Tumor Diagnosis and Classification
exposures or outcomes progresses, and the quality, quantity, and/​or In LMICs, the completeness and specificity of tumor diagnosis var-
specificity of evidence improves. For example, the contining global ies depending on economic resources and medical infrastructure.
increase in obesity, metabolic syndrome, and type II diabetes, com- Less than 10% of people in Africa and South America are covered
bined with improvements in laboratory assays to measure hormones by population-​ based tumor registries (Chapter  8). In high-​ income
in large population studies, has created new opportunities to study countries, tumor classification is more advanced because of earlier
metabolic and hormonal effects on cancer. Thus, the discussion of application of diagnostic innovations and revised classification sys-
“Hormones and Cancer” (Chapter 22) has been expanded to consider tems. Classification systems evolve with the introduction of new histo-
endogenous as well as exogenous exposures and peptide hormones chemical and molecular markers and advances in understanding tumor
(insulin, insulin-​like growth factors, growth hormone, leptin, adipo- biology. Even in high-​income countries, there is wide variability in
nectin, resistin, ghrelin, etc.) in addition to steroidal sex hormones. the proportion of tumors incompletely or inadequately characterized.
Several agents recently classified as Group  1 human carcinogens Molecular profiling at major cancer centers may include a range of
affect massive numbers of people. These include outdoor air pollution established tumor markers, exome or whole genome sequencing, copy
4

4 Introduction
number, messenger and micro RNA sequencing, DNA methylation, to early childhood infection with Epstein-​Barr virus (Chapters 26 and
and proteomics analysis (Hoadley et al., 2014). While this information 39). Although these hypotheses are important, they are difficult to test
may be useful clinically, it is not yet available for most cancer patients, without biomarkers or experiments of nature that demarcate the timing
nor are the data routinely incorporated into cancer surveillance sys- of exposure. Serial acquisition of biological samples over many years
tems (Chapter 8). Even cancers that have undergone intensive multi- of follow-​up would be informative, although this approach must be
disciplinary review to improve classification, such as hematopoietic tempered by feasibility and cost considerations.
and lymphoproliferative malignancies, include subtypes characterized
as “not otherwise specified” or provisionally classified (Swerdlow
et al., 2016). Thus epidemiologic studies of cancer must collect and FUTURE RESEARCH DIRECTIONS
archive tumor tissue in order to ensure a uniform, more complete, and
contemporary approach to molecular testing. While individual chapters outline future research directions for spe-
cific exposures or cancers, we highlight here selected cross-​cutting
issues that apply broadly to many cancers.
Over-​diagnosis
“Over-​diagnosis” refers to the incidental detection of small and/​or Team Science
indolent cancers that otherwise might not cause clinical problems
during the patient’s lifetime (Chapter  8). Extreme examples of this One of the benefits of GWAS and pooled risk factor studies has been
have been the sudden increase in prostate cancer diagnoses follow- the formation of multi-​institutional international consortia to share
ing the introduction of PSA screening (Chapter 53), and the increase biospecimens and primary data in order to maximize statistical power
in thyroid cancer diagnoses due to screening programs using ultra- for discovery and robust replication (Boffetta et al., 2007). These con-
sound (Chapter 44). Overdiagnosis is most problematic if it leads to sortia have been instrumental in creating new models of funding, lead-
unnecessary treatment and serious adverse effects. The introduction ership, and authorship, and in sharing and harmonizing primary data
of new screening tests can also distort temporal trends in incidence across studies. The formation of larger and more complex data sets has
and bias observational studies of the impact of screening (Chapter 63). stimulated innovations in informatics and the development and appli-
The likelihood of over-​diagnosis varies by cancer site and depends cation of novel analytic methods. Further advances in high-​throughput
on the baseline incidence and risk characteristics of a population. laboratory technology will continue to create new opportunities to
An estimated 10–​30% of newly diagnosed breast cancers identified explore the complex biology of genomics, metabolomics, proteomics,
with screening mammography may reflect over-​diagnosis (Bleyer & and so on, in large population studies. This will generate vast amounts
Welch, 2012; Loeb et al., 2014; Vickers et al., 2014). In the absence of of data to be analyzed. It will also require insights from diverse disci-
molecular markers that reliably distinguish indolent from aggressive plines to plan analyses and to interpret the results.
tumors, clinicians must grapple with the potential for “over-​treatment” “Transdisciplinary research” signifies a level of collaboration in
(Chapter 3). Over-​diagnosis poses a greater clinical dilemma for early which researchers from different scientific disciplines come together
stage cancers in internal organs than for premalignant lesions detected to identify the most important research questions, design the optimal
by colorectal or cervical screening, because the treatment is more approach, and collect, analyze, and publish the study results jointly
invasive. (Rosenfield, 1992). This level of team science transcends disciplinary
boundaries and benefits all parties. For example, the application of
haplotype analyses based on the structure of genetic linkage disequi-
Exposure Measurement Issues librium, initially proposed by geneticists, greatly accelerated explor-
atory analyses across the entire genome in large population studies.
Regardless of the epidemiologic study design used, it is challenging
Similarly, the involvement of epidemiologists in tumor genomics has
to characterize accurately many types of acquired exposures due to
brought a population science perspective to this field, increasing atten-
a lack of comprehensive measurements during the relevant exposure
tion to population sampling, sex, and racial/​ethnic differences, and
window. This presents a greater problem for some exposures than oth-
exposures such as smoking that can affect the mutational spectrum
ers. For example, as described in Chapters 19–​21, misclassification of
of various cancers. There are many opportunities for collaborations
exposure is of great concern in characterizing patterns of nutrition and
involving laboratory scientists, analytic chemists, epidemiologists,
physical activity, especially at earlier stages of life. It presents less of a
and others to accelerate research on etiologic issues related to can-
problem in studying cigarette smoking, menopausal hormonal therapy,
cer. One example would be transdisciplinary research to understand
or exposure to microbial agents, since these exposures can be reason-
hormonal carcinogenesis at the molecular level in human populations
ably well defined qualitatively, and biomarkers exist to supplement
(Chapter 22).
questionnaire data. Certain exposures are experienced in multiple
settings, including workplace, residential, recreational, medical, war
zone, and other settings. Chemical exposures often occur as mixtures Cancer Survivorship
in air or water. Surrogate measures, such as job titles for occupational
exposures and administrative databases for residential exposures, do The number of people surviving after a diagnosis of cancer has
not capture variations among individuals or over time. increased rapidly in high-​income and many middle-​income countries
The timing of exposure is an area of special interest and challenge. due to improvements in treatment and the effects of screening on both
Exposures that occur during a particular time window or a susceptible early diagnosis and more complete ascertainment. In the United States,
stage of development may have adverse effects that are not evident the estimated number of people alive for at least 5 years after a diagno-
when exposure occurs later in life. A classic example of this involved sis of cancer increased from 4 million in 1978 (~1.8% of the popula-
in utero exposure to high doses of the hormone di-​ethyl stilbesterol tion) to 13.7 million in 2012 (~4% of the population), and is predicted
(DES) in the daughters of mothers treated to prevent pregnancy com- to approach 18 million by 2022 (de Moor et al., 2013; Harrop et al.,
plications, who subsequently developed vaginal carcinoma in adoles- 2011). A substantial proportion of these are long-​term survivors. Forty
cence (Chapter 49). For breast cancer, it is hypothesized that hormonal percent of individuals who survived for at least 5  years were alive
exposures received in utero or immediately postnatally may affect 10 or more years after diagnosis, and 15% had survived 20 or more
early stages in tumor development, or that breast tissue may be more years (Howlader et al., 2015). To address the rapidly increasing pop-
susceptible to certain exposures (e.g., ionizing radiation, cigarette ulation of cancer survivors, the NCI Office of Cancer Survivorship
smoking, or alcohol consumption) during the period between men- and a 2006 publication from the Institute of Medicine (Committee
arche and first full-​term pregnancy, when cells are proliferating but not on Cancer Survivorship, 2006) provided a framework for identifying
fully differentiated (Chapter  45). Similar hypotheses have been pro- and addressing the unmet needs of this growing population. Disease
posed for nasopharyngeal cancer and Hodgkin lymphoma in relation and treatment affect multiple health domains (e.g., medical, physical
 5

Introduction 5
function, psychiatric and psychosocial, cognitive, work, sexual, and gastrointestinal system, yet in the United States accounts for only
reproductive). These factors and heath-​related quality of life should 3.2% of digestive system cancer cases (Howlader et al., 2015).
be assessed at discrete intervals following diagnosis. Studies should The tools for studying complex microbial communities are only now
examine whether quality of life and survivorship issues differ for can- becoming available. There is great interest, however, in characterizing
cers in children from those in later life. Research on survivorship is alterations in the microbiome caused by changes in diet, antibiotic
largely still in its infancy (de Moor et al., 2013; Harrop et al., 2011; use, and other exposures, and in the inflammatory responses that result
Richardson et al., 2011). The occurrence of multiple primary and sec- from the influx of pathogenic organisms into organ-​specific microbi-
ondary cancers following treatment is an important area of research omes (Dale and Moran, 2006). Future epidemiologic and experimental
(Chapter  60). Cancer epidemiologists should have a major research research on the human microbiome will likely continue to focus on
role in cancer survivorship because of their expertise in observational the carcinogenic effects of disturbed homeostasis and the disruption
study designs and exposure assessment. of the normal diversity of the microbial flora (Bultman, 2016), and
as well as on potential role(s) of the microbiome on the pathogenesis
of obesity, diabetes (Okeke et  al., 2014), and autoimmune diseases
Risk Prediction Models (Hooper et al., 2012).
Risk prediction models estimate the absolute risk of being diagnosed
with or dying from a specific condition during a defined time period
Development of New Cohorts
for individuals with defined demographic characteristics and risk fac-
tors. These models are widely used to predict individual risk and to There is an ongoing need to develop new cohorts to complement exist-
guide treatment decisions in cardiovascular medicine. Unfortunately, ing cohorts, with a particular focus on collecting and banking biologi-
the models currently available for cancer provide reasonably accu- cal samples needed for broader OMICs studies (e.g., tumor tissue,
rate predictions for groups of people but not for individuals (Amir blood, urine, stool), the inclusion of state-​of-​the-​art exposure mea-
et al., 2010). surements and data collection strategies, data on new and emerging
Site-​specific chapters discuss research on risk prediction models exposures, and repeated assessments over time to capture important
for cancers of the lung (Chapter 28), colorectum (Chapter 36), breast changes in exposures, behaviors, or physiological factors. Besides
(Chapter  45), and multiple primary cancers (Chapter  60). Brinton enrolling participants from more recent birth cohorts, there is also a
and colleagues in Chapter 45 review the research on factors that may critical need to increase the racial/​ethnic and socioeconomic diversity
improve the discriminatory accuracy of existing models of breast can- of participants. Careful consideration must be given to cost-​effective
cer. These include endogenous hormone levels, proliferative benign data sources and methods of data collection to address these needs in
biopsy diagnoses, behavioral risk factors, mammographic density, and the long-​term follow-​up of future cohorts. Data access and data shar-
genetic polymorphisms. Although the associations with genetic poly- ing also need to be considered since data collected from more recent
morphisms identified by GWAS are modest (per allele ORs: 1.5–​2.0) cohorts (such as the UK Biobank, the NCI Cohort Consortium, and
or weak (ORs <1.5), as discussed earlier, it is hoped that in the aggre- the Childhood Cancer Survivor Study) are increasingly made broadly
gate the addition of these loci to the model can more accurately predict available as a resource for qualified investigators. Finally, new models
risk for individuals, thereby improving targeted approaches for screen- of governance and participant engagement and interaction will need to
ing and prophylactic treatment (Garcia-​Closas et al., 2014). be developed for the next generation of cohort studies.
Risk prediction models of colorectal cancer have been used to esti-
mate the independent and combined effects of behavioral, medical,
familial and genetic risk factors on attributable risks. One such study New and Non-​Traditional Data Sources
estimated that a population with the optimal distribution of established A variety of new data sources are enriching the opportunities for
modifiable risk factors for colorectal cancer would have a 43% percent descriptive and analytic epidemiologic studies. The expanding use of
lower (95% CI:  0.14, 0.65) incidence of disease (Lee et  al., 2016). electronic health records, along with a movement toward interoperabil-
Meester et al. (2015) estimated the effect of screening on deaths from ity and standardized content, can potentially provide individual-​level
colorectal cancer and concluded that the non-​use of screening methods medical data on tumor characteristics, comorbidity, and treatment.
among people aged ≥ 50 years in the United States accounted for more Data linkage between population-​based cancer registries, vital status
than 50% of colorectal cancer deaths. and mortality records, hospital and other medical records, and admin-
istrative data sets provides an important resource for studies of cancer
survivorship. Personal devices such as accelerometers or smart phones
The Microbiome to track movement can improve the measurement of physical activity.
Remarkable advances have been achieved in identifying microbial In parallel, many other types of digital data are increasingly avail-
pathogens that influence cancer risk (Chapter 24). This research has able. Potentially linkable data from diverse fields of science (e.g.,
focused historically on disease-​causing organisms, rather than on the OMICs; environmental and geographic sciences) and areas outside
trillions of “commensal” organisms that live in or on the human body. of science (e.g., business, finance, telecommunications, social media,
Increased interest in the human microbiome has broadened the scope and the Internet of things) are being promoted under the umbrella of
of inquiry. The greatest concentration of commensal microorgan- “Big Data.” Non-​traditional data sources have been touted as a rev-
isms is in the upper and lower gastrointestinal tract, although there olutionary development in the future of epidemiology (Khoury and
are well-​ characterized symbiotic communities in the skin, mouth, Ioannidis, 2014; Mooney et al., 2015). It should be recognized that the
sinonasal cavities, and vagina. Diverse species of bacteria, archaea, use of non-​traditional data sources involves much more than working
viruses (including bacteriophages), and fungi can be identified. Some with large volumes of data. These sources have been characterized as
species have coevolved with humans for millennia and maintain essen- high variety (secondary use from many diverse sources), high volume
tial physiologic enzymatic functions, such as the synthesis of essential (both in numbers of observations and/​or variables per observation) and
vitamins and the metabolism of carbohydrates, bile acids, and xeno- high velocity (real time) (Douglas, 2012). It is important that epide-
biotics. Commensal flora may have detrimental as well as beneficial miologists collaborate with informaticians and computer scientists
effects on cancer. The colonic microbiome is thought to account for to explore the promise of Big Data. Expertise in observational study
the approximately 40-​fold higher incidence of adenocarcinoma in the design, along with efforts to assess validity and reproducibility, will
colon than in the small intestine (Chapter 35). While the small intes- be essential to avoid “big error” driven by chance, bias (e.g., selection
tine is not sterile, it has comparatively few commensal organisms. The bias, measurement error, confounding) and lack of external validity.
difference in cancer risk between the two sites is remarkable, given Cancer epidemiologists bring the subject matter expertise needed to
that both organs undergo rapid cell replication. The small intestine frame and interpret Big Data results for clinical and public health rel-
comprises about 90% of the mucosal absorptive surface area of the evance. While the long-​term impact of Big Data on cancer surveillance
6

6 Introduction
and analytical epidemiology is not yet clear, we anticipate that this Howlader N, Noone AM, Krapcho M, et  al. 2015. SEER Cancer Statistics
may have a major impact on the field over the next decade. Review, 1975–​2012, National Cancer Institute. Bethesda, MD, http://​seer.
cancer.gov/​csr/​1975_​2012/​, based on November 2014 SEER data submis-
sion, posted to the SEER Web site, April 2015.
References
Khoury MJ, and Ioannidis JP. 2014. Medicine: Big Data meets public health.
Amir E, Freedman OC, Seruga B, and Evans DG. 2010. Assessing women at Science, 346(6213), 1054–​1055. PMCID: PMC4684636.
high risk of breast cancer:  a review of risk assessment models. J Natl Lee DH, Keum N, and Giovannucci EL. 2016. Colorectal cancer epidemiology
Cancer Inst, 102(10), 680–​691. PMID: 20427433. in the Nurses’ Health Study. Am J Public Health, 106(9), 1599–​1607.
Armitage P, and Doll R. 1954. The age distribution of cancer and a multi-​ PMCID: PMC4981802.
stage theory of carcinogenesis. Br J Cancer, 8(1), 1–​ 12. PMCID: Loeb S, Bjurlin MA, Nicholson J, et  al. 2014. Overdiagnosis and
PMC2007940. overtreatment of prostate cancer. Eur Urol, 65(6), 1046–​ 1055.
Bleyer A, and Welch HG. 2012. Effect of three decades of screening mammog- PMCID: PMC4113338.
raphy on breast-​cancer incidence. N Engl J Med, 367(21), 1998–​2005. Meester RG, Doubeni Ca, Lansdorp-​Vogelaar I, et  al. 2015. Colorectal can-
Boffetta P, Armstrong B, Linet M, et al. 2007. Consortia in cancer epidemiol- cer deaths attributable to nonuse of screening in the United States. Ann
ogy: lessons from Interlymph. Cancer Epidemiol Biomarkers Prev, 16(2), Epidemiol, 25(3), 208–​213 E201. PMCID: PMC4554530.
197–​199. Mooney SJ, Westreich DJ, and El-​ Sayed AM. 2015. Commentary:  epi-
Bultman SJ. 2016. The microbiome and its potential as a cancer preventive demiology in the era of big data. Epidemiology, 26(3), 390–​ 394.
intervention. Semin Oncol, 43(1), 97–​106. PMCID: PMC4789109. PMCID: PMC4385465.
Committee on Cancer Survivorship. 2006. Improving Care and Quality Norum JH, Andersen K, and Sorlie T. 2014. Lessons learned from the intrinsic
of Life, National Cancer Policy Board. From Cancer Patient to subtypes of breast cancer in the quest for precision therapy. Br J Surg,
Cancer Survivor:  Lost in Transition. Washington, DC:  National 101(8), 925–​938.
Academies Press. Okeke F, Roland BC, and Mullin GE. 2014. The role of the gut microbiome in
Dale C, and Moran NA. 2006. Molecular interactions between bacterial symbi- the pathogenesis and treatment of obesity. Glob Adv Health Med, 3(3),
onts and their hosts. Cell, 126(3), 453–​465. 44–​57. PMCID: PMC4030605.
De Moor JS, Mariotto AB, Parry C, et  al. 2013. Cancer survivors in the Richardson A, Addington-​Hall J, Amir Z, et al. 2011. Knowledge, ignorance
United States:  prevalence across the survivorship trajectory and impli- and priorities for research in key areas of cancer survivorship:  find-
cations for care. Cancer Epidemiol Biomarkers Prev, 22(4), 561–​570. ings from a scoping review. Br J Cancer, 105 Suppl 1, S82–​ S94.
PMCID: PMC3654837. PMCID: PMC3251948.
Douglas L. 2012. The Importance of Big Data:  A  Definition. Stamford, Rosenfield PL. 1992. The potential of transdisciplinary research for sustaining
CT: Gartner. and extending linkages between the health and social sciences. Soc Sci
Ferlay J, Soerjomataram I, Ervik M, et  al. 2013. GLOBOCAN 2012 V1.0, Med, 35(11), 1343–​1357.
Cancer Incidence and Mortality Worldwide:  IARC Cancerbase No. 11 Schottenfeld D, and Fraumeni JF. 1982. Cancer Epidemiology and Prevention.
[Internet]. Available from: http://​globocan.iarc.fr. 1st ed. Philadelphia: Saunders.
Fritz A, Percy C, Jack A, et al. 2000. International Classification of Diseases Schottenfeld D, and Fraumeni JF. 1996. Cancer Epidemiology and Prevention.
for Oncology, 3rd ed. Geneva: World Health Organization. 2nd ed. Oxford: Oxford University Press.
Garcia-​Closas M, Gunsoy NB, and Chatterjee N. 2014. Combined associations Schottenfeld D, and Fraumeni JF. 2006. Cancer Epidemiology and Prevention.
of genetic and environmental risk factors: implications for prevention of 3rd ed. Oxford: Oxford University Press.
breast cancer. J Natl Cancer Inst, 106(11). PMCID: PMC4271030. Smith MT, Guyton KZ, Gibbons CF, et  al. 2016. Key characteristics of car-
Harrop JP, Dean JA, and Paskett ED. 2011. Cancer survivorship research:  a cinogens as a basis for organizing data on mechanisms of carcinogenesis.
review of the literature and summary of current NCI-​designated cancer Environ Health Perspect, 124(6), 713–​721. PMCID: PMC4892922.
center projects. Cancer Epidemiol Biomarkers Prev, 20(10), 2042–​2047. Swerdlow SH, Campo E, Pileri SA, et  al. 2016. The 2016 revision of the
PMCID: PMC3191883. World Health Organization classification of lymphoid neoplasms. Blood,
Hoadley KA, Yau C, Wolf DM, et al. 2014. Multiplatform analysis of 12 cancer 127(20), 2375–​2390. PMCID: PMC4874220.
types reveals molecular classification within and across tissues of origin. Vickers AJ, Sjoberg DD, Ulmert D, et al. 2014. Empirical estimates of prostate
Cell, 158(4), 929–​944. PMCID: PMC4152462. cancer overdiagnosis by age and prostate-​specific antigen. BMC Med, 12,
Hooper LV, Littman DR, and Macpherson AJ. 2012. Interactions between the 26. PMCID: PMC3922189.
microbiota and the immune system. Science, 336(6086), 1268–​1273. Whiteman DC, and Wilson LF. 2016. The fractions of cancer attributable to
PMCID: PMC4420145. modifiable factors: a global review. Cancer Epidemiol, 44, 203–​221.
Hornsby C, Page KM, and Tomlinson IP. 2007. What can we learn from the Wu S, Powers S, Zhu W, and Hannun YA. 2016. Substantial contribution of
population incidence of cancer? Armitage and Doll revisited. Lancet extrinsic risk factors to cancer development. Nature, 529(7584), 43–​47.
Oncol, 8(11), 1030–​1038. PMCID: PMC4836858.
 7

I
BASIC CONCEPTS
8
 9

2 Biology of Neoplasia

MICHAEL DEAN AND KAROBI MOITRA

OVERVIEW major genetic and external risk factors for most cancers, the principal
genes that are altered somatically in tumors, and many of the mecha-
The biology of tumors is being understood in unprecedented detail in nisms. While external factors, like mutagens and infectious agents, initi-
terms of the development, growth, survival, and spread of cancer cells ate cancer, in the later stages of tumor development we find the tumor
in the body, and their interaction with the host. Analyses of familial cell tapping into diverse tools and pathways that are the basis of life itself.
cancer syndromes and in vitro and animal assays have revealed the
major tumor suppressor genes and oncogenes that regulate cell growth
and survival. More recent scans of large numbers of cancer cases and Definition of Cancer
controls with single nucleotide polymorphism (SNP) arrays, known as • “Cancer” refers to a large, heterogeneous group of diseases with
genome-​wide association studies (GWAS), have identified additional common underlying pathology characterized by uncontrolled cel-
genetic loci, mostly in regulatory regions that influence cancer risk. lular growth and division.
The genomic characterization of tumors through exome and whole • Cancer is inherently a genetic disease at the cellular level.
genome sequencing, transcriptome, and DNA methylation analyses • Genetic and epigenetic changes accumulate in localized (somatic)
are identifying further complexity in the somatic alterations present tissue and dysregulate genetic control over basic cellular functions.
in tumor cells. A new field of tumor signature analysis seeks to cor-
relate exposures (tobacco, carcinogens, radiation) to specific classes Cancer involves all of the organ systems and cell types of the body
of mutations in the tumor genome. Alterations in the DNA repair pro- (although some tissues are more susceptible than others) and is there-
cesses of the cell also can be shown to leave a signature in the point fore inherently more complicated than other major disease such as car-
mutations and structural alterations found in cancer. A major new class diovascular or neurological disease.
of somatically altered genes are those encoding proteins that modify Cancer is a genetic disease in the sense that the tumor cell has
histones and other components of chromatin and/​or alter the methyla- suffered from multiple lesions in its DNA, nearly always involving
tion of DNA. Such epigenetic alterations appear to be central to late multiple gene mutations, chromosome rearrangements, and extensive
events in cancer, such as metastasis and therapy resistance, and further alteration of the epigenome, through changes in DNA methylation and
understanding of these alterations will be the key to effective therapy. histone modification (Figure 2–2). Depending on the tumor type, up to
Single-​cell analyses are identifying complex patterns of cancer cell 10% of cancers are caused by inherited germ-​line mutations that are
heterogeneity and evolution and have supported the concept of the moderately to highly penetrant. Common germline variants raising or
tumor cell as a multi-​cellular entity. Finally, the cancer cell arises from lowering cancer risk 0.1–​4-​fold have also been described for nearly
the body’s normal cells, and interacts throughout the entire lifetime of every tumor type.
the tumor with surrounding host cells, blood supply, and the immune Cancers begin as localized growths or pre-​malignant lesions. In a
system. Inflammation is both a major cause and consequence of can- number of cancer types, we can see these growths in the form of colon
cer, and increasingly modulators of the immune system are being used polyps, cervical dysplasia, enlarged moles, and ductal carcinoma in
in cancer therapy. situ, among others. Many of these early lesions will not progress or
can regress or disappear spontaneously. Early colon cancer lesions are
among the best studied of these growths. Nearly all colon polyps have
INTRODUCTION been observed to have inactivating mutations in the APC tumor sup-
pressor gene (Kinzler and Vogelstein, 1996). The APC protein func-
tions as an antagonist of WNT signaling, and was first identified in
The Odyssey of a Tumor familial adenomatous polyposis (FAP), an autosomal dominant dis-
ease characterized by a large number of colon polyps and a high risk
In the epic poem The Odyssey, Homer details the long, adventurous
for colon cancer.
journey of Odysseus as he returns home from the Trojan Wars. Over
A very critical transition is the evolution or progression of the
the course of this decade-​long, arduous journey, Odysseus must use
pre-​malignant lesion into a local, malignant tumor. This stage
all his talents, physical strength, prowess as a soldier, and intelligence
involves the accumulation of yet more genetic insults. Few local
to survive. At times the gods are against him, and at other times they
malignant lesions can be eliminated by the body. The final transition
actually come to his aid. In the end, he is the only surviving member
is to an invasive and/​or metastatic tumor that is capable of invad-
of his army.
ing adjacent tissues or spreading across the body. The vast majority
There is a similar long and difficult path of a tumor from a single
of cancer morbidity and mortality is due to cancers in this third,
abnormal cell to a mature and often lethal growth (Figure 2–1). In the
metastatic stage.
odyssey of the tumor, though, our own cells are the villains of the epic,
and it is science and medicine (rather than the gods) that are called
upon to wage the epic battle against our own cells. The initial tumor Inherited Cancer Syndromes
can arise at virtually any time of our life, from just after birth until old Family history has been noted as a risk factor since the early days
age, and in fact cancer risk continues to increase at least until 80 years of medicine, but the major inherited cancer syndromes began to be
of age (Harding et al., 2008; Pedersen et al., 2016). described in the 1900s. For example, the von Hippel Lindau syndrome
Tumors can arise from virtually any cell type or tissue in the body. was independently described by von Hippel and Landau in 1904 and
Despite this diversity, there are certain traits in common with tumors in 1927 and involves tumors of the kidney and pheochromocytomas
different individuals and from different tissues. We have identified the inherited in an autosomal dominant fashion.

9
10

10 Part I:  Basic Concepts


(d)
(a)

Normal
Cell (b) (c)

Therapy
Resistant
Tumor
Germline
Mutant
Cell

Pre-Malignant
Lesion
Locally
Genetic Invasive
Susceptible Cell Lesion

Metastatic
Tumor

Figure 2–1.  Odyssey of a cancer cell. (a) Cancer cells derive from normal cells and form a pre-malignant lesion. Inherited mutations can predispose to
pre-malignant lesions and cancers (germline mutations), and over 500 cancer susceptibility alleles are known. (b) Pre-malignant lesion can be studied
(colon polyps, cervical lesions), and in some cases regress. (c) Or, pre-malignant lesions can progress to local invasive cancer (carcinoma in situ). (d) The
final phase involves the development of the fully malignant, invasive, and metastatic tumor, with the potential to develop resistance to therapy (radiation,
chemotherapy, targeted or immune therapy).

The Role of Tumor Suppressor Genes are almost always genes that are mutated in sporadic cancers (Table 2–1).
Most of the genes causing these inherited syndromes have turned out to
A major conceptual breakthrough came in 1975 when Al Knudson pub- play critical roles in the cell cycle or growth control, and therefore their
lished an analysis of retinoblastoma and correctly predicted that indi- loss imparts a lack of control of the cell-​division process and/​or loss of
viduals inherited a defective copy of a gene (that we now call a tumor recognition of damage and checkpoints to cell cycle progression. Calling
suppressor gene) and that the tumor cells suffer a somatic loss or loss them tumor suppressor genes explains the role of these genes in cancer
of function of the normal copy (Knudson, 1971). Therefore, individuals formation, but not their role in normal growth and development.
affected by these syndromes often display multiple tumors such as bilat- Genes involved in DNA repair constitute another class of genes that
eral breast cancer or retinoblastoma, multiple kidney or colon tumors cause inherited cancer. Again, they are mutated in the germline, but
(Knudson, 2002). Although the inheritance pattern is dominant, at the the tumor has a further mutation or loss of the normal allele (the sec-
cellular level, both copies of the gene must be inactivated or disabled. ond hit). These tumors contain lesions in critical components of DNA
The same tumor suppressor gene can also suffer mutation or loss of repair and therefore can accumulate lesions that can lead to cancer.
both alleles in a person without an inherited defect (spontaneous muta- A critical conclusion made by Knudson was that the same gene that
tion). Therefore, the genes identified from inherited cancer syndromes causes an inherited cancer syndrome can suffer somatic mutation to
both copies and contribute to sporadic cancer. He demonstrated math-
ematically that this can explain the tendency for inherited retinoblasto-
(a) (b) (c) (d) (e) mas to occur bilaterally and at an earlier age of onset, whereas tumors
Genetic Point Mutation Point Insertion/ Gene Gene in patients without a family history are more likely to be unilateral and
Lesions (SNV) Deletion Translocation Deletion/Gain appear at a later age. This model has held true for many of these genes,
(indel) (CNA)
and APC (colon cancer), RB1 (retinoblastoma), VHL (renal tumors),
PTCH1 (basal cell carcinoma), and TP53 (multiple tumor types) are
GGG GGG very frequently somatically mutated (Hahn et al., 1996; Kinzler et al.,
Gly Gly
Genetic 1991; Latif et al., 1993; Murphree and Benedict, 1984; Varley, 2003).
Susceptible
Several of these genes have been termed “gatekeepers,” as they are
GAG GG often the first gene mutated in the tumor and/​or are inactivated in close
Germline
Mutant
Ser X to 100% of specific tumor types (Kinzler and Vogelstein, 1997). This
is consistent with the critical role that the gatekeeper genes play in
Figure  2–2. Genetic lesions in cancer cells. (a) Some individuals have the formation of the pre-​malignant lesion, the necessary precursor to
germline mutations (rare, highly penetrant mutations) or susceptibility the local malignancy. For example, patients with germline APC muta-
alleles. (b) Point mutations result in the replacement of a single nucleo- tions develop hundreds of colon polyps, and virtually all sporadic
tide, a single-nucleotide variant (SNV). These can cause the replacement colon polyps display somatic mutation of the APC gene (Kinzler and
of a single amino acid in a protein (non-synonymous variant), create a new Vogelstein, 1996).
stop codon (nonsense mutation) or alter a splice site or regulatory element. There are exceptions to this paradigm, mostly in the DNA repair
(c) Insertions or deletions (indels) of one or more nucleotide in the coding genes. Germline mutations in the BRCA1 and BRCA2 genes confer
region of a gene can result in a shift in the reading frame and an inactive high penetrance for breast cancer, moderate penetrance for ovar-
protein. (d) Chromosome translocations result in the formation of a fusion ian cancer, and risk for prostate, pancreas, and other tumors. In the
protein containing portions of two previously distinct genes. (e) An entire tumors of such germline-​mutated patients, there is virtually always
gene can be deleted or amplified. Copy number alterations (CNA). inactivation of the normal allele, consistent with the two-​hit model
 1

Biology of Neoplasia 11
Table 2–1. Major Inherited Cancer Genes

Type of Cancer Syndrome Gene(s)

All cancers Bloom syndrome BLM


Breast, ovarian, pancreatic, prostate Breast-​ovarian cancer syndrome BRCA1, BRCA2
Breast, thyroid, endometrial Cowden syndrome PTEN
Colorectal Familial adenomatous polyposis (FAP) APC
Colorectal Hereditary non-​polyposis colorectal MLH1, MSH2, MHS6, PMS2
cancer/​Lynch syndrome
Retinal Familial retinoblastoma RB1
Melanoma Familial atypical multiple mole-​melanoma CDKN2A
syndrome (FAMM)
Leukemia Fanconi’s anemia FACC, FACA
Pancreatic Hereditary pancreatitis/​familial pancreatitis PRSS1, SPINK1
Leukemias, breast, brain and soft Li-​Fraumeni TP53
tissues cancer
Pancreatic cancers, pituitary adenomas, Multiple endocrine neoplasia 1 MEN1
benign skin and fat tumors
Skin and brain Nevoid basal cell carcinoma syndrome PTCH1
Thyroid, pheochromocytoma Multiple endocrine neoplasia 2 RET, NTRK1
Pancreatic, liver, lung, breast, ovarian, Peutz-​Jeghers syndrome STK11/​LKB1
uterine, and testicular
Tumors of the spinal cord, cerebellum, Von Hippel-​Lindau syndrome VHL
retina, adrenals, and kidneys
Kidney Wilms tumor WT1
Skin Xeroderma pigmentosum XPD, XPB, XPA
Leukemias and lymphomas Ataxia telangiectasia ATM

Source: Adapted from: http://dceg.cancer.gov/research/what-we-study/gene-host/hereditary-cancer-syndrome and the AACR Cancer Progress


Report, 2015.

(Couch et al., 2014; Miki et al., 1994; Moran et al., 2012; Struewing 1973). The hybrid protein contains a portion of BCR at the N termi-
et al., 1997; Wooster et al., 1995). However, BRCA1 and BRCA2 are nus and the kinase portion of ABL at the C terminus. This leads to the
not gatekeeper genes for breast and ovarian cancer, as they are rarely production of an activated ABL tyrosine kinase constitutively signal-
somatically mutated in breast or ovarian tumors in non-​hereditary ing and stimulating cell division (Clark et al., 1988). Certain cancer cell
cases (Cancer Genome Atlas Research Network, 2011, 2012). types are known to be dependent on this oncogene signal for sustained
At the molecular level, both inherited and somatic mutations of cell growth and are known as “oncogene addicted.” In the case of the
tumor suppressor genes serve to inactivate the protein. They are fre- Philadelphia translocation, ABL tyrosine kinase inhibitors can inhibit
quently frameshift or nonsense mutations generating a truncated pro- CML cells and, barring the appearance of drug resistance, can perma-
tein, and are less likely to be point mutations causing the change of a nently keep the tumor cells suppressed (Druker et al., 2001a, 2001b).
single critical amino acid.
Oncogenic Driver Genes
Somatic Mutations and Oncogenes The advent of large-​scale exome and whole genome sequencing of
Oncogenes are the other major class of genes that are frequently tumors has led to an explosion of information on the extent of somatic
mutated in cancer. These are genes that are activated by mutation, cre- mutations in tumors (Vogelstein et al., 2013). There is a high variation
ating a molecule with a dominant function in the cell (Croce, 2008; in the number of somatic mutations per tumor, with pediatric tumors
Dean, 1997). These genes are almost never found to be mutated in the and most hematopoietic tumors showing low mutation load, and solid
germline, and are often lethal when introduced in the activated form tumors higher mutation loads (Kandoth et al., 2013). Critical to this work
in the germline of a mouse. Most oncogenes were discovered as the is the elucidation of the functional mutations in a given tumor and the
cellular homologues of viral oncogenes (Stehelin et al., 1976; Watson separation of these mutations (driver mutations) from random mutations
et al., 1982), or by cellular assays in which DNA from a tumor cell was accumulating in the tumor that may have little or no effect on the tumor-
introduced into immortal cells in culture and a morphological change igenesis process (passenger mutations). Vogelstein et al. developed an
was noted, or the cells became tumorigenic in the mouse (Der et al., operational definition to define oncogenes and tumor suppressor genes
1982; Pulciani et al., 1982). based on the frequency of certain types of mutations (Lawrence et al.,
Most oncogenes have a direct role in cell growth and can be growth 2013; Vogelstein et al., 2013). But mathematical calculations have also
factors (PDGFA, KITL), growth factor receptors (MET, EGFR, been performed to determine if a gene is significantly mutated in a given
PDGFR), intracellular growth signaling molecules (RAS, PIK3CA, tumor type based on the size of the gene (Lawrence et al., 2013). For
RAF, MAPK1), or transcription factors mediating growth signals most of the well-​known oncogenes and tumor suppressor genes, there is
(MYC, FOS) (Croce, 2008; Dean, 1997; Dean et al., 1985; Weinberg, experimental evidence in cell culture or model systems to demonstrate
1996). The mutations in these genes tend to be either specific muta- that they drive cell growth or tumor formation at the cellular level. But
tions that activate the protein (G12E in RAS, H1047Y in PIK3CA) genome sequencing of tumors has identified many frequently mutated
or gene amplifications creating an overabundance of the protein. The genes that have not been experimentally validated.
mutations activating oncogenes are among the most common somatic
events in multiple cancer types.
Chromatin Remodeling Genes
Oncogenes can also be activated by chromosomal translocations
that generate fusion proteins. In the example of the Philadelphia chro- The genes most frequently mutated in cancers that were not discovered
mosome, the t(9;21) event joins the BCR protein on chromosome 9 to as oncogenes or tumor suppressors are the class of genes involved in
the ABL oncogene on chromosome 21 (de Klein et al., 1982; Rowley, the modification of histone molecules found in chromatin, or proteins
12

12 Part I:  Basic Concepts


directly involved in the position and remodeling of nucleosomes on Table 2–2. Continued
DNA. The role of nearly all of these genes as frequently mutated genes
was discovered during exome or whole genome sequencing of large Gene Symbol RefSeq ID Chromosome Location
numbers of specific tumors (Kishimoto et  al., 2005; Muraoka et  al., CXXC1 NM_​001101654.1 18q21.1
1996; Santos-​Rosa and Caldas, 2005). For example, the exome sequenc- DEK NM_​001134709.1 6p22.3
ing of bladder tumors led to the identification of KDM6A, a previously DMAP1 NM_​001034023.1 1p34.1
obscure enzyme functioning as a lysine methylase of histones, as one of DPF1 NM_​001135155.2 19q13.2
the most frequently mutated genes in this tumor type (Gui et al., 2011; DPF2 NM_​006268.4 11q13.1
Guo et al., 2013). In fact, four of the five mutated genes in bladder can- DPF3 NM_​001280543.1 14q24.2
cer come from this class. And for nearly every solid tumor, one or more EP400 NM_​015409.4 12q24.33
chromatin remodeling genes are commonly inactivated (Table 2–2). ERCC6 NM_​000124.3 10q11.23
GADD45A NM_​001199741.1 1p31.2
GADD45B NM_​015675.3 19p13.3
Epigenetic and Regulatory Changes in Cancer GADD45G NM_​006705.3 9q22.1-​q22.2
At the molecular level, most of these chromatin remodeling genes HCFC1 NM_​005334.2 Xq28
have the same spectrum of mutations as the tumor suppressor genes, HELLS NM_​001289067.1 10q23.33
frameshift, and nonsense mutations; and both alleles are inactivated HLTF NM_​003071.3 3q25.1-​q26.1
in the tumor cell. However, they are not found as germline mutations HMG20B NM_​006339.2 19p13.3
causing cancer, but they often cause developmental disorders (Santos-​ IKZF1 NM_​001220766.1 7p12.2
Rosa and Caldas, 2005). ING3 NM_​019071.2 7q31.31
Therefore, tumor genome sequencing has uncovered a major new INO80 NM_​017553.2 15q15.1
class of genes (epigenetic modifiers) that is commonly mutated in can- INO80B NM_​031288.3 2p13.1
cer. In fact, in nearly every tumor type, there is at least one gene from INO80C NM_​001098817.1 18q12.2
this class that is frequently mutated. While in a few cases, genes from INO80D NM_​017759.4 2q33.3
this class have been shown to accelerate cell growth, invasion, and INO80E NM_​173618.1 16p11.2
tumor growth in model systems, in the majority of these genes, we KAT5 NM_​001206833.1 11q$
do not have a molecular mechanism explaining their function in the LRWD1 NM_​152892.1 7q21.1
cancer process. One exciting idea is that the tumor cell may need to MBD5 NM_​018328.4 2q23.1
undergo a wholesale change in gene expression and regulation and that MBD6 NM_​052897.3 12q13.3
the loss of critical chromatin regulatory genes allows the tumor cell to MTA1 NM_​001203258.1 14q32.33
be more plastic and to turn on programs of gene expression that enable MTA3 NM_​004689.3 2p21
the more complex phenotypes of invasion and metastasis. MYBBP1A NM_​001105538.1 17p13.3
Except for rare embryonic mutations and certain pediatric MYO1C NM_​001080779.1 17p13.3
tumors, cancers develop over decades and involve the accumulation NASP NM_​001195193.1 1p34.1
NCOA2 NM_​006540.2 8q13.3
NFRKB NM_​001143835.1 11q24.3
Table 2–2. Chromatin Remodeling Genes PHF10 NM_​018288.3 6q27
PHF17 NM_​001287437.1 4q28.2
Gene Symbol RefSeq ID Chromosome Location PHF19 NM_​001009936.2 9q33.2
PIWIL4 NM_​152431.2 11q21
ACTB NM_​001101.3 7p22.1
PSIP1 NM_​001128217.1 9p22.3
ACTL6A NM_​004301.3 3q26.33
RAD54B NM_​001205262.2 8q22.1
ACTL6B NM_​016188.4 7q22.1
RAD54L NM_​001142548.1 1p32
ACTR3B NM_​001040135.2 7q36.1
RAD54L2 NM_​015106.2 3p21.2
ACTR5 NM_​024855.3 20q11.23
RB1 NM_​000321.2 13q14.2
ACTR6 NM_​022496.4 12q23.1
RSF1 NM_​016578.3 11q14.1
ARID1A NM_​006015.4 1p36.11
RUVBL1 NM_​003707.2 3q21.3
ARID2 NM_​152641.2 12q12
RUVBL2 NM_​006666.1 19q13.3
ATAD2 NM_​014109.3 8q24.13
SETD6 NM_​001160305.1 16q21
ATRX NM_​000489.3 Xq21.1
SMARCA1 NM_​001282874.1 Xq25
BAHD1 NM_​014952.3 15q15.1
SMARCA5 NM_​003601.3 4q31.21
BAZ2A NM_​013449.3 12q13.3
SMARCAD1 NM_​001128429.2 4q22.3
BRD4 NM_​014299 19p13.12
SMARCAL1 NM_​001127207.1 2q35
BPTF NM_​004459.6 17q24.3
SMARCC1 NM_​003074.3 3p21.31
BRMS1 NM_​001024957.1 11q13.2
SMARCC2 NM_​001130420.1 12q13.2
CHAF1A NM_​005483.2 19p13.3
SMARCD1 NM_​003076.4 12q13.12
CHAF1B NM_​005441.2 21q22.13
SMARCD2 NM_​001098426.1 17q23.3
CHD1 NM_​001270.2 5q15-​q21
SMARCD3 NM_​001003801.1 7q36.1
CHD1L NM_​001256337.1 1q21.1
SMARCE1 NM_​003079.4 17q21.2
CHD2 NM_​001042572.2 15q26.1
SRCAP NM_​006662.2 16p11.2
CHD3 NM_​001005273.2 17p13.1
TFPT NM_​013342.3 19q13
CHD4 NM_​001273.2 12p13.31
TOP2A NM_​001067.3 17q21-​q22
CHD5 NM_​015557.2 1p36.31
TOP2B NM_​001068.3 3p24.2
CHD6 NM_​032221.4 20q12
TP73 NM_​017818.3 1p36.32
CHD7 NM_​017780.3 8q12.1-​q12.2
VPS72 NM_​001271087.1 1q21.3
CHD8 NM_​020920.3 14q11.2
YY1 NM_​003403.4 14q32.2
CHD9 NM_​025134.4 16q12.2
ZHX1 NM_​001017926.2 8q24.13
CTBP1 NM_​001328.2 4p16.3
ZNHIT1 NM_​006349.2 7q22.1
CTCF NM_​001191022.1 16q22.1
ZRANB3 NM_​001286568.1 2q21.3
CTCFL NM_​001269041.1 20q13.31
 13

Biology of Neoplasia 13
of genetic and epigenetic alterations in interplay with the immune (e) (f) (g)
system.

HALLMARKS OF CANCER CELLS

From the careful dissection of the properties of many tumor types,


Weinberg and Hanahan, in two review articles over a decade apart,
described the most common features, or hallmarks, of the tumor cell
(Hanahan and Weinberg, 2000, 2011) (Figures 2–3a and 2–3b). These
are common characteristics of most tumor types. At the individual
Altered energetics/
cell level, the most important hallmarks are self-​renewal, uncontrolled Metabolism
Loss of
growth, absence of growth regulation, loss of DNA repair, and loss of
Immune Control
programmed cell death in the face of these lesions. These hallmarks Angiogenesis
are especially important at the initial stages of carcinogenesis in the
formation of the pre-​malignant and later malignant lesion.
At later stages, a tumor acts as a multicellular entity that interacts
(h) (l)
with multiple normal cells of the immune system, stimulates the pro-
duction of new blood vessels (angiogenesis), and develops the ability
to invade other tissues and metastasize.

Cancer Stem Cell Model


The most important hallmark of cancer cells is the continual growth
of the tumor, or self-​renewal. In the body’s normal stem cells, self-​ Epigenetic
renewal is a mechanism to generate cells identical to the stem cell and Reprogramming
maintain a population of pluripotent, multipotent, or oligopotent cells.
One model of cancer places a major emphasis on the ability of cancer Invasion and Metastasis
cells to undergo self-​renewing replication like that which occurs in
stem cells (Al-​Hajj et al., 2004; Dean et al., 2005). In certain tumor Figure 2–3b.  Hallmarks of cancer cells: late stages. (e) The immune system
types and tumor models, cancer stem cells or tumor initiating cells can recognize abnormal or infected cells and destroy them. Cancer cells can
have been isolated (Al-​Hajj et al., 2003). evade the immune system. One mechanism is the secretion of inhibitory
One pathway to tumor development starts with a chronically rep- factors such as checkpoints and cytokines. (f) A growing tumor requires a
licating normal stem cell. These cells naturally possess several of the blood supply, and tumors can stimulate the production of new blood ves-
“hallmarks of cancer.” Sustained or chronic tissue damage and/​or sels. (g) Many tumors switch to glycolosis as an energy source (Warburg
inflammation can lead to the activation of stem cells and their con- effect). (h) Invasion into surrounding tissues and other parts of the body
tinuous replication. Such a population of replicating stem cells can (metastasis) are common tumor properties. (i) Tumor cells mutate one or
become a target for somatic mutations that cause sustained prolifera- more genes that modify the histone proteins that form chromatin (chromatin
tive signaling and/​or evasion of growth suppression. remodeling genes) and permit epigenetic reprogramming of the genome.
This leads to a pre-​malignant cancer stem cell, a target for further
mutation and uncontrolled proliferation and loss of growth control.
Early mutations can also promote defective DNA repair, and the loss defective DNA repair promote further genome instability and mutation
of checkpoints in the cell cycle preventing cells with damaged DNA and tumor evolution.
or aneuploid chromosomes from replicating. Genome instability and However, most morbidity and mortality from cancer is the result of
tumor invasion and/​or metastasis, and tumor progression involves the
activation of invasion and metastasis, the induction of angiogenesis
(a) (b) (c) (d) to provide a blood supply to the growing tumor, the evasion of the
immune system, and altered cell energetics.

Self-
Renewal
Epigenetic Reprogramming
Uncontrolled In virtually all tumor types, a newly described class of frequently
Growth mutated genes consists of enzymes involved in histone modification
Replication of Defective DNA Repair/ and components of chromatin remodeling complexes. One model of
Damaged Cell Chromosome Integrity cancer involves a stage of cellular reprogramming of expression state
(epigenetic state), altering entire programs of gene expression. The
Oncogene Tumor DNA Repair ability to turn on or off whole programs of expression allows the tumor
Activation Suppressor Gene Mutation cell to adapt to virtually all encountered barriers
Loss
• Invasion:  The cancer cell can turn on (or the cancer stem cells
Figure 2–3a.  Hallmarks of cancer cells: early stages. Tumors often show already possess) the ability to respond to homing signals, migrate
properties common across cancer types. (a) Self-renewal is a type of cell through tissue into the bloodstream, and travel to other parts of the
division that generates daughter cells identical to the original cell. Stem body. Portions of the tumor may establish a niche whereby other
cells also undergo this type of division. These cells have been termed can- cells in the tumor can proliferate and grow.
cer stem cells, or cancer-initiating cells. (b) Whereas most normal cells • Immune evasion: Tumor cells can express immune evasion signals
will stop dividing as they come in contact with other cells (contact inhibi- (such as PD-​L1) and inhibit immune cells that recognize the tumor
tion), many tumor cells continue to grow. (c) Reduced cell death. Cell death as foreign.
is a normal process for many cells in the body, but many tumor cells are • Cell metabolism: Many tumor cells alter their energetics and metab-
defective in this process and continue to survive in the presence of damage. olism. For example, initiating programs of gene expression that
(d) Tumor cells can be defective in DNA repair their telomeres. allow the use of glucose, or anaerobic glycolysis.
14

14 Part I:  Basic Concepts

Tumor Differentiation and Evolution cysts, facial and bone abnormalities, and occasionally medulloblastomas
(Anderson et al., 1967; Gorlin and Goltz, 1960). Genetic linkage studies
At first glance, the tumor seems like one of the mythical beasts that localized the gene for NBCCS to chromosome 9, and the gene responsi-
Ulysses encountered on his journey, a Hydra or a Cyclops. But in fact ble was identified as Patched (PTCH1) (Gailani et al., 1992; Hahn et al.,
the cancer cell is a human cell and therefore utilizes tools that our 1996; Johnson et al., 1996). This gene was first discovered in drosophila
normal cells employ and is subject to most of the same constraints as part of a group of genes required for normal embryonic development
and restrictions. A human being goes from a single fertilized egg to an of the fly, and mutants cause the patchy formation of bristles. Patched is
adult with thousands of cell types and tissues with diverse function, a cell surface protein and the receptor for a class of secreted, modified
without changing the DNA of the cells (Figure 2–4). This development peptides, Hedgehogs (SHH) (Dean, 1996). Patched repressed the activ-
and differentiation are all accomplished by the coordinated activation ity of a G-​protein-​coupled receptor Smoothened (SMO), and the binding
and repression of distinct programs of gene expression, the secretion of SHH to PTCH1 releases SMO from repression and leads eventually
of proteins and other factors that influence neighboring cells, and the to changes in gene regulation in the nucleus through the GLI family of
contact and interaction of cells with each other. The tumor likewise transcription factors (van den Heuvel and Ingham, 1996).
evolves from a single cell, forms a network of self-​renewing and com- All of the major components of this signaling pathway are conserved
mitted cells, secretes factors that stimulate other cells in the tumor, in insects, vertebrates, and other organisms. The correct functioning of
and interacts with other tumor cells as well as normal cells. these proteins is required for the development of the growing embryo
The transition from a normal fetus to an adult human involves the and the polarity and symmetry of cells and structures during develop-
continued growth and differentiation of cells to form multiple organs ment. We now recognize all the components of this pathway as either
and specialized cell types. Cells migrate in the growing human, estab- tumor suppressor genes (PTCH1) or oncogenes (SHH, SMO, GLI). In
lish niches for stem cells, develop blood supplies, and in certain cases nearly all cases of NBCCS, we find germline mutations in the PTCH1
alter metabolism and energy utilization. In a parallel fashion, the cells gene, and in nearly all sporadic basal cell carcinomas and a portion of
of a growing and developing tumor alter gene expression profiles in medulloblastomas PTCH1 is mutated. This work provided a clear con-
subpopulations of cells, migrate to different locations, adapt to new nection to the functions of development and differentiation of embry-
environments, survive over years or decades, and adapt to therapeutic onic stem cells and the formation of tumors (Bale and Yu, 2001).
strategies to eliminate the tumor cell.
A key finding in the connection between the developing embryo and
the tumor was the discovery of oncogenes and tumor suppressors that
Evolution and the Tumor
are also key embryonic differentiation factors. Gorlin syndrome, or
nevoid basal cell carcinoma syndrome (NBCCS), is an inherited disor- Our species and all other vertebrates are the product of a billion years
der characterized by hundreds to thousands of basal cell carcinoma, jaw of evolution. Through this process we evolved unique characteristics,

Blood Cells

(a)

Neurons

Muscle Cells
Normal
Stem Cell
Embryo Stem
Fetus Kidney Cells
Cell

(b)
Angiogenic Cells

Tumor Metastatic Cells


Cell

Immune Resistant Cells


Pre-Malignant
Lesion Locally
Invasive
Lesion Tumor Therapy Resistant Cells
Stem
Cell

Figure 2–4.  Cancer development and differentiation. The progression of the tumor cell, from single cell to fully developed malignancy, displays paral-
lels to the development of the organism and differentiation. The embryo is a mix of self-renewing stem cells and cells committed to discrete pathways
of differentiation. A pre-malignant lesion also contains these two classes of cells. (a) The fetus displays an increasing array of differentiating cells types;
formation of blood vessels, altered metabolism, and energy utilization. The local malignant tumor accumulates necrotic cells, and can be angiogenic. The
development from fetus to adult organism requires a large-scale reprogramming of gene expression networks to carry out the generation of cells of multiple
organ and tissue types by epigenetic reprogramming. Epigenetic plasticity in the developing tumor can also lead to the development and differentiation
of a large number of cell types. This can allow spread of the tumor, evasion of cellular processes such as immune attack, and/or survival from therapeutic
approaches. (b) The tumor also undergoes evolutionary selection. The tumor cell progresses from a single-cell organism to a small multi-cellular organ-
ism. Just as evolution allowed diverse classes and species of organisms to evolve from largely the same sets of genes, the tumor can undergo selection for
altered, duplicated, or deleted genes. To adapt to new conditions and survive therapy, the tumor adapts resistance mechanisms. Many of the same principles
of natural selection apply to tumors as apply to populations of species.
 15

Biology of Neoplasia 15
adapted to changing environments, individuals were selectively and more broadly the genetic architecture of genetic risk in popula-
removed, and surviving individuals went on to reproduce and survive. tions in relation to cancer (Chung and Chanock, 2011). Currently there
A tumor also undergoes this same selective process, only in the are almost 500 loci identified in the genome that modify risk of one
context of our body and over the time scale of years, decades, or our or more cancers. However, in only a few cases do we understand the
lifespan. Many cells in the tumor die, and necrotic areas of larger molecular basis of these effects.
tumors are often seen. Radiation, chemotherapy, and other treatments
can kill off large portions of the cancer, and in some cases all cells,
resulting in cures. However, those cells that survive treatment and Occupational/​Environmental Epidemiology
retain the property of self-​renewal can continue to grow, resulting in
remission. Many of the principles and concepts that were developed The role of environment (broadly defined, including lifestyle factors)
and used to understand and explain evolution can also be applied to and occupation in the development of cancer has been critical. Acute
the evolution of cancer. DNA sequencing of tumor cells at different or long-​term environmental or occupational exposures can contribute
sites of the body and/​or stages of the disease is resulting in deeper to chronic inflammation or tissue damage (smoking, asbestos expo-
understanding of these processes, sometimes at the resolution of sin- sure, benzene, etc.). Agents may cause tissue damage/​inflammation
gle cells (Xu et al., 2012). (UV exposure, radiation, asbestos, infectious agents), or may induce
mutations (radiation, mutagenic chemicals, mutagenic viruses), or
Key Points commonly both. In some cases (tobacco, UV exposure, viral infec-
• Cancer cells within a tumor display tremendous diversity. tions), we can now detect specific mutational signatures in cancer
• New mutations, epigenetic changes, and gene rearrangements occur genomes.
continually during the cancer process.
• Cancer cells face barriers to survival that include immune evasion, Radiation Epidemiology
therapy resistance, and development of invasion/​metastasis. Radiation exposure can occur through the environment, occupation,
accidents, or medical treatment, and can often be accurately measured
and modeled in many organisms. While in acute or concentrated doses
Tumor Microenvironment radiation can cause tissue damage, the majority of the carcinogenesis
of radiation is due to DNA damage and mutation.
The cancer cell is a human cell, with many properties in common with
non-​malignant human cells. In the body of the individual, the cancer
cell interacts in a complex manner with the host cells. Therefore, there Hormonal Epidemiology
exists a microenvironment surrounding the tumor that is composed of
host cells interacting and responding to the tumor and is subject to Many major cancers involve reproductive organs or organs exclu-
modification by the tumor cells. The major classes of cells in the tumor sive or largely to one gender (cervix, prostate, breast, ovary). These
microenvironment (TME) include the following: tumor types involve major hormonal effects. For example, many breast
tumors are dependent on estrogens and/​ or progesterone, and this
Vasculature:  For tumor cells to advance beyond a size typically knowledge has aided both prevention and therapy.
greater than 2 millimeters requires the development of new blood However, there is also a large gender discordance for many other
vessels (angiogenesis). However, the interior of many tumors is types:  male bias in bladder cancer, any pediatric cancers, and liver
hypoxic and may contribute to tumor cell properties including ther- cancer; female bias in chronic lymphocytic leukemia and thyroid can-
apy resistance. cer. In most cases we do not understand these effects. As hormone
Stromal cells: Fibroblasts can be recruited into the TME and their func- production and exposure change dramatically over time for males and
tions altered to support tumor growth (Hanahan and Coussens, 2012; females, this is a critical area of cancer epidemiology.
Pickup et al., 2013). Myeloid-​derived suppressor cells (MDSCs) are
cells derived from the myeloid lineage’s T cell repressive properties
(Talmadge and Gabrilovich, 2013). Tumor-​infiltrating lymphocytes Infectious Disease Epidemiology
are T cells with reactivity against the tumor cells. In certain cases,
these cells have been expanded and used therapeutically (Hinrichs and Many of the world’s most common cancers and the cancers that dis-
Rosenberg, 2014). play the greatest disparities due to income involve infectious agents
(cervical and HPV, liver and HBV and HCV, and gastric and H. pylori)
Cancer cells need to evade the immune system’s constant probing (Klein, 2002; Schiffman and Castle, 2005; Torre et al., 2015; Uemura
for virally infected or otherwise foreign cells. This can be accom- et al., 2001; zur Hausen and de Villiers, 1994). The list of direct causes
plished by not expressing foreign antigens or suppressing the immune of cancer by infectious agents and pathogens is impressive. And the
response. Many of the new checkpoint inhibitor cancer strategists role of other chronic infections remains to be fully explored.
using PD-​L1 or PD-​1 inhibitors seek to reverse the immune suppres-
sion of tumors (Sunshine and Taube, 2015).
Nutritional Epidemiology
The nutritional state of the individual, diet, body mass index (BMI),
FIELDS OF EPIDEMIOLOGY AND and related conditions such as diabetes and obesity are increasingly
THE CANCER PROCESS recognized as major risk factors for cancers. Studies show that virtu-
ally all cancer types increase with increasing BMI. Furthermore, the
Our understanding of the process of cancer development not only has increase in BMI and obesity in world populations has led to projec-
been shaped by, but also influences all areas of epidemiology, and tions that this will be a more significant risk factor than tobacco in the
increasingly genetic and genomic studies of cancer are applied to sub- future (Calle et al., 2003).
divisions of the field. The mechanisms by which increased body mass contributes to can-
cer are poorly studied. However, some of the hormones regulated by
energy consumption, such as insulin and insulin-​like growth factors,
Genetic Epidemiology are tied to cellular pathways for growth and division. Many tumor
Classical genetic epidemiology involved the identification of rare can- suppressor and oncogene networks connect to metabolic pathways,
cer families, the study of genetic conditions that contain cancer as a and some enzymes of the tricarboxylic acid (TCA) cycle are tumor
phenotype, and the determination of heritability. More recent genetic suppressor genes. In addition, fat deposition and obesity can cause an
epidemiology has involved the study of genome-​ wide association inflammatory state and may therefore be analogous to other inflamma-
studies (GWAS), identification of common but low relative risk alleles, tory states such as viral and bacterial infections.
16

16 Part I:  Basic Concepts

AGING AND CANCER the telomeres are shortened because the conventional DNA polymer-
ases are unable to replicate the ends of chromosomes. This is known as
Just as cancer is a genetic disease, it is also a disease of aging. Age is the “end replication problem” (Donate and Blasco, 2011). This is why
the most important risk factor for most adult cancers, and many of the a cellular enzyme called telomerase may be recruited in certain situa-
processes that we understand are operative in cancer development can tions to add TTAGGG repeats to the chromosome ends. The enzyme
be thought of in light of aging. telomerase consists of a subunit Tert that possesses reverse transcrip-
tase activity, an RNA element called Terc that is the template on which
DNA is synthesized, and a protein called dyskerin (DKc1) that has the
DNA Damage ability to bind and stabilize Terc. Upregulated telomerase expression
is a feature of pluripotent stem cells and also of early stage embryonic
With time, the cells of our body accumulate DNA damage. Much
development; however, telomerase activity can also be present in adult
of this occurs through natural processes, such as the deamination of
stem cell compartments (Blasco, 2005).
5-​methyl-​cytosine found in many DNA regions; 5-​methyl-​cytosine
Telomere length and Tert are critical factors determining the mobili-
spontaneously deaminates to uracil, and the body has an enzyme/​DNA
zation of stem cells. However, it was also found that shelterin, a major
repair system to recognize this damage and repair it. However, this
protein complex bound to mammalian telomeres, may play a role in the
process is error prone, and DNA lesions accumulate over time, leading
initiation of neoplasia (Donate and Blasco, 2011). Mice conditionally
in cases to the accumulation of sufficient mutations in a single cell to
deleted for the shelterin proteins TRF1, TPP1, and Rap1 were gener-
start the cancer process.
ated, and this demonstrated that telomere dysfunction is sufficient to
One of the new areas of cancer genome study is the ability to exam-
produce premature tissue degeneration, acquisition of chromosomal
ine the whole genomes of cancer cells and discern the signatures of
aberrations, and the initiation of neoplastic lesions (Blasco, 2005).
mutational processes (Alexandrov et al., 2013). By tabulating all muta-
Based on further experiments, a stem-​cell-​based model for the role
tions in the tumor genome, and noting the nature of the base changes
of telomeres related to cancer and aging was proposed (Donate and
and the local context of the mutation, signatures can be identified.
Blasco, 2011). It was proposed that in young or adult organisms, stem
The most common cancer signature is indeed one in which a CpG is
cells have the ability to repopulate certain tissues as needed. During
mutated (Shiraishi et al., 2015).
this process, the stem cells undergo telomere shortening, which is
only partially counterbalanced by the action of telomerase. However,
in older organisms, the stem cell telomeres are actually too short.
Chronic Infection Critically short telomeres are recognized by the body as DNA damage.
Especially for cervical, gastric, and liver cancer, chronic infections This recognition of potential DNA damage activates a p53-​mediated
by HPV, H. pylori, and hepatitis viruses (HBV, HCV) are the prin- DNA damage signaling response that can impair stem cell mobiliza-
cipal cause. None of these infections causes cancer shortly after tion, leading to suboptimal tissue regeneration, which may ultimately
infection; all require a long latency period in which the infectious lead to organ failure. This phenomenon of decreased stem cell mobi-
agent stimulated tissue damage, inflammation, local repair, and lization reduces the probability that abnormal cells will accumulate in
activation of tissue stem cells before pre-​malignant and malignant tissues and thus provides a mechanism for cancer protection. On the
lesions arise. other hand, if stem cells express aberrantly high levels of the enzyme
It is recently appreciated that activation of innate anti-​viral infection telomerase (by the acquisition of tumorigenic, telomerase-​reactivating
mechanisms, such as the ABOBEC family of anti-​viral proteins, over mutations), stem cell mobilization is more efficient and tissue fitness
long periods of time can lead to somatic mutations in tumor cells. The is maintained for a longer time. This would increase life span, but
second most common signature is one consistent with APOBEC activa- would also increase the probability of initiating a tumor (Donate and
tion (Lawrence et al., 2013; Shiraishi et al., 2015). It is indeed commonly Blasco, 2011).
found in cervical cancer and head and neck cancers associated with HPV
infection, but also with acute lymphocytic leukemia (ALL), bladder,
breast, kidney, pancreas, and some forms of lung cancer. Whether there
The Aging Immune System and Cancer
are unappreciated viral infections influencing these cancers, or alternate
pathways to activate APOBECs, remains to be determined. It has been found that the incidence of most common cancers increases
with age and may possibly be caused by a decline in immune func-
tion. This phenomenon is called “immune senescence” (Foster et al.,
Chronic Inflammation 2011). The causal determinant for this phenomenon, although highly
controversial, may be deleterious changes to T-​cell subsets that over
A large number of cancers are caused by chronic inflammation caused time may impair immunity. In one murine study, tumors were able to
by radiation, irritants, toxins, or immune inflammatory processes. For limit the function of CD8 + T cells, including specific responses to
example, a person with ulcerative colitis or Crohn’s disease has an tumor antigens specific for prostate tumors (Anderson et  al., 2007).
elevated risk to develop colorectal cancer (Jess et al., 2012). The role Another study showed that microvesicles isolated from human tumor
of asbestos in lung cancer, UV radiation and sunburn in skin cancer, cells were able to induce the generation and expansion of Treg (T regu-
and irritants in tobacco products in lung and oral cancers are well doc- latory cells) that are capable of suppressing T-​cell responses (Szajnik
umented. As with viral infections, these cancers develop over many et al., 2010). In relation to this, the immunoregulatory effects of the
decades, and the aspects of aging on the development of cancer due to tumor (and its environment) on T cells, along with the decreasing abil-
inflammation are poorly understood. ity of the body to replace naïve T-​cell populations, may allow cancers
to progress because T cells seem to have a fixed immune repertoire
associated with age-​related immunosenescence. The intricate mecha-
Cell Senescence and Telomere Regulation nisms that lead to declining cellular immunity are very unclear, but
Telomeres are the protective elements at the chromosome ends that the progressive decline in thymic output of T cells may be the primary
aid in preventing unscheduled DNA repair and degradation. They are event leading to immune senescence in old age. With age, the produc-
composed of repetitive DNA sequences (TTAGGG repeats) bound to tion of new naïve T cells reduces, resulting in the peripheral T cells
an array of proteins with specialized functions (Donate and Blasco, having to undergo repeated rounds of cell division in order to main-
2011). The protection afforded to the chromosome depends on the tain the status quo so that the overall size of the T-​cell compartment
length of telomeric repeats and the integrity of the telomere-​binding remains relatively stable. This prolonged survival of the peripheral
proteins. The length and integrity of telomeres are regulated by specific T cells results in defects in all types of activation states of T cells, from
epigenetic modifications, indicating that a higher order control of telo- the naïve to the terminally differentiated. This reduction in functional
mere function exists. After each round of cell division is completed, immunity can be permissive to tumor formation and might promote
 17

Biology of Neoplasia 17
tumor formation by contributing to low-​level inflammation. In order suggest that cancer cells can acquire a plastic state that would allow
to circumvent this pro-​inflammatory environment, specific cytokine the generation of cells in the tumor capable of diverse functions and
blockers may be beneficial to both neoplasia and other inflammatory evolution. A therapeutic attraction to this model is that this may be a
disease states, but have the drawback of compromising the immune reversible process that could be manipulated.
system (Zidi et al., 2010). Thus, more research needs to be carried out Single cell genomic analyses of tumors are revealing an enormous
to determine the specific mechanisms of action of cytokine blockers to level of complexity and heterogeneity. These studies can be used to
allow for the development of targeted therapies. follow the evolution of cancer cells over both time and location in the
Another strategy would be to delay replicative senescence in T cells, cancer patient.
utilizing strategies that sustain telomerase activity (Effros, 2011).
While we have learned a lot about the role of age-​related declining References
immunity as it relates to cancer incidence, the precise cellular mecha- Al-​Hajj M, Becker MW, Wicha M, Weissman I, and Clarke MF. 2004.
nisms through which this occurs still remain elusive. As research pro- Therapeutic implications of cancer stem cells. Curr Opin Genet Dev,
gresses, from animal models to human clinical trials, new therapeutic 14(1), 43–​47. PMID: 15108804.
strategies to enhance immune function may become powerful tools to Al-​Hajj M, Wicha MS, Benito-​Hernandez A, Morrison SJ, and Clarke MF.
reduce cancer incidence (Foster et al., 2011). 2003. Prospective identification of tumorigenic breast cancer cells. Proc
Natl Acad Sci U S A, 100(7), 3983–​3988. PMCID: PMC153034.
Alexandrov LB, Nik-​Zainal S, Wedge DC, et  al. 2013. Signatures of muta-
CONCLUSIONS tional processes in human cancer. Nature, 500(7463), 415–​ 421.
PMCID: PMC3776390.
Many of the genetic and biological processes that occur in cancer cells are Anderson DE, Taylor WB, Falls HF, and Davidson RT. 1967. The nevoid
now well documented. Through the study of animal models, cell culture basal cell carcinoma syndrome. Am J Hum Genet, 19(1), 12–​ 22.
systems and human tumors we have detailed catalogs of the following: PMCID: PMC1706134.
Anderson MJ, Shafer-​Weaver K, Greenberg NM, and Hurwitz AA. 2007.
• The genes in which germline mutations cause highly penetrant can- Tolerization of tumor-​specific T cells despite efficient initial priming in a
cer syndromes; primary murine model of prostate cancer. J Immunol, 178(3), 1268–​1276.
• Genetic loci from GWAS studies that influence cancer risk; PMID: 17237372.
Bale AE, and Yu KP. 2001. The hedgehog pathway and basal cell carcinomas.
• Genes frequently altered somatically in cancer.
Hum Mol Genet, 10(7), 757–​762. PMID: 11257109.
There is considerable work still in progress to identify rare alleles Blasco MA. 2005. Telomeres and human disease: ageing, cancer and beyond.
involved in cancer risk, to understand the function of cancer GWAS Nat Rev Genet, 6(8), 611–​622. PMID: 16136653.
Calle EE, Rodriguez C, Walker-​Thurmond K, and Thun MJ. 2003. Overweight,
loci, and to understand complex copy number changes and gene rear-
obesity, and mortality from cancer in a prospectively studied cohort of
rangements in cancer cells. U.S. adults. N Engl J Med, 348(17), 1625–​1638. PMID: 12711737.
Most of the major environmental and occupational risk factors associ- Cancer Genome Atlas Research Network. 2011. Integrated genomic analyses of
ated with cancer have been identified and can now be at least partially ovarian carcinoma. Nature, 474(7353), 609–​615. PMCID: PMC3163504.
understood at the molecular level from current data. The exciting new Cancer Genome Atlas Network. 2012. Comprehensive molecular portraits of
area of mutation signatures promises to further reveal the effect of spe- human breast tumours. Nature, 490(7418), 61–​70. PMCID: PMC3465532.
cific exposures on somatic alterations in tumors. However, even the well-​ Chung CC, and Chanock SJ. 2011. Current status of genome-​wide association
known cancer-​causing infections, such as human papillomavirus (HPV), studies in cancer. Hum Genet, 130(1), 59–​78. PMID: 21678065.
hepatitis B virus (HBV), and hepatitis C virus (HCV), are only partially Clark SS, McLaughlin J, Timmons M, et al. 1988. Expression of a distinctive
BCR-​ABL oncogene in Ph1-​positive acute lymphocytic leukemia (ALL).
understood, and there is much left to be learned regarding agents such
Science, 239(4841 Pt 1), 775–​777. PMID: 3422516.
as asbestos, tobacco smoke, and chemical carcinogens. The long latency Couch FJ, Nathanson KL, and Offit K. 2014. Two decades after BRCA: set-
of cancer and the complexity of the human immune system and tumor ting paradigms in personalized cancer care and prevention. Science,
microenvironment leave enormous areas for future investigation. 343(6178), 1466–​1470. PMCID: PMC4074902.
Epigenetic modification has emerged as a major factor in the devel- Croce CM. 2008. Oncogenes and cancer. N Engl J Med, 358(5), 502–​511.
opment of cancer, largely in the later stages of the disease. Nearly PMID: 18234754.
all cancers have mutations in one or more histone-​modifying enzyme de Klein A, van Kessel AG, Grosveld G, et  al. 1982. A cellular oncogene is
affecting chromatin structure, and/​or enzyme-​regulating DNA methyl- translocated to the Philadelphia chromosome in chronic myelocytic leu-
ation (Figure 2–5). An intriguing outcome of these findings is that the kaemia. Nature, 300(5894), 765–​767. PMID: 6960256.
Dean M. 1996. Polarity, proliferation and the hedgehog pathway. Nat Genet,
deregulation of the epigenome, and/​or “return to an embryonic state,”
14(3), 245–​247. PMID: 8896548.
may be a newly appreciated hallmark of cancer. This model would Dean M. 1997. Towards a unified model of tumor suppression: lessons learned
from the human patched gene. Biochim Biophys Acta, 1332(2), M43–​
M52. PMID: 9141460.
(a) Naked DNA Dean M, Fojo T, and Bates S. 2005. Tumor stem cells and drug resistance. Nat
Rev Cancer, 5(4), 275–​284. PMID: 15803154.
Dean M, Park M, Le Beau MM, et  al. 1985. The human met oncogene is
(b) DNA covered in histones related to the tyrosine kinase oncogenes. Nature, 318(6044), 385–​388.
and other chromatin proteins PMID: 4069211.
Der CJ, Krontiris TG, and Cooper GM. 1982. Transforming genes of human
bladder and lung carcinoma cell lines are homologous to the ras genes of
Harvey and Kirsten sarcoma viruses. Proc Natl Acad Sci U S A, 79(11),
(c) Histones modified by adding or
3637–​3640. PMCID: PMC346478.
removing methyl or acyl groups
Donate LE, and Blasco MA. 2011. Telomeres in cancer and ageing. Philos
Trans R Soc Lond B Biol Sci, 366(1561), 76–​84. PMCID: PMC3001316.
(d) Remodeling of histones to Druker BJ, Sawyers CL, Kantarjian H, et al. 2001a. Activity of a specific inhibi-
create “open” chromatin or tor of the BCR-​ABL tyrosine kinase in the blast crisis of chronic myeloid
closed, inaccessible regions leukemia and acute lymphoblastic leukemia with the Philadelphia chro-
mosome. N Engl J Med, 344(14), 1038–​1042. PMID: 11287973.
Figure  2–5.  Chromatin alteration and remodeling (a) DNA is a charged Druker BJ, Talpaz M, Resta DJ et al. 2001b. Efficacy and safety of a specific
molecule. (b) DNA is covered by histones and other proteins to form inhibitor of the BCR-​ABL tyrosine kinase in chronic myeloid leukemia.
nucleosomes and chromatin. (c) Histones undergo extensive modification. N Engl J Med, 344(14), 1031–​1037. PMID: 11287972.
(d) Other protein complexes (chromatin remodelers) can alter the location Effros RB. 2011. Telomere/​telomerase dynamics within the human immune
of nucleosomes, make DNA open to transcription factors turning genes on, system:  effect of chronic infection and stress. Exp Gerontol, 46(2–​3),
or, covering regulatory regions, turning genes off. 135–​140. PMCID: PMC3246363.
18

18 Part I:  Basic Concepts


Foster AD, Sivarapatna A, and Gress RE. 2011. The aging immune sys- Muraoka M, Konishi M, Kikuchi-​Yanoshita R, et al. 1996. p300 gene altera-
tem and its relationship with cancer. Aging Health, 7(5), 707–​ 718. tions in colorectal and gastric carcinomas. Oncogene, 12(7), 1565–​1569.
PMCID: PMC3222953. PMID: 8622873.
Gailani MR, Bale SJ, Leffell DJ, et al. 1992. Developmental defects in Gorlin Murphree AL, and Benedict WF. 1984. Retinoblastoma: clues to human onco-
syndrome related to a putative tumor suppressor gene on chromosome 9. genesis. Science, 223(4640), 1028–​1033. PMID: 6320372.
Cell, 69(1), 111–​117. PMID: 1348213. Pedersen JK, Engholm G, Skytthe A, Christensen K. 2016. Cancer and aging:
Gorlin RJ, and Goltz RW. 1960. Multiple nevoid basal-​ cell epithelioma, epidemiology and methodological challenges. Acta Oncol, 55(Suppl 1),
jaw cysts and bifid rib. A  syndrome. N Engl J Med, 262, 908–​912. 7–12. PMID: 26825001.
PMID: 13851319. Pickup M, Novitskiy S, and Moses HL. 2013. The roles of TGFbeta in
Gui Y, Guo G, Huang Y, et al. 2011. Frequent mutations of chromatin remodel- the tumour microenvironment. Nat Rev Cancer, 13(11), 788–​ 799.
ing genes in transitional cell carcinoma of the bladder. Nat Genet, 43(9), PMCID: PMC4025940.
875–​878. PMID: 21822268. Pulciani S, Santos E, Lauver AV, et  al. 1982. Oncogenes in human tumor cell
Guo G, Sun X, Chen C et al. 2013. Whole-​genome and whole-​exome sequenc- lines:  molecular cloning of a transforming gene from human blad-
ing of bladder cancer identifies frequent alterations in genes involved der carcinoma cells. Proc Natl Acad Sci U S A, 79(9), 2845–​ 2849.
in sister chromatid cohesion and segregation. Nat Genet, 45(12), 1459–​ PMCID: PMC346303.
1463. PMID: 24121792. Rowley JD. 1973. Letter:  A  new consistent chromosomal abnormality in
Hahn H, Wicking c, Zaphiropoulous PG, et al. 1996. Mutations of the human chronic myelogenous leukaemia identified by quinacrine fluorescence
homolog of Drosophila patched in the nevoid basal cell carcinoma syn- and Giemsa staining. Nature, 243(5405), 290–​293. PMID: 4126434.
drome. Cell, 85(6), 841–​851. PMID: 8681379. Santos-​Rosa H, and Caldas C. 2005. Chromatin modifier enzymes, the histone
Hanahan D, and Coussens LM. 2012. Accessories to the crime:  functions of code and cancer. Eur J Cancer, 41(16), 2381–​2402. PMID: 16226460.
cells recruited to the tumor microenvironment. Cancer Cell, 21(3), 309–​ Schiffman M, and Castle PE. 2005. The promise of global cervical-​cancer pre-
322. PMID: 22439926. vention. N Engl J Med, 353(20), 2101–​2104. PMID: 16291978.
Hanahan D, and Weinberg RA. 2000. The hallmarks of cancer. Cell, 100(1), Shiraishi Y, Tremmel G, Miyano S, and Stephens M. 2015. A simple model-​
57–​70. PMID: 10647931. based approach to inferring and visualizing cancer mutation signatures.
Hanahan D, and Weinberg RA. 2011. Hallmarks of cancer: the next generation. PLoS Genet, 11(12), e1005657. PMCID: PMC4667891.
Cell, 144(5), 646–​674. PMID: 21376230. Stehelin D, Varmus HE, Bishop JM, and Vogt PK. 1976. DNA related to the
Harding C, Pompei F, Lee EE, and Wilson R. 2008. Cancer suppression at old transforming gene(s) of avian sarcoma viruses is present in normal avian
age. Cancer Res, 68(11), 4465–​4478. PMID: 18519710. DNA. Nature, 260(5547), 170–​173. PMID: 176594.
Hinrichs CS, and Rosenberg SA. 2014. Exploiting the curative potential Struewing JP, Hartge P, Wacholder S, et  al. 1997. The risk of cancer
of adoptive T-​ cell therapy for cancer. Immunol Rev, 257(1), 56–​ 71. associated with specific mutations of BRCA1 and BRCA2 among
PMCID: PMC3920180. Ashkenazi Jews [see comments]. N Engl J Med, 336(20), 1401–​1408.
Jess T, Rungoe C, and Peyrin-​Biroulet L. 2012. Risk of colorectal cancer in PMID: 9145676.
patients with ulcerative colitis: a meta-​analysis of population-​based cohort Sunshine J, and Taube JM. 2015. PD-​1/​PD-​L1 inhibitors. Curr Opin Pharmacol,
studies. Clin Gastroenterol Hepatol, 10(6), 639–​645. PMID: 22289873. 23, 32–​38. PMCID: PMC4516625.
Johnson RL, Rothman AL, Xie J, et al. 1996. Human homolog of patched, a Szajnik M, Czystowska M, Szczepanski MJ, Mandapathil M, and Whiteside
candidate gene for the basal cell nevus syndrome. Science, 272(5268), TL. 2010. Tumor-​derived microvesicles induce, expand and up-​regulate
1668–​1671. PMID: 8658145. biological activities of human regulatory T cells (Treg). PLoS One, 5(7),
Kandoth C, McLellan MD, Vandin F, et  al. 2013. Mutational landscape and e11469. PMCID: PMC2908536.
significance across 12 major cancer types. Nature, 502(7471), 333–​339. Talmadge JE, and Gabrilovich DI. 2013. History of myeloid-​derived suppressor
PMCID: PMC3927368. cells. Nat Rev Cancer, 13(10), 739–​752. PMCID: PMC4358792.
Kinzler KW, Nilbert MC, Su LK, et al. 1991. Identification of FAP locus genes Torre LA, Bray F, Siegel RL, et al. 2015. Global cancer statistics, 2012. CA
from chromosome 5q21. Science, 253(5020), 661–​665. PMID: 1651562. Cancer J Clin, 65(2), 87–​108. PMID: 25651787.
Kinzler KW, and Vogelstein B. 1996. Lessons from hereditary colorectal can- Uemura N, Okamoto S, Yamamoto S, et al. 2001. Helicobacter pylori infection
cer. Cell, 87(2), 159–​170. PMID: 8861899. and the development of gastric cancer. N Engl J Med, 345(11), 784–​789.
Kinzler KW, and Vogelstein B. 1997. Cancer-​susceptibility genes: gatekeepers PMID: 11556297.
and caretakers. Nature, 386(6627), 761–​763. PMID: 9126728. van den Heuvel M, and Ingham PW. 1996. Smoothened encodes a receptor-​like
Kishimoto M, Kohno T, Okudela K et al. 2005. Mutations and deletions of the serpentine protein required for hedgehog signalling. Nature, 381(6591),
CBP gene in human lung cancer. Clin Cancer Res, 11(2 Pt 1), 512–​519. 547–​551. PMID: 8700230.
PMID: 15701835. Varley JM. 2003. Germline TP53 mutations and Li-​Fraumeni syndrome. Hum
Klein G. 2002. Perspectives in studies of human tumor viruses. Front Biosci, 7, Mutat, 21(3), 313–​320. PMID: 12619118.
d268–​274. PMID: 11779700. Vogelstein B, Papadopoulos N, Velculescu VE, et  al. 2013. Cancer genome
Knudson AG, Jr. 1971. Mutation and cancer: statistical study of retinoblastoma. landscapes. Science, 339(6127), 1546–​1558. PMCID: PMC3749880.
Proc Natl Acad Sci U S A, 68(4), 820–​823. PMCID: PMC389051. Watson R, Oskarsson M, and Vande Woude, GF. 1982. Human DNA
Knudson AG. 2002. Cancer genetics. Am J Med Genet, 111(1), 96–​ 102. sequence homologous to the transforming gene (mos) of Moloney
PMID: 12124744. murine sarcoma virus. Proc Natl Acad Sci U S A, 79(13), 4078–​4082.
Latif F, Tory K, Gnarra J, et al. 1993. Identification of the von Hippel-​Lindau disease PMCID: PMC346580.
tumor suppressor gene. Science, 260(5112), 1317–​1320. PMID: 8493574. Weinberg RA. 1996. How cancer arises. Sci Am, 275(3), 62–​70. PMID: 8701295.
Lawrence MS, Stojanov P, Polak P, et al. 2013. Mutational heterogeneity in can- Wooster R, Bignell G, Lancaster J, et al. 1995. Identification of the breast cancer
cer and the search for new cancer-​associated genes. Nature, 499(7457), susceptibility gene BRCA2. Nature, 378(6559), 789–​792. PMID: 8524414.
214–​218. PMCID: PMC3919509. Xu X, Hou Y, Yin X, et al. 2012. Single-​cell exome sequencing reveals single-​
Miki Y, Swensen J, Shattuck-​Eidens D, et  al. 1994. A strong candidate for nucleotide mutation characteristics of a kidney tumor. Cell, 148(5), 886–​
the breast and ovarian cancer susceptibility gene BRCA1. Science, 895. PMID: 22385958.
266(5182), 66–​71. PMID: 7545954. Zidi I, Mestiri S, Bartegi A, and Amor NB. 2010. TNF-​alpha and its inhibitors
Moran A, O’Hara C, Khan S, et al. 2012. Risk of cancer other than breast or in cancer. Med Oncol, 27(2), 185–​198. PMID: 19277912.
ovarian in individuals with BRCA1 and BRCA2 mutations. Fam Cancer, zur Hausen H, and de Villiers EM. 1994. Human papillomaviruses. Annu Rev
11(2), 235–​242. PMID: 22187320. Microbiol, 48, 427–​447. PMID: 7826013.
 19

3 Morphological and Molecular Classification of Human Cancer

MARK E. SHERMAN, MELISSA A. TROESTER, KATHERINE A. HOADLEY,


AND WILLIAM F. ANDERSON

OVERVIEW The “basket trial” concept has received less attention in the context
of etiological and prevention research than in the realm of treatment
Defining etiological, biological, and clinical heterogeneity are impor- trials, but recognition that cancers of different sites may share com-
tant goals in epidemiologic research. To achieve these aims, epide- mon etiologic exposures and molecular mechanisms of carcinogene-
miologists conduct trans-​ disciplinary studies to relate both newly sis suggest that this concept may have similar potential. For cancers
identified and established cancer risk factors to specific cancer sub- that are causally linked to an infectious agent, such as carcinogenic
types defined by histopathological and molecular features. Thus, the human papillomavirus (HPV) genotypes (Jemal et al., 2013), highly
traditional model of identifying cancer risk factors by organ site alone effective prophylactic HPV vaccines may lower risks of HPV-​related
has been supplemented, and largely supplanted. cancers of the cervix, vagina, vulva, anus, penis, and head and neck
Relating specific cancer risk factors to groups of tumors that share (Herrero et al., 2015). For cancers driven by common molecular
a common molecular pathogenesis offers opportunities to strengthen mechanisms, such as specific DNA repair defects (e.g., Lynch syn-
risk associations and provides a strong basis for translational research drome and BRCA1/​2 mutations), hormonal imbalances or metabolic
on early detection or prevention based on underlying biology. The disturbances (e.g., breast and gynecologic cancers), the possibility of
promise of this strategy has been revealed through efforts to offer identifying individuals at risk for a set of cancers that are related to
carriers of deleterious mutations in high-​penetrance genes, such as common etiological and/​or mechanistic factors offers the prospect of
BRCA1/​2, comprehensive strategies to prevent breast and ovarian lowering incidence rates of several cancers with a single intervention.
cancers, and the administration of prophylactic human papillomavi- Achieving this aim will require epidemiologists to extend their efforts
rus (HPV) vaccines to dramatically reduce risks of cervical cancer from accurate assessment of exposures to detailed characterization of
and HPV-​induced cancers arising at other anogenital sites and in the endpoints.
head and neck. Evolving methods of tumor classification integrate data across mul-
This chapter provides a primer on approaches that use pathology tiple scales, encompassing populations, patient-​level factors, primary
and molecular biology to subclassify cancers in epidemiologic studies. anatomical site, histopathological typing, and molecular characteriza-
Given the breadth of this topic and its rapid evolution with changing tion. Research at each of these scales, as well as synthesis of informa-
technologies, the goal is to emphasize important principles and pro- tion across them, is needed to gain the comprehensive understanding
vide illustrative examples, rather than to comprehensively review this of cancer etiology and pathogenesis that will be required to develop
subject. Readers are encouraged to consult specific chapters through- improved strategies for cancer prevention and lower cancer mortality.
out the text that demonstrate the application of the principles presented The aims of this chapter are to introduce the breadth of approaches
to cancers of specific organs or exposures. for tumor classification, comment on their strengths and weaknesses,
and illustrate their applications in epidemiological research. Given the
immensity of this subject, and its rapid evolution, this chapter empha-
INTRODUCTION sizes histopathologic and molecular classifications of cancer and their
relationships with complementary approaches (Hoadley et al., 2014).
Cancer is a biologically and clinically heterogeneous class of dis- Specific examples are provided to demonstrate principles. Detailed
eases. Much of epidemiological research is concerned with under- discussions of individual organ sites are presented throughout this
standing the causal and mechanistic sources of this diversity volume.
because identification of etiologically “refined” cancer subtypes
may increase the specificity of risk associations and predictions,
enabling more effective screening, prevention, and population CHARACTERISTICS AND DEFINITIONS
surveillance. OF NEOPLASMS (NEW GROWTHS), CANCERS,
Cancers arising from different organs are associated with distinct AND CANCER PRECURSORS
clinical presentations and behaviors, and therefore primary ana-
tomic site of origin is a critical determinant of clinical management. The synonymous terms “neoplasm” and “tumor” refer to new growths
However, expanding knowledge of the epidemiology, pathology, and that do not respond physiologically to growth signals and disrupt nor-
molecular profiles of cancer, and the intersection of these factors, has mal anatomy. Tumors are further categorized as benign or malignant.
led to development of novel tumor classifications, which categorize Benign neoplasms (and tumor-​like lesions, such as hamartomas) are
tumors within and across organ sites and disciplines. This new per- circumscribed, remain confined to the primary site of origin, and
spective has had a major influence on therapeutic research, and has are usually curable with surgical removal. In contrast, lesions desig-
led to development of “basket trials,” in which participant eligibility nated with the synonymous terms “malignant neoplasm” or “cancer”
is based on molecular characteristics of tumors, rather than primary are defined by the potential to invade and destroy benign tissues and
sites. In treatment trials, such as the MATCH Trial (McNeil, 2015), metastasize (spread) to other sites.
individuals with cancers that demonstrate particular candidate driver Metastatic lesions cause death by damaging normal tissues, leading
mutations are selected to receive targeted therapies, irrespective of site to organ dysfunction, and by producing deleterious systemic effects,
of origin. Basket trials are innovative, but their promise is tempered such as coagulation abnormalities or metabolic disturbances that dis-
by data suggesting that primary site remains an indicator of response, rupt homeostasis. Even small metastases may prove lethal, if deposits
even among cancers that share a common mutation in a putative driver impair critical physiological functions, such as occurs with metasta-
gene, such as BRAF (Hyman et al., 2015). ses to cardiorespiratory centers in the brainstem. In some instances,

19
20

20 Part I:  Basic Concepts


cancers result in poorly understood metabolic defects leading to have irregular shapes and uneven chromatin distribution, and appear
multiple organ failures, arrhythmias, cardiorespiratory arrest, hem- hyperchromatic (dark) on hematoxylin and eosin-​stained sections (a
orrhage, or thrombosis. Other cancers behave indolently initially and correlate of aneuploidy or polyploidy).
may remain asymptomatic for long periods of time, even if untreated. Histopathology aims to predict biological behavior by analyzing tis-
Asymptomatic cancers may be detected through targeted screening sues, whereas cytopathology relies on the microscopic appearance of
or incidentally via routine clinical practices such as blood tests or individual cells and small cell clusters. Newer morphologic computer-​
radiologic imaging. The behavior of certain cancers, particularly a based methods are demonstrating the possibility of predicting cellular
subset of breast and prostate cancers, is characterized by variable behavior based on characteristics beyond unaided human visualization
periods of dormancy at metastatic sites, which may become clinically (Beck et al., 2011). Further, improved methods for identifying and/​or
symptomatic years following initial diagnosis and treatment (Yeh isolating rare circulating tumor cells based on physical properties and
and Ramaswamy, 2015). Thus, defining the mechanisms and mark- molecular markers may eventually enable detailed characterization of
ers associated with asymptomatic cancers and late recurrences is an such cells to predict prognosis or guide treatment, especially in the
important challenge. setting of therapy-​resistant or recurrent cancer. With respect to isolat-
The definition of metastasis is sometimes complex. For example, ing circulating tumor cells, developing methods to improve recovery,
endometriosis is a benign disease that is hypothesized to develop purity, viability, relative enrichment, and throughput remain as chal-
from sloughed benign menstrual endometrium (uterine lining) that lenges (Esmaeilsabzali et al., 2013). Analysis of cell free macromol-
passes through the fallopian tube, implants, and grows outside of the ecules (DNA, RNA, etc.) derived from cancers represent an alternate
uterus, especially in the ovaries (Vercellini, 2015). Epidemiologic and approach.
molecular data suggest that in rare instances, endometriotic lesions A goal of quantitative pathology techniques is to convert sub-
may undergo malignant change, thereby giving rise to ovarian endo- jective semi-​quantitative and qualitative observations (i.e., ana-
metrioid or clear cell carcinomas (Pearce et al., 2012). Further, tubal log data) to measured quantities (digital data) that are amenable
ligation is protective for ovarian cancer, particularly the endometrioid to statistical analysis and interpretation. While this process brings
subtype, which could reflect a reduction in endometriotic lesions from the power of analytical methods to bear, unrecognized subjective
which these tumors arise, among other postulated mechanisms (Rice clues that experienced pathologists use may be lost. Conversely,
et al., 2012). Other non-​metastatic processes may also “transport” identification of quantitative features with image analysis may help
benign or malignant cells to distant sites, either naturally or secondary pathologists refine microscopic diagnostic criteria. Nonetheless,
to instrumentation, but if such lesions do not cause harm, they may not under certain states, benign cells may demonstrate morphologic
constitute metastases. and non-​morphologic features that overlap with cancer, including
mutations in cancer-​associated genes, such as p53 in the fallopian
tube, PTEN in endometrium, and mutations in sun-​exposed eyelid
PATHOLOGY IN CANCER DIAGNOSIS AND RESEARCH skin (Lee et  al., 2007; Martincorena and Campbell, 2015; Mutter
et al., 2001).
In diagnostic pathology, macroscopic, microscopic, and molecu- Proliferation is among the best-​recognized features of cancer cells,
lar features in conjunction with clinical data are used to distinguish but like many other cancer hallmarks (Hanahan and Weinberg, 2011),
non-​neoplastic from neoplastic lesions, and to divide the latter into it is not a pathognomonic or specific diagnostic feature. Benign cells
benign or malignant tumors. Although pathologic criteria are gener- located at the base of epithelia and immature cells in benign bone
ally useful in predicting the average behavior of groups of tumors, the marrow may also be highly proliferative. However, in contrast to can-
performance of histopathology alone in estimating the prognosis and cer cells, mitotically active benign cells help preserve homeostasis
treatment response of individual cancers is more variable. The power by responding appropriately to the balance of multiple signals (e.g.,
of morphology derives from the concept that it reflects the integrated growth factors, hormones), reflect the intrinsic state of cells (e.g.,
interaction of tumor and host characteristics over time (i.e., form fol- senescence, dormancy), and interact physiologically with the systemic
lows function). However, the force of this advantage is limited by the milieu and the microenvironment, rather than dividing autonomously.
inability to accurately measure specific features, which reflects the Cancer treatments, such as chemotherapy and radiation, that non-​
subjectivity of visual assessment, the enormous morphologic vari- specifically damage dividing cells may also injure benign dividing
ability of malignant and benign tissues under varying physiologic and cells, accounting for some adverse effects of these treatments (e.g.,
pathophysiologic states, and the effects of technical artifacts. In addi- bone marrow toxicity or mucositis). In response to injury (e.g., radia-
tion, different molecular alterations may yield similar morphologic tion or chemotherapy) or hormone exposures, benign cells may show
features. nuclear pleomorphism (variable appearances) that mimics cancer
Benign neoplasms show greater cellular organization than malig- cells (Arias-​Stella, 2002; Oliva et  al., 2013). The broad overlapping
nant tumors and are composed of cells that more closely resemble range of morphologic appearances of benign and malignant cells may
normal cells of the organ site of origin (Table 3–​1). Microscopically, pose challenges for classifying lesions using morphologic and non-​
benign tumor cells are generally smaller than cancers cells and con- morphologic methods.
tain smaller nuclei with modest variability in size and shape, mini-
mal hyperchromasia, fine chromatin detail, a lower ratio of nuclear
to cytoplasmic area, and a low mitotic index. Malignant tumors are CLASSIFICATION/​TERMINOLOGY OF MALIGNANT
composed of cells in disordered arrangements, which are character- TUMORS (CANCERS)
ized by a higher ratio of nuclear to cytoplasmic area and nuclei that
Overview
Table 3–​1. Distinctive Features of Benign Versus Malignant Tumors Cancers are divided according to states or stages of progression
(Cancers) (American Joint Committee on Cancer [AJCC], 2011), such as in situ
versus invasive, and the latter are grouped according to patterns of
Feature Benign Malignant differentiation as carcinomas (epithelial) versus sarcomas (mesenchy-
mal or connective tissue). Lymphoid/​hematologic tumors (technically
Differentiation High—​resembles normal Variable—​basis for grading
sarcomas), germ cell tumors, and melanomas (technically carcinomas)
Pleomorphism Generally low Variable—​generally high
are typically considered separately because of their distinctive biologi-
Mitoses Variable Variable—​may be abnormal
cal and clinical features (Figure 3–​1). The basic morphologic classi-
Organization High Disorganized
fication of cancers is important because it is a fundamental indicator
Growth Circumscribed or Invasive, metastatic potential
of tumor biology and is related to etiology, pathogenesis, molecular
encapsulated
alterations, and clinical management.
 21

Morphological and Molecular Classification of Human Cancer 21


Tumor = Neoplasm lined by squamous epithelium, but glandular metaplasia in the form
of Barrett’s mucosa, often occurring in the context of chronic gastric
reflux, may undergo dysplasia and progress to adenocarcinoma (gland
Benign Malignant Tumor forming cancer) (Runge et al., 2015). Analogous to many other organ
• “omas” Malignant Neoplasm sites, more meticulous evaluation (endoscopy and biopsy) has led to
• Adenoma (glands) Cancer
• Fibroma, Chondroma, increased detection of Barrett’s mucosa with dysplasia (i.e., putative
etc. (Any Stromal cancer precursor), but lowered risks of progression to cancer (Runge
Component)
• Tumor-like
Carcinoma Sarcoma Other et al., 2015). Similarly, the development of squamous metaplasia and
• Lymphoma
• Papillomas • Epithelial • Stroma
• Melanoma
dysplasia in large bronchi of smokers may contribute to the high fre-
• Polyps
• Germ Cell quency of squamous cell carcinomas centrally in the lung (Ishizumi
et al., 2010). It is notable that the inter-​observer reproducibility of dys-
Adenocarcinoma Squamous Cell Carcinoma Other plasia diagnoses in Barrett’s mucosa, bronchial epithelium, and other
Undifferentiated sites is often imperfect, if not suboptimal.
Neuroendocrine
Carcinosarcoma

Definition and Significance of Carcinoma in Situ (CIS)


Figure 3–​1.  Terminology of neoplasms.
Carcinomas arise from alterations in benign epithelium, denoted by
terms such as “dysplasia,” “atypia,” or “atypical hyperplasia.” The
most proximate antecedent of invasive carcinoma is referred to as “in
Carcinomas Are Malignant Tumors That Demonstrate situ carcinoma” (carcinoma in situ [CIS]). In CIS, “malignant appear-
Epithelial Differentiation ing” cells are confined within a preexisting epithelial lining or struc-
ture, whereas in invasive carcinoma, malignant cells invade adjacent
Carcinomas arise predominantly from the surface linings of hollow tissue compartments (Figure 3–​2). The boundaries between epithelia
viscera, including the respiratory, gastrointestinal, gynecological, and (e.g., linings of visceral lumens) and underlying tissues (walls of vis-
genitourinary tracts, or solid organs, such as breast, prostate liver, pan- cera or dermis) are demarcated by basement membranes, which are
creas and kidney, and skin. Given that the lumens of hollow viscera complex acellular structures composed mainly of collagens and pro-
are connected, at least indirectly, to the environment outside the body, teins (Halfter et al., 2015). These acellular structures contribute impor-
these surfaces are exposed to potential carcinogens derived from both tantly to tissue compartmentalization, organization, and biophysical
the external (exogenous) environment (e.g., inhaled, ingested, ascend- properties, and change with aging and pathologic states.
ing genital and genitourinary pathogens) and those generated within Clinically, the term “cancer” generally refers to invasive carcinoma,
the body (endogenous). The thickness, cellular composition, and a lesion that has metastatic potential by virtue of breaching the base-
organization of visceral epithelium are highly variable, but a major ment membrane and gaining access to blood vessels and lymphatic
distinction is whether the lining is composed of squamous cells or channels. Considering CIS as “cancer” is arguably a misnomer because
glandular cells. CIS lacks these features, and generally does not pose risks of lethal-
Maintenance of epithelia requires continuous regeneration to replace ity. The majority of CIS lesions are asymptomatic and are detected
cells that have died, have been shed, or have undergone senescence. by screening with methods such as Papanicolaou (Pap) smear, mam-
Epithelium is characterized by a more highly proliferative basal com- mography, colonoscopy, and so on. The term “CIS” has engendered
partment (away from lumen) and a non-​dividing compartment charac- substantial controversy because of its somewhat arbitrary distinction
terized by cellular maturation or differentiation (specialized function). from less-​developed precursor lesions of invasive carcinomas and its
Current knowledge about number, distribution, and plasticity of stem threatening connotation.
cells, factors that may influence expansion, contraction, or elimination Cancer registries maintain variable practices with respect to record-
in the context of mutations, and their tissue context-​dependent func- ing CIS lesions at different organ sites. Monitoring incidence rates of
tions (i.e., stem cell niches) continues to evolve (Frede et al., 2014). CIS is important in organ systems that are subjected to cancer screen-
The number of divisions in normal stem cells required for epithelial ing because the diagnosis of these lesions typically prompts treat-
maintenance in specific organs is hypothesized to correlate with can- ment to prevent progression. However, progression of CIS to invasive
cer risk (Tomasetti and Vogelstein, 2015). Thus, the predominance of
carcinomas as opposed to other types of cancer among humans may
reflect the total number of cells comprising epithelia, the requirement
for cells to proliferate to maintain these epithelia (perhaps increasing
the risk of replication errors and clonal expansion of such cells), expo-
sure to carcinogens, and human longevity, with its attendant decline in
functions, such as immunity, DNA repair, telomere maintenance, epig-
enomic markers, and other mechanisms that promote genetic stability
(Franzke et al., 2015).
Visceral epithelium is typically multilayered and coated with vari-
ous acellular substances, such as keratin or mucins that protect under-
lying parenchyma from injurious agents (i.e., mucosae). In addition,
the epithelia are in contact with a microbiome film, which is receiving
increasing attention with respect to its role in cancer and other dis-
eases. Metaplasia, the process by which one type of benign epithelium
is replaced by another, occurs at multiple organ sites. Anatomic sites of
metaplasia are often sites of cell turnover and cancer formation.
Metaplastic transitions between squamous and glandular epithe-
lium occur normally at the junctions of the ectocervix and endocervix,
rectum and anus, larynx and trachea, and esophagus and stomach. In Figure 3–​2.  Left: Colon polyp forming exophytic mass projecting from
general, squamous cell carcinomas arise from squamous epithelium colonic mucosa. Note well-​formed glands invading stalk of polyp (arrow).
and adenocarcinomas from glandular epithelium. However, the devel- Right: High power view showing invasive adenocarcinoma (malignant
opment of metaplasia at aberrant sites, often in response to epithe- glands) associated with inflammation and stromal changes. From website:
lial injury, may result in carcinomas with differentiation that does not Juan Rosai’s Collection of Surgical Pathology Seminars (http://​rosaicol-
match that of the native epithelium. For example, the esophagus is lection.org/​)
2

22 Part I:  Basic Concepts


carcinoma is neither certain nor necessarily rapid, and some CIS CARCINOMAS: MACROSCOPIC
lesions may regress. However, methods to accurately predict which AND HISTOPATHOLOGIC FEATURES
CIS lesions will become invasive and possibly metastasize during a
patient’s lifetime are lacking. If detecting and treating CIS is beneficial, Most carcinomas form discrete, ill-​defined, firm masses, although
then these interventions should lead over time to reduced population-​ diffusely infiltrating carcinomas that merge imperceptibly with sur-
based incidence rates of invasive carcinomas (in the absence of other rounding tissues are characteristic of tumors such as lobular carci-
changes in population risk) and, eventually, lower mortality. noma of the breast and signet ring cell gastric carcinoma (Figure
Removal or destruction of screen-​detected cancer precursors in the 3–​3). Carcinomas may be associated with desmoplastic stroma that
colon and cervix has led to substantial reductions in the incidence of includes abundant collagen, and imparts a firm consistency (scir-
invasive cancers at these sites (Brenner et al., 2014; Peirson et al., 2013). rhous). Other carcinomas, including medullary carcinomas, are
Given that only a fraction of cancer precursors is destined to progress, soft secondary to high cellularity with scant stroma. Central soften-
screening inevitably results in “over-​diagnosis” and “over-​treatment” ing is also characteristic of rapidly growing cancers that have out-
of many innocuous lesions. Controversies in mammography screening grown their blood supply and have undergone necrosis, which may
for breast cancer exemplify this issue. During 1980–​2004, diagnoses lead to metabolic changes and acquisition of aggressive features
of ductal carcinoma in situ (DCIS) increased seven-​fold, and rates of (Cairns, 2015).
early-​stage invasive cancer have increased, whereas incidence rates of Carcinomas are characteristically composed of cohesive cells that
metastatic breast cancer have remained stable, supporting the impor- express specific molecular weight keratin proteins (Ordonez, 2013) and
tance of over-​diagnosis (Bleyer and Welch, 2013; Sherman et  al., show evidence of differentiation, such as intra-​or extra-​cellular keratin
2014; Welch et al., 2015). (squamous cell carcinoma) or gland formation or secretions (adeno-
Risk factor associations for CIS and invasive cancer in many organs carcinoma) (Figure 3–​4). Squamous cell carcinomas form distinctive
are similar, as might be expected for factors that are genetic, devel- cell-​cell attachments referred to as intercellular bridges.
opmental, or act early in adult life. Nonetheless, risk associations for Invasive carcinoma may undergo epithelial mesenchymal transi-
“cases” may differ, depending on whether CIS is included, especially tion, which is characterized by reduced intercellular cohesion and loss
in populations with screening. Most CIS lesions, even in organs that of polarity, increased migration, and shape change from cuboidal to
can be palpated such as the breast, are found by screening (Claus et al., spindly, consistent with a mesenchymal phenotype (Pasquier et  al.,
2001); thus, differences in screening practices across populations 2015). These changes are best appreciated in vitro and are not reli-
could affect the consistency of risk factor associations in the epide- ably defined by microscopy in clinical specimens. Invasion results in
miologic literature, particularly for CIS. epithelial-​stromal interactions, which may involve fibroblasts, adipo-
Issues related to the validity and utility of CIS as an endpoint are cytes, immune and non-​immune inflammatory cells, and lymphatic
further complicated by variability in the biology of CIS. Some CIS and blood vascular channels. Historically, stroma was viewed as a pas-
lesions are composed of cells that already possess the capacity to sive contributor in carcinogenesis, but this view has been supplanted
invade and metastasize, whereas others lack invasive potential. For by a model that emphasizes the important role of tumor–​stromal inter-
example, incidence rates of cervical intraepithelial neoplasia 3 (i.e., actions in either promoting or inhibiting cancer progression. In fact,
CIN 3, severe dysplasia, or CIS) among screened women in their late the formation of metabolically activated cancer-​associated fibroblasts
twenties or thirties are comparatively high, but invasive squamous and cancer-​associated adipocytes may contribute to tumor growth by
cell carcinomas are uncommon until women reach their forties and producing factors that stimulate proliferation (Marcucci et al., 2014).
fifties, and the morphology, molecular alterations, and biology of Cancer-​associated fibroblasts often appear enlarged, express smooth
these lesions are different (den Boon et al., 2015; Hariri et al., 2015). muscle proteins (e.g., actin), and are rich in organelles when examined
Accordingly, most CIN 3 lesions have neither invaded nor possess that by electron microscopy.
capacity. In contrast, observational findings suggest that some intraep- The composition of stromal matrix has received limited attention in
ithelial lesions (endometrial intraepithelial carcinoma or serous tubal cancer epidemiology, but factors such as hyaluronan, which interacts
intraepithelial carcinoma) may exfoliate cells that pass into the peri- with CD44 receptors, may promote epithelial mesenchymal transition,
toneal cavity (either through or from the fallopian tube), implant on a stem cell–​like phenotype, and features associated with aggressive-
the peritoneum, and grow as metastases (Rabban and Zaloudek, 2007; ness and treatment resistance (Chanmee et al., 2015). In contrast, spe-
Seidman et al., 2011; Wheeler et al., 2000). cific types of immune infiltrates may improve prognosis for several
cancer types, and efforts to expand breast cancer staging to include a
characterization of the density of such infiltrates has been proposed
Terminology of Cancer Precursors, Including (Salgado et al., 2015). Biophysical properties of tumor stroma, specifi-
Carcinoma in Situ cally stiffness, have emerged as important in carcinogenesis and can-
cer progression and may contribute broadly to the hallmarks of cancer
Epithelial intraepithelial neoplasia (EIN) terminology is conceptu- (Pickup et al., 2014). Further, the topography and alignment of colla-
alized as a universal nomenclature for carcinoma precursor lesions, gen fibers may be important in invasion. For example, data suggest that
ranging from least to most severe (EIN 1, 2, 3), which can be applied to
all organ sites (e.g., cervical [CIN], vaginal [VAIN], prostatic intraepi-
thelial neoplasia [PIN], etc.). The term “neoplasia” has been criticized
because it may be misconstrued as cancer. In addition, the numbering
of EIN lesions might suggest an orderly progression through levels of
increasing severity, but evidence that such progression occurs in tis-
sues is lacking for many organ systems. Further, risk of progression of
EIN to invasive carcinoma may differ by organ site, characteristics of
lesions, patient characteristics, and exposures.
Prompted by concerns related to over-​diagnosis of mammographi-
cally detected DCIS, the novel term “indolent lesions of epithelial ori-
gin” (IDLE) has been proposed for these lesions (Esserman et al., 2014).
A fundamental dilemma is that the natural history of these putative cancer
precursors is ill-​defined, in part because biopsy and resection alter their
natural history. Further, the nature of intraepithelial lesions may change Figure 3–​3. Left: Gastric wall expanded by diffuse infiltrate of tumor
as the sensitivity of screening methods increases over time. For example, cells. Right: High power view demonstrating signet ring cell adenocarci-
CIN 3 lesions detected with sensitive HPV DNA testing are often so noma. Blue-​grey material is mucin. From website: Juan Rosai’s Collection
small that they are removed by diagnostic biopsy (Sherman et al., 2003). of Surgical Pathology Seminars (http://​rosaicollection.org/​)
 23

Morphological and Molecular Classification of Human Cancer 23

Figure 3–​4.  Left: Squamous cell carcinoma demonstrating abundant keratin formation and keratin pearls (arrow). Right: Well-​formed glands of adenocar-
cinoma comprised of malignant appearing cells surrounding gland lumens. From website: Juan Rosai’s Collection of Surgical Pathology Seminars (http://​
rosaicollection.org/​)

parallel fibers at the boundary between breast cancer and surrounding and organization similar to that of corresponding benign cells.
tissues may create a path of “least resistance” for tumor migration and Histopathologic grade, as assessed microscopically, is associated with
portend a worse survival (Riching et al., 2014). genomic instability, and in many cancers with proliferation. In breast
Immune infiltrates in the tumor microenvironment can be charac- cancer, grade is related to Ki67 index, assessed by immunohistochem-
terized using immunohistochemistry, which may provide information istry (proliferation marker) and molecular panels that reflect prolifera-
about prognosis and treatment responses. Immune cells can be either tion, but genomic grade, which is based on molecular analysis, may
pro-​tumorigenic or tumoricidal, and a major goal of certain forms of provide refined prognostic information (Ignatiadis et al., 2016).
cancer immunotherapy is to “educate” the immune system to respond In some tumors, such as prostatic adenocarcinoma, the Gleason
with tumoricidal mechanisms. Immune signatures are also prominent grade is particularly important for prognosis and choosing manage-
in the molecular classification of many tumor types. Dense immune ment. Men diagnosed on biopsy with well-​differentiated adenocarci-
infiltrates are characteristic of medullary carcinomas arising at vari- nomas (low Gleason scores) may opt for observation alone, whereas
ous organ sites, such as breast and pancreas. Cancers associated with men with poorly differentiated adenocarcinomas (higher Gleason
defects in DNA mismatch repair may produce abundant neoantigens scores) generally require aggressive management. However, increased
that elicit dense lymphoid infiltrates, but expression of immune check- understanding of the molecular pathogenesis of prostate cancer and
point ligands on the tumor cells diminishes the effectiveness of immune its progression under treatment suggest the value of a new classifi-
surveillance. Recent data suggest that treatment of these cancers with cation with immediate treatment implications, reflecting whether a
checkpoint inhibitors may unleash effective immune responses (Diaz tumor is androgen-​dependent, and the hormone stimulation is endo-
and Le, 2015). Ironically, the term “inflammatory breast carcinoma” crine or paracrine, or has progressed to a more autonomous, androgen-​
refers to a cancer that mimics mastitis clinically, but the erythematous independent growth phase (Logothetis et  al., 2013). Similarly,
and edematous appearance of the breast on physical examination pri- premenopausal women with low-​ grade endometrioid endometrial
marily reflects dermal lymphatic obstruction with tumor emboli, rather carcinomas may opt for fertility-​ preserving medical management,
than inflammation. whereas for other women, hysterectomy and adjuvant therapy are rec-
Most instances of inflammation represent rapid, non-​ specific ommended (Colombo et al., 2016).
innate immune responses (see Chapter  25 on immunologic factors). Stage refers to tumor size and extent of spread from local-​regional
Inflammation may disrupt basement membranes and alter tissue to distant sites. Grade can be a predictor of stage, but the latter is gen-
organization, resulting in epithelial–​ stromal interactions that may erally the stronger prognostic predictor. Although high stage augurs a
be pro-​carcinogenic, as reflected in the conceptualization of cancers poor prognosis, some tumors that are clinically considered low stage at
as “wounds that do not heal” (Dvorak, 2015). Observational epide- diagnosis later demonstrate metastases and result in death. For exam-
miological data suggests that anti-​ inflammatory medication may ple, a low-​stage estrogen receptor (ER)-​negative breast cancer may
lower risks of some cancers, but estimated effect sizes are variable have a worse prognosis than a higher stage well-​differentiated ER-​
and often small, and interpretations are limited in post hoc analyses, positive cancer. The impetus for developing molecular marker panels
which may misclassify the amount, frequency, and timing of drug to predict prognosis derives from frustrations with the shortcomings
use in relation to potentially pro-​carcinogenic inflammatory insults. of traditional assessment of grade and stage. Thus, adjuvant systemic
The suggested role of inflammation in carcinogenesis underpins the treatment is often given even after surgery for early-​stage tumors, if
exploration of anti-​ inflammatory agents for chemoprevention (see risk of recurrence is considered unacceptably high.
Chapter 23 on pharmaceutical drugs other than hormones). The role For some tumors, neoadjuvant therapy is administered prior to
of adaptive immunity in cancer is more complex. Antigen-​specific resection to assess tumor response (and to facilitate surgery). However,
immunity responses may inhibit tumor development or progression in the biology of metastatic tumor cells or circulating tumor cells may
some instances and elevate risk in others (e.g., autoimmune disease differ from that of the tumor at its primary site, especially after treat-
and lymphoma, and inflammatory bowel disease and colon cancer). ment. Thus, the development of “liquid biopsy” methods that enable
Adaptive immune responses provide the basis for the development of repeated molecular analysis of circulating tumor cells during treatment
vaccines and immune therapies for treatment and prevention (Finn, is emerging as a potential method to detect chemotherapy resistance
2014; Mandal et al., 2014). (Dawson et al., 2013).
Most carcinomas that metastasize spread via the lymphatic system;
accordingly, removal and examination of draining lymph nodes for
Predictors of Carcinoma Clinical Behavior: Grade metastatic deposits is often performed to stage carcinomas. The pri-
and Stage mary purpose of staging lymphadenectomy is to guide the need for sys-
temic therapy, rather than to remove the cancer deposits, although the
Grade refers to the degree of tumor differentiation—​the extent to which latter may have debatable effects on risk of recurrence. Demonstration
carcinomas cells show cytologic features, functional specialization, that patterns of lymphatic drainage are predictable has enabled the
24

24 Part I:  Basic Concepts


development of “sentinel node” techniques in which examination of diagnostic criteria and immunohistochemistry has improved the accu-
one or a few nodes may provide required staging information with- racy of pathology diagnosis, but misclassification persists, particularly
out full lymph node dissection (Niebling et  al., 2016). Specifically, in studies that rely on self-​reported data to define primary site. As the
if sentinel nodes are negative, the entire nodal basin is considered molecular profiles of different tumor types have become more com-
cancer-​free, and a complete dissection is avoided, reducing morbid- prehensively defined, the possibilities for distinguishing primary from
ity and operative time. If sentinel nodes are positive, the remaining metastatic sites has increased.
nodes in the region are typically removed to provide complete staging.
Carcinomas may also spread hematogenously to sites such as liver,
lung, and bone, or invade through viscera, leading to spread via direct Cancers Demonstrating Stromal
extension or release of cancer cells into body cavities, producing fluid
Differentiation: Sarcomas and Carcinosarcomas
collections containing malignant cells (i.e., malignant ascites or pleu-
ral effusions). Among adults, sarcomas are extremely rare, whereas these tumors
Although anatomy and lymphovascular drainage are likely impor- account for a greater percentage of non-​hematologic cancers among
tant determinants of sites of metastases, accumulating data indicate children. Incidence rates of osteogenic sarcomas spike during ado-
that molecular characteristics of primary tumor cells and microen- lescence (Ward et al., 2014). Sarcomas also occur among adults fol-
vironments at potential metastatic sites are also significant (Langley lowing treatment for cancer. Sarcomas are classified based upon the
and Fidler, 2011). Building on the “seed and soil” hypothesis that was matrix products they produce or the mesenchymal differentiation they
first proposed by Stephen Paget in the late nineteenth century (Paget, display: cartilage (chondrosarcoma); osteoid (osteogenic sarcoma);
1889), investigators have proposed the “cancer diaspora” concept, collagen (fibrosarcoma); smooth muscle (leiomyosarcoma), skeletal
which analogizes cancer spread with phases of emigration, migration, muscle (rhabdomyosarcoma); or blood vessels (angiosarcoma) (Figure
and immigration in human populations (Pienta et al., 2013). This view 3–​5). Grossly, these tumors present as large masses, sometimes with
proposes continued interactions between metastatic cells and primary focal softening if partly necrotic. However, some large benign mes-
tumors, including homing of circulating tumor cells back to primary enchymal tumors, such as leiomyomas, may also infarct. Sarcoma
sites, and has prompted the theoretical suggestion that inducing cells cells are detached, and have been classified as round cell, epithelioid
to migrate to sites unfavorable for growth and survival might represent (morphology that mimics carcinoma), or spindle cell, as opposed to
a therapeutic strategy. the typical cuboidal or columnar shape of carcinoma cells. Sarcomas
Knowledge of usual patterns of metastases may aid in the recogni- usually spread hematogenously; hence, lymph node staging is not per-
tion of misclassification of primary tumor sites. For example, gastro- formed routinely for these tumors.
intestinal cancers often metastasize via the draining portal circulation The molecular classification of sarcomas includes tumors with trans-
to the liver, and misclassification of metastatic adenocarcinoma as locations and relatively few alterations, tumors with complex karyo-
primary hepatocellular carcinoma may affect the interpretation of types, and others with point mutations or amplifications in suspected
epidemiological studies, especially when the former are numerically driver oncogenic mutations (Hameed, 2014). Testing for translocations
predominant. Metastatic lobular carcinomas of the breast may mimic in genes such as KMT2A/​MLL, ETV6, and EWSR1 is performed rou-
primary linitis plastica of the stomach (Cormier et al., 1980). Similarly, tinely in many centers. Interestingly, gene fusions do not necessarily
data suggest that metastases to the ovary from occult gastrointestinal correspond with specific histopathologic subtypes, such that classifi-
carcinomas have been frequently misclassified as primary mucinous cation requires integration of clinical, histopathological, and molecu-
ovarian carcinomas historically (Soslow, 2011). Use of improved lar data. Some sarcomas may form bone or involve bone and require

Figure 3–​5.  Sarcomas are malignant tumors displaying mesenchymal differentiation: top left: chrondrosarcoma; top right: osteogenic sarcoma; bottom
left: rhabdomyosarcoma; bottom right: leiomyosarcoma. From website: Juan Rosai’s Collection of Surgical Pathology Seminars (http://​rosaicollection.org/​)
 25

Morphological and Molecular Classification of Human Cancer 25


special processing prior to histologic processing, which may interfere [leukemias], 39 [Hodgkin lymphoma, 40 [non-​Hodgkin lymphoma],
with immunohistochemistry and other molecular testing. In contrast to and 41 [multiple myeloma]).
epithelia, which are bound by a well-​defined basement membrane, the
organization of benign mesenchymal tissue is less well understood. Categorization of Hematolymphoid Cancers
Consequently, sarcoma precursors are largely uncharacterized.
Carcinosarcomas are tumors composed of mixed components, Lymphoma.  The WHO lymphoma classification was updated in
including areas of carcinoma and sarcoma. Uterine carcinosarco- 2008 to reflect evolving concepts, including multidisciplinary clas-
mas, sometimes referred to as malignant mixed mullerian tumors sification, inclusion of borderline categories, and provisional entities
(MMMTs), are among the best studied (Berton-​Rigaud et al., 2014). requiring further study (Campo et al., 2011; Menon et al., 2012; Xie
Homologous MMMTs produce malignant stroma native to the uterus et  al., 2015). Like other tumor classifications, questions about what
(e.g., leiomyosarcomas [smooth muscle] or fibrosarcoma [fibrous constitutes a specific “entity” and whether distinctive molecular mark-
tissue]), whereas heterologous types produce stromal tissues not ers alone are sufficient to justify separate classification continue to
normally found in the uterus (e.g., osteogenic sarcoma [bone] or chon- evolve. Lymphomas are divided into HL or NHL (Figure 3–​6), and
drosarcoma [cartilage]). At other sites (e.g., breast), tumors contain- may affect both children and adults, but the biology and behavior of
ing malignant epithelial and mesenchymal elements are referred to these tumors differ substantially among younger and older persons.
as “metaplastic carcinomas.” Data suggest that many uterine carcino- Although screening per se is not performed routinely for hemato-
sarcomas are clonal, and the final diagnostic classification may not lymphoid malignancies, routine blood testing may prompt evaluation
indicate the subtype of cancer that was present when the tumor first of abnormal cell counts (e.g., lymphocytosis) and lead to discovery of
formed (D’Angelo and Prat, 2011). Diagnostic classification of can- asymptomatic (chronic lymphocytic leukemia or small lymphocytic
cer is based on the most clinically aggressive component because this lymphoma [CLL]) and precursor (monoclonal B-​cell lymphocytosis)
is paramount for determining prognosis and treatment; however, this disease. Similarly, detection of high levels of a particular immuno-
approach may not be ideal for etiologic studies. globulin or light chain in serum or urine may suggest an occult tumor
(e.g., plasma cell myeloma). These clinical results raise concerns about
over-​diagnosis, the need to treat, and the appropriateness of manage-
ment. In contrast to solid tumors, the natural history of hematolym-
Malignant Tumors of the Immune System phoid tumors and their putative precursors generally remains unaltered
and Blood: Lymphoma and Leukemia by procedures leading to their diagnosis; therefore, these tumors are
especially well suited to natural history studies that aim to identify
Overview markers and mechanisms associated with disease progression.
Historically, the hematolymphoid malignancies were divided into lym- Lymphomas can be further divided into tumors of immature (lin-
phomas (lymphopoietic cancers), which were defined mainly as solid eage precursor) or mature (peripheral) B-​cells or T/​NK-​cells, which
tumors arising in lymph node, spleen, or mucosal-​associated lymphoid reflect differences in cell of origin, etiological factors, and clinical
tissue, and leukemias (hematopoietic cancers), which were character- behavior. Factors considered in clinical classification of B-​cell lym-
ized by a predominance of tumor cells in blood and bone marrow. phomas include anatomic site of presentation, association with immu-
However, since 2001, the World Health Organization (WHO) has clas- nodeficiency states (ascribed to HIV infection or aging), morphologic
sified these malignancies according to lineage: myeloid, lymphoid, and features, detection of Epstein-​Barr virus or human Herpesvirus-​8,
histiocytic/​dendritic cell (Harris et al., 2000). The leukemias are now and expression of immunohistochemical markers of differentiation.
divided into two major groupings—​myeloid and lymphoid neoplasms. Less progress has been made in understanding T-​cell than B-​cell lym-
Myeloid neoplasms derive from granulocytic (neutrophil, eosinophil, phomas, perhaps reflecting the rarity of these neoplasms, incomplete
basophil), monocytic/​ macrophage, erythroid, megakaryocytic, and knowledge of normal T/​NK-​cell development, and the greater ten-
mast cell lineages, and include acute myeloid leukemia (AML), myelo- dency of these tumors to contain an admixture of tumor cells and reac-
dysplastic syndromes (MDS), myeloproliferative neoplasms (MPNs), tive cells (Campo et al., 2011).
and “overlap” disorders termed “myelodysplastic/​myeloproliferative In Hodgkin lymphoma, histopathologic features and systemic
neoplasms” (MDS/​MPN). Lymphoid neoplasms derive from B-​cells, symptoms are the mainstay of diagnosis (Kuppers et  al., 2012).
T-​cells, and natural killer (NK) cells and include Hodgkin (HL) and Hodgkin lymphoma is distinctive in that it forms masses composed of
non-​Hodgkin lymphomas (NHL), plasma cell neoplasms (e.g., mul- rare monoclonal Reed Sternberg cells in a background of many reac-
tiple myeloma), and lymphoid leukemias. B-​and T/​NK-​cell leukemias tive cells (variably including lymphocytes, plasma cells, and eosin-
are now classified with the lymphomas due to several of these entities ophils). These features pose challenges for molecular analysis. The
having circulating (blood) and solid (tissue) phases that represent dif- nature of Reed Sternberg cells remains uncertain; these cells share
ferent manifestations of the same disease (e.g., CLL [blood phase] and markers with germinal B-​cells, but also demonstrate dysregulation of
small lymphocytic lymphoma [tissue phase]; lymphoblastic leukemia NF-​ĸB and JAK/​STAT signaling and commonly latent infection with
[blood phase] and lymphoblastic lymphoma [tissue phase]). Epstein-​Barr virus.
Taxonomy of the hematolymphoid cancers demonstrates the value
of interdisciplinary, integrated approaches for tumor categorization.
Classification incorporates etiologic factors, clinical presentation, his-
topathologic features, and molecular markers that reflect cell type, cel-
lular growth patterns, and molecular characteristics related to aberrant
differentiation, incomplete maturation, and dysregulated proliferation.
A central diagnostic feature is identification of a monoclonal cell pop-
ulation with an aberrant phenotype, whereas diagnosis of most other
cancers relies more heavily on histopathologic and clinical evidence.
Rapid progress in understanding normal blood and lymphoid cell
development and maturation, combined with tools for defining clonal
proliferations, enabled the early development of the molecular hema-
tolymphoid taxonomies. As sensitive methods for detecting clonal cell
populations have developed, concerns about what constitutes a hema-
tologic cancer precursor or a cancer have arisen, analogous to concerns Figure 3–​6.  Left: Non-​Hodgkin diffuse large B-​cell lymphoma (left) and
about over-​diagnosis in other organ systems. The sections that follow Right: Hodgkin Lymphoma with prominent Reed Sternberg cell showing
briefly contrast approaches for classifying lymphomas and leukemias; binucleation with prominent nucleoli (arrow). From website: Juan Rosai’s
more details are provided elsewhere in this volume (Chapters  38 Collection of Surgical Pathology Seminars (http://​rosaicollection.org/​)
26

26 Part I:  Basic Concepts

Leukemia.  Leukemia is divided into acute forms, which are more Historically, stage has been the best predictor of prognosis and a
common among children and young adults, and chronic forms, which critical determinant of management, though cancer staging is actually
occur later in life. Approximately 70% of acute lymphoblastic leuke- more of a chronological than biological characteristic (Winer et  al.,
mia cases (ALL) occur among children, and these are further divided 2005). US staging efforts are led by the American Joint Committee
into B-​cell and T-​cell types. The pathogenesis of ALL is broadly con- on Cancer (AJCC, 2011). The AJCC’s TNM (tumor, node, and metas-
ceptualized as resulting from arrested B-​cell or T-​cell development, tasis) staging system is based on the extent of disease in the primary
coupled with uncontrolled cellular proliferation. B-​cell ALL is largely tumor (T), regional lymph nodes (N), and distant or systemic metasta-
subclassified according to detection of specific translocations (Paolini ses (M). In brief, Stage 0 refers to carcinoma in situ (CIS), Stage I inva-
et al., 2011). sive cancers refer to “early” lesions that are confined to the primary
Hematologic malignancies provide well-​characterized examples in organ site of origin, Stage II tumors have spread to the regional lymph
which a chronic, low-​grade cancer evolves into high-​grade, aggressive nodes, whereas “late” Stages III–​IV lesions have greater degrees of
disease. The development of blast crisis (i.e., ALL) among individuals local extension and/​or distant metastases. Missing T, N, or M is coded
with chronic myelogenous leukemia (Chereda and Melo, 2015)  and as “X.”
conversion of low-​grade to high-​grade lymphomas in Richter’s syn-
drome (Jain and O’Brien, 2012)  represent two important examples.
In contrast, the progression from low-​to high-​grade carcinoma is less MULTIPLE APPROACHES FOR CANCER
well described in solid tumors, although some carcinomas with mixed CLASSIFICATION: FROM THE GENERAL POPULATION
patterns of differentiation may represent tumors in transition. In con- TO MOLECULAR CATALOGUING SCHEMES
trast to the relative importance of gene expression patterns in develop-
ing classification of most solid tumors, cytogenetic changes have been Choosing fit-​for-​purpose cancer classification systems involves trade-​
more important in the classification of hematologic cancers, which offs between statistical power, assay costs, logistical constraints related
in some instances has revealed strong evidence of progression of one to availability of tissues, and philosophical uncertainties about “lump-
tumor subtype into another, such as the identification of BCR-​ABL1 ing” versus “splitting.” Classifications optimized to achieve clinical
fusion gene in both chronic myelogenous leukemia and blast crisis research aims, such as determining prognosis or predicting treatment
(Chereda and Melo, 2015). responses, reflect mechanisms that govern tumor behavior, whereas
Acute myeloid leukemia is classified based on morphology, cytoge- the goal of etiological classifications is to group cancers according to
netics, mutations in oncogenes, and clinical features, such as identifi- mechanisms that transform normal tissues into cancer precursors and
cation of leukemogenic exposures or pre-​leukemic syndromes, which cancers (Table 3–​2).
augur a poor prognosis (Hasserjian, 2013). Detection of characteristic Developing useful classifications for high-​grade cancers is com-
molecular changes in cases with prior treatment of myelodsyplastic plex because tumors are genetically unstable and spawn sub-​clones,
syndromes may provide the basis for exploring novel etiologic associ- which can emerge as dominant under the influence of selection pres-
ations or mechanisms associated with progression. Characterization of sures. From an etiologic perspective, downstream tumor progression
molecular abnormalities in non-​neoplastic background bone marrow obscures the relationships between the initial risk-​factor exposures and
elements invites parallels with malignant field effects in epithelium the characteristics of the cancer when first formed. Clinically, high-​
surrounding in situ or invasive carcinomas. grade tumors demonstrate great plasticity in response to therapy, such
Chronic lymphocytic leukemia (CLL) is among the most com- that minor cell subpopulations that were present within the cancer
mon hematologic malignant tumors, accounting for an estimated at diagnosis or cells bearing newly acquired mutations post-​therapy
15,000 incident cases annually in the United States, with a median may emerge as dominant. Cancers arising among carriers of BRCA1/​
age at diagnosis of 70 years (Hallek, 2013). The diagnosis is based 2 mutations offer a notable example; these cancers may arise from
on detection of a monoclonal population of mature B-​cells. The loss of DNA repair functions, and often respond to poly-​(ADP-​ribose)
prognosis of these tumors is quite variable, and discovery of the eti- polymerase (PARP) inhibitors, which further impair these processes,
ologic factors, markers, and mechanisms that characterize aggres- leading to “synthetic lethality.” However, following treatment, some
sive versus indolent types could facilitate the development of early of these tumors re-​express BRCA1/​2, restore DNA repair mechanisms,
detection tests and possibly improved treatments. This mirrors and become resistant (Lord and Ashworth, 2013).
themes in solid tumors, such as prostate cancer, in which identify- Classification systems that broadly lump tumors based on funda-
ing men with indolent cancers who are candidates for conservative mental biological similarities leverage statistical power gained from
treatment (including monitoring) is important. Similar to findings larger sample sizes per category, which improves precision, whereas
for solid tumors, TP53 mutations are associated with genetic insta- finer classifications gain strength from specificity of association. Inter-​
bility and a poor prognosis. The molecular findings in CLL are relationships between approaches for tumor classification are pre-
identical to those of small lymphocytic lymphoma, and the two are sented here, followed by additional comments on strategies that use
distinguished based on whether the predominant manifestation is in morphologic and molecular methods.
the circulation or solid organs (lymph nodes or spleen). Given that
leukemia is by definition a systemic disease, the staging models
used for solid tumors, which mainly reflect anatomical distribution, Age and Morphological and Molecular
do not apply. Among asymptomatic patients with CLL, deferred Classification of Cancer
treatment has been considered for many years, and as such, this
approach may provide a model for limited intervention for other Age is the single most important risk factor for cancer overall and is
indolent cancer types. also reflected in the morphologic and molecular heterogeneity of spe-
cific types of cancer. Our understanding of the biology of aging, cancer
risk, and cancer heterogeneity has evolved since Armitage and Doll
CANCER CLASSIFICATION AND STAGING first drew attention to the exponential growth pattern and linear rise
of most cancer incidence rates on a log rate by log age graphic scale
The WHO International Classification of Diseases for Oncology (ICD-​ (Armitage and Doll, 1954, 1957). This observation provided the theo-
O, 3rd edition) is a dual numerical classification system for coding retical foundation for the concept of long-​term and multistep carcino-
topography (tumor site) and morphology (histopathology). Topography genesis (i.e., tumor initiation, promotion, progression). In this context,
describes the anatomical location of the tumor. Topography codes are “chronological age” has emerged as a surrogate for “biological age,”
prefixed by a letter “C,” followed by two numeric digits for the pri- which affects both the risk of cancer developing and the specific biol-
mary site and a single digit for the subsite. Morphology codes describe ogy of the tumors that emerge.
the histopathological cell type with four digits and a single digit for Each phase of the life course is related to specific tumor types, which
benign or malignant behavior. in turn reflect critical etiological factors that are operative during these
 27

Morphological and Molecular Classification of Human Cancer 27


Table 3–​2. Approaches for Classifying Cancers

Method of Categorization Illustrations Strengths Limitations

Age at diagnosis Pediatric vs. adult; age at onset; Defines important patterns in large Cancers with similar biology may present at
menopausal status population-​based data sets different periods of life
Genetic factors High penetrance genes, familial or Reflects strong causative mechanisms Unknown status for many individuals
sporadic
Environmental Radiation, complications of prior Defines important etiological factor with Modified by other exposures; specialized
exposures treatment, natural or man-​made public health implications application
disasters, carcinogens
Mode of detection Screen detected vs. interval vs. Important for clinical translation Subject to undetected biases
symptomatic
Pathology Histologic type, degree of Routinely available Imperfect reproducibility and limited
differentiation (i.e., grade) mechanistic implications
Molecular pathology DNA, RNA, protein and integrated Provides detailed snapshot of biology Costs and effort, may reflect current biology
classifications rather than mechanisms of development,
intratumoral heterogeneity
Clinical behavior Aggressive vs. indolent Clinically determined Reflects complex interaction between
individual and cancer biology; affected by
method of detection and clinical care
Molecular treatment Somatic mutations or pathways Reflects mechanisms required for tumor Inconstant relationship with etiology and
targets growth with important treatment pathogenesis
implications (i.e., predictive markers)

periods of a person’s lifetime (Figure 3–​7). Absolute incidence rates post-​pubertal development. The temporal association between osteo-
of all pediatric cancers are low in the general population; moreover, sarcoma and the growth spurt is consistent with the higher incidence of
childhood cancers are dominated by leukemias, brain tumors, and sar- these tumors among African American than white children (reflecting
comas (Ward et al., 2014). In contrast, these tumor types are compar- earlier onset of puberty), boys than girls (greater increase in height
atively uncommon among adults relative to the greater incidence of and weight), and its predilection to affect large dog breeds (Ottaviani
carcinomas. Certain tumor types, such as acute lymphoblastic leuke- and Jaffe, 2009).
mia and rare solid tumors composed of undifferentiated cells (often Some cancers with a peak incidence during adolescence have a
referred to as “blastomas”; e.g., neuroblastoma, hepatoblastoma, etc.), bimodal age distribution, which includes a second wave of later onset
are almost exclusively diseases of young children, suggesting the tumors superimposed on an underlying primary non-​neoplastic dis-
likely importance of heritable syndromes, genetic defects, or carcino- ease. For example, the development of osteosarcoma at older ages is
genic exposures in utero as etiologic factors. sometimes related to preexisting Paget’s disease, a benign bone abnor-
Other cancer types that demonstrate a strong predilection for occur- mality, or therapeutic radiation for a preceding cancer (Ottaviani and
ring during late childhood and early adulthood include germ cell Jaffe, 2009). The biology of osteosarcomas and response to chemo-
tumors, specific subtypes of lymphoma, such as Hodgkin lymphoma, therapy differ by age, with best outcomes among youngest patients.
and sarcomas such as osteogenic sarcoma (Parsons et  al., 2008). Similarly, leukemias and lymphomas that may be related to cancer
These relationships may reflect exposures associated with puberty and treatment may differ biologically and clinically from spontaneously

A: Females B: Males
500 1000
Breast Prostate
Lung and Bronchus Lung and Bronchus
Colon and Rectum Colon and Rectum
Rate per 100,000 women/men per year

Melanoma Melanoma of the Skin


400 Cervix 800 Testis

300 600

200 400

100 200

0 0
0 20 40 60 80 100 0 20 40 60 80 100
*******************Age at diagnosis (years) ********************

Figure 3–​7.  Age-​specific incidence patterns for different types of cancers by sex.


28

28 Part I:  Basic Concepts


occurring tumors. Understanding risk factors for treatment-​related and is strongly associated with mutations in BRCA1 (Anderson et al.,
tumors may help define factors that predispose to the development of 2006, 2014). In summary, age categories (or life-​course periods) define
de novo disease (Bueso-​Ramos et al., 2015). groups of certain cancers that are enriched for particular phenotypes,
Among adult onset cancers, age may be interrelated with histo- but these age-​defined groups rarely specify pure categories.
pathologic subtypes and markers suggestive of specific etiologic
mechanisms. The identification of subsets of tumors that occur in
earlier adulthood than expected, particularly when multiple, involv-
Classifying Cancer by Environmental Exposures
ing more than one organ site and/​or occurring among multiple fam-
ily members, has contributed to the identification of a carcinogenic Cancers may be classified according to specific environmental expo-
role of heritable high-​penetrance mutations in BRCA1/​2, mismatch sures, particularly if these exposures result in distinctive tumor biology.
repair genes that cause Lynch syndrome and other cancer syndromes. Examples include radiation-​or chemotherapy-​induced cancers, can-
Further, cancers that are related to inherited mutations demonstrate cers associated with infection (hepatitis B, HPV, Helicobacter pylori),
stereotypical biological and clinical features, and potentially improved and cancers associated with powerful carcinogens (e.g., aflatoxin,
treatment responses to targeted therapies (e.g., PARP inhibitors and smoking) that produce specific molecular signatures (Alexandrov
ovarian/​tubal high-​grade serous carcinomas among BRCA1 carriers and Stratton, 2014). The identification of such connections between
[Bao et al., 2016] and immunologic treatments for colon carcinomas exposures and outcomes can aid in population surveillance and
among Lynch syndrome patients [Le et  al.,  2015]). Identification of cancer control. For example, transplant recipients may develop hema-
associations between specific germline mutations and cancers has led tologic cancers that respond to lowering immunosuppression, but they
to recognition of the importance of somatic alterations in the same develop squamous cell carcinomas of the skin that are more likely
genes or molecular pathways among individuals who are not carriers to recur and metastasize than those occurring among non-​transplant
of germline mutations, especially in the context of genes involved in recipients. Geographically defined exposures (atom bomb, chemical
DNA repair (Jacinto and Esteller, 2007). or nuclear power plants, etc.) may aid in the recognition of exposure-​
Studies of breast cancer age-​specific incidence exemplify the power specific morphologic or molecular patterns. Further, population-​
and limitations of age-​specific models (reviewed in Anderson et  al., restricted exposures may reveal links that transcend individual organ
2014). Breast cancer rates increase rapidly during early reproductive sites and present possibilities to identify characteristic molecular sig-
life, plateau near age 50 years (a surrogate for menopause), and then natures (Hoadley et al., 2014). For example, smokers are at markedly
rise again at a slower pace. Pike and colleagues introduced the concept elevated relative and absolute risk of developing bladder, lung, and
of “tissue aging” or biologic aging, as opposed to chronologic aging, head and neck cancers, and molecular analysis has revealed common
to account for effects of risk factors upon the peculiar shape of the genetic alterations.
breast cancer age-​specific incidence rate curve (Pike et al., 1983). This High-​throughput genomic platforms (methylation, copy number,
model may be conceptualized in three phases:  (1)  puberty to meno- mutation, RNA and protein expression) in the Cancer Genome Atlas
pause, when women are exposed to factors that increase breast cancer Project (TCGA) have helped link molecular changes with the causal
risk and incidence rates rise at an accelerating pace; (2)  menopause importance of smoking. In TCGA, molecular analysis of cancers origi-
transition, when the influence of these factors wanes and the rate of nating at 12 different sites (organs) demonstrated strong relationships
rise of breast cancer incidence slows; and finally (3) menopause, when between molecular alterations and site of origin. However, TCGA also
rates increase (albeit at a slower rate than at earlier ages) in response identified a group of cancers, termed “squamous-​like,” which included
to new exposures and conditions. Differences among women with squamous cell carcinomas from different sites (and divergent squamous
respect to risk factors means that women of the same “chronological differentiation, not matching that of native epithelium), including the
age” may have different “tissue ages” and, consequently, different can- head and neck, lung, and bladder. This group could reflect similar his-
cer risks. However, Pike’s early conceptualizations of aging and can- topathology and/​or common etiologic exposures. Furthermore, the vast
cer incidence did not account for etiologic heterogeneity. Thus, what majority of bronchial squamous cell carcinomas show a squamous-​
appears as a single age-​specific incidence rate pattern may result from like/​smoking-​associated molecular pattern (206/​238, 87%), as do head
the confluence of two or more independent age incidence patterns (see and neck carcinomas (302/​305, 99%), as compared with only 31 of
Chapter 45 on breast cancer). 120 of bladder cancers (30%) (The Cancer Genome Network, 2012;
Lilienfeld and others provided evidence that the overall breast can- Hoadley et al., 2014). While TCGA has emphasized histopathology in
cer incidence rate curve represents the confluence of two distinctive naming cancers with this molecular profile (terming them “squamous-​
incidence rate patterns, prompting a search for molecular pathologic like”), it is striking that these three sites are those with the strongest
correlates of these two types (De Waard et  al., 1964; Lilienfeld and links to smoking, and that the strength of the smoking association is
Johnson, 1955). As the specificity of data available in cancer registries roughly proportional to the percentage of site-​specific cancers that
such as National Cancer Institute’s Surveillance, Epidemiology, and are included in the squamous-​like cluster. Other epidemiological data
End Results (SEER) Program has increased, the potential for complex support the classification of a smoking-​related etiologic grouping that
tumor subtype analyses has grown, substantiating the hypothesis that transcends site of origin (referred to in TCGA as “convergence”).
overall breast cancer incidence patterns are misleading in that they While all major histologic types of lung cancer are strongly smoking
represent the combined effects of two or more very different cancer related, adenocarcinomas occur notably more frequently among a sub-
subtypes (Anderson et al., 2014). set of women who are non-​smokers (Pesch et al., 2012). As such, the
Registry datasets are suited to teasing apart effects of age, but lung adenocarcinoma samples in the TCGA study clustered separately
individual-​level data for patients and tumor pathology are typically from the other smoking-​associated cancers (Cancer Genome Atlas
limited. However, the large sample sizes and lengthy periods of cov- Research, 2012, 2014; Hoadley et al., 2014).
erage enable assessment of calendar-​ period effects (which reflect If smoking-​ related cancers could be identified more accurately
interventions such as screening) and birth-​cohort effects that are more across sites, surveillance for these etiologically related cancers could
closely linked to etiology (risk factors) (Rosenberg and Anderson, be unified and risk stratification might be improved. The potential to
2011). Just as chronological age may not adequately reflect biologi- pool data for smoking-​related cancers defined by molecular signatures
cal age, the implications of attained age may vary by year of birth or might enable identification of risk factors and interactions among them
calendar year of diagnosis. Analyses of registry data have consistently that apply across tumor sites and could identify a target population for
shown that younger-​onset and older-​onset tumors are biologically and preventive interventions. In population surveillance, “lumping” etio-
etiologically distinct in breast cancer and other tumor types. logically related cancers could improve the ability to detect incidence
Nonetheless, given that age at diagnosis is simply a surrogate of trends more quickly and appreciate the public health importance of
complex biology, it is rarely sufficient to define a highly homogeneous such changes at the population level. The utility of this approach is
set of cancers. An exception in breast cancer that proves the rule might exemplified in analysis of second smoking-​associated cancers in which
be medullary breast cancer, which show a unimodal age distribution, lung (stage I), bladder, kidney, and head/​neck cancers were combined
 29

Morphological and Molecular Classification of Human Cancer 29


as a single case definition in an analysis that demonstrated that smok- DCIS, and mass lesions produced by most types of invasive cancers,
ers who survive a first smoking-​related cancer have an increased risk which demonstrate ductal histology. However, invasive lobular carci-
of developing a second smoking-​related tumor (Shiels et al., 2014). nomas, which grow diffusely, may produce fewer radiologic footprints
The value for cancer surveillance, cancer control, and public health and are harder to detect. Further, factors such as higher mammographic
policy is more clearly demonstrated when the attributable fraction of density, which is associated with younger age and lean body mass,
an exposure for a particular tumor type is high, as in smoking-​related may obscure small lesions, increasing the probability of an interval
cancers. For example, an analysis of data for 1976–​2005 demonstrated cancer (Boyd et al., 2007).
that lung cancer incidence rates or deaths among women increased in In colon cancer screening, distal tumors are more readily detectable
18 states, 16 of which were located in the South or Midwest, where the because they arise within exophytic polyps and are more easily identi-
prevalence of smoking is higher and annual percentage rates of dis- fied by digital rectal examination, sigmoidoscopy, or colonoscopy. In
continuation were lower than in other parts of the United States (Jemal contrast, right-​sided cancers, such as those that develop in the more
et  al., 2008). Future analyses might also consider combining cancer expansive cecal lumen, may be less readily detected, and such tumors
subtypes in analyses to assess smoking-​related deaths and evaluating may differ histologically from their left-​sided counterparts (Brenner
lung cancers for smoking-​related molecular signatures. et al., 2014). Similarly, prior to the development of better sampling and
A similar concept has been considered with respect to human papil- HPV DNA testing methods, the movement of the cervical transforma-
lomavirus (HPV)-​related cancers, which account for nearly all ano- tion zone posteriorly with age resulted in a higher frequency of false
genital cancers and a proportion of head and neck cancers, and may negative results at older ages (Sherman et al., 2003).
be preventable through prophylactic HPV vaccination. Given that the Defining tumor types that are less readily detected with current
incidence of HPV-​related cancers is comparatively low in the United screening methods may prompt the development of alternative early
States, pooling might offer an important increase in the statistical detection approaches or may refocus approaches for cancer prevention
power to recognize early trends (Jemal et al., 2013). and control (Miglioretti et al., 2015). Awareness of mode of detection
Another example of etiologically related convergence of multiple may also have implications for external validity in epidemiological
sites is the associations between hormonal exposures and women’s analyses because adjustment for pathology variables may not fully
cancers. Following the report from the Women’s Health Initiative of negate the influence of screening on the representativeness of tumors
a link between use of combined estrogen and progestin menopausal detected. This may have profound importance in international stud-
hormone replacement and increased risk of breast cancer and non-​ ies that include comparisons across extremely variable health systems,
neoplastic diseases, use of these products declined precipitously in the leading to false conclusions that observed data reflect etiology, when
United States, and incidence rate patterns of hormone-​associated can- mode of detection is an important contributor.
cers among postmenopausal women changed rapidly. The associations
were organ and histopathologic subtype specific:  for breast cancers,
declines were found in ER-​positive breast cancers, including specific USING PATHOLOGY TECHNIQUES
histopathologic subtypes, such as lobular and tubular carcinomas TO CLASSIFY CANCERS
(Ravdin et  al., 2007), whereas for ovarian cancers, reductions were
greatest for tumors that are linked to endometriosis, including endo- Histopathology in conjunction with clinical history provided the
metrioid and clear cell carcinomas (Yang et  al., 2013). Since 2002, basis for the early development of tumor classifications. Introduction
endometrial cancer rates have increased among women aged 50–​ of electron microscopy and histochemical and immunohistochemi-
74 years, particularly for subtypes most strongly linked to hormonal cal stains expanded the armamentarium of diagnostic technologies
exposures (Wartko et al., 2013). beyond pure visual assessment.
Potential reasons for the increasing rates of endometrial cancers in Methods to prepare reagent monoclonal antibodies with specificity
the face of declines in rates for cancers arising at other organ sites against protein epitopes that are adequately preserved in routinely pre-
could reflect etiological differences, such as differences in risk asso- pared formalin-​fixed paraffin-​embedded tissue sections transformed
ciations for obesity and/​or the risk modification of obesity by exog- diagnostic pathology from a discipline based on visual interpretation
enous hormone use, although multiple other explanations are possible alone to one that integrates microscopy with cell-​based assessment
(Cote et al., 2015). Obesity increases the risk of both postmenopausal of biomarker expression. Improved antigen retrieval protocols, sig-
breast cancer and endometrial cancer; however, exogenous proges- nal amplification methods, and labeling of antibodies (fluorescence
tins increase risk of the former, while lowering the risk of the latter and chromogenic tags) have increased the sensitivity and specificity
(Anderson et  al., 2003; Brinton and Felix, 2014; Chlebowski and of immunohistochemical assays. Further, in situ hybridization meth-
Anderson, 2014; Collaborative Group on Epidemiological Studies of ods can enable visual assessment of RNA or DNA species that reflect
Ovarian Cancer et al., 2015; Dobbins et al., 2013; Hou et al., 2013). gene amplifications, translocations, or other alterations. Novel meth-
ods allow identification of co-​expression of biomarkers within cells,
interaction of markers, and simultaneous detection of different types
of molecules, such as nucleic acid and protein.
Classifying Cancer by Mode of Detection Despite advances, various tissue assay methods have strengths and
Among organ sites for which there is population-​based cancer screen- limitations that reflect sample preparation, storage, and other factors.
ing, mode of detection has important influences on the morphologi- Analytical specificity and sensitivity of immunohistochemical stains
cal and molecular classification of cancer and cancer precursors that and other approaches that combine morphology with molecular probes
are diagnosed. Screen-​detected cancers are more often slow grow- continue to present challenges, which are often magnified in epidemio-
ing and indolent compared with those that present symptomatically logical studies, which include tissues prepared at different laboratories
(Weiss, 2003). In the extreme, the detection of more indolent cancers using variable methods of fixation and processing.
by screening (length time bias) leads to over-​diagnosis, the discovery Performance of immunohistochemical assays may be impaired by
of tumors that would never have manifested symptomatically. This multiple technical issues related to fixation, processing, staining, and
impact of screening can lead to a number of paradoxical effects (Jatoi interpretation (True, 2014). Guidelines for reporting prognostic mark-
and Anderson, 2005), including increased cancer incidence rates, ers to improve the quality of reported research have been promulgated
impression of longer survival (lead time bias), and apparently better as the “REMARK” criteria, which may be required by journals for
treatment responses. Healthy volunteer effects among screened indi- publication (McShane et al., 2005).
viduals may also skew results toward favorable outcomes and are a Strengths of immunohistochemistry include substantial experi-
concern in clinical trials and cohort studies (selection bias). ence with the method, availability, microscopic assessment of marker
The effect of screening on the tumor types identified varies with the expression by cell type and intracellular compartment, and identifica-
technology employed and patient characteristics. For example, mam- tion of heterogeneity of expression within a sample. Although non-​
mography can identify suspicious calcifications that are common in specificity may be a problem with some assays, pathologists can often
30

30 Part I:  Basic Concepts


identify these problems when patterns of expression are inconsistent
with established knowledge.
Quantification of protein expression by immunohistochemistry
is challenging (Pozner-​Moulis et  al., 2007), and in most diagnostic
applications, cell identification (e.g., distinguishing carcinoma from
lymphoma) or gross scoring of negative or positive is the main goal
(e.g., ER in breast cancer). Immunofluorescence is considered to pro-
vide more accurate quantitation of markers than immunohistochemis-
try. Fluorescence offers advantages for multi-​probe labeling, and this
approach is employed in methods that combine probes to define cell
types and cellular compartments (e.g., membrane, nucleus, cytoplasm)
in which the target probe is measured. For example, by simultane-
ously applying an anti-​keratin reagent antibody to identify epithelial
cells, DAPI (DNA stain) to define the nuclear compartment within
these cells and an anti-​ER antibody, ER expression can be quantified
in breast cancer cells by comparing the nuclear ER and DAPI signals
within keratin-​positive cells (Camp et  al., 2002). However, working
with fluorescent markers has trade-​offs, including reduced ability to
characterize morphology and issues related to preparation of reagent
antibodies and loss of fluorescence over time. Novel methods that Figure  3–​8. Tissue microarray demonstrating immunohistochemistry
combine metal tagged reagent antibodies with multi-​plexed ion beam staining for ER in breast cancer samples.
imaging have also been developed to enable quantification of multiple
markers per cell (Levenson et al., 2015).
In contrast with microscopy-​ based methods, high-​ throughput
sections, such as dipping in paraffin, placing sections in anhydrous or
molecular techniques typically evaluate markers (e.g., DNA, RNA)
anoxic environments, and cold storage, provide unpredictable preser-
derived from mixed cell populations and cannot assess co-​expression
vation of antigenicity and that minimizing the interval from cutting
or topographic localization. The importance of cell-​ type specific-
to staining is the best strategy. Standardized handling and batching
ity depends upon the degree to which tumor cells and surrounding
are critical in studies designed to sensitively measure changes in bio-
benign cells are admixed, whether analyses are discovery-​oriented
marker expression in serial samples, such as neoadjuvant trials.
or hypothesis-​driven, the sensitivity of the assay, and the power of
Improvements in digital scanning technology, web-​based software
analytic methods to separate signals from mixed cell populations. In
for image viewing and scoring, and automated image analysis enhance
tumors that infiltrate into benign tissues (e.g., breast), the challenges
the utility of TMAs in large studies. However, the utility of TMAs
are more pronounced than in circumscribed cancers. If the goal is to
is limited by the same considerations that apply to all immunohisto-
understand tumor biology, signals from the surrounding stroma may
chemical assays: (1) representativeness of tissue samples; (2) represen-
be informative. In contrast, if one wishes to define a cancer-​specific
tativeness of the individual cores (typically 0.6–​1.0 mm); (3) quality
biomarker that may also be found in blood (e.g., circulating tumor
of tissue fixation and preservation of target antigens; (4)  specificity
DNA), identifying markers specific for cancer cells may be of greater
of reagent antibodies; (5) performance of staining protocols; (6) accu-
interest.
racy, and reproducibility of scoring and required effort; and (7) issues
related to data recording, managing, and integrating with other types
of participant data. In research studies, typically only one block per
Tissue Microarrays tumor is available, and some markers may show topographic hetero-
geneity; areas of tumor that are more widely separated may be more
The development of tissue microarray (TMA) technology has poten-
likely to capture variability (Rimm Lab) (Chung et al., 2007). Finally,
tiated opportunities for analysis of immunohistochemistry, in situ
TMAs are best suited to studies of cancer tissues, although TMAs
hybridization, and related methods in large epidemiologic studies
specifically prepared to study cancer precursors, normal structures,
(Kononen et  al., 1998). TMAs are prepared by matching up micro-
stroma surrounding cancer, or immune infiltrates can be prepared
scope slides and blocks to enable localization, removal, and transfer
(Yang et al., 2006).
of tissue cores from targets in many donor blocks into a single recip-
ient block that can be sectioned for immunohistochemistry, thereby
facilitating batch staining and analysis of multiple tumors (Figure Scoring and Interpretation of Immunostains
3–​8). If the sample set is large and/​or multiple immunostains are
planned, this approach can reduce effort and costs. However, multiple Automated scoring of immunohistochemical assays offers potential
cores per tumor are generally prepared and placed in separate blocks advantages, including standardization and quantification. In automated
because any one section typically lacks some cores or contains cores analysis, cancer cells may be identified visually or by computer algo-
with inadequate representation of well-​preserved evaluable cells. Use rithm based on morphometric features and/​or staining for markers.
of TMAs has highlighted critical issues related to sectioning formalin Interactive approaches are less efficient because each sample requires
fixed paraffin-​embedded tissue blocks and loss of immunoreactivity on visual review, whereas in fully automated systems, up-​front effort to
cut sections (Fergenbaum et al., 2004). Loss of immunoreactivity on “tune” the algorithm enables the computer to score all cases without
cut sections is a greater problem than loss of antigenicity in paraffin input, although quality-​control reads of a subset are highly advisable.
blocks (Combs et al., 2016). When blocks are sectioned, the first sec- Analysis of tissues processed in different laboratories that use
tions that are cut are not perfectly parallel to the surface of the block variable tissue preparation, sectioning, and staining protocols poses
and are usually discarded because they do not represent the entire tis- challenges for automated image analyses of immunostains because
sue in the block (referred to as “facing”). Thus, each time a block is algorithms may perform differently in these samples. In multi-​center
sectioned, there is wastage. Accordingly, given that batch sectioning studies, comparing results among laboratories where samples were
conserves tissue, it is advantageous to plan forward by cutting multiple prepared may identify problems. Comparison of scores from replicate
sections at one session and applying validated immunohistochemical cores in tissue microarrays with different results may help identify
protocols as quickly as possible. Loss of antigenicity varies with condi- images requiring visual review. In the end, standardization and optimi-
tions related to tissue fixation and block preparation, the antigen target, zation are essential for generating usable data.
and the immunohistochemical reagent antibody and assay procedures. A comprehensive overview of all possible morphological classifica-
Most pathologists agree that strategies to preserve antigenicity in cut tions of cancer is beyond the scope of this chapter and is accessible
 31

Morphological and Molecular Classification of Human Cancer 31


in existing texts related to oncology and pathology or in site-​specific of atrophic gastritis associated with glandular metaplasia and dyspla-
chapters in this text. Many such writings relate morphological tumor sia (Yakirevich and Resnick, 2013). In contrast, diffusely infiltrating
subtyping to clinical considerations or molecular characteristics, but signet ring cell adenocarcinomas are not associated with metaplas-
concepts to consider in epidemiological research have received less tic changes, and have been linked to CDH1 mutations. Increases in
attention. Examples of general principles are provided in the follow- esophageal adenocarcinoma are occurring worldwide and have been
ing, emphasizing several of the most common types of cancers. attributed to an increased prevalence of Barrett’s mucosa, which is
characterized by replacement of squamous epithelium by glandular
epithelium, typically in the lower esophagus (Runge et al., 2015).
Anatomic Tumor Location
Intra-​individual asymmetries in paired organs are common, as are sys- Macroscopic and Microscopic Growth Patterns
tematic differences in cancer incidence by laterality (Roychoudhuri of Carcinoma
et al., 2006). These differences may reflect biological factors or detec-
tion biases, but convincing explanations are lacking. For example, Carcinomas that form masses that project from a mucosal surface into
breast cancer incidence varies by quadrant of the breast, and while a cavity or lumen of an organ are described as exophytic or polypoid.
this may represent a biological difference in the amount or composi- Whereas some carcinomas grow as polypoid masses, others develop
tion of tissues by anatomic location, this remains unproven. Low-​dose within preexisting benign polyps, which undergo malignant change, as
computed tomography (CT) lung cancer screening may be less sensi- is recognized in the colon and uterus. Polypoid lesions are often asso-
tive in detecting squamous cell carcinomas, which tend to occur in the ciated with cysts or found in viscera with cavities (i.e., uterus) or tubu-
central portion of the lung, than adenocarcinomas, which are relatively lar anatomy (colon). Cysts, which are defined as benign fluid-​filled
more frequent in the better visualized periphery of the lung (National structures lined by epithelium, are extremely common throughout the
Lung Screening Trial Research et al., 2013). Given that smoking risks body and rarely undergo malignant change, whereas “cystic change”
are higher for squamous cell carcinomas than for adenocarcinomas, (often necrotic cavitation, rather than true cyst formation) in malignant
performance of screening may vary among populations with different tumors is common. Gross architectural patterns are important because
percentages of smokers. they affect clinical presentation, detection by screening, and epide-
The frequency of other specific tumor types varies by anatomic loca- miologic associations. In the Prostate, Lung, Colorectal and Ovarian
tion. In the upper esophagus, squamous cell carcinomas predominate, (PLCO) cancer screening trial, cysts detected by transvaginal ultra-
whereas adenocarcinomas predominate in the lower part of the organ, sound were not associated with ovarian cancer risk factors, suggesting
often arising in metaplastic and dysplastic Barrett’s mucosa. Cancer that ovarian cancers do not arise typically from benign cysts, as histor-
incidence is low in the small intestine, particularly for adenocarcino- ically supposed. (Hartge et al., 2000). The lack of association between
mas; however, the anatomy, physiologic function, and luminal con- benign cysts lined by ovarian surface epithelium and carcinoma may
tents of the intestines change radically beyond the junction between account for the poor performance of transvaginal ultrasound as an
the small and large intestine, a frequent site of cancer development in ovarian cancer screening test (Buys et al., 2011; Jacobs et al., 2016).
Western populations. Carcinomas exhibit patterns of differentiation, including squa-
Although most colon carcinomas arise in adenomatous polyps, mous, glandular, neuroendocrine, and mixed; patterns of differ-
tumors that arise in the context of mismatch repair deficiency (i.e., entiation are associated with specific clinical features, and in some
Lynch syndrome) are distinctive, often arising in serrated polyps that instances, with molecular alterations that represent therapeutic targets.
were unrecognized historically (Patai et  al., 2013; Snover, 2010). Histopathologic typing of well-​differentiated carcinomas is reliable,
Microscopically, these cancers are typically high-​grade, mucinous, but distinguishing histopathologic type becomes increasingly difficult
and associated with lymphoid infiltrates. Historically, Lynch syn- as tumors become less differentiated. Special stains may help distin-
drome cancers have been proposed to have a predilection for the right guish squamous cell carcinoma from adenocarcinoma of the lung,
colon, although some part of this excess could reflect delayed identifi- which is important because further testing of adenocarcinomas may
cation and eradication of precancerous polyps. Right-​sided tumors are identify specific mutations that predict responses to targeted therapies.
more frequently exophytic, and therefore tend to remain asymptom- In 2011, a new multidisciplinary classification of lung adenocarci-
atic, whereas tumors arising in the narrower left colon may affect bowl noma was proposed to reflect increasing understanding of the biologi-
habits and produce symptoms. cal complexity of these tumors (Travis et al., 2011).
In pancreatic adenocarcinoma, tumors that arise in the tail of the
organ are nearly always detected when metastatic, whereas a subset Heterogeneity in the Pathology
of adenocarcinomas arising in the head of the pancreas at the junction
of Adenocarcinoma: Etiologic and
of the biliary system and small intestine (ampulla of Vater) may cause
biliary destruction, leading to jaundice and earlier detection (Bond-​ Clinical Associations
Smith et al., 2012). The distinction of different patterns of adenocarcinoma can relate to
etiologic associations and differences in clinical behavior. Selected
examples include the following:
Microscopic Patterns Associated with Carcinoma
• Endometrial carcinoma:  endometrioid adenocarcinomas are
Many carcinomas are proposed to arise in the context of field effects strongly related to hormonal influences and generally have a favor-
that comprise regions of molecular alterations (e.g., genetic, epige- able prognosis, whereas serous carcinomas occur at older ages, are
netic, telomere shortening, and other changes) that provide a back- less strongly related to hormonal factors, and spread aggressively
drop against which additional changes that initiate tumorigenesis are like their ovarian/​tubal homologues (Brinton et al., 2013).
superimposed (Chai and Brown, 2009). Accordingly, recurrence fol- • Gastric carcinoma: gland-​forming adenocarcinomas are associated
lowing tumor excision, even with microscopically clear margins, may with atrophic gastritis and metaplasia, whereas diffusely infiltrat-
result from residual “benign appearing” epithelium that retains pre- ing carcinomas have a different risk-​factor profile and presentation,
cancerous changes. Molecular mapping of cancers and surrounding and are linked to germline or somatic CDH1 mutations (E-​cadherin)
tissues represents a promising approach to explore the evolution of (Yakirevich and Resnick, 2013).
cancer, although the sequence with which molecular alterations occur • Breast carcinoma: lobular carcinomas grow diffusely as single can-
(and therefore causality) cannot be determined from cross-​sectional cer cells or in small linear arrangements, and are more difficult to
samples. detect mammographically than ductal carcinomas (Johnson et  al.,
In the cervix, anus, and bronchi, squamous cell carcinomas arise 2015). Lobular carcinomas are linked to CDH1 mutations, as are
at sites of squamous metaplasia. In the stomach, adenocarcinomas similar appearing diffusely infiltrating gastric carcinomas (“signet
composed of well-​formed glands commonly develop in the setting ring” cell adenocarcinomas); ductal carcinomas form glands or solid
32

32 Part I:  Basic Concepts


masses and have variable risk-​ factor associations, depending in standardization entails costs, logistical concerns, collaborations, and
part on estrogen receptor status (Acs et al., 2001). (Note: given that appropriate internal review board (IRB) approvals.
pathologists may use immunostains for e-​cadherin to diagnose lobu- Agreement between clinical and central research testing results is
lar carcinoma, the morphologic distinctions between lobular versus high for some markers, such as ER and PR in breast cancer (Badve
ductal and e-​cadherin negative or positive are blurred.) et al., 2008); however, for other markers, such as HER-​2, assessing the
• Ovarian/​tubal carcinoma:  these include multiple histologic types, status is more complex (Perez et al., 2014). Knowledge of the repro-
some of which have been linked to endometriosis and tend to pres- ducibility of biomarker studies has potential utility in determining the
ent with stage I  disease (endometrioid and clear cell carcinomas), value of repeating assays centrally to reduce misclassification.
whereas high-​grade serous carcinomas typically present with stage Diagramming the “tissue life cycle” provides a blueprint for
III of IV disease and ascites (Kurman and Shih Ie, 2016). planning pathology components in epidemiologic studies (Figure
• Lung carcinoma: tumors measuring up to 3 cm that coat preexist- 3–​9) (Sherman et  al., 2010). Patient specimens are removed by
ing benign pulmonary architecture (“lepidic growth”) are referred to clinicians to diagnose the cause of a specific symptom, clinical
as “adenocarcinoma in situ” to emphasize the excellent prognosis; finding, radiologic abnormality, or test result, and thus the aims
most adenocarcinomas form masses that destroy underlying benign of diagnostic pathology efforts are narrowly focused. Methods
architecture (Travis et al., 2011). of obtaining samples vary by method used to localize the target,
access to technology, and indication, which affect the quality and
size of the sample.
Neuroendocrine Carcinomas Within studies, the characteristics of participants with available tis-
Carcinomas showing neuroendocrine differentiation occur throughout sue should be compared with those for whom samples are unavailable
the body, although adenomas and well-​differentiated carcinomas that to address representativeness and possible selection bias. For some
produce abundant secretory products occur most frequently within cancer types (i.e., breast), the smallest tumors may be insufficient for
neuroendocrine organs, sometimes as part of a multiple endocrine research, whereas for other cancers (pancreatic or lung) the diagno-
neoplasia syndrome. Neuroendocrine tumors (carcinoids, islet cell sis may be made without surgical removal, limiting the availability
tumors, thyroid medullary carcinomas, etc.) generally appear as uni- of research samples. These biases could affect analyses of risk-​factor
form tumor cells, containing small round nuclei with fine chromatin, associations or prognosis by molecular tumor classification.
arranged in geographic islands, surrounding by a vascular network. A comprehensive review of all considerations is beyond the scope
High-​grade neuroendocrine carcinomas composed of small ovoid of this writing, but a list of candidate questions for consideration is
or elongated cells with hyperchromatic nuclei growing in solid sheets, provided here.
with frequent mitoses and necrosis, may occur throughout the body. • Who collected the sample? Specifically, what was the subspecialty
These “small cell carcinomas” characteristically present with meta- of the clinician who removed the sample? This may be related to the
static disease that responds initially to chemotherapy and radiation, but technique for collection, the clinical setting, geographic setting, and
later recurs. Cervical small cell carcinomas are more strongly associ- participant characteristics.
ated with HPV 18 than squamous cell carcinomas, in which HPV 16 is • What tissue was removed? Was the tissue removed for biopsy diag-
numerically predominant (Atienza-​Amores et al., 2014). nosis, or was the whole organ removed for known cancer?
• Why was the tissue removed? Did the patient have symptoms or
a positive screening test, reflected in an abnormal imaging study,
Tissue Morphology as Putative biochemical or molecular test? Was the surgery performed for risk
Intermediate Endpoints reduction in a high-​risk individual?
• Where was the tissue removed? Tissues removed outside the hospi-
Biomarkers may be useful as intermediate or surrogate endpoints. tal are often sent to the laboratory fixed, whereas tissues removed
Establishing that a biomarker is on the causal pathway is powerful in the hospital may arrive fixed or fresh. Does the setting represent
because preventing the intermediate may portend a benefit in cancer a general practice setting, high-​risk clinic, or referral for a second
prevention. Intermediates also develop more quickly than cancer, and opinion? In international studies, it is important to understand how
therefore can provide more rapid approaches for demonstrating effi- clinical practices vary by country and by center within country,
cacy in prevention studies. However, intermediates often fall short which can profoundly influence quantity, type, and quality of tissues
of expectations because the associations with cancer are not always available for research.
strong, and blocking one carcinogenic pathway may not prevent carci-
nogenesis by alternative mechanisms (Schatzkin and Gail, 2002).
In some carcinomas, cancer precursors may serve as useful inter-
mediate markers, although not every precursor lesion will progress
to invasion, and some cancers may arise from multiple different Tissue
precursors. Dysplastic changes, such as may occur in inflammatory Sampling Fixation
bowel disease, may also serve as intermediates and help guide sur-
veillance for colon cancer. At anogenital sites, detection of persistent
type-​specific HPV DNA infection may provide a useful intermediate
biomarker, and serve as an endpoint in screening and prevention stud- Subject Storage with
ies (Lowy et al., 2015). Although not every persistent infection pro- Optimal Processing
gresses, it is commonly held that all HPV-​related cancers are linked to Risk Preservation
a persistent infection. Prognosis

The Tissue Life Cycle: Practical Consideration Database


in Planning Analysis of Tissue Biomarkers Analyses Scoring Assay
in Epidemiology
Increasingly, epidemiologic studies involve multiple centers; conse- Figure 3–​9. Tissue life cycle. Permission and citation may be needed.
quently, performing central pathology review and molecular analy- Figure 1 adapted from Sherman ME, Howatt W, Blows FM, Pharoah P,
ses in a single laboratory using common validated methods may Hewitt SM, Garcia-​Closas M. Molecular pathology in epidemiologic stud-
improve the consistency of cancer classification. Securing improved ies: a primer on key considerations. Cancer Epidemiol Biomarkers Prev.
2010 Apr;19(4):966–​972. doi:10.1158/​1055-​9965.EPI-​10-​0056.
 3

Morphological and Molecular Classification of Human Cancer 33


• When was the tissue removed? Molecular preservation of frozen tis- that cancer subtypes defined by these methods extended classifications
sue is generally optimal, but fixed tissues (if well prepared) may be based on robust clinical markers by providing additional prognostic
useful for many assays decades later. Changes in clinical practice, information. The Human Genome Project accelerated progress by pro-
environmental/​lifestyle factors, and pathology practices over time viding the framework for designing probes to evaluate the transcrip-
may affect assay feasibility and results. tome (Lander et al., 2001).
• How was the tissue removed? Was the tissue removed as a small In breast cancer, DNA microarrays were used to identify an “intrin-
biopsy that was targeted with direct visualization, or was it image-​ sic gene list.” The early arrays were performed on macrodissected
guided? If surgically removed, was an open approach used with samples of tumor cells, stroma, immune cells, and adipose tissue.
intact removal, or was a minimally invasive procedure used that led The intrinsic gene list identified genes that were inherent to the tumor
to removal of tissue in fragments without orientation? because they showed similar expression between multiple samples of
the same tumor or tumor/​metastasis pairs. This approach identified five
Larger tissue specimens received in the diagnostic pathology labora- robust breast cancer subtypes, which were repeatedly identified, sug-
tory are sampled for histopathologic examination based on the clinical gesting that these categories reflected fundamental divisions in breast
context and gross appearance. Selected tissue fragments are used to cre- cancer (Perou et al., 2000). This classification distinguished a category
ate formalin fixed paraffin-​embedded tissue blocks that are sectioned with a good prognosis (luminal A) from classes with poorer outcomes,
(approximately 5 um), mounted on glass slides, stained, and reviewed designated as luminal B, HER2, and basal-​like (Sorlie et  al., 2001).
microscopically. Each block can yield dozens of tissue sections, and the This taxonomy has been reproduced in multiple populations and using
uncut material in the block is general archived in pathology laboratories different RNA-​based technologies, which has transformed thinking
for variable numbers of years, although a minimum of 10 years is gen- about breast biology and treatment (Dunbier et  al., 2011; Hu et  al.,
erally required for laboratory accreditation in the United States. 2006; Huo et  al., 2009; Rouzier et  al., 2005; Sorlie et  al., 2001; Yu
Pathologists review sections microscopically and formulate diag- et al., 2004).
nostic reports that provide information required for management Improved technology has expanded expression profiling to include
and assessing prognosis. Pathology reports generally include tumor more genes and non-​coding RNAs, and has improved production of
size, histopathologic subtype, grade, and clinical biomarker test- more specific stable probe sequences with quality control. RNA label-
ing. Abstracting pathology reports poses challenges related to non-​ ing and hybridization are relatively easy and can be performed in
standardized protocols, idiosyncratic differences among specimens, 2  days. Data analysis can be performed on desktop computers with
and variation in formatting and systems of alphanumeric coding for widely available packages, including those freely available from
specimens and individual blocks. Differences in diagnostic terminol- Bioconductor (Gentleman et al., 2004).
ogy may necessitate translation to common nomenclature to permit Microarrays are useful for measuring expression of many genes
aggregation of data across study centers. Over the years, standardiza- concurrently; however, the outputs are relative gene expression val-
tion of pathology reports has increased, which often includes synoptic ues that are based on hybridization and fluorescent values, which are
reporting that systematically includes all major features related to a semi-​quantitative. These methods also depend upon amplification and
particular cancer type. synthesis of cDNAs, which can be challenging for fragmented RNAs,
Changes in the specificity of diagnoses reported in pathology prac- such as those extracted from formalin fixed paraffin-​embedded tissues.
tice poses challenges for assessing temporal trends in cancer incidence Further, these methods can only detect probes represented on arrays,
rates or analytic studies of cancer incidence in long-​term cohort stud- limiting discovery. New technologies, such as RNA-​counting meth-
ies using cancer registry data. Nomenclature may also evolve to reflect ods (e.g., Nanostring; Geiss et al., 2008), do not rely on amplifying
revised understanding of the clinical behavior of pathologic entities, as the RNA and can count the number of RNA molecules directly for
was found for the encapsulated follicular variant of papillary thyroid up to 800 genes using a barcoded system. These methods can detect
carcinoma. Based on a lack of adverse outcomes for individuals with fragmented RNAs, and therefore can be applied to both fresh frozen
these cancers, an expert panel recommended renaming these lesions as and formalin fixed paraffin-​embedded tissues and samples with low
“noninvasive follicular thyroid neoplasm with papillary-​like nuclear (cellularity) yields.
features,” thereby revoking the cancer status of such tumors (Nikiforov In the late 2000s, use of RNA sequencing further increased analyti-
et al., 2016). Of note, despite this designation, many of these tumors cal capabilities, including more sensitivity for detecting lower levels
show clonal mutations in genes linked to cancer, supporting the view of transcripts. Further, sequencing can detect uncharacterized RNA
that molecular testing, like morphologic classification, may have limi- sequences that can be mapped to the genome to identify novel genes,
tations for predicting tumor biology. assess alternative splicing, find somatic gene fusions, and measure
expression levels of mutations.

EVOLUTION OF HIGH THROUGHPUT MOLECULAR


METHODS FOR CLASSIFYING CANCER Next-​Generation Sequencing
Molecular testing for single or limited panels of prognostic and predic- Next-​generation sequencing of somatic tissues offers advantages over
tive markers to stratify patients with respect to prognosis and to iden- earlier generation Sanger Sequencing, including greater through-
tify optimal treatment is an established and growing part of clinical put, partly enabled by parallel read detection, increased capacity to
medicine. With technological advances in array technology and, more sequence bases, and reduced costs per base. Using newer methods
recently, next-​generation sequencing, the biomarker field has moved has enabled detection of new mutations of clinical consequence. For
into more comprehensive molecular characterization of tumor samples. example, Sanger sequencing of mutations in 601 selected genes in 91
As costs for sequencing decline, this trend is anticipated to continue, glioblastoma multiforme tissues and matched blood samples identi-
although bioinformatic challenges remain limiting. Table 3–​3 reviews fied genes that were recurrently mutated in glioblastoma multiforme
some of the current technologies for analyzing RNA, DNA, DNA (Cancer Genome Atlas Research, 2008). Using next-​generation meth-
methylation, and protein, highlighting strengths and weaknesses. ods to sequence the genome of only 22 glioblastoma multiforme,
The move toward classifications of cancer based on comprehen- another group identified IDH1 mutations, which emerged as impor-
sive molecular characterization began with the development of DNA tant in understanding the pathogenesis of glioblastoma multiforme,
microarrays (previously referred to as cDNA microarrays) for measur- with potential for predicting prognosis and improving treatment
ing gene expression in the late 1990s to early 2000s. Early microarray (Parsons et al., 2008). IDH1 and IDH2 mutations are found frequently
studies classified cancer samples into “subtypes” sharing similar gene in lower grade gliomas and are associated with better survival (Brat
expression, as determined by agnostic statistical methods, such as et al., 2015).
hierarchical clustering (Alizadeh et al., 2000; Perou et al., 2000; Sorlie Next-​generation DNA sequencing methods include whole genome
et al., 2001). Enthusiasm for this research was driven by recognition sequencing (WGS), whole exome sequencing (WES), and targeted
34

34 Part I:  Basic Concepts


Table 3–​3. Strengths and Weaknesses of Selected Molecular Methods for Classifying Cancers

Analyte Technology Strengths Limitations

RNA RT-​PCR Sensitive quantitative Low throughput; requires gene sequence


Nanostring Good dynamic range; suitable for FFPE Medium throughput; limited number of targets
Minimal bioinformatics
Microarrays High throughput; low cost Low dynamic range; not suitable for FFPE
Quick Genes detected limited to specificity and selection
Minimal bioinformatics of probes
RNAseq (mRNA, miRNA, High throughput; large dynamic range Expensive; time intensive
total RNA) Suitable for FFPE Requires intensive bioinformatics support
DNA Whole genome sequencing Mutations, structural variations, copy number Expensive; time intensive
(WGS) variation, for both coding and non-​coding regions Requires intensive bioinformatics support
More uniform coverage
Whole exome sequencing Mutations (SNVs, indels) for coding regions; less Limited to coding regions in capture set; variability
(WES) expensive than WGS in capture efficiency
Changes in capture kits over time
Variable detection of structural or copy number
variations
Targeted sequencing Less expensive; mutations Limited to regions captured by probes
High coverage of selected regions
Copy number arrays Quick and inexpensive Not full genome; resolution is about a marker every
Copy number and LOH 700bp
SNP information
DNA methylation Arrays High throughput; less bioinformatics demand Inexpensive; analysis limited to selected probes
(currently up to 450K)
BiSulfite sequencing Whole methylome; high sensitivity Expensive; low throughput
Intensive bioinformatics analysis
Protein Western blots Can detect bands based on molecular weight Low throughput; dependent on antibodies
Reverse phase protein High throughput samples ~150 antibodies Antibodies must be specific; proteome incompletely
arrays (RPPA) covered
Mass spectrometry Large number of proteins quantified; good for Expensive; limited sample throughput
known or novel proteins De novo methods not always accurate
Not all peptides visible
Intensive bioinformatics

sequencing (TS), which differ predominately in the amount of the lower throughput and at a significantly higher price. Molecular clas-
genome that is sequenced. Methodologic differences related to nucleic sification using DNA methylation has played a major role in sev-
acid preparation and amplification prior to sequencing may affect results eral tumor types. A CpG island methylator phenotype (CIMP) was
between studies, posing challenges for data pooling. WGS aims to first characterized in colorectal cancers (Toyota et  al., 1999)  and
sequence the entire genome and can find variants in both coding and subsequently in other tumor types. TCGA established CIMP in gli-
non-​coding regions, detect structural variations, and determine copy oma, which was associated with younger age at diagnosis, proneu-
number states. Typically, coverage is around 30X, although the purity ral subtype, IDH1 mutations, and improved outcomes (Noushmehr
of the sample might require additional sequencing to sensitively detect et al., 2010).
alterations, and deeper reads may also be required to identify sub-
clones. Applications of WGS have been constrained by cost, but these
have dropped dramatically from ~$10  million for a whole genome Proteomics
in 2007 to $1000–​$4000 in 2015. WES sequences the exomes (cod-
ing regions), which comprise about 1% of the human genome. WES To date, most molecular classifications of carcinomas have been
reduces the amount of genomic regions sequenced, and therefore cov- developed using RNA analysis, but proteomic data are important in
erage is typically higher, approximately 30X–​100X. Finally, TS usu- targeted drug development. Immunohistochemistry and Western blots
ally includes smaller gene panels for focused analyses, and coverage is have practical limitations for assessing expression of many different
usually 500X–​1000X. proteins in large studies. Reverse phase protein arrays (RPPA) allow
DNA sequencing methods have also been used for classification. detection of ~200–​300 proteins and phosphoproteins in up to 1000 dif-
TCGA analyses of melanoma resulted in a proposed classification ferent samples with higher sensitivity than Western blots (Tibes et al.,
based on mutations—​BRAF, RAS, NF1, and Triple-​WT (The Cancer 2006; Zhang et al., 2009). Mass spectrometry methods can detect more
Genome Network, 2015). DNA sequencing can yield mutational spec- proteins, but have limited sample throughput. Protein-​based methods
tra that may ultimately be linked with etiological exposures that are are sensitive, but can be problematic for large studies because stored
linked to a specific somatic mutation, such as UV induced, APOBEC, samples are susceptible to protein degradation.
POLE, age, and smoking signatures (Alexandrov et  al., 2013; Nik-​
Zainal et al., 2012).
RESEARCH APPLICATIONS OF BIOMARKERS
While most current research has focused on translating genomic
Analysis of DNA Methylation discoveries to clinical applications (e.g., prognosis and treatment
Epigenetic alterations, such as DNA methylation, can play an response), biomarkers may be measured in cancer tissues to address
important role in regulating gene expression. Two platforms used different research aims. Other purposes include early detection,
currently to assess DNA methylation include arrays, which evalu- screening, and etiology; general issues are covered in Chapter 6, but
ate up to 450,000 probes, and bisulfite sequencing, which can cover we highlight a few examples here, in particular those related to diag-
most of the CpG and methylation sites in the genome, but with nosis and classification.
 35

Morphological and Molecular Classification of Human Cancer 35


Multifunctional Biomarkers: HPV DNA and ER variation, methylation, or even amplification or overexpression of
other TP53-​regulatory genes, is a cardinal feature of many cancers.
Biomarkers may serve singular functions in early detection, prognosis, A  TP53 signature based on gene expression that reflects the down-
or prediction, but many useful biomarkers function in multiple capaci- stream effects of alteration in TP53 function, rather than the underly-
ties, such as HPV 16 DNA in the cervix and ER in breast cancer. The ing molecular mechanisms, has been developed as a useful classifier
cervix is susceptible to infection with over 13 carcinogenic HPV types, (Troester et al., 2006).
the vast majority of which occur among young women and become
undetectable within 2  years (Moscicki et  al., 2012). However, HPV
16 genotypes are more virulent, with a greater tendency to progress High-​Throughput Genomic Classification
to cervical cancer than other genotypes, accounting for approximately
50% of these cancers overall. As a result of its importance as an eti- The Cancer Genome Atlas (TCGA), a National Cancer Institute–​
ologic agent and prognostic marker, clinical guidelines recommend sponsored project to develop molecular taxonomies of cancers arising
immediate colposcopy for women who test positive for HPV 16 (or at multiple organ sites, provides the gold standard for high-​dimensional
18) DNA, whereas for all other carcinogenic HPV types, triage with genomic cancer classification based on multi-​ platform molecular
cytology is acceptable (Huh et al., 2015). profiling data (see Chapter  4). Further, TCGA has provided unique
ER status of breast cancers delineates etiologically distinct tumor opportunities to assess molecular genomic and epigenomic changes
subtypes, as defined by risk-​factor associations, incidence rate pat- across tumors of different organ sites, demonstrating unexpected and
terns, and molecular profiles (Althuis et  al., 2004; Anderson et  al., potentially important biological relationships among cancers through-
2014; Perou et al., 2000). Positive ER status predicts improved out- out the body. For example, data suggest common genomic alterations
comes among postmenopausal women (especially in early follow-​up); for serous carcinomas of the pelvis and basal carcinomas of the breast,
ER therefore represents a single-​gene molecular marker that is a useful including a high prevalence of mutations in TP53 and a similar pattern
classifier across the spectrum from etiology and prediction (response of copy number alterations (The Cancer Genome Network, 2012).
to treatment) to prognosis (survival) (Henderson and Patek, 1998). ER TCGA has resulted in landmark achievements in molecular clas-
status also demonstrates the blurring of prognosis and predictive mark- sifications; nonetheless, the results of the project are not optimal for
ers. Prior to the introduction of tamoxifen, a highly effect adjuvant addressing all types of scientific questions. For example, TCGA was
therapy for ER-​positive breast cancer, the hazard rates of both ER-​ not designed to investigate etiological heterogeneity, prognosis, or pre-
positive and ER-​negative cancers were more similar, with a sharp peak diction of treatment. The study does not represent a population-​based
in the first years following diagnosis, followed by a marked decline. sample and tends to over-​represent larger and more aggressive tumors.
Following introduction of adjuvant tamoxifen therapy, the hazard rate Moreover, TCGA includes only limited risk factor information, and
of ER-​positive breast cancers flattened, but remained above baseline although the data sets have been used to analyze outcomes, follow-​up
for many years (Berry et al., 2006). is not mature and available treatment data are minimal. Importantly,
the study was designed to collect optimally preserved tissue samples,
and therefore, the tumors may not be representative of those occurring
in the general population, at least with regard to frequency of occur-
Single Biomarkers rence. Therefore, TCGA provides an important biomarker discovery
Clinical testing for many single biomarkers is important for assessing engine, but molecular epidemiology and clinical research are pivotal
prognosis and determining management, as outlined throughout this for applying biomarkers to answer important translational research
chapter; however, recognition that none of these biomarkers performs questions.
flawlessly has prompted searches for expanded panels to improve Integrated multi-​platform molecular analysis, while providing the
performance. For example, KRAS, BRAF, and NRAS are important most detailed molecular classifications, is expensive and unaffordable
prognostic indicators in colon cancer, and may predict responses to in most epidemiological studies. However, anticipated reductions in
EGFR-​targeted therapies; however, predictions are limited by molecu- sequencing costs may increase the feasibility of these approaches in
lar redundancy in the EGFR pathway (Tran et  al., 2015). In breast the future. Further, molecular panels based on immunohistochemistry,
cancer, ER-​positive status predicts responses to endocrine therapies, DNA sequencing, or mRNA expression that approximate classifica-
but treatments may fail because of ER mutations, alterations in other tions based on comprehensive molecular analysis can be applied in
growth pathways, or other reasons (Zhao and Ramaswamy, 2014). many epidemiologic projects, particularly as methods for analyzing
Ovarian/​tubal serous carcinomas with BRCA1/​2 dysfunction resulting paraffin embedded tissues improve (Tyekucheva et al., 2015). TCGA
from germline mutations or somatic alterations respond favorably to is currently leveraging methods, such as elastic nets and LASSO, to
platinum chemotherapy or PARP inhibitors, but such tumors are rarely identify the minimal number of features needed to distinguish tumor
cured (Bao et al., 2016). classes identified with the full data. These types of studies are a neces-
Development of more accurate multi-​gene molecular classifiers has sary next step in translating the TCGA findings to large population-​
been enabled by the availability of high-​throughput methods for geno- based studies. Given power requirements that result in part from
mic characterization. While some biomarker panels are standardized multiple comparisons, future research in this area will likely require
and commercialized, other tests performed for research or clinical consortia of multiple studies. These approaches will necessitate either
management are considered “laboratory-​developed tests,” which are standardization of methodology or demonstration of comparability in
intended for “in-​house” use only. As precision medicine advances, the classification across genomic platforms.
need for such tests will likely increase, raising concerns about vali-
dation (accuracy, analytical sensitivity and specificity, and reproduc-
ibility), clinical predictive performance, and quality assurance, with INTEGRATION OF CLASSIFICATION SCHEMA
implications for regulation and insurance coverage.
Biomarker panels that assess multiple carcinogenic pathways may Researchers addressing the challenge of developing useful cancer clas-
provide improved assessment of critical mechanisms (e.g., prolifera- sifications rely on different types of data, ranging from population-​
tion, apoptosis, etc.) by assessing processes with several related mark- based incidence rates through intensive molecular profiling. Each
ers (“metagenes”). Several multi-​gene biomarker panels for breast approach offers strengths and weaknesses, but none is likely to fully
cancer have been approved as clinical tests, and it may be possible capture the heterogeneity of cancer from every perspective: etiological,
to repurpose the results to study etiological questions (Gyorffy et al., biological, and clinical. Consequently, integrating information from
2015). Developing gene signatures that reflect function of pathways multiple disciplines may improve the utility of cancer classifications.
may represent powerful approaches for understanding carcinogene- Classification of breast cancer provides an illustrative example of
sis and improving prevention and treatment strategies. For example, where different approaches reinforce one another to provide a coherent
loss of TP53 functions, as a consequence of mutation, copy number picture of carcinogenesis. In breast cancer, from a clinical perspective,
36

36 Part I:  Basic Concepts


there are three primary types: hormone receptor positive, HER2 over- nuclei with a particular immunohistochemical stain and measure stain
expressing, and a third group encompassing all other tumors. Etiologic intensity in each nucleus, but cannot distinguish invasive cancer cells
hypotheses based on descriptive population-​based statistics suggest from CIS and benign cells without applying sophisticated algorithms.
that breast cancer may represent two main types: an early-​onset can- Interactive image analysis can sometimes be used to harness the visual
cer, enriched for ER-​negative cancers with a peak incidence at age identification skills of pathologists and the ability of computer pro-
50 years, and a larger group of late-​onset tumors, which is mainly ER-​ grams to count and measure morphological features. For some pur-
positive and peaks at age 70 years (Anderson et al., 2014). In analytical poses, this synergistic power may enable improved morphologic
epidemiology, cancers are often split based on age and menopausal classification. Further, results of image analysis may lead to improve-
status, relationship to estrogenic risk factors, ER status, and associa- ments in diagnostic criteria used by pathologists.
tion with mutations in high-​penetrance genes (e.g., BRCA1/​2) (Althuis An important application of morphologic classification is that it can
et  al., 2004; Anderson et  al., 2014; The Cancer Genome Network, be merged with molecular testing to assess relationships of cells within
2012; Yang et  al., 2011). Basic biologists that embrace a stem cell intact tissues and co-​expression of proteins within individual cells. To
model of carcinogenesis might envision two main types:  luminal or achieve these goals, several methods are being developed to enable
basal, reflecting proposed progenitor cells of origin. labeling with dozens of probes on a single section. If combined with
Following the identification of the breast cancer intrinsic subtypes quantification of signals from target probes, this approach may iden-
based on expression profiling, TCGA extended the characterization of tify various subclones within cancers and map these topographically to
breast cancer using multiple genomic platforms (mRNA and miRNA assess intratumoral heterogeneity, and potentially its regional drivers.
gene expression, DNA sequencing, DNA methylation, copy number Finally, in large studies of cancer that are assessing robust markers,
variation) (The Cancer Genome Network, 2012), which affirmed the automated analysis may provide standardized reads more quickly than
validity of the original classification. Further, the data demonstrated might be achieved by visual interpretation. However, the advantages of
that the three most prevalent somatic mutations in breast cancer are this approach are tempered by the upfront effort to develop and apply
TP53 (37%), PIK3CA (36%), and GATA3 (11%), and that the preva- algorithms and the need to perform quality assurance.
lence of these mutations differ by subtype. Almost 80% of basal carci-
nomas showed a mutation or loss of TP53, and those that did not share
these defects showed defects in other TP53 pathway members, such
Molecular Classification
as the TP53 negative-​regulator MDM2. Compared with basal carcino-
mas, a lower percentage of luminal breast cancers demonstrate TP53 Many epidemiologic studies attempt to link known cancer risk fac-
defects, whereas the percentage of PIK3CA and GATA3 mutations was tors to cancers of particular organ systems, stratified by molecular
much higher (The Cancer Genome Network, 2012). Approximately tumor subtypes, as defined using marker panels. This approach can
35 genes were mutated with appreciable frequency in breast cancers. be expanded to evaluating etiologic heterogeneity based on subtypes
However, little is known about associations between these mutations, defined by molecular profiling, and further, to relating risk factors or
risk factors, and prognosis. Thus, additional analyses encompassing a exposures to expression of individual genes, gene mutations, cytoge-
multidisciplinary perspective are needed. netic changes, or molecular signatures. Relating risk factors to causal
Subsequent studies have shown that genomic data and epidemio- mechanisms and strongly associated biomarkers may offer opportuni-
logic data can be integrated, yielding improved inferences. In a TCGA ties for early detection and prevention.
report that evaluated multiple genomic platforms from 12 different Assessing risk factors in relation to increasingly complex tumor
cancers, the differences in the genetic characteristics of luminal versus classifications poses challenges for achieving sufficient statistical
basal breast cancers were on the order of differences between sites power to distinguish true associations from chance findings found
(e.g., bladder vs. kidney) and were greater than differences between among multiple comparisons. Addressing these issues will require
some sites (e.g., colon and rectum) (Hoadley et al., 2014). This under- large data sets compiled in collaborative consortia or data pooling, an
scores, from a genomic perspective, that the two breast cancer sub- approach that will pose complexities related to differences in sample
types are best considered as distinct diseases. Further, basal breast preparation, profiling methodologies, logistics, and cost. Ideally, once
cancers and high-​grade tubal/​ovarian serous carcinomas shared an biomarker associations are identified, they should be independently
unexpectedly high number of molecular similarities. replicated in different studies to support their validity, and then evalu-
ated in diverse populations to assess generalizability. Further, for
many cancers, subtypes of particular etiologic interest may comprise
CHALLENGES AND FUTURE DIRECTIONS a minority of tumors (e.g., basal breast cancers, endometrial serous
carcinomas, etc.).
Attempting to relate risk factors to specific changes at the DNA or
Classical Pathology Methods
RNA level may prove challenging because numerous stochastic events
Histopathology was the mainstay of cancer classification for many related to genetic instability, an intrinsic property of cancer, may prove
years. Ongoing research seeks to overcome the inherent subjectivity, difficult to distinguish from changes related to cancer etiology or
inconsistency, and variability of visual assessment, while preserving pathogenesis. Complex biostatistical analysis of multiple samples per
the rich data that morphology provides by reflecting the composite tumor may be required to distinguish driver from passenger molecular
dynamic effects of complex biology over time. When pathologists events in established cancers, but such samples are rarely available and
score histopathological features, they are relying on a visual, men- the work is costly. Analysis of cancer precursors could reduce difficul-
tal database of thousands of images and applying pattern recognition ties related to distinguishing molecular drivers from passengers related
skills and diagnostic criteria. This process is at best semi-​quantitative, to genetic instability (Campbell et al., 2016), but once tissues are sam-
and often merely descriptive. Differences in pathologists’ biases and pled, the natural history of such lesions may be unknown with regard
experiences may contribute to subtle, and sometimes more than subtle, to progression to invasive cancer, regression, or dormancy. Further,
differences in interpretation. Further, these opinions may be reinforced precursor lesions are typically asymptomatic, and the types of such
among pathologists who work together and strive for reproducibility. lesions may vary with screening methods.
A number of objective, automated algorithms have been developed to Another possible approach to improve the distinction of molecular
address these concerns. alterations causally related to carcinogenesis is to study field effects
Automated algorithms and computer-​based image analysis offer surrounding cancers, cancer precursors, or normal tissues (Chai and
inherent strengths, including the ability to count events and measure Brown, 2009). Further, integrating such findings in the context of
features, neither of which is readily achieved visually. However, algo- epidemiological studies may be useful for understanding the “etio-
rithms are often challenged to make distinctions that are readily made logical field effect,” which links molecular alterations in tissues to
by pathologists, such as identifying artifacts and distinguishing cell potential carcinogenic exposures (Lochhead et  al., 2015). Although
types. Simple computer algorithms can count the number of positive such cross-​sectional studies cannot distinguish whether a specific
 37

Morphological and Molecular Classification of Human Cancer 37


molecular alteration preceded or followed cancer development, it of cancer progression is highest among lesions with the highest degree
may aid the identification of biomarkers that could be tested in other of regional genetic heterogeneity (Merlo et al., 2010).
sample sets without cancer (e.g., tissues removed for diagnosis) and Among heterogeneous tumors, there are at least two types of hetero-
known outcomes in follow-​up. The development of a pre-​TCGA geneity, which may have different consequences. Heterogeneity can
project may enable this research in the future, although technical be stochastic and dispersed, reflecting genetic instability across the
challenges remain, related to molecular studies of small samples, tumor. In a dispersed heterogeneous tumor, various clones show fitness
the likely need to work with fixed tissues, and difficulties related to and/​or become extinct via a linear, randomly evolving (stochastic),
knowing the natural history of lesions once removed (Campbell et al., time-​variant process. Heterogeneity can also be segregated or hierar-
2016). Nonetheless, such research may distinguish etiological factors chical, possibly defined by key geographic landmarks or microenvi-
and mechanisms involved in initiation from those that may be related ronmental zones, with tumor cells found in one area showing distinct
to later phases of carcinogenesis, such as clonal expansion, invasion, evolutionary pressures and therefore distinct genetic characteristics.
and metastasis. The latter type of heterogeneity was termed “allopatric” in reference
Cancer risk factors that are genetic or related to development may to ecologic speciation that is branched due to geographic segregation
operate by shaping normal tissues to make them more susceptible to (Burrell et al., 2013).
cancer initiation. In the breast, data suggest that differences in physi- Segregated heterogeneity is particularly problematic for molec-
ological exposures, ranging from attained age or age at menarche and ular diagnostics. Sampling of one “species” or zone of a hetero-
menopausal status to recent pregnancy and childbirth, may alter the geneous tumor would not represent the tumor’s overall biological
microanatomy, molecular characteristics, and function of normal cells, potential and could lead to treatments that lack the breadth of
resulting in a spectrum of “molecular histology” (Figueroa et al., 2014; effects required to inhibit all driver molecular pathways. Thus,
Sherman et al., 2012). tumors could be of at least three types: (1) homogeneous; (2) het-
Changes in molecular histology may represent prevention targets erogeneous, dispersed; and (3) heterogeneous, segregated. Despite
in healthy populations. Well-​developed surrogate endpoint biomark- the potential for prognostic and/​or treatment decision differences
ers have been identified for some cancers (Schatzkin and Gail, 2002), between the first two categories, both homogeneous and heteroge-
but for most cancers, such endpoints are lacking. Identification of neous, dispersed tumors could reliably be characterized by assaying
surrogate endpoints could facilitate evaluation of novel prevention samples from a single location. Recognition of dispersed heteroge-
approaches in clinical trials and allow monitoring of the effectiveness neity may have important prognostic and predictive implications,
of established approaches in clinical practice. but at least sampling is straightforward. Segregated molecular
heterogeneity would require multiple samplings to be adequately
described and targeted.
CHALLENGES AND FUTURE DIRECTIONS: Another area of contemporary interest is in further characterizing
CLINICAL RESEARCH tumors according to the characteristics of their microenvironments.
This can mean characterizing stromal or immune cell infiltrations
As our understanding of the molecular biology of cancer advances, around a tumor, but can also encompass better understanding of the
important topics with translational implications are emerging. Recent systemic milieu of the person with disease. For example, obesity-​and
research has emphasized the key role of intratumoral (within tumor) diabetes-​associated cancers may have different metabolic or tissue
spatial or temporal heterogeneity. A central translational goal of can- characteristics that affect their growth (Gucalp et al., 2016). Similarly,
cer classification is optimized, targeted treatments, which requires recent pregnancy may affect the milieu in which a reproductive or
accurate clinical subtyping. Clinical subtyping has historically uti- breast cancer arises, resulting in distinct tumor characteristics, modu-
lized single tissue samples collected at diagnosis and selected based lated by the post-​pregnancy microenvironment, as suggested in animal
upon high tumor viability and cellularity. However, tumors may have models (Lyons et  al., 2011). Tumors associated with local immune
mixed phenotypes or may be spatially heterogeneous, and this could responses may be more amenable to immunotherapy, as has recently
confound accurate clinical subtyping and treatment. Understanding been demonstrated for colon carcinomas associated with microsatellite
intratumoral genotypic and phenotypic heterogeneity is a critical topic instability, a defect that may produce multiple tumor antigens that are
of cross-​disciplinary concern, although to date, this has received most recognizable by the immune system and breast cancer (Le et al., 2015;
attention with regard to predicting therapeutic responses (Alizadeh Savas et al., 2016).
et al., 2015).
As an example of the importance of intratumoral heterogeneity,
consider the estrogen receptor, arguably the most important prog- Implications for Cancer Epidemiology
nostic/​predictive biomarker in breast cancer clinical care. Tumors Historically, cancers were analyzed purely by site of origin. As appre-
that express ER strongly in every cell tend to be well differentiated, ciation of the biological heterogeneity of cancer increased, etiologi-
indolent, and more readily cured; however, many tumors classified as cal research expanded to assessment of cancer subtypes, first based
ER-​positive include substantial subpopulations of ER-​negative cells on histology, and then on limited numbers of key biomarkers. As the
(at least based on immunohistochemical assays performed at any capacity for high-​throughput molecular analysis has increased, and the
given time point), and the behavior and treatment response of such costs for performing such analyses have decreased, questions arise as
tumors is less predictable. Understanding which ER-​positive tumors to the best approach for tumor classification. In tandem with the arrival
are composed of biologically diverse clones is critical because initial of the “molecular age,” clinical practice has moved toward using less
treatments may inadvertently select for tumor subpopulations that are invasive biopsy methods, which remove smaller samples that are often
resistant to targeted therapy. Thus, identifying cancers with clonal consumed by intensive molecular testing. Further, neoadjuvant therapy
diversity and developing algorithms to classify such tumors are critical has rendered many surgically removed tissues less useful for etiologic
to the success of precision medicine; tailored treatments should target studies. Consequently, obtaining “residual” tissue for research has
the Achilles’ heel of all potentially clinically significant clones, not become more difficult, which poses challenges for conducting large
merely the dominant one. epidemiological studies.
Spatial heterogeneity ultimately arises from temporal heterogeneity As suggested in this writing, optimal classification may depend on
(i.e., because tumors change through clonal expansion and “evolution” the research questions posed: etiological, mechanistic, prognostic, or
over time), but spatial heterogeneity is particularly important because predictive. At the extreme, every cancer might be viewed as a “one of a
initial treatment success, based on accurate molecular characterization kind,” but this approach is not amenable to understanding associations
and appropriate treatment selection at diagnosis, is the best predictor using statistical methods. Accordingly, efforts to define the optimal
of treatment outcome. Spatially, some tumors are homogeneous, and parsimonious biomarker panels, which group cancers that are highly
such tumors might be anticipated to respond more uniformly, portend- similar biologically, even if not identical, are needed. To accomplish
ing a more indolent course. Previous studies have shown that the risk this goal, molecular epidemiologists will move increasingly into the
38

38 Part I:  Basic Concepts


world of trans-​disciplinary research, partnering with both laboratory Boyd NF, Guo H, Martin LJ, et  al. 2007. Mammographic density and the
scientists at the cutting edge of technology, and bioinformaticists and risk and detection of breast cancer. N Engl J Med, 356(3), 227–​236.
biostatisticians who can provide guidance in interpreting enormous PMID: 17229950.
amounts of data. Brat DJ, Verhaak RG, Aldape KD et  al. 2015. Comprehensive, integrative
genomic analysis of diffuse lower-​grade gliomas. N Engl J Med, 372(26),
References 2481–​2498. PMCID: PMC4530011.
Brenner H, Stock C, and Hoffmeister M. 2014. Effect of screening sigmoid-
Acs G, Lawton TJ, Rebbeck TR, LiVolsi VA, and Zhang PJ. 2001. Differential oscopy and screening colonoscopy on colorectal cancer incidence and
expression of E-​cadherin in lobular and ductal neoplasms of the breast mortality:  systematic review and meta-​analysis of randomised con-
and its biologic and diagnostic implications. Am J Clin Pathol, 115(1), trolled trials and observational studies. BMJ, 348, g2467. PMCID:
85–​98. PMID: 11190811. PMC3980789.
AJCC. 2011. AJCC cancer staging manual (7th ed.). Edge SB, Byrd Brinton LA, and Felix AS. 2014. Menopausal hormone therapy and risk
DR, Compton CC, Fritz AG, Greene FL, and Trotti A (Eds.). of endometrial cancer. J Steroid Biochem Mol Biol, 142, 83–​ 89.
New York: Springer. PMCID: PMC3775978.
Alexandrov LB, Nik-​Zainal S, Wedge DC, et  al. 2013. Signatures of muta- Brinton LA, Felix AS, McMeekin DS, et al. 2013. Etiologic heterogeneity in
tional processes in human cancer. Nature, 500(7463), 415–​ 421. endometrial cancer: evidence from a Gynecologic Oncology Group trial.
PMCID: PMC3776390. Gynecol Oncol, 129(2), 277–​284. PMCID: PMC4006113.
Alexandrov LB, and Stratton MR. 2014. Mutational signatures: the patterns of Bueso-​Ramos CE, Kanagal-​Shamanna R, Routbort MJ, and Hanson CA. 2015.
somatic mutations hidden in cancer genomes. Curr Opin Genet Dev, 24, Therapy-​related myeloid neoplasms. Am J Clin Pathol, 144(2), 207–​218.
52–​60. PMCID: PMC3990474. PMID: 26185306.
Alizadeh AA, Aranda V, Bardelli A, et  al. 2015. Toward understand- Burrell RA, McGranahan N, Bartek J, and Swanton C. 2013. The causes and
ing and exploiting tumor heterogeneity. Nat Med, 21(8), 846–​ 853. consequences of genetic heterogeneity in cancer evolution. Nature,
PMCID: PMC4785013. 501(7467), 338–​345. PMID: 24048066.
Alizadeh AA, Eisen MB, Davis RE et  al. 2000. Distinct types of diffuse Buys SS, Partridge E, Black A, et al. 2011. Effect of screening on ovarian can-
large B-​cell lymphoma identified by gene expression profiling. Nature, cer mortality: the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer
403(6769), 503–​511. PMID: 10676951. Screening Randomized Controlled Trial. JAMA, 305(22), 2295–​2303.
Althuis MD, Fergenbaum JH, Garcia-​ Closas M, et  al. 2004. Etiology of PMID: 21642681.
hormone receptor-​ defined breast cancer:  a systematic review of the Cairns RA. 2015. Drivers of the Warburg phenotype. Cancer J, 21(2), 56–​61.
literature. Cancer Epidemiol Biomarkers Prev, 13(10), 1558–​ 1568. PMID: 25815844.
PMID: 15466970. Camp RL, Chung GG, and Rimm DL. 2002. Automated subcellular localiza-
Anderson GL, Judd HL, Kaunitz AM, et al. 2003. Effects of estrogen plus pro- tion and quantification of protein expression in tissue microarrays. Nat
gestin on gynecologic cancers and associated diagnostic procedures: the Med, 8(11), 1323–​1327. PMID: 12389040.
Women’s Health Initiative randomized trial. JAMA, 290(13), 1739–​1748. Campbell JD, Mazzilli SA, Reid ME, et al. 2016. The case for a Pre-​Cancer
PMID: 14519708. Genome Atlas (PCGA). Cancer Prev Res (Phila), 9(2), 119–​ 124.
Anderson WF, Pfeiffer RM, Dores GM, and Sherman ME. 2006. Comparison PMID: 26839336.
of age frequency distribution patterns for different histopathologic Campo E, Swerdlow SH, Harris NL et  al. 2011. The 2008 WHO classifica-
types of breast carcinoma. Cancer Epidemiol Biomarkers Prev, 15(10), tion of lymphoid neoplasms and beyond: evolving concepts and practical
1899–​1905. applications. Blood, 117(19), 5019–​5032. PMCID: PMC3109529.
Anderson WF, Rosenberg PS, Prat A, Perou CM, and Sherman ME. 2014. How Cancer Genome Atlas Research Network. 2008. Comprehensive genomic
many etiological subtypes of breast cancer: two, three, four, or more? J characterization defines human glioblastoma genes and core pathways.
Natl Cancer Inst, 106(8). PMCID: PMC4148600. Nature, 455(7216), 1061–​1068. PMCID: PMC2671642.
The Cancer Genome Network. 2012. Comprehensive molecular portraits of Cancer Genome Atlas Research Network. 2012. Comprehensive genomic char-
human breast tumours. Nature, 490(7418), 61–​70. PMCID: PMC3465532. acterization of squamous cell lung cancers. Nature, 489(7417), 519–​525.
The Cancer Genome Network. 2015. Genomic classification of cutaneous mel- PMCID: PMC3466113.
anoma. Cell, 161(7), 1681–​1696. PMCID: PMC4580370. Cancer Genome Atlas Research Network. 2014. Comprehensive molecu-
Arias-​Stella J. 2002. The Arias-​Stella reaction: facts and fancies four decades lar profiling of lung adenocarcinoma. Nature, 511(7511), 543–​ 550.
after. Adv Anat Pathol, 9(1), 12–​23. PMID: 11756756. PMCID: PMC4231481.
Armitage P, and Doll R. 1954. The age distribution of cancer and a multi-​stage Chai H, and Brown RE. 2009. Field effect in cancer-​an update. Ann Clin Lab
theory of carcinogenesis. Br J Cancer, 8(1), 1–​12. Sci, 39(4), 331–​337. PMID: 19880759.
Armitage P, and Doll R. 1957. A two-​stage theory of carcinogenesis in relation Chanmee T, Ontong P, Kimata K, and Itano N. 2015. Key roles of hyaluronan
to the age distribution of human cancer. Br J Cancer, 11(2), 161–​169. and its CD44 receptor in the stemness and survival of cancer stem cells.
Atienza-​Amores M, Guerini-​Rocco E, Soslow RA, Park KJ, and Weigelt B. Front Oncol, 5, 180. PMCID: PMC4530590.
2014. Small cell carcinoma of the gynecologic tract: a multifaceted spec- Chereda B, and Melo JV. 2015. Natural course and biology of CML. Ann
trum of lesions. Gynecol Oncol, 134(2), 410–​418. PMID: 24875120. Hematol, 94(Suppl 2), S107–​121. PMID: 25814077.
Badve SS, Baehner FL, Gray RP, et  al. 2008. Estrogen-​and progesterone-​ Chlebowski RT, and Anderson GL. 2014. Menopausal hormone therapy and
receptor status in ECOG 2197:  comparison of immunohistochemistry cancer: changing clinical observations of target site specificity. Steroids,
by local and central laboratories and quantitative reverse transcription 90, 53–​59. PMCID: PMC4192019.
polymerase chain reaction by central laboratory. J Clin Oncol, 26(15), Chung GG, Zerkowski MP, Ghosh S, Camp RL, and Rimm DL. 2007.
2473–​2481. PMID: 18487567. Quantitative analysis of estrogen receptor heterogeneity in breast cancer.
Bao Z, Cao C, Geng X, et  al. 2016. Effectiveness and safety of poly (ADP-​ Lab Invest, 87(7), 662–​669. PMID: 17334408.
ribose) polymerase inhibitors in cancer therapy: a systematic review and Claus EB, Stowe M, and Carter D. 2001. Breast carcinoma in situ:  risk fac-
meta-​analysis. Oncotarget, 7(7), 7629–​7639. PMID: 26399274. tors and screening patterns. J Natl Cancer Inst, 93(23), 1811–​1817.
Beck M, Komis G, Ziemann A, Menzel D, and Samaj J. 2011. Mitogen-​ PMID: 11734598.
activated protein kinase 4 is involved in the regulation of mitotic and Collaborative Group on Epidemiological Studies of Ovarian Cancer, Beral V,
cytokinetic microtubule transitions in Arabidopsis thaliana. New Phytol, Gaitskell K, et  al. 2015. Menopausal hormone use and ovarian cancer
189(4), 1069–​1083. PMID: 21155826. risk: individual participant meta-​analysis of 52 epidemiological studies.
Berry DA, Cirrincione C, Henderson IC, et al. 2006. Estrogen-​receptor status Lancet, 385(9980), 1835–​1842. PMCID: PMC4427760.
and outcomes of modern chemotherapy for patients with node-​positive Colombo N, Creutzberg C, Amant F, et  al. 2016. ESMO-​ ESGO-​ ESTRO
breast cancer. JAMA, 295(14), 1658–​1667. PMID: 16609087. Consensus Conference on Endometrial Cancer: diagnosis, treatment and
Berton-​Rigaud D, Devouassoux-​Shisheboran M, Ledermann JA, et  al. 2014. follow-​up. Ann Oncol, 27(1), 16–​41. PMID: 26634381.
Gynecologic Cancer InterGroup (GCIG) consensus review for uterine Combs SE, Han G, Mani N, et al. 2016. Loss of antigenicity with tissue age in
and ovarian carcinosarcoma. Int J Gynecol Cancer, 24(9 Suppl 3), S55–​ breast cancer. Lab Invest, 96(3), 264–​269. PMID: 26568292.
60. PMID: 25341582. Cormier WJ, Gaffey TA, Welch JM, Welch JS, and Edmonson JH. 1980. Linitis
Bleyer A, and Welch HG. 2013. Effect of screening mammography on breast plastica caused by metastatic lobular carcinoma of the breast. Mayo Clin
cancer incidence. N Engl J Med, 368(7), 679. PMID: 23406034. Proc, 55(12), 747–​753. PMID: 6261047.
Bond-​Smith G, Banga N, Hammond TM, and Imber CJ. 2012. Pancreatic ade- Cote ML, Ruterbusch JJ, Olson SH, Lu K, and Ali-​Fehmi R. 2015. The grow-
nocarcinoma. BMJ, 344, e2476. PMID: 22592847. ing burden of endometrial cancer:  a major racial disparity affecting
 39

Morphological and Molecular Classification of Human Cancer 39


black women. Cancer Epidemiol Biomarkers Prev, 24(9), 1407–​1415. factors:  preliminary data from the prostate, lung, colon, and ovar-
PMID: 26290568. ian cancer screening trial. Am J Obstet Gynecol, 183(5), 1232–​1237.
D’Angelo E, and Prat J. 2011. Pathology of mixed Mullerian tumours. Best PMID: 11084571.
Pract Res Clin Obstet Gynaecol, 25(6), 705–​718. PMID: 21742560. Hasserjian RP. 2013. Acute myeloid leukemia: advances in diagnosis and clas-
Dawson SJ, Tsui DW, Murtaza M, et al. 2013. Analysis of circulating tumor sification. Int J Lab Hematol, 35(3), 358–​366. PMID: 23590662.
DNA to monitor metastatic breast cancer. N Engl J Med, 368(13), 1199–​ Henderson IC, and Patek AJ. 1998. The relationship between prognostic and
1209. PMID: 23484797. predictive factors in the management of breast cancer. Breast Cancer Res
De Waard F, Baanders-​Vanhalewijn EA, and Huizinga J. 1964. The bimodal Treat, 52(1–​3), 261–​288.
age distribution of patients with mammary carcinoma:  evidence for Herrero R, Gonzalez P, and Markowitz LE. 2015. Present status of human
the existence of 2 types of human breast cancer. Cancer, 17, 141–​151. papillomavirus vaccine development and implementation. Lancet Oncol,
PMID: 14123673. 16(5), e206–​216. PMID: 25943065.
den Boon JA, Pyeon D, Wang SS, et al. 2015. Molecular transitions from pap- Hoadley KA, Yau C, Wolf DM, et al. 2014. Multiplatform analysis of 12 cancer
illomavirus infection to cervical precancer and cancer:  role of stromal types reveals molecular classification within and across tissues of origin.
estrogen receptor signaling. Proc Natl Acad Sci U S A, 112(25), E3255–​ Cell, 158(4), 929–​944. PMCID: PMC4152462.
3264. PMCID: PMC4485108. Hou N, Hong S, Wang W, et al. 2013. Hormone replacement therapy and breast
Diaz LA, Jr., and Le DT. 2015. PD-​1 blockade in tumors with mismatch-​repair cancer:  heterogeneous risks by race, weight, and breast density. J Natl
deficiency. N Engl J Med, 373(20), 1979. PMID: 26559582. Cancer Inst, 105(18), 1365–​1372. PMCID: PMC3776262.
Dobbins M, Decorby K, and Choi BC. 2013. The association between obesity Hu Z, Fan C, Oh DS, et  al. 2006. The molecular portraits of breast tumors
and cancer risk:  a meta-​analysis of observational studies from 1985 to are conserved across microarray platforms. BMC Genomics, 7, 96.
2011. ISRN Prev Med, 2013, 680536. PMCID: PMC4062857. PMCID: PMC1468408.
Dunbier AK, Anderson H, Ghazoui Z, et al. 2011. Association between breast Huh WK, Ault KA, Chelmow D, et al. 2015. Use of primary high-​risk human
cancer subtypes and response to neoadjuvant anastrozole. Steroids, 76(8), papillomavirus testing for cervical cancer screening:  interim clinical
736–​740. PMID: 21447351. guidance. J Low Genit Tract Dis, 19(2), 91–​96. PMID: 25574659.
Dvorak HF. 2015. Tumors:  wounds that do not heal-​redux. Cancer Immunol Huo D, Ikpatt F, Khramtsov A, et  al. 2009. Population differences in breast
Res, 3(1), 1–​11. PMCID: PMC4288010. cancer: survey in indigenous African women reveals over-​representation
Esmaeilsabzali H, Beischlag TV, Cox ME, Parameswaran AM, and Park EJ. of triple-​negative breast cancer. J Clin Oncol, 27(27), 4515–​ 4521.
2013. Detection and isolation of circulating tumor cells:  principles and PMID: 19704069.
methods. Biotechnol Adv, 31(7), 1063–​1084. PMID: 23999357. Hyman DM, Puzanov I, Subbiah V, et al. 2015. Vemurafenib in multiple non-
Esserman LJ, Thompson IM, Reid B, et al. 2014. Addressing overdiagnosis and melanoma cancers with BRAF V600 mutations. N Engl J Med, 373(8),
overtreatment in cancer: a prescription for change. Lancet Oncol, 15(6), 726–​736. PMID: 26287849.
e234–​242. PMCID: PMC4322920. Ignatiadis M, Azim HA, Jr., Desmedt C, et al. 2016. The genomic grade assay
Fergenbaum JH, Garcia-​Closas M, Hewitt SM, et al. 2004. Loss of antigenicity compared with Ki67 to determine risk of distant breast cancer recurrence.
in stored sections of breast cancer tissue microarrays. Cancer Epidemiol JAMA Oncol, 2(2), 217–​224. PMID: 26633571.
Biomarkers Prev, 13(4), 667–​672. PMID: 15066936. Ishizumi T, McWilliams A, MacAulay C, Gazdar A, and Lam S. 2010. Natural
Figueroa JD, Pfeiffer RM, Patel DA, et  al. 2014. Terminal duct lobular unit history of bronchial preinvasive lesions. Cancer Metastasis Rev, 29(1),
involution of the normal breast: implications for breast cancer etiology. J 5–​14. PMCID: PMC3417071.
Natl Cancer Inst, 106(10). PMCID: PMC4200067. Jacinto FV, and Esteller M. 2007. Mutator pathways unleashed by epi-
Finn OJ. 2014. Vaccines for cancer prevention:  a practical and feasible genetic silencing in human cancer. Mutagenesis, 22(4), 247–​ 253.
approach to the cancer epidemic. Cancer Immunol Res, 2(8), 708–​713. PMID: 17412712.
PMCID: PMC4163937. Jacobs IJ, Menon U, Ryan A, et  al. 2016. Ovarian cancer screening and
Franzke B, Neubauer O, and Wagner KH. 2015. Super DNAging: new insights mortality in the UK Collaborative Trial of Ovarian Cancer Screening
into DNA integrity, genome stability and telomeres in the oldest old. (UKCTOCS):  a randomised controlled trial. Lancet, 387(10022), 945–​
Mutat Res Rev Mutat Res, 766, 48–​57. PMID: 26596548. 956. PMCID: PMC4779792.
Frede J, Adams DJ, and Jones PH. 2014. Mutation, clonal fitness and Jain P, and O’Brien S. 2012. Richter’s transformation in chronic lym-
field change in epithelial carcinogenesis. J Pathol, 234(3), 296–​301. phocytic leukemia. Oncology (Williston Park), 26(12), 1146–​ 1152.
PMID: 25046364. PMID: 23413591.
Geiss GK, Bumgarner RE, Birditt B, et al. 2008. Direct multiplexed measure- Jatoi I, and Anderson WF. 2005. Cancer screening. Curr Prob Surgery, 42(9),
ment of gene expression with color-​coded probe pairs. Nat Biotechnol, 620–​682.
26(3), 317–​325. PMID: 18278033. Jemal A, Simard EP, Dorell C, et al. 2013. Annual report to the nation on the
Gentleman RC, Carey VJ, Bates DM, et al. 2004. Bioconductor: open software status of cancer, 1975–​2009, featuring the burden and trends in human
development for computational biology and bioinformatics. Genome papillomavirus(HPV)-​associated cancers and HPV vaccination coverage
Biol, 5(10), R80. PMCID: 545600. levels. J Natl Cancer Inst, 105(3), 175–​201. PMCID: PMC3565628.
Gucalp A, Iyengar NM, Hudis CA, and Dannenberg AJ. 2016. Targeting Jemal A, Thun MJ, Ries LA, et  al. 2008. Annual report to the nation on the
obesity-​related adipose tissue dysfunction to prevent cancer development status of cancer, 1975–​2005, featuring trends in lung cancer, tobacco
and progression. Semin Oncol, 43(1), 154–​160. PMCID: PMC4789163. use, and tobacco control. J Natl Cancer Inst, 100(23), 1672–​ 1694.
Gyorffy B, Hatzis C, Sanft T, et  al. 2015. Multigene prognostic tests PMCID: PMC2639291.
in breast cancer:  past, present, future. Breast Cancer Res, 17, 11. Johnson K, Sarma D, and Hwang ES. 2015. Lobular breast cancer series: imag-
PMCID: PMC4307898. ing. Breast Cancer Res, 17, 94. PMCID: PMC4499185.
Halfter W, Oertle P, Monnier CA, et al. 2015. New concepts in basement mem- Kononen J, Bubendorf L, Kallioniemi A, et  al. 1998. Tissue microarrays for
brane biology. FEBS J, 282(23), 4466–​4479. PMID: 26299746. high-​throughput molecular profiling of tumor specimens. Nat Med, 4(7),
Hallek M. 2013. Chronic lymphocytic leukemia:  2013 update on diagno- 844–​847. PMID: 9662379.
sis, risk stratification and treatment. Am J Hematol, 88(9), 803–​816. Kuppers R, Engert A, and Hansmann ML. 2012. Hodgkin lymphoma. J Clin
PMID: 23720127. Invest, 122(10), 3439–​3447. PMCID: PMC3534167.
Hameed M. 2014. Molecular diagnosis of soft tissue neoplasia: clinical appli- Kurman RJ, and Shih Ie M. 2016. The dualistic model of ovarian carcinogen-
cations and recent advances. Expert Rev Mol Diagn, 14(8), 961–​977. esis:  revisited, revised, and expanded. Am J Pathol, 186(4), 733–​747.
PMID: 25109831. PMID: 27012190.
Hanahan D, and Weinberg RA. 2011. Hallmarks of cancer: the next generation. Lander ES, Linton LM, Birren B, et al. 2001. Initial sequencing and analysis
Cell, 144(5), 646–​674. PMID: 21376230. of the human genome. Nature, 409(6822), 860–​921. PMID: 11237011.
Hariri S, Johnson ML, Bennett NM, et  al. 2015. Population-​based trends in Langley RR, and Fidler IJ. 2011. The seed and soil hypothesis revisited: the
high-​grade cervical lesions in the early human papillomavirus vaccine role of tumor-​stroma interactions in metastasis to different organs. Int J
era in the United States. Cancer, 121(16), 2775–​2781. PMID: 26098295. Cancer, 128(11), 2527–​2535. PMCID: PMC3075088.
Harris NL, Jaffe ES, Diebold J, et  al. 2000. The World Health Organization Le DT, Uram JN, Wang H et  al. 2015. PD-​ 1 blockade in tumors with
classification of hematological malignancies report of the Clinical mismatch-​repair deficiency. N Engl J Med, 372(26), 2509–​ 2520.
Advisory Committee Meeting, Airlie House, Virginia, November 1997. PMCID: PMC4481136.
Mod Pathol, 13(2), 193–​207. PMID: 10697278. Lee Y, Miron A, Drapkin R, et al. 2007. A candidate precursor to serous carci-
Hartge P, Hayes R, Reding D, et  al. 2000. Complex ovarian cysts in post- noma that originates in the distal fallopian tube. J Pathol, 211(1), 26–​35.
menopausal women are not associated with ovarian cancer risk PMID: 17117391.
40

40 Part I:  Basic Concepts


Levenson RM, Borowsky AD, and Angelo M. 2015. Immunohistochemistry Ottaviani G, and Jaffe N. 2009. The epidemiology of osteosarcoma. Cancer
and mass spectrometry for highly multiplexed cellular molecular imag- Treat Res, 152, 3–​13. PMID: 20213383.
ing. Lab Invest, 95(4), 397–​405. PMID: 25730370. Paget S. 1889. The distribution of secondary growths in cancer of the breasts.
Lilienfeld AM, and Johnson EA. 1955. The age distribution in female breast Lancet (March 23, 1889), 571–​573.
and genital cancers. Cancer, 8(5), 875–​882. PMID: 13261037. Paolini S, Gazzola A, Sabattini E, et al. 2011. Pathobiology of acute lympho-
Lochhead P, Chan AT, Nishihara R, et  al. 2015. Etiologic field effect:  reap- blastic leukemia. Semin Diagn Pathol, 28(2), 124–​134. PMID: 21842698.
praisal of the field effect concept in cancer predisposition and progres- Parsons DW, Jones S, Zhang X, et  al. 2008. An integrated genomic analysis
sion. Mod Pathol, 28(1), 14–​29. PMCID: PMC4265316. of human glioblastoma multiforme. Science, 321(5897), 1807–​ 1812.
Logothetis CJ, Gallick GE, Maity SN, et  al. 2013. Molecular classifica- PMCID: 2820389.
tion of prostate cancer progression:  foundation for marker-​ driven Pasquier J, Abu-​Kaoud N, Al Thani H, and Rafii A. 2015. Epithelial to mes-
treatment of prostate cancer. Cancer Discov, 3(8), 849–​861. PMCID: enchymal transition in a clinical perspective. J Oncol, 2015, 792182.
PMC3926428. PMCID: PMC4575734.
Lord CJ, and Ashworth A. 2013. Mechanisms of resistance to therapies tar- Patai AV, Molnar B, Tulassay Z, and Sipos F. 2013. Serrated pathway: alterna-
geting BRCA-​mutant cancers. Nat Med, 19(11), 1381–​1388. PMID: tive route to colorectal cancer. World J Gastroenterol, 19(5), 607–​615.
24202391. PMCID: PMC3574586.
Lowy DR, Herrero R, Hildesheim A, and Participants in the IARC/​ NCI Pearce CL, Templeman C, Rossing MA, et  al. 2012. Association between
Workshop on Primary Endpoints for Prophylactic HPV Vaccine Trials. endometriosis and risk of histological subtypes of ovarian cancer:  a
2015. Primary endpoints for future prophylactic human papillomavirus pooled analysis of case-​control studies. Lancet Oncol, 13(4), 385–​394.
vaccine trials:  towards infection and immunobridging. Lancet Oncol, PMCID: PMC3664011.
16(5), e226–​233. PMID: 25943067. Peirson L, Fitzpatrick-​Lewis D, Ciliska D, and Warren R. 2013. Screening for
Lyons TR, O’Brien J, Borges VF, et al. 2011. Postpartum mammary gland invo- cervical cancer: a systematic review and meta-​analysis. Syst Rev, 2, 35.
lution drives progression of ductal carcinoma in situ through collagen and PMCID: PMC3681632.
COX-​2. Nat Med, 17(9), 1109–​1115. PMCID: PMC3888478. Perez EA, Cortes J, Gonzalez-​Angulo AM, and Bartlett JM. 2014. HER2 test-
Mandal S, Pile K, Chacko RT, and Danda D. 2014. Malignancy and autoim- ing: current status and future directions. Cancer Treat Rev, 40(2), 276–​
munity: causally or casually related? Int J Rheum Dis, 17(6), 601–​605. 284. PMID: 24080154.
PMID: 25346148. Perou CM, Sorlie T, Eisen MB, et al. 2000. Molecular portraits of human breast
Marcucci F, Bellone M, Caserta CA, and Corti A. 2014. Pushing tumor cells tumours. Nature, 406(6797), 747–​752. PMID: 10963602.
towards a malignant phenotype:  stimuli from the microenvironment, Pesch B, Kendzia B, Gustavsson P, et  al. 2012. Cigarette smoking and lung
intercellular communications and alternative roads. Int J Cancer, 135(6), cancer:  relative risk estimates for the major histological types from a
1265–​1276. PMID: 24174383. pooled analysis of case-​control studies. Int J Cancer, 131(5), 1210–​1219.
Martincorena I, and Campbell PJ. 2015. Somatic mutation in cancer and nor- PMCID: PMC3296911.
mal cells. Science, 349(6255), 1483–​1489. PMID: 26404825. Pickup MW, Mouw JK, and Weaver VM. 2014. The extracellular matrix
McNeil C. 2015. NCI-​MATCH launch highlights new trial design in precision-​ modulates the hallmarks of cancer. EMBO Rep, 15(12), 1243–​ 1253.
medicine era. J Natl Cancer Inst, 107(7). PMID: 26142446. PMCID: PMC4264927.
McShane LM, Altman DG, Sauerbrei W, et al. 2005. Reporting recommenda- Pienta KJ, Robertson BA, Coffey DS, and Taichman RS. 2013. The cancer dias-
tions for tumor marker prognostic studies (REMARK). J Natl Cancer pora: metastasis beyond the seed and soil hypothesis. Clin Cancer Res,
Inst, 97(16), 1180–​1184. PMID: 16106022. 19(21), 5849–​5855. PMCID: PMC3835696.
Menon MP, Pittaluga S, and Jaffe ES. 2012. The histological and biological spec- Pike MC, Krailo MD, Henderson BE, Casagrande JT, and Hoel DG. 1983.
trum of diffuse large B-​cell lymphoma in the World Health Organization “Hormonal” risk factors, “breast tissue age” and the age-​incidence of
classification. Cancer J, 18(5), 411–​420. PMCID: PMC3458515. breast cancer. Nature, 303(5920), 767–​770. PMID: 6866078.
Merlo LM, Shah NA, Li X, et  al. 2010. A comprehensive survey of clonal Pozner-​Moulis S, Cregger M, Camp RL, and Rimm DL. 2007. Antibody
diversity measures in Barrett’s esophagus as biomarkers of progression to validation by quantitative analysis of protein expression using expres-
esophageal adenocarcinoma. Cancer Prev Res (Phila), 3(11), 1388–​1397. sion of Met in breast cancer as a model. Lab Invest, 87(3), 251–​260.
PMCID: PMC3004782. PMID: 17260003.
Miglioretti DL, Zhu W, Kerlikowske K, et al. 2015. Breast tumor prognostic Rabban JT, and Zaloudek CJ. 2007. Minimal uterine serous carcinoma:  cur-
characteristics and biennial vs annual mammography, age, and meno- rent concepts in diagnosis and prognosis. Pathology, 39(1), 125–​133.
pausal status. JAMA Oncol, 1(8), 1069–​1077. PMCID: PMC4644100. PMID: 17365828.
Moscicki AB, Schiffman M, Burchell A, et al. 2012. Updating the natural his- Ravdin PM, Cronin KA, Howlader N, et al. 2007. The decrease in breast-​cancer
tory of human papillomavirus and anogenital cancers. Vaccine, 30(Suppl incidence in 2003 in the United States. N Engl J Med, 356(16), 1670–​
5), F24–​33. PMCID: PMC3700362. 1674. PMID: 17442911.
Mutter GL, Ince TA, Baak JP, et  al. 2001. Molecular identification of latent Rice MS, Murphy MA, and Tworoger SS. 2012. Tubal ligation, hysterec-
precancers in histologically normal endometrium. Cancer Res, 61(11), tomy and ovarian cancer:  a meta-​ analysis. J Ovarian Res, 5(1), 13.
4311–​4314. PMID: 11389050. PMCID: PMC3386864.
National Lung Screening Trial Research Team, Church TR, Black WC, Riching KM, Cox BL, Salick MR, et al. 2014. 3D collagen alignment limits
et al. 2013. Results of initial low-​dose computed tomographic screen- protrusions to enhance breast cancer cell persistence. Biophys J, 107(11),
ing for lung cancer. N Engl J Med, 368(21), 1980–​1991. PMCID: 2546–​2558. PMCID: PMC4255204.
PMC3762603. Rosenberg PS, and Anderson WF. 2011. Age-​period-​cohort models in can-
Niebling MG, Pleijhuis RG, Bastiaannet E, et al. 2016. A systematic review and cer surveillance research:  ready for prime time? Cancer Epidemiol
meta-​analyses of sentinel lymph node identification in breast cancer and Biomarkers Prev, 20(7), 1263–​1268. PMCID: PMC3132831.
melanoma, a plea for tracer mapping. Eur J Surg Oncol, 42(4), 466–​473. Rouzier R, Perou CM, Symmans WF, et al. 2005. Breast cancer molecular sub-
PMID: 26853759. types respond differently to preoperative chemotherapy. Clin Cancer Res,
Nik-​Zainal S, Alexandrov LB, Wedge DC, et  al. 2012. Mutational processes 11(16), 5678–​5685. PMID: 16115903.
molding the genomes of 21 breast cancers. Cell, 149(5), 979–​ 993. Roychoudhuri R, Putcha V, and Moller H. 2006. Cancer and laterality: a study
PMID: 22608084. of the five major paired organs (UK). Cancer Causes Control, 17(5), 655–​
Nikiforov YE, Seethala RR, Tallini G, et al. 2016. Nomenclature Revision for 662. PMID: 16633912.
Encapsulated follicular variant of papillary thyroid carcinoma:  a para- Runge TM, Abrams JA, and Shaheen NJ. 2015. Epidemiology of Barrett’s
digm shift to reduce overtreatment of indolent tumors. JAMA Oncol, 2(8), esophagus and esophageal adenocarcinoma. Gastroenterol Clin North
1023–​1029. PMID: 27078145. Am, 44(2), 203–​231. PMCID: PMC4449458.
Noushmehr H, Weisenberger DJ, Diefes K, et al. 2010. Identification of a CpG Salgado R, Denkert C, Demaria S, et  al. 2015. The evaluation of tumor-​
island methylator phenotype that defines a distinct subgroup of glioma. infiltrating lymphocytes (TILs) in breast cancer:  recommendations by
Cancer Cell, 17(5), 510–​522. PMCID: 2872684. an International TILs Working Group 2014. Ann Oncol, 26(2), 259–​271.
Oliva E, Pinheiro NF, Heney NM, et  al. 2013. Immunohistochemistry as an PMID: 25214542.
adjunct in the differential diagnosis of radiation-​induced atypia versus Savas P, Salgado R, Denkert C, et al. 2016. Clinical relevance of host immu-
urothelial carcinoma in situ of the bladder:  a study of 45 cases. Hum nity in breast cancer: from TILs to the clinic. Nat Rev Clin Oncol, 13(4),
Pathol, 44(5), 860–​866. PMID: 23199526. 228–​241. PMID: 26667975.
Ordonez NG. 2013. Broad-​spectrum immunohistochemical epithelial mark- Schatzkin A, and Gail M. 2002. The promise and peril of surrogate end points
ers: a review. Hum Pathol, 44(7), 1195–​1215. PMID: 23427873. in cancer research. Nat Rev Cancer, 2(1), 19–​27. PMID: 11902582.
 41

Morphological and Molecular Classification of Human Cancer 41


Seidman JD, Zhao P, and Yemelyanova A. 2011. “Primary peritoneal” high-​ Tyekucheva S, Martin NE, Stack EC, et  al. 2015. Comparing platforms for
grade serous carcinoma is very likely metastatic from serous tubal messenger RNA expression profiling of archival formalin-​fixed, paraffin-​
intraepithelial carcinoma: assessing the new paradigm of ovarian and pel- embedded tissues. J Mol Diagn, 17(4), 374–​381. PMCID: PMC4483460.
vic serous carcinogenesis and its implications for screening for ovarian Vercellini P. 2015. Introduction: management of endometriosis: moving toward
cancer. Gynecol Oncol, 120(3), 470–​473. PMID: 21159368. a problem-​oriented and patient-​centered approach. Fertil Steril, 104(4),
Sherman ME, Figueroa JD, Henry JE, et al. 2012. The Susan G. Komen for the 761–​763. PMID: 26363383.
Cure Tissue Bank at the IU Simon Cancer Center: a unique resource for Ward E, DeSantis C, Robbins A, Kohler B, and Jemal A. 2014. Childhood
defining the “molecular histology” of the breast. Cancer Prev Res (Phila), and adolescent cancer statistics, 2014. CA Cancer J Clin, 64(2), 83–​103.
5(4), 528–​535. PMCID: PMC4159749. PMID: 24488779.
Sherman ME, Howatt W, Blows FM, et al. 2010. Molecular pathology in epi- Wartko P, Sherman ME, Yang HP, et al. 2013. Recent changes in endometrial
demiologic studies:  a primer on key considerations. Cancer Epidemiol cancer trends among menopausal-​age U.S.  women. Cancer Epidemiol,
Biomarkers Prev, 19(4), 966–​972. PMCID: PMC2852464. 37(4), 374–​377. PMCID: PMC3679300.
Sherman ME, Lorincz AT, Scott DR et  al. 2003. Baseline cytology, human Weiss NS. 2003. Adjusting for screening history in epidemiologic studies of
papillomavirus testing, and risk for cervical neoplasia: a 10-​year cohort cancer: why, when, and how to do it. Am J Epidemiol, 157(11), 957–​961.
analysis. J Natl Cancer Inst, 95(1), 46–​52. PMID: 12509400. PMID: 12777356.
Sherman ME, Mies C, and Gierach GL. 2014. Opportunities for molecular Welch HG, Gorski DH, and Albertsen PC. 2015. Trends in metastatic breast
epidemiological research on ductal carcinoma in-​situ and breast car- and prostate cancer—​lessons in cancer dynamics. N Engl J Med, 373(18),
cinogenesis:  interdisciplinary approaches. Breast Dis, 34(3), 105–​116. 1685–​1687. PMID: 26510017.
PMCID: PMC4558907. Wheeler DT, Bell KA, Kurman RJ, and Sherman ME. 2000. Minimal uterine
Shiels MS, Gibson T, Sampson J, et al. 2014. Cigarette smoking prior to first serous carcinoma: diagnosis and clinicopathologic correlation. Am J Surg
cancer and risk of second smoking-​associated cancers among survivors Pathol, 24(6), 797–​806. PMID: 10843281.
of bladder, kidney, head and neck, and stage I lung cancers. J Clin Oncol, Winer EP, Carey LA, Dowsett M, and Tripathy D. 2005. Beyond anatomic
32(35), 3989–​3995. PMCID: PMC4251962. staging:  are we ready to take the leap to molecular classification? In:
Snover DC. 2010. Maximizing the value of the endoscopist-​pathologist partner- Perry MC (Ed.), American Society of Clinical Onocolgy education book
ship in the management of colorectal polyps and carcinoma. Gastrointest (pp. 46–​59). Alexandria, VA: American Society of Clinical Oncology.
Endosc Clin N Am, 20(4), 641–​657. PMID: 20889069. Xie Y, Pittaluga S, and Jaffe ES. 2015. The histological classification
Sorlie T, Perou CM, Tibshirani R, et al. 2001. Gene expression patterns of breast of diffuse large B-​ cell lymphomas. Semin Hematol, 52(2), 57–​ 66.
carcinomas distinguish tumor subclasses with clinical implications. Proc PMCID: PMC4374126.
Natl Acad Sci U S A, 98(19), 10869–​10874. PMCID: PMC58566. Yakirevich E, and Resnick MB. 2013. Pathology of gastric cancer and its precursor
Soslow RA. 2011. Mucinous ovarian carcinoma:  slippery business. Cancer, lesions. Gastroenterol Clin North Am, 42(2), 261–​284. PMID: 23639640.
117(3), 451–​453. PMID: 20845480. Yang HP, Anderson WF, Rosenberg PS, et  al. 2013. Ovarian cancer inci-
Tibes R, Qiu Y, Lu Y, et al. 2006. Reverse phase protein array: validation of a dence trends in relation to changing patterns of menopausal hormone
novel proteomic technology and utility for analysis of primary leukemia therapy use in the United States. J Clin Oncol, 31(17), 2146–​2151.
specimens and hematopoietic stem cells. Mol Cancer Ther, 5(10), 2512–​ PMCID: PMC3731982.
2521. PMID: 17041095. Yang XR, Chang-​Claude J, Goode EL, et al. 2011. Associations of breast can-
Tomasetti C, and Vogelstein B. 2015. Cancer etiology: variation in cancer risk cer risk factors with tumor subtypes: a pooled analysis from the Breast
among tissues can be explained by the number of stem cell divisions. Cancer Association Consortium studies. J Natl Cancer Inst, 103(3), 250–​
Science, 347(6217), 78–​81. PMCID: PMC4446723. 263. PMCID: PMC3107570.
Toyota M, Ahuja N, Ohe-​Toyota M, et al. 1999. CpG island methylator pheno- Yang XR, Charette LA, Garcia-​Closas M, et al. 2006. Construction and vali-
type in colorectal cancer. Proc Natl Acad Sci U S A, 96(15), 8681–​8686. dation of tissue microarrays of ductal carcinoma in situ and terminal
PMCID: PMC17576. duct lobular units associated with invasive breast carcinoma. Diagn Mol
Tran NH, Cavalcante LL, Lubner SJ, et al. 2015. Precision medicine in colorec- Pathol, 15(3), 157–​161. PMID: 16932071.
tal cancer: the molecular profile alters treatment strategies. Ther Adv Med Yeh AC, and Ramaswamy S. 2015. Mechanisms of cancer cell dor-
Oncol, 7(5), 252–​262. PMCID: PMC4543854. mancy:  another hallmark of cancer? Cancer Res, 75(23), 5014–​5022.
Travis WD, Brambilla E, Noguchi M, et  al. 2011. International Association PMCID: PMC4668214.
for the Study of Lung Cancer/​ American Thoracic Society/​ European Yu K, Lee CH, Tan PH, and Tan P. 2004. Conservation of breast cancer
Respiratory Society: international multidisciplinary classification of lung molecular subtypes and transcriptional patterns of tumor progression
adenocarcinoma:  executive summary. Proc Am Thorac Soc, 8(5), 381–​ across distinct ethnic populations. Clin Cancer Res, 10(16), 5508–​5517.
385. PMID: 21926387. PMID: 15328190.
Troester MA, Herschkowitz JI, Oh DS, et  al. 2006. Gene expression pat- Zhang L, Wei Q, Mao L, et al. 2009. Serial dilution curve: a new method for
terns associated with p53 status in breast cancer. BMC Cancer, 6, 276. analysis of reverse phase protein array data. Bioinformatics, 25(5), 650–​
PMCID: PMC1764759. 654. PMCID: PMC2647837.
True LD. 2014. Methodological requirements for valid tissue-​based biomarker Zhao M, and Ramaswamy B. 2014. Mechanisms and therapeutic advances in
studies that can be used in clinical practice. Virchows Arch, 464(3), 257–​ the management of endocrine-​resistant breast cancer. World J Clin Oncol,
263. PMCID: PMC4009398. 5(3), 248–​262. PMCID: PMC4127598.
42
 43

4 Genomic Landscape of Cancer

Insights for Epidemiologists

CHRISTOPHER J. MAHER AND ELAINE R. MARDIS

OVERVIEW in some cases rarely) affected by cancer. Indeed, several discover-


ies have indicated surprising new genes and pathways that, when
The past decade has seen rapid advances in the study of cancer altered, lead to cancer onset and progression. For example, spliceo-
genomics using next-​generation or massively parallel technology some proteins, cohesin complex genes, and metabolic enzymes have
for DNA sequencing. The resulting knowledge has transformed our been identified as predominantly mutated in certain tumor types and
understanding of the genetic underpinnings of cancer susceptibility, have been shown by follow-​on functional experiments to contrib-
onset, and progression. New technologies have increased the speed ute to carcinogenesis. There also have been surprises regarding the
and have lowered the cost of DNA sequencing, and have enabled germline contribution to cancer susceptibility. Several studies have
high-​volume characterization of RNA, DNA methylation, DNA-​ demonstrated the contribution of de novo mutations in known can-
protein complexes, DNA conformation, and a host of other fac- cer susceptibility genes to the onset of disease. These discoveries
tors that, when altered, can lead to cancer onset and progression. indicate that, while a predisposition to cancer as ascertained by fam-
Technological advances have greatly expanded research on somatic ily history provides important clinical data, the absence of family
changes in tumor tissue, revealing both the singularity of individual history cannot solely rule out a germline contribution. Finally, by
cancer genomes and the commonality of genetic alterations that comparing genomic alterations across different tissue sites, so-​called
drive cancer in different tissues. Regulatory processes involve not pan cancer analyses have demonstrated the commonality of genomic
only changes in nucleotide sequence but also functionally relevant alterations across cancer genomes and yet have emphasized in sin-
effects mediated through epigenetics, transcription factors, non-​cod- gularity that each patient’s cancer genome is composed of a unique
ing RNAs, and chromatin packaging. Combined with our enhanced set of genomic alterations. Beyond genes and their alterations, a
understanding of cancer cell-​specific alterations in the genome is a richer understanding of gene regulatory mutations and other types of
growing understanding of how our constitutional genome impacts regulatory alterations has been revealed using genome-​wide assays
cancer susceptibility. Hence, the introduction to the current frame- that are read out by MPS to characterize the cancer epigenome.
work of cancer genomics we provide here will inform future studies These efforts include studies of newer types of non-​coding RNAs
of the genetic epidemiology of cancer. that are only beginning to be classified as overexpressed in cancers
and to have associated regulatory roles. Taken together, the cancer
genome landscape is incredibly complex and varied, yet these newer
INTRODUCTION approaches are increasing our knowledge with each new study.

Over the past 10  years, the study of cancer genomics using next-​
generation or massively parallel technology for DNA sequencing THE IMPACT OF TECHNOLOGY
has transformed our understanding of the genetic underpinnings
of cancer susceptibility, onset, and progression. Expanding beyond Massively Parallel Sequencing
the detection of genomic alterations, methods development aimed
at characterizing RNA, DNA methylation, DNA-​protein complexes, Following completion of the human genome reference sequence in
DNA conformation, and a host of other aspects of cancer-​altered 2004 (International Human Genome Sequencing, 2004), capillary
nucleic acids has resulted from the increasing throughput and sequencing technology was used to study mutations in known cancer
decreasing cost of DNA sequencing. One vital component of this genes following directed polymerase chain reaction (PCR) ampli-
progress was the initial sequence, and ever-​improving completion, of fication of the gene’s exons. Increasing the numbers of PCR prim-
the human reference genome that provides the framework for inter- ers across all annotated exons of every human gene was possible,
preting the data generated by these increasingly complex methods but there were significant challenges due to the cost of sequencing,
to evaluate changes that are contributing to the onset and progres- the scalability of the molecular biology, and the need for extraordi-
sion of cancer. The human reference genome (International Human narily large amounts of DNA from the tumor to accomplish many
Genome Sequencing, 2004) is indispensable in this new era of cancer thousands of PCRs done individually. As such, most early studies
genomics discovery since the short read lengths produced by mas- of cancer genomes focused on known genes in relatively large num-
sively parallel sequencing (MPS) instruments require alignment of bers of tumors, or sequenced large numbers of genes (or all of them)
sequencing reads to the reference genome as a first step to data anal- in a small number of tumors (Ding et  al., 2008; Ley et  al., 2003;
ysis. In other words, rather than being able to assemble the individual Wood et  al., 2007). In the first decade of the 2000s, new sequenc-
chromosomes from the sequencing data reads, one must first align ing technologies were introduced that were called “next-​generation”
to the fixed reference genome and then use a variety of interpreta- or “massively parallel” sequencing instruments. These technologies
tional algorithms to evaluate the data according to the experimental were distinctly different from the combination of Sanger sequenc-
“question” being asked by the upfront preparatory assays (Mardis, ing and capillary electrophoresis-​based separation and detection of
2013). Hence, by using this general paradigm for data generation and reaction products because, rather than a decoupled set of reactions to
analysis, cancer genomics efforts have tremendously enhanced our produce sequencing fragments that were subsequently separated and
understanding of the differences between tumor and normal genomes detected on the sequencing instrument, these instruments permitted
across the breadth of adult and pediatric tissue sites commonly (and sequencing and detection in situ, without the need for electrophoretic

43
4

44 Part I:  Basic Concepts


fragment separation. As such, one could combine, for example, all RNAseq
PCR products resulting from primer-​directed amplification in indi-
vidual reactions, and produce sequencing data for all products at RNA can be sequenced using massively parallel approaches as well,
once. The result was a massive increase in throughput and a dramatic following an initial conversion to DNA by reverse transcriptase
decrease in the cost of the sequencing reactions, but still required (Mortazavi et al., 2008). This can be accomplished from total RNA,
large amounts of tumor-​isolated DNA to feed the upfront PCRs. which subsequently is treated prior to sequencing to subtract ribo-
The alternative approach, prior to 2009, was to sequence the entire somal RNA (rRNA) sequences due to their incredible abundance in the
genome of the tumor using these new instruments, but the large human transcriptome. rRNA subtraction takes a number of forms and
amount of data produced by this approach, the expense of producing can include the use of an exome hybrid capture step, described earlier,
that data, and the lack of a computational analysis method to make or a number of other selection approaches. When RNA is abundant, an
sense of the hundreds of millions of short read–​length sequences initial step to isolate polyadenylated mRNAs from total RNA can be
made the analysis seem implausible. In 2008, however, the first such accomplished with poly-​T labeled magnetic beads that hybridize the
study was reported for a single patient whole genome, comparing polyadenylated mRNAs and isolate them from solution by applying a
the acute myeloid leukemia (AML) genome to the matched skin-​ magnetic field. After washing away the non-​polyadenylated RNAs, a
derived normal genome (Ley et  al., 2008), using the Solexa (now sequencing library is generated from reverse transcriptase-​converted
Illumina) technology. In this study, as in all to follow, the human sequences prior to sequencing. Since gene-​coding RNAs are not the
reference genome provided the template against which short reads only RNAs of potential value to study in comparing cancer to nor-
were aligned and variants detected, and then algorithm-​driven com- mal tissues, as outlined later in this chapter, a variety of approaches to
parison approaches were used to identify somatic alterations unique generate libraries from non-​coding RNAs has evolved in the context
to the tumor. Many aspects of the data provided by MPS have of MPS to permit the characterization of different classes of ncRNAs,
changed since this early study, including read-​length increases (cur- such as microRNAs and lncRNAs. In all RNA studies of tumor tissue,
rently Illumina produces 150 bp), the capability to produce sequence the challenge of data interpretation lies in placing results into context,
data from both ends of each library fragment (“paired-​end reads”), which typically requires an appropriate and parallel study from a com-
and the instrument throughput (coverage for multiple whole human parative (i.e., adjacent, non-​malignant) normal tissue.
genomes can be produced in a few days per instrument). Due to these
types of improvements, algorithms that examine the aligned reads to
identify different types of alterations have also been developed and
LARGE-​SCALE CANCER GENOME DISCOVERY
applied to tease out focused insertions or deletions of one to several
nucleotides (“indels”), to identify either increased coverage indica- Data Deluge
tive of an amplified chromosomal region or decreased coverage at
deleted regions, as well as structural variations (inversions, trans- The result of these rapidly developing methods and the decreasing
locations) based on paired end reads that do not map as anticipated cost of MPS has been an explosion in our characterization of can-
onto the genome. cer genomes, exomes, and transcriptomes, revealing catalogs of new
mutations and other somatic alterations in genes and alterations in
transcription that can lead to cancer onset or progression. In particular,
Exome Hybrid Capture two large-​scale cancer genomics studies that are investigating adult
cancers are The Cancer Genome Atlas (TCGA; www.cancergenome.
In 2009, several groups developed methods based on nucleic acid nih.gov) and the International Cancer Genome Consortium (ICGC;
hybridization that acted to “subset” the genome by capturing only www.icgc.org). TCGA is a largely US-​based project that was funded
the coding portions (the “exome”) of known genes (Bainbridge by the National Cancer Institute (NCI) and the National Human
et al., 2010; Gnirke et al., 2009; Hodges et al., 2009). Exome hybrid Genome Research Institute (NHGRI). This project has studied over
capture is accomplished by synthetic DNA or RNA probes that 10,000 cancer samples, but due to cost limitations, most were stud-
are designed to hybridize library fragments from a whole genome ied only by exome sequencing, with only around 10% of each cancer
library based on nucleic acid sequence similarity. The synthetic type sequenced by whole genome methods. By contrast, ICGC is an
probes have biotin molecules covalently linked to them during syn- international project that requires individual groups to ascertain fund-
thesis, permitting a post-​hybridization pull-​down of probe:library ing to pursue genomic discovery in specific cancer types. Their aim
hybrids by mixing in streptavidin-​coated magnetic beads to bind is to have 50 cancer types studied from over 25,000 patients. Most
the biotin and subsequently applying a magnetic field. Captured ICGC projects use whole genome sequencing (WGS) to characterize
library fragments are reclaimed by denaturing them away from the the genome. The two projects also are cooperating in a pan-​cancer
probes, after which they are quantitated and sequenced to provide analysis, currently underway, that includes data from over 2000 can-
the exome data. In practice, hybrid capture is performed for each cers across many tissue sites that were profiled by WGS, RNAseq,
tumor and matched normal exome, whereby increasing throughput and other types of next-​generation sequencing (NGS)–​based analyses
on sequencing instruments has enabled pools of hybrid capture frag- as well as proteomics (www.dcc.icgc.org). Cumulatively, these large
ments from many such exomes to be mixed prior to hybrid capture projects have effectively defined the catalog of cancer-​specific changes
and sequencing. The use of a DNA barcode on each library adapter that occur across most common and some rare tissue sites, and have
set permits downstream computational binning of sequences derived led to remarkable discoveries of the genetic contributors to can-
from individual patient samples for separate alignment and analysis cer’s development. Another large-​scale project, the Pediatric Cancer
of somatic alterations. In essence, the exome capture technique pro- Genome Project (PCGP; https://​www.stjude.org/​research/​pediatric-​
vides a focused and readily interpretable data set that is smaller and cancer-​genome-​project.html) was a privately funded collaborative
less expensive to produce than a whole genome data set and is easier project carried out by St. Jude Children’s Research Hospital and the
to analyze and interpret. The disadvantage of exome sequencing is Genome Institute at Washington University. This 5-​year project, now
that structural variants are extremely difficult to identify, copy num- completed, focused on genomic discovery in pediatric cancer types
ber analyses are possible but at very low resolution, and regulatory and resulted in sequencing of nearly 1000 cancer and paired normal
variants are missed since they commonly lie in non-​coding regions samples by WGS in comparison to a matched normal. The final 2 years
of the genome. Further subsetting of the exome is possible only if a of this project included RNAseq of the tumor, and whole genome
discrete set of genes is of interest, rather than all known genes. For bisulfite sequencing data to elucidate somatic methylation changes.
example, hybrid capture reagents that target genes linked to can- While the matched normal genome is most commonly used as a
cer susceptibility are commercially available for research and also comparator to help define what is truly somatic, a separate analysis
are offered as commercial CLIA (Clinical Laboratory Improvement of only the normal genome has also provided insights regarding the
Amendments) assays. genetic susceptibility to cancer (Lu et al., 2015; Zhang et al., 2015).
 45

Genomic Landscape of Cancer 45


In addition to susceptibility, a data mining study of TCGA solid tis- regions, such as promoters or enhancers. Such regions have typically
sue germline exomes, derived from peripheral blood lymphocytes, not been identified by large-​scale cancer genomics studies, due to the
led to an interesting series of observations regarding the increased preponderance of exome versus WGS data, the difficulty of delineat-
presence of rare truncation variants and known hotspot variants in ing the putative functions of these loci, and their imprecisely defined
cancer genes in these blood normal (Xie M et al., 2014). The study boundaries and functional regulatory components. One large study
demonstrated a positive correlation between blood-​specific mutations that attempted to better define the regulatory genome of human is
in leukemia/​lymphoma genes and age of the individuals, and deter- ENCODE (Encyclopedia of DNA Elements), which utilized human
mined that these mutations were detectable largely due to the growth cell lines including cancer cell lines (e.g., HeLa, K562) and a variety
advantage they conferred on the hematopoetic stem cell that carried of assays like chromatin immunoprecipitation (ChIP) and RNAseq,
them and its subsequent clonal expansion. These results were verified to characterize the functional portions of the non-​ coding genome
in a non-​cancer data set from the National Heart, Lung, and Blood (Consortium, 2012). These assignments are based on the evaluation of
Institute (NHLBI) exomes study’s WHISP (Women’s Health Initiative protein: DNA binding for specific transcription factors, on chromatin
Sequencing Project) cohort. McCarroll’s group reported a similar packaging by different histones with differential methylation status,
finding in 10% of persons older than 65 when analyzing exome data and by correlative analyses that link different DNA configurations to
for 12,380 Swedish persons that were blood derived (Genovese et al., the resulting RNA expression data for the genes at these loci. Further
2014). Here, health outcomes analysis from 2 to 7 years following refinements of the ENCODE data have ensued, including a cancer-​spe-
blood sampling showed that clonal hematopoiesis was a strong risk cific functional analysis approach devised by Gerstein and colleagues
factor for subsequent hematologic cancer. (Khurana et al., 2013). Here, the initial approach combined ENCODE
regions with data from the 1000 Genomes Project individuals to define
regulatory regions that are particularly sensitive to mutation or that
Remaining Challenges clearly disrupt transcription factor binding. The resulting patterns
One clear challenge that accompanies the data deluge that is result- identified were then used to develop a computational tool (FunSeq)
ing from large-​scale cancer genomics data is a means to predict the to examine data from ~90 cancer whole genome sequences, revealing
functional consequences of the many discovered somatic variants on around 100 candidate non-​coding drivers. While the amount of data in
the final protein produced in the cancer cell. This conundrum has led whole genome data sets can provide large numbers of possible non-​
to improved computational algorithms that consider a variety of lines coding drivers using these approaches, in a refined version of FunSeq,
of evidence to predict functional consequences (Gonzalez-​Perez et al., called FunSeq2 (Fu et al., 2014), Gerstein and colleagues devised a
2013; Schroeder et al., 2014; Tamborero et al., 2013). Such methods prioritization scheme that reduces the number of highly likely func-
typically are implemented as a first classifier of functional predic- tional drivers one might need to pursue with biological studies to
tion for novel mutations without supporting evidence of the resulting support their ability to impact tumor biology. Certainly the increas-
mutant protein function, mainly because defining this impact by bio- ing numbers of whole genomes being sequenced in cancer discovery
logical measures is a daunting task, given the magnitude of the somatic efforts will improve the identification of significantly mutated regula-
alteration data that now exists and continues to grow. However, biolog- tory drivers in the future (Piraino and Furney, 2016). Perhaps the most
ical data that demonstrate the impact of altered amino acid sequence common regulatory alteration known to occur across multiple tissue
on protein function remain the gold standard. sites is in the promoter region of the TERT gene. This gene encodes a
ribonucleoprotein polymerase that maintains telomere ends by addi-
tion of the telomere repeat TTAGGG. The promoter mutations create
Functional Studies a new binding motif for Ets transcription factors and ternary complex
factors (TCFs) near the transcription start site of the gene, resulting
While newer tools for genome editing, such as TALENs and Crispr/​ in increased gene expression levels. Two groups initially reported
Cas9, are enabling scalable functional studies of specific mutations, the TERT promoter mutations: one studying a family with inherited
supporting data sets have been generated to provide resources for func- melanoma predisposition by linkage analysis approaches coupled with
tional predictions. Two early efforts that provided a resource for cor- NGS (Horn et al., 2013), and a second studying melanoma somatic
relating cancer cell genome mutations and function were published alterations by NGS-​based WGS approaches (Huang et al., 2013). This
in 2012. One study was the Cancer Cell Line Encyclopedia (CCLE), provides an interesting and likely not unique example of how con-
published by Garraway and colleagues (Barretina et al., 2012). This stitutional and somatic alterations can lead to the same disease and
study compiled gene expression, chromosomal copy number, and highlights yet another discovery from large-​scale comparative cancer
exome sequencing data from 947 cancer cell lines and released the genomics: namely, the large degree of overlap between germline sus-
data to public availability. Furthering the correlation of mutational pro- ceptibility genes and their mutations with the common somatically
files and drug responsiveness, these authors also studied 24 anticancer mutated genes in human cancer genomes.
drugs across 479 of the cell lines to catalog drug sensitivity in the To this end, Snyder and colleagues have evaluated WGS data
context of genotype and gene expression-​based prediction. Coincident from 436 patients in TCGA to identify point mutations in regulatory
publication of a similar study of 639 human tumor cell lines by Benes regions with evidence for positive selection in transcription factor
and colleagues (Garnett et al., 2012) determined the genotypes of 64 binding sites. Their efforts have identified recurrently mutated sites
commonly mutated cancer genes obtained by NGS, with copy num- across these individuals that include the known non-​coding regulatory
ber analysis from single nucleotide polymorphism (SNP) microarrays mutations at the TERT promoter, as well as several new sites (Melton
and expression profiling by microarrays. In this study, 130 drugs were et al., 2015).
screened to determine drug sensitivity and dose–​response curves. One
challenge in sequencing cancer cell lines is that the comparator nor-
mal cell line or DNA source is largely not available, so the resulting RNA Expression Resource
genotypes are no doubt predominantly, but not exclusively, somatic. One final resource worth mentioning in the context of correlative
genomic and RNAseq data is the Genotype-​Tissue Expression (GTEx)
resource (https://​commonfund.nih.gov/​GTEx/​index) that aims to
Regulatory Region Functions provide a combination of genomic data and RNAseq-​based expres-
Beyond functional aspects of mutations relating to the resulting pro- sion data and analyses from healthy human tissues. This consortium
teins, there are higher-​level impacts of somatic mutations that lie out- recently published their pilot analysis of gene regulation in humans
side of known gene coding sequences in the genome. Namely, some (Consortium, 2015), presenting their analysis of RNA sequencing data
mutations may alter RNA expression and hence interfere with normal in the context of tissue site and genotype from 1641 samples across 42
cellular control of growth and division by disrupting gene regulatory tissues from 175 individuals. While the clear aim of this resource is to
46

46 Part I:  Basic Concepts


help put genomic variation into context relative to its impact on gene primary and recurrent disease can be striking, especially taken in the
expression, GTEx data already have proved valuable as a resource by context of a specific treatment. One example is shown in Figure 4–2,
which to gauge the cancer-​specific RNAseq expression data that often comparing an AML patient genome at diagnosis and after treatment
are lacking comparative RNAseq data from an adjacent non-​malignant by induction chemotherapy, consolidation therapy, and a greater than
tissue. 1-​year disease-​free remission. As evidenced by Figure 4.2, the recur-
rent disease is significantly less heterogeneous than the primary, where
one minor subclone has survived the chemotherapy regimen and has
DEEPER INSIGHTS FROM CANCER GENOMICS emerged with additional genomic alterations as the predominant clone
in the recurrent disease presentation (Ding et al., 2012).
In addition to advanced computational methods and visualization
Genomic Heterogeneity
tools (Grundberg et al., 2013; Hajirasouliha et al., 2014; Miller et al.,
Beyond enhancing our understanding of the genomic underpinnings 2014; Nik-​Zainal et al., 2012; Roth et al., 2014) to calculate and dis-
of cancer, the use of NGS in these studies has shed light on aspects play genomic heterogeneity, the pursuit of heterogeneity in cancer has
of cancer that were long suspected, based on other data, including resulted in a multitude of interesting findings. Studies of intratumoral
microscopy-​based staining (e.g., immunohistochemistry). One such heterogeneity have evaluated the comparative clonal architecture of
area is genomic heterogeneity in cancer samples. While it is intuitive tumors from the same tissue site, sampled at different locations in
that as cancer cells rapidly grow and divide during disease progres- the tumor, and also have evaluated the somatic mutations for their
sion, daughter cells will acquire, in addition to the initial set of found- likelihood of response to a targeted therapy (a.k.a. “druggability”)
ing mutations that led to the cancer phenotype, new mutations that (Gerlinger et al., 2012; Gerlinger et al., 2014; Haffner et al., 2013;
may confer additional growth advantage, may be neutral, or may aid McGranahan et al., 2015; Zhang et al., 2014). The latter provides
in treatment resistance once therapy begins. The unique characteristic insights into whether single core or needle biopsy assays for targeted
of NGS data that has led to improved abilities to identify such genomic therapy susceptibility are likely to provide sufficient information upon
heterogeneity is the digital nature of the data. In particular, when DNA which to base treatment decisions, and establishes the proportion of
is isolated from a tumor mass, the isolate contains genomic representa- variants identified or missed based on single versus multiple samples/​
tion from each cell’s genome in that mass. Since each DNA fragment assays of different tissue biopsies. While such studies have only been
in this isolate contributes to the sequencing library, and each result- conducted in a limited number of tissue sites, in general most studies
ing sequencing read originates from one unique genomic fragment, have advocated for more than one biopsy being assayed per tumor in
the data are in essence quantitative, or digital in nature. One way to the clinical setting (i.e., druggability).
illustrate this is by evaluating copy number altered regions, where the Intertumoral heterogeneity data have emerged as a natural conse-
magnitude of the amplification is exactly proportional to the (-​fold) quence of thousands of tumors being studied by NGS-​based genomics
increase in data coverage of the amplified chromosomal region once and have illustrated the myriad combinations of driver mutations,
the data are aligned to the reference genome (Figure 4–1). In the con- tumor suppressor alterations, germline susceptibility, and other non-​
text of genomic heterogeneity, we can simply examine the level of data DNA-​ based (methylation, chromatin configuration, RNA over-​or
coverage for each identified somatic variant and calculate the number underexpression absent obvious copy number alteration) somatic
of sequencing data reads that represent the variant and those that rep- changes that lead to cancer. As intertumoral heterogeneity studies have
resent the wildtype (or unmutated) allele. The percentage of variant-​ progressed and have begun to compare primary to recurrent disease
containing reads for each variant is referred to as the variant allele changes in heterogeneity (de Bruin et al., 2014; Johnson et al., 2014;
fraction (VAF), whereby the founder variants carried in every cancer Xie T et al., 2014) across multiple patients’ samples, we have come to
cell have a VAF of around 50% (since most variants are heterozygous). understand that primary tissue sites can display a panoply of combina-
Using a variety of approaches to group or cluster all identified variants tions of different subclones in the temporal samples taken at metasta-
with shared VAFs, the heterogeneity of the cancer cell population con- sis, most of which are discernable in the primary tissue. This temporal
tributing to the genomic DNA isolate can be characterized. Estimates comparison of heterogeneity becomes increasingly interesting in the
of genomic heterogeneity are improved by increased coverage depth, context of targeted therapy and acquired resistance (Awad et al., 2013;
which enables more sensitive detection of low proportion cell popu- Juric et al., 2015; Katayama et al., 2012; Pao et al., 2005; Poulikakos
lations or “subclones” and by increasing numbers of variants, where et al., 2011; Sequist et al., 2011; Wagle et al., 2011). One critical clin-
WGS with high coverage at variant sites yields higher confidence clus- ical question that remains to be answered is whether primary tumors
tering of VAFs due to the larger number of somatically mutated sites. that are more heterogeneous predict a worse outcome for the patient
Temporal or spatial changes in heterogeneity also can be evaluated in the setting of a consistent therapeutic approach to the disease (e.g.,
if the appropriate samples exist. For example, the differences between standard of care). Large-​scale analyses across cancer types indicate

Figure 4–1.  An indicated HER2 locus amplification from whole genome sequencing (WGS) of a human breast cancer genome. The panel provides data on
the focal amplifications along chromosome 17. This patient had a known HER2 amplification by conventional clincopathologic assay (HER2 fluorescent
in situ hybridization).
 47

Genomic Landscape of Cancer 47

Figure 4–2.  Changes in tumor heterogeneity over time. This figure illustrates the changes in AML tumor cell genomes in the primary compared to the
relapse population. Effectively, the initial or founder mutations develop in a hematopoetic stem cell (HSC) to provide a growth advantage. As this popula-
tion of cells rapidly grows and divides, additional mutations are added to a subpopulation of cells, creating a subclone. Each subclone of cells carries a
genotype that includes the founder clone mutations and mutations unique to that subclone. After chemotherapy treatment, the patient suffers a relapse that
represents a minor subclone from the primary disease presentation that has acquired additional mutations and a chromosomal translocation, expanding to
represent the single homogeneous cell population in the relapse disease.

that heterogeneity can impact outcomes (Andor et  al., 2016; Morris with the typical clinical trial and drug approval paradigm by which
et  al., 2016). Understanding the role of heterogeneity in the context therapies are tested one tissue site at a time. As evidence mounts for
of outcome for similarly treated cancers may be impactful within the the shared nature of alterations in specific cancer genes across tissue
clinical setting of disease treatment, especially in the context of tar- sites (Cancer Genome Atlas Research et  al., 2013; Hoadley et  al.,
geted therapy and acquired resistance. 2014; Leiserson et al., 2015), the practice of treating cancer based on
data from a genotyping assay to help determine the treatment for each
patient is becoming increasingly common. For example, 3%–​4% of
Single Cell Studies human breast cancers are driven by point mutations in HER2 rather
One technology-​based pursuit of genomic heterogeneity that has been than by its amplification (Bose et al., 2013), and 1%–​2% of prostate
enabled by NGS and specific devices is the isolation and characteri- cancers are driven by the BRAF mutations so common in human mela-
zation of the genomes of single cells. This interesting question about nomas (Palanisamy et  al., 2010). By profiling somatic alterations in
how genomic heterogeneity can be characterized at the single cell level tumor tissues, we also have discovered that several known cancer sus-
is a technical challenge, since the amount of DNA from each cell is ceptibility genes (BRCA1/​2, TP53, NF1) are also germline mutated
below the limit of the amount required to make an NGS library or in multiple tissue sites (Lu et al., 2015; Schrader et al., 2016; Zhang
even for site-​specific PCR and sequencing of known hotspot muta- et al., 2015). This fact emphasizes the need for comparing tumor to
tions, for example. As a result, these studies require an initial step of normal DNA in cancer genotyping assays because the nature of the
whole genome amplification (WGA) to increase the amount of geno- mutation is critically important for determining both treatment and the
mic DNA prior to library construction. However, the WGA procedure need for genetic counseling (Jones et al., 2015).
is enzymatic and thus introduces biases during amplification that result While the individual combinations of altered genes in any one human
in semi-​random areas that are not amplified completely (“amplifica- cancer are highly unique, by layering the information about genes that
tion dropout”) (Navin, 2014). Initially, yields from WGA were insuf- are activated or inactivated by their alterations onto known biological
ficient for high coverage sequencing suitable for mutation detection, pathways, we can identify that there is a common set of biological
so only copy number-​based comparisons were possible (Navin et al., pathways that are frequently altered in cancer. Initially described in
2011). As library methods have improved to enable low DNA input, 2001 (Hanahan and Weinberg, 2000), our growing knowledge about
sufficient depth of coverage for mutation calls has resulted (Hughes cancer genomics that emerged from large-​scale studies resulted in the
et al., 2014; Wang et al., 2014). However, although tens to hundreds of addition of several new pathways in a revised “hallmarks” manuscript
cells from each tumor typically are studied, it is often unclear whether in 2011 (Hanahan and Weinberg, 2011).
mutations that are not seen in every cell are representing heterogene-
ity or are false negative results due to amplification dropout. While
increasing numbers will add statistical confidence to estimates of het- DNA Damage
erogeneity, the cost of single cell sequencing across hundreds of cells
Another discovery from large-​scale genomic studies is the identifi-
calls into question the value of this approach when compared to high-​
cation of DNA damage signatures that can be attributed to specific
depth NGS coverage of mutated sites in DNA isolated from a tumor
mutagens or mutational influences. In particular, the DNA from
mass, as discussed earlier (Hughes et al., 2014).
smoking-​associated lung cancers has not only a characteristic signa-
ture from benz-​pyrene adduct formation with the DNA, but also an
Pan-​Cancer Genomics extraordinarily elevated mutation number across the genome due to
the long-​term exposure typical of these patients (Pleasance et  al.,
Large-​ scale cancer genomics discovery also has fundamentally 2010). Never-​ smokers, by contrast, have relatively few mutations
changed our notions about the uniqueness of cancer in terms of its in their tumor genomes (Govindan et al., 2012). Similarly, UV light
tissue site specificity. While tissue biology remains an important con- leaves a characteristic pattern of damage due to thymine dimerization
sideration to treatment paradigms (Prahallad et  al., 2012), we also and repair, and hence UV-​induced melanomas and basal cell carcino-
know that many cancer genes are frequently mutated in different tissue mas have the highest mutation numbers of all cancers due to lifelong
sites, and in some cases, cancers driven by the same mutations will sunlight exposure (Alexandrov et al., 2013). By contrast, melanomas
respond to treatment with an appropriate targeted therapy. In princi- in protected areas do arise (e.g., bottoms of feet) yet have a paucity of
ple, the shared nature of druggable targets across tissue sites conflicts mutations and no characteristic signature of UV damage. In studies
48

48 Part I:  Basic Concepts


comparing treatment-​naive primary to post-​treatment recurrent tumors Double mutations 3%
where a DNA-​damaging chemotherapy was utilized following the EML4-ALK BRAF 2%
primary diagnosis, several studies have demonstrated a characteristic 8%
increase in a specific DNA damage motif or signature (Ding et  al.,
2012; Johnson et al., 2014) in the post-​therapy tumor genome, as well
as an elevated mutation number.
Unknown

Targeting the Cancer Genome KRAS 25%


Coincident with our increasing knowledge of genomic alterations that EGFR 17%
result in cancer development or progression is the increasing shift
away from broad-​spectrum cytotoxic chemotherapy toward target-
ing specific genes known to frequently develop mutations that acti-
vate the corresponding protein, disrupt normal cellular processes and
controls, and drive cancer development. In this approach, designing
small molecule inhibitors to specifically bind to and inhibit the acti-
vated variant protein (or protein family members) can bring dramatic Unknown/no mutation detected EGFR KRAS EML4-ALK
results to metastatic cancer patients with a significant disease bur- Double mutations BRAF MET ROS1 MET
den. Initially, the targets of many small molecule inhibitors were not NRAS AKT1 PIK3CA HER2 MEK1
known, but in 2004, three studies were published that illustrated how
genomic approaches could help to identify markers of response. Here,
the research groups utilized the newly completed human genome Figure  4–3. Genomic drivers of cancer development in non-​small-​cell
sequence, which had provided precise locations and annotations to adenocarcinoma of the lung. The pie graph illustrates that a large propor-
many of the cancer-​related tyrosine kinase oncogenes. Their work tion of the genomic drivers of NSCLC have been identified by large-​scale
studied specific tyrosine kinase genes in small numbers of patients cancer genomics discovery efforts. Many of the driver alterations can be
who had enrolled in clinical trials and either did or did not respond to targeted by specific therapies.
small molecule tyrosine kinase inhibitor drugs. These studies identi-
fied that most lung non-​small-​cell adenocarcinoma (NSCLC) patients practical challenges do still remain with respect to this molecular pro-
with a response to a tyrosine kinase inhibitor carried specific somatic filing paradigm. In particular, not all metastatic sites are readily biop-
mutations in the tyrosine kinase domain of the gene encoding epider- sied, and insurance may not cover the cost of re-​biopsy if there is no
mal growth factor receptor (EGFR) (Lynch et  al., 2004; Paez et  al., demonstrated clinical benefit. Also, while the number of targeted ther-
2004; Pao et  al., 2004). While these were painstaking studies done apies is growing, the number of gene targets remains relatively small.
by PCR amplification and sequencing of suspect target genes, the Furthermore, depending upon the number of genes or sites tested, only
widespread use of massively parallel sequencing has vastly increased around 25%–​40% of patients have a clearly indicated therapeutic tar-
our understanding of target genes, mutations, and their relationship to get in their tumor genome that might qualify them to be treated with
therapeutic response (Dienstmann et  al., 2015; Meador et  al., 2014; that drug or class of drugs. If a trial is available and accruing patients,
Wagner et al., 2016), and even has identified frequent genetic changes they may not be eligible due to other factors, such as their performance
that result in acquired resistance to small molecule inhibitor therapies. status, the number of prior therapies, or other qualification metrics. If
In particular, these approaches have accelerated the identification of they are eligible, patients sometimes do not have the financial means
patients most likely to respond to a targeted therapy such that first-​line to pay for their own costs of travel and hotel stays required to partici-
treatment with a tyrosine kinase inhibitor in newly diagnosed NSCLC pate in the trial.
patients carrying the corresponding EGFR mutations is now FDA
approved in the United States. In fact, the large number of somatic
alterations now recognized for driving NSCLC development and their CANCER EPIGENOMICS
corresponding therapies have made the case for testing multiple genes
and fusion gene/​protein drivers in patients diagnosed with the disease Another significant area of large-​scale research has been to under-
(Figure 4–3) using NGS and computational analysis to detect drug- stand functionally relevant changes to the genome that do not involve
gable gene targets. a change in the nucleotide sequence but can ultimately alter gene
expression, such as DNA methylation or histone modifications, yet
are essential for fully understanding many biological processes and
Diagnostic Multi-​Gene Assays disease states. This can be exemplified by international collaborations
Such multi-​gene assays or panels take on a wide ranging number of such as The ENCODE (http://​www.genome.gov/​encode/​) Consortium
genes and different types of somatic alterations that can be remarka- and the National Institutes of Health (NIH) Roadmap Epigenetics
ble in their scope and breadth, from testing tens of genes to all known Project (http://​www.roadmapepigenomics.org). These pioneering
human genes (the exome) and assaying for all mutation types from efforts highlight how epigenetic information complements genotype
point mutations to structural rearrangements that produce targetable and expression analysis and its potential importance in cancer. They
fusion gene drivers (Hyman et al., 2015; Siu et al., 2015; Van Allen further emphasize the need for discovery genomics centered on epi-
et al., 2014). Clinically, these assays are more conservative of precious genetics (gene regulation) that has resulted in a variety of novel tech-
tumor tissue biopsies than performing multiple single-​gene tests, and niques coupled with specific analytical methods according to different
the results are more quickly and cheaply obtained. Furthermore, given epigenetic marks that can be evaluated. Here we highlight several
the relatively large number of frequently mutated genes now identi- recent technological advances that have facilitated high-​throughput
fied as being shared across multiple tissue sites, NGS-​based multiplex assessment of functional regulatory elements that may act across a
gene assays are achieving analytical and clinical validity as diag- range from proximal (i.e., within the promoter region) to distant, and
nostics. Downsides include the fact that these assays also will reveal provide examples of their application to cancer epigenomic discovery.
so-​called VUS (variants of unknown significance) in genes that are
cancer-​related yet for which the impact of the variant, especially on
drug response, is unknown for that tissue site (or for any site) (Andre
DNA-​Protein Binding
et al., 2014; Van Allen et al., 2014). Reimbursement of the costs asso- Currently, there are numerous strategies to interrogate chromatin
ciated with these assays also has been slower to emerge, and more biology. One of the initial applications for determining how proteins
 49

Genomic Landscape of Cancer 49


interact with DNA to regulate gene expression is chromatin immuno- binding following hormone therapy increases chromatin accessibility
precipitation (ChIP) coupled with massively parallel DNA sequenc- (Tewari et al., 2012).
ing, referred to as ChIP-​Seq. To date, it has been widely used to
map precisely the genome-​wide binding sites for proteins (transcrip-
tion factors, modified histones, RNA polymerase, etc.). As one of Distant Chromatin Conformation Epigenomics
the earlier applications developed for epigenetic discovery, there To this point we have focused on applications commonly used for
already exist ChIP-​seq guidelines and best practices released by understanding regulatory processes occurring in close proximity to a
ENCODE (Landt et al., 2012) to maximize performance. Likewise, gene. However, recent advances have made it possible to understand the
there are numerous open-​source tools for ChIP-​seq data analysis contribution of the three-​dimensional organization of chromatin to reg-
(Fejes et al., 2008; Qin et al., 2010; Zhang et al., 2008). However, ulating gene expression. The first such high-​throughput method, called
for the remainder of applications discussed, often the availability of chromatin conformation capturing (3C), is used to study the nuclear
experimental and analytical resources is much more limited. A sig- organization in the native cellular state ((Miele et al., 2006); Splinter et
nificant advancement in the application of ChIP-​Seq has been the al., 2004). In brief, 3C involves (1) cross-​linking proteins and DNA that
transition from initial cancer studies focusing on cell lines to study- are in close proximity in the nuclear space, (2) a restriction digest to
ing the epigenomes of human tissues. This transition enables gen- separate cross-​linked from non-​cross-​linked DNA fragments, (3) DNA
erating epigenetic maps across patient cohorts to understand how ligation under conditions favorable to ligate junctions between cross-​
epigenetic changes correlate with clinical and epidemiological data. linked DNA fragments, (4) reversal of cross-​links, and (5) quantifica-
Such an approach is exemplified by a breast cancer study focused on tion (PCR, microarray, high-​throughput sequencing). This method also
understanding the differences in estrogen receptor (ER) binding in spurred the development of circularized chromosome conformation
primary breast cancers during the progression to distant metastases. capture (4C) (Zhao et al., 2006) and carbon-​copy chromosome confor-
Notably, the resulting data demonstrated that patients resistant to ER mation capture (5C) (Dostie and Dekker, 2007). One advantage of 4C
agonist therapies still have active recruitment of ER to chromatin. is that it enables a genome-​wide screen for interactions with a specific
Additionally, patients more likely to progress to metastatic disease locus of interest; 5C builds upon this approach by enabling parallel
acquired ER binding to unique genomic regions not bound by ER in analysis of interactions with a selected set of loci. The application of
patients reported to have good clinical outcomes (Ross-​Innes et al., these technologies can be witnessed by our improved understanding
2012). Overall, this study demonstrates the biological and clinical of hormone regulation in prostate and breast cancer. In the context of
importance of studying the epigenome in broader patient populations breast cancer, it was observed that estrogen is able to induce chromatin
with corresponding clinical data. “looping” by bringing two genes not normally located near one another
in the genome into close spatial proximity (Hu et al., 2008). Such find-
ings have been replicated in prostate cancer studies that couple 3C with
Proximal Chromatin Conformation Epigenomics ChIP (ChIP-​3C), to explore AR-​induced chromatin “looping.” Multiple
studies have reported distal AR binding sites capable of regulating
As chromatin is tightly packaged into an array of nucleosomes, the nucle- well-​known target genes through chromatin looping, some of which
osome positioning plays a significant regulatory function by facilitating have been shown to be directly involved in promoting cell growth in
the “openness” or accessibility of binding sites to transcription factors castration-​resistant prostate cancer (Chen et al., 2011; Wang et al.,
(TFs) and to the general transcription machinery. Therefore, chromatin 2007, 2009). In addition to gene regulation, AR has been implicated
accessibility assays have been developed to correlate regulatory DNA in the formation of gene fusions, which commonly occur in prostate
sequences that coincide with open or accessible genomic DNA. In con- cancer. Notably, androgen treatment appears to induce the co-​localiza-
trast to ChIP-​seq, chromatin accessibility assays do not require anti- tion of TMPRSS2 and ERG, to generate the most common gene fusion
bodies specific for the DNA binding protein of interest, and the results in prostate cancer, and also activates TMPRSS2 transcription (Mani
can directly report the effect of chromatin structure alterations on gene et al., 2009; Wang et al., 2009). Taken together, a deeper understand-
transcription. Currently, there are three common methods to assay open ing of three-​dimensional chromatin organization has offered profound
chromatin. The first approach, DNase-​seq, allows for the global discov- insights into gene regulation.
ery of nucleosome-​free genomic regions that are hypersensitive to nucle-
ase treatment with the non-​specific double strand endonuclease DNase I,
also known as DNase I hypersensitive sites (DHSs) (Boyle et al., 2011;
Hesselberth et  al., 2009). The second method, formaldehyde-​assisted
Methylation Epigenomics
isolation of regulatory elements, or FAIRE-​Seq (Bianco et  al., 2015), DNA methylation is a process by which methyl groups are added to
uses phenol-​chloroform to separate nucleosome-​bound from free geno- cytosine nucleotides in DNA and is an important regulator of gene
mic DNA. The last method, assay for transposase-​accessible chromatin transcription. Aberrant DNA methylation patterns in cancerous tis-
with high-​throughput sequencing (ATAC-​seq), measures open chroma- sue genomes by comparison to normal tissues have been identified as
tin through the ability of hyperactive Tn5 transposase to integrate into recurrent in a large number of human malignancies. The two common
active or open chromatin regions (Buenrostro et al., 2015). As the most states are (1) hypermethylation, or enhanced DNA methylation in the
recently introduced approach, ATAC-​seq has been used the least to date. promoter region, which results in gene silencing; and (2) hypometh-
However, it is an appealing strategy due to the simplicity of the method, ylation, or reduced methylation, which results in gene overexpression
its sensitivity, and the fact that the method does not require a large quan- and globally has been associated with tumor development and progres-
tity of genomic DNA, which is often a limiting factor when studying sion. Unlike many genetic mutations in cancer, aberrant DNA meth-
clinical samples. While the application of these assays has primarily ylation can be reversed, therefore making it an appealing therapeutic
focused on single gene studies in cell lines, already the results of these target. Currently there are a number of approaches for identifying
studies have led to the discovery of critical epigenetic changes respon- methylation changes that can be divided into three main categories.
sible for differential gene expression in cancers. For instance, by inte- DNA is either pretreated with a restriction enzyme, is enriched for
grating DNase I hypersensitivity and expression data, we have gained a methylated DNA using an affinity based approach (i.e., anti-​methyl-
deeper understanding of the interplay between chromatin accessibility cytosine antibodies), or is treated with sodium bisulphite to convert
and androgen receptor (AR) function, which is particularly important in unmethylated cytosines to thymines. Initial high-​throughput methods
prostate cancer. As expected, AR binding is associated with chromatin utilized arrays to monitor methylation changes (i.e., MeDIP [Weber
accessibility, and ultimately this affects gene expression following hor- et al., 2005]; MCA [Estecio et al., 2007]; MIRA [Rauch et al., 2006])
mone induction. However, in contrast to other previously studied DNA-​ before being adapted for NGS (i.e., MeDIP-​Seq [Harris et al., 2010];
binding proteins, AR binding appears to have multiple modes. In fact, and MIRA-​Seq [Choi et al., 2010]). Whole genome shotgun bisulfite
half of the AR binding sites identified by this study were not accessible sequencing (WGBS-​seq) is currently the most exhaustive approach for
to DNase I  cleavage prior to hormone induction, suggesting that AR monitoring the whole methylome landscape, although it is also the
50

50 Part I:  Basic Concepts


most costly. WGBS-​seq has been successfully used to map the com- CONCLUSIONS
plete methylomes of several cancer samples (Berman et al., 2012;
Hansen et al., 2011), demonstrating that hypermethylated domains Taken together, the combination of clever molecular biology
occur primarily at CpG islands and are concentrated within regions approaches with massively parallel sequencing and genome-​ scale
of long-​range hypomethylation that span across greater than 100 kb. analyses have provided a rapid and remarkable improvement in our
These same hypomethylated regions coincide with attachment sites to understanding of the role of the genome in cancer development and
the nuclear lamina, leading to the hypothesis that widespread DNA progression. This chapter has attempted to introduce some of these
methylation changes in cancer cells are linked to gene silencing pro- approaches and the resulting breadth of knowledge gained from them
grams according to three-​dimensional chromatin organization in the to date. There is undoubtedly still much to learn. However, we already
nucleus (Berman et al., 2012). are seeing evidence that these methods and the results obtained from
them are beginning to make inroads to large-​scale epidemiological
studies, to our understanding of how environmental factors such as
Long Non-​Coding RNA Regulation exposure to known carcinogens and mutagens are represented in the
For decades, cancer genetics research has largely focused on the impor- resulting tumor DNA, and to further defining the correlation between
tance of protein-​coding genes, such as oncogenes and tumor suppres- epigenetic changes and cancer development.
sors that can serve as biomarkers and therapeutic targets. The more
recent discovery of microRNAs (miRNAs) and of long non-​coding References
RNAs (lncRNAs) have led to multiple discoveries regarding the role of Alexandrov LB, Nik-​Zainal S, Wedge DC, et  al. 2013. Signatures of muta-
non-​coding RNAs in gene regulation. LncRNAs are typically greater tional processes in human cancer. Nature, 500(7463), 415–​ 421.
than 200 nucleotides in length, transcribed with RNA polymerase II, PMCID: PMC3776390.
and produce transcripts that are 5' capped, polyadenylated, spliced Andor N, Graham TA, Jansen M, et al. 2016. Pan-​cancer analysis of the extent
(like mRNAs), lack coding potential, and only recently have emerged and consequences of intratumor heterogeneity. Nat Med, 22(1), 105–​113.
as critical regulators in tumorigenesis. As part of the ENCODE proj- PMID: 26618723.
ect, the GENCODE consortium has already manually curated 9227 Andre F, Mardis E, Salm M, et al. 2014. Prioritizing targets for precision cancer
medicine. Ann Oncol, 25(12), 2295–​2303. PMID: 25344359.
human lncRNAs (Derrien et  al., 2012). However, by leveraging the Awad MM, Engelman JA, and Shaw AT. 2013. Acquired resistance to crizo-
unbiased view of RNA transcription products offered by transcriptome tinib from a mutation in CD74-​ROS1. N Engl J Med, 369(12), 1173.
sequencing (RNA-​Seq), many additional lncRNAs have been detected, PMID: 24047072.
resulting in the annotation of greater than 50,000 expressed lncRNA Bainbridge MN, Wang M, Burgess DL, et al. 2010. Whole exome capture in solu-
genes, many of which had eluded discovery by microarray-​based strat- tion with 3 Gbp of data. Genome Biol, 11(6), R62. PMCID: PMC2911110.
egies (Cabanski et al., 2015; Iyer et al., 2015; Simonetti et al., 1989). Barretina J, Caponigro G, Stransky N, et  al. 2012. The Cancer Cell Line
Overall, the characterization of RNA species, elucidating their func- Encyclopedia enables predictive modelling of anticancer drug sensitivity.
tion, and assessing their clinical applicability have become a major Nature, 483(7391), 603–​607. PMCID: PMC3320027.
area of biological and clinical importance in cancer research. Berman BP, Weisenberger DJ, Aman JF, et  al. 2012. Regions of focal DNA
hypermethylation and long-​range hypomethylation in colorectal cancer
While many classes of non-​coding RNAs have been implicated coincide with nuclear lamina-​associated domains. Nat Genet, 44(1), 40–​
in cancer, lncRNAs are still largely unexplored and recently have 46. PMCID: PMC4309644.
emerged as a new aspect of biology. Several well-​described examples, Bianco S, Rodrigue S, Murphy BD, and Gevry N. 2015. Global mapping of
such as HOTAIR (Gupta et al., 2010) and ANRIL (Yap et al., 2010), open chromatin regulatory elements by formaldehyde-​assisted isolation
indicate that lncRNAs may be essential actors in cancer biology, of regulatory elements followed by sequencing (FAIRE-​seq). Methods
typically facilitating epigenetic gene repression through chromatin-​ Mol Biol, 1334, 261–​272. PMID: 26404156.
modifying complexes. Importantly, newer methods have enabled a Bose R, Kavuri SM, Searleman AC, et al. 2013. Activating HER2 mutations
deeper understanding of lncRNA-​based regulation, both by determin- in HER2 gene amplification negative breast cancer. Cancer Discov, 3(2),
ing the proteins with which different lncRNAs interact and where 224–​237. PMCID: PMC3570596.
Boyle AP, Song L, Lee BK, et al. 2011. High-​resolution genome-​wide in vivo
lncRNAs bind chromatin to epigenetically regulate gene expression. footprinting of diverse transcription factors in human cells. Genome Res,
There are two major strategic approaches that have developed over 21(3), 456–​464. PMCID: PMC3044859.
the past few years. The first strategy focuses on identifying spe- Buenrostro JD, Wu B, Chang HY, and Greenleaf WJ. 2015. ATAC-​seq:  a
cific proteins that interact with lncRNAs by using antibodies to pull method for assaying chromatin accessibility genome-​wide. Curr Protoc
down a protein complex and its associated lncRNAs. These methods Mol Biol, 109, 21–​29. PMCID: PMC4374986.
include RNA-​immunoprecipitation (RIP; Zhao et al., 2010) and UV-​ Cabanski CR, White NM, Dang HX, et  al. 2015. Pan-​cancer transcriptome
crosslinked immunoprecipitation (CLIP) coupled with transcriptome analysis reveals long noncoding RNAs with conserved function. RNA
sequencing (Murigneux et al., 2013). While the identification of pro- Biol, 12(6), 628–​642. PMCID: PMC4615893.
tein partners can implicate an lncRNA in a specific functional mecha- Cancer Genome Atlas Research N, Weinstein JN, Collisson EA, et  al. 2013.
The Cancer Genome Atlas Pan-​ Cancer analysis project. Nat Genet,
nism or biological process, it requires a priori knowledge of the protein 45(10), 1113–​1120. PMCID: PMC3919969.
to be studied and a corresponding high-​specificity antibody. In con- Chen Z, Zhang C, Wu D, et al. 2011. Phospho-​MED1-​enhanced UBE2C locus
trast, the second strategy takes an lncRNA-​centric approach by focus- looping drives castration-​ resistant prostate cancer growth. EMBO J,
ing on studying the interactions occurring with a specific lncRNA of 30(12), 2405–​2419. PMCID: PMC3116285.
interest. Here, oligonucleotides are designed to tile across the entire Choi JH, Li Y, Guo J, et  al. 2010. Genome-​wide DNA methylation maps in
target lncRNA transcript, enabling the selective hybridization of RNA follicular lymphoma cells determined by methylation-​enriched bisulfite
or DNA to the lncRNA. Subsequently, the lncRNA-​bound interactors sequencing. PLoS One, 5(9). PMCID: PMC2947499.
can undergo sequencing to identify either the RNA transcripts that Chu C, Qu K, Zhong FL, Artandi SE, and Chang HY. 2011. Genomic maps
interact with it or to reveal where the lncRNA was bound to chromatin of long noncoding RNA occupancy reveal principles of RNA-​chromatin
interactions. Mol Cell, 44(4), 667–​678. PMCID: PMC3249421.
throughout the genome. Consortium. 2012. An integrated encyclopedia of DNA elements in the human
Two widely adopted methods of this type include chromatin isola- genome. Nature, 489(7414), 57–​74. PMCID: PMC3439153.
tion by RNA purification (ChIRP; Chu et al., 2011) and RNA antisense Consortium. 2015. Human genomics. The Genotype-​Tissue Expression (GTEx)
purification (RAP; Engreitz et al., 2015). While largely similar meth- pilot analysis: multitissue gene regulation in humans. Science, 348(6235),
odologies, ChIRP uses 20-​mer oligonucleotide probes, whereas RAP 648–​660. PMCID: PMC4547484.
uses much longer probes (120 nucleotides). The RAP strategy was de Bruin EC, McGranahan N, Mitter R, et  al. 2014. Spatial and temporal
used to successfully elucidate how HOTAIR interacts with chromatin diversity in genomic instability processes defines lung cancer evolution.
to promote breast cancer metastasis (Gupta et al., 2010). Science, 346(6206), 251–​256. PMCID: PMC4636050.
 51

Genomic Landscape of Cancer 51


Derrien T, Johnson R, Bussotti G, et al. 2012. The GENCODE v7 catalog of Hodges E, Rooks M, Xuan Z, et al. 2009. Hybrid selection of discrete genomic
human long noncoding RNAs: analysis of their gene structure, evolution, intervals on custom-​designed microarrays for massively parallel sequenc-
and expression. Genome Res, 22(9), 1775–​1789. PMCID: PMC3431493. ing. Nat Protoc, 4(6), 960–​974. PMCID: PMC2990409.
Dienstmann R, Jang IS, Bot B, Friend S, and Guinney J. 2015. Database of Horn S, Figl A, Rachakonda PS, et  al. 2013. TERT promoter mutations
genomic biomarkers for cancer drugs and clinical targetability in solid in familial and sporadic melanoma. Science, 339(6122), 959–​ 961.
tumors. Cancer Discov, 5(2), 118–​123. PMID: 25656898. PMID: 23348503.
Ding L, Getz G, Wheeler DA, et  al. 2008. Somatic mutations affect key Hu Q, Kwon YS, Nunez E, et  al. 2008. Enhancing nuclear receptor-​induced
pathways in lung adenocarcinoma. Nature, 455(7216), 1069–​ 1075. transcription requires nuclear motor and LSD1-​ dependent gene net-
PMCID: PMC2694412. working in interchromatin granules. Proc Natl Acad Sci U S A, 105(49),
Ding L, Ley TJ, Larson DE, et  al. 2012. Clonal evolution in relapsed acute 19199–​19204. PMCID: PMC2593616.
myeloid leukaemia revealed by whole-​ genome sequencing. Nature, Huang FW, Hodis E, Xu MJ, et  al. 2013. Highly recurrent TERT pro-
481(7382), 506–​510. PMCID: PMC3267864. moter mutations in human melanoma. Science, 339(6122), 957–​ 959.
Dostie J, and Dekker J. 2007. Mapping networks of physical interactions PMCID: PMC4423787.
between genomic elements using 5C technology. Nat Protoc, 2(4), 988–​ Hughes AE, Magrini V, Demeter R, et  al. 2014. Clonal architecture of sec-
1002. PMID: 17446898. ondary acute myeloid leukemia defined by single-​cell sequencing. PLoS
Engreitz J, Lander ES, and Guttman M. 2015. RNA antisense purification Genet, 10(7), e1004462. PMCID: PMC4091781.
(RAP) for mapping RNA interactions with chromatin. Methods Mol Biol, Hyman DM, Solit DB, Arcila ME, et al. 2015. Precision medicine at Memorial
1262, 183–​197. PMID: 25555582. Sloan Kettering Cancer Center:  clinical next-​ generation sequencing
Estecio MR, Yan PS, Ibrahim AE, et  al. 2007. High-​throughput methylation enabling next-​ generation targeted therapy trials. Drug Discov Today,
profiling by MCA coupled to CpG island microarray. Genome Res, 20(12), 1422–​1428. PMID: 26320725.
17(10), 1529–​1536. PMCID: PMC1987348. International Human Genome Sequencing. 2004. Finishing the euchro-
Fejes AP, Robertson G, Bilenky M, et al. 2008. FindPeaks 3.1: a tool for identi- matic sequence of the human genome. Nature, 431(7011), 931–​945.
fying areas of enrichment from massively parallel short-​read sequencing PMID: 15496913.
technology. Bioinformatics, 24(15), 1729–​1730. PMCID: PMC2638869. Iyer MK, Niknafs YS, Malik R, et  al. 2015. The landscape of long noncod-
Fu Y, Liu Z, Lou S, et al. 2014. FunSeq2: a framework for prioritizing non- ing RNAs in the human transcriptome. Nat Genet, 47(3), 199–​ 208.
coding regulatory variants in cancer. Genome Biol, 15(10), 480. PMCID: PMC4417758.
PMCID: PMC4203974. Johnson BE, Mazor T, Hong C, et al. 2014. Mutational analysis reveals the ori-
Garnett MJ, Edelman EJ, Heidorn SJ, et al. 2012. Systematic identification of gin and therapy-​driven evolution of recurrent glioma. Science, 343(6167),
genomic markers of drug sensitivity in cancer cells. Nature, 483(7391), 189–​193. PMCID: PMC3998672.
570–​575. PMCID: PMC3349233. Jones S, Anagnostou V, Lytle K, et  al. 2015. Personalized genomic analyses
Genovese G, Kahler AK, Handsaker RE, et al. 2014. Clonal hematopoiesis and for cancer mutation discovery and interpretation. Sci Transl Med, 7(283),
blood-​cancer risk inferred from blood DNA sequence. N Engl J Med, 283ra253. PMCID: PMC4442685.
371(26), 2477–​2487. PMCID: PMC4290021. Juric D, Castel P, Griffith M, et  al. 2015. Convergent loss of PTEN leads to
Gerlinger M, Horswell S, Larkin J, et al. 2014. Genomic architecture and evolu- clinical resistance to a PI(3)Kalpha inhibitor. Nature, 518(7538), 240–​
tion of clear cell renal cell carcinomas defined by multiregion sequencing. 244. PMCID: PMC4326538.
Nat Genet, 46(3), 225–​233. PMCID: PMC4636053. Katayama R, Shaw AT, Khan TM, et al. 2012. Mechanisms of acquired crizo-
Gerlinger M, Rowan AJ, Horswell S, et al. 2012. Intratumor heterogeneity and tinib resistance in ALK-​rearranged lung Cancers. Sci Transl Med, 4(120),
branched evolution revealed by multiregion sequencing. N Engl J Med, 120ra117. PMCID: PMC3385512.
366(10), 883–​892. PMID: 22397650. Khurana E, Fu Y, Colonna V, et  al. 2013. Integrative annotation of variants
Gnirke A, Melnikov A, Maguire J, et al. 2009. Solution hybrid selection with from 1092 humans: application to cancer genomics. Science, 342(6154),
ultra-​long oligonucleotides for massively parallel targeted sequencing. 1235587. PMCID: PMC3947637.
Nat Biotechnol, 27(2), 182–​189. PMCID: PMC2663421. Landt SG, Marinov GK, Kundaje A, et al. 2012. ChIP-​seq guidelines and prac-
Gonzalez-​Perez A, Mustonen V, Reva B, et al. 2013. Computational approaches tices of the ENCODE and modENCODE consortia. Genome Res, 22(9),
to identify functional genetic variants in cancer genomes. Nat Methods, 1813–​1831. PMCID: PMC3431496.
10(8), 723–​729. PMCID: PMC3919555. Leiserson MD, Vandin F, Wu HT, et  al. 2015. Pan-​cancer network analysis
Govindan R, Ding L, Griffith M, et al. 2012. Genomic landscape of non-​small identifies combinations of rare somatic mutations across pathways and
cell lung cancer in smokers and never-​smokers. Cell, 150(6), 1121–​1134. protein complexes. Nat Genet, 47(2), 106–​114. PMCID: PMC4444046.
PMCID: PMC3656590. Ley TJ, Mardis ER, Ding L, et al. 2008. DNA sequencing of a cytogenetically
Grundberg I, Kiflemariam S, Mignardi M, et al. 2013. In situ mutation detec- normal acute myeloid leukaemia genome. Nature, 456(7218), 66–​72.
tion and visualization of intratumor heterogeneity for cancer research and PMCID: PMC2603574.
diagnostics. Oncotarget, 4(12), 2407–​2418. PMCID: PMC3926836. Ley TJ, Minx PJ, Walter MJ, et  al. 2003. A pilot study of high-​throughput,
Gupta RA, Shah N, Wang KC, et al. 2010. Long non-​coding RNA HOTAIR sequence-​based mutational profiling of primary human acute myeloid
reprograms chromatin state to promote cancer metastasis. Nature, leukemia cell genomes. Proc Natl Acad Sci U S A, 100(24), 14275–​
464(7291), 1071–​1076. PMCID: PMC3049919. 14280. PMCID: PMC283582.
Haffner MC, Mosbruger T, Esopi DM, et  al. 2013. Tracking the clonal ori- Lu C, Xie M, Wendl MC, et al. 2015. Patterns and functional implications of
gin of lethal prostate cancer. J Clin Invest, 123(11), 4918–​ 4922. rare germline variants across 12 cancer types. Nat Commun, 6, 10086.
PMCID: PMC3809798. PMCID: PMC4703835.
Hajirasouliha I, Mahmoody A, and Raphael BJ. 2014. A combinatorial Lynch TJ, Bell DW, Sordella R, et al. 2004. Activating mutations in the epidermal
approach for analyzing intra-​tumor heterogeneity from high-​throughput growth factor receptor underlying responsiveness of non-​small-​cell lung
sequencing data. Bioinformatics, 30(12), i78–​86. PMCID: PMC4058927. cancer to gefitinib. N Engl J Med, 350(21), 2129–​2139. PMID: 15118073.
Hanahan D, and Weinberg RA. 2000. The hallmarks of cancer. Cell, 100(1), Mani RS, Tomlins SA, Callahan K, et al. 2009. Induced chromosomal prox-
57–​70. PMID: 10647931. imity and gene fusions in prostate cancer. Science, 326(5957), 1230.
Hanahan D, and Weinberg RA. 2011. Hallmarks of cancer: the next generation. PMCID: PMC2935583.
Cell, 144(5), 646–​674. PMID: 21376230. Mardis ER. 2013. Next-​generation sequencing platforms. Annu Rev Anal Chem
Hansen KD, Timp W, Bravo HC, et al. 2011. Increased methylation variation (Palo Alto Calif), 6, 287–​303. PMID: 23560931.
in epigenetic domains across cancer types. Nat Genet, 43(8), 768–​775. McGranahan N, Favero F, de Bruin EC, et al. 2015. Clonal status of actionable
PMCID: PMC3145050. driver events and the timing of mutational processes in cancer evolution.
Harris RA, Wang T, Coarfa C, et  al. 2010. Comparison of sequencing-​based Sci Transl Med, 7(283), 283ra254. PMCID: PMC4636056.
methods to profile DNA methylation and identification of monoal- Meador CB, Micheel CM, Levy MA, et al. 2014. Beyond histology: translating
lelic epigenetic modifications. Nat Biotechnol, 28(10), 1097–​ 1105. tumor genotypes into clinically effective targeted therapies. Clin Cancer
PMCID: PMC2955169. Res, 20(9), 2264–​2275. PMCID: PMC4008689.
Hesselberth JR, Chen X, Zhang Z, et al. 2009. Global mapping of protein-​DNA Melton C, Reuter JA, Spacek DV, and Snyder M. 2015. Recurrent somatic
interactions in vivo by digital genomic footprinting. Nat Methods, 6(4), mutations in regulatory regions of human cancer genomes. Nat Genet,
283–​289. PMCID: PMC2668528. 47(7), 710–​716. PMCID: PMC4485503.
Hoadley KA, Yau C, Wolf DM, et al. 2014. Multiplatform analysis of 12 cancer Miele A, Gheldof N, Tabuchi TM, Dostie J, and Dekker J. 2006. Mapping chro-
types reveals molecular classification within and across tissues of origin. matin interactions by chromosome conformation capture. Curr Protoc
Cell, 158(4), 929–​944. PMCID: PMC4152462. Mol Biol, 74(21.11), 21.11.1–​21.11.20.
52

52 Part I:  Basic Concepts


Miller CA, White BS, Dees ND, et al. 2014. SciClone: inferring clonal archi- Simonetti I, Rossen JD, Winniford MD, and Marcus ML. 1989. Biphasic
tecture and tracking the spatial and temporal patterns of tumor evolution. effect of nitroglycerin on coronary hemodynamics in normal subjects. Z
PLoS Comput Biol, 10(8), e1003665. PMCID: PMC4125065. Kardiol, 78(Suppl 2), 52–​55; discussion 64–​57. PMID: 2511694.
Morris LG, Riaz N, Desrichard A, et al. 2016. Pan-​cancer analysis of intratu- Siu LL, Conley BA, Boerner S, and LoRusso PM. 2015. Next-​generation
mor heterogeneity as a prognostic determinant of survival. Oncotarget, 7, sequencing to guide clinical trials. Clin Cancer Res, 21(20), 4536–​4544.
10051–​10063. PMCID: PMC4891103. PMCID: PMC4610034.
Mortazavi A, Williams BA, McCue K, Schaeffer L, and Wold B. 2008. Mapping Splinter E, Grosveld F, and de Laat W. 2004. 3C technology:  analyzing the
and quantifying mammalian transcriptomes by RNA-​Seq. Nat Methods, spatial organization of genomic loci in vivo. Methods Enzymol, 375, 493–​
5(7), 621–​628. PMID: 18516045. 507. PMID: 14870685.
Murigneux V, Sauliere J, Roest Crollius H, and Le Hir H. 2013. Transcriptome-​ Tamborero D, Gonzalez-​Perez A, Perez-​Llamas C, et al. 2013. Comprehensive
wide identification of RNA binding sites by CLIP-​seq. Methods, 63(1), identification of mutational cancer driver genes across 12 tumor types. Sci
32–​40. PMID: 23545196. Rep, 3, 2650. PMCID: PMC3788361.
Navin N, Kendall J, Troge J, et al. 2011. Tumour evolution inferred by single-​ Tewari AK, Yardimci GG, Shibata Y, et al. 2012. Chromatin accessibility reveals
cell sequencing. Nature, 472(7341), 90–​94. PMCID: PMC4504184. insights into androgen receptor activation and transcriptional specificity.
Navin NE. 2014. Cancer genomics: one cell at a time. Genome Biol, 15(8), 452. Genome Biol, 13(10), R88. PMCID: PMC3491416.
PMCID: PMC4281948. Van Allen EM, Wagle N, Stojanov P, et  al. 2014. Whole-​exome sequencing
Nik-​Zainal S, Van Loo P, Wedge DC, et al. 2012. The life history of 21 breast and clinical interpretation of formalin-​fixed, paraffin-​embedded tumor
cancers. Cell, 149(5), 994–​1007. PMCID: PMC3428864. samples to guide precision cancer medicine. Nat Med, 20(6), 682–​688.
Paez JG, Janne PA, Lee JC, et al. 2004. EGFR mutations in lung cancer: cor- PMCID: PMC4048335.
relation with clinical response to gefitinib therapy. Science, 304(5676), Wagle N, Emery C, Berger MF, et al. 2011. Dissecting therapeutic resistance to
1497–​1500. PMID: 15118125. RAF inhibition in melanoma by tumor genomic profiling. J Clin Oncol,
Palanisamy N, Ateeq B, Kalyana-​Sundaram S, et al. 2010. Rearrangements of 29(22), 3085–​3096. PMCID: PMC3157968.
the RAF kinase pathway in prostate cancer, gastric cancer and melanoma. Wagner AH, Coffman AC, Ainscough BJ, et al. 2016. DGIdb 2.0: mining clini-
Nat Med, 16(7), 793–​798. PMCID: PMC2903732. cally relevant drug-​gene interactions. Nucleic Acids Res, 44(D1), D1036–​
Pao W, Miller V, Zakowski M, et al. 2004. EGF receptor gene mutations are 1044. PMCID: PMC4702839.
common in lung cancers from “never smokers” and are associated with Wang Q, Li W, Liu XS, et al. 2007. A hierarchical network of transcription fac-
sensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci U S A, tors governs androgen receptor-​dependent prostate cancer growth. Mol
101(36), 13306–​13311. PMCID: PMC516528. Cell, 27(3), 380–​392. PMCID: PMC3947890.
Pao W, Miller VA, Politi KA, et al. 2005. Acquired resistance of lung adeno- Wang Q, Li W, Zhang Y, et al. 2009. Androgen receptor regulates a distinct tran-
carcinomas to gefitinib or erlotinib is associated with a second mutation scription program in androgen-​independent prostate cancer. Cell, 138(2),
in the EGFR kinase domain. PLoS Med, 2(3), e73. PMCID: PMC549606. 245–​256. PMCID: PMC2726827.
Piraino SW, and Furney SJ. 2016. Beyond the exome:  the role of non-​ Wang Y, Waters J, Leung ML, et  al. 2014. Clonal evolution in breast cancer
coding somatic mutations in cancer. Ann Oncol, 27(2), 240–​ 248. revealed by single nucleus genome sequencing. Nature, 512(7513), 155–​
PMID: 26598542. 160. PMCID: PMC4158312.
Pleasance ED, Stephens PJ, O’Meara S, et al. 2010. A small-​cell lung cancer Weber M, Davies JJ, Wittig D, et al. 2005. Chromosome-​wide and promoter-​
genome with complex signatures of tobacco exposure. Nature, 463(7278), specific analyses identify sites of differential DNA methylation in
184–​190. PMCID: PMC2880489. normal and transformed human cells. Nat Genet, 37(8), 853–​ 862.
Poulikakos PI, Persaud Y, Janakiraman M, et al. 2011. RAF inhibitor resistance PMID: 16007088.
is mediated by dimerization of aberrantly spliced BRAF(V600E). Nature, Wood LD, Parsons DW, Jones S, et  al. 2007. The genomic landscapes of
480(7377), 387–​390. PMCID: PMC3266695. human breast and colorectal cancers. Science, 318(5853), 1108–​1113.
Prahallad A, Sun C, Huang S, et al. 2012. Unresponsiveness of colon cancer to PMID: 17932254.
BRAF(V600E) inhibition through feedback activation of EGFR. Nature, Xie M, Lu C, Wang J, et al. 2014. Age-​related mutations associated with clonal
483(7387), 100–​103. PMID: 22281684. hematopoietic expansion and malignancies. Nat Med, 20(12), 1472–​
Qin ZS, Yu J, Shen J, et al. 2010. HPeak: an HMM-​based algorithm for defining 1478. PMCID: PMC4313872.
read-​enriched regions in ChIP-​Seq data. BMC Bioinformatics, 11, 369. Xie T, Cho YB, Wang K, et al. 2014. Patterns of somatic alterations between
PMCID: PMC2912305. matched primary and metastatic colorectal tumors characterized by whole-​
Rauch T, Li H, Wu X, and Pfeifer GP. 2006. MIRA-​assisted microarray analy- genome sequencing. Genomics, 104(4), 234–​241. PMID: 25066378.
sis, a new technology for the determination of DNA methylation patterns, Yap KL, Li S, Munoz-​Cabello AM, et al. 2010. Molecular interplay of the non-
identifies frequent methylation of homeodomain-​containing genes in lung coding RNA ANRIL and methylated histone H3 lysine 27 by polycomb
cancer cells. Cancer Res, 66(16), 7939–​7947. PMID: 16912168. CBX7 in transcriptional silencing of INK4a. Mol Cell, 38(5), 662–​674.
Ross-​Innes CS, Stark R, Teschendorff AE, et al. 2012. Differential oestrogen PMCID: PMC2886305.
receptor binding is associated with clinical outcome in breast cancer. Zhang J, Fujimoto J, Zhang J, et al. 2014. Intratumor heterogeneity in localized
Nature, 481(7381), 389–​393. PMCID: PMC3272464. lung adenocarcinomas delineated by multiregion sequencing. Science,
Roth A, Khattra J, Yap D, et  al. 2014. PyClone:  statistical inference of 346(6206), 256–​259. PMCID: PMC4354858.
clonal population structure in cancer. Nat Methods, 11(4), 396–​ 398. Zhang J, Walsh MF, Wu G, et  al. 2015. Germline mutations in predisposi-
PMID: 24633410. tion genes in pediatric cancer. N Engl J Med, 373(24), 2336–​ 2346.
Schrader KA, Cheng DT, Joseph V, et al. 2016. Germline variants in targeted PMCID: PMC4734119.
tumor sequencing using matched normal DNA. JAMA Oncol, 2(1), 104–​ Zhang Y, Liu T, Meyer CA, et  al. 2008. Model-​based analysis of ChIP-​Seq
111. PMID: 26556299. (MACS). Genome Biol, 9(9), R137. PMCID: PMC2592715.
Schroeder MP, Rubio-​Perez C, Tamborero D, Gonzalez-​Perez A, and Lopez-​ Zhao J, Ohsumi TK, Kung JT, et  al. 2010. Genome-​wide identification of
Bigas N. 2014. OncodriveROLE classifies cancer driver genes in loss of polycomb-​ associated RNAs by RIP-​ seq. Mol Cell, 40(6), 939–​ 953.
function and activating mode of action. Bioinformatics, 30(17), i549–​555. PMCID: PMC3021903.
PMCID: PMC4147920. Zhao Z, Tavoosidana G, Sjolinder M et  al. 2006. Circular chromosome con-
Sequist LV, Waltman BA, Dias-​Santagata D, et al. 2011. Genotypic and histo- formation capture (4C) uncovers extensive networks of epigenetically
logical evolution of lung cancers acquiring resistance to EGFR inhibitors. regulated intra-​and interchromosomal interactions. Nat Genet, 38(11),
Sci Transl Med, 3(75), 75ra26. PMCID: PMC3132801. 1341–​1347. PMID: 17033624.
 53

5 Genetic Epidemiology of Cancer

KATHRYN L. PENNEY, KYRIAKI MICHAILIDOU, DEANNA ALEXIS CARERE,


CHENAN ZHANG, BRANDON PIERCE, SARA LINDSTRÖM, AND PETER KRAFT

OVERVIEW to the incidence in the general population.) For most cancers, λs falls
between 1.5 and 2.5, although there are exceptions: testicular cancer,
Cancer is a genetic disease at the cellular level. Tumors occur when for example, has λs = 8.6. For comparison, the λs for early-​onset dis-
variations in DNA sequence and aberrant gene expression disrupt the eases or diseases that affect fitness can be much larger than for most
normal cellular processes that control cell division, survival, clonal cancers: the λs for type 1 Diabetes is 13.7 and for schizophrenia is
growth, and migration. Most of the genomic variants that affect can- 9.0 (Wray et  al., 2010). The lower λs for many cancers may reflect
cer growth and progression are somatic mutations in tumor tissue that that they are late-​onset diseases that typically affect men and women
are acquired rather than inherited (see Chapter 4). At the same time, after reproductive age, so that most (but not all) alleles associated with
genetic variants inherited in germline DNA can affect susceptibility to cancer risk may not be under strong negative selection, and hence do
cancer. This chapter reviews the evidence linking inherited genetic var- not exhibit the strong biological effects typical of negatively selected
iation to cancer incidence and mortality. The literature includes studies alleles.
of rare variants in high-​risk families, risk concordance in twins, and There is also evidence that having a relative with cancer of one type
association studies of unrelated individuals in search of common, less increases risk of other types of cancers. For example, first-​degree rela-
penetrant susceptibility loci. Despite considerable variability across tives of cases with prostate cancer are at 1.1-​fold increased risk of colon
cancers, most cancers exhibit familial clustering, driven partly by a cancer and 1.3-​fold increased risk of thyroid cancer (Amundadottir
small number of known rare variants that powerfully affect risk and a et al., 2004). Fraternal (dizygotic) twins of individuals with any type
larger number of common variants that, in combination or at the popu- of cancer are at 1.1-​fold increased risk of any cancer, while identical
lation level, could significantly affect risk. We discuss the implications (monozygotic) twins of individuals with any type of cancer are at 1.4-​
of these findings for clinical care, public health, and basic biology. We fold increased risk.
close with a discussion of open questions, most notably the puzzle of Some of the increased risk of cancer among family members may be
“missing heritability”: the fact that—​despite tremendous advances in due to shared environmental risk factors. However, in order to explain
our understanding of the genetic epidemiology of cancer over the last most of the familial aggregation, the magnitude of environmental
10 years—​multiple lines of evidence suggest that many specific risk effects and strength of the correlation of exposures among family
variants, both rare and common, have yet to be discovered. members would have to be much larger than is empirically observed
(Khoury et al., 1988). This suggests that much familial aggregation is
due to inherited genetic factors.
EVIDENCE SUPPORTING INHERITED SUSCEPTIBILITY Twin studies are one tool that can help disentangle inherited genetic
TO CANCER susceptibility from shared environmental exposures. Under certain
mathematical modeling assumptions (Falconer, 1965; Falconer and
Scientists have long reported that cancer aggregates in families. Paul Mackay, 1996; Khoury et al., 1988; Sham, 1998)—​for example, that
Broca described a large family with a high burden of various cancers in an individual’s cancer risk is determined by an unobserved, normally
his 1866 book, Traité des Tumeurs (Figure 5–​1) (Broca, 1866). At least distributed “liability,” which is made up of independent genetic and
three types of systematic studies provide evidence for an inherited environment components—​the difference in the proportion of disease-​
genetic contribution to risk of cancer: studies showing that relatives of concordant monozygotic (identical) twin pairs and the proportion of
cancer cases have a higher risk of cancer than the general population; disease-​concordant dizygotic (fraternal) twin pairs can be used to esti-
studies comparing cancer incidence patterns across different types of mate the inherited genetic contribution to familial aggregation, as dis-
relative pairs (e.g., comparing concordance rates in monozygotic ver- tinct from shared environmental factors. In particular, such studies can
sus dizygotic twins); and studies that test whether the inheritance of estimate the heritability of different cancers, defined as the proportion
cancer in extended pedigrees is consistent with a simple Mendelian of total variance in liability due to genetic factors. The genetic contri-
model for genetic inheritance, such as autosomal dominant or reces- bution can range dramatically for different cancers. A  recent analy-
sive. These studies also shed light on the genetic architecture of differ- sis of 203,691 individual twins from the Scandinavian twin registries
ent cancers, that is, the number of distinct genetic loci that influence (Mucci et al., 2016) determined overall cancer heritability to be 33%
cancer risk, their allele frequencies, and the distribution of the magni- (95% confidence interval [CI]: 30%, 37%). A high degree of heritabil-
tude of their effects. ity was seen for several specific cancers, including melanoma (58%;
Close relatives of cancer cases are themselves at increased risk of 95% CI:  43%, 73%), prostate cancer (57%; 95% CI:  51%, 63%),
cancer. For example, a woman with a first-​degree female relative with ovarian cancer (39%; 95% CI: 23%, 55%), kidney cancer (38%; 95%
breast cancer has a 2-​fold higher risk of developing breast cancer com- CI: 21%, 55%), and breast cancer (31%; 95% CI: 11%, 51%). These
pared to a woman without a family history (Collaborative Group on results provide evidence for an inherited genetic contribution to these
Hormonal Factors in Breast, 2001; Kharazmi et  al., 2014; Pharoah cancers and a rough guide to the relative importance of inherited ver-
et al., 1997). Men whose father had prostate cancer are more than twice sus acquired factors to risk. However, heritability values should be
as likely to develop prostate cancer (Kicinski et al., 2011). Individuals interpreted with care, because procedures for estimating heritability
with a first-​degree relative with colon cancer have a approximately are sensitive to modeling assumptions (Falconer and Mackay, 1996;
one-​third increased risk of developing the disease (Slattery et  al., Lewontin, 1995; Sham, 1998; Visscher et al., 2008), and because her-
2003). Table 5–​1 shows the sibling recurrence risk ratio λs for a range itability is defined in the context of a particular mathematical model
of cancers. (λs is the ratio of the incidence in siblings of a cancer case and need not have direct biologic or epidemiologic interpretability

53
54

54 Part I:  Basic Concepts


Mrs. Z
b 1728
d 1788
Mrs. D
b 1773
d 1827
Mrs. C
b 1763
d 1814

d 55 d 61
d 40 d 47
1848 1856
1822 1837
Liver
Abdomen

Figure 5–​1.  Portion of a pedigree describing breast and other cancers running in Paul Broca’s family. Broca (1866).

(Hoover, 2000; Visscher et al., 2008). In particular, a high heritability studies (Figure 5–​2). Linkage studies (described in more detail later
does not imply that there are no environmental risk factors for disease in the chapter) use the co-​segregation of disease status and alleles at
or that most cases are due to genetics alone: heritability should not be specific loci within families to identify genomic regions harboring
interpreted as a population attributable fraction. Nor do differences in causal variants. Association studies (described later in the chapter)
heritability across populations imply that the genetic contribution to use population-​level (across-​family) association between an allele and
disease is necessarily greater or smaller in one population or the other disease, typically estimated using large series of unrelated cases and
(Hoover, 2000): differences in the distribution of environmental risk controls. Before reviewing these two approaches in more detail, we
factors likely account for much of the differences in heritability. introduce two conceptual models relating genetic variants to cancer
risk: a Mendelian model and a polygenic model. In broad terms, link-
age studies are better suited to identify Mendelian variants, and associ-
GENETIC MODELS FOR CANCER RISK ation studies are better suited to identify polygenic variants.

Although the data on patterns of cancer incidence within and across


families suggest that inherited genetic factors influence risk for most Mendelian (Rare Variant Large Effect) Model
cancers, they do not identify particular genetic loci and variants associ-
Under a Mendelian model, the association between disease variants
ated with risk. Identifying cancer susceptibility variants requires meas-
and cancer risk closely follows one of the classical Mendelian domi-
uring and comparing DNA variation across individuals. Over the last
nance patterns, with nearly complete penetrance and few phenocopies.
25 years, two basic approaches have identified most of the currently
(Penetrance refers to the probability that an individual with the risk
known cancer susceptibility variants: linkage studies and association
genotype contracts disease. Phenocopies are individuals diagnosed
with the disease who do not have the risk genotype.) For example, a
risk allele might act in a dominant fashion: those who carry at least
Table 5–​1.  Sibling Recurrence Risk Ratios for Various one copy of the risk allele are at very high lifetime risk of cancer.
Cancers Mendelian risk alleles are presumed to be rare—​typically with an
allele frequency below 1/​500.
Site λs Family-​based linkage studies have been relatively effective at iden-
tifying rare, high-​penetrance Mendelian risk alleles for several rea-
Aerodigestive tract 6.80 sons. First, by focusing on families with multiple affected individuals,
ALL 5.00 they enrich the sample for risk alleles. The sample sizes needed to
Bladder 1.53 detect associations with rare alleles in a population-​based case-​control
Brain/​CNS 1.97 study are much larger, even for alleles with large effects. Second, high
Breast 1.83 penetrance and a low phenocopy rate ensure that disease status is an
CLL 4.50 effective proxy for the unknown and unmeasured risk allele. This
Colorectal 2.54 makes co-​segregation of a measured genetic marker and disease status
Female genitalia 2.22 informative for co-​segregation between the marker and the risk allele.
Granulocytic leukemia 2.94
Hodgkin lymphoma 1.25
Lung 2.55 Polygenic (Common Variant Small Effect) Model
Melanoma 2.10
Multiple myeloma 4.29 Under a polygenic model, disease risk is determined by the cumulative
Non-​Hodgkin lymphoma 1.68 effects of many common alleles, each of which has a small individual
Oral cavity 1.82 effect on disease risk. Although made up of discrete effects, the cumu-
Prostate 2.21 lative polygenic components of risk can be thought of as continuous
Soft tissue sarcoma 2.00 factors with a known correlation structure among relatives. This model
Testicular 8.57 underlies the calculations used to estimate heritabilities from twin and
Thyroid 8.48 other family-​based studies.
Uterine 1.32 Alleles with small effect are difficult to detect using linkage stud-
ies: low penetrance and a large number of phenocopies make disease
status a poor proxy for any single contributing locus. On the other
ALL = acute lymphoblastic leukemia (childhood); CLL = chronic
lymphocytic leukemia; CNS = central nervous system.
hand, because they are common, these alleles can be effectively stud-
Bhat et al. (2015); Chang et al. (2005); Risch (2001). ied in population-​ based case-​control studies, despite their smaller
 5

Genetic Epidemiology of Cancer 55


(a) (b)

44 AT
AT AT
AA AA AA

AA AT
AT TT
65 45 39 AT
BD AE BA BD

AA
AA AA
AA AT

41 23 39 fT,cases = 0.33
fT,controls = 0.17
BA BE DE

Figure 5–​2.  Strategies for localizing cancer susceptibility variants. (a) Linkage studies rely on the co-​segregation of alleles and disease status within a
family. (b) Association studies compare allele frequencies between samples of unrelated cases and controls.

effects. The relative efficiency of association approaches relative to genes (CPGs) have been discovered, and together account for at least
linkage approaches for common risk alleles with modest effects was 3% of cancer diagnoses worldwide (Rahman, 2014).
one of the main arguments justifying the expansion of genetic associ- CPGs employ a diversity of molecular mechanisms. Most function
ation studies over the last 20 years. normally as tumor suppressors: gatekeepers or caretakers in the cell
cycle that inhibit cell proliferation and prevent tumor development
(Oliveira et al., 2005). The best known of these is TP53, which encodes
Genetic Architecture of Cancer a tumor suppressor active in many human tissues. Dominantly inherited
Genetic architecture refers to the number of risk alleles that influence loss-​of-​function mutations in TP53 lead to Li-​Fraumeni syndrome, a
a particular cancer and the distribution of their frequencies and effect notable cancer syndrome characterized by both rare and common can-
sizes. The genetic architecture will influence the relative efficacy of cers, with pediatric and adult onset, and multiple primary cancers over
different study designs (Bodmer and Bonilla, 2008). For example, if the lifespan (Kamihara et al., 2014). It is the tumor suppressor class of
both common and rare variants contribute to cancer risk, but the effect CPGs (and the RB1 gene in pediatric retinoblastoma, in particular) that
sizes for both common and rare variants are modest, then linkage stud- formed the basis of Knudson’s two-​hit hypothesis—​that is, that carci-
ies will be less effective than association studies at identifying risk nogenesis requires accumulation of loss-​of-​function mutations in both
loci (and very large sample sizes will be needed to reliably identify copies of a tumor suppressor gene, and that familial cancers develop
rare variants). On the other hand, if Mendelian mutations account for at younger ages because genetically predisposed individuals start out
most cases, association studies looking for common risk alleles may with the first “hit” already present in all germline cells, with the second
be ineffective. Patterns of disease segregation within families and con- hit occurring in the tumor (Knudson, 1971). Carcinogenesis addition-
cordance rates across different degrees of relationship provide some ally requires the activation of proto-​oncogenes, and a small number of
information about the genetic architecture. However, these data are CPGs (e.g., RET, associated with familial medullary thyroid cancer)
rarely strong enough to rule out either a Mendelian or polygenic com- fall into this category, predisposing to cancer when germline gain-​of-​
ponent, and suggest that both rare variants with large effects and com- function mutations cause constitutional activation of the gene product
mon variants with smaller effects contribute to many cancers. As we (Kouvaraki et al., 2005).
review in the following, findings from linkage and association studies At the family level, a majority of CPGs exhibit autosomal dominant
are consistent with this mixed genetic architecture, combining both inheritance (e.g., BRCA1/​2, causing hereditary breast and ovarian can-
rare and common risk alleles (Figure 5–​3). cer syndrome), while a sizable minority are autosomal recessive (e.g.,
MUTYH: colorectal polyposis), and a handful are X-​linked (e.g., WAS
[Wiskott-​Aldrich syndrome]), Y-​linked (SRY:  gonadal dysgenesis
LINKAGE STUDIES and cancers of the underdeveloped gonadal tissues), or show parent-​
of-​origin effects via differential methylation (e.g., SDHD: hereditary
One of the short-​term goals of the Human Genome Project when it paraganglioma/​pheochromocytoma syndrome) (Bardella et al., 2011).
was launched in 1990 was the development of a panel of microsatellite Gene dosage effects are variable: for example, while homozygous or
markers spaced across the genome. Over the next decade, research- compound heterozygous mutations in BRCA1 are unobserved and are
ers used linkage studies to track co-​segregation of disease with these assumed to be lethal in utero (Gowen et al., 1996), double mutations
markers to identify broad regions of the genome that harbored rare, in BRCA2 lead to Fanconi anemia, a condition characterized by cer-
high-​penetrance cancer predisposition genes. Linkage studies have tain physical features, bone marrow failure, and childhood cancers
focused on families with an often striking aggregation of disease. (Howlett et al., 2002).
Owing to this method of case ascertainment, successful investigations Familial cancer clusters caused by CPG mutations share some or
of these families have typically yielded rare genetic mutations confer- all of the following features:  early age at diagnosis (e.g., colorec-
ring a moderate to high risk of particular cancers. (Relative risks for tal cancer in adolescence); multiple primary tumors in one individ-
those with high-​risk genotypes range from approximately 3 to > 100 ual (e.g., bilateral breast cancer, or both breast and ovarian cancer
[Nagy et al., 2004].) However, while the identified mutations are often diagnoses); uncommon sex distribution (e.g., breast cancer in men);
limited in relevance to specific families or ethnic groups, the genes rare tumors (e.g., adrenocortical cancer) or an unusual constella-
identified have in most cases had broad application to other families tion of features (e.g., renal cancer and pneumothorax); and multi-
with similar phenotypes. To date, over 114 such cancer predisposition ple affected individuals across generations. These unique patterns of
56

56 Part I:  Basic Concepts

Linkage

CDH1 BRCA1
TP53

10 BRCA2

STK11
Relative Risk

PALB2

PTEN

CHEK2
ATM

GWAS

Risk SNPs

1
0.000001 0.00001 0.0001 0.001 0.01 0.1 1
Allele frequency

Figure 5–​3.  Relative risk and frequency of known breast cancer risk alleles. BRCA1 and BRCA2 were discovered via linkage studies. The highlighted
alleles in the lower right-​hand corner were discovered via genome-​wide association studies (GWAS). Modified from a figure courtesy of Peter Devilee
and Douglas Easton.

disease—​distinguishable even against the backdrop of cancer as a for genetic linkage and estimate map distance. Map distance, distinct
common disease of older age—​make high-​risk cancer families ame- but related to physical distance on a chromosome, reflects the tight-
nable to identification and systematic classification (Fitzgerald et al., ness of linkage between two loci, and is estimated by recombination
2010; Umar et al., 2004). These families have historically served as a frequency (ϴ). When two loci are unlinked, they are inherited inde-
fruitful resource for genetic studies, particularly those employing link- pendently of each other, and ϴ = 0.5; when two loci are linked, they
age analysis (Hall et al., 1990; Khan et al., 1988). are more often inherited together, and ϴ < 0.5. The recombination
Linkage analysis relies on the co-​segregation of a measured genetic fraction ϴ is typically estimated using maximum likelihood meth-
marker and an unmeasured disease allele, and uses statistical methods ods:  LOD (“logarithm of the odds”) scores are then computed for
to test for and measure genetic linkage between the marker and the each value of ϴ, comparing the likelihood of observing the family
causal allele. In this way, linkage analysis can provide both (a) a test- data assuming the loci are linked to the likelihood assuming the loci
able marker to predict disease risk status by proxy when the CPG itself are unlinked. Positive LOD scores favor linkage, and a LOD score of
is not known, and (b) physical mapping of a CPG to a particular chro- +3 or greater has traditionally been considered strong evidence that
mosomal region, through repeated linkage analyses with increasingly two loci are linked. The ϴ value with the highest LOD score is con-
proximate markers. To date, linkage analysis has led to the identifica- sidered the best estimate of ϴ.
tion of at least 59 CPGs (Rahman, 2014). For rare phenotypes, a model-​based (parametric) method of link-
Linkage analysis requires two sets of data: first, accurate documen- age analysis is typically used, which assumes a particular mode of
tation of family relationships and phenotype in affected and unaffected inheritance. Another critical assumption is that a single genetic locus
relatives; and second, genetic samples from all available relatives, is responsible for the phenotype in the family, so the utility of linkage
genotyped for at least one highly polymorphic genetic marker. Highly analysis is determined by the extent to which having the phenotype is
polymorphic markers are essential because it is the variability in marker strongly predictive of having the underlying risk allele. A limitation of
status that permits tracking of the marker through a family. (Highly linkage analysis is that it may be less effective for clusters of common
polymorphic markers are genetic variants with many relatively com- cancers with probable phenocopies, where multiple genes and/​or envi-
mon alleles—​say, each with frequency greater than 1%. Most individ- ronmental factors contribute to risk. Extensions of linkage analysis to
uals in the population will be heterozygous for these markers: they will these situations typically make use of non-​parametric methods that are
have inherited two different alleles from their parents.) Homozygous agnostic about mode of inheritance, and make no assumptions about
individuals (e.g., marker genotype = AA) will be uninformative, while the number of risk loci that contribute to a trait. The utility of linkage
a marker with only two or three possible alleles (e.g., A, B, C) may be analysis is further limited, however, by small family size, unavailabil-
difficult to track if there are many matings between people with the ity of relatives’ genetic samples, and the general work-​intensive and
same combination of alleles (e.g., AB & AB) (Figure 5–​4). costly nature of a procedure involving dozens to hundreds of individ-
Informally, one can often visually track disease and marker sta- ual samples and case histories (Klein, 2005). Today, linkage analysis
tus in a pedigree and identify the likely disease-​linked marker, as in is largely being supplanted by other technologies (e.g., whole exome
Figure 5–​4. Formally, linkage analysis uses likelihood ratios to test sequencing) in the field of CPG discovery.
 57

Genetic Epidemiology of Cancer 57


(a) (b)

44

71 31 45 36 49
DD EH BC ED EH FG DD

65 45 39
BD AE BA BD

28 29 48 43 37 40
EB AH EB HC HC EF HF

41 23 39
25 BA BE DE
EA

Figure 5–​4.  Co-​segregation of breast cancer and alleles for a marker on chromosome 17q21 near BRCA1 (Hall et al., 1990). In family (a), the E allele
and the breast cancer risk allele are inherited together (denoting absence of recombination); in family (b), the B allele and the risk allele are inherited
together.

ASSOCIATION STUDIES markers near the FTO gene, while the third study—​which matched
cases and controls on body mass index, unlike the other studies—​
Association studies are typically conducted as case-​control studies, in did not (Diabetes Genetics Initiative et  al., 2007; Scott et  al., 2007;
which genotype frequencies are compared between unrelated patients Wellcome Trust Case Control Consortium, 2007). These markers are
with the disease of interest (cases) and individuals who are free of the strongly associated with body mass index. By conditioning on a medi-
disease (controls). Cases may be drawn from population-​based studies ator of the marker-​diabetes association, the power of the third study
(prospective cohorts or cancer registries) or from hospital/​clinic (retro- to identify these markers was greatly reduced. Although most GWAS
spective)–​based series (Cordell and Clayton, 2005; Newton-​Cheh and have been conducted by conditioning on a minimal set of covariates, it
Hirschhorn, 2005). Cohort studies are conducted when a large number is important to note that in some situations, the inclusion of covariates
of individuals are recruited and a comprehensive collection of baseline as potential effect modifiers may lead to increased power, as discussed
data are collected. Individuals are followed, and those who develop the later in this chapter in the section on gene–​environment interactions.
disease of interest are considered cases in the association analysis. The Considering that the initial aim for many genetic association stud-
cost of genotyping an entire cohort is usually prohibitive, so cohort ies is discovery—​establishing that a genetic variant is associated with
studies are typically analyzed as nested case-​control studies. Such a cancer risk—​some investigators have adopted designs that provide
design is particularly useful if other (non-​genetic) risk factors, which valid tests of the null hypothesis but that have biased measures of asso-
may not be reliably assessed in retrospective case-​control studies, need ciation under the alternative, in order to increase statistical power. For
to be taken into account (Cordell and Clayton, 2005; Newton-​Cheh example, many association studies oversample cases with a positive
and Hirschhorn, 2005). In hospital-​based retrospective case-​control family history and compare these to general-​population controls. This
studies, cases are ascertained by disease status, and other phenotypic approach can improve the power to detect associations, because such
characteristics are also collected (Cordell and Clayton, 2005; Newton-​ cases are more likely to carry susceptibility alleles, but it can lead to
Cheh and Hirschhorn, 2005). biased estimates of the effect size (Antoniou and Easton, 2003).
In principle, genetic association studies are a special case of an Disease-​free controls need to be drawn from the same population
observational epidemiologic study: the exposure of interest just hap- as the cases (in particular, they need to be of the same ethnicity), or
pens to be genetic variation. Accordingly, basic principles of epide- differences in genotype frequencies between cases and controls may
miologic design for valid inference should be considered (Manolio simply reflect population variation in frequencies. A variety of analytic
et al., 2006). However, in practice, some of the biases that affect obser- methods have been developed to adjust for latent population structure
vational studies are not present or are minimized by the nature of the and cryptic relatedness (Kang et al., 2010; Price et al., 2010); of these,
genetic exposure. For example, exposure suspicion bias (Manolio adjusting for the top principal components of genetic variation is the
et al., 2006) is unlikely to affect genotype measurement, as the expo- most widely used (Price et al., 2006). Controls also need to be repre-
sure is measured using automated techniques by lab technicians who sentative of the spectrum of individuals at risk for disease so that bias
are blind to the case-​control status of the individual who supplied the is avoided and appropriate adjustments need to be made to avoid asso-
DNA sample. With the exception of population stratification bias, ciations due to confounders (Pearson and Manolio, 2008; Zondervan
genetic associations are unlikely to be strongly confounded, because and Cardon, 2007). For example, case-​control studies in prostate can-
genotypes are distributed independently of potential confounders cer that used “supernormal” controls (i.e., with very low PSA levels)
within ancestrally homogeneous, outbred populations. Consequently, identified markers that in subsequent studies in controls alone were
many genetic association studies apply minimal adjustment for covari- shown to be associated with PSA levels rather than prostate cancer
ates, as this adjustment is not needed for valid tests of the null hypoth- (Knipe et al., 2014). A well-​defined but non-​representative set of con-
esis of no cancer-​genotype association. In fact, care is required when trols can sometimes increase the power of the study, but it can also
deciding which covariates to adjust for, as adjustment for some known introduce selection bias.
cancer risk factors can reduce power to detect a genetic association An alternative approach for studying low-​penetrance alleles is a
or even create a spurious genetic association (e.g., due to collider family-​based association design. Rather than utilizing unrelated cases
bias) (Aschard et al., 2015; Day et al., 2016; Kuo and Feingold, 2010; and controls, family-​based designs involve genotyping relatives. The
Pirinen et al., 2012). For example, two of the three initial genome-​wide most common approach involves genotyping affected cases together
association studies (GWAS) of type 2 diabetes identified diabetes risk with their parents (“trio design”). An association test is then derived
58

58 Part I:  Basic Concepts


by comparing the frequency of allelic transmission from heterozygous www.internationalgenome.org/​home) or the Haplotype Reference
parents with its expectation (1/​2) (Laird and Lange, 2006; Spielman Consortium (McCarthy et al., 2016; http://​www.haplotype-​reference-​
et al., 1993). This design has the advantage that, because the case gen- consortium.org), there was no exhaustive catalog of common genetic
otypes are matched to the un-​transmitted alleles from the same par- variation in humans. Instead, investigators relied on existing databases
ents, it is not subject to bias due to population stratification. However, such as dbSNP to identify variants that putatively altered the function
collecting sufficient parent-​offspring trios can be problematic, espe- or expression of the gene—​primarily non-​synonymous coding SNPs.
cially for an adult-​onset disease (Laird and Lange, 2006). (It is worth noting that a variant that is predicted to alter a gene’s pro-
tein product may not change the cancer-​relevant function of the gene;
additional experimental work is needed to establish the downstream
Linkage Disequilibrium impact of changes in protein structure [Rebbeck et al., 2004].) After
The genetic phenomenon of linkage disequilibrium has made associa- including putative functional variants, investigators might then attempt
tion studies aimed at discovering novel variants associated with dis- to indirectly capture other common variants in or near the gene by
ease relatively cost-​effective. It also makes association studies more carefully selecting a set of “tag SNPs”: a set of SNPs chosen such that
precise than linkage studies, in the sense that markers associated with any variant in the target region is either itself a tag SNP or is in high
disease tend to be physically closer to the causal variants than mark- linkage disequilibrium with a tag SNP (or a known combination of tag
ers that co-​segregate with disease within families. The resolution from SNPs). Selecting tag SNPs requires a reference panel of densely geno-
linkage studies is approximately one to two million base pairs, while typed or sequenced individuals from a population with similar ancestry
the resolution for association studies is on the order of tens of thou- to the study population. (Before the advent of the HapMap, investiga-
sands of base pairs (Schaid et al., 1999). tors had to generate these panels for their candidate genes themselves
In brief, linkage disequilibrium refers to the correlation between [Hunter et al., 2005].) Target regions for candidate gene studies typi-
alleles at two varying sites on the same chromosome. Most common cally included a small amount of sequence up-​and downstream of
single nucleotide polymorphisms (SNPs, pronounced “snips”) are the gene (10–​20 kb), so by default—​and often by explicit choice—​
strongly correlated with at least one other SNP, and in some cases, candidate gene studies focused on coding regions of the genes, and did
dozens of SNPs may be in strong linkage disequilibrium with each not investigate medium-​and long-​range gene-​regulation mechanisms.
other. (SNPs are the most commonly studied markers in genetic asso- Because of these constraints, candidate gene studies could only
ciation studies. They are single-​base-​pair sites in the genome that vary focus on a small number of genes, and a small number of variants
across individuals.) Thus, genotyping a carefully chosen subset of within those genes, and could not reliably make any claims regarding
common SNPs can provide useful information about the majority of the comprehensiveness of these tests: if none of the variants tested
common SNPs in a region or across the whole genome. showed any association with disease, it was still possible that an
Patterns of linkage disequilibrium are influenced by a number of fac- untyped and unknown variant in the gene was associated with dis-
tors, including recombination, selection, and demographics. Because ease, but was not detected because it was not in strong linkage dis-
of this, a SNP that is a good proxy for another in one population may equilibrium with any of the typed variants. Despite some success,
not be a good proxy in another population. Similarly, the accuracy e.g., ALDH2 and ADH genes in esophageal cancer; (Brooks et  al.,
of imputation (discussed later) from a fixed set of genotyped SNPs 2009; Cui et al., 2009; Hashibe et al., 2008; Wu et al., 2012), CHEK2,
may differ across populations. In particular, Africans and African PALB2, and CASP8 in breast cancer; (Cox et  al., 2007; Lin et  al.,
Americans exhibit lower levels of linkage disequilibrium—​on aver- 2015; Southey et al., 2016), the majority of the associations in these
age, SNPs in Africans are strongly correlated with fewer other SNPs genes failed to replicate in subsequent independent studies (Ioannidis
than SNPs in Europeans or East Asians. As a consequence, more SNPs et al., 2001), and often candidate gene studies reached apparently con-
need to be genotyped in Africans and African Americans to achieve the tradictory conclusions. There are a number of reasons for these incon-
same level of coverage as in Europeans or East Asians. sistencies and lack of replication (Hirschhorn and Altshuler, 2002;
Kraft et  al., 2009b; NCI-NHGRI Working Group on Replication in
Association Studies et  al., 2007). First, the initial report may have
been a false positive. Many early candidate gene association studies
Candidate Gene Association Studies failed to appropriately adjust significance thresholds for the number
Before advances in our catalogs of genetic variation and in genotyping of markers tested. Small sample sizes and implicitly overoptimistic
technology made GWAS feasible, most association studies focused priors also likely contributed to false positive results (Hirschhorn
on particular candidate genes. These studies required investigators to and Altshuler, 2002; Hirschhorn et al., 2002; Ioannidis, 2005, 2006;
make several key decisions about which genes to study and what varia- Wacholder et  al., 2004). Second, replication studies may also have
tion in those genes to measure—​decisions that were constrained by the been underpowered to detect modest true associations. Third, studies
available knowledge and technologies (Tabor et al., 2002). looking at the same candidate gene may also have genotyped different
First, the choice of which candidate genes to study should ide- markers, making comparison of results difficult, especially before the
ally have been driven by biological hypotheses linking gene prod- advent of comprehensive surveys of linkage disequilibrium. Fourth,
ucts to carcinogenesis. DNA repair genes were common candidates, differences in allele frequencies or linkage disequilibrium patterns
for example—​over 140 publications on just XRCC1 and lung neo- across populations can lead to non-​replication: a marker that was a
plasms are listed in the Centers for Disease Control and Prevention good proxy for the causal variant in the initial population is a poor
Genopedia (https://​phgkb.cdc.gov/​HuGENavigator/​home.do)—​as proxy in the second. Finally, in principle, differences in non-​genetic
were steroid-​hormone metabolism genes for hormone-​related cancers exposures across study populations could modify the effect of the
(Hunter et  al., 2005). However, some studies chose candidate genes genetic variant (although in practice, clear examples of this phenom-
primarily because they contained a well-​known and easily genotyped enon are rare).
polymorphism, with a post hoc hypothesis relating the gene to cancer. As discussed in the next section, GWAS incorporated some of the
Furthermore, the choice of biologically motivated candidate genes was lessons learned from the inconsistencies of candidate gene studies,
(and still is) limited by our knowledge of cancer biology. Candidate including the following: studies should be sufficiently powered; sig-
gene studies could not systematically explore genes that might have an nificance thresholds should appropriately account for the number of
unanticipated link to cancer. markers tested or the low prior probability that any particular marker
Second, after having chosen candidate genes, investigators had is associated with disease risk; and standardized genotyping panels
to choose which genetic markers to type in and around these genes. should be used as far as possible to facilitate meta-​analysis. Arguably
Even ignoring cost constraints, exhaustively genotyping all com- most important, GWAS allowed investigators to investigate the entire
mon variation in a gene was not possible; until very recently, with the genome in a hypothesis-​free manner, enabling them to discover cancer-​
advent of large whole-​genome sequencing reference panels like the associated variants in genes and pathways not previously hypothesized
1,000 Genomes Project (Genomes Project Consortium, 2015; http://​ to be associated with the development of cancer.
 59

Genetic Epidemiology of Cancer 59


GENOME-​WIDE ASSOCIATION STUDIES:  magnitude > 1.3. More recently identified loci have ORs below 1.2 and
DESIGN PRINCIPLES even below 1.1, suggesting that samples sizes larger than 10,000 cases
and 10,000 controls will be required to detect additional associations
In GWAS, the concept of association studies is extended to the whole with such modest effects (Figure 5–​5).
genome; an agnostic approach is followed, in which no prior hypoth-
esis about the location or type of genetic variants or genes involved Phenotype Choice and Definition
in the disease of interest is assumed (Evangelou and Ioannidis, 2013;
Hirschhorn and Daly, 2005; Ioannidis et  al., 2009; Marchini and In GWAS and genetic association studies it is very important to have
Howie, 2010; Pearson and Manolio, 2008; Price et al., 2010). High-​ well-​defined phenotypes, as this will increase the power of detecting
throughput genotyping technologies developed in the first decade associations, especially for specific diseases where the classification
of the 2000s are used, allowing for the simultaneous assay of hun- is not always straightforward. Misclassification of cases and controls
dreds of thousands to millions of polymorphisms (Genomes Project can largely bias the study results toward no association (Pearson and
Consortium, 2012; Pearson and Manolio, 2008). GWAS capture the Manolio, 2008). The large majority of the GWAS that are conducted
large majority of genome variation by utilizing the linkage disequilib- usually take into consideration cases that are representative of the
rium structure from publicly available databases such as the HapMap population-​level disease. This allows for selection of a large number of
project or the 1000 Genomes Project (International HapMap, 2005). samples and is powerful to identify variants that are significantly asso-
SNPs on a GWAS chip are selected so that they “tag” most of the ciated in all the different subtypes of the disease. Different disease sub-
known common variants across the genome. Hence, rather than geno- types, though, might have different characteristics. Thus restricting the
typing all SNPs, GWAS use the genotype for the tag SNPs to give analyses to specific subtypes can increase power for detecting associa-
proxy information about the non-​genotyped SNPs which they tag. tions that predispose to those subtypes (Kraft and Haiman, 2010). For
The tagging SNPs on GWAS platforms usually have a population fre- example, restricting breast cancer cases to samples that carry deleteri-
quency of the minor allele greater than 1%. Based on data from the ous mutations in BRCA1 (which are mainly ER-​negative cases) led to
HapMap—​a project that genotyped a dense set of markers in reference the identification of further breast cancer susceptibility loci that were
samples from multiple ethnicities—​the majority of the commonly not identifiable in the general population cases (Antoniou et al., 2010).
occurring SNPs in European populations can be tagged using a subset This strategy has also proven successful in ovarian cancer, as analy-
of 500,000–​1,000,000 SNPs (International HapMap, 2005). A similar ses restricted to epithelial ovarian cancer (EOC) and a meta-​analysis
number of SNPs is needed for Asian ancestry populations, whereas a with ovarian cancer in patients with deleterious mutations in BRCA1/​2
larger number is needed to tag all common variation in African ances- (which have a high percentage of EOC) identified further susceptibil-
try populations (International HapMap, 2005). ity loci that could not be identified using all subtypes (Kuchenbaecker
The 1000 Genomes Project and the Haplotype Consortium data sets et al., 2015).
now provide more comprehensive resources, both because they are
based on whole-​genome sequencing and because they include more
Multiple Testing
individuals across more populations. These panels allow for the bet-
ter tagging of the rarer variants (Genomes Project Consortium, 2012; GWAS explore thousands to millions of polymorphisms in a large
McCarthy et al., 2016; Nelson et al., 2013). number of samples. The conventional p-​value < 0.05 for significance
Large numbers of samples are needed in order to identify associa- cannot be used in GWAS, as the number of tests is large—​tens of thou-
tions with diseases, as the effects of the variants that are being observed sands of variants will be flagged as statistically significant using that
are usually small: odds ratio (OR) < 1.5, and most of the times < 1.3 threshold, most of which will be false positives, and far too many to
(see section on GWAS results later in this chapter). Power is an impor- carry on to conventional functional studies. The usual “GWAS signif-
tant issue that must be taken into account when designing GWAS. icance” threshold that has been used is 5 × 10–​8, corresponding to a
The first published GWAS identified loci with a moderate number of Bonferroni correction for about one million independent tests (Pearson
cases/​controls since the variants originally identified had larger ORs of and Manolio, 2008). This threshold was determined via simulation

(a) Target power: 80% (b) Sample size: n = 25,000 cases

1.8 1.20
Smallest Detectable Odds Ratio
Smallest Detectable Odds Ratio

1.6 1.15

n = 1,000
1.4 1.10
power = 80%

1.2 1.05 power = 25%


n = 5,000
power = 1%
n = 25,000
1.0 n = 125,000 1.00

0.0 0.2 0.4 0.6 0.8 1.0 0.0 0.2 0.4 0.6 0.8 1.0
Risk Allele Frequency Risk Allele Frequency

Figure 5–​5.  Smallest detectable per-​allele odds ratios in a case-​control GWAS, assuming 1:1 case:control ratio and a significance threshold of p < 5 ×
10–​8. Panel (a) depicts the smallest detectable odds ratios with 80% power as a function of sample size and risk allele frequency. Panel (b) depicts smallest
detectable odds ratios for a study of 25,000 cases and 25,000 controls as a function of the target power and the risk allele frequency.
60

60 Part I:  Basic Concepts


and considers all common variants segregating in a European-​ancestry adequate power to detect true associations. Several different strategies
population (not just the tag SNPs on a GWAS array) and the empir- have been applied to increase power to detect associations. One of the
ical linkage disequilibrium among them (Dudbridge and Gusnanto, most common practices is to use a multistage design. In the first stage,
2008; Hoggart et al., 2008; Pe’er et al., 2008). Thus it is an appropriate a GWAS is performed using a large number of SNPs and a smaller
threshold, even when the number of markers tested is larger than one set of individuals. In the second stage, the most significantly associ-
million (e.g., when more than one million common markers are geno- ated variants from the first stage are brought forward for replication
typed or imputed; see later discussion in this chapter). Bayesian meth- in a larger independent set of samples. Three-​and four-​stage designs
ods have also been proposed to select markers that are truly associated are also performed, where the most significant variants in the previ-
with disease with high posterior probability; considering the low prior ous stages are explored in further independent samples (Pearson and
probability that a random common marker is truly disease-​associated Manolio, 2008).
and the small expected effect sizes, these Bayesian methods typically We wish to clarify two potential points of confusion regarding mul-
require large test statistics—​of similar magnitude to those required to tistage GWAS. First, the approach of only testing “promising” mark-
achieve p < 5 × 10–​8—​in order to flag a credible disease-​marker asso- ers from the first stage of GWAS is primarily a cost-​saving measure.
ciation (Wakefield, 2007, 2012, 2014). Empirically, the p < 5 × 10–​8 If GWAS data are available on all the studies that contribute to the
threshold has proven very specific: markers that pass that threshold in various stages, then the most efficient design and analysis will be to
a GWAS tend to replicate in adequately powered replication studies. conduct a genome-​wide meta-​analysis, and to study all markers in all
It is worth noting that such stringent significance thresholds are studies. Early GWAS adopted a multistage design because genome-​
driven by the desire to minimize the experiment-​wide Type I  error wide genotyping arrays were still many times more expensive than
rate:  the probability of incorrectly declaring even one marker to be assays typing tens to thousands of markers. Now that GWAS arrays
associated with disease when it is not. This is arguably an appropriate are much less expensive, the cost savings from a multistage design are
goal in the context of locus discovery, when one wants to say with nil or negligible and arguably are offset by the power gains of running
reasonable certainty that yes, this locus is associated with cancer risk, a GWAS array on as many samples as possible. Second, a study can
because one then wants to use that result to make inferences regard- meet the goals of replication without conducting a formal replication
ing disease biology. In particular, if one plans to conduct additional (i.e., genotyping the significant SNPs from the GWAS in yet another
expensive and time-​intensive functional studies to better understand set of samples). Most recent large GWAS have been meta-​analyses of
the mechanism linking genetic variation at a cancer-​associated locus to many studies, each of which had been genotyped. Associations that
cancer initiation and development, then one should be certain the asso- achieve genome-​wide significance in these studies meet both of the
ciation is real before investing considerable resources in functional criteria for replication: they are unlikely to be due to sampling vari-
studies. That said, requiring such a stringent significance threshold ability, and they are unlikely to reflect design artifact or other biases
ensures that some truly cancer-​associated markers will fail to reach that are restricted to one or a few contributing studies. Genotyping
statistical significance (controlling Type I error will increase Type II promising markers in yet another study may not provide much addi-
error). Multiple lines of evidence discussed in the following suggest tional information regarding the robustness of the finding, especially
that there are many cancer-​associated loci that show some evidence for considering that the size of any study available for replication is likely
association in GWAS, but do not achieve p < 5 × 10–​8. In some situa- much smaller than the original GWAS meta-​analysis.
tions, where there is greater tolerance for false-​positive results, it may In order to assemble such large samples sizes and replicate asso-
be appropriate to relax the genome-​wide significance threshold (relax- ciations in diverse studies and populations, investigators from many
ing control of Type I error in order to minimize Type II error). This studies have organized large, multi-​institutional, and multidisciplinary
could be the case in the setting where many promising markers can consortia. These consortia can pool financial and scientific resources,
be followed up cheaply and quickly—​for example, by genotyping all coordinate work, and ensure that credit gets apportioned fairly. Aside
independent markers with p < 10–​4 in a large replication set. We cannot from assembling large sample sizes, these collaborations can harmo-
be sure which of the markers with p between 10–​4 and 10–​8 are truly nize phenotype and exposure data, and make prospective decisions
disease associated, but we can be confident that some of those mark- regarding genotyping platforms to ensure data compatibility (Amos
ers are truly associated with disease (assuming the GWAS was ade- et al., 2016; Bennett et al., 2011). Collaborations across consortia can
quately powered). Similarly, a risk-​prediction model using only those also make use of economies of scale when ordering genotyping arrays
SNPs that reach genome-​wide significance may be less predictive than and assays.
a model that includes SNPs passing a lower significance threshold, Figure 5–​6 illustrates the impact the collaborative approach can
when the signal added by truly associated sub-​genome-​wide signifi- have on the pace of discovery, using breast cancer as an example. Over
cant SNPs overpowers the noise from non-​associated SNPs. Thus, it the first few years, several research groups undertook GWAS of breast
can be useful to consider the appropriate significance threshold for cancer, independently expanding the size of their distinct replication
genetic association studies as a decision theoretic problem, balancing efforts (both in terms of number of markers studied and sample size)
the relative costs of false positives and false negatives in a particular and publishing separately. This led to a steady stream of discoveries,
research setting. so that from 2007 to 2012 approximately 27 independent breast can-
cer risk loci had been discovered. By conducting a meta-​analysis of
existing GWAS and designing a genotyping array to follow up 29,807
Replication and Power in GWAS: The Importance promising SNPs in a large replication sample (45,290 cases and
of Collaboration 41,880 controls), researchers were able to more than double the num-
ber of identified loci in 2013 (Garcia-​Closas et al., 2013a; Michailidou
Given that many reported associations in early candidate gene studies
et al., 2013). Imputation in the replication sample further increased the
failed to replicate, the GWAS community quickly adopted standards to
power of the study, so that another 15 loci were discovered in 2015
maximize the chances of successful replication (NCI-​NHGRI Working
(Michailidou et al., 2015a). Finally, as part of the OncoArray initiative
Group on Replication in Association Studies et al., 2007). Replication
(Amos et al., 2016), investigators in more than 90 studies were able to
serves two primary goals: first, to establish that the reported asso-
double the sample size of the largest breast cancer GWAS conducted
ciation is real, and not solely due to random sampling; and second,
to date. This study is projected to almost double the number of known
to establish that the association is consistent across different studies
breast cancer risk loci.
and does not reflect a bias present in one particular study (Kraft et al.,
2009b).
Given the stringent significance thresholds needed to distinguish Imputation
chance from true associations (see previous section), and given
the expected modest effect sizes (see the section “Genome-​ Wide Genotype imputation is a method that can be used to estimate geno-
Association Studies: Results”), large sample sizes are needed to obtain types for SNPs that are not directly genotyped in the study sample
 61

Genetic Epidemiology of Cancer 61

2007
2017 (projected)
Easton (2007) Nature
150 Hunter (2007) Nat Genet
Stacey (2007) Nat Genet
2010
Stacey (2008) Nat Genet

Cumulative number of GWAS-discovered markers


Ahmed (2009) Nat Genet
Zheng (2009) Nat Genet
Thomas (2009) Nat Genet
Turnbull (2010) Nat Genet
100 Antoniou (2010) Nat Genet
2015
2012
Haiman (2011) Nat Genet
Siddiq (2012) Hum Mol Genet
Ghoussaini (2012) Nat Genet
2013 2013
Michailidou (2013) Nat Genet
50 2015
Michailidou (2015) Nat Genet

2012
2010
2007

1,000 10,000 100,000


Number of cases in initial discovery GWAS

Figure 5–​6.  Number of GWAS-​discovered breast-​cancer risk markers as a function of largest initial GWAS sample size. Projected numbers for 2017 are
based on K. Michailidou (2015).

(Howie et al., 2012; Li et al., 2010; Marchini and Howie, 2010; Zheng with an R2 > 0.8 using the 1,000 Genomes Project Phase 3 v5 ref-
et  al., 2011). Imputation in GWAS works because SNPs are often erence panel. (R2 is a metric that estimates the correlation between
correlated due to linkage disequilibrium (LD), so that missing SNP imputed SNP genotypes and true SNP genotypes.) Different eth-
genotypes can often be inferred with high accuracy. Typically, study nicities produce different qualities of imputation accuracy of com-
samples are genotyped in a large number of SNPs (e.g., in a GWAS), mon and rare variants. The HapMap and the 1000 Genomes Project
and missing SNPs are inferred from a reference panel of haplotypes. included samples from different ethnicity backgrounds, whereas the
To date, the reference populations most widely used are the HapMap Haplotype Reference Consortium reference panel consists largely of
Consortium project data (International HapMap, 2005) and the 1000 samples with mainly European ancestry.
Genomes Project data (Genomes Project Consortium, 2012). More
recently, the Haplotype Reference Consortium has released a refer-
ence panel of more than 32,000 sequenced samples (McCarthy et al., GENOME-​WIDE ASSOCIATION STUDIES: RESULTS
2016). The 1000 Genomes and the Haplotype Reference Consortium
data are based on whole genome sequencing of large number of sam- GWAS have been extremely successful at identifying common vari-
ples. Stretches of shared haplotypes are identified between the ref- ants with modest effects associated with risk of complex disease. As
erence panel and the study samples, and missing genotypes for each noted in an early review, “There have been few, if any, similar bursts
sample are then “filled in” by copying alleles in the matching refer- of discovery in the history of medical research” (Hunter and Kraft,
ence haplotypes (Li et  al., 2009). There are several reasons to use 2007). Cancer has been no exception. As of November 2016, over 200
genotype imputation, but a major use is to facilitate combining results published GWAS on more than 30 different cancers were listed in the
from different GWAS based on different genotyping platforms, since NHGRI-​ EBI Catalog of published GWAS (https://​www.ebi.ac.uk/​
results are then obtained for all SNPs in the reference panel. In addi- gwas/​). These papers reported 938 SNP-​cancer associations at the p <
tion, it increases the power of association analysis for studies based 5 × 10–​8 level of statistical significance. An exhaustive review of these
on a single platform and fine-​mapping studies, since an imputed SNP results is beyond the scope of this chapter—​and any detailed review
that is only partially correlated with typed SNPs may give a stronger would be out of date by the time it is published. Instead, we focus on
association than directly genotyped SNPs. Until recently, common general trends, illustrated by a few particular examples.
variants (minor allele frequency [MAF] > 0.01) could be reliably
imputed with high accuracy, whereas variants with MAF < 0.01 were
Effect Sizes for Common Alleles Are Modest
not imputed as well (Howie et al., 2012; Marchini and Howie, 2010).
Improvements to the different imputation algorithms, as well as the With few exceptions, the per-​allele odds ratio associated with common
increase in the numbers of the sequenced samples in the reference risk alleles (frequencies between 5% and 95%) are modest (Figure 5–​8).
panels, have increased the accuracy of the imputation of rarer variants Although the relatively small number of low-​frequency SNPs that
dramatically; variants down to MAF = 0.1% can now be imputed with have been identified through GWAS tend to have larger effect sizes,
high accuracy (Figure 5–​7) (Genomes Project Consortium, 2015; that may reflect the fact that the GWAS to date have only been pow-
McCarthy et al., 2016). In European-​ancestry samples, approximately ered to detect low frequency variants with large per-​allele odds ratios.
9.5 million SNPs can be imputed from commonly used GWAS arrays It is an open empirical question whether low frequency and rare alleles
62

62 Part I:  Basic Concepts


(a) One Thousand Genomes Project (b) Haplotype Reference Consortium

31.6 31.6
0.00–0.01
0.01–0.05
0.05–0.10

Number of Markers (millions)


Number of Markers (millions) 0.10–0.20
10.0 0.20–0.35 10.0
0.35–0.50

3.2 3.2

1.0 1.0

0.3 0.3

0.00 0.30 0.50 0.70 0.80 0.95 0.00 0.30 0.50 0.70 0.80 0.95
Rsq Threshold Rsq Threshold

Figure 5–​7.  Performance of imputation using (a) the 1000 Genomes Project (Phase 3) reference panel and (b) the Haplotype Reference Consortium panel,
for samples genotyped using the OncoArray (Amos et al., 2016). The y-​axis denotes the number of markers that can be imputed with estimated accuracy
(imputation R-​squared) at or above the specified threshold. Each line denotes the performance for markers in distinct minor allele frequency bins.

generally have larger effects than common variants (and, if so, how More Common Cancer Risk Alleles Have Yet to Be
much larger). Discovered
This is illustrated in Figure 5–​10, which presents results of a series of
Most Cancers Appear to Include a Polygenic simulated GWAS with increasing sample sizes for a cancer with a pol-
Component ygenic component. None of the 100 loci with a per allele-​odds ratio
of 1.05 was discovered in studies of 1000 or even 10,000 cases, due
For most of the cancers with at least one published genome-​wide sig- to low power. One risk allele reaches genome-​wide significance in the
nificant association, GWAS have identified at least 10 unique risk SNPs study with 50,000 cases, and 28 reach genome-​wide significance in
in a median of seven distinct risk loci (Table 5–​2). For the cancers that the study with 100,000 cases. There are two things to note from this
have been studied in the largest sample sizes (breast and prostate cancer), example. First, those 28 genome-​wide significant SNPs in the study
over 100 independent genome-​wide significant associations have been with 100,000 cases and 100,000 controls represent the “tip of the ice-
reported. The number of risk loci and the distribution of their effect sizes berg.” The power to detect a particular causal SNP in Figure  5–​10
vary somewhat across cancers, although because of differences in sample (panel d) is modest (about 25%); some luck was involved for these 28
sizes across cancer GWAS it is difficult to say how much the genetic archi- SNPs to achieve p < 5 × 10–​8. It is likely that SNPs with modest effects
tecture varies (Table 5–​2 and Figure 5–​9). Of particular note are ALL, (which had low power to be detected) are drawn from a larger pool
CLL, and testicular cancer, which have larger effect sizes on average (con- of SNPs, many (or most) of which have gone undetected. Second, as
sistent with their larger sibling recurrence risk ratios; see Table 5–​1). sample size increases, the distribution of p-​values for causal SNPs

4.0

3.5
Per-Allele Odds Ratio

3.0

2.5

2.0

1.5

1.0

0.0 0.2 0.4 0.6 0.8 1.0


Risk Allele Frequency

Figure 5–​8.  Per-​allele odds ratios of cancer risk alleles from published genome-​wide association studies (restricted to markers with association p < 5 ×
10–​8). Data from Burdett T (EBI), version v1.0 (accessed November 25, 2016).
 63

Genetic Epidemiology of Cancer 63


Table 5–​2.  Summary of Risk Markers from Published Genome-​Wide 2013; de los Campos et al., 2010; Finucane et al., 2015; Gusev et al.,
Association Studies (GWAS) of Cancer 2014; Vilhjalmsson et  al., 2015). Several lines of evidence suggest
that the scenario presented in Figure 5–​10 holds for many cancers,
Site npapers nSNPs nregions σg2 and that more common risk alleles with modest effects remain to be
discovered. Techniques that estimate hg2, the amount of variability in
ALL 8 19 10 0.58–​1.02 disease liability that could be explained by all the SNPs measured in a
Bladder 9 16 15 0.25–​0.17 GWAS, suggest that currently known genome-​wide significant SNPs
Breast 26 107 59 0.47–​0.82 account for a fraction of hg2 (Lu et al., 2014; Sampson et al., 2015).
Cervical 2 7 5 0.07–​0.08 Similarly, inverse-​ probability-​
of-​
discovery methods suggest that
CLL 5 31 23 0.77–​0.89 many common variants with effect sizes similar to those already dis-
CML 1 1 1 0.19–​0.19 covered remain unknown (Chatterjee et al., 2013; Krier et al., 2016;
Colorectal 20 55 39 0.30–​0.33 Park et al., 2011, 2012).
Melanoma 1 7 7 0.04–​0.04
Endometrial 1 1 1 0.02–​0.02
Esophageal 4 21 18 0.37–​0.39 Some Regions Harbor Multiple Statistically
Ewing sarcoma 1 3 3 0.39–​0.39 Independent Signals
Gastric 4 10 7 0.11–​0.18
Glioma 7 23 12 0.33–​0.39 The 8q24 region, for example, contains at least five SNPs that are
Hepatocellular carcinoma 3 5 4 0.19–​0.22 independently associated with prostate cancer risk (in the sense that
Hodgkin lymphoma 5 12 9 0.27–​0.26 the SNPs remain statistically significant after mutual adjustment in a
Lung 15 26 17 0.31–​0.39 multivariable regression model) (Al Olama et al., 2009; Gudmundsson
Lymphoma 8 23 12 0.58–​0.89 et  al., 2007a; Haiman et  al., 2007; Witte, 2007; Yeager et  al., 2007,
Multiple myeloma 3 10 7 0.17–​0.21 2009). This may indicate that there are distinct mechanisms whereby
Non-​melanoma skin 9 31 25 0.63–​0.66 different SNPs disrupt gene function or expression in the region
Ovarian 8 24 22 0.38–​0.37 (allelic heterogeneity), or it may indicate that the SNPs jointly tag a
Pancreatic 5 24 17 0.20–​0.23 rare variant with a larger effect (Anderson et al., 2011; Dickson et al.,
Prostate 24 142 77 0.78–​1.61 2010; Wray et al., 2011). More research is needed to understand the
Renal cell carcinoma 5 8 6 0.08–​0.05 functional mechanisms at these loci.
Testicular 7 24 20 0.89–​1.07
Thyroid 4 8 4 0.27–​0.47
Some Regions Harbor Variants Associated
npapers = number of distinct published GWAS containing at least one genetic association with Multiple Cancers and Other Complex Traits
with p < 5 × 10–​8; nSNPs = number of distinct single nucleotide polymorphisms with p <
5 × 10–​8 in at least one paper; nregions = number of distinct regions (cytobands) containing
The 8q24 region is again an example of this phenomenon, as it con-
at least one SNP with p < 5 × 10–​8 in at least one paper; σg2 = the sum of the effect tains SNPs associated with prostate, breast, colon, ovarian, thyroid,
sizes for the published risk markers, Σi 2 qi (1-​qi) βi2. The range for σg2 is defined by and kidney cancers and glioma (Ghoussaini et al., 2008; Yeager et al.,
first considering only one marker per region (the lower bound), then considering all the 2009). The region near the TERT and CLPTM1L genes also contains
unique markers in the regions, averaging the effect sizes for markers reported more than
once (the upper bound).
SNPs associated with lung, pancreatic, and at least four other distinct
ALL = acute lymphoblastic leukemia (childhood); CLL = chronic lymphocytic cancers (Wang et al., 2014). The HLA region and a region on 11q13
leukemia; CML = chronic myeloid leukemia also contain variants associated with multiple cancers (Chung and
Data from Burdett et al. (2016). Chanock, 2011).
The biological implications of these findings remain unclear.
Some of these regions contain multiple SNPs that are asso-
starts to differ from that of the background “noise” SNPs. This fact ciated with different cancers:  for example, rs4449583 at the
could be leveraged to improve risk prediction models (see later dis- TERT-​CLPTM1L locus is associated with prostate cancer, while
cussion) or to identify functional elements across the genome that rs13170453 at the same locus is associated with lung cancer (the
are more likely to harbor cancer-​associated alleles (Chatterjee et al., two SNPs are in low LD, r2 < 0.10) (Wang et  al., 2014). Some
regions contain individual SNPs that are associated with multiple
cancers: rs4430796 at HNF1B is associated with both prostate can-
cer and endometrial cancer (Gudmundsson et  al., 2007b; Spurdle
ALL et al., 2011). Some alleles that are associated with multiple cancers
CLL increase the risk of one cancer but lower the risk of another:  the
Testicular A allele of rs451360 increases risk of pancreatic and testicular can-
Esophageal cer, for example, but decreases risk of lung cancer (Wang et  al.,
Ovarian 2014). These findings may indicate that variants at these loci affect
Lung multiple cancer-​related pathways, or a single pathway that affects
Pancreatic
different cancers, some in different ways. Further work pinpointing
the causal variants in these regions and how they perturb biologic
Prostate
function is needed.
Colorectal
Cancer risk SNPs also co-​ localize with SNPs associated with
Breast other complex traits. For example, the G allele of rs4430796 near
HNF1B is associated with an increase in risk of type 2 diabetes, but
0.00 0.05 0.10 0.15 0.20 0.25 a decrease in prostate and endometrial cancer risks (Elliott et  al.,
Effect size: 2 q (1–q) β2 2010; Gudmundsson et  al., 2007b; Machiela et  al., 2012; Pierce
and Ahsan, 2010; Spurdle et al., 2011). The T allele of rs505922 is
Figure 5–​9.  Boxplots of effect sizes for cancer risk alleles from published a proxy for the “O” allele of the ABO gene and is associated with
GWAS (restricted to markers with association p < 5 × 10–​8). The effect size lower risk of pancreatic cancer, lower risk of venous thromboembo-
of a risk allele is its contribution to the variance in log odds of disease. lism (Germain et al., 2015; Wolpin et al., 2010), markedly higher lev-
ALL = acute lymphoblastic leukemia (childhood); CLL = chronic lymphocytic els of e-​selectin (Qi et al., 2010), and an increased risk of duodenal
leukemia; q = minor allele frequency; β = log per-​allele odds ratio. ulcer (Amundadottir et al., 2006; Tanikawa et al., 2012). The breast
Data from Burdett T (EBI), version v1.0 (accessed November 25, 2016). cancer risk SNP rs11075995 is located near FTO, but is apparently
64

64 Part I:  Basic Concepts


(a) n = 1,000 cases (b) n = 10,000 cases
12 12
100 causal loci,
10 MAF = 0.10, 10
–log 10 p-value OR = 1.04

–log 10 p-value
8 8

6 6

4 4

2 2
0 0

Position Position
(c) n = 50,000 cases (d) n = 100,000 cases
12 12

10 10
–log 10 p-value

–log 10 p-value
8 8

6 6

4 4

2 2

0 0

Position Position

Figure 5–​10.  Manhattan plots from four simulated GWAS. Each point represents one of 100,000 independent markers; highlighted points represent 100
true causal variants (with a minor allele frequency of 0.1 and per-​allele OR of 1.04); all other markers are assumed to have the same allele frequencies in
cases and controls. The x-​axis denotes order along the genome; the y-​axis is the –​log10 p-​value for association. Dashed line represents the genome-​wide
statistical significance threshold (p < 5 × 10–​8).

independent of the obesity-​related SNPs in that region (Garcia-​Closas The Causal Variant(s) at Most GWAS-​Identified Loci
et al., 2013a). Remain Unknown
Such findings raise intriguing biological hypotheses. They also
suggest that combining GWAS results across multiple cancers can Although there is evidence that SNPs in regulatory regions as a class
increase power to detect SNPs that are in fact associated with multiple are associated with cancer risk, identifying the specific variants that
cancers or cancer-​related traits (Bhattacharjee et al., 2012; Fehringer influence cancer development and how they do so remains a difficult
et al., 2016; Kar et al., 2016). This possibility is supported by cross-​ task. While linkage disequilibrium helps identify cancer-​associated
phenotype heritability analyses. These analyses estimate the correla- regions (by enabling cost-​effective design and accurate imputation), it
tion in SNP effects across cancers (and between cancers and other hinders the identification of the relevant functional SNPs within these
phenotypes). They show that many cancers share a genetic component regions. SNPs that are in strong LD will be statistically indistinguish-
with each other and other traits (e.g., lung cancer and smoking behav- able due to colinearity, even in large sample sizes. Modern statistical
ior), although the genetic correlation is not as strong as that observed approaches to “fine mapping” integrate association results from large
for some psychiatric traits (e.g., schizophrenia and bipolar disease) studies (typically GWAS meta-​analyses), linkage disequilibrium pat-
(Sampson et al., 2015). terns, and genomic functional annotations in order to identify a (hope-
fully small) set of SNPs likely to contain the causal variant(s) in a
region (Coetzee et  al., 2012; Kichaev et  al., 2014, 2016; Ward and
Most GWAS-​Identified Markers Do Not Clearly Tag Kellis, 2016). Still, even when these analyses return a small number
Coding Variants of candidate variants, additional functional analyses will be needed to
confirm that they are biologically related to cancer risk and to under-
As with other complex traits, the majority of SNPs that are associated stand the mechanism behind the association.
with cancer are not themselves in coding regions, nor are they indi-
vidually in strong linkage disequilibrium with coding variants. Instead,
these SNPs and their strong proxies are mostly found in putative regu- Most Genome-​Wide Association Studies Have Been
latory regions (Maurano et al., 2012; Schaub et al., 2012). Heritability
Conducted in European-​Ancestry Populations
enrichment analyses that use all GWAS markers (not only those reach-
ing genome-​wide significance) to partition the total genetic contribution This creates several important gaps in our knowledge of the genetic
to cancer liability across different regions of the genome (e.g., coding, epidemiology of cancer. To begin with, the impact on non-​European
promoter, UTR, repressed) have consistently found that regulatory ele- populations of markers discovered in Europeans is less clear. When
ments explain most of the genetic contribution to cancer risk. The 8q24 SNPs that are significantly associated with cancer in Europeans are
region yet again provides a concrete example of this phenomenon. This present in other ethnicities, some associations replicate (though often
region is “gene desert”:  all of the cancer-​associated SNPs there are with weaker magnitude of association), while some do not (Fejerman
about one million base pairs removed from the nearest gene. There is et  al., 2014; Han et  al., 2015b; Murillo-​Zamora et  al., 2013; Waters
some experimental evidence suggesting that these SNPs disrupt long-​ et al., 2009). Taken in aggregate, the variants discovered in European-​
range binding between the 8q24 region and MYC, an oncogene. ancestry populations are typically associated with cancer in other
 65

Genetic Epidemiology of Cancer 65


ethnic groups, although these associations (and hence their ability to other complex traits) were criticized for poor discriminatory power
discriminate cases and controls) are generally lower. As an example, and misleading presentation (conflating the statistical significance of
a score composed of 105 known prostate cancer risk SNPs (most dis- an association test with clinical utility) (Kraft et al., 2009a). The lim-
covered in European-​ancestry samples) was tested across multiple eth- ited discriminatory power in early reports was in large part due to the
nicities: “Comparing the highest to lowest risk score deciles, the OR relatively small number of genome-​wide significant markers known at
was 6.22 for non-​Hispanic whites, 5.82 for Latinos, 3.77 for African-​ the time. Now, for cancers with scores of genome-​wide significant risk
Americans, and 3.38 for East Asians” (Hoffmann et  al., 2015). The alleles, the discriminatory ability of genetic risk scores is on par with
lack of replication, or weaker associations, may be due to differences or surpasses the discriminatory ability of other widely used risk tools
in allele frequencies or LD patterns across different ethnic groups. or biomarkers. For example, the discrimination of 86 breast-​cancer
Moreover, European-​ancestry GWAS are underpowered to discover risk SNPs already surpasses that of the Gail model, based on estab-
variants that are rare in Europeans but common in non-​Europeans. lished (non-​genetic) breast cancer risk factors (area under the ROC
Studying non-​ European populations can increase power to detect curve of 0.63 for a model including 86 risk SNPs, compared to 0.59
these loci, identifying variants that are directly relevant to the popu- for the Gail model; Maas et al., 2016). The area under the ROC curve
lations where they are polymorphic and more generally in terms of for 133 prostate-​cancer risk SNPs is 0.68, equivalent to the area under
our understanding of cancer biology. Increasing effort is being made the curve for prostate-​specific antigen testing (Szulkin et al., 2015b;
to perform GWAS in non-​European ancestry populations, including Thompson et al., 2005).
those of African, East Asian, and Latino American ancestry. Several avenues for improving genetic risk-​prediction models are
currently being explored. First, larger GWAS sample sizes will lead
to greater power to identify new risk loci and sort signal from noise
APPLICATIONS OF GWAS RESULTS (Krier et al., 2016; Park et al., 2010, 2011). Second, more advanced
statistical methods can improve risk prediction. Several groups have
Although additional epidemiologic studies, bioinformatic analyses, proposed models that do not restrict to genome-​wide significant SNPs,
and biologic experiments are needed to understand the mechanisms either by raising the p-​value threshold for including markers in a risk
linking genetic variation to variation in cancer risks, GWAS have score or by applying penalized regression methods (Chatterjee et al.,
already led to biologic insights, and GWAS results may have immedi- 2013; Dudbridge, 2013, 2016; Vilhjalmsson et al., 2015). These meth-
ate clinical implications and may open up new approaches to under- ods can improve discrimination relative to a risk score that includes
standing cancer epidemiology. For example, GWAS have highlighted only genome-​wide significant SNPs by taking advantage of truly asso-
the importance of regulatory variation relative to protein-​coding varia- ciated SNPs that fail to reach genome-​wide significance (Figure 5–​10).
tion, overturning some of the common wisdom from the candidate However, large training samples are needed to realize this potential
gene era. Here we highlight two contemporary applications of GWAS gain, and care must be taken to avoid overfitting (ideally by reporting
results: genetic risk prediction and Mendelian Randomization studies. model performance in an independent test data set).
Most genetic prediction models assume that the effects of risk alleles
add within and across loci on the log relative risk scale. Relaxing this
assumption to allow for gene-​gene interactions (non-​additive effects)
Risk Prediction might improve model performance. However, the log additive model
Rare, high-​penetrance variants in cancer predisposition genes (whether appears to be a good fit to empirical data (Hsu et al., 2015; Jiao et al.,
identified via genetic testing or inferred based on family history) have 2012; Joshi et al., 2014; Lindstrom et al., 2012; Mavaddat et al., 2015),
been used to counsel men and women from high-​risk families regard- and including non-​additive effects (which will require very large sam-
ing their options for cancer screening and prevention for over 30 years ples sizes to detect and estimate accurately [Zuk et  al.,  2012]) may
(Biesecker et  al., 1993; Fitzgibbons et  al., 1987; Lynch HT, 1967; not greatly improve model discrimination (Aschard et al., 2012a). It is
Yandell DW et  al., 1989). Recently, Mary-​Claire King and others important to note that additivity on the log relative risk scale implies
have proposed the implementation of population screening for known that the relative risk of cancer increases exponentially with the number
cancer-​related risk variants (regardless of family history) (King et al., of risk alleles carried. This can lead to a small subset of individuals in
2014; Manchanda et al., 2015; Metcalfe et al., 2015; Plon, 2015); sep- the tails of the risk distribution with markedly elevated (or lowered)
arately, there are initiatives to expand standard testing to include less risk. For example, the 1% of men with the highest values of a 77-​SNP
well-​characterized cancer predisposition genes (Couch et  al., 2015; genetic risk score for prostate cancer are at 4.7 times the average risk
Easton et  al., 2015; Kapoor et  al., 2015; Tung et  al., 2015). These of prostate cancer (Eeles et al., 2013), and the 1% of women with the
proposals highlight several gaps in empirical data on rare variants in highest values of a 77-​SNP genetic risk score for breast cancer are at 3
cancer predisposition genes. First, for known deleterious mutations, times the average risk (Mavaddat et al., 2015).
current estimates of lifetime risk are largely based on high-​risk fami- Finally, it should be noted that the clinical or public health utility
lies with multiple affected family members. The impact of these muta- of a genetic risk prediction algorithm cannot be determined by any
tions in the general population remains unknown. Second, it remains one of the usual statistical metrics used to assess model performance
difficult to predict the impact of novel variants in known predisposition (area under the ROC curve; variance in log relative risks; prediction
genes. For example, while only 2% of BRCA1/​2 variants identified in r2; etc. [Chatterjee et al., 2016; Gail and Pfeiffer, 2005; Gerds et al.,
the context of genetic counseling are variants of unknown significance 2008; Kerr et  al., 2014; Pfeiffer and Gail,  2011]) taken in isolation.
(previously unreported variants with a predicted function that could be The utility will depend on the balance of risks, costs, and benefits asso-
deleterious or benign) (Eggington et al., 2014), currently 30%–​40% of ciated with a particular application of the risk model. The benefits of
variants identified are of unknown significance when large multi-​gene using predictive genetic profiles to define high-​risk subjects who might
panels are employed (Tung et al., 2015). Third, the evidence linking receive intervention (and low-​risk subjects who might not) should out-
some proposed predisposition genes to particular cancers is inconclu- weigh the costs of the testing and the intervention. This net benefit thus
sive. All of these questions are currently active areas of research. depends on the specifics of the proposed intervention. (“An expensive
The success of GWAS in identifying common risk alleles raises or invasive intervention, such as prophylactic surgery, to prevent a rare
the possibility that these alleles could also be used to predict absolute outcome requires a very high specificity. High specificity may not
risk of cancer, especially in the general-​population setting. Although be as important for a less expensive and less risky intervention, such
the effects of these common risk alleles are individually small, taken as providing additional encouragement to adopt a healthier lifestyle
together they can describe a larger gradient in disease risk. Typically, to individuals at high lifetime risk” [Kraft et al., 2009a]). Moreover,
genetic risk models combine information from independent genome-​ assessments of clinical or public health utility typically involve param-
wide significant SNPs in a genetic risk score: a sum of the number of eters beyond the risk model itself, such as uptake of testing, adherence
risk alleles an individual carries, weighted by the allele-​specific log to treatment or screening guidelines given genetic risk results, effi-
relative risks. Early reports of the genetic risk scores for cancer (and cacy of treatment or screening modality (which may itself depend on
6

66 Part I:  Basic Concepts


genetic risk), and so on (Chowdhury et al., 2013, 2015; Marcus et al., build comprehensive data sets of intermediate phenotypes for develop-
2016). Modeling the impact of genetic risk prediction in a variety of ment of better IVs.
settings is currently an area of active interest.

FUTURE DIRECTIONS
Mendelian Randomization Although the last 10  years have seen tremendous advances in our
Epidemiological studies are used to estimate associations between knowledge of the genetic epidemiology of cancer, many important
exposures of interest and cancer risk. However, these associations may questions remain open. To begin with, the known rare and common
be biased estimates of the effect of an exposure on cancer risk due risk variants represent only a part of the inherited genetic contribution
to unmeasured confounding and/​or reverse causality. Mendelian ran- to cancer risk; more risk variants have yet to be discovered. Several
domization (MR) is an approach that can be used to avoid such biases. lines of evidence suggest this. First, meaningfully larger GWAS still
MR methods can estimate a causal relationship between an exposure identify novel loci for most cancers. Second, the effect sizes for many
and a disease if the exposure has a known genetic determinant, which newly detected risk loci are small enough that luck played some role
is used as an instrumental variable (IV) for the exposure (Didelez and in their discovery, implying that there are many more loci with simi-
Sheehan, 2007; Lawlor, 2008). Genetic variants serve as good can- larly small effects yet to be discovered. Finally, the known loci do not
didate IVs because they are time-​invariant and randomly assigned at account for observed familial clustering: GWAS-​identified SNPs can
conception; thus, they are not susceptible to the effects of potentially explain 16% of the sibling relative risk of breast cancer, for example,
confounding environmental factors and are not affected by disease sta- with known rare variants accounting for approximately another 16%
tus. Rather than directly estimating the association between an expo- (Michailidou et al., 2015a). There are a number of reasons for this the
sure and outcome, MR involves estimating the association between gap between the inherited genetic component due to known genetic
the IV and the outcome, and using this estimate to infer the effect risk variants and that inferred from family studies, often referred to as
of the exposure on the outcome. These inferences are valid under the the “missing heritability” (Maher, 2008; Manolio et al., 2009).
assumptions that the IV is (1) predictive of the exposure, (2) indepen- Unknown common variants account for some of the “missing heri-
dent of confounders that bias the exposure–​outcome association, and tability,” as do unknown rare variants. How much of the “missing heri-
(3) independent of the outcome given the exposure and confounders tability” is due to undiscovered common risk variants and how much
of the exposure-​outcome association (Didelez and Sheehan, 2007; is due to undiscovered rare variants is an open empirical question.
Glymour et al., 2012; Lawlor, 2008). Current data are sparse and conflicting, largely due to the paucity of
Prior to the proliferation of GWAS, only a few MR studies had been large-​scale sequencing association studies of complex human diseases
reported, and these typically used IVs that were well-​described, single and traits. A recent study of over 15,000 subjects with whole-​genome
genetic predictors of exposures. As large-​scale GWAS revealed new and whole-​exome sequencing data (plus imputed genotypes on over
potential IVs for exposures and biomarkers of interest, the use of MR 111,000 subjects with GWAS data) found that lower-​frequency vari-
methods has grown rapidly. As of October 2016, at least 27 MR stud- ants do not have a major role in predisposition to type 2 diabetes
ies reporting significant effects of various exposures and biomarkers (Fuchsberger et al., 2016). On the other hand, targeted sequencing of
on cancer risk factors had been published (Boccia et al., 2009; Bonilla 63 regions (comprising 12 Mb of sequence) in 9200 men suggested
et al., 2013, 2016; Brennan et al., 2009; Carreras-​Torres et al., 2016; that SNPs with MAFs of 0.1%–​1% “explain a substantial fraction of
Day et al., 2015; Dixon et al., 2016; Gao et al., 2016; Guo et al., 2016; prostate cancer risk in men of African ancestry.” However, the same
Iles et  al., 2014; Khankari et  al., 2016a, 2016b; Lewis and Smith, study found little evidence that rare variants substantially contributed
2005; Nead et al., 2015; Nimptsch et al., 2015; Ojha et al., 2016; Ong to risk in other ethnicities (Mancuso et  al., 2016). A  clear answer
et al., 2016; Painter et al., 2016b; Taylor et al., 2017; Thompson et al., regarding the relative contribution of rare variation to population-​level
2016; Thrift et al., 2015a, 2015b; Walsh et al., 2015, 2016; Wang et al., variation in cancer risk will require very large samples to be sequenced
2011; Zhang B et al., 2015; Zhang C et al., 2015). These studies have (on the order of more than 25,000 cases and a similar number of con-
reported effects for age at onset of puberty, anthropometric traits, alco- trols; Zuk et al., 2014). Considering the substantial costs involved in
hol and coffee consumption, estradiol levels, polyunsaturated fatty acid sequencing this number of samples, future discovery efforts should
levels, vitamin D levels, and telomere length on various cancer types. take several complementary approaches, including additional GWAS
Methodological extensions of the MR method have also been (especially for cancers and racial/​ethnic populations where GWAS
developed, including (1) the use of multiple genetic variants as multi-​ sample sizes have been relatively modest), sequencing of high-​risk
SNP IVs (Palmer et  al., 2012; Pierce et  al., 2011); (2)  two-​sample families (especially those known not to segregate a known risk muta-
approaches in which IV associations with the exposure and cancer risk tion), and large-​scale sequencing efforts.
are measured in two independent data sets (Pierce and Burgess, 2013); Finally, for completeness, we note that some of the “missing herita-
(3) the use of summarized (rather than individual-​level) data to obtain bility” may be due to an apples-​to-​oranges comparison. The estimates
MR estimates (Burgess et  al., 2013); and (4)  methods for detecting from GWAS and sequencing studies of the genetic contribution of
violations of MR assumptions and reducing the impact of bias due to known, measured genetic variation to population variability in cancer
violations (Bowden et al., 2015a, 2016). risk and shared familial cancer risk (i.e., estimates of hg2) typically
MR has become a widely used methodology and has provided assume that variants interact additively; they do not include domi-
important insights regarding the causality of risk factors for cancer nance or epistatic (gene-​gene interaction) effects (Pharoah et al., 2002;
and many other disease phenotypes. “Hypothesis-​free” MR is a poten- Risch, 1990; Yang et al., 2011). Estimates of heritability from family
tial future direction involving mining of genotyping and phenotyping studies can include both dominance and epistatic effects. Estimates
data sets for evidence of causal relationships within biological net- from twin studies, for example, include both (Falconer and Mackay,
works without a priori specification of exposure or outcome (Evans 1996; Zuk et  al., 2012). Moreover, family studies are susceptible to
and Davey Smith, 2015). Relevant databases and methods are being bias from shared environment (Visscher et al., 2008). Thus, while pro-
developed (http://​www.mrbase.org/​) (Voight, 2014). MR is also being viding some guidance as to the genetic architecture of cancer, herita-
explored as a tool for informing drug discovery by its use in assessing bilities from family studies should not be taken as the final standard for
the probable efficacy of pursuing a target, investigating on-​and off-​ the success of current or future genetic discoveries.
target drug effects, and to better target existing agents through drug Aside from the remaining unknown variants associated with cancer
repurposing (Evans and Davey Smith, 2015). However, future progress risk, much remains to be learned about the genetic variants associated
depends on the continued expansion and improvement of methodology with cancer survival and prognosis, response to treatment, the interac-
to address issues arising from potential violations of assumptions in tions between genetic and environmental variation, and the biologic
the selection of IVs such as pleiotropy (Bowden et al., 2015b; Burgess, mechanisms linking these risk-​associated loci to cancer etiology. We
2015), and the advancement of high-​throughput omics technologies to discuss these areas in the next sections.
 67

Genetic Epidemiology of Cancer 67


Cancer Survival and “nurture” contribute to cancer initiation, growth, and prognosis.
Thus there is great interest in studying the joint effects of genetics and
Although it is well known that cancer risk has a heritable compo- environmental, lifestyle, and other non-​genetic exogenous and endog-
nent, little is known about the heritability of cancer progression and enous exposures using epidemiological data (Aschard et  al., 2012b;
ultimately cancer-​specific death. A  Swedish study (Lindstrom et  al., Chatterjee and Mukherjee, 2009; Garcia-​Closas et  al., 2011; Hutter
2007) of more than three million families, including one million cancer et al., 2013; Kraft and Hunter, 2009; Mechanic et al., 2012). Such stud-
cases, noted an increased risk for cancer-​specific death in children with ies can address at least three goals. First, by accounting for the pos-
poor parental survival compared to those with good parental survival sibility that genetic effects may differ across exposure strata, studies
for breast, colorectal, lung, and prostate cancer. In addition, a recent can leverage gene–​environment interaction to increase power to detect
study (Fejerman et  al., 2013)  in a Hispanic population showed that loci that standard marginal genetic association tests (which average
individuals with higher Indigenous American ancestry had increased genetic effects over exposure strata) fail to detect (Aschard et al., 2010,
risk of breast cancer–​specific mortality, and this association persisted 2013; Dai et al., 2012; Gauderman et al., 2013; Hsu et al., 2012; Kraft
after adjusting for demographic factors. et  al., 2007; Lindstrom et  al., 2009; Pare et  al., 2010; Soave et  al.,
Despite this, studies aiming at identifying specific genetic variants 2015). Second, a better understanding of the joint effects of genotypes
associated with cancer survival have been conflicting. Recognizing the and exposures can help identify individuals who might disproportion-
heterogeneity in prognosis among cancer patients, many studies have ately benefit from clinical interventions (or groups that might dispro-
focused on specific subgroups, defined by either disease characteris- portionately benefit from public health interventions) (Garcia-​Closas
tics or treatment. A GWAS of survival among 3494 colorectal cancer et  al., 2013b; Maas et  al., 2016; Siemiatycki and Thomas, 1981).
cases identified SNPs at 6p12.1 to be associated with survival among Finally, understanding the pattern of disease risk across strata defined
individuals with distant-​metastatic disease on a genome-​wide level by genotypes and exposures may provide some insight into the bio-
(HR = 1.8; 95% CI: 1.5, 2.2; p = 7.6 × 10–​10); however, these SNPs were logical mechanisms through which genotypes and exposures influence
not significant among all cases (Phipps et al., 2016). SNPs at 5q23.2 disease.
and 6q21 were found to be associated with overall survival among The study of gene–​environment interaction in epidemiological stud-
1537 diffuse large B-​cell lymphoma patients treated with immunoche- ies often focuses on statistical interaction, but it is important to note
motherapy (HR = 1.49; 95% CI: 1.29, 1.72; p = 3.53 × 10–​8), although that statistical interaction alone provides, at most, circumstantial sup-
the risk estimates for disease-​specific survival were lower and did not port for any of these three goals (Kraft and Hunter, 2009; Siemiatycki
reach genome-​wide significance (Ghesquieres et al., 2015). For breast and Thomas, 1981; Thompson, 1991). Statistical interaction refers to
cancer, variants on 11q24 (Guo et al., 2015) have been associated with departures from additivity on a particular outcome scale. Changing
survival among 6881 cases with estrogen receptor negative (ER–​) dis- the outcome scale typically changes inference regarding the presence
ease (HR = 1.95; 95% CI = 1.55, 2.47; p = 1.91 × 10–​8). or absence of statistical interaction. For example, in the top two pan-
All of these findings have not yet been replicated in independent stud- els of Figure 5–​11, there is no interaction on the absolute risk scale,
ies. More evidence will be needed to confidently confirm them. Survival which implies that there is an antagonistic interaction on the log odds
GWAS for ovarian (Johnatty et al., 2015), prostate (Szulkin et al., 2015a), scale. Similarly, in the bottom panels, there is no interaction on the log
lung (Wu X et al., 2013), and pancreatic (Wu et al., 2014) cancer have odds scale, which implies that there is a synergistic interaction on the
all failed in returning genome-​wide significant findings. It is important absolute risk scale. The interpretation of a statistical interaction thus
to note that GWAS of cancer survival are lagging cancer risk GWAS in depends on the choice of outcome scale, and this scale should be cho-
terms of sample sizes. In addition, they are struggling with additional sen with the research goals in mind.
heterogeneity issues due to difference in treatment and disease stage. As In the context of locus discovery, the goal is not to identify statistical
sample sizes become larger, we will be able to maintain adequate sta- interaction per se, but to maximize power to detect loci associated with
tistical power while stratifying patients according to treatment or tumor disease. This has led some to propose scale-​free joint tests of genetic
characteristics; thus, novel loci for cancer survival are likely to emerge. main effects and interactions, which are geared to detect markers that
are associated with disease in at least one exposure stratum (Aschard
et al., 2010, 2013; Kraft et al., 2007; Lindstrom et al., 2009; Pare et al.,
Pharmacogenetics and Treatment Response 2010; Soave et al., 2015). Such tests are well powered across a wide
Pharmacogenetics, the study of the relationship between genetics range of alternative scenarios, but may not be optimal to identify those
and drug response, has shown that variants within drug-​metabolizing markers that interact with exposure but have modest marginal effects
genes can influence the rate at which the drug is converted to its metab- (i.e., markers that a standard GWAS might fail to identify). Developing
olites, thus impacting its effectiveness or leading to unwanted side tests that maximize power to detect statistical interactions (typically
effects. Most cancer genetic research regarding treatment response has departures from additivity on the log odds scale) is an active area of
focused on somatic mutations. However, there is compelling evidence research (Boonstra et al., 2016; Cornelis et al., 2012; Dai et al., 2012;
that inherited genetics may influence the efficacy of cancer therapy and Gauderman et  al., 2013; Hsu et  al., 2012; Mukherjee et  al., 2012;
management (Bell et al., 2015; Patel et al., 2013; Walko and McLeod, Thomas et al., 2012).
2014). Currently, a handful of anticancer drugs have labels related to When the goal is to identify individuals or groups who might dis-
specific germline genomic loci and outcomes (Pesenti et al., 2015). proportionately benefit from an intervention, the absolute risk scale
As an example of the potential impact of germline variation, a recent is the natural choice: departures from additivity in this case implies
study determined that men with prostate cancer with germline (or that the change in disease risk due to a change in exposure depends on
somatic) mutations in BRCA2 and other DNA repair genes were much individuals’ genotypes (see Figure 5–​11, panel [c]). Consequently, the
more likely to respond to a PARP inhibitor, olaparib, than those without absence of interactions on the log-​odds scale (as is typically reported)
a DNA repair mutation (88% vs. 4% of patients) (Mateo et al., 2015). is potentially an interesting result, as it implies a supra-​additive inter-
In addition to impacting the response of drugs, SNPs may influence action on the absolute risk scale.
the response to other types of cancer therapy, including radiation. The Finally, there is no scale that would allow one to make universal
field of radiogenomics focuses on the impact of genetic variants on inferences about the underlying biological mechanisms through which
normal tissue radiosensitivity, which may result in decreases in quality genotype and exposure influence disease; statistical interaction need
of life and disability (Kerns et al., 2014, 2015). not imply biological interaction, and vice versa (Siemiatycki and
Thomas, 1981; Thompson, 1991). Ideally, in the setting where the
putative biological effects of genotype and exposure are known or sus-
Gene–​Environment Interactions pected, one could develop a falsifiable hypothesis about what the dis-
tribution of disease risks would look like across genotype-​by-​exposure
“Gene–​environment interaction” is widely accepted to be ubiquitous in strata, and then test whether the observed data were consistent with
the development of most cancers, in the broad sense that both “nature” that hypothesis. But in the setting of a GWAS, where the biologic
68

68 Part I:  Basic Concepts


(a) (b)

0.5 0

Log odds of disease


Risk of disease
Carrier

Noncarrier

0 –3

Unexposed Exposed Unexposed Exposed

(c) (d)

0
0.5

Log odds of disease


Risk of disease

0 –3

Unexposed Exposed Unexposed Exposed

Figure 5–​11.  Two gene–​environment interaction scenarios for a binary disease, binary genotype, and binary exposure, and their representations on two
different scales. Panels (a) and (b) assume “no additive interaction” (i.e., no departure from an additive gene–​environment interaction model on the absolute
scale). Panel (a) shows this scenario on the absolute risk scale; panel (b) shows the same four disease probabilities on the log odds scale. Panels (c) and
(d) assume “no multiplicative interaction” (i.e., no departure from an additive gene–​environment interaction model on the log odds scale). Panel (c) shows
this scenario on the absolute risk scale, and panel (d) shows it on the log odds scale.

function of a tested SNP is typically unknown, it is very hard to infer a 2014; Figueroa et al., 2014; Garcia-​Albeniz et al., 2016; Gong et al.,
SNP’s biologic function from the presence of statistical SNP–​exposure 2016; Rudolph et al., 2013; Wu et al., 2012). Evidence for departures
interaction. from a multiplicative gene–​environment odds ratio model involving
Empirical results on gene–​environment interactions in cancer have GWAS-​identified risk variants has also been modest, either consider-
been mixed. There are a handful of long-​established examples from ing risk variants alone or aggregated in a genetic risk score (Campa
candidate gene studies involving variants known to alter the metabo- et al., 2011; Joshi et al., 2014; Kantor et al., 2014; Lindstrom et al.,
lism of a carcinogen. These include a variant in NAT2 interacting with 2011; Nickels et al., 2013; Pearce et al., 2013; Rudolph et al., 2016).
smoking exposure to influence risk of bladder cancer (Garcia-​Closas However, as noted earlier, absence of departures from additivity on
et al., 2005, 2013b; Rothman et al., 2007) and variants in ALDH and the log-​odds scale typically implies departures from additivity on
ADH genes interacting with alcohol to influence risk of esophageal the absolute risk scale, and this has potential implications for risk-​
squamous cell carcinoma (Brooks et al., 2009; Cui et al., 2009; Hashibe stratified public health and clinical interventions (Garcia-​Closas et al.,
et al., 2008; Wu et al., 2012). A review of the candidate gene literature 2013b; Maas et al., 2016; Siemiatycki and Thomas, 1981). There are
prior to 2013 found little compelling evidence for gene–​environment a number of possible reasons for the relative paucity of identified
interactions outside of these two examples, although modest sample gene–​environment interactions in cancer, including the need for large
sizes, combined with gaps in the studied genes, exposures, and can- sample sizes to detect modest interaction effects, exposure measure-
cers, make these results difficult to generalize (Simonds et al., 2016). ment error, lack of data on exposure during relevant time windows,
Genome-​ wide screens for variants involved in gene–​ environment and limited range of exposure variability in the populations studied
interactions in cancer have been inconclusive. A number of possible to date (Hutter et al., 2013; Kraft and Aschard, 2015; Simonds et al.,
interactions have been identified, including several affecting risk of 2016). Efforts to improve the power of gene–​environment interaction
colorectal cancer:  an interaction between a variant near CYP24A1 studies in cancer have largely focused on increasing the sample size,
and hormone therapy affecting colorectal cancer risk (Garcia-​Albeniz study diversity, and exposure measurement. A  better understanding
et  al., 2016)  and an interaction between a variant near MGST1 and of the biologic mechanisms driving known population-​level associa-
aspirin use (Nan et  al., 2015). Most of the suggestive interactions tions may also help identify specific exposures likely to be involved in
from GWAS need to be replicated (Du et al., 2014; Figueiredo et al., gene–​environment interactions.
 69

Genetic Epidemiology of Cancer 69


Biological Mechanisms familial risk (“missing heritability”). This suggests that a range of
study designs is needed to discover new risk variants, including larger
Now that hundreds of risk alleles have been associated with cancer GWAS (especially for non-​European ancestry populations and for
risk, the current challenge is to determine the biological function of cancers where the current sample sizes are small) and population-​
these variants. The significant variants identified from GWAS may and family-​based whole-​exome and whole-​genome sequencing stud-
not actually be the causal SNPs; it is likely that they are only cor- ies. Methodological lessons from the last 10  years—​including the
related with the true causal SNP. In an attempt to identify the causal importance of well-​powered studies, and appropriate consideration of
variant, fine mapping of the region is performed (Darabi et al., 2015; multiple testing or low priors—​will continue to be relevant. Although
Edwards et  al., 2013; Ghoussaini et  al., 2016; Glubb et  al., 2015; current costs for whole-​exome and whole-​genome sequencing make
Meyer et al., 2013; Painter et al., 2016a). This can be accomplished some study designs prohibitively expensive now, a combination of
using publicly available resources such as the 1000 Genomes Project, clever use of existing data and alternate technologies (e.g., impu-
as well as by performing additional resequencing or dense genotyping tation) and technological developments (cheaper sequencing) will
of the region. With these data, all SNPs in LD with the GWAS vari- provide opportunities for future advances. The next 10 years should
ant will be identified. Depending on the LD structure, this may result bring quantitative and qualitative changes to our understanding of the
in numerous plausible causal variants. The differences in LD struc- inherited genetic contribution to cancer risk, as we discover more risk
ture across ethnic groups can potentially help to narrow the region variants and better understand their biologic effects.
of interest (Franceschini et  al., 2016; Han et  al., 2015a; Lindstrom
et al., 2016; Liu et al., 2014; Wu Y et al., 2013). Additionally, detailed References
annotation of the functional elements of the genome (enhancers, tran-
scription factor binding sites, etc.) is available through resources like Ahmed S, Thomas G, Ghoussaini M, et al. 2009. Newly discovered breast can-
cer susceptibility loci on 3p24 and 17q23.2. Nat Genet, 41(5), 585–​590.
ENCODE (Encode Project Consortium, 2012; Schaub et al., 2012).
PMCID: PMC2748125.
Tools such as PAINTOR (Kichaev et  al., 2014, 2016), HaploReg Al Olama AA, Kote-​Jarai Z, Giles GG, et al. 2009. Multiple loci on 8q24 asso-
(Ward and Kellis, 2016), and funciSNP (Coetzee et al., 2012; Hazelett ciated with prostate cancer susceptibility. Nat Genet, 41(10), 1058–​1060.
et al., 2014; Rhie et al., 2013) incorporate information on the strength PMID: 19767752.
of the association and the potential function of the genomic location Amos CI, Dennis J, Wang Z, et al. 2016. The OncoArray Consortium: a net-
in order to prioritize candidates within the identified list of possible work for understanding the genetic architecture of common cancers.
causal variants. Cancer Epidemiol Biomarkers Prev; 26(1), 126–​135. PMID: 27697780.
The next step is to characterize the function of the putative causal Amundadottir LT, Sulem P, Gudmundsson J, et  al. 2006. A common variant
variants (Freedman et al., 2011). Functional characterization of these associated with prostate cancer in European and African populations. Nat
Genet, 38(6), 652–​658. PMID: 16682969.
SNPs can involve many different types of studies and data. The major-
Amundadottir LT, Thorvaldsson S, Gudbjartsson DF, et al. 2004. Cancer as a
ity of significant GWAS loci are in non-​coding regions, so their func- complex phenotype: pattern of cancer distribution within and beyond the
tion and the gene they impact are not generally obvious. However, nuclear family. PLoS Med, 1(3), e65. PMCID: PMC539051.
that many are located in potential gene regulatory regions helps to Anderson CA, Soranzo N, Zeggini E, and Barrett JC. 2011. Synthetic asso-
guide functional studies. Many risk variants function as expression ciations are unlikely to account for many common disease genome-​wide
Quantitative Trait Loci (eQTLs), loci that are associated with tran- association signals. PLoS Biol, 9(1), e1000580. PMCID: PMC3022527.
script abundance. eQTLs can be either located near the gene they Antoniou AC, and Easton DF. 2003. Polygenic inheritance of breast can-
influence, or far from it (even on a different chromosome); however, cer:  Implications for design of association studies. Genet Epidemiol,
eQTLs near a gene typically have a larger effect than those that are 25(3), 190–​202. PMID: 14557987.
Antoniou AC, Wang X, Fredericksen ZS, et al. 2010. A locus on 19p13 mod-
distant. An important consideration is that many eQTLs may be tissue
ifies risk of breast cancer in BRCA1 mutation carriers and is associated
specific. The GTEx (Genotype-​Tissue Expression) project has created with hormone receptor-​negative breast cancer in the general population.
a publicly available database of gene expression across numerous nor- Nat Genet, 42(10), 885–​892. PMCID: PMC3130795.
mal tissue types to address the issue of tissue specificity (http://​www. Aschard H, Hancock DB, London SJ, and Kraft P. 2010. Genome-​wide meta-​
gtexportal.org/​home/​) (The GTEx Consortium, 2015). However, gen- analysis of joint tests for genetic and gene-​ environment interaction
eration of data for additional tissue and cell types may be necessary effects. Hum Hered, 70(4), 292–​300. PMCID: PMC3085519.
depending on the cancer being studied. Another consideration is that Aschard H, Chen J, Cornelis MC, et  al. 2012a. Inclusion of gene-​gene and
eQTLs may be context and timing specific, such that it is difficult to gene-​ environment interactions unlikely to dramatically improve risk
obtain data from the relevant time period and environmental condi- prediction for complex diseases. Am J Hum Genet, 90(6), 962–​972.
PMCID: PMC3370279.
tions. Once a candidate gene is identified, more functional studies,
Aschard H, Lutz S, Maus B, et  al. 2012b. Challenges and opportunities in
including in vitro cancer models to determine the gene’s impact on the genome-​wide environmental interaction (GWEI) studies. Hum Genet,
cells behavior, are warranted. 131(10), 1591–​1613. PMCID: PMC3677711.
Aschard H, Vilhjalmsson BJ, Joshi AD, Price AL, and Kraft P. 2015. Adjusting
for heritable covariates can bias effect estimates in genome-​wide associ-
SUMMARY ation studies. Am J Hum Genet, 96(2), 329–​339. PMCID: PMC4320269.
Aschard H, Zaitlen N, Tamimi RM, Lindstrom S, and Kraft P. 2013. A non-
Our knowledge of the inherited genetic contribution to cancer risk parametric test to detect quantitative trait loci where the phenotypic
has expanded tremendously over the last 10 years, from a handful of distribution differs by genotypes. Genet Epidemiol, 37(4), 323–​ 333.
very rare high-​risk mutations affecting a small proportion of the pop- PMCID: PMC4088942.
Bardella C, Pollard PJ, and Tomlinson I. 2011. SDH mutations in cancer.
ulation to hundreds of modest-​effect alleles influencing risk across
Biochim Biophys Acta, 1807(11), 1432–​1443. PMID: 21771581.
the entire population. These advances have opened up many new Bell GC, Donovan KA, and McLeod HL. 2015. Clinical implications of opi-
research questions, from basic biology—​understanding the mecha- oid pharmacogenomics in patients with cancer. Cancer Control, 22(4),
nisms through which genetic variation influences carcinogenesis and 426–​432. PMID: 26678969.
progression—​to clinical and public health practice—​for example, Bennett SN, Caporaso N, Fitzpatrick AL, et al. 2011. Phenotype harmonization
using genetic risk information for precision prevention, screening, or and cross-​study collaboration in GWAS consortia: the GENEVA experi-
treatment. At the same time, much remains to be learned about the ence. Genet Epidemiol, 35(3), 159–​173. PMCID: PMC3055921.
genetic architecture of cancer: much of the apparent inherited com- Bhat GA, Shah IA, Rafiq R, et al. 2015. Family history of cancer and the risk of
ponent of risk remains unidentified, even for cancers that have been squamous cell carcinoma of oesophagus: a case-​control study in Kashmir,
India. Br J Cancer, 113(3), 524–​532. PMCID: PMC4522628.
studied in very large sample sizes, and the genetic contribution to can-
Bhattacharjee S, Rajaraman P, Jacobs KB, et al. 2012. A subset-​based approach
cer risk in non-​European populations remains understudied. Current improves power and interpretation for the combined analysis of genetic
evidence is consistent with the hypothesis that a mixture of rare high-​ association studies of heterogeneous traits. Am J Hum Genet, 90(5), 821–​
risk and common moderate-​risk alleles contributes to the unidentified 835. PMCID: PMC3376551.
70

70 Part I:  Basic Concepts


Biesecker BB, Boehnke M, Calzone K, et  al. 1993. Genetic counseling for Coetzee SG, Rhie SK, Berman BP, Coetzee GA, and Noushmehr H. 2012.
families with inherited susceptibility to breast and ovarian cancer. JAMA, FunciSNP:  an R/​ bioconductor tool integrating functional non-​ coding
269(15), 1970–​1974. PMID: 8352830. data sets with genetic association studies to identify candidate regulatory
Boccia S, Hashibe M, Galli P, et al. 2009. Aldehyde dehydrogenase 2 and head SNPs. Nucleic Acids Res, 40(18), e139. PMCID: PMC3467035.
and neck cancer:  a meta-​analysis implementing a Mendelian random- Collaborative Group on Hormonal Factors in Breast Cancer. 2001. Familial
ization approach. Cancer Epidemiol Biomarkers Prev, 18(1), 248–​254. breast cancer:  collaborative reanalysis of individual data from 52 epi-
PMID: 19124505. demiological studies including 58,209 women with breast cancer and
Bodmer W, and Bonilla C. 2008. Common and rare variants in multifacto- 101,986 women without the disease. Lancet, 358(9291), 1389–​1399.
rial susceptibility to common diseases. Nat Genet, 40(6), 695–​ 701. PMID: 11705483.
PMCID: PMC2527050. Cordell HJ, and Clayton DG. 2005. Genetic association studies. Lancet,
Bonilla C, Gilbert R, Kemp JP, et al. 2013. Using genetic proxies for lifecourse 366(9491), 1121–​1131. PMID: 16182901.
sun exposure to assess the causal relationship of sun exposure with circu- Cornelis MC, Tchetgen EJ, Liang L, et al. 2012. Gene-​environment interactions
lating vitamin d and prostate cancer risk. Cancer Epidemiol Biomarkers in genome-​wide association studies: a comparative study of tests applied
Prev, 22(4), 597–​606. PMCID: PMC3616836. to empirical studies of type 2 diabetes. Am J Epidemiol, 175(3), 191–​202.
Bonilla C, Lewis SJ, Martin RM, et al. 2016. Pubertal development and prostate PMCID: PMC3261439.
cancer risk: Mendelian randomization study in a population-​based cohort. Couch FJ, Hart SN, Sharma P, et  al. 2015. Inherited mutations in 17 breast
BMC Med, 14, 66. PMCID: PMC4820939. cancer susceptibility genes among a large triple-​negative breast cancer
Boonstra PS, Mukherjee B, Gruber SB, et al. 2016. Tests for gene-​environment cohort unselected for family history of breast cancer. J Clin Oncol, 33(4),
interactions and joint effects with exposure misclassification. Am J 304–​311. PMCID: PMC4302212.
Epidemiol, 183(3), 237–​247. PMCID: PMC4724093. Cox A, Dunning AM, Garcia-​Closas M, et al. 2007. A common coding variant
Bowden J, Davey Smith G, and Burgess S. 2015a. Mendelian randomization with in CASP8 is associated with breast cancer risk. Nat Genet, 39(3), 352–​
invalid instruments:  effect estimation and bias detection through Egger 358. PMID: 17293864.
regression. Int J Epidemiol, 44(2), 512–​525. PMCID: PMC4469799. Cui R, Kamatani Y, Takahashi A, et al. 2009. Functional variants in ADH1B
Bowden J, Davey Smith G, Haycock PC, and Burgess S. 2016. Consistent and ALDH2 coupled with alcohol and smoking synergistically
estimation in mendelian randomization with some invalid instruments enhance esophageal cancer risk. Gastroenterology, 137(5), 1768–​1775.
using a weighted median estimator. Genet Epidemiol, 40(4), 304–​314. PMID: 19698717.
PMCID: PMC4849733. Dai JY, Logsdon BA, Huang Y, et al. 2012. Simultaneously testing for marginal
Bowden J, Smith GD, and Burgess S. 2015b. Mendelian randomiza- genetic association and gene-​environment interaction. Am J Epidemiol,
tion with invalid instruments:  effect estimation and bias detec- 176(2), 164–​173. PMCID: PMC3499112.
tion through Egger regression. Int J Epidemiol, 44(2), 512–​ 525. Darabi H, McCue K, Beesley J, et  al. 2015. Polymorphisms in a putative
PMID: WOS:000357106100015. enhancer at the 10q21.2 breast cancer risk locus regulate NRBF2 expres-
Brennan P, McKay J, Moore L, et al. 2009. Obesity and cancer: Mendelian ran- sion. Am J Hum Genet, 97(1), 22–​34. PMCID: PMC4572510.
domization approach utilizing the FTO genotype. Int J Epidemiol, 38(4), Day FR, Ruth KS, Thompson DJ, et al. 2015. Large-​scale genomic analyses
971–​975. PMCID: PMC2734066. link reproductive aging to hypothalamic signaling, breast cancer suscepti-
Broca P. 1866. Traité des tumeurs, vols, 1, 2. Paris: Asselin. bility and BRCA1-​mediated DNA repair. Nat Genet, 47(11), 1294–​1303.
Brooks PJ, Enoch MA, Goldman D, Li TK, and Yokoyama A. 2009. The alco- PMCID: PMC4661791.
hol flushing response: an unrecognized risk factor for esophageal cancer Day FR, Loh PR, Scott RA, Ong KK, and Perry JR. 2016. A robust example
from alcohol consumption. PLoS Med, 6(3), e50. PMCID: PMC2659709. of collider bias in a genetic association study. Am J Hum Genet, 98(2),
Burdett T, Hall PN, Hastings E, et al. The NHGRI-​EBI Catalog of Published 392–​393. PMCID: PMC4746366.
Genome-​Wide Association Studies, version v1.0., http://​www.ebi.ac.uk/​ de los Campos G, Gianola D, and Allison DB. 2010. Predicting genetic pre-
gwas/​, accessed November 25, 2016. disposition in humans: the promise of whole-​genome markers. Nat Rev
Burgess S, Butterworth A, and Thompson SG. 2013. Mendelian randomiza- Genet, 11(12), 880–​886. PMID: 21045869.
tion analysis with multiple genetic variants using summarized data. Genet Diabetes Genetics Initiative of Broad Institute of Harvard and MIT, Lund
Epidemiol, 37(7), 658–​665. PMID: WOS:000325980600002. University, and Novartis Institutes of BioMedical Research et  al.
Burgess S, Bowden J. 2015. Integrating summarized data from multiple genetic 2007. Genome-​wide association analysis identifies loci for type 2
variants in Mendelian randomization:  bias and coverage properties of diabetes and triglyceride levels. Science, 316(5829), 1331–​ 1336.
inverse-​variance weighted methods. ArXiv Prepr. ArXiv151204486. PMID: 17463246.
Campa D, Kaaks R, Le Marchand L, et  al. 2011. Interactions between Dickson SP, Wang K, Krantz I, Hakonarson H, and Goldstein DB. 2010. Rare
genetic variants and breast cancer risk factors in the breast and pros- variants create synthetic genome-​ wide associations. PLoS Biol, 8(1),
tate cancer cohort consortium. J Natl Cancer Inst, 103(16), 1252–​1263. e1000294. PMCID: PMC2811148.
PMCID: PMC3156803. Didelez V, and Sheehan N. 2007. Mendelian randomization as an instrumental
Carreras-​Torres R, Haycock PC, Relton CL, et al. 2016. The causal relevance of variable approach to causal inference. Stat Methods Med Res, 16(4), 309–​
body mass index in different histological types of lung cancer: a Mendelian 330. PMID: WOS:000248753000001.
randomization study. Sci Rep, 6, 31121. PMCID: PMC4973233. Dixon SC, Nagle CM, Thrift AP, et al. 2016. Adult body mass index and risk
Chang ET, Smedby KE, Hjalgrim H, et  al. 2005. Family history of hemato- of ovarian cancer by subtype:  a Mendelian randomization study. Int J
poietic malignancy and risk of lymphoma. J Natl Cancer Inst, 97(19), Epidemiol, 45(3), 884–​895. PMID: 27401727.
1466–​1474. PMID: 16204696. Du M, Zhang X, Hoffmeister M, et  al. 2014. No evidence of gene-​calcium
Chatterjee N, and Mukherjee B. 2009. Statistical approaches to studies of gene-​ interactions from genome-​ wide analysis of colorectal cancer risk.
gene and gene-​environment interactions. In: Khoury MJ, Bedrosian S, Gwinn Cancer Epidemiol Biomarkers Prev, 23(12), 2971–​ 2976. PMCID:
M, Higgins J, Ioannidis J, Little J. (Eds.), Human Genome Epidemiology, PMC4257872.
2nd ed.: Building the Evidence for Using Genetic Information to Improve Dudbridge F. 2013. Power and predictive accuracy of polygenic risk scores.
Health and Prevent Disease. New York: Oxford University Press. PLoS Genet, 9(3), e1003348. PMCID: PMC3605113.
Chatterjee N, Shi J, and Garcia-​Closas M. 2016. Developing and evaluating Dudbridge F. 2016. Polygenic epidemiology. Genet Epidemiol, 40(4), 268–​
polygenic risk prediction models for stratified disease prevention. Nat Rev 272. PMCID: PMC4982028.
Genet, 17(7), 392–​406. PMID: 27140283. Dudbridge F, and Gusnanto A. 2008. Estimation of significance thresholds
Chatterjee N, Wheeler B, Sampson J, et al. 2013. Projecting the performance of for genomewide association scans. Genet Epidemiol, 32(3), 227–​234.
risk prediction based on polygenic analyses of genome-​wide association PMCID: PMC2573032.
studies. Nat Genet, 45(4), 400–​405. PMCID: PMC3729116. Easton DF, Pharoah PD, Antoniou AC et al. 2015. Gene-​panel sequencing and
Chowdhury S, Dent T, Pashayan N, et al. 2013. Incorporating genomics into the prediction of breast-​cancer risk. N Engl J Med, 372(23), 2243–​2257.
breast and prostate cancer screening:  assessing the implications. Genet PMCID: PMC4610139.
Med, 15(6), 423–​432. PMCID: PMC3941015. Easton DF, Pooley KA, Dunning AM, et al. 2007. Genome-​wide association
Chowdhury S, Henneman L, Dent T, et al. 2015. Do health professionals need study identifies novel breast cancer susceptibility loci. Nature, 447(7148),
additional competencies for stratified cancer prevention based on genetic 1087–​1093. PMCID: PMC2714974.
risk profiling? J Pers Med, 5(2), 191–​212. PMCID: PMC4493496. Edwards SL, Beesley J, French JD, and Dunning AM. 2013. Beyond
Chung CC, and Chanock SJ. 2011. Current status of genome-​wide association GWASs:  illuminating the dark road from association to function. Am J
studies in cancer. Hum Genet, 130(1), 59–​78. PMID: 21678065. Hum Genet, 93(5), 779–​797. PMCID: PMC3824120.
 71

Genetic Epidemiology of Cancer 71


Eeles RA, Olama AA, Benlloch S, et al. 2013. Identification of 23 new prostate Garcia-​Closas M, Rothman N, Figueroa JD, et  al. 2013b. Common genetic
cancer susceptibility loci using the iCOGS custom genotyping array. Nat polymorphisms modify the effect of smoking on absolute risk of bladder
Genet, 45(4), 385–​391, 391e381–​382. PMCID: PMC3832790. cancer. Cancer Res, 73(7), 2211–​2220. PMCID: PMC3688270.
Eggington JM, Bowles KR, Moyes K, et al. 2014. A comprehensive laboratory-​ Gauderman WJ, Zhang P, Morrison JL, and Lewinger JP. 2013. Finding novel
based program for classification of variants of uncertain significance in genes by testing G x E interactions in a genome-​wide association study.
hereditary cancer genes. Clin Genet, 86(3), 229–​237. PMID: 24304220. Genet Epidemiol, 37(6), 603–​613. PMCID: PMC4348012.
Elliott KS, Zeggini E, McCarthy MI, et  al. 2010. Evaluation of association Genomes Project Consortium, Abecasis GR, Auton A, et al. 2012. An integrated
of HNF1B variants with diverse cancers:  collaborative analysis of data map of genetic variation from 1,092 human genomes. Nature, 491(7422),
from 19 genome-​ wide association studies. PLoS One, 5(5), e10858. 56–​65. PMCID: PMC3498066.
PMCID: PMC2878330. Genomes Project Consortium, Auton A, Brooks LD, et  al. 2015. A global
Encode Project Consortium. 2012. An integrated encyclopedia of DNA elements reference for human genetic variation. Nature, 526(7571), 68–​ 74.
in the human genome. Nature, 489(7414), 57–​74. PMCID: PMC3439153. PMCID: PMC4750478.
Evangelou E, and Ioannidis JP. 2013. Meta-​analysis methods for genome-​ Gerds TA, Cai T, and Schumacher M. 2008. The performance of risk prediction
wide association studies and beyond. Nat Rev Genet, 14(6), 379–​389. models. Biom J, 50(4), 457–​479. PMID: 18663757.
PMID: 23657481. Germain M, Chasman DI, de Haan H, et  al. 2015. Meta-​analysis of 65,734
Evans DM, and Davey Smith G. 2015. Mendelian randomization: new applica- individuals identifies TSPAN15 and SLC44A2 as two susceptibility
tions in the coming age of hypothesis-​free causality. Annu Rev Genomics loci for venous thromboembolism. Am J Hum Genet, 96(4), 532–​542.
Hum Genet, 16, 327–​350. PMID: 25939054. PMCID: PMC4385184.
Falconer DS. 1965. The inheritance of liability to certain diseases, estimated Ghesquieres H, Slager SL, Jardin F, et  al. 2015. Genome-​ wide associa-
from the incidence among relatives. Ann Hum Genet, 29(51), 51–​76. tion study of event-​ free survival in diffuse large B-​ cell lymphoma
Falconer DS, and Mackay TFC. 1996. Introduction to Quantitative Genetics, treated with immunochemotherapy. J Clin Oncol, 33(33), 3930–​3937.
4th ed. Harlow: Pearson Prentice Hall. PMCID: PMC4879713.
Fehringer G, Kraft P, Pharoah PD, et  al. 2016. Cross-​cancer genome-​wide Ghoussaini M, Fletcher O, Michailidou K, et al. 2012. Genome-​wide associa-
analysis of lung, ovary, breast, prostate, and colorectal cancer reveals tion analysis identifies three new breast cancer susceptibility loci. Nat
novel pleiotropic associations. Cancer Res, 76(17), 5103–​ 5114. Genet, 44(3), 312–​318. PMCID: PMC3653403.
PMCID: PMC5010493. Ghoussaini M, French JD, Michailidou K, et al. 2016. Evidence that the 5p12
Fejerman L, Ahmadiyeh N, Hu D, et al. 2014. Genome-​wide association study variant rs10941679 confers susceptibility to estrogen-​receptor-​positive
of breast cancer in Latinas identifies novel protective variants on 6q25. breast cancer through FGF10 and MRPS30 regulation. Am J Hum Genet,
Nat Commun, 5, 5260. PMCID: PMC4204111. 99(4), 903–​911. PMCID: PMC5065698.
Fejerman L, Hu D, Huntsman S, et al. 2013. Genetic ancestry and risk of mor- Ghoussaini M, Song H, Koessler T, et al. 2008. Multiple loci with different can-
tality among U.S. Latinas with breast cancer. Cancer Res, 73(24), 7243–​ cer specificities within the 8q24 gene desert. J Natl Cancer Inst, 100(13),
7253. PMCID: PMC3881587. 962–​966. PMCID: PMC2902819.
Figueiredo JC, Hsu L, Hutter CM, et al. 2014. Genome-​wide diet-​gene interac- Glubb DM, Maranian MJ, Michailidou K, et  al. 2015. Fine-​ scale map-
tion analyses for risk of colorectal cancer. PLoS Genet, 10(4), e1004228. ping of the 5q11.2 breast cancer locus reveals at least three indepen-
PMCID: PMC3990510. dent risk variants regulating MAP3K1. Am J Hum Genet, 96(1), 5–​20.
Figueroa JD, Han SS, Garcia-​Closas M, et al. 2014. Genome-​wide interaction PMCID: PMC4289692.
study of smoking and bladder cancer risk. Carcinogenesis, 35(8), 1737–​ Glymour MM, Tchetgen Tchetgen EJ, and Robins JM. 2012. Credible
1744. PMCID: PMC4123644. Mendelian randomization studies:  approaches for evaluating the instru-
Finucane HK, Bulik-​Sullivan B, Gusev A, et al. 2015. Partitioning heritability mental variable assumptions. Am J Epidemiol, 175(4), 332–​ 339.
by functional annotation using genome-​wide association summary statis- PMCID: PMC3366596.
tics. Nat Genet, 47(11), 1228–​1235. PMCID: PMC4626285. Gong J, Hutter CM, Newcomb PA, et  al. 2016. Genome-​ wide interac-
Fitzgerald RC, Hardwick R, Huntsman D, et  al. 2010. Hereditary diffuse tion analyses between genetic variants and alcohol consumption and
gastric cancer:  updated consensus guidelines for clinical manage- smoking for risk of colorectal cancer. PLoS Genet, 12(10), e1006296.
ment and directions for future research. J Med Genet, 47(7), 436–​444. PMCID: PMC5065124.
PMCID: PMC2991043. Gowen LC, Johnson BL, Latour AM, Sulik KK, and Koller BH. 1996. BRCA1
Fitzgibbons RJ, Jr., Lynch HT, Stanislav GV, et  al. 1987. Recognition deficiency results in early embryonic lethality characterized by neuroepi-
and treatment of patients with hereditary nonpolyposis colon can- thelial abnormalities. Nat Genet, 12(2), 191–​194. PMID: 8563759.
cer (Lynch syndromes I  and II). Ann Surg, 206(3), 289–​ 295. Gudmundsson J, Sulem P, Manolescu A, et al. 2007a. Genome-​wide associa-
PMCID: PMC1493182. tion study identifies a second prostate cancer susceptibility variant at
Franceschini N, Carty CL, Lu Y, et al. 2016. Variant discovery and fine map- 8q24. Nat Genet, 39(5), 631–​637. PMID: 17401366.
ping of genetic loci associated with blood pressure traits in Hispanics and Gudmundsson J, Sulem P, Steinthorsdottir V, et  al. 2007b. Two variants on
African Americans. PLoS One, 11(10), e0164132. PMCID: PMC5063457. chromosome 17 confer prostate cancer risk, and the one in TCF2 protects
Freedman ML, Monteiro AN, Gayther SA, et al. 2011. Principles for the post-​ against type 2 diabetes. Nat Genet, 39(8), 977–​983. PMID: 17603485.
GWAS functional characterization of cancer risk loci. Nat Genet, 43(6), Guo Q, Schmidt MK, Kraft P, et  al. 2015. Identification of novel genetic
513–​518. PMCID: PMC3325768. markers of breast cancer survival. J Natl Cancer Inst, 107(5).
Fuchsberger C, Flannick J, Teslovich TM, et al. 2016. The genetic architecture PMCID: PMC4555642.
of type 2 diabetes. Nature, 536(7614), 41–​47. PMCID: PMC5034897. Guo Y, Warren Andersen S, Shu XO, et al. 2016. Genetically predicted body
Gail MH, and Pfeiffer RM. 2005. On criteria for evaluating models of absolute mass index and breast cancer risk:  Mendelian randomization analyses
risk. Biostatistics, 6(2), 227–​239. PMID: 15772102. of data from 145,000 women of European descent. PLoS Med, 13(8),
Gao C, Patel CJ, Michailidou K, et al. 2016. Mendelian randomization study e1002105. PMCID: PMC4995025.
of adiposity-​related traits and risk of breast, ovarian, prostate, lung and Gusev A, Lee SH, Trynka G, et al. 2014. Partitioning heritability of regulatory
colorectal cancer. Int J Epidemiol, 45(3), 896–​908. PMID: 27427428. and cell-​type-​specific variants across 11 common diseases. Am J Hum
Garcia-​Albeniz X, Rudolph A, Hutter C, et  al. 2016. CYP24A1 variant Genet, 95(5), 535–​552. PMCID: PMC4225595.
modifies the association between use of oestrogen plus progesto- Haiman CA, Chen GK, Vachon CM, et  al. 2011. A common variant at the
gen therapy and colorectal cancer risk. Br J Cancer, 114(2), 221–​229. TERT-​ CLPTM1L locus is associated with estrogen receptor-​ negative
PMCID: PMC4815813. breast cancer. Nat Genet, 43(12), 1210–​1214. PMCID: PMC3279120.
Garcia-​Closas M, Couch FJ, Lindstrom S, et al. 2013a. Genome-​wide associa- Haiman CA, Patterson N, Freedman ML, et al. 2007. Multiple regions within
tion studies identify four ER negative-​specific breast cancer risk loci. Nat 8q24 independently affect risk for prostate cancer. Nat Genet, 39 (5),
Genet, 45(4), 392–​398, 398e391–​392. PMCID: PMC3771695. 638–​644. PMCID: PMC2638766.
Garcia-​Closas M, Jacobs K, Kraft P, and Chatterjee N. 2011. Analysis of epi- Hall JM, Lee MK, Newman B, et  al. 1990. Linkage of early-​onset familial
demiologic studies of genetic effects and gene-​environment interactions. breast cancer to chromosome 17q21. Science, 250(4988), 1684–​1689.
IARC Sci Publ 163, 281–​301. PMID: 22997868. PMID: 2270482.
Garcia-​Closas M, Malats N, Silverman D, et  al. 2005. NAT2 slow acetyla- Han Y, Hazelett DJ, Wiklund F, et al. 2015a. Integration of multiethnic fine-​
tion, GSTM1 null genotype, and risk of bladder cancer: results from the mapping and genomic annotation to prioritize candidate functional SNPs
Spanish Bladder Cancer Study and meta-​analyses. Lancet, 366(9486), at prostate cancer susceptibility regions. Hum Mol Genet, 24(19), 5603–​
649–​659. PMCID: PMC1459966. 5618. PMCID: PMC4572069.
72

72 Part I:  Basic Concepts


Han Y, Signorello LB, Strom SS, et al. 2015b. Generalizability of established Prostate Cancer Cohort Consortium. Am J Epidemiol, 180(10), 1018–​
prostate cancer risk variants in men of African ancestry. Int J Cancer, 1027. PMCID: PMC4224360.
136(5), 1210–​1217. PMCID: PMC4268262. Kamihara J, Rana HQ, and Garber JE. 2014. Germline TP53 mutations and the
Hashibe M, McKay JD, Curado MP, et  al. 2008. Multiple ADH genes are changing landscape of Li-​Fraumeni syndrome. Hum Mutat, 35(6), 654–​
associated with upper aerodigestive cancers. Nat Genet, 40(6), 707–​709. 662. PMID: 24706533.
PMID: 18500343. Kang HM, Sul JH, Service SK, et  al. 2010. Variance component model to
Hazelett DJ, Rhie SK, Gaddis M, et al. 2014. Comprehensive functional anno- account for sample structure in genome-​wide association studies. Nat
tation of 77 prostate cancer risk loci. PLoS Genet, 10(1), e1004102. Genet, 42(4), 348–​354. PMCID: PMC3092069.
PMCID: PMC3907334. Kantor ED, Hutter CM, Minnier J, et  al. 2014. Gene-​environment inter-
Hirschhorn JN, and Altshuler D. 2002. Once and again-​issues surrounding rep- action involving recently identified colorectal cancer susceptibil-
lication in genetic association studies. J Clin Endocrinol Metab, 87(10), ity Loci. Cancer Epidemiol Biomarkers Prev, 23(9), 1824–​ 1833.
4438–​4441. PMID: 12364414. PMCID: PMC4209726.
Hirschhorn JN, and Daly MJ. 2005. Genome-​wide association studies for Kapoor NS, Curcio LD, Blakemore CA, et al. 2015. Multigene panel testing
common diseases and complex traits. Nat Rev Genet, 6(2), 95–​ 108. detects equal rates of pathogenic BRCA1/​2 Mutations and has a higher
PMID: 15716906. diagnostic yield compared to limited BRCA1/​2 analysis alone in patients
Hirschhorn JN, Lohmueller K, Byrne E, and Hirschhorn K. 2002. A compre- at risk for hereditary breast cancer. Ann Surg Oncol, 22(10), 3282–​3288.
hensive review of genetic association studies. Genet Med, 4(2), 45–​61. PMID: 26219241.
PMID: 11882781. Kar SP, Beesley J, Amin Al Olama A, et al. 2016. Genome-​wide meta-​analyses
Hoffmann TJ, Van Den Eeden SK, Sakoda LC, et al. 2015. A large multiethnic of breast, ovarian, and prostate cancer association studies identify mul-
genome-​wide association study of prostate cancer identifies novel risk tiple new susceptibility loci shared by at least two cancer types. Cancer
variants and substantial ethnic differences. Cancer Discov, 5(8), 878–​ Discov, 6(9), 1052–​1067. PMCID: PMC5010513.
891. PMCID: PMC4527942. Kerns SL, Kundu S, Oh JH, et  al. 2015. The prediction of radiotherapy
Hoggart CJ, Clark TG, De Iorio M, Whittaker JC, and Balding DJ. 2008. toxicity using single nucleotide polymorphism-​based models:  a step
Genome-​wide significance for dense SNP and resequencing data. Genet toward prevention. Semin Radiat Oncol, 25(4), 281–​ 291. PMCID:
Epidemiol, 32(2), 179–​185. PMID: 18200594. PMC4576690.
Hoover RN. 2000. Cancer:  nature, nurture, or both. N Engl J Med, 343(2), Kerns SL, West CM, Andreassen CN, et al. 2014. Radiogenomics: the search
135–​136. PMID: 10891523. for genetic predictors of radiotherapy response. Future Oncol, 10(15),
Howie B, Fuchsberger C, Stephens M, Marchini J, and Abecasis GR. 2012. Fast 2391–​2406. PMID: 25525847.
and accurate genotype imputation in genome-​wide association studies Kerr KF, Wang Z, Janes H, et al. 2014. Net reclassification indices for evaluat-
through pre-​phasing. Nat Genet, 44(8), 955–​959. PMCID: PMC3696580. ing risk prediction instruments:  a critical review. Epidemiology, 25(1),
Howlett NG, Taniguchi T, Olson S, et al. 2002. Biallelic inactivation of BRCA2 114–​121. PMCID: PMC3918180.
in Fanconi anemia. Science, 297(5581), 606–​609. PMID: 12065746. Khan PM, Tops CMJ, Breukel C, et al. 1988. Close linkage of a highly poly-
Hsu L, Jeon J, Brenner H, et al. 2015. A model to determine colorectal cancer morphic marker (D5s37) to familial adenomatous polyposis (FAP) and
risk using common genetic susceptibility loci. Gastroenterology, 148(7), confirmation of FAP localization on chromosome 5q21–​Q22. Human
1330–​1339 e1314. PMCID: PMC4446193. Genetics, 79(2), 183–​185. PMID: WOS:A1988P119100019.
Hsu L, Jiao S, Dai JY, et  al. 2012. Powerful cocktail methods for detecting Khankari NK, Murff HJ, Zeng C, et  al. 2016a. Polyunsaturated fatty
genome-​wide gene-​environment interaction. Genet Epidemiol, 36(3), acids and prostate cancer risk:  a Mendelian randomisation analysis
183–​194. PMCID: PMC3654520. from the PRACTICAL consortium. Br J Cancer, 115(5), 624–​ 631.
Hunter DJ, and Kraft P. 2007. Drinking from the fire hose:  statistical issues PMCID: PMC4997551.
in genomewide association studies. N Engl J Med, 357(5), 436–​439. Khankari NK, Shu XO, Wen W, et al. 2016b. Association between adult height
PMID: 17634446. and risk of colorectal, lung, and prostate cancer:  results from meta-​
Hunter DJ, Kraft P, Jacobs KB, et al. 2007. A genome-​wide association study analyses of prospective studies and mendelian randomization analyses.
identifies alleles in FGFR2 associated with risk of sporadic postmeno- PLoS Med, 13(9), e1002118. PMCID: PMC5012582.
pausal breast cancer. Nat Genet, 39(7), 870–​874. PMCID: PMC3493132. Kharazmi E, Chen T, Narod S, Sundquist K, and Hemminki K. 2014. Effect of
Hunter DJ, Riboli E, Haiman CA, et al. 2005. A candidate gene approach to multiplicity, laterality, and age at onset of breast cancer on familial risk of
searching for low-​penetrance breast and prostate cancer genes. Nat Rev breast cancer: a nationwide prospective cohort study. Breast Cancer Res
Cancer, 5(12), 977–​985. PMID: 16341085. Treat, 144(1), 185–​192. PMID: 24487690.
Hutter CM, Mechanic LE, Chatterjee N, et al. 2013. Gene-​environment inter- Khoury MJ, Beaty TH, and Liang KY. 1988. Can familial aggregation of dis-
actions in cancer epidemiology: a National Cancer Institute Think Tank ease be explained by familial aggregation of environmental risk factors?
report. Genet Epidemiol, 37(7), 643–​657. PMCID: PMC4143122. Am J Epidemiol, 127(3), 674–​683. PMID: 3341366.
Iles MM, Bishop DT, Taylor JC, et al. 2014. The effect on melanoma risk of Kichaev G, Roytman M, Johnson R, et al. 2016. Improved methods for multi-​
genes previously associated with telomere length. J Natl Cancer Inst, trait fine mapping of pleiotropic risk loci. Bioinformatics, 33(2), 248–​
106(10). PMCID: PMC4196080. 255. PMID: 27663501.
International HapMap C. 2005. A haplotype map of the human genome. Nature, Kichaev G, Yang WY, Lindstrom S, et al. 2014. Integrating functional data to
437(7063), 1299–​1320. PMCID: PMC1880871. prioritize causal variants in statistical fine-​mapping studies. PLoS Genet,
Ioannidis JP, Ntzani EE, Trikalinos TA, and Contopoulos-​Ioannidis DG. 2001. 10(10), e1004722. PMCID: PMC4214605.
Replication validity of genetic association studies. Nat Genet, 29(3), 306–​ Kicinski M, Vangronsveld J, and Nawrot TS. 2011. An epidemiological reap-
309. PMID: 11600885. praisal of the familial aggregation of prostate cancer:  a meta-​analysis.
Ioannidis JP. 2005. Why most published research findings are false. PLoS Med, PLoS One, 6(10), e27130. PMCID: PMC3205054.
2(8), e124. PMCID: PMC1182327. King MC, Levy-​Lahad E, and Lahad A. 2014. Population-​based screening for
Ioannidis JP, Thomas G, and Daly MJ. 2009. Validating, augmenting and refin- BRCA1 and BRCA2: 2014 Lasker Award. JAMA, 312 (11), 1091–​1092.
ing genome-​wide association signals. Nat Rev Genet, 10(5), 318–​329. PMID: 25198398.
PMID: 19373277. Klein AP. 2005. Overview of Linkage Analysis Pancreatic Cancer (pp. 329–​
Ioannidis JP, Trikalinos TA, and Khoury MJ. 2006. Implications of small effect 341). New York: Springer.
sizes of individual genetic variants on the design and interpretation of Knipe DW, Evans DM, Kemp JP, et  al. 2014. Genetic variation in prostate-​
genetic association studies of complex diseases. Am J Epidemiol, 164(7), specific antigen-​detected prostate cancer and the effect of control selec-
609–​614. PMID: 16893921. tion on genetic association studies. Cancer Epidemiol Biomarkers Prev,
Jiao S, Hsu L, Berndt S, et  al. 2012. Genome-​ wide search for gene-​ 23(7), 1356–​1365. PMCID: PMC4082405.
gene interactions in colorectal cancer. PLoS One, 7(12), e52535. Knudson AG, Jr. 1971. Mutation and cancer: statistical study of retinoblastoma.
PMCID: PMC3530500. Proc Natl Acad Sci U S A, 68 (4), 820–​823. PMCID: PMC389051.
Johnatty SE, Tyrer JP, Kar S, et al. 2015. Genome-​wide analysis identifies novel Kouvaraki MA, Shapiro SE, Perrier ND, et al. 2005. RET proto-​oncogene: a
loci associated with ovarian cancer outcomes: findings from the Ovarian review and update of genotype-​phenotype correlations in hereditary med-
Cancer Association Consortium. Clin Cancer Res, 21(23), 5264–​5276. ullary thyroid cancer and associated endocrine tumors. Thyroid, 15(6),
PMCID: PMC4624261. 531–​544.
Joshi AD, Lindstrom S, Husing A, et al. 2014. Additive interactions between Kraft P, and Hunter DJ. 2009. The challenge of assessing complex gene-​gene
susceptibility single-​nucleotide polymorphisms identified in genome-​ and gene-​environment interactions. In: Khoury M, Bedrosian S, Gwinn M,
wide association studies and breast cancer risk factors in the Breast and Higgins J, Ioannidis J, and Little J (Eds.), Human Genome Epidemiology,
 73

Genetic Epidemiology of Cancer 73


2nd ed.: Building the Evidence for Using Genetic Information to Improve and Prostate Cancer Cohort Consortium. Am J Epidemiol, 176(12), 1121–​
Health and Prevent Disease. New York: Oxford University Press. 1129. PMCID: PMC3571230.
Kraft P, Wacholder S, Cornelis MC, et  al. 2009a. Beyond odds ratios-​ -​ Maher B. 2008. Personal genomes: the case of the missing heritability. Nature,
communicating disease risk based on genetic profiles. Nat Rev Genet, 456(7218), 18–​21. PMID: 18987709.
10(4), 264–​269. PMID: 19238176. Manchanda R, Legood R, Burnell M, et al. 2015. Cost-​effectiveness of popula-
Kraft P, Yen YC, Stram DO, Morrison J, and Gauderman WJ. 2007. Exploiting tion screening for BRCA mutations in Ashkenazi Jewish women com-
gene-​environment interaction to detect genetic associations. Hum Hered, pared with family history-​based testing. J Natl Cancer Inst, 107(1), 380.
63(2), 111–​119. PMID: 17283440. PMCID: PMC4301704.
Kraft P, Zeggini E, and Ioannidis JP. 2009b. Replication in genome-​wide asso- Mancuso N, Rohland N, Rand KA, et al. 2016. The contribution of rare variation
ciation studies. Stat Sci, 24(4), 561–​573. PMCID: PMC2865141. to prostate cancer heritability. Nat Genet, 48(1), 30–​35. PMID: 26569126.
Kraft P, and Aschard H. 2015. Finding the missing gene-​environment interac- Manolio TA, Bailey-​Wilson JE, and Collins FS. 2006. Genes, environment and
tions. Eur J Epidemiol, 30(5), 353–​355. PMCID: PMC4905576. the value of prospective cohort studies. Nat Rev Genet, 7(10), 812–​820.
Kraft P, and Haiman CA. 2010. GWAS identifies a common breast can- PMID: 16983377.
cer risk allele among BRCA1 carriers. Nat Genet, 42(10), 819–​820. Manolio TA, Collins FS, Cox NJ, et al. 2009. Finding the missing heritability of
PMID: 20877320. complex diseases. Nature, 461(7265), 747–​753. PMCID: PMC2831613.
Krier J, Barfield R, Green RC, and Kraft P. 2016. Reclassification of genetic-​ Marchini J, and Howie B. 2010. Genotype imputation for genome-​wide asso-
based risk predictions as GWAS data accumulate. Genome Med, 8(1), 20. ciation studies. Nat Rev Genet, 11(7), 499–​511. PMID: 20517342.
PMCID: PMC4756503. Marcus PM, Pashayan N, Church TR, et  al. 2016. Population-​based preci-
Kuchenbaecker KB, Ramus SJ, Tyrer J, et al. 2015. Identification of six new sion cancer screening:  a symposium on evidence, epidemiology, and
susceptibility loci for invasive epithelial ovarian cancer. Nat Genet, 47(2), next steps. Cancer Epidemiol Biomarkers Prev, 25(11), 1449–​ 1455.
164–​171. PMCID: PMC4445140. PMCID: PMC5165650.
Kuo CL, and Feingold E. 2010. What’s the best statistic for a simple test of Mateo J, Carreira S, Sandhu S, et  al. 2015. DNA-​repair defects and olapa-
genetic association in a case-​control study? Genet Epidemiol, 34(3), 246–​ rib in metastatic prostate cancer. N Engl J Med, 373(18), 1697–​1708.
253. PMID: 20025064. PMID: 26510020.
Laird NM, and Lange C. 2006. Family-​based designs in the age of large-​ Maurano MT, Humbert R, Rynes E, et al. 2012. Systematic localization of com-
scale gene-​ association studies. Nat Rev Genet, 7(5), 385–​ 394. mon disease-​associated variation in regulatory DNA. Science, 337(6099),
PMID: 16619052. 1190–​1195. PMCID: PMC3771521.
Lawlor DA, Harbord RM, Sterne JAC, Timpson N, and Davey Smith G. 2008. Mavaddat N, Pharoah PD, Michailidou K, et al. 2015. Prediction of breast can-
Mendelian randomization: using genes as instruments for making causal cer risk based on profiling with common genetic variants. J Natl Cancer
inferences in epidemiology. Stat Med, 27, 1133–​1163. Inst, 107(5). PMCID: PMC4754625.
Lewis SJ, and Smith GD. 2005. Alcohol, ALDH2, and esophageal cancer:  a McCarthy S, Das S, Kretzschmar W, et al. 2016. A reference panel of 64,976
meta-​analysis which illustrates the potentials and limitations of a haplotypes for genotype imputation. Nat Genet, 48(10), 1279–​ 1283.
Mendelian randomization approach. Cancer Epidemiol Biomarkers Prev, PMID: 27548312.
14(8), 1967–​1971. PMID: 16103445. Mechanic LE, Chen HS, Amos CI, et al. 2012. Next generation analytic tools
Lewontin R. 1995. Human Diversity. New York: Scientific American Library. for large scale genetic epidemiology studies of complex diseases. Genet
Li Y, Willer CJ, Ding J, Scheet P, and Abecasis GR. 2010. MaCH:  using Epidemiol, 36(1), 22–​35. PMCID: PMC3368075.
sequence and genotype data to estimate haplotypes and unobserved geno- Metcalfe KA, Eisen A, Lerner-​Ellis J, and Narod SA. 2015. Is it time to offer
types. Genet Epidemiol, 34(8), 816–​834. PMCID: PMC3175618. BRCA1 and BRCA2 testing to all Jewish women? Curr Oncol, 22(4),
Li Y, Willer C, Sanna S, and Abecasis G. 2009. Genotype imputation. Annu Rev e233–​e236. PMCID: PMC4530819.
Genomics Hum Genet, 10, 387–​406. PMCID: PMC2925172. Meyer KB, O’Reilly M, Michailidou K, et  al. 2013. Fine-​scale mapping of
Lin WY, Camp NJ, Ghoussaini M, et  al. 2015. Identification and character- the FGFR2 breast cancer risk locus:  putative functional variants differ-
ization of novel associations in the CASP8/​ALS2CR12 region on chro- entially bind FOXA1 and E2F1. Am J Hum Genet, 93(6), 1046–​1060.
mosome 2 with breast cancer risk. Hum Mol Genet, 24(1), 285–​298. PMCID: PMC3852923.
PMCID: PMC4334820. Michailidou K, Beesley J, Lindstrom S, et  al. 2015a. Genome-​wide associ-
Lindstrom LS, Hall P, Hartman M, et al. 2007. Familial concordance in cancer ation analysis of more than 120,000 individuals identifies 15 new sus-
survival: a Swedish population-​based study. Lancet Oncol, 8(11), 1001–​ ceptibility loci for breast cancer. Nat Genet, 47(4), 373–​380. PMCID:
1006. PMID: 17921068. PMC4549775.
Lindstrom S, Ablorh A, Chapman B, et al. 2016. Deep targeted sequencing of Michailidou K, Hall P, Gonzalez-​Neira A, et al. 2013. Large-​scale genotyping
12 breast cancer susceptibility regions in 4611 women across four differ- identifies 41 new loci associated with breast cancer risk. Nat Genet, 45(4),
ent ethnicities. Breast Cancer Res, 18(1), 109. PMCID: PMC5097387. 353–​361. PMCID: PMC3771688.
Lindstrom S, Schumacher FR, Cox D, et al. 2012. Common genetic variants in Michailidou K, Lindstrom S, Dennis J, et  al. 2015b. Meta-​ analysis of
prostate cancer risk prediction: results from the NCI Breast and Prostate OncoArray, iCOGS, and GWAS data for more than 220,000 women iden-
Cancer Cohort Consortium (BPC3). Cancer Epidemiol Biomarkers Prev, tifies more than 50 novel breast cancer susceptibility loci. Paper presented
21(3), 437–​444. PMCID: PMC3318963. at the 65th Annual Meeting of the American Society of Human Genetics,
Lindstrom S, Schumacher F, Siddiq A, et  al. 2011. Characterizing associa- Baltimore, MD.
tions and SNP-​environment interactions for GWAS-​identified prostate Mucci LA, Hjelmborg JB, Harris JR, et al. 2016. Familial risk and heritabil-
cancer risk markers:  results from BPC3. PLoS One, 6(2), e17142. ity of cancer among twins in Nordic countries. JAMA, 315(1), 68–​76.
PMCID: PMC3044744. PMID: 26746459.
Lindstrom S, Yen YC, Spiegelman D, and Kraft P. 2009. The impact of gene-​ Mukherjee B, Ahn J, Gruber SB, and Chatterjee N. 2012. Testing gene-​
environment dependence and misclassification in genetic association environment interaction in large-​ scale case-​control association stud-
studies incorporating gene-​environment interactions. Hum Hered, 68(3), ies: possible choices and comparisons. Am J Epidemiol, 175(3), 177–​190.
171–​181. PMCID: PMC2869071. PMCID: PMC3286201.
Liu CT, Buchkovich ML, Winkler TW, et al. 2014. Multi-​ethnic fine-​mapping Murillo-​Zamora E, Moreno-​ Macias H, Ziv E, et  al. 2013. Association
of 14 central adiposity loci. Hum Mol Genet, 23(17), 4738–​ 4744. between rs2981582 polymorphism in the FGFR2 gene and the risk
PMCID: PMC4119415. of breast cancer in Mexican women. Arch Med Res, 44(6), 459–​466.
Lu Y, Ek WE, Whiteman D, et al. 2014. Most common “sporadic” cancers have PMCID: PMC4049087.
a significant germline genetic component. Hum Mol Genet, 23(22), 6112–​ Nagy R, Sweet K, and Eng C. 2004. Highly penetrant hereditary cancer syn-
6118. PMCID: PMC4271103. dromes. Oncogene, 23(38), 6445–​6470. PMID: 15322516.
Lucas AO. 1976. Surveillance of communicable diseases in tropical Africa. Int Nan H, Hutter CM, Lin Y, et  al. 2015. Association of aspirin and NSAID
J Epidemiol, 5 (1), 39–​43. PMID: 1262110. use with risk of colorectal cancer according to genetic variants. JAMA,
Lynch HT AD, Krush AJ, Mukerjee D. 1967. Cancer, heredity and genetic 313(11), 1133–​1142. PMCID: PMC4382867.
counseling. Xeroderma pigmentosum. Cancer, 20(10), 1796–​1801. NCI-​NHGRI Working Group on Replication in Association Studies, Chanock
Maas P, Barrdahl M, Joshi AD, et al. 2016. Breast cancer risk from modifiable SJ, Manolio T, et al. 2007. Replicating genotype-​phenotype associations.
and nonmodifiable risk factors among white women in the United States. Nature, 447(7145), 655–​660. PMID: 17554299.
JAMA Oncol, 2 (10), 1295–​1302. PMID: 27228256. Nead KT, Sharp SJ, Thompson DJ, et al. 2015. Evidence of a causal association
Machiela MJ, Lindstrom S, Allen NE, et al. 2012. Association of type 2 diabe- between insulinemia and endometrial cancer: a Mendelian randomization
tes susceptibility variants with advanced prostate cancer risk in the Breast analysis. J Natl Cancer Inst, 107(9). PMCID: PMC4572886.
74

74 Part I:  Basic Concepts


Nelson SC, Doheny KF, Pugh EW, et al. 2013. Imputation-​based genomic cov- Pierce BL, Ahsan H, and VanderWeele TJ. 2011. Power and instru-
erage assessments of current human genotyping arrays. G3 (Bethesda), ment strength requirements for Mendelian randomization stud-
3(10), 1795–​1807. PMCID: PMC3789804. ies using multiple genetic variants. Int J Epidemiol, 40(3), 740–​752.
Newton-​Cheh C, and Hirschhorn JN. 2005. Genetic association studies of PMID: WOS:000293618300025.
complex traits: design and analysis issues. Mutat Res, 573(1–​2), 54–​69. Pierce BL, and Burgess S. 2013. Efficient design for Mendelian randomization
PMID: 15829237. studies: subsample and 2-​sample instrumental variable estimators. Am J
Nickels S, Truong T, Hein R, et  al. 2013. Evidence of gene-​environment Epidemiol, 178(7), 1177–​1184. PMCID: PMC3783091.
interactions between common breast cancer susceptibility loci and Pirinen M, Donnelly P, and Spencer CC. 2012. Including known covariates can
established environmental risk factors. PLoS Genet, 9(3), e1003284. reduce power to detect genetic effects in case-​control studies. Nat Genet,
PMCID: PMC3609648. 44(8), 848–​851. PMID: 22820511.
Nimptsch K, Aleksandrova K, Boeing H, et al. 2015. Association of CRP genetic Plon SE. 2015. BRCA1/​2 population screening: embracing the benefits. Curr
variants with blood concentrations of C-​reactive protein and colorectal Oncol, 22(4), e230–​e231. PMCID: PMC4530817.
cancer risk. Int J Cancer, 136(5), 1181–​1192. PMID: 25043606. Price AL, Patterson NJ, Plenge RM, et al. 2006. Principal components analysis
Ojha J, Codd V, Nelson CP, et al. 2016. Genetic variation associated with longer corrects for stratification in genome-​wide association studies. Nat Genet,
telomere length increases risk of chronic lymphocytic leukemia. Cancer 38(8), 904–​909. PMID: 16862161.
Epidemiol Biomarkers Prev, 25(7), 1043–​1049. PMCID: PMC5008454. Price AL, Zaitlen NA, Reich D, and Patterson N. 2010. New approaches to
Oliveira AM, Ross JS, and Fletcher JA. 2005. Tumor suppressor genes in breast population stratification in genome-​wide association studies. Nat Rev
cancer: the gatekeepers and the caretakers. Am J Clin Pathol, 124 Suppl, Genet, 11(7), 459–​463. PMCID: PMC2975875.
S16–​S28. PMID: 16468415. Qi L, Cornelis MC, Kraft P, et al. 2010. Genetic variants in ABO blood group
Ong JS, Cuellar-​Partida G, Lu Y, et  al. 2016. Association of vitamin D lev- region, plasma soluble E-​selectin levels and risk of type 2 diabetes. Hum
els and risk of ovarian cancer:  a Mendelian randomization study. Int J Mol Genet, 19(9), 1856–​1862. PMCID: PMC2850622.
Epidemiol, 45(5), 1619–​1630. PMCID: PMC5100621. Rahman N. 2014. Realizing the promise of cancer predisposition genes. Nature,
Painter JN, Kaufmann S, O’Mara TA, et al. 2016a. A common variant at the 505(7483), 302–​308. PMCID: PMC4975511.
14q32 endometrial cancer risk locus activates AKT1 through YY1 bind- Rebbeck TR, Spitz M, and Wu X. 2004. Assessing the function of genetic vari-
ing. Am J Hum Genet, 98(6), 1159–​1169. PMCID: PMC4908177. ants in candidate gene association studies. Nat Rev Genet, 5(8), 589–​597.
Painter JN, O’Mara TA, Marquart L, et al. 2016b. Genetic risk score Mendelian PMID: 15266341.
randomization shows that obesity measured as body mass index, but Rhie SK, Coetzee SG, Noushmehr H, et al. 2013. Comprehensive functional
not waist:hip ratio, is causal for endometrial cancer. Cancer Epidemiol annotation of seventy-​ one breast cancer risk Loci. PLoS One, 8(5),
Biomarkers Prev, 25(11), 1503–​1510. PMCID: PMC5093082. e63925. PMCID: PMC3661550.
Palmer TM, Lawlor DA, Harbord RM, et al. 2012. Using multiple genetic vari- Risch N. 1990. Linkage strategies for genetically complex traits. I. Multilocus
ants as instrumental variables for modifiable risk factors. Stat Methods models. Am J Hum Genet, 46(2), 222–​228. PMCID: PMC1684987.
Med Res, 21(3), 223–​242. PMID: WOS:000304231900002. Risch N. 2001. The genetic epidemiology of cancer:  interpreting fam-
Pare G, Cook NR, Ridker PM, and Chasman DI. 2010. On the use of variance ily and twin studies and their implications for molecular genetic
per genotype as a tool to identify quantitative trait interaction effects: a approaches. Cancer Epidemiol Biomarkers Prev, 10(7), 733–​ 741.
report from the Women’s Genome Health Study. PLoS Genet, 6(6), PMID: 11440958.
e1000981. PMCID: PMC2887471. Rothman N, Garcia-​Closas M, and Hein DW. 2007. Commentary: Reflections
Park JH, Gail MH, Greene MH, and Chatterjee N. 2012. Potential usefulness on G. M. Lower and colleagues’ 1979 study associating slow acetylator
of single nucleotide polymorphisms to identify persons at high cancer phenotype with urinary bladder cancer: meta-​analysis, historical refine-
risk: an evaluation of seven common cancers. J Clin Oncol, 30(17), 2157–​ ments of the hypothesis, and lessons learned. Int J Epidemiol, 36(1), 23–​
2162. PMCID: PMC3397697. 28. PMID: 17510073.
Park JH, Gail MH, Weinberg CR, et al. 2011. Distribution of allele frequen- Rudolph A, Hein R, Lindstrom S, et  al. 2013. Genetic modifiers of meno-
cies and effect sizes and their interrelationships for common genetic sus- pausal hormone replacement therapy and breast cancer risk:  a
ceptibility variants. Proc Natl Acad Sci U S A, 108(44), 18026–​18031. genome-​wide interaction study. Endocr Relat Cancer, 20(6), 875–​887.
PMCID: PMC3207674. PMCID: PMC3863710.
Park JH, Wacholder S, Gail MH, et al. 2010. Estimation of effect size distribu- Rudolph A, Chang-​ Claude J, and Schmidt MK. 2016. Gene-​ environment
tion from genome-​wide association studies and implications for future interaction and risk of breast cancer. Br J Cancer, 114(2), 125–​133.
discoveries. Nat Genet, 42(7), 570–​575. PMCID: PMC4615599. PMCID: PMC4815812.
Patel JN, McLeod HL, and Innocenti F. 2013. Implications of genome-​wide Sampson JN, Wheeler WA, Yeager M, et al. 2015. Analysis of heritability and
association studies in cancer therapeutics. Br J Clin Pharmacol, 76(3), shared heritability based on genome-​wide association studies for thirteen
370–​380. PMCID: PMC3769665. cancer types. J Natl Cancer Inst, 107(12), djv279. PMCID: PMC4806328.
Pe’er I, Yelensky R, Altshuler D, and Daly MJ. 2008. Estimation of the multiple Schaid DJ, Buetow K, Weeks DE, et al. 1999. Discovery of cancer susceptibil-
testing burden for genomewide association studies of nearly all common ity genes: study designs, analytic approaches, and trends in technology. J
variants. Genet Epidemiol, 32(4), 381–​385. PMID: 18348202. Natl Cancer Inst Monogr, 26, 1–​16. PMID: 10854480.
Pearce CL, Rossing MA, Lee AW, et  al. 2013. Combined and interactive Schaub MA, Boyle AP, Kundaje A, Batzoglou S, and Snyder M. 2012. Linking
effects of environmental and GWAS-​ identified risk factors in ovar- disease associations with regulatory information in the human genome.
ian cancer. Cancer Epidemiol Biomarkers Prev, 22(5), 880–​ 890. Genome Res, 22(9), 1748–​1759. PMCID: PMC3431491.
PMCID: PMC3963289. Scott LJ, Mohlke KL, Bonnycastle LL, et al. 2007. A genome-​wide association
Pearson TA, and Manolio TA. 2008. How to interpret a genome-​wide associa- study of type 2 diabetes in Finns detects multiple susceptibility variants.
tion study. JAMA, 299(11), 1335–​1344. PMID: 18349094. Science, 316(5829), 1341–​1345. PMCID: PMC3214617.
Pesenti C, Gusella M, Sirchia SM, and Miozzo M. 2015. Germline oncophar- Sham P. 1998. Statistics in Human Genetics. London: Arnold.
macogenetics, a promising field in cancer therapy. Cell Oncol (Dordr), Siddiq A, Couch FJ, Chen GK, et  al. 2012. A meta-​analysis of genome-​
38(1), 65–​89. PMID: 25573079. wide association studies of breast cancer identifies two novel suscep-
Pfeiffer RM, and Gail MH. 2011. Two criteria for evaluating risk prediction tibility loci at 6q14 and 20q11. Hum Mol Genet, 21(24), 5373–​5384.
models. Biometrics, 67(3), 1057–​1065. PMCID: PMC3135785. PMCID: PMC3510753.
Pharoah PD, Antoniou A, Bobrow M, et al. 2002. Polygenic susceptibility to Siemiatycki J, and Thomas DC. 1981. Biological models and statistical interac-
breast cancer and implications for prevention. Nat Genet, 31(1), 33–​36. tions: an example from multistage carcinogenesis. Int J Epidemiol, 10(4),
PMID: 11984562. 383–​387. PMID: 7327838.
Pharoah PD, Day NE, Duffy S, Easton DF, and Ponder BA. 1997. Family his- Simonds NI, Ghazarian AA, Pimentel CB, et  al. 2016. Review of the gene-​
tory and the risk of breast cancer: a systematic review and meta-​analysis. environment interaction literature in cancer:  what do we know? Genet
Int J Cancer, 71(5), 800–​809. PMID: 9180149. Epidemiol, 40(5), 356–​365. PMCID: PMC4911236.
Phipps AI, Passarelli MN, Chan AT, et  al. 2016. Common genetic variation Slattery ML, Levin TR, Ma K, et  al. 2003. Family history and colorectal
and survival after colorectal cancer diagnosis: a genome-​wide analysis. cancer:  predictors of risk. Cancer Causes Control, 14(9), 879–​ 887.
Carcinogenesis, 37(1), 87–​95. PMCID: PMC4715234. PMID: 14682445.
Pierce BL, and Ahsan H. 2010. Genetic susceptibility to type 2 diabetes is asso- Soave D, Corvol H, Panjwani N, et al. 2015. A joint location-​scale test improves
ciated with reduced prostate cancer risk. Hum Hered, 69(3), 193–​201. power to detect associated SNPs, gene sets, and pathways. Am J Hum
PMCID: PMC2866577. Genet, 97(1), 125–​138. PMCID: PMC4572492.
 75

Genetic Epidemiology of Cancer 75


Southey MC, Goldgar DE, Winqvist R, et al. 2016. PALB2, CHEK2 and ATM Wakefield J. 2007. A Bayesian measure of the probability of false discovery
rare variants and cancer risk:  data from COGS. J Med Genet, 53(12), in genetic epidemiology studies. Am J Hum Genet, 81(2), 208–​227.
800–​811. PMCID: PMC5200636. PMCID: PMC1950810.
Spielman RS, McGinnis RE, and Ewens WJ. 1993. Transmission test for Wakefield J. 2012. Commentary:  Genome-​ wide significance thresh-
linkage disequilibrium:  the insulin gene region and insulin-​dependent olds via Bayes factors. Int J Epidemiol, 41(1), 286–​291. PMCID:
diabetes mellitus (IDDM). Am J Hum Genet, 52(3), 506–​ 516. PMC3304534.
PMCID: PMC1682161. Wakefield J, Skrivankova V, Hsu FC, Sale M, and Heagerty P. 2014. Detecting
Spurdle AB, Thompson DJ, Ahmed S, et al. 2011. Genome-​wide association signals in pharmacogenomic genome-​ wide association studies.
study identifies a common variant associated with risk of endometrial Pharmacogenomics J, 14(4), 309–​315. PMCID: PMC4085158.
cancer. Nat Genet, 43(5), 451–​454. PMCID: PMC3770523. Walko CM, and McLeod HL. 2014. Personalizing medicine in geriatric oncol-
Stacey SN, Manolescu A, Sulem P, et al. 2007. Common variants on chromo- ogy. J Clin Oncol, 32(24), 2581–​2586. PMID: 25071134.
somes 2q35 and 16q12 confer susceptibility to estrogen receptor-​positive Walsh KM, Codd V, Rice T, et al. 2015. Longer genotypically-​estimated leu-
breast cancer. Nat Genet, 39(7), 865–​869. PMID: 17529974. kocyte telomere length is associated with increased adult glioma risk.
Stacey SN, Manolescu A, Sulem P, et al. 2008. Common variants on chromo- Oncotarget, 6(40), 42468–​42477. PMCID: PMC4767445.
some 5p12 confer susceptibility to estrogen receptor-​positive breast can- Walsh KM, Whitehead TP, de Smith AJ, et  al. 2016. Common genetic
cer. Nat Genet, 40(6), 703–​706. PMID: 18438407. variants associated with telomere length confer risk for neuroblas-
Szulkin R, Karlsson R, Whitington T, et al. 2015a. Genome-​wide association toma and other childhood cancers. Carcinogenesis, 37(6), 576–​ 582.
study of prostate cancer-​specific survival. Cancer Epidemiol Biomarkers PMCID: PMC4876988.
Prev, 24(11), 1796–​1800. PMID: 26307654. Wang J, Wang H, Chen Y, Hao P, and Zhang Y. 2011. Alcohol ingestion and
Szulkin R, Whitington T, Eklund M, et  al. 2015b. Prediction of individual colorectal neoplasia: a meta-​analysis based on a Mendelian randomiza-
genetic risk to prostate cancer using a polygenic score. Prostate, 75(13), tion approach. Colorectal Dis, 13(5), e71–​e78. PMID: 21114754.
1467–​1474. PMID: 26177737. Wang Z, Zhu B, Zhang M, et al. 2014. Imputation and subset-​based association
Tabor HK, Risch NJ, and Myers RM. 2002. Candidate-​gene approaches for analysis across different cancer types identifies multiple independent risk
studying complex genetic traits: practical considerations. Nat Rev Genet, loci in the TERT-​CLPTM1L region on chromosome 5p15.33. Hum Mol
3(5), 391–​397. PMID: 11988764. Genet, 23(24), 6616–​6633. PMCID: PMC4240198.
Tanikawa C, Urabe Y, Matsuo K, et al. 2012. A genome-​wide association study Ward LD, and Kellis M. 2016. HaploReg v4:  systematic mining of puta-
identifies two susceptibility loci for duodenal ulcer in the Japanese popu- tive causal variants, cell types, regulators and target genes for human
lation. Nat Genet, 44 (4), 430–​434, S431–​S432. PMID: 22387998. complex traits and disease. Nucleic Acids Res, 44(D1), D877–​D881.
Taylor AE, Martin RM, Geybels MS, et  al. 2017. Investigating the possible PMCID: PMC4702929.
causal role of coffee consumption with prostate cancer risk and progres- Waters KM, Le Marchand L, Kolonel LN, et al. 2009. Generalizability of asso-
sion using Mendelian randomization analysis. Int J Cancer, 140(2), 322–​ ciations from prostate cancer genome-​wide association studies in mul-
328. PMCID: PMC5132137. tiple populations. Cancer Epidemiol Biomarkers Prev, 18(4), 1285–​1289.
Thomas DC, Lewinger JP, Murcray CE, and Gauderman WJ. 2012. Invited PMCID: PMC2917607.
commentary: GE-​Whiz! Ratcheting gene-​environment studies up to the Wellcome Trust Case Control Consortium. 2007. Genome-​wide association
whole genome and the whole exposome. Am J Epidemiol, 175(3), 203–​ study of 14,000 cases of seven common diseases and 3,000 shared con-
207; discussion 208–​209. PMCID: PMC3261438. trols. Nature, 447(7145), 661–​678. PMCID: PMC2719288.
Thomas G, Jacobs KB, Kraft P, et al. 2009. A multistage genome-​wide associa- Witte JS. 2007. Multiple prostate cancer risk variants on 8q24. Nat Genet,
tion study in breast cancer identifies two new risk alleles at 1p11.2 and 39(5), 579–​580. PMID: 17460686.
14q24.1 (RAD51L1). Nat Genet, 41(5), 579–​584. PMCID: PMC2928646. Wolpin BM, Kabrhel C, Varraso R, et al. 2010. Prospective study of ABO blood
Thompson DJ, O’Mara TA, Glubb DM, et al. 2016. CYP19A1 fine-​mapping type and the risk of pulmonary embolism in two large cohort studies.
and Mendelian randomization: estradiol is causal for endometrial cancer. Thromb Haemost, 104(5), 962–​971. PMCID: PMC3150546.
Endocr Relat Cancer, 23 (2), 77–​91. PMCID: PMC4697192. Wray NR, Purcell SM, and Visscher PM. 2011. Synthetic associations cre-
Thompson IM, Ankerst DP, Chi C, et  al. 2005. Operating characteristics of ated by rare variants do not explain most GWAS results. PLoS Biol, 9(1),
prostate-​specific antigen in men with an initial PSA level of 3.0 ng/​ml or e1000579. PMCID: PMC3022526.
lower. JAMA, 294(1), 66–​70. PMID: 15998892. Wray NR, Yang J, Goddard ME, and Visscher PM. 2010. The genetic interpre-
Thompson WD. 1991. Effect modification and the limits of biological infer- tation of area under the ROC curve in genomic profiling. PLoS Genet,
ence from epidemiologic data. J Clin Epidemiol, 44(3), 221–​ 232. 6(2), e1000864. PMCID: PMC2829056.
PMID: 1999681. Wu C, Kraft P, Stolzenberg-​Solomon R, et al. 2014. Genome-​wide association
Thrift AP, Gong J, Peters U, et al. 2015a. Mendelian randomization study of study of survival in patients with pancreatic adenocarcinoma. Gut, 63(1),
height and risk of colorectal cancer. Int J Epidemiol, 44(2), 662–​672. 152–​160. PMCID: PMC3816124.
PMCID: PMC4481609. Wu C, Kraft P, Zhai K, et  al. 2012. Genome-​wide association analyses of
Thrift AP, Gong J, Peters U, et  al. 2015b. Mendelian randomization study esophageal squamous cell carcinoma in Chinese identify multiple sus-
of body mass index and colorectal cancer risk. Cancer Epidemiol ceptibility loci and gene-​environment interactions. Nat Genet, 44(10),
Biomarkers Prev, 24(7), 1024–​1031. PMCID: PMC4490960. 1090–​1097. PMCID: PMC3513832.
Tung N, Battelli C, Allen B, et al. 2015. Frequency of mutations in individuals with Wu X, Wang L, Ye Y, et al. 2013. Genome-​wide association study of genetic
breast cancer referred for BRCA1 and BRCA2 testing using next-​generation predictors of overall survival for non-​small cell lung cancer in never
sequencing with a 25-​gene panel. Cancer, 121(1), 25–​33. PMID: 25186627. smokers. Cancer Res, 73(13), 4028–​4038. PMCID: PMC3719971.
Turnbull C, Ahmed S, Morrison J, et al. 2010. Genome-​wide association study Wu Y, Waite LL, Jackson AU, et al. 2013. Trans-​ethnic fine-​mapping of lipid
identifies five new breast cancer susceptibility loci. Nat Genet, 42(6), loci identifies population-​specific signals and allelic heterogeneity that
504–​507. PMCID: PMC3632836. increases the trait variance explained. PLoS Genet, 9(3), e1003379.
Umar A, Boland CR, Terdiman JP et  al. 2004. Revised Bethesda Guidelines PMCID: PMC3605054.
for hereditary nonpolyposis colorectal cancer (Lynch syndrome) Yandell DW, Campbell TA, Dayton SH, et al. 1989. Oncogenic point mutations
and microsatellite instability. J Natl Cancer Inst, 96(4), 261–​ 268. in the human retinoblastoma gene: Their applications to genetic counsel-
PMCID: PMC2933058. ing. N Engl J Med, 321(25), 1689–​1695.
Vilhjalmsson BJ, Yang J, Finucane HK, et al. 2015. Modeling linkage disequi- Yang J, Lee SH, Goddard ME, and Visscher PM. 2011. GCTA:  a tool for
librium increases accuracy of polygenic risk scores. Am J Hum Genet, genome-​wide complex trait analysis. Am J Hum Genet, 88(1), 76–​82.
97(4), 576–​592. PMCID: PMC4596916. PMCID: PMC3014363.
Visscher PM, Hill WG, and Wray NR. 2008. Heritability in the genom- Yeager M, Chatterjee N, Ciampa J, et al. 2009. Identification of a new pros-
ics era:  concepts and misconceptions. Nat Rev Genet, 9(4), 255–​266. tate cancer susceptibility locus on chromosome 8q24. Nat Genet, 41(10),
PMID: 18319743. 1055–​1057. PMCID: PMC3430510.
Voight BF. 2014. MR_​predictor: a simulation engine for Mendelian randomiza- Yeager M, Orr N, Hayes RB, et al. 2007. Genome-​wide association study of
tion studies. Bioinformatics, 30(23), 3432–​3434. PMID: 25165093. prostate cancer identifies a second risk locus at 8q24. Nat Genet, 39(5),
Wacholder S, Chanock S, Garcia-​Closas M, El Ghormli L, and Rothman N. 645–​649. PMID: 17401363.
2004. Assessing the probability that a positive report is false: an approach Zhang B, Shu XO, Delahanty RJ, et al. 2015. Height and breast cancer risk: evi-
for molecular epidemiology studies. J Natl Cancer Inst, 96(6), 434–​442. dence from prospective studies and mendelian randomization. J Natl
PMID: 15026468. Cancer Inst, 107(11). PMCID: PMC4643630.
76

76 Part I:  Basic Concepts


Zhang C, Doherty JA, Burgess S, et al. 2015. Genetic determinants of telomere Zondervan KT, and Cardon LR. 2007. Designing candidate gene and genome-​
length and risk of common cancers:  a Mendelian randomization study. wide case-​control association studies. Nat Protoc, 2(10), 2492–​2501.
Hum Mol Genet, 24(18), 5356–​5366. PMCID: PMC4550826. PMCID: PMC4180089.
Zheng J, Li Y, Abecasis GR, and Scheet P. 2011. A comparison of approaches Zuk O, Hechter E, Sunyaev SR, and Lander ES. 2012. The mystery of miss-
to account for uncertainty in analysis of imputed genotypes. Genet ing heritability: genetic interactions create phantom heritability. Proc Natl
Epidemiol, 35(2), 102–​110. PMCID: PMC3143715. Acad Sci U S A, 109(4), 1193–​1198. PMCID: PMC3268279.
Zheng W, Long J, Gao YT, et al. 2009. Genome-​wide association study identi- Zuk O, Schaffner SF, Samocha K, et al. 2014. Searching for missing heritabil-
fies a new breast cancer susceptibility locus at 6q25.1. Nat Genet, 41(3), ity: designing rare variant association studies. Proc Natl Acad Sci U S A,
324–​328. PMCID: PMC2754845. 111(4), E455–​E464. PMCID: PMC3910587.
 7

6 Application of Biomarkers in Cancer Epidemiology

ROEL VERMEULEN, DOUGLAS A. BELL, DEAN P. JONES, MONTSERRAT GARCIA-​CLOSAS,


AVRUM SPIRA, TERESA W. WANG, MARTYN T. SMITH, QING LAN,
AND NATHANIEL ROTHMAN

OVERVIEW chapter we described (1) how biomarkers could be used in epidemio-


logical studies, (2) the importance of accurate biomarker characteriza-
Advancements in OMICs (e.g., genomics, epigenomics, transcrip- tion, including knowledge about inter-​individual and intra-​individual
tomics, metabolomics, adductomics, proteomics) have enabled inves- variability in biomarker response, and (3) how laboratory assay vari-
tigators to explore comprehensively the biological consequences of ability could impact the biomarker disease associations. These fun-
exogenous and endogenous exposures, to detect (1) molecular sig- damentals, rooted in measurement error theory, still apply in the era
natures of exposure, (2) early signs of adverse biological effects, and of large-​scale OMICs analyses, with the exception that for non-​fixed
(3) preclinical disease, and (4) to further classify subtypes of can- biomarker endpoints we now need to assess the distribution of inter-​
cer at the molecular level. These new technologies have proven to individual, intra-​individual, and laboratory variability for hundreds to
be invaluable for assembling a comprehensive molecular portrait of thousands of signals before commencing a study. As such, the inter-
human exposure, health, and disease. This includes hypothesis-​driven pretation of studies using high-​dimensional platforms should be done
biomarkers, as well as platforms that can comprehensively analyze with some care where positive findings may have meaning (i.e., if
entire biologic processes and “compartments” agnostically, includ- replicated and biases can be excluded) but where null findings have
ing the measurement of small molecules (metabolomics), DNA limited meaning. In the absence of knowing the intraclass correla-
polymorphisms, and rarer inherited variants (genomics), methyla- tions coefficients (ICCs  =  intra-​individual variance /​total variance)
tion and microRNA (epigenomics), chromosome-​wide alterations, of these null findings, an interpretation of the findings cannot be given
mRNA (transcriptomics), proteins (proteomics), the microbiome and subsequent pathway analyses may be biased. (For a more detailed
(microbiomics), and other biologic components within the human discussion on the effect of measurement error on the interpretation of
body. Although the implementation of these technologies in cancer molecular data, see Garcia-​Closas et al., 2006; International Agency
epidemiology already has shown great promise, some challenges of for Research on Cancer [IARC], 2011).
particular importance still need to be addressed to exploit the full In the current update of the chapter we focus on describing these
potential of these platforms. Non-​genetic OMICs markers vary over new generations of high-​dimensional OMICs technologies and dis-
time; therefore there is an urgent need for cohorts to collect and repeat cuss their implementation in the analysis of non-​germline biomarkers
biological samples over time. In addition, due to the growing insights in healthy tissue in epidemiological studies, with a focus on etio-
that early-​life events have great importance for disease development logic studies. A  discussion of infectious, nutritional, immunologic,
later in life, it is desirable to follow up cohort studies of children germline, and somatic tumor markers is provided in Chapters  2–​4,
and adolescents with biologic samples over their lifetime. As there 5, 19, 24, and 25, respectively, of the current textbook. Biomarkers
is growing evidence that many of the OMIC signals are tissue or to assess other categories of exposures and lifestyle factors are
cell-​specific, more insights in transferability of OMIC signals across described in additional chapters. For a comprehensive review of con-
organs, tissues, and cells are necessary. Lastly, the use of these high-​ ceptual issues in the application of biomarkers in epidemiological
dimensional platforms will result in vast amounts of interconnected studies, a detailed discussion of technologies used to measure a wide
data. Therefore, procedures and methodologies to collect, manage, range of biomarkers, and their application in epidemiologic studies,
and analyze such data will need to be generated. see the textbook Molecular Epidemiology: Principles and Practices
(IARC, 2011).

INTRODUCTION
BIOMARKER CATEGORIES
As noted in the third edition of Schottenfeld and Fraumeni’s Cancer
Epidemiology and Prevention, the prediction that biomarkers are Classically, biomarkers have been classified in discrete entities along
increasingly being incorporated into cancer epidemiology (Garcia-​ the pathway from exposure to disease. The National Research Council
Closas et al., 2006) has truly materialized in the last decade. A search in 1987 (Committee on Biological Markers of the National Research
in PubMed of the keywords “epidemiology,” “biomarkers,” and “can- Council, 1987) subdivided the process into markers of (1) exposure,
cer” provides us with a convincing picture of a steady increase in the (2)  internal dose, (3)  biologically effective dose, (4)  early biologi-
number of studies incorporating biomarkers in cancer epidemiology cal effect, (5)  altered structure or function, and (6)  clinical disease;
research, with a sharp increase in the number of publications after each of these steps could be influenced by host susceptibility, includ-
2010 (Figure 6–​1). ing genetic traits and effect modifiers (e.g., social economic status,
This trend follows quite well the technological advances that have gender, or diet) (Figure 6–​3). This framework has been very useful
been made in array and sequence-​based OMICs analyses where the in the pre-​OMIC era, where many of the hypothesis-​driven markers
number of markers being measured can be described by an exponential measured fit in these discrete steps with, for example, urinary metabo-
function following closely Moore’s law (Figure 6–​2). lites reflecting internal dose, DNA adducts biologically effective dose,
Since the publication of the third edition of Cancer Epidemiology cultured peripheral lymphocyte chromosomal aberrations reflecting
and Prevention, many of the OMICs technologies have been marked early biological effects, and tumor mutations in specific genes reflect-
by tremendous progress in the collection, analysis, and interpretation ing the disease. However, in the era of OMICs these distinctions
of OMICs data. In addition, new OMICs technologies have emerged have become less clear, as many of these technologies may measure
(e.g., adductomics, microbiomics). In the previous edition of this more than one process. For example, metabolomics has been used

77
78

78 Part I:  Basic Concepts


1600

1400

1200

Number of publications
1000

800

600

400

200

0
1985 1990 1995 2000 2005 2010 2015 2020
Year

Figure 6–​1.  Number of publications based on the keywords “epidemiology,” “biomarkers,” and “cancer” in PubMed.

to measure both exogenous exposures and biological effects (Jones, different levels of biological regulation, providing a comprehensive
2016). Likewise, epigenetic changes have been linked to both exog- multidimensional picture of disease mechanisms. The use of OMICs
enous exposures, early biological effects, and disease (Guida et  al., technologies to measure an exposure either directly or by its imprints
2015; Joehanes et al., 2016; Shenker et al., 2013). These new technolo- in the biological system has been termed exposomics and may enhance
gies therefore provide, on one hand, the unique opportunity to truly exposure assessment (Vineis et  al., 2016; Wild, 2005, 2012)  and at
measure the exposures and associated biological processes across the the same time provide mechanistic insights (Figure 6–​3). Other bio-
continuum from exposure to disease; on the other hand, because of markers, often more downstream in the exposure–​disease continuum
the fact that they reflect potentially different steps of the multistage and lacking identifiable exposure specificity, can provide mechanis-
process from exposure to disease, interpretations may not always be tic insights and, potentially, have clinical applications (Figure 6–​3).
straightforward, and questions concerning the specificity of the signals At the same time, with the availability of next generation sequenc-
will need to be answered. Of course the major scientific leap that has ing data on whole exomes and whole genomes from a wide range of
been offered by OMICs technologies is the change from hypothesis-​ tumors, and the application of new bioinformatic methods, a series
driven marker research to agnostic screening of hundreds to hundreds of mutational signatures have recently been identified that have been
of thousands of biological markers (Figure 6–​2). This now allows, as linked to a number of exposures, including particularly striking and
in genomics, broad screening of biomarkers along the continuum from consistent findings from certain components of tobacco smoking
exposure to disease. It furthermore allows the integration of signals at (Alexandrov et al., 2016). This suggests that full characterization of

Development of OMIC technologies


10,00,00,000 10000000

100,00,000 1000000
Number of Analytes (other OMICs)

10,00,000 100000
KB per Day (Genomics)

100,000 10000

10,000 1000

1000 100

100 10

10 1

1 0.1
1996–2000 2001–2005 2006–2010 2011–2015 2016–2020

Genomics Epigenetics Metabolomics Adductomics Proteomics

Figure 6–​2.  Number of KB per day and analytes per day for the different OMICs platforms by 5-​year intervals.
 79

Application of Biomarkers in Cancer Epidemiology 79


Early Altered
Internal
Exposure biological structure/ Disease
dose
effects functions

Exposure Assessment Mechanistic/Clinical


(Exposomics)

Genetic/other sources
of susceptibility

Figure 6–​3.  Defining a continuum of biomarker categories that reflect the carcinogenic process.

somatic alterations in tumors could provide important information for others, contain more than 50,000 metabolites. The total number of
at least some of the exposures that drive cancer risk. The use of mark- metabolites appears to be more than one million when including the
ers to identify mechanisms of disease and for early detection is the large number of lipids, complex carbohydrates, phytochemicals, and
domain of mechanistic and clinical research. In exposomics, the inten- environmental chemicals. Uncertainties in chemical identification and
sity, duration, persistence, and specificity of the exposure signal are quantification discussed earlier, along with limitations in databases
the most important characteristics of a successful exposure marker. In and knowledge bases, necessitate caution at every step in analysis and
the next section we discuss several of these new technologies and their interpretation. Additionally, because of the multiple and changing ana-
implementation in cancer epidemiology. lytical platforms, extra effort is warranted in providing metadata for
sample collection, processing, and analysis, to facilitate evaluation of
reproducibility between studies.
NEW BIOMARKER TECHNOLOGIES
High-​Resolution Metabolomics for Exogenous
Metabolomics and Endogenous Exposure Screening
Due to its large dynamic range, metabolomic measurements now cover
Metabolomics refers to the study of small molecules, typically with both exogenous (environmental, occupational, lifestyle) and endoge-
molecular mass less than 2000 Dalton. This includes the molecular nous molecules. This allows metabolomics to be used to directly meas-
fuels and building blocks for all macromolecular components, as ure not only internal doses of exogenous and endogenous compounds,
well as degradation and waste products generated by the body and but also the biological perturbations these exposures have on the bio-
exogenous chemicals that can cause or protect against carcinogene- logical system. An example of using metabolomics to measure the
sis, including non-​nutritive components of food, microbiome-​related internal dose of exogenous compounds can be found in Walker et al.
metabolites, drugs and related metabolites, and commercial and envi- (2016), where, in a population of individuals occupationally exposed
ronmental chemicals. to trichloroethylene (TCE), an industrial degreasing agent and envi-
Contemporary metabolomics builds upon a strong history of ana- ronmental contaminant, known and unknown TCE metabolites in
lytical chemistry, which provides capabilities for the targeted analysis plasma were identified. Subsequently, these TCE metabolites were
of most metabolites and chemicals known to be relevant to cancer. linked to metabolites and biological measures indicative of immune-​
Recent studies have yielded promising leads (Armitage and Barbas, and nephrotoxicity, two known adverse-​outcome-​pathways of TCE.
2014) from targeted and non-​targeted analyses with mass spectrome- This proof-​of-​principle study provides promise that metabolomics is
try (MS), nuclear magnetic resonance (NMR) spectroscopy, and other able to screen broadly for exogenous compounds and that interactions
analytical approaches. Targeted methods are often used to test specific of these compounds with adverse-​outcome-​pathways can be identified
hypotheses, while non-​targeted methods are used in an agnostic man- (Figure 6–​4).
ner to search for differences between groups or associations with other
variables. Both approaches are useful, but more powerful analytic High-​Resolution Metabolomics for Cancer
methods now enable the measurement of thousands of chemicals in Many putative biomarker candidates have been found to differ in
biologic samples, and some believe that tens of thousands will become tissue-​specific cancer studies (Armitage and Barbas, 2014). These
practical as human exposome research develops (Uppal et al., 2016). include a diverse range of metabolic pathways, which could imply that
Terminology can be confusing for metabolomics because some different tissue-​specific tumors have very different metabolic char-
methods provide a single signal derived from multiple chemicals, acteristics and/​or that adaptive responses to cancer and cancer treat-
while others give multiple signals from one chemical. Additionally, ments are heterogeneous. Some studies show that citric acid cycle,
some data are provided with absolute measures of abundance, while glycolytic metabolites and ketone bodies differ, as expected from the
others provide only relative quantification. In discussion of data, the well-​known Warburg effect (Allegra et al., 2016). In addition, many
term metabolite is often used generically to refer to any small mol- of the essential amino acids (phenylalanine, tryptophan, lysine, his-
ecule in a living organism, regardless of origin. The term feature tidine, isoleucine, leucine, valine, methionine, threonine) as well as
or metabolic feature is used to refer to something measured, which non-​essential amino acids (glutamine, glutamate, alanine, aspartate,
has insufficient characterization to determine whether it represents a glycine, tyrosine, cysteine), differ for cancer at different sites. Amino
known chemical, a mixture of chemicals, or a product of a chemi- acid metabolites (4-​ hydroxyphenylacetic acid, homovanillic acid,
cal generated during the assay (Miller and Jones, 2014). The Human 5-​hydroxyindoleacetic acid, hydroxyproline, isovaleric acid, tau-
Metabolomics Database (HMDB), available at http://​www.hmdb.ca, is rine) also differ, as do some nucleotides, purines (methyladenosine;
an important resource for human metabolomics data; it contains infor- hypoxanthine, 8-​hydroxydeoxyguanosine), and pyrimidines (uridine,
mation on common metabolites and their concentrations in biologic methyluridine, 5-​hydroxymethyl-​2-​deoxyuridine). Additional meta-
materials. bolic differences include formate, putrescine, bile acid metabolites,
Only about 2000 metabolites are present in central human meta- and metabolites from a range of lipid pathways (free fatty acids, ultra
bolic pathways of the Kyoto Encyclopedia of Genes and Genomes long-​chain fatty acids, oxidized fatty acids, acylcarnitines, phosphati-
(KEGG), but the combined resources of KEGG, HMDB, Metlin, and dylcholines, sphingolipids, gangliosides, retinol). The information to
80

80 Part I:  Basic Concepts


Immune responsive biomarkers
TCE exposure biomarkers

Correlation coefficient

–0.7 0 0.6

Urinary TCE exposure biomarker


Urinary renal biomarker
Renal damage biomarkers Immunological markers
Unidentified halogenated m/z, by isotope pair
Identified metabolite

Figure 6–​4.  Network correlation analysis of molecular markers previously tested for association with TCE exposure and metabolic features detected
in this study. Only correlations with Spearman |r| ≥ 0.3 and p-​value ≤ 0.05 corresponding to identified metabolites or probable halogens were included.
Immune markers: CD4 = CD4+ T-​cell count; IgG = immunoglobulin G; IgM = immunoglobulin M; LY = lymphocyte cell count; mtDNA = mitochondrial DNA
turnover rate; sCD27 = soluble CD27; sCD30 = soluble CD30; WBC = white blood cell count. Nephrotoxicity markers: aGST = urinary α glutathione-​s-​transferase;
piGST = urinary pi glutathione-​s-​transferase; KIM1 = urinary kidney injury marker 1; NAG = urinary n-​acetyl-​beta-​glucosaminidase. TCE exposure markers: Conj.
TCEOH = urinary trichloroethanol glucuronide; Free TCEOH = urinary trichloroethanol; Tot TCEOH = sum of urinary trichloroethanol and trichloroethanol gluc-
uronide; TCA = urinary trichloroacetic acid. Source: Walker et al. (2016).

date suggests that metabolic responses to cancer and cancer treatment as well as useful targets to complement therapeutics targeting IGF-​1-​
are too complex to expect single metabolic biomarkers to approach the related signaling pathways.
utility of some genetic and proteomic markers. Similar arguments can be made for mitochondria-​related metabo-
Despite this, the cumulative findings provide a foundation for lites and other metabolites found to differ in cancer studies.
improved experimental and analytical design and point to new stra­
tegies for biomarker development. The data show that differences
exist in most of the essential amino acids. Protein synthesis requires High-​Resolution Metabolomics for Cancer
essential amino acids for rapid cell growth, so the data indicate that Therapeutics
differences in circulating essential amino acid concentrations could As precision medicine methods are developed, metabolomics is
reveal characteristics of tumor vulnerability to anti-​metabolites that assuming a central role because of the central role of metabolism
disrupt essential amino acid transport or utilization. Essential amino in bioenergetics, catabolism, and anabolism. Newer high-​resolution
acid metabolism is directly linked to the mTOR pathway, known to methods are likely to play an increasingly important role because the
be responsive to growth factors, nutrients, energy and stress signals, cost for analysis of > 10,000 metabolic features is less than that for
and essential signaling pathways, such as PI3K, MAPK and AMPK, analysis of 300 metabolites by more conventional methods. A major
to control cell growth, proliferation, and survival. Inhibitors of mTOR difference is the use of computational methods for data extraction
are under active investigation for tumor therapeutics, and the metabol- (Yu et al., 2013). With ultra-​high resolution MS, rigorously defined
omics data suggest that some of the essential amino acids may prove chromatographic procedures, and replicate analyses of single sam-
to be useful biomarkers for therapeutic efficacy, as well as indicators ples, improved detection and reproducibility are obtained. With this
of useful targets to complement mTOR inhibitors with inhibitors of approach, unidentified features (ions) can be defined in terms of m/​z
essential amino acid utilization. (mass-​to-​charge ratio), retention time (seconds), and ion intensity.
Differences in non-​essential amino acids may also provide useful For features that are present in a pooled reference sample, such
predictive biomarkers for the use of therapeutic adjuvants. Glutamine as NIST SRM1950 from the National Institute of Standards and
is an anabolic amino acid linked to IGF-​1 signaling that inhibits apop- Technology, identity can be retrospectively assigned upon curation
tosis and supports tumor cell growth. Glutamate and aspartate are of the pooled reference. Recent advances further establish a refer-
mitochondrial energy substrates, but perhaps more important, nucle- ence standardization protocol to quantify chemicals that are con-
otide biosynthesis depends upon these amino acids. Non-​essential firmed and quantified in a pooled reference analyzed concurrently
amino acids also provide a sparing effect for glucose, which is needed with unknown samples. In principle, this can be used for hundreds
for NADPH supply for rapid cell growth. Cysteine is a precursor for to thousands of metabolites and includes most of the amino acids,
the antioxidant GSH, also known to support tumor resistance to cancer water-​soluble vitamins, and a broad spectrum of energy metabo-
therapeutics. Thus, like the essential amino acids, the non-​essential lites, lipids, metabolic degradation products, and markers of organ
amino acids could provide useful biomarkers for therapeutic efficacy, function.
 81

Application of Biomarkers in Cancer Epidemiology 81


Key issues for the use of high-​resolution metabolomics data for As indicated earlier, an important advantage of high-​resolution
epidemiology are similar to those faced for other OMICs technolo- metabolomics is that measures are included for most metabolic
gies and include the need to protect against false discovery from large pathways, which is an advantage over the more targeted analyses
numbers of statistical tests, the means to include unidentified measures using platforms such as Biocrates ​or Brainshake. Thus, data are often
into analysis, and strategies to link multiple significant features into obtained with many apparently significant features, many of which
common conceptual models. Large numbers of tests are often sum- have not been curated and all of which could be true or could repre-
marized in terms of a metabolome-​wide association study (MWAS) sent false discovery. Some aspects, such as multiple features derived
(Figure 6–​5(a)) in which the negative log p is plotted against the m/​z from one chemical, deviate from assumptions upon which statistical
or chromatographic retention time, analogous to genome-​wide associ- methods are based. Thus, statistical tests and respective p values may
ation study (GWAS). be better viewed as a way to prioritize features for more detailed
In a standard workflow, selected features are used with a clustering examination. With this in mind, one has a choice to select the top fea-
algorithm (e.g., hierarchical cluster analysis; HCA) or principal com- tures using either raw p value or false discovery rate (FDR) threshold
ponent analysis (PCA) to determine how effectively individuals are criteria. An advantage of the former is that all of these features pass
classified or separated by the selected features (Figure 6–​5(c)). Insight threshold criteria for significance (i.e., this list includes all features
into underlying disease etiology and therapeutic mechanisms can be that truly differ). The disadvantage is that there is certainty that many
obtained from the metabolomics data using additional biostatistics, of these have been detected by chance. To overcome this limitation,
bioinformatics, and pathway analysis tools (Figure 6–​5(d)). one can use pathway enrichment to test for significant pathways. For

(a) MWAS of TB disease (b) Selected metabolites that differ

m/z 148.0594 m/z 399.2116 m/z 801.5767


9 Glutamate D-series resolvins αTrehalose-6-mycolate
6 20 20
8

Log (2) Intensity


7
0
6

5 0 0
– log P

–6 –4 –4
4
m/z 851.5953 m/z 237.0192 m/z 271.2319
Phosphatidylinositol Unidentified Unidentified
3 20 20 6
2
Log (2) Intensity

0
1

0
0
200

800

1400

2000

0
–1 –4 –16
HC TB HC TB HC TB
m/z

(c) 2-Way hierarchical cluster analysis


(d) KEGG Brite Classification

Raw Z Score
Color Key 5%
3%

–2 0 2
17% Commercial products

Control Environmental chemicals


1
Intermediatory metabolites
TB 47%
2 2% Microbial metabolites
3 Pharmaceuticals
m/z Clusters

4 Plant-derived metabolites

5 17%

6
7 9%
8

Figure 6–​5.  High-​resolution metabolomics workflow. (a) An MWAS was performed for pulmonary tuberculosis (TB) patients compared to uninfected
household controls. Respective broken lines, from bottom: raw p = 0.05; FDR = 0.2; FDR = 0.05. (b) Selected metabolites that differ in A are plotted using
box and whiskers plots. (c) Two-​way hierarchical cluster analysis of metabolites that differ in A shows that the metabolites separate the individuals and
that the metabolites are associated into clusters. (d) Use of the Kyoto Encyclopedia of Genes and Genomes (KEGG) Brite Classification of metabolom-
ics database matches gives a depiction of the types of chemicals that differ between patients and controls. FDR: false discovery rate. Source: Data are from
Frediani et al. (2014).
82

82 Part I:  Basic Concepts


instance, by testing all features significant at raw p < 0.05 for enrich- on or off of expression of lineage-​determining transcription factors
ment of metabolites in specific pathways relative to that obtained by (see Figure 6–​6 for a simplified depiction) (Bernstein et  al., 2007;
a random selection of features from the total feature table, one can Gifford et al., 2013; Ziller et al., 2013).
identify pathways of interest. Mummichog (Li et al., 2013) is a pro- New patterns are established by de novo methyltransferases and
gram designed to facilitate the use of high-​resolution data by using histone-​modifying complexes, and once these are established, cell-​
all selected features to test for pathway enrichment without require- type identity is preserved by epigenetic maintenance factors. Signals
ment for chemical identity prior to statistical analysis. from the environment (diet, metabolic stress, or exposure-​related) can
An alternative approach uses features that achieve a desired FDR perturb these epigenetic patterns over a short period of time (Philibert
threshold along with MetabNet (Uppal et  al., 2015), a program that et al., 2012), such as during fetal development (Joubert et al., 2012,
tests for features that are significantly correlated with selected metabo- 2014; Marsit, 2015)  and the effects may be transient or persistent
lites. This approach is based upon the observation that the concentra- (Richmond et  al., 2015; Tobi et  al., 2014; Wan et  al., 2012). Aging
tions of metabolites within a metabolic pathway are often correlated. drives epigenetic changes in blood and tissues (Horvath, 2013), and
By selecting metabolites that are correlated with a feature that is sig- these changes have been associated with mortality (Marioni et  al.,
nificant at an appropriate FDR threshold, pathway enrichment analysis 2015). It has been proposed that environmentally caused epigenetic
by Mummichog can provide evidence for important pathways, even for changes may be an important mechanism connecting exposure to later
unidentified features associated with disease. An example is provided chronic disease risk (Barker and Clark, 1997; Heijmans et al., 2008;
by a pentachlorofuran associated with age-​related macular degenera- Herceg et al., 2013; Tobi et al., 2014). In animal model systems there
tion (Osborn et al., 2013). In principle, this provides means to detect is some evidence that epigenetic effects may persist across multiple
pathway associations of unidentified metabolites associated with can- generations (Jirtle and Skinner, 2007; Rando and Verstrepen, 2007).
cer for further evaluation. Tumor studies have demonstrated that tumor-​associated epigenetic
patterns, including epigenetic inactivation of tumor suppressor genes,
Integration of Metabolomics with Other are transmitted as part of clonal growth and, along with somatic muta-
OMICs Platforms tions, are important determinants of the tumor transcriptome and phe-
As indicated earlier, metabolomics data can provide useful insight into notype (Christensen et al., 2011; Hansen et al., 2011; Landau et al.,
disease etiology, but evolving methods indicate that these data may 2014; McDonald et  al., 2011). In addition, characteristics of tumors
be even more useful in combination with phenotypic markers and/​or include a disorganized epigenetic state, loss of epigenetic lineage
genomics, transcriptomics, and other OMICs data. Two advantages are identity, and stem cell-​like proliferative capacity (Hansen et al., 2011;
evident from schema describing the functional genome and its interac- Jones and Baylin, 2007; Pujadas and Feinberg, 2012).
tions with the environment (diet, microbiome, chemical exposures). First,
the metabolome includes metabolic products of the other components DNA Methylation, Chromatin State, and
of the functional genome. This means that metabolic associations with Transcriptomics
genetic, epigenetic, transcription, or proteomic variation can be used
to infer functional relationships. Thus, corresponding metabolic asso- Recent studies indicate that each of these interacting features have
ciations can provide insight into genome or transcriptome associations genomic patterns that can be used to distinguish cell type, cell lineage,
with cancer. A second advantage is that the metabolome includes dietary, and disease tissue (Bernstein et al., 2007; Bock et al., 2012; Schubeler,
microbiome-​related, and environmental chemicals. These metabolites 2015; Thurman et  al., 2012; Ziller et  al., 2013), but each provides
provide measures of exogenous influences relevant to cancer. different information and has advantages and disadvantages as a bio-
In summary, an accumulating literature demonstrates potential for marker (including methodological). Transcriptome patterns (mRNA,
metabolomics to provide new cancer biomarkers. While well founded, lncRNA, eRNA, miRNA) are highly informative as to current cell
this interpretation also must be viewed within the reality of the com- state, but patterns may be transient and there are many situations when
plexity of cancer. At the very best, metabolomics biomarkers could be it is difficult to obtain high-​quality RNA. The ENCODE and Roadmap
useful to define risk of cancer, to support early detection of cancer, to Epigenome projects have clearly demonstrated that specific histone
improve ways to predict benefit and risk from specific treatment strat- modifications (e.g., H3K4me3) are strongly correlated with transcrip-
egies, and to provide means to follow treatment efficacy and disease tion (Ernst et  al., 2011), while other modifications (H3K27Ac) are
progression. At the very worst, however, failed biomarkers can mislead highly correlated with the DNA methylation status of gene regulatory
in cancer prevention, cause misdiagnosis, contribute to cancer develop- regions (Ziller et al., 2013). However, chromatin studies have primar-
ment and progression, or misinform clinicians and patients concerning ily focused on large scale multi-​tissue characterization (Kundaje et al.,
treatment efficacy and disease progression. These realities force caution 2015) or on mechanisms using model systems (Bernstein et al., 2007).
and skepticism in the quest for metabolomics biomarkers for cancer. Techniques that provide more general genome-​wide chromatin state
information (like DNaseI-​sequencing or the transposase-​based ATAC-​
seq) have advanced rapidly and are being applied in larger and larger
Epigenetics studies. But these chromatin approaches are still technically challeng-
Epigenetics is the transmission of gene regulatory information from ing, relatively costly, and generally require the processing of freshly
parent cell to daughter cell (either germline or somatic) via modifi- isolated cells. Although the relationships are complex, we can assume
cations of cytosines in DNA or histones rather than DNA sequence that persistent changes of chromatin state and the transcriptome are
(Bernstein et al., 2007). Epigenomic studies examine this information, likely to be accompanied by altered DNA methylation either within
be it DNA methylation or histone modification (chromatin state) pat- the promoter, the gene body, or in associated enhancers (Schmidl
terns, at a genome-​wide scale (Fazzari and Greally, 2004). As with et al., 2009; Schubeler, 2015). In addition, the relative ease of use and
other biomarkers, epigenetic measurements have potential for detect- cost-​ effectiveness of measuring DNA methylation on microarrays
ing signatures of exposure, early signs of cellular pathology, dis- have rapidly expanded their potential application in population stud-
ease susceptibility, or preclinical disease (Feinberg and Fallin, 2015; ies, to detect exposure-​related effects as well as disease susceptibility
Joubert et al., 2012; Nelson et al., 2011; Reynolds et al., 2015). Rather (Michels et al., 2013).
than comprehensively annotating many studies, we will outline gen-
eral concepts and principles for epigenetic studies in populations and Methods for  Measuring 5-​Methylcytosine.  Cytosines in
review several examples. CpG dinucleotides are either methylated or not; however, the measure-
ment of methylation at a single CpG dinucleotide, or groups of CpGs,
Transmission of Epigenetic Information is a continuous variable that is expressed as the ratio or percent of
Normal development and differentiation of embryonic cells into tis- methylated signal to unmethylated signal (e.g., 0.0–​1.0, or 0–​100%).
sues involve dynamic, concerted genome-​wide changes of DNA meth- Measurement typically involves bisulfite treatment to convert unmeth-
ylation, chromatin state, and transcription, and the subsequent turning ylated cytosines to uracils, and following amplification of the signal,
 83

Application of Biomarkers in Cancer Epidemiology 83


(a) (b)
Repressed Repressed
CpG Enhancer Promoter
No DNA methylation, chromatin, TF
5mC 5mC Transcription binding, and gene expression change
TSS with lineage commitment
5mCpGs
Gene exon
CpG
TF Binding Sites Promoter Pluripotent Stem Cell
Island

Progenitor Cell

Lineage Specific
Progenitor Cell
+ H3K27me3
DNMT HMT HDAC

TET HDM HAT


Terminally Differentiated Cells

Active Enhancer Active Promoter


eRNA (c) (d) (e)
TF TSS Transcription
5OHmCpG Pol2 Pol2 Exposure Exposure
DNA
Gene exon
Methylation
TF Binding Sites CpG Island Promoter Biomarker of
DNA Methylation Risk or
Biomarker of Preclinical
Pathology Disease
* * * # # # DNA
Pol2 Pol2
Methylation
# # Biomarker
H3K4me3
* H3K27ac
Disease Disease Disease
# H3K4me3

DNMT - DNA methyltransferase, TET - demethylase


HMT - Histone methyltransferase, HDM - Histone demethylase
HAT - Histone acetyltransferase, HDAC - Histone deacetylase

Figure 6–​6.  (a) Repressed enhancers and genes display high levels of DNA methylation, repressive histone modifications (H3K27me3), and no tran-
scription. Conversion to active state is accompanied by demethylation of the enhancer, transcription factor binding, activating histone modification
(H3K27ac, H3K4me3), and transcription. (b) Cell-​type lineage specification is accompanied by the repression and activation of lineage-​specific
genes, particularly transcription factors. (c) Exposure may drive methylation changes that are independent of the disease pathway. (d) Exposure-​
induced change in DNA methylation may represent an intermediate development of disease. (e) Altered DNA methylation in a tissue may be a marker
of susceptibility or an early sign of disease tissue.

these are detected as thymine using genotyping or sequencing meth- these studies have not always been consistent (Brennan and Flanagan,
ods. Most assays cannot distinguish 5-​methylcytosine from 5-​hydroxy 2012; Hsiung et al., 2007; Nelson et al., 2011) and the lack of speci-
methylcytosine (a demethylation intermediate step). About 10% of ficity is problematic.
the 20 million or so CpGs in the genome are concentrated into CpG Methylation-​ specific polymerase chain reaction (Herman et  al.,
“islands” (CGIs) that are associated with genes. Typically these CGIs 1996) and pyrosequencing (Colella et al., 2003) are both useful tech-
are unmethylated; however, fully methylated CGIs are often a feature niques for quantifying methylation levels in a small target region.
of repressed or silenced genes, and many CGIs become methylated There are numerous varieties of these approaches (Accomando et al.,
in differentiated tissues (Fazzari and Greally, 2004). Fully methylated 2012; Leng et al., 2012; Ma et al., 2015; Wan et al., 2012). The gold
CpG sequences located in the > 500,000 retrotransposon repeat ele- standard for qualitative genome-​wide CpG analysis is whole genome
ments in the genome are also associated with the repression of expres- bisulfite sequencing (WGBS; Bernstein et al., 2007) to 30–​50 x depth
sion of these elements. Global methylation techniques measure the of coverage. While this technique has primarily been used to character-
average level of methylation of all CpGs (or a subsample) across the ize differences among tumors or purified cell types, it is increasingly
genome, and the methylation levels of CpG islands and repeat ele- being used to profile different individuals as the cost of sequencing
ments contribute greatly to measurements of global methylation level. whole genomes comes down (Hansen et  al., 2011; Kundaje et  al.,
Important early studies of methylation found tumors to be hypometh- 2015; Ziller et al., 2013). At this time, the technical variability and ana-
ylated relative to normal tissue (reviewed in Jones and Baylin, 2007). lytical reproducibility of whole genome sequencing methods have not
The concept that global methylation levels might predict risk has been been evaluated. Reduced representation bisulfite sequencing (RRBS),
pursued in blood DNA, primarily using PCR-​based pyrosequencing which can measure methylation at several million sites near CpG-​rich
methods that examine CpG methylation in repetitive elements such as regions, has recently been used effectively in a population study (Tobi
Alu repeats and long interspersed nucleotide elements (LINE; Yang et al., 2014) with validation by PCR-​mass spectrometry. However, the
et al., 2004), which sample the genome in hundreds of thousands of use of microarrays for reproducibly quantifying CpG methylation lev-
locations. This approach can detect differences in methylation levels els, such as the 450K Human Methylation or 850K MethylationEPIC
among controls and cancer cases (Moore et al., 2008), but results from arrays, has revolutionized epigenomic studies in populations, and the
84

84 Part I:  Basic Concepts


newer version provides additional informative CpGs located in gene and find methylation level at AHRR to be a robust biomarker with
regulatory regions. regard to misclassification, dose-​response, and time since quitting
(Shenker et  al., 2013). However, as reported by Bauer et  al. (2015)
Epigenomics in  Population Studies. Study designs that and Su et  al. (2016), some smoking-​associated differential methyl-
compare differences in DNA methylation between groups of unex- ated regions (DMRs) may be specific to a cell-​type lineage, and this
posed and exposed individuals, or cases and controls, should follow needs to be considered in the interpretation of results. With regard to
established GWAS recommendations involving appropriate sample other environmental exposures, relatively few published studies have
size, multiple comparisons, or FDR, controlling for population struc- both detailed quantitative measurements of exposure (Ruiz-​Hernandez
ture, gender, age, and other demographic variables. However, there et al., 2015) and genome-​wide DNA methylation, but the number of
are a number of additional issues that must be considered, and these studies is increasing rapidly, particularly for arsenic (Argos et  al.,
have been reviewed in detail (Michels et al., 2013). It is known that 2015; Kile et al., 2014; Koestler et al., 2013; Laine et al., 2015; Rojas
CpGs are the most polymorphic dinucleotides in the genome (Tomso et al., 2015; Seow et al., 2014b) and mercury (Maccani et al., 2015).
and Bell, 2003); thus any CpGs that overlap sequence polymorphisms A number of studies have examined the influence of maternal diet,
must be filtered out prior to analyses. Importantly, whereas genotype including folic acid, and famine on DNA methylation in offspring and
will be invariant across tissues, methylation varies dramatically by have linked these exposures with various outcomes such as birthweight
cell type and the choice of tissue sample for testing will greatly affect and metabolism (Kupers et al., 2015; Steegers-​Theunissen et al., 2009;
downstream analysis and generalizability. Methylation arrays may be Tobi et al., 2014).
subject to batch effects at each of these steps: DNA extraction, bisul-
fite treatment, amplification, and hybridization; therefore normaliza- Disease Detection and Disease Association.  If changes in
tion and batch correction should be applied. Numerous programs are DNA methylation occur in nascent diseased cells, then they may be
available for this (Aryee et al., 2014; Morris and Beck, 2015; Morris useful as an intermediate biological marker of pathology, preclinical
et al., 2014; Teschendorff et al., 2013; Zhuang et al., 2012). disease, or clinical disease (Figure 6–​6 (e)). The potential benefit of
early detection approaches is enormous, but clinical application will
depend greatly on cross-​validation and characterizing the nature of the
Heterogeneity of Cell Mixtures and Limit of Detection. methylation difference. For example, it is important to determine if a
Whether DNA samples are obtained from a mixture of cells like blood, detected methylation effect represents a susceptibility that increases risk
or from purified cell types, detecting a methylation difference will of disease, a response to the presence of diseased cells, or the presence
depend on the proportion of cells displaying the altered methylated of actual diseased cells (i.e., circulating tumor DNA). Belinsky and col-
state. Thus, to have a stable measurement of unmethylated and methyl- laborators (Belinsky et al., 2005, 2007; Leng et al., 2015; Palmisano
ated sequences, there must be a sufficient number of intact genomes in et al., 2000) have developed assays that detect altered methylation sta-
the sample prior to any amplification step. Both technical variation and tus of genes involved in lung tumorigenesis and have applied these to
limit of detection will be improved if the sample contains DNA from sputum DNA to detect tumors prior to clinical diagnosis. Langevin
20,000–​100,000 cells (~0.1–​0.7 μg). If whole blood DNA is analyzed, et al. identified methylation patterns in blood associated with various
DNA methylation changes may represent an expansion of a specific solid tumors (Langevin et  al., 2014) and have recently reviewed the
cell type, a clone of immunologically stimulated cells, or groups of potential use of epigenetic alterations, including DNA methylation, for
cells from different lineages that share a similar response, but it may be early detection, diagnosis, and assessing risk of lung cancer (Langevin
that only a specific lineage and small number of cells actually display et al., 2015). Chan et al. (2013) applied bisulfite sequencing to DNA
altered methylation. In any case, accounting for heterogeneity is nec- isolated from plasma from a number of different cancer patients and
essary to differentiate between methylation differences driven by cell proposed this for early detection of cancers. Success of these methods
number or distribution changes, and those associated with the variable of early detection will depend very much on additional validation and
of interest (Houseman et al., 2012, 2015; Jaffe and Irizarry, 2014; Liu predictive accuracy relative to other clinical markers.
et al., 2013). However, if one knows that the etiology of the disease Using DNA methylation patterns to detect inherited or acquired
involves a specific blood cell type that occurs at low frequency (such disease susceptibility has important study design requirements. As
as CD56+ natural killer cells), accounting for cell type heterogene- pointed out recently (Garcia-​Closas et  al., 2013), prospectively col-
ity in whole blood may not help in finding the signal, and it may be lected samples (preferably > 10 years prior to disease diagnosis) are
important to enrich for the cell type of interest using flow sorting or critical because the presence of incipient disease may affect the meas-
immunomagnetic beads. ured endpoints, in particular, altering immune cell-​type composition.
The rheumatoid arthritis study of Liu et al. (2013), which compares
Toxicant Exposure-​ Associated Changes in DNA whole blood DNA from prevalent cases to non-​diseased controls,
Methylation.  Studies in model organisms (Jirtle and Skinner, presents a striking example. Even after adjustment for gross leuko-
2007) and human cells (Christensen et al., 2009; Joubert et al., 2012; cyte cell-​type changes, more than 50,000 CpGs displayed differential
Langevin et al., 2011; Marsit, 2015) have clearly demonstrated that a methylation at genome-​wide significance levels (p <1.2 × 10–​7) and
variety of exposures are associated with altered DNA methylation pat- the authors attribute these changes to the consequences of rheumatoid
terns in non-​tumor cells. As cells respond and adapt to exposure condi- arthritis on the immune system.
tions, remodeling of methylation patterns across the genome occurs, Identifying specific CpG loci associated with cancer risk at genome-​
and in blood, specific immune cells may be affected. Exposure-​ wide significance levels has proven difficult, but a number of reports
induced changes in DNA methylation may represent a useful bio- from prospective studies have described methylation patterns asso-
marker of exposure that is independent of toxicity or disease (Figure ciated with breast cancer (Brennan et  al., 2012; Severi et  al., 2014;
6–​6(c)), or such change may be functionally intermediate in the dis- van Veldhoven et al., 2015; Xu et al., 2013). Van Veldhoven reported
ease process (Figure 6–​6(d)). Individuals displaying exposure-​induced on WGBS on pooled prediagnostic samples in one cohort and 450K
DNA methylation changes may be at higher risk for developing dis- arrays in two others and found that decreased average methylation
ease. The detection of DNA methylation changes as a biomarker of across the genome was associated with risk, particularly in gene bod-
tobacco smoke exposure has been highly successful, clearly identi- ies (van Veldhoven et  al., 2015). Severi et  al. compared averaged
fying individuals with low levels of exposure, such as in cord blood methylation of functional promoters and regions outside promoters in
(Joubert et al., 2012), and light smokers (Philibert et al., 2012), and breast cancer cases and controls and found that higher methylation of
also those with previous exposure such as former smokers (Guida promoters was associated with increased risk, while higher methyla-
et al., 2015; Joehanes et al., 2016; Lee et al., 2015; Shenker et al., 2013; tion of non-​promoter CpGs was protective (Severi et  al., 2014). Xu
Wan et  al., 2012). Many of these tobacco smoke studies have com- et al. reported on a set of 250 differentially methylated CpGs in breast
pared self-​reported smoking, or cotinine levels, with the methylation cancer (FDR Q < 0.05) and calculated prediction accuracy relative to
changes at various loci (particularly the CpG in AHRR cg05575921), the Gail model (Xu et al., 2013). In a study looking at a CpGs near
 85

Application of Biomarkers in Cancer Epidemiology 85


2191 microRNAs, Cordero et al. identified a set of CpGs associated sequencing technologies circumvent the traditional use of annotated
with breast cancer in the EPIC study (Cordero et al., 2015). probe sets, its detection capabilities offer high dynamic ranges and
The application of epigenetic analysis to population studies of expo- additional investigative opportunities such as the detection of novel
sure, disease detection, and disease risk is increasing rapidly, and as transcripts or isoforms (Garber et  al., 2011; Trapnell et  al., 2010;
the science evolves along with the technology and analysis methods, Veneziano et  al., 2016; Wang et  al., 2009). Nevertheless, the ulti-
this approach is likely to produce important research discoveries and mate decision of selecting a profiling method should include multi-
useful biomarkers. ple considerations, including a study’s resources, design, and research
objectives.
We will highlight several studies that have employed transcrip-
tional profiling to capture the physiologic response to carcinogenic
Transcriptomics exposures. A number of chemical compounds and environmental pol-
As previously mentioned, high throughput OMICs technologies are lutants have been shown to alter the transcriptional profiles of whole
invaluable for assembling a comprehensive molecular portrait of blood as well as other tissues (Olsen et al., 2015; Rager et al., 2011;
human health and disease. This section will focus on transcriptomics, van Leeuwen et al., 2006; Wang et al., 2005). Of note, tobacco smoke
or the study of total RNA in a cell or tissue (Willingham and Gingeras, is one of the few exposures to have been thoroughly examined using
2006). In the previous edition of this chapter, transcriptomics was still gene expression profiling studies in human populations (Wild et al.,
an emerging concept with promising yet limited applications. The 2013). Several key findings in the space of cigarette use and lung
decade following its publication has been marked by tremendous prog- health have begun to underscore the promise of transcriptomics in
ress in the collection, analysis, and interpretation of transcriptomic minimally invasively monitoring exposure–​ response relationships.
data. Specifically, characterization of the human transcriptome across Specifically, whole genome profiling of histologically normal-​
a spectrum of chronic diseases has been instrumental for the identifica- appearing airway epithelial cells obtained from large and small air-
tion of clinical subtypes in cancer (Bhattacharjee et al., 2001; Golub way brushings of current and former smokers have revealed distinct
et  al., 1999), the development of diagnostic and prognostic markers mRNA patterns associated with tobacco use (Harvey et  al., 2007;
(Silvestri et al., 2015; van de Vijver et al., 2002), and the prioritization Spira et  al., 2004). Similar studies have shown that ncRNAs, such
of drug targets (Evans and Guy, 2004). as miRNAs, are also impacted by smoking and lung cancer (Beane
The application of transcriptomics in examining molecular changes et  al., 2011; Perdomo et  al., 2011; Schembri et  al., 2009); miRNA
associated with established or potential disease risk factors is promis- dysregulation, in particular, has been heavily implicated in the role of
ing for a number of reasons. For one, an individual’s transcriptome lung disease development and progression (Angulo et al., 2012). In
is highly dynamic and responsive to external stressors (Coughlin, addition, there is some evidence that exercise (Flowers et al., 2015)
2014). An intermediate between genes and protein products, total and environmental exposures such as air pollution can alter miRNA
mRNA abundance in particular is regarded as a molecular phenotype patterns (Vrijens et al., 2015). There is also some emerging evidence
that provides a biologically relevant snapshot of cellular activity. The that miRNA patterns in white blood cells may be related to future risk
measurement of changes in transcript abundance can provide critical of cancer development from prospective studies (Muti et al., 2014).
insight into the status of key biological mechanisms by which an expo- As an extension of this “field of injury,” gene expression differ-
sure might be exerting its effects. Rather than reflect the total amount ences in the airway epithelium of smokers can serve as a classifier
of exposure that comes into contact with host’s cells or target organ, for the diagnostic evaluation of lung cancer (Silvestri et al., 2015;
transcriptional profiling provides a proximal assessment of the host Spira et al., 2007), as well as a guide for the clinical management
response to an exposure’s biologically effective dose. of chronic obstructive pulmonary disease (COPD) (Steiling et  al.,
Transcriptomics can also influence the molecular underpinnings of 2013). Moreover, transcriptomic profiling studies have pinpointed
long-​term structural and functional alterations within different target concordant smoking-​induced changes in epithelial cells from the
systems (Flavell and Greenberg, 2008). Furthermore, these molecular bronchial epithelium and those obtained non-​invasively from the
profiles can be derived to account for inter-​individual variability in buccal and nasal epithelium (Sridhar et  al., 2008; Zhang X et  al.,
processes such as metabolism and repair (Rogue et al., 2012). Since 2010). Thus, a growing body of evidence suggests that transcrip-
the impact of environmental exposures can be different for each indi- tionally profiling samples collected from the relatively accessible
vidual due to a variety of genetic and non-​genetic factors, this is vital epithelial cell lining of the nose and mouth can provide additional
for accurately relating exposures to health outcomes and disease risk opportunities to examine the host response to complex environmen-
(Idaghdour and Awadalla, 2013). Accordingly, transcriptomic profil- tal exposures.
ing can constructively mediate the interplay between external expo- A framework of this environmental assessment paradigm was
sures and the physiologic host response. recently applied in the household setting of indoor air pollution stud-
Importantly, recent large-​scale genomic studies have demonstrated ies, wherein buccal epithelial brushings were used to non-​invasively
that while the majority of the human genome is transcribed into RNA, examine the host response among non-​smoking women exposed to
only a small fraction is accounted for by protein-​coding genes (Djebali smoky (bituminous) and smokeless (anthracitic) coal (Wang et  al.,
et  al., 2012). In contrast, a large proportion of transcripts belong to 2015). Specifically, whole genome microarray expression profiling
the vast array of non-​coding RNAs (ncRNAs) (Cech and Steitz, 2014; identified a signature of genes differentially expressed among female
Iyer et  al., 2015). MicroRNAs (miRNAs), for instance, are a family residents who burned smoky coal in their residences of Fuyuan and
of small ncRNAs that can regulate the transcription, translation, or Xuanwei, China. Enrichment analysis, an analytic approach that can
degradation of mRNAs (He and Hannon, 2004); miRNA are com- be executed using an evolving compendium of computational methods
monly studied in conjunction with mRNA expression levels to pro- for identifying representative functional or biologic pathways in silico
vide biologic insight to regulatory networks and targeting mechanisms (Huang da et al., 2009), indicated that this smoky coal signature was
(Devaraj and Natarajan, 2011). Relative to miRNAs, the study of other enriched for pro-​inflammatory mediators as well as changes associated
ncRNAs (e.g., piRNAs, snoRNAs, lincRNAs) in the context of disease with tobacco smoke exposure. These results provided biologic context
etiology and screening has been limited, though their contribution to to epidemiologic investigations that previously reported an association
gene regulation and the modulation of disease-​related phenotypes are of smoky coal exposure with lung cancer risk (Barone-​Adesi et  al.,
steadily being revealed (Esteller, 2011; Meseure et al., 2015). 2012; Downward et al., 2014; Lan et al., 2005; Mumford et al., 1987).
To date, global transcriptional profiling studies have largely fea- Notably, human peripheral blood is another source of relatively
tured cDNA and oligonucleotide-​based microarray platforms, with accessible biological material whose transcriptomic utility should
a shifting focus toward deep sequencing methodologies such as not be overlooked. For instance, previous studies of publicly avail-
RNA-​sequencing (Mutz et al., 2013; Woo et al., 2004). Studies have able gene expression data reveal that white blood cell (WBC) tran-
examined the comparability and reproducibility of microarrays and scriptomes can serve to a certain extent as surrogate biopsy material
RNA-​sequencing results (Marioni et al., 2008; Zhao et al., 2014). As to identify biomarkers for other organs (Liew et al., 2006; Mohr and
86

86 Part I:  Basic Concepts


Liew, 2007). The utility of WBC transcriptomes is further augmented currently to allow large-​scale sampling of the microbiome (Flores
by the technical observation that human blood-​derived samples in et  al., 2015). This, together with ever-​ increasing next-​ generation
long-​term storage in biobanks generally have the potential to be ana- sequencing techniques, will allow in the near future the agnostic
lyzed using high-​throughput OMICs technologies; so long as the buffy screening of microbiota in different parts of the body, and the evalua-
coats have been deep-​frozen within a few hours of blood collection, tion of their interaction with environmental stressors and cancer risk.
RNA isolation from frozen buffy coats originally stored without RNA
preservatives can still be accomplished for transcriptomic profiling Adductomics
(Hebels et  al., 2013). As such, there will be many opportunities for DNA and protein adducts have been used for decades as biomark-
conducting studies of the transcriptome and cancer risk in prospective ers of exposure. They are especially useful for measuring exposure
cohort studies. An example of such a study is the study on chronic to electrophiles or chemicals that are metabolically activated to elec-
lymphocytic leukemia (CLL), where prediagnostic transcriptomic pro- trophiles. These electrophilic molecules are potentially toxic because
files were found to predict disease more than 10 years before diagnosis they react with nucleophilic sites in DNA and proteins to produce
(Chadeau-​Hyam et al., 2014). mutations and post-​ translational modifications, respectively, and
In summary, the thoughtful application of transcriptomics can their degradation and partitioning behaviors can contribute to reactive
provide physiologically relevant, molecular insights to complement toxicity (Grigoryan et al., 2016). While previous techniques focused
traditional exposure assessment strategies. Additionally, biomark- on measuring single DNA or protein adducts, current technology is
ers generated by any given technology can be used in a number of starting to allow for the measurement of the adductome, which can be
ways depending on the status (e.g., healthy or diseased) of the source defined as the totality of adducts on a given nucleophilic target. The
tissue. Continued integration of these methodologies into the canon term adductome was first used to describe all of the adducts on DNA
of molecular epidemiology can enable the public health community (Kanaly et al., 2006). However, because of their greater abundance and
to rapidly develop sensitive and specific markers of exposure, and to residence times in human blood, adducts on the circulating proteins
more comprehensively evaluate the health consequences of exposure hemoglobin (Hb) and human serum albumin (HSA) are preferable to
interventions in environmental, occupational, or other targeted settings those of DNA for characterizing adductomes (Rappaport et al., 2012).
among various populations. The nucleophilic hotspot represented by the only free sulfhydryl group
in HSA (HSA-​Cys(34)) offers particular advantages for adductomic
experiments. Although targeted adducts of HSA-​Cys(34) have been
Other OMICs Platforms monitored for decades, an unbiased method has only recently been
reported for visualizing the HSA-​ Cys(34) adductome. Cys34, the
Microbiome only free thiol in HSA, is a nucleophilic hotspot that accounts for
Studying the microbiome has recently gained much attention, in part about 80% of the antioxidant capacity of serum. Different analytical
facilitated by the rapid advancements in sequencing (Zhernakova methods have been proposed to measure the adductome, for example,
et al., 2016). Humans carry in various parts of the body, such as the triple-​
quadrupole mass spectrometry (TQMS) with selected reac-
gastrointestinal tract, lung, mouth, vagina, and skin, multitudes of tion monitoring (SRM) and LC-​high-​resolution-​mass-​spectrometry
microorganisms. It is estimated that the human-​associated microbi- (Grigoryan et al., 2016). Although some proof of principle has been
ota consists of at least 40,000 bacterial strains in 1800 genera, which described by showing interactions between exogenous exposures (i.e.,
collectively harbor at least 9.9  million non-​human genes (Walker, smoking), lifestyle, and anthropometric measures, the value in cancer
2016). It is increasingly being recognized that microbiota have large epidemiology still needs to be established.
influences on biological processes and play a significant role in the
metabolism of exogenous compounds. While culture-​based tech- Proteomics
niques are still relevant, the advent of sequence-​based analytical Proteomics measures all proteins and peptides. As many biologi-
techniques has made it possible to broadly explore interactions of cal functions are transmitted through proteins, proteomics can yield
the microbiome with exposures and diseases; 16S (and 18S) rRNA is valuable insights into disease biology. Proteomic technologies have
the most commonly used universal marker gene method, exploiting been developing from two-​dimensional electrophoreses, enabling the
the fact that there are highly conserved regions of DNA sequence, analyses of a few tenths of proteins to thousands of proteins using
but also regions that are more variable and unique to certain micro- liquid chromatography–​MS (LC–​MS) techniques. LC–​MS is a power-
bial groups or genera (Woese and Fox, 1977). Based on the S rRNA ful technique that it is oriented toward the identification of proteins in
analyses, data can be clustered into operational taxonomic units complex mixtures (Horvatovich et  al., 2010). Several LC-​MS tech-
(OTUs). Next-​generation sequencing allows a deeper investigation niques can be distinguished, varying from digestion of proteins to pep-
in the microbiome by simultaneously sequencing many microbial tides and direct LC-​MS analyses to quantitative proteomics techniques
genomes (Fleischmann et  al., 1995). Next-​generation sequencing using isobaric tags for relative and absolute quantitation (iTRAQ),
also allows measurement of viruses, which are not covered in S stable isotope labeling by amino acids in cell culture (SILAC), and
rRNA sequencing. the use of aptamers in the systematic evolution of ligands by exponen-
Given the influence that the microbiota has on innate immunity, it is tial enrichment (SELEX) (Priyadharshini and Teran, 2016). For the
very plausible that the microbiome elicits an effect on both cancer risk detection of low-​abundant proteins, targeted protein microarrays with
and host response to cancer therapy. An example of microbes influ- well-​characterized molecules with specific activity, such as antibodies,
encing colon cancer risk is Helicobacter hepaticus causing the exces- peptide-​MHC complexes, or lectins, are used as immobilized probes
sive release of nitric oxide from immune cells. (Thaiss et al., 2016). (Jain, 2016).
Ahn et  al. (2013), in a case-​control study, investigated agnostically Several studies have incorporated proteomic approaches to iden-
the association between the gut microbiome and colorectal cancer and tify novel proteins for disease screening, such as for cancers of the
described that cases had overall lower microbial community diversity. bladder (Frantzi et al., 2016), pancreas (Pan et al., 2015), liver (Tsai
In a small pilot study, Hosgood et al. described a potential influence et al., 2015), lung (Shiels et al., 2013), and ovary (Trabert et al., 2014).
of indoor air pollution on the respiratory microbiome and lung cancer However, only few of these have been conducted in prospective stud-
risk (2014). ies. Two prospective studies using targeted immune microarrays have
Although sequencing techniques have improved considerably, most found predictive protein markers for lung and ovarian cancer (Shiels
studies published to date are based on hospital-​based case-​control et al., 2013; Trabert et al., 2014). These studies hold promise for pro-
studies, hampering separation of cause and effect. Many prospective teomics, especially if LC-​MS platforms mature to be able to have
studies to date have not collected biological samples of the gut (i.e., direct quantification and measure low-​abundance proteins, to provide
feces), or respiratory tracts (e.g., buccal, nasal swabs) hampering eti- pictures of the protein networks involved in the pathophysiology of
ological research on the interaction between the microbiome and can- cancer, providing opportunities for early screening and novel treat-
cer. However, easy-​to-​use sampling techniques are being developed ment options.
 87

Application of Biomarkers in Cancer Epidemiology 87


Chromosomal Alterations, Aberrations, comparison of hematological effects of benzene, TCE, and formalde-
and Mutations hyde (Bassig et al., 2016), and comparison of transcriptomic effects of
The assessment of chromosomal alterations in healthy tissue has been exposure to indoor combustion of coal with tobacco smoking (Wang
extensively reviewed (Shuga et  al., 2011). There are many reports et  al., 2015). Furthermore, such studies can evaluate whether there
linking chromosomal aberrations and micronuclei in peripheral blood are early biologic perturbations caused by new exposures or recent
lymphocytes to various exogenous exposures, and some of these changes in lifestyle factors that have not been present long enough to
events have been associated with future risk of cancer (Bonassi et al., have been evaluated for their association with cancer (Rothman et al.,
2008, 2013; Murgia et al., 2008; Shuga et al., 2011). New approaches 1995). For instance, due to the increasing use of nanoparticles in both
to measuring chromosome damage are available, such as array-​based research and manufacturing operations, the assessment of preclinical
cytogenetics and single cell sequencing (Shuga et  al., 2013). For indicators of disease, such as biomarkers of pulmonary inflammation,
example, GWAS data generated from the analysis of peripheral white is important for identifying potential adverse health effects in asymp-
blood cell DNA can be used to detect large structural genetic mosa- tomatic individuals at an early stage (Schulte et al., 2008; Vlaanderen
icism in human autosomes and partial or complete loss of chromo- et al., 2017).
somes X and Y. It has been shown that these events increase with age, Longitudinal designs collect repeated biomarker and exposure infor-
but their association with risk of cancer has not yet been demonstrated mation from the same individuals at different points in time. These
(Machiela et  al., 2015b, 2016b; Zhou et  al., 2016). Another type of studies can be used to assess intra-​individual variation in exposure
chromosomal characteristic that can be measured in peripheral white levels, which are critical to the application and interpretation of expo-
blood cell DNA is chromosomal telomere length. Longer white blood sure biomarkers in prospective cohort studies, and to directly compare
cell telomere length phenotype was associated with increased risk of changes in exposure to changes in biomarkers within the same indi-
several cancers in prospective cohort studies, including non-​Hodgkin viduals. For instance, longitudinal study designs have demonstrated
lymphoma (NHL; Hosnijeh et  al., 2014; Lan et  al., 2009)  and lung that changes in diet are directly related to changes in hemoglobin
cancer (Seow et al., 2014a). Further, a genetic risk score of telomere-​ adducts of acrylamide (Vikstrom et al., 2012). Additionally, longitu-
length-​associated genetic variants obtained from GWAS data was used dinal studies can be used to determine the effectiveness of interven-
to predict longer telomere length and was associated with risk of NHL, tion studies by studying the changes in exposure and/​or intermediate
especially chronic lymphocytic leukemia, and lung cancer in a study of endpoints. For example, changes in mammographic breast density
non-​smoking females in Asia (Machiela et al., 2015a, 2016a). 12–​18 months after starting Tamoxifem therapy have been shown to
be predictive of the response to this drug in preventing breast cancer
(Cuzick et al., 2011).
INCORPORATION OF BIOMARKERS A distinct advantage of the cross-​sectional study is that detailed and
IN EPIDEMIOLOGIC STUDIES accurate information can be collected on current exposure patterns,
potential confounders, and effect modifiers. This can enable the evalu-
ation of the extent to which a wide range of exogenous and endog-
Cross-​Sectional Studies with Biomarker Endpoints
enous exposures leave either short-​term or longer-​term “imprints” on
Cross-​sectional or longitudinal designs on healthy subjects exposed to biomarker endpoints, and the degree of specificity of such associa-
particular exogenous or endogenous agents can be used to investigate tions. As noted previously, the long-​term relationship between tobacco
factors associated with biomarkers, which are treated as the outcome smoking and specific methylation patterns (Joehanes et al., 2016) pro-
variable. These studies focus on exposure and intermediate endpoint vides an example of how an exposure signature may be discerned
biomarkers and often evaluate modifiers of the exposure–​endpoint using an OMIC technology. The ability to identify specific exposure
relationship. Issues in the validation of studies of intermediate end- signatures is likely to aid exposure assessment in future nested case-​
points were discussed previously (Garcia-​Closas et al., 2006). control studies.
Cross-​sectional studies collect biomarker and exposure informa- Selection of an appropriate unexposed group can be challenging in
tion at a single point in time. Cross-​sectional studies are often used to a cross-​sectional study. Apart from coming from the same base popu-
answer questions such as whether a given population has been exposed lation as the exposed group, efficiency is increased by some level of
to a particular compound, the level of exposure, the range of the expo- matching based on age, sex, ethnicity, socioeconomic status, and per-
sure, and the external and internal determinants of the exposure. The haps smoking, as these studies are often relatively small. For biomark-
National Health and Nutrition Examination Survey (NHANES) con- ers with large inter-​individual variation among unexposed individuals,
ducted since 1999 biannually in the United States provides the most an alternative study design in which subjects are used as their own ref-
comprehensive cross-​sectional survey to date in which over 400 infec- erent and followed from the start of the exposure up to a certain length
tious, biochemical, physiological, and environmental variables are of exposure (or vice versa) might be more efficient for biomarker end-
measured in large samples of the general population, allowing detailed points with short half-​lives. An example of such a study is a study
analyses of the exposure and biomarker distribution, its determinants, among farmers exposed to pesticides, with intermediate endpoints that
and interrelations (Patel and Manrai, 2015). were spaced across an entire growing season (Vermeulen et al., 2005).
Cross-​sectional studies can evaluate intermediate biologic effects By having both repeated samples over time among exposed farmers
from a wide range of exposures in the diet and environment, as well and controls, group and individual differences over time can be ana-
as from lifestyle factors such as obesity and reproductive status. For lyzed simultaneously using an instrumental variable approach. Such
instance, cytogenetic markers, including chromosomal aberrations an approach leverages both the inter-​and intra-​individual differences
and micronuclei, have been used as intermediate endpoints in cross-​ in exposures between groups and within individuals overtime (Lerro
sectional studies to demonstrate DNA-​damaging effects derived from et al., 2016).
environmental and dietary exposures (Bonassi et  al., 2005). This The explosion of OMICs technologies is providing unprecedented
design can provide mechanistic insight into well-​established expo- opportunities to comprehensively assess all endogenous and exogenous
sure–​disease relations and to supplement suggestive but inconclusive exposures during an individual’s lifetime, or what has been referred
evidence of the carcinogenicity of an exposure. For example, cross-​ to as the exposome (Rappaport, 2012; Rappaport et  al., 2014; Wild,
sectional molecular epidemiology studies of formaldehyde and tri- 2005, 2012). In this context, cross-​sectional and longitudinal studies
chloroethylene provided insights into the ability of these chemicals are critical to characterize the determinants of these biomarkers in the
to cause hematologic, immunologic, and/​or nephrotoxic alterations population. For instance, studies of the impact of blood collection fac-
(Lan et al., 2010, 2015; Vermeulen et al., 2012; Zhang L et al., 2010). tors (such as fasting time, season, and time of the day) on OMICs mea-
In addition, effects from comparable studies of the same endpoints surements are critical for the design and interpretation of studies and
across multiple types of exposures, including those that are established disease risk (Hebels et al., 2013; Townsend et al., 2016). Longitudinal
carcinogens, can also complement studies of cancer as an endpoint studies of biomarkers such as DNA methylation to evaluate the tempo-
by comparing and contrasting underlying mechanisms of action (e.g., ral variation in repeated specimens from the same individual are also
8

88 Part I:  Basic Concepts


critical, particularly when subsequent studies with disease risk as the involves collecting biologic specimens. Collection of biologic materi-
outcome only have biological specimens collected at a single point in als using non-​invasive approaches (e.g., collecting genomic DNA from
time for each individual (Flanagan et al., 2015). buccal cells) (Hansen et  al., 2007)  might help increase participation
rates. Lack of participation biases the study results when the reason
for non-​participation is directly or indirectly related to the factors
Case-​Control and Case-​Series Studies under study. Because this condition is often difficult to prove, espe-
cially when many factors are under study, high participation rates
The advantages and disadvantages of case-​control and cohort study become the only means to ensure comparability of participants and
designs have been discussed in detail elsewhere (Caporaso et  al., non-​participants. When participation rates are lower than desirable, it
1999; Clayton and McKeigue, 2001; Langholz et al., 1999; Rothman is important to seek some basic risk factor information, and biospeci-
and Greenland, 1998; Wacholder et al., 2002b), and we have previ- mens when possible, from non-​participants to identify differences
ously compared these designs in detail with regard to the collection between participants and non-​participants (Mezei and Kheifets, 2006).
and use of biologic samples for studies of cancer etiology (Garcia-​ In the case–​case, case series, or case-​only design, only subjects
Closas et al., 2006). We summarize several key issues here. Important with the disease of interest and no controls are enrolled in the study.
advantages of case-​control studies compared to prospective cohort This design has been proposed to evaluate etiologic heterogeneity
studies are their ability to enroll large numbers of cancer cases quickly using tumor markers (Begg and Zhang, 1994; Sherman et al., 2010).
and the potential to study uncommon tumors that do not occur in large The degree of etiologic heterogeneity can be quantified by the ratio
enough numbers in cohort studies. Because only one disease is eval- of the relative risk for the association of exposure on marker-​positive
uated, questionnaires can be used for a more detailed and focused tumors to the relative risk for marker-​negative tumors. This parameter
exposure assessment than in cohort studies, which address multiple is equivalent to the odds ratio for the association between exposure
outcomes. and tumor marker in the cases. However, case-​only studies are limited
However, because case-​control studies collect exposure information to an estimation of the ratio of the relative risks and cannot be used
and biologic specimens after diagnosis and sometimes after treatment to obtain estimates of the relative risk for developing different tumor
of the disease, they are vulnerable to differential misclassification and types. It should be noted that the relative risk from a case-​only design
to uncertainties in the temporal relation between the cancer devel- underestimates the relative risk derived in a case-​control design when
opment and the biomarker under study. The influence of the disease the exposure of interest is associated with more than one tumor type
process and treatment on biomarkers of interest is often unknown. in the study.
Although this is not a concern for genetic susceptibility markers that The case-​only study has also been proposed as a valid design to
do not change over time, it can be an important problem for other evaluate multiplicative gene–​ gene (Yang et  al., 1999)  and gene–​
types of biomarkers, including epigenetic, transcriptomic, proteomic, environment (Khoury and Flanders, 1996) interactions. However, this
metabolomic and microbiomic markers (see section on prospective design has important limitations; most notably, it cannot be used to
cohort studies later in this chapter). obtain estimates of risk for disease or additive interactions, is suscep-
The hospital-​based case-​control design has been popular in molecu- tible to misinterpretation of the interaction parameter (Schmidt and
lar epidemiology because it facilitates subject enrollment and intense Schaid, 1999), and is highly dependent on the assumption of inde-
collection and processing of biologic specimens. Personal contact with pendence between the exposure and the genotype under study (Albert
the study participants by doctors, nurses, or interviewers is made easy, et  al., 2001). Because of these limitations, case-​control designs are
which usually results in higher participation rates for interviews and preferable to designs using only cases.
collection of biologic specimens (Morton et al., 2006). In addition, the
easier access to health professionals and laboratories allows collection
of different types and larger quantities of biologic specimens and more
elaborate processing protocols, such as cryopreservation of lympho- Prospective Cohort Studies
cytes. Because study subjects generally are geographically less spread Establishing a cohort study is initially extremely costly and time-​
out than those in population-​based or cohort studies, rapid shipment of consuming; however, in the long run it becomes more cost-​efficient
specimens to central laboratories for more extensive processing proto- because it can study multiple disease endpoints and provides a well-​
cols is facilitated. In addition, because cases are usually diagnosed in defined population that can be easily sampled for efficiency (Potter,
fewer hospitals than population-​based studies, the collection of tumor 1997). Biologic specimens are collected before disease diagnosis and,
samples for molecular pathology studies (Sherman et al., 2010) is also ideally, before the beginning of the disease process. Therefore, it is
facilitated. Furthermore, it is easier to enroll cases before surgery and well suited to study biomarkers that are directly or indirectly affected
establish collaborations with pathology departments at the hospital(s) by the disease process (Hunter, 1997).
for more intense, specialized tissue collections (e.g., freshly fro- As in case-​control studies, the collection of biospecimens to mea-
zen tissue, alcohol-​fixed tissue). Another advantage of the hospital-​ sure biomarkers in cohort studies may adversely affect participation
based design is the easier follow-​up of the patients to determine their rates. Cohort studies can avoid selection biases by collecting ques-
response to treatment and their survival. Follow-​up of cases can be tionnaires and biological samples at baseline or before disease onset,
extremely valuable in molecular epidemiology studies, as many of the provided that specimens remain available for each participant and
biomarkers under study (e.g., tumor characteristics, genetic polymor- follow-​up is complete. However, subjects with biological specimens
phisms) may be especially relevant for clinical outcomes in addition collected after the cohort has been formed might have different charac-
to cancer etiology (Berrington de Gonzalez and Morton, 2012; Elena teristics from the rest of the cohort. Increasingly, concerns over privacy
et al., 2013; Ransohoff, 2013). have also affected the willingness of participants to take part in some
There are established criteria for selecting controls in hospital-​based research studies.
studies to estimate the disease association of a single factor without With adequate infrastructure, cohort studies can be used to collect
bias, but the criteria for assessing associations between more than one prospective serial biological specimens to evaluate how changes in bio-
factor (e.g., gene–​environment interactions) are less clear. Wacholder markers over time relate to disease risk. However, many large cohort
et al. discussed the appropriate exclusion criteria in control selection studies have only been able to collect a single biologic sample at one
for estimating main and subgroup effects as well as additive and mul- point in time. Although this is not a concern for DNA-​based assays of
tiplicative gene–​environment interactions (Wacholder et  al., 2002a). inherited susceptibility markers, it poses some limitations for several
Other study design and analytical considerations in the study of inter- other categories of markers, particularly for exposure biomarkers that
actions include sample size considerations and the impact of measure- have relatively short half-​lives in the presence of exposures that vary
ment error (Garcia-​Closas et al., 2011). substantially from day to day. Notably, advances in OMICs technolo-
One of the main challenges of population-​based case-​control stud- gies are providing unprecedented opportunities to integrate informa-
ies has been obtaining high participation, especially when participation tion from a wide range of endogenous and exogenous exposures,
 89

Application of Biomarkers in Cancer Epidemiology 89


through analyses of the epigenome (Mill and Heijmans, 2013), adduc- establishment of new cohorts and biobanks such as UK Biobank,
tome, transciptome, proteome, metabolome, and microbiome. Because China Kadoorie Biobank, German National Cohort (2014), Cancer
of the changing nature of the exposome, longitudinal cohort studies Prevention Study 3 (Cancer Prevention Study 3), and the National
with repeated biological specimens processed and stored using appro- Institutes of Health’s Precision Medicine Initiative are steps toward
priate protocols are essential to study their relationship with disease these goals.
risk. In addition, tools that allow integration of the large amounts of
data, including pattern recognition and visualization tools, are required
to analyze increasingly complex and large data sets. Careful consider- Incorporation of Biomarker Information
ation of the principles of study design (Spitz and Bondy, 2010) will in Risk-​Prediction Models
still be critical to fulfill the promise of the growing number of X-​wide
Genome-​wide association studies (GWAS) have uncovered hundreds
association studies (Bock, 2012; Chadeau-​Hyam et al., 2010; Foxman
of common single nucleotide polymorphisms (SNPs) related to sus-
and Martin, 2015; Garcia-​Closas et al., 2013; Hanage, 2014; Homuth
ceptibility to common diseases. In addition, targeted, exome, and
et al., 2015; Michels et al., 2013). Finally, observational cohort studies
whole genome DNA sequencing studies, performed in increasingly
with serial collections that also collected clinical data can be highly
large study populations, are extending the search for susceptibility
valuable to identify and validate early detection biomarkers (Schully
markers to uncommon variants. This work is leading to the emergence
et al., 2015).
of gene-​panel and whole-​genome tests to assess polygenic disease
Given that most members of a cohort do not develop cancer, nested
risk, which is likely to increase in the future. Information on genetic
case-​ control and less commonly case-​ cohort studies are used to
and other biomarkers of risk can be integrated with information on
improve efficiency (Wacholder, 1991). With these designs, only sam-
environmental factors in risk prediction models (Garcia-​Closas et al.,
ples from cases and a random subset of non-​cases are analyzed, reduc-
2014). Thus, continuous development and evaluation of risk models
ing the laboratory requirements and cost considerably. The nested
incorporating increasing knowledge on risk factors will be required
case-​control design includes all cases identified in the cohort up to a
(Chatterjee et  al., 2016). Prospective cohort studies play a key role
particular point in time and a random sample of subjects free of dis-
both in the discovery of new biomarkers, as well as in the evaluation
ease at the time of the case diagnosis. On the other hand, a case-​cohort
of the ability of improved models to provide accurate estimates of the
design includes a random sample of the cohort population at the onset
probability of developing disease in the future.
of the study and all cases identified in the cohort up to a particular
point in time. Increasing the case to control ratio to two or three con-
trols per case can easily increase the efficiency of nested case-​control CONCLUSIONS
studies. The case-​cohort design is simpler and allows evaluation of
several disease endpoints; however, because in case-​cohort studies the There has been a confluence of events that, when taken together, will
same disease-​free subjects are repeatedly used as “controls” for dif- provide unprecedented opportunities to extend our knowledge about
ferent disease endpoints, it is difficult to perform assays on matched the environmental and genetic forces that drive cancer risk (Smith
samples from diseased and disease-​free subjects; thus, depletion of et al., 2011).
samples from the disease-​free group may be an issue. These study These include the enhancement of the ability to measure external
designs have been compared with respect to other biomarker-​related exposures through improved questionnaires linked to databases of
issues that arise in study implementation, such as batch effect, length environmental, occupational, nutritional, and other exposures and the
of storage, and freeze-​thaw cycles (Rundle et al., 2005). development of advanced technology that has produced a wide range
The number of subjects enrolled in existing (e.g., http://​epi.grants. of wearable devices capable of measuring a diverse array of exposure
cancer.gov/​Consortia/​cohort.html; UK Biobank:  http://​www.bbmri-​ and lifestyle characteristics such as environmental exposures, physical
lpc-​biobanks.eu/​cohorts.html; China Kadoorie Biobank: https://​www. activity, and sleep quality (DeBord et  al., 2016). Second, as noted,
asiacohort.org), ongoing (Precision Medicine Initiative), and planned there has been an exponential increase in the type and quantity of
prospective cohort studies among adults will most likely approach or molecules that can be measured and identified in biologic samples
exceed 10 million people within the next 5 or so years. These studies that have been collected, processed, and stored appropriately. These
will provide an unprecedented opportunity to apply a wide range of include hypothesis-​driven biomarkers as well as platforms that can
hypothesis-​driven biomarkers and OMIC platforms on large numbers comprehensively analyze entire biologic processes and “compart-
of patients for both common and less common cancers, allow the study ments” agnostically, including the measurement of small molecules
of even relatively uncommon tumors with pooling across cohorts, and (metabolomics), DNA polymorphisms and rarer inherited variants
enable replication and extension of findings. The vast majority of these (genomics), methylation and miRNA (epigenomics), chromosome-​
cohorts are located in North America, Europe, Asia, and Australia, and wide alterations, mRNA (transcriptomics), proteins (proteomics), the
these cohorts provide important opportunities for replication within microbiome, and other biologic components within the human body.
and across these populations. At the same time, there remain impor- These advancements, and their continuing evolution, will enable
tant opportunities to study the etiology of diseases present in South investigators to comprehensively study the biological consequences
America and Africa. of exogenous and endogenous exposures, to detect (1) molecular sig-
Cohort studies often have limited ability to collect tumor samples natures of exposure, (2) early signs of adverse biological effects, and
from large numbers of subjects and to follow up cases for the purpose (3) preclinical disease, and (4) to identify clinical subtypes in cancer.
of carrying out survival studies. New cohort studies based on large Finally, there will soon be 10 million or more adult subjects enrolled in
institutions such as integrated health providers could enable access prospective cohort studies on several continents that have collected and
to tumor samples and easier follow-​up of cases to study treatment stored one or more types of biological samples. These three forces will
response and survival. New prospective cohort studies can take advan- be combined in unprecedented ways to provide profound new insights
tage of increasingly available electronic medical records, technologi- into cancer etiology and at the same time provide myriad opportunities
cal advances in the collection and analysis of biological specimens, for translational research and applications that have great potential to
wearables, and Big Data management platforms and analytics. The reduce the burden of disease through primary prevention, including
collection of biologic samples in newer large prospective cohort stud- public health guidelines, standards, and regulations, as well as second-
ies has stimulated the testing, evolution, and optimization of biologi- ary and tertiary prevention, including the emerging concept of preci-
cal sample collection and storage as well as biorepository technology sion medicine (Collins and Varmus, 2015).
(Hubel et al., 2011; UK Biobank; Vaught, 2016). In addition, improved Some challenges of particular importance include the need for
infrastructure should facilitate repeated biospecimen collections over cohorts to collect repeat biological samples over time; the desirability
time (e.g. blood, urine, buccal and stool samples), and allow special of following up cohort studies of children and adolescents with bio-
collection or processing of biologic samples that might be needed logic samples over their lifetime; and the need to be able to collect,
for the optimum application of new and emerging technologies. The manage, and analyze in meaningful ways the vast amount of data that
90

90 Part I:  Basic Concepts


future studies will generate. Cohorts that are able to collect repeat bio- Bernstein BE, Meissner A, and Lander ES. 2007. The mammalian epigenome.
logical samples, especially blood, as well as pre-​neoplastic tissue sam- Cell, 128(4), 669–​681. PMID: 17320505.
ples when possible, will be especially well positioned to apply new Berrington de Gonzalez A, and Morton LM. 2012. Converting epidemio-
and emerging technologies to study non-​fixed biological endpoints logic studies of cancer etiology to survivorship studies:  approaches
and challenges. Cancer Epidemiol Biomarkers Prev, 21(6), 875–​880.
that show substantial intra-​individual variation over time, reflecting PMID: 22426143.
random fluctuation, secular trends, and early disease processes. The Bhattacharjee A, Richards WG, Staunton J, et al. 2001. Classification of human
ability to derive individual trajectories of the disease processes would lung carcinomas by mRNA expression profiling reveals distinct adeno-
be a major step toward precision medicine that allows targeted pri- carcinoma subclasses. Proc Natl Acad Sci U S A, 98(24), 13790–​13795.
mary and secondary prevention programs. In addition, a comprehen- PMCID: PMC61120.
sive life-​course approach to studying the etiology of cancer and other Bhuvaneshwar K, Belouali A, Singh V, et al. 2016. G-​DOC Plus: an integrative
chronic diseases would add considerably to these efforts, and could be bioinformatics platform for precision medicine. BMC Bioinformatics,
accomplished in part if prospective cohort studies of children and ado- 17(1), 193. PMCID: PMC4851789.
lescents could be followed up into adulthood and beyond. Finally, the Bock C. 2012. Analysing and interpreting DNA methylation data. Nat Rev
Genet, 13(10), 705–​719. PMID: 22986265.
huge data generated from the application of multiple OMICs platforms Bock C, Beerman I, Lien WH, et al. 2012. DNA methylation dynamics during
to each subject are providing an unprecedented challenge to integrate in vivo differentiation of blood and skin stem cells. Mol Cell, 47(4), 633–​
and interpret the resulting data in a meaningful way. There are many 647. PMCID: PMC3428428.
ongoing attempts to come to grips with the tsunami of data we will Bonassi S, Norppa H, Ceppi M, et al. 2008. Chromosomal aberration frequency
be facing over the coming years, and new methods, including more in lymphocytes predicts the risk of cancer: results from a pooled cohort
explicit integration of mechanistic evidence and biology, will no doubt study of 22 358 subjects in 11 countries. Carcinogenesis, 29(6), 1178–​
be developed over time to study the wealth of information these stud- 1183. PMCID: PMC2443275.
ies will generate (Bhuvaneshwar et al., 2016; Kristensen et al., 2014). Bonassi S, Taioli E, and Vermeulen R. 2013. Omics in population studies:  a
molecular epidemiology perspective. Environ Mol Mutagen, 54(7), 455–​
460. PMID: 23908054.
References
Bonassi S, Ugolini D, Kirsch-​Volders M, et al. 2005. Human population studies
Accomando WP, Wiencke JK, Houseman EA, et al. 2012. Decreased NK cells with cytogenetic biomarkers: review of the literature and future prospec-
in patients with head and neck cancer determined in archival DNA. Clin tives. Environ Mol Mutagen, 45(2-​3), 258–​270. PMID: 15688363.
Cancer Res, 18(22), 6147–​6154. PMCID: PMC3500449. Brennan K, and Flanagan JM. 2012. Is there a link between genome-​wide
Ahn J, Sinha R, Pei Z, et  al. 2013. Human gut microbiome and risk for hypomethylation in blood and cancer risk? Cancer Prev Res (Phila),
colorectal cancer. J Natl Cancer Inst, 105(24), 1907–​1911. PMCID: 5(12), 1345–​1357. PMID: 23135621.
PMC3866154. Brennan K, Garcia-​Closas M, Orr N, et al. 2012. Intragenic ATM methylation
Albert PS, Ratnasinghe D, Tangrea J, and Wacholder S. 2001. Limitations of in peripheral blood DNA as a biomarker of breast cancer risk. Cancer
the case-​only design for identifying gene-​environment interactions. Am J Res, 72(9), 2304–​2313. PMID: 22374981.
Epidemiol, 154(8), 687–​693. PMID: 11590080. Cancer Prevention Study 3. Available from:  http://​www.cancer.org/​research/​
Alexandrov LB, Ju YS, Haase K, et al. 2016. Mutational signatures associated researchtopreventcancer/​ c urrentcancerpreventionstudies/​ c ancer-​
with tobacco smoking in human cancer. Science, 354(6312), 618–​622. prevention-​study-​3. Accessed October 11, 2016.
Allegra A, Innao V, Gerace D, Bianco O, and Musolino C. 2016. The metabo- Caporaso N, Rothman N, and Wacholder S. 1999. Case-​control studies of com-
lomic signature of hematologic malignancies. Leuk Res, 49, 22–​ 35. mon alleles and environmental factors. J Natl Cancer Inst Monogr 26,
PMID: 27526405. 25–​30. PMID: 10854482.
Angulo M, Lecuona E, and Sznajder JI. 2012. Role of MicroRNAs in lung Cech TR, and Steitz JA. 2014. The noncoding RNA revolution-​trashing old
disease. Arch Bronconeumol, 48(9), 325–​330. PMCID: PMC3855881. rules to forge new ones. Cell, 157(1), 77–​94. PMID: 24679528.
Argos M, Chen L, Jasmine F, et al. 2015. Gene-​specific differential DNA meth- Chadeau-​Hyam M, Ebbels TM, Brown IJ, et  al. 2010. Metabolic profiling
ylation and chronic arsenic exposure in an epigenome-​wide association and the metabolome-​ wide association study:  significance level for
study of adults in Bangladesh. Environ Health Perspect, 123(1), 64–​71. biomarker identification. J Proteome Res, 9(9), 4620–​4627. PMCID:
PMCID: PMC4286273. PMC2941198.
Armitage EG, and Barbas C. 2014. Metabolomics in cancer biomarker discov- Chadeau-​Hyam M, Vermeulen R, Hebels D, et  al. 2014. Prediagnostic tran-
ery:  current trends and future perspectives. J Pharm Biomed Anal, 87, scriptomic markers of Chronic lymphocytic leukemia reveal perturba-
1–​11. PMID: 24091079. tions 10  years before diagnosis. Ann Oncol, 25(5), 1065–​1072. PMID:
Aryee MJ, Jaffe AE, Corrada-​Bravo H, et  al. 2014. Minfi:  a flexible and 24558024.
comprehensive Bioconductor package for the analysis of Infinium Chan KC, Jiang P, Chan CW, et  al. 2013. Noninvasive detection of cancer-​
DNA methylation microarrays. Bioinformatics, 30(10), 1363–​ 1369. associated genome-​wide hypomethylation and copy number aberrations
PMCID: PMC4016708. by plasma DNA bisulfite sequencing. Proc Natl Acad Sci U S A, 110(47),
Barker DJ, and Clark PM. 1997. Fetal undernutrition and disease in later life. 18761–​18768. PMCID: PMC3839703.
Rev Reprod, 2(2), 105–​112. PMID: 9414472. Chatterjee N, Shi J, and Garcia-​Closas M. 2016. Developing and evaluating
Barone-​Adesi F, Chapman RS, Silverman DT, et al. 2012. Risk of lung cancer polygenic risk prediction models for stratified disease prevention. Nat Rev
associated with domestic use of coal in Xuanwei, China:  retrospective Genet, 17(7), 392–​406. PMID: 27140283.
cohort study. BMJ, 345, e5414. PMCID: PMC3431444. China Kadoorie Biobank. Available from:  http://​www.ckbiobank.org/​site/​.
Bassig BA, Zhang L, Vermeulen R, et al. 2016. Comparison of hematological Accessed October 11, 2016.
alterations and markers of B-​cell activation in workers exposed to ben- Christensen BC, Houseman EA, Marsit CJ, et al. 2009. Aging and environmen-
zene, formaldehyde and trichloroethylene. Carcinogenesis, 37(7), 692–​ tal exposures alter tissue-​specific DNA methylation dependent upon CpG
700. PMCID: PMC4936387. island context. PLoS Genet, 5(8), e1000602. PMCID: PMC2718614.
Bauer M, Linsel G, Fink B, et  al. 2015. A varying T cell subtype explains Christensen BC, Smith AA, Zheng S, et al. 2011. DNA methylation, isocitrate
apparent tobacco smoking induced single CpG hypomethylation in whole dehydrogenase mutation, and survival in glioma. J Natl Cancer Inst,
blood. Clin Epigenetics, 7(1), 81. PMCID: PMC4526203. 103(2), 143–​153. PMCID: PMC3022619.
Beane J, Vick J, Schembri F, et al. 2011. Characterizing the impact of smok- Clayton D, and McKeigue PM. 2001. Epidemiological methods for studying
ing and lung cancer on the airway transcriptome using RNA-​Seq. Cancer genes and environmental factors in complex diseases. Lancet, 358(9290),
Prev Res (Phila), 4(6), 803–​817. PMCID: PMC3694393. 1356–​1360. PMID: 11684236.
Begg CB, and Zhang ZF. 1994. Statistical analysis of molecular epidemiology Colella S, Shen L, Baggerly KA, Issa JPJ, and Krahe R. 2003. Sensitive and
studies employing case-​series. Cancer Epidemiol Biomarkers Prev, 3(2), quantitative universal pyrosequencing (TM) methylation analysis of CpG
173–​175. PMID: 8049640. sites. Biotechniques, 35(1), 146–​. PMID: WOS:000184021300015.
Belinsky SA, Grimes MJ, Casas E, et al. 2007. Predicting gene promoter meth- Collins FS, and Varmus H. 2015. A new initiative on precision medicine. N
ylation in non-​small-​cell lung cancer by evaluating sputum and serum. Br Engl J Med, 372(9), 793–​795. PMID: 25635347.
J Cancer, 96(8), 1278–​1283. PMCID: PMC2360148. Committee on Biological Markers of the National Research Council. 1987.
Belinsky SA, Klinge DM, Dekker JD, et al. 2005. Gene promoter methylation Biological markers in environmental health research. Committee on
in plasma and sputum increases with lung cancer risk. Clin Cancer Res, Biological Markers of the National Research Council. Environ Health
11(18), 6505–​6511. PMID: 16166426. Perspect, 74, 3–​9. PMCID: PMC1474499.
 91

Application of Biomarkers in Cancer Epidemiology 91


Cordero F, Ferrero G, Polidoro S, et  al. 2015. Differentially methylated Cancer Epidemiology and Prevention (3rd ed., pp. 70–​88). New  York:
microRNAs in prediagnostic samples of subjects who developed breast Oxford University Press.
cancer in the European Prospective Investigation into Nutrition and German National Cohort (GNC) Consortium. 2014. The German National
Cancer (EPIC-​Italy) cohort. Carcinogenesis, 36(10), 1144–​1153. PMID: Cohort:  aims, study design and organization. Eur J Epidemiol, 29(5),
26168820. 371–​382. PMCID: PMC4050302.
Coughlin SS. 2014. Toward a road map for global -​omics: a primer on -​omic Gifford CA, Ziller MJ, Gu H, et  al. 2013. Transcriptional and epigenetic
technologies. Am J Epidemiol, 180(12), 1188–​1195. PMID: 25409464. dynamics during specification of human embryonic stem cells. Cell,
Cuzick J, Warwick J, Pinney E, et  al. 2011. Tamoxifen-​induced reduction in 153(5), 1149–​1163. PMCID: PMC3709577.
mammographic density and breast cancer risk reduction: a nested case-​ Golub TR, Slonim DK, Tamayo P, et al. 1999. Molecular classification of can-
control study. J Natl Cancer Inst, 103(9), 744–​752. PMID: 21483019. cer: class discovery and class prediction by gene expression monitoring.
DeBord DG, Carreon T, Lentz TJ, et  al. 2016. Use of the “exposome” in Science, 286(5439), 531–​537. PMID: 10521349.
the practice of epidemiology:  a primer on -​omic technologies. Am J Grigoryan H, Edmands W, Lu SS, et al. 2016. Adductomics pipeline for untar-
Epidemiol, 184(4), 302–​314. PMCID: PMC5025320. geted analysis of modifications to Cys34 of human serum albumin. Anal
Devaraj S, and Natarajan J. 2011. miRNA-​mRNA network detects hub mRNAs Chem, 88(21), 10504–10512. Epub 2016 Oct 10. PMID: 27684351.
and cancer specific miRNAs in lung cancer. In Silico Biol, 11(5-​6), 281–​ Grimes DA, and Schulz KF. 2002. Bias and causal associations in observational
295. PMID: 23202429. research. Lancet, 359(9302), 248–​252. PMID: 11812579.
Djebali S, Davis CA, Merkel A, et  al. 2012. Landscape of transcription in Guida F, Sandanger T, Castagné R, et al. 2015. Dynamics of smoking-​induced
human cells. Nature, 489(7414), 101–​108. PMCID: PMC3684276. genome-​wide methylation changes with time since smoking cessation.
Downward GS, Hu W, Large D, et al. 2014. Heterogeneity in coal composition Hum Mol Genet, 24(8), 2349–​2359. PMID: 25556184.
and implications for lung cancer risk in Xuanwei and Fuyuan counties, Hanage WP. 2014. Microbiology: microbiome science needs a healthy dose of
China. Environ Int, 68, 94–​104. PMID: 24721117. scepticism. Nature, 512(7514), 247–​248. PMID: 25143098.
Elena JW, Travis LB, Simonds NI, et  al. 2013. Leveraging epidemiology Hansen KD, Timp W, Bravo HC, et al. 2011. Increased methylation variation
and clinical studies of cancer outcomes:  recommendations and oppor- in epigenetic domains across cancer types. Nat Genet, 43(8), 768–​775.
tunities for translational research. J Natl Cancer Inst, 105(2), 85–​94. PMCID: PMC3145050.
PMCID: PMC3545903. Hansen TV, Simonsen MK, Nielsen FC, and Hundrup YA. 2007. Collection
Ernst J, Kheradpour P, Mikkelsen TS, et  al. 2011. Mapping and analysis of of blood, saliva, and buccal cell samples in a pilot study on the Danish
chromatin state dynamics in nine human cell types. Nature, 473(7345), nurse cohort:  comparison of the response rate and quality of genomic
43–​49. PMCID: PMC3088773. DNA. Cancer Epidemiol Biomarkers Prev, 16(10), 2072–​2076. PMID:
Esteller M. 2011. Non-​coding RNAs in human disease. Nat Rev Genet, 12(12), 17932355.
861–​874. PMID: 22094949. Harvey BG, Heguy A, Leopold PL, et al. 2007. Modification of gene expression
Evans WE, and Guy RK. 2004. Gene expression as a drug discovery tool. Nat of the small airway epithelium in response to cigarette smoking. J Mol
Genet, 36(3), 214–​215. PMID: 14988717. Med (Berl), 85(1), 39–​53. PMID: 17115125.
Fazzari MJ, and Greally JM. 2004. Epigenomics: beyond CpG islands. Nat Rev He L, and Hannon GJ. 2004. MicroRNAs: small RNAs with a big role in gene
Genet, 5(6), 446–​455. PMID: 15153997. regulation. Nat Rev Genet, 5(7), 522–​531. PMID: 15211354.
Feinberg AP, and Fallin MD. 2015. Epigenetics at the Crossroads of Hebels D, Georgiadis P, Keun H, et al. 2013. Performance in omics analyses
Genes and the Environment. JAMA, 314(11), 1129–​ 1130. PMID: of blood samples in long-​term storage: opportunities for the exploitation
WOS:000361284500019. of existing biobanks in environmental health research. Environ Health
Flanagan JM, Brook MN, Orr N, et  al. 2015. Temporal stability and deter- Perspect, 121(4), 480–​487.
minants of white blood cell DNA methylation in the breakthrough gen- Heijmans BT, Tobi EW, Stein AD, et al. 2008. Persistent epigenetic differences
erations study. Cancer Epidemiol Biomarkers Prev, 24(1), 221–​ 229. associated with prenatal exposure to famine in humans. Proc Natl Acad
PMID: 25371448. Sci U S A, 105(44), 17046–​17049. PMCID: PMC2579375.
Flavell SW, and Greenberg ME. 2008. Signaling mechanisms linking neuronal Herceg Z, Lambert MP, van Veldhoven K, et al. 2013. Towards incorporating
activity to gene expression and plasticity of the nervous system. Annu Rev epigenetic mechanisms into carcinogen identification and evaluation.
Neurosci, 31, 563–​590. PMCID: PMC2728073. Carcinogenesis, 34(9), 1955–​1967. PMCID: PMC3765050.
Fleischmann RD, Adams MD, White O, et  al. 1995. Whole-​genome ran- Herman JG, Graff JR, Myohanen S, Nelkin BD, and Baylin SB. 1996.
dom sequencing and assembly of Haemophilus influenzae Rd. Science, Methylation-​ specific PCR:  a novel PCR assay for methylation sta-
269(5223), 496–​512. PMID: 7542800. tus of CpG islands. Proc Natl Acad Sci U S A, 93(18), 9821–​9826.
Flores R, Shi J, Yu G, et  al. 2015. Collection media and delayed freezing PMID: WOS:A1996VF61400091.
effects on microbial composition of human stool. Microbiome, 3, 33. Homuth G, Wahl S, Muller C, et al. 2015. Extensive alterations of the whole-​
PMCID: PMC4534027. blood transcriptome are associated with body mass index: results of an
Flowers E, Won GY, and Fukuoka Y. 2015. MicroRNAs associated with mRNA profiling study involving two large population-​based cohorts.
exercise and diet:  a systematic review. Physiol Genomics, 47(1), 1–​11. BMC Med Genomics, 8, 65. PMCID: PMC4608219.
PMID: 25465031. Horvath S. 2013. DNA methylation age of human tissues and cell types.
Foxman B, and Martin ET. 2015. Use of the microbiome in the practice of Genome Biol, 14(10), R115. PMCID: PMC4015143.
epidemiology: a primer on -​omic technologies. Am J Epidemiol, 182(1), Horvatovich P, Hoekman B, Govorukhina N, and Bischoff R. 2010.
1–​8. PMCID: PMC4498138. Multidimensional chromatography coupled to mass spectrometry in
Frantzi M, van Kessel KE, Zwarthoff EC, et al. 2016. Development and vali- analysing complex proteomics samples. J Sep Sci, 33(10), 1421–​1437.
dation of urine-​based peptide biomarker panels for detecting bladder PMID: 20486207.
cancer in a multi-​center study. Clin Cancer Res, 22(16), 4077–​4086. Hosgood Hr, Sapkota A, Rothman N, et  al. 2014. The potential role of lung
PMID: 27026199. microbiota in lung cancer attributed to household coal burning exposures.
Frediani JK, Jones DP, Tukvadze N, et  al. 2014. Plasma metabolomics in Environ Mol Mutagen, 55(8), 643–651. doi: 10.1002/em.21878. Epub
human pulmonary tuberculosis disease: a pilot study. PLoS One, 9(10), 2014 Jun 3. PMID: 24895247.
e108854. PMCID: PMC4198093. Hosnijeh FS, Matullo G, Russo A, et al. 2014. Prediagnostic telomere length
Garber M, Grabherr MG, Guttman M, and Trapnell C. 2011. Computational and risk of B-​cell lymphoma-​Results from the EPIC cohort study. Int J
methods for transcriptome annotation and quantification using RNA-​seq. Cancer, 135(12), 2910–​2917. PMID: 24771230.
Nat Methods, 8(6), 469–​477. PMID: 21623353. Houseman EA, Accomando WP, Koestler DC, et  al. 2012. DNA meth-
Garcia-​Closas M, Gail MH, Kelsey KT, and Ziegler RG. 2013. Searching for ylation arrays as surrogate measures of cell mixture distribution. BMC
blood DNA methylation markers of breast cancer risk and early detection. Bioinformatics, 13, 86. PMCID: PMC3532182.
J Natl Cancer Inst, 105(10), 678–​680. PMCID: PMC3653825. Houseman EA, Kim S, Kelsey KT, and Wiencke JK. 2015. DNA methylation in
Garcia-​Closas M, Gunsoy NB, and Chatterjee N. 2014. Combined associations whole blood: uses and challenges. Curr Environ Health Rep, 2(2), 145–​
of genetic and environmental risk factors: implications for prevention of 154. PMID: 26231364.
breast cancer. J Natl Cancer Inst, 106(11). PMCID: PMC4271030. Hsiung DT, Marsit CJ, Houseman EA, et al. 2007. Global DNA methylation level
Garcia-​Closas M, Jacobs K, Kraft P, and Chatterjee N. 2011. Analysis of epi- in whole blood as a biomarker in head and neck squamous cell carcinoma.
demiologic studies of genetic effects and gene-​environment interactions. Cancer Epidemiol Biomarkers Prev, 16(1), 108–​114. PMID: 17220338.
IARC Sci Publ 163, 281–​301. PMID: 22997868. Huang da W, Sherman BT, and Lempicki RA. 2009. Bioinformatics enrichment
Garcia-​Closas M, Vermeulen R, Sherman ME, et al. 2006. Application of bio- tools: paths toward the comprehensive functional analysis of large gene
markers in epidemiology. In:  Schottenfeld D, and Fraumeni JF (Eds.), lists. Nucleic Acids Res, 37(1), 1–​13. PMCID: PMC2615629.
92

92 Part I:  Basic Concepts


Hubel A, Aksan A, Skubitz AP, Wendt C, and Zhong X. 2011. State of the art in soluble CD27 and CD30 markers. Carcinogenesis, 31(9), 1592–​1596.
preservation of fluid biospecimens. Biopreserv Biobank, 9(3), 237–​244. PMCID: PMC2930801.
PMID: 24850337. Landau DA, Clement K, Ziller MJ, et al. 2014. Locally disordered methylation
Hunter DJ. 1997. Methodological issues in the use of biological markers forms the basis of intratumor methylome variation in chronic lympho-
in cancer epidemiology:  cohort studies. IARC Sci Publ 142, 39–​ 46. cytic leukemia. Cancer Cell, 26(6), 813–​825. PMCID: PMC4302418.
PMID: 9354910. Langevin SM, Houseman EA, Accomando WP, et al. 2014. Leukocyte-​adjusted
International Agency for Research on Cancer (IARC). 2012. In Rothman N, epigenome-​wide association studies of blood from solid tumor patients.
Hainaut P, Schulte P, Smith M, Boffetta P, Perera F (Eds.), IARC Scientific Epigenetics, 9(6), 884–​895. PMCID: PMC4065186.
Publication, No 163. Geneva, Switzerland: WHO Press. Langevin SM, Houseman EA, Christensen BC, et  al. 2011. The influence
Idaghdour Y, and Awadalla P. 2013. Exploiting gene expression variation to of aging, environmental exposures and local sequence features on the
capture gene-​environment interactions for disease. Front Genet, 3, 228. variation of DNA methylation in blood. Epigenetics, 6(7), 908–​ 919.
PMCID: PMC3668192. PMCID: PMC3154431.
Iyer MK, Niknafs YS, Malik R, et  al. 2015. The landscape of long noncod- Langevin SM, Kratzke RA, and Kelsey KT. 2015. Epigenetics of lung cancer.
ing RNAs in the human transcriptome. Nat Genet, 47(3), 199–​ 208. Transl Res, 165(1), 74–​90. PMCID: PMC4162853.
PMCID: PMC4417758. Langholz B, Rothman N, Wacholder S, and Thomas DC. 1999. Cohort studies
Jaffe AE, and Irizarry RA. 2014. Accounting for cellular heterogeneity is criti- for characterizing measured genes. J Natl Cancer Inst Monogr 26, 39–​42.
cal in epigenome-​wide association studies. Genome Biol, 15(2), R31. PMID: 10854484.
PMCID: PMC4053810. Lee KW, Richmond R, Hu P, et al. 2015. Prenatal exposure to maternal ciga-
Jain KK. 2016. Role of proteomics in the development of personalized rette smoking and DNA methylation:  epigenome-​wide association in a
medicine. Adv Protein Chem Struct Biol, 102(doi), 41–​52. PMID: discovery sample of adolescents and replication in an independent cohort
26827601. at birth through 17 years of age. Environ Health Perspect, 123(2), 193–​
Jirtle RL, and Skinner MK. 2007. Environmental epigenomics and disease sus- 199. PMCID: PMC4314251.
ceptibility. Nat Rev Genet, 8(4), 253–​262. PMID: 17363974. Leng S, Do K, Yingling CM, et al. 2012. Defining a gene promoter methylation
Joehanes R, Just AC, Marioni RE, et al. 2016. Epigenetic signatures of ciga- signature in sputum for lung cancer risk assessment. Clin Cancer Res,
rette smoking. Circ Cardiovasc Genet, 9(5), 436–447. Epub 2016 Sep 20. 18(12), 3387–​3395. PMCID: PMC3483793.
PMID: 27651444. Leng S, Liu Y, Weissfeld JL, et  al. 2015. 15q12 variants, sputum gene pro-
Jones DP. 2016. Sequencing the exposome:  a call to action. Toxicol Rep, 3, moter hypermethylation, and lung cancer risk:  a GWAS in smokers. J
29–​45. PMCID: PMC4692045. Natl Cancer Inst, 107(5). PMCID: PMC4555640.
Jones PA, and Baylin SB. 2007. The epigenomics of cancer. Cell, 128(4), 683–​ Lerro C, Beane Freeman LE, Portengen L, et al. 2016. A longitudinal study of
692. PMCID: PMC3894624. atrazine and 2,4-​D exposure and oxidative stress markers among Iowa
Joubert BR, Haberg SE, Bell DA, et  al. 2014. Maternal smoking and DNA corn farmers.
methylation in newborns:  in utero effect or epigenetic inheritance? Li S, Park Y, Duraisingham S, et  al. 2013. Predicting network activity from
Cancer Epidemiol Biomarkers Prev, 23(6), 1007–​ 1017. PMCID: high throughput metabolomics. PLoS Comput Biol, 9(7), e1003123.
PMC4140220. PMCID: PMC3701697.
Joubert BR, Haberg SE, Nilsen RM, et al. 2012. 450K epigenome-​wide scan Liew CC, Ma J, Tang HC, Zheng R, and Dempsey AA. 2006. The periph-
identifies differential DNA methylation in newborns related to maternal eral blood transcriptome dynamically reflects system wide biol-
smoking during pregnancy. Environ Health Perspect, 120(10), 1425–​ ogy:  a potential diagnostic tool. J Lab Clin Med, 147(3), 126–​132.
1431. PMCID: PMC3491949. PMID: 16503242.
Kanaly RA, Hanaoka T, Sugimura H, et al. 2006. Development of the adduc- Liu Y, Aryee MJ, Padyukov L, et al. 2013. Epigenome-​wide association data
tome approach to detect DNA damage in humans. Antioxid Redox Signal, implicate DNA methylation as an intermediary of genetic risk in rheu-
8(5–​6), 993–​1001. PMID: 16771689. matoid arthritis. Nat Biotechnol, 31(2), 142–​147. PMCID: PMC3598632.
Khoury MJ, and Flanders WD. 1996. Nontraditional epidemiologic approaches Ma Y, Zhang H, Liu F, et al. 2015. Highly sensitive detection of DNA meth-
in the analysis of gene-​environment interaction: case-​control studies with ylation levels by using a quantum dot-​based FRET method. Nanoscale,
no controls! Am J Epidemiol, 144(3), 207–​213. PMID: 8686689. 7(41), 17547–​17555. PMID: 26446775.
Kile ML, Houseman EA, Baccarelli AA, et al. 2014. Effect of prenatal arse- Maccani JZ, Koestler DC, Lester B, et  al. 2015. Placental DNA meth-
nic exposure on DNA methylation and leukocyte subpopulations in cord ylation related to both infant toenail mercury and adverse neurobe-
blood. Epigenetics, 9(5), 774–​782. PMCID: PMC4063836. havioral outcomes. Environ Health Perspect, 123(7), 723–​ 729.
Koestler DC, Avissar-​Whiting M, Houseman EA, Karagas MR, and Marsit CJ. PMCID: PMC4492267.
2013. Differential DNA methylation in umbilical cord blood of infants Machiela MJ, Hsiung CA, Shu XO, et  al. 2015a. Genetic variants associ-
exposed to low levels of arsenic in utero. Environ Health Perspect, 121(8), ated with longer telomere length are associated with increased lung
971–​977. PMCID: PMC3733676. cancer risk among never-​smoking women in Asia:  a report from the
Kristensen VN, Lingjaerde OC, Russnes HG, et al. 2014. Principles and meth- female lung cancer consortium in Asia. Int J Cancer, 137(2), 311–​319.
ods of integrative genomic analyses in cancer. Nat Rev Cancer, 14(5), PMCID: PMC4733320.
299–​313. PMID: 24759209. Machiela MJ, Lan Q, Slager SL, et al. 2016a. Genetically predicted longer telo-
Kundaje A, Meuleman W, Ernst J, et  al. 2015. Integrative analysis of mere length is associated with increased risk of B-​cell lymphoma sub-
111 reference human epigenomes. Nature, 518(7539), 317–​ 330. types. Hum Mol Genet, 25(8), 1663–​1676. PMCID: PMC4854019.
PMCID: PMC4530010. Machiela MJ, Zhou W, Karlins E, et al. 2016b. Female chromosome X mosa-
Kupers LK, Xu X, Jankipersadsing SA, et al. 2015. DNA methylation medi- icism is age-​related and preferentially affects the inactivated X chromo-
ates the effect of maternal smoking during pregnancy on birthweight of some. Nat Commun, 7, 11843. PMCID: PMC4909985.
the offspring. Int J Epidemiol, 44(4), 1224–​1237. PMCID: PMC4588868. Machiela MJ, Zhou W, Sampson JN, et  al. 2015b. Characterization of large
Laine JE, Bailey KA, Rubio-​Andrade M, et al. 2015. Maternal arsenic expo- structural genetic mosaicism in human autosomes. Am J Hum Genet,
sure, arsenic methylation efficiency, and birth outcomes in the Biomarkers 96(3), 487–​497. PMCID: PMC4375431.
of Exposure to ARsenic (BEAR) pregnancy cohort in Mexico. Environ Marioni JC, Mason CE, Mane SM, Stephens M, and Gilad Y. 2008. RNA-​seq: an
Health Perspect, 123(2), 186–​192. PMCID: PMC4314242. assessment of technical reproducibility and comparison with gene expres-
Lan Q, Cawthon R, Shen M, et  al. 2009. A prospective study of telomere sion arrays. Genome Res, 18(9), 1509–​1517. PMCID: PMC2527709.
length measured by monochrome multiplex quantitative PCR and risk Marioni RE, Shah S, McRae AF, et  al. 2015. DNA methylation age of
of non-​ Hodgkin lymphoma. Clin Cancer Res, 15(23), 7429–​ 7433. blood predicts all-​cause mortality in later life. Genome Biol, 16, 25.
PMCID: PMC2787641. PMCID: PMC4350614.
Lan Q, Shen M, Berndt SI, et  al. 2005. Smoky coal exposure, NBS1 poly- Marsit CJ. 2015. Influence of environmental exposure on human epigenetic
morphisms, p53 protein accumulation, and lung cancer risk in Xuan Wei, regulation. J Exp Biol, 218(Pt 1), 71–​79. PMCID: PMC4286705.
China. Lung Cancer, 49(3), 317–​323. PMID: 15921821. McDonald OG, Wu H, Timp W, Doi A, and Feinberg AP. 2011. Genome-​scale
Lan Q, Smith MT, Tang X, et al. 2015. Chromosome-​wide aneuploidy study epigenetic reprogramming during epithelial-​to-​mesenchymal transition.
of cultured circulating myeloid progenitor cells from workers occu- Nat Struct Mol Biol, 18(8), 867–​874. PMCID: PMC3150339.
pationally exposed to formaldehyde. Carcinogenesis, 36(1), 160–​167. Meseure D, Drak Alsibai K, Nicolas A, Bieche I, and Morillon A. 2015. Long
PMCID: PMC4291049. noncoding RNAs as new architects in cancer epigenetics, prognostic bio-
Lan Q, Zhang L, Tang X, et  al. 2010. Occupational exposure to trichlo- markers, and potential therapeutic targets. Biomed Res Int, 2015, 320214.
roethylene is associated with a decline in lymphocyte subsets and PMCID: PMC4584070.
 93

Application of Biomarkers in Cancer Epidemiology 93


Mezei G, and Kheifets L. 2006. Selection bias and its implications for case-​ Rando OJ, and Verstrepen KJ. 2007. Timescales of genetic and epigenetic
control studies:  a case study of magnetic field exposure and childhood inheritance. Cell, 128(4), 655–​668. PMID: 17320504.
leukaemia. Int J Epidemiol, 35(2), 397–​406. PMID: 16303812. Ransohoff DF. 2013. Cultivating cohort studies for observational transla-
Michels KB, Binder AM, Dedeurwaerder S, et  al. 2013. Recommendations tional research. Cancer Epidemiol Biomarkers Prev, 22(4), 481–​484.
for the design and analysis of epigenome-​wide association studies. Nat PMCID: PMC4378958.
Methods, 10(10), 949–​955. PMID: 24076989. Rappaport SM. 2012. Biomarkers intersect with the exposome. Biomarkers,
Mill J, and Heijmans BT. 2013. From promises to practical strategies in epi- 17(6), 483–​489. PMCID: PMC4763608.
genetic epidemiology. Nat Rev Genet, 14(8), 585–​594. PMID: 23817309. Rappaport SM, Barupal DK, Wishart D, Vineis P, and Scalbert A. 2014. The
Miller GW, and Jones DP. 2014. The nature of nurture: refining the definition of blood exposome and its role in discovering causes of disease. Environ
the exposome. Toxicol Sci, 137(1), 1–​2. PMCID: PMC3871934. Health Perspect, 122(8), 769–​774. PMCID: PMC4123034.
Mohr S, and Liew CC. 2007. The peripheral-​blood transcriptome: new insights Rappaport SM, Li H, Grigoryan H, Funk WE, and Williams ER. 2012.
into disease and risk assessment. Trends Mol Med, 13(10), 422–​432. Adductomics: characterizing exposures to reactive electrophiles. Toxicol
PMID: 17919976. Lett, 213(1), 83–​90. PMCID: PMC4758449.
Moore LE, Pfeiffer RM, Poscablo C, et  al. 2008. Genomic DNA hypometh- Reynolds LM, Wan M, Ding J, et  al. 2015. DNA methylation of the aryl
ylation as a biomarker for bladder cancer susceptibility in the Spanish hydrocarbon receptor repressor associations with cigarette smoking
Bladder Cancer Study: a case-​control study. Lancet Oncology, 9(4), 359–​ and subclinical atherosclerosis. Circ Cardiovasc Genet, 8(5), 707–​716.
366. PMID: WOS:000254689000023. PMID: 26307030.
Morris TJ, and Beck S. 2015. Analysis pipelines and packages for Infinium Richmond RC, Simpkin AJ, Woodward G, et  al. 2015. Prenatal exposure
HumanMethylation450 BeadChip (450k) data. Methods, 72, 3–​ 8. to maternal smoking and offspring DNA methylation across the life-
PMCID: PMC4304832. course: findings from the Avon Longitudinal Study of Parents and Children
Morris TJ, Butcher LM, Feber A, et  al. 2014. ChAMP:  450k Chip (ALSPAC). Hum Mol Genet, 24(8), 2201–​2217. PMCID: PMC4380069.
Analysis Methylation Pipeline. Bioinformatics, 30(3), 428–​ 430. Rogue A, Lambert C, Spire C, Claude N, and Guillouzo A. 2012. Interindividual
PMCID: PMC3904520. variability in gene expression profiles in human hepatocytes and com-
Morton LM, Cahill J, and Hartge P. 2006. Reporting participation in epide- parison with HepaRG cells. Drug Metab Dispos, 40(1), 151–​ 158.
miologic studies: a survey of practice. Am J Epidemiol, 163(3), 197–​203. PMID: 21994436.
PMID: 16339049. Rojas D, Rager JE, Smeester L, et  al. 2015. Prenatal arsenic exposure
Mumford JL, He XZ, Chapman RS, et al. 1987. Lung cancer and indoor air pollu- and the epigenome:  identifying sites of 5-​ methylcytosine alterations
tion in Xuan Wei, China. Science, 235(4785), 217–​220. PMID: 3798109. that predict functional changes in gene expression in newborn cord
Murgia E, Ballardin M, Bonassi S, Rossi AM, and Barale R. 2008. Validation blood and subsequent birth outcomes. Toxicol Sci, 143(1), 97–​ 106.
of micronuclei frequency in peripheral blood lymphocytes as early cancer PMCID: PMC4274382.
risk biomarker in a nested case-​control study. Mutat Res, 639(1–​2), 27–​ Rothman KJ, and Greenland S. 1998. Modern Epidemiology.
34. PMID: 18155071. Philadelphia: Lippincott-​Raven.
Muti P, Sacconi A, Hossain A, et al. 2014. Downregulation of microRNAs 145-​ Rothman N, Stewart WF, and Schulte PA. 1995. Incorporating biomarkers into
3p and 145-​5p is a long-​term predictor of postmenopausal breast cancer cancer epidemiology: a matrix of biomarker and study design categories.
risk: the ORDET prospective study. Cancer Epidemiol Biomarkers Prev, Cancer Epidemiol Biomarkers Prev, 4(4), 301–​311. PMID: 7655323.
23(11), 2471–​2481. PMID: 25073626. Ruiz-​Hernandez A, Kuo CC, Rentero-​Garrido P, et  al. 2015. Environmental
Mutz KO, Heilkenbrinker A, Lonne M, Walter JG, and Stahl F. 2013. chemicals and DNA methylation in adults:  a systematic review of the
Transcriptome analysis using next-​ generation sequencing. Curr Opin epidemiologic evidence. Clin Epigenetics, 7, 55. PMCID: PMC4433069.
Biotechnol, 24(1), 22–​30. PMID: 23020966. Rundle AG, Vineis P, and Ahsan H. 2005. Design options for molecular epide-
Nelson HH, Marsit CJ, and Kelsey KT. 2011. Global methylation in exposure miology research within cohort studies. Cancer Epidemiol Biomarkers
biology and translational medical science. Environ Health Perspect, Prev, 14(8), 1899–​1907. PMID: 16103435.
119(11), 1528–​1533. PMCID: PMC3226501. Schembri F, Sridhar S, Perdomo C, et al. 2009. MicroRNAs as modulators of
Olsen KS, Skeie G, and Lund E. 2015. Whole-​blood gene expression profiles smoking-​induced gene expression changes in human airway epithelium.
in large-​scale epidemiological studies: what do they tell? Curr Nutr Rep, Proc Natl Acad Sci U S A, 106(7), 2319–​2324. PMCID: PMC2650144.
4(4), 377–​386. PMCID: PMC4639574. Schmidl C, Klug M, Boeld TJ, et  al. 2009. Lineage-​specific DNA methyla-
Osborn MP, Park Y, Parks MB, et  al. 2013. Metabolome-​wide association tion in T cells correlates with histone methylation and enhancer activity.
study of neovascular age-​related macular degeneration. PLoS One, 8(8), Genome Res, 19(7), 1165–​1174. PMCID: PMC2704427.
e72737. PMCID: PMC3754980. Schmidt S, and Schaid DJ. 1999. Potential misinterpretation of the case-​only
Palmisano WA, Divine KK, Saccomanno G, et al. 2000. Predicting lung can- study to assess gene-​environment interaction. Am J Epidemiol, 150(8),
cer by detecting aberrant promoter methylation in sputum. Cancer Res, 878–​885. PMID: 10522659.
60(21), 5954–​5958. PMID: 11085511. Schubeler D. 2015. Function and information content of DNA methylation.
Pan S, Brentnall TA, and Chen R. 2015. Proteomics analysis of bodily Nature, 517(7534), 321–​326. PMID: 25592537.
fluids in pancreatic cancer. Proteomics, 15(15), 2705–​ 2715. Schully SD, Carrick DM, Mechanic LE, et al. 2015. Leveraging biospecimen
PMCID: PMC4526406. resources for discovery or validation of markers for early cancer detec-
Patel CJ, and Manrai AK. 2015. Development of exposome correlation globes tion. J Natl Cancer Inst, 107(4). PMCID: PMC4342676.
to map out environment-​wide associations. Pac Symp Biocomput, 231–​ Schulte P, Geraci C, Zumwalde R, Hoover M, and Kuempel E. 2008.
242. PMCID: PMC4299925. Occupational risk management of engineered nanoparticles. J Occup
Perdomo C, Spira A, and Schembri F. 2011. MiRNAs as regulators of the Environ Hyg, 5(4), 239–​249. PMID: 18260001.
response to inhaled environmental toxins and airway carcinogenesis. Seow WJ, Cawthon RM, Purdue MP, et  al. 2014a. Telomere length in white
Mutat Res, 717(1-​2), 32–​37. PMID: 21549133. blood cell DNA and lung cancer: a pooled analysis of three prospective
Philibert RA, Beach SR, and Brody GH. 2012. Demethylation of the aryl cohorts. Cancer Res, 74(15), 4090–​4098. PMCID: PMC4119534.
hydrocarbon receptor repressor as a biomarker for nascent smokers. Seow WJ, Kile ML, Baccarelli AA, et al. 2014b. Epigenome-​wide DNA meth-
Epigenetics, 7(11), 1331–​1338. PMCID: PMC3499333. ylation changes with development of arsenic-​induced skin lesions in
Potter JD. 1997. Logistics and design issues in the use of biological samples in Bangladesh: a case-​control follow-​up study. Environ Mol Mutagen, 55(6),
observational epidemiology. IARC Sci Publ, 142, 31–​37. PMID: 9354909. 449–​456. PMCID: PMC4082746.
Precision Medicine Initiative. Available from:  http://​www.nih.gov/​precision-​ Severi G, Southey MC, English DR, et al. 2014. Epigenome-​wide methylation
medicine-​initiative-​cohort-​program. Accessed October 11, 2016. in DNA from peripheral blood as a marker of risk for breast cancer. Breast
Priyadharshini VS, and Teran LM. 2016. Personalized medicine in respiratory Cancer Res Treat, 148(3), 665–​673. PMID: 25407397.
disease: role of proteomics. Adv Protein Chem Struct Biol, 102(doi), 115–​ Shenker NS, Ueland PM, Polidoro S, et al. 2013. DNA methylation as a long-​
146. PMID: 26827604. term biomarker of exposure to tobacco smoke. Epidemiology, 24(5), 712–​
Pujadas E, and Feinberg AP. 2012. Regulated noise in the epigenetic 716. PMID: WOS:000323107600012.
landscape of development and disease. Cell, 148(6), 1123–​ 1131. Sherman ME, Howatt W, Blows FM, et al. 2010. Molecular pathology in epi-
PMCID: PMC3488344. demiologic studies:  a primer on key considerations. Cancer Epidemiol
Rager JE, Smeester L, Jaspers I, Sexton KG, and Fry RC. 2011. Epigenetic Biomarkers Prev, 19(4), 966–​972. PMCID: PMC2852464.
changes induced by air toxics:  formaldehyde exposure alters miRNA Shiels MS, Pfeiffer RM, Hildesheim A, et al. 2013. Circulating inflammation
expression profiles in human lung cells. Environ Health Perspect, 119(4), markers and prospective risk for lung cancer. J Natl Cancer Inst, 105(24),
494–​500. PMCID: PMC3080931. 1871–​1880. PMCID: PMC3888091.
94

94 Part I:  Basic Concepts


Shuga J, Hainaut P, and Smith MT. 2011. Assessment of genetic damage in van de Vijver MJ, He YD, van’t Veer LJ, et al. 2002. A gene-​expression signa-
healthy and diseased tissue. In: Rothman NHP, Schulte P, Smith M, ture as a predictor of survival in breast cancer. N Engl J Med, 347(25),
Boffetta P, and Perera FP (Eds.), Molecular Epidemiology: Principles and 1999–​2009. PMID: 12490681.
Practices. Lyon, France: IARC Sci Publ, 163:63–97. van Leeuwen DM, van Herwijnen MH, Pedersen M, et al. 2006. Genome-​wide
Shuga J, Zeng Y, Novak R, et  al. 2013. Single molecule quantitation and differential gene expression in children exposed to air pollution in the
sequencing of rare translocations using microfluidic nested digital PCR. Czech Republic. Mutat Res, 600(1–​2), 12–​22. PMID: 16814814.
Nucleic Acids Res, 41(16), e159. PMCID: PMC3763562. van Veldhoven K, Polidoro S, Baglietto L, et al. 2015. Epigenome-​wide asso-
Silvestri GA, Vachani A, Whitney D, et al. 2015. A bronchial genomic classi- ciation study reveals decreased average methylation levels years before
fier for the diagnostic evaluation of lung cancer. N Engl J Med, 373(3), breast cancer diagnosis. Clin Epigenetics, 7, 67. PMCID: PMC4524428.
243–​251. PMCID: PMC4838273. Vaught J. 2016. Biobanking comes of age: the transition to biospecimen sci-
Smith MT, Hainaut P, Perera FP, et  al. 2011. Future perspectives on molec- ence. Annu Rev Pharmacol Toxicol, 56, 211–​228. PMID: 26514206.
ular epidemiology. In: Rothman NHP, Schulte P, Smith M, Boffetta P, Veneziano D, Di Bella S, Nigita G, et  al. 2016. Noncoding RNA: current
and Perera FP (Eds.), Molecular Epidemiology: Principles and Practices. deep sequencing data analysis approaches and challenges. Hum Mutat,
Lyon, France: IARC, 493–499. 37(12):1283–1298. doi: 10.1002/humu.23066. Epub 2016 Sep 5. PMID:
Spira A, Beane J, Shah V, et al. 2004. Effects of cigarette smoke on the human 27516218.
airway epithelial cell transcriptome. Proc Natl Acad Sci U S A, 101(27), Vermeulen R, De Roos AJ, Bakke B, et al. 2005. A study on immunological
10143–​10148. PMCID: PMC454179. responses to exposures encountered in corn farming. J Biochem Mol
Spira A, Beane JE, Shah V, et al. 2007. Airway epithelial gene expression in Toxicol, 19(3), 172. PMID: 15977189.
the diagnostic evaluation of smokers with suspect lung cancer. Nat Med, Vermeulen R, Zhang L, Spierenburg A, et al. 2012. Elevated urinary levels of kid-
13(3), 361–​366. PMID: 17334370. ney injury molecule-​1 among Chinese factory workers exposed to trichlo-
Spitz MR, and Bondy ML. 2010. The evolving discipline of molecular epi- roethylene. Carcinogenesis, 33(8), 1538–​1541. PMCID: PMC3499056.
demiology of cancer. Carcinogenesis, 31(1), 127–​ 134. PMCID: Vikstrom AC, Warholm M, Paulsson B, et al. 2012. Hemoglobin adducts as a mea-
PMC2802669. sure of variations in exposure to acrylamide in food and comparison to ques-
Sridhar S, Schembri F, Zeskind J, et al. 2008. Smoking-​induced gene expres- tionnaire data. Food Chem Toxicol, 50(7), 2531–​2539. PMID: 22525869.
sion changes in the bronchial airway are reflected in nasal and buccal Vineis P, Chadeau-​Hyam M, Gmuender H, et al. 2016. The exposome in prac-
epithelium. BMC Genomics, 9, 259. PMCID: PMC2435556. tice: design of the EXPOsOMICS project. Int J Hyg Environ Health, 2017
Steegers-​Theunissen RP, Obermann-​ Borst SA, Kremer D, et  al. 2009. Mar;220(2 Pt A):142-151. doi: 10.1016/j.ijheh.2016.08.001. Epub 2016
Periconceptional maternal folic acid use of 400 microg per day is related Aug 19.
to increased methylation of the IGF2 gene in the very young child. PLoS Vlaanderen J, Pronk A, Rothman N, et  al. 2017. A cross-​sectional study of
One, 4(11), e7845. PMCID: PMC2773848. changes in markers of immunological effects and lung health due to expo-
Steiling K, van den Berge M, Hijazi K, et al. 2013. A dynamic bronchial airway sure to multi-​walled carbon nanotubes. Nanotoxicology, 11(3):395–404.
gene expression signature of chronic obstructive pulmonary disease and doi: 10.1080/17435390.2017.1308031. Epub 2017 Apr 3.
lung function impairment. Am J Respir Crit Care Med, 187(9), 933–​942. Vrijens K, Bollati V, and Nawrot TS. 2015. MicroRNAs as potential signatures
PMCID: PMC3707363. of environmental exposure or effect: a systematic review. Environ Health
Su D, Wang X, Campbell MR, et  al. 2016. Distinct Epigenetic Effects of Perspect, 123(5), 399–​411. PMCID: PMC4421768.
Tobacco Smoking in Whole Blood and among Leukocyte Subtypes. Wacholder S. 1991. Practical considerations in choosing between the case-​
PLoS One, 11(12), e0166486. doi:  10.1371/​ journal.pone.0166486. cohort and nested case-​control designs. Epidemiology, 2(2), 155–​158.
eCollection 2016. PMID: 1932316.
Teschendorff AE, Marabita F, Lechner M, et  al. 2013. A beta-​mixture quan- Wacholder S, Chatterjee N, and Hartge P. 2002a. Joint effect of genes and envi-
tile normalization method for correcting probe design bias in Illumina ronment distorted by selection biases:  implications for hospital-​based
Infinium 450 k DNA methylation data. Bioinformatics, 29(2), 189–​196. case-​control studies. Cancer Epidemiol Biomarkers Prev, 11(9), 885–​
PMCID: PMC3546795. 889. PMID: 12223433.
Thaiss CA, Zmora N, Levy M, and Elinav E. 2016. The microbiome and innate Wacholder S, Garcia-​Closas M, and Rothman N. 2002b. Study of genes and
immunity. Nature, 535(7610), 65–​74. PMID: 27383981. environmental factors in complex diseases. Lancet, 359(9312), 1155;
Thurman RE, Rynes E, Humbert R, et  al. 2012. The accessible chroma- author reply 1157. PMID: 11943292.
tin landscape of the human genome. Nature, 489(7414), 75–​ 82. Walker AW. 2016. Studying the human microbiota. Adv Exp Med Biol, 902(doi),
PMCID: PMC3721348. 5–​32. PMID: 27161348.
Tobi EW, Goeman JJ, Monajemi R, et al. 2014. DNA methylation signatures Walker DI, Uppal K, Zhang L, et al. 2016. High-​resolution metabolomics of
link prenatal famine exposure to growth and metabolism. Nat Commun, occupational exposure to trichloroethylene. Int J Epidemiol, 45(5):1517–
5, 5592. PMCID: PMC4246417. 1527. Epub 2016 Oct 5.
Tomso DJ, and Bell DA. 2003. Sequence context at human single nucleotide Wan ES, Qiu W, Baccarelli A, et  al. 2012. Cigarette smoking behaviors and
polymorphisms: overrepresentation of CpG dinucleotide at polymorphic time since quitting are associated with differential DNA methylation
sites and suppression of variation in CpG islands. J Mol Biol, 327(2), across the human genome. Hum Mol Genet, 21(13), 3073–​3082. PMCID:
303–​308. PMID: 12628237. PMC3373248.
Townsend MK, Bao Y, Poole EM, et  al. 2016. Impact of pre-​ analytic Wang TW, Vermeulen RC, Hu W, et  al. 2015. Gene-​expression profiling of
blood ­ sample collection factors on metabolomics. Cancer Epidemiol buccal epithelium among non-​smoking women exposed to household
Biomarkers, 25(5):823–829. doi: 10.1158/1055-9965.EPI-15-1206. Epub air pollution from smoky coal. Carcinogenesis, 36(12), 1494–​ 1501.
2016 Mar 3. PMCID: PMC4675833.
Trabert B, Pinto L, Hartge P, et al. 2014. Pre-​diagnostic serum levels of inflam- Wang Z, Gerstein M, and Snyder M. 2009. RNA-​Seq: a revolutionary tool for
mation markers and risk of ovarian cancer in the prostate, lung, colorectal transcriptomics. Nat Rev Genet, 10(1), 57–​63. PMCID: PMC2949280.
and ovarian cancer (PLCO) screening trial. Gynecol Oncol, 135(2), 297–​ Wang Z, Neuburg D, Li C, et  al. 2005. Global gene expression profiling in
304. PMCID: PMC4254357. whole-​blood samples from individuals exposed to metal fumes. Environ
Trapnell C, Williams BA, Pertea G, et  al. 2010. Transcript assembly and Health Perspect, 113(2), 233–​241. PMCID: PMC1277870.
quantification by RNA-​Seq reveals unannotated transcripts and isoform Wild CP. 2005. Complementing the genome with an “exposome”: the outstand-
switching during cell differentiation. Nat Biotechnol, 28(5), 511–​515. ing challenge of environmental exposure measurement in molecular epide-
PMCID: PMC3146043. miology. Cancer Epidemiol Biomarkers Prev, 14(8), 1847–​1850. PMID:
Tsai TH, Song E, Zhu R, et  al. 2015. LC-​MS/​MS-​based serum proteomics 16103423.
for identification of candidate biomarkers for hepatocellular carcinoma. Wild CP. 2012. The exposome: from concept to utility. Int J Epidemiol, 41(1),
Proteomics, 15(13), 2369–​2381. PMCID: PMC4490019. 24–​32. PMID: 22296988.
UK Biobank. Available from: http://​www.ukbiobank.ac.uk. Accessed October Wild CP, Scalbert A, and Herceg Z. 2013. Measuring the exposome: a power-
11, 2016. ful basis for evaluating environmental exposures and cancer risk. Environ
Uppal K, Soltow QA, Promislow DE, et  al. 2015. MetabNet:  an R package Mol Mutagen, 54(7), 480–​499. PMID: 23681765.
for metabolic association analysis of high-​resolution metabolomics data. Willingham AT, and Gingeras TR. 2006. TUF love for “junk” DNA. Cell,
Front Bioeng Biotechnol, 3, 87. PMCID: PMC4464066. 125(7), 1215–​1220. PMID: 16814704.
Uppal K, Walker DI, Liu K, et al. 2016. Computational metabolomics: a frame- Woese CR, and Fox GE. 1977. Phylogenetic structure of the prokaryotic
work for the million metabolome. Chem Res Toxicol, 29(12):1956–1975. domain: the primary kingdoms. Proc Natl Acad Sci U S A, 74(11), 5088–​
Epub 2016 Oct 12. 5090. PMCID: PMC432104.
 95

Application of Biomarkers in Cancer Epidemiology 95


Woo Y, Affourtit J, Daigle S, et al. 2004. A comparison of cDNA, oligonucle- Zhang X, Sebastiani P, Liu G, et al. 2010. Similarities and differences between
otide, and Affymetrix GeneChip gene expression microarray platforms. J smoking-​ related gene expression in nasal and bronchial epithelium.
Biomol Tech, 15(4), 276–​284. PMCID: PMC2291701. Physiol Genomics, 41(1), 1–​8. PMCID: PMC2841495.
Xu Z, Bolick SC, DeRoo LA, et al. 2013. Epigenome-​wide association study Zhao S, Fung-​Leung WP, Bittner A, Ngo K, and Liu X. 2014. Comparison of
of breast cancer using prospectively collected sister study samples. J Natl RNA-​Seq and microarray in transcriptome profiling of activated T cells.
Cancer Inst, 105(10), 694–​700. PMCID: PMC3653821. PLoS One, 9(1), e78644. PMCID: PMC3894192.
Yang AS, Estecio MR, Doshi K, et al. 2004. A simple method for estimating Zhernakova A, Kurilshikov A, Bonder MJ, et  al. 2016. Population-​ based
global DNA methylation using bisulfite PCR of repetitive DNA elements. metagenomics analysis reveals markers for gut microbiome composition
Nucleic Acids Res, 32(3), e38. PMCID: PMC373427. and diversity. Science, 352(6285), 565–​569. PMID: 27126040.
Yang Q, Khoury MJ, Sun F, and Flanders WD. 1999. Case-​ only design Zhou W, Machiela MJ, Freedman ND, et al. 2016. Mosaic loss of chromosome
to measure gene-​ gene interaction. Epidemiology, 10(2), 167–​ 170. Y is associated with common variation near TCL1A. Nat Genet, 48(5),
PMID: 10069253. 563–​568. PMCID: PMC4848121.
Yu T, Park Y, Li S, and Jones DP. 2013. Hybrid feature detection and infor- Zhuang J, Widschwendter M, and Teschendorff AE. 2012. A comparison of
mation accumulation using high-​resolution LC-​MS metabolomics data. J feature selection and classification methods in DNA methylation stud-
Proteome Res, 12(3), 1419–​1427. PMCID: PMC3624888. ies using the Illumina Infinium platform. BMC Bioinformatics, 13, 59.
Zhang L, Tang X, Rothman N, et al. 2010. Occupational exposure to formal- PMCID: PMC3364843.
dehyde, hematotoxicity, and leukemia-​specific chromosome changes in Ziller MJ, Gu H, Muller F, et  al. 2013. Charting a dynamic DNA meth-
cultured myeloid progenitor cells. Cancer Epidemiol Biomarkers Prev, ylation landscape of the human genome. Nature, 500(7463), 477–​481.
19(1), 80–​88. PMCID: PMC2974570. PMCID: PMC3821869.
96
 97

7 Causal Inference in Cancer Epidemiology

STEVEN N. GOODMAN AND JONATHAN M. SAMET

OVERVIEW of association has reached a critical threshold of certainty and that a


reduction of exposure can be expected to be followed by reduced dis-
Judgments about causality are central to the development of interven- ease occurrence. Over the last 50 years, identification of the external
tions intended to prevent or reduce exposure to risk factors that cause causes of cancer has been the primary focus of most epidemiologic
cancer. Because causation is not directly observable in medicine, sci- research on cancer; only relatively recently has attention shifted toward
entists and philosophers have had to develop sets of constructs and identifying inherited and acquired genetic determinants of susceptibil-
heuristics that define “cause” operationally. Just as the Henle-Koch ity, metabolic factors that affect risk, intermediate markers of the early
criteria are used to confirm that a specific microorganism causes an stages of carcinogenesis, and evaluation of preventive interventions.
infectious disease, a parallel framework has been developed to evaluate There are numerous examples in which the identification of a cause
whether associations observed in epidemiologic studies of non-com- of cancer has led to intervention and reduction of disease occurrence.
municable diseases represent causation, rather than spurious correla- Tobacco use and cancer of the lung is a notable example for its histori-
tions or chance. The criteria in this framework, often attributed to the cal precedence and for the framework applied to the scientific evidence
British medical statistician Sir Austin Bradford Hill or to the 1964 as the causality of the association was evaluated (US Department of
Report of the US Surgeon General on tobacco, include consistency, Health, Education and Welfare, 1964; White, 1990). A broad range
strength of association, specificity, temporality, coherence/plausibility/ of causal risk factors has been linked to diverse forms of cancer.
analogy, biological gradient, and experiment. This chapter reviews These include infectious agents (e.g., human papillomavirus and cer-
these criteria in depth and considers the challenges of applying them vical cancer), physical agents (e.g., ionizing radiation and leukemia
to population research on cancer. It discusses the concepts of causation and solid tumors), inhaled agents (e.g., tobacco smoke or radon and
in the context of the multistage nature of cancer, the “counterfactual” lung cancer), pharmaceutical agents and hormones (e.g., diethylstil-
notion of causation, the component cause model for understanding dis- bestrol and adenocarcinoma of the vagina), food contaminants (e.g.,
eases with multiple causes, and the “weight of the evidence” approach aflatoxin and liver cancer), workplace exposures (e.g., asbestos and
for integrating information from multiple lines of research. It also mesothelioma), behavioral exposures (e.g., alcohol consumption and
describes the increasing power of mechanistic information to reveal liver cancer), and inherited genetic mutations (e.g., Li-​Fraumeni syn-
the processes of carcinogenesis at the molecular level. It posits that drome and cancer of multiple sites). These and other factors have been
mechanistic information, such as the discovery of molecular “signa- judged to be causal only after the accumulation of sufficient evidence
tures” of specific exposures, will continue to grow in importance as that, in most instances, represents multiple lines of research including
one of the “twin pillars” of causal inference, and will in some cases epidemiologic studies, clinical research, laboratory experiments and
reduce our reliance on empirical, population-based data. preventive interventions that have reduced risk.
This chapter provides an overview of causal inference with a focus
on the interpretation of epidemiologic data on cancer risk. It begins
INTRODUCTION with an introduction to the centuries-​old discussion on cause and
causation and next considers the epidemiologic concept of causation,
The prevention of disease has long been based implicitly on taking setting the discussion in the context of current understanding of car-
action on the assumption that a disease is caused by a factor that can cinogenesis as a multistep process. The criteria for causation, often
be controlled. Early examples include the experimental evidence gen- attributed to the Britis medical statistician Sir Austin Bradford Hill
erated by Lind, showing that consumption of oranges and lemons (1965) or to the 1964 Report of the US Surgeon General on tobacco
prevented scurvy, and Snow’s observations on cholera occurrence in (US Department of Health, Education and Welfare, 1964), have pro-
London, showing a disease pattern consistent with water-​borne trans- vided a framework for the evaluation of evidence in judging the cau-
mission (Johnson, 2007; Rosen, 1993). In these examples, preventive sality of associations.
steps followed: After Lind’s experiment, the diets of the British navy These criteria are addressed in depth, and their application is illus-
were supplemented with citrus fruits; and after Snow’s observational trated with the example of tobacco smoking, both active and passive, in
study, steps were taken to ensure that the source of water was changed relation to lung cancer. The chapter concludes with a consideration of
in the affected areas of London. Over the ensuing centuries, infec- emerging issues concerned with causation, including the interpretation
tious agents were causally linked to specific diseases, and preven- of data coming from the new “-​omics” (e.g., genomics, epigenomics,
tion was accomplished by interrupting transmission and by vaccines. metabolomics, exposomics, and proteomics), the expanding technolo-
During the twentieth century, public health was threatened by parallel gies of contemporary “molecular epidemiology,” and new approaches
epidemics of several chronic diseases, including cancer; and as the to modeling epidemiological data and evaluating causation.
causal agents were identified, a broad range of preventive strategies
was implemented. Underlying these actions was the determination that
a causal relationship existed between some agent and disease risk. CONCEPTS OF CAUSATION
The concept of causation has long had a central and pivotal role in
the application of epidemiologic evidence for controlling cancer. The At its foundation, “cause” is not knowable with certainty. This fact under-
designation of a risk factor as “causal” has been the starting point for lies much of the methodologic and conceptual confusion that often swirls
initiating cancer prevention programs based on reducing exposure to around claims of causation based on scientific evidence. The fundamental
the risk factor. Although the concept of causation itself remains a mat- intuition underlying the causal concept is that event A somehow produces
ter of continuing discussion among philosophers and others, use of the another event, B. However, the “production” of B is not observable. The
term in public health implies that the evidence supporting causality philosopher Bryan Magee summarized this conundrum eloquently.

97
98

98 Part I:  Basic Concepts


It seems to be impossible for us to form any conception of an In observational studies of humans, however, researchers must try to
ordered world at all without the idea of there being causal con- infer what the outcome would be in a counterfactual state by study-
nections between events. But when we pursue this idea seriously ing another group of persons who, at least on average, are substan-
we find that causal connection is not anything we ever actually tively different from the exposed group in only one variable:  the
observe, nor ever can observe. We may say that Event A causes exposure under study. The outcome of this second group is used to
Event B, but when we examine the situation we find that what we represent what would have occurred in the original group if it were
actually observed is Event A  followed by Event B.  There is not observed with an exposure different from that which actually existed
some third entity between them, a casual link, which we also (Greenland, 1990). For example, in the case of smoking and disease,
observe… . So we have this indispensable notion of cause at the this comparison is between disease risk in smokers and nonsmokers.
very heart of our conception of the world, and of our understanding Simply observing a difference in the probability of an outcome
of our own experience, which we find ourselves quite unable to between two groups that differ on X is not sufficient evidence for cau-
validate by observation or experience… . It actually purports to tell sation because it does not distinguish between causation and spurious
us how specific material events are related to each other in the real or indirect association, produced by “confounders,” or ancillary causes.
world, yet it is not derived from, nor can it be validated by, observa- The notion of “causation” requires that the cause somehow actively
tion of that world. This is deeply mysterious. (Magee, 2001) “produce” its effect, which is captured operationally by the requirement
that active manipulation of the cause should produce a change in the
The fact that causation is not directly observable means that sci- probability of the outcome. For example, if one saw that students with
entists and philosophers have had to develop a set of constructs and poor visual acuity typically sat closer to the front of a classroom, one
heuristics by which to define a “cause” operationally. These constructs would not call the seating arrangement a “cause” of their poor eyesight
typically have two components:  a predictive or associational one, unless it could be shown that seating them farther back improved it. The
determined empirically from variations in the probability of disease notation that captures this idea is one that introduces an operator, not
occurrence, and an explanatory one, based on a proposed underlying part of traditional statistical notation “Set (X = x),” which corresponds
mechanism. All causal claims rest on these twin pillars; an association to actively setting a risk factor X equal to some value x, rather than
with no plausible mechanistic basis is typically not accepted as causal, simply observing that the factor is equal to x. Thus, the counterfactual
and a proposed mechanism, however well founded, cannot be accepted notion of probabilistic causation for a risk factor X requires condition 2.
as the basis for a causal claim without empirical demonstration that
the outcome occurs more often in the presence of the purported cause Condition 2: no confounding
than in its absence. However, these components need not contribute Pr[outcome | set(X = x)] = Pr(outcome | X = x)
equally, and various causal claims may rest on quite different balances
of contributions of empirical and mechanistic information. If we put together condition 1 (that there is an observed association
Underlying any operational definition of causality must be an onto- between cause and effect) with condition 2 (that there is no other extra-
logic one: that is, how a cause is defined in principle. A particularly use- neous cause responsible for the observed effect), we have the coun-
ful, widely accepted definition in both philosophy and epidemiology is terfactual condition for probabilistic causation, expressed as follows.
the “counterfactual” notion of causation. This concept had its origins at
least as far back as the English philosopher David Hume (1711–​1776) Condition 1 + Condition 2 = Causality condition
(Hume, 1739). During the twentieth century, this concept was further Pr[outcome | set(X = y)] ≠ Pr[outcome | set(X = z)]
developed and applied by statisticians, philosophers, and epidemiolo-
gists (Bunge, 1959; Glass et  al., 2013; Greenland, 1990; Greenland This condition states that if the probability of an outcome changes
et al., 1999; Hernán and Robins, 2016; Lewis, 1973; Neyman, 1990; when risk factor X is actively changed from z to y, then X is regarded
Pearl, 2000; Robins, 1986; 1987; Rubin, 1974). The counterfactual as a cause of the outcome.
definition holds that something is a cause of a given outcome if, when In the randomized controlled trial, a risk factor is actively manipu-
the same individual is observed with and without a purported cause and lated. Understanding the role of randomization can deepen insights
without changing any other characteristic of that individual, a different into the interpretation of nonrandomized designs used in observational
outcome would be observed; for example, the counterfactual state for studies. Randomization has two critical consequences:  (1)  it makes
a smoker is the same individual never having smoked. The state that exposure to a proposed causal factor independent of potentially con-
cannot be observed is called the counterfactual state: literally, counter founding factors; and (2) it provides a known probability distribution
to the observed facts. The impossibility of observing the counterfactual for the potential outcomes in each group under a given mathematical
state is what makes all causal claims subject to uncertainty. hypothesis (i.e., the null) (Greenland, 1990). Randomization does not
In practice, the risk caused by most exposures is not so extreme that necessarily free the inference from an individual randomized study
it can be observed in every individual. Rather, the definition is probabi- from unmeasured confounding (it does so only on average, assum-
listic, meaning that the outcome is more likely to occur in the presence ing proper randomization). Randomization does imply that measures
of the cause than in its absence. Health research rarely deals with causes of uncertainty about causal estimates from randomized studies have
that inevitably produce certain outcomes, or with outcomes that never an experimental foundation. In the absence of randomization, uncer-
occur absent specific causes. For example, smokers do not always get tainty about causal effects depends in part on the confidence that all
lung cancer, and never-​smokers do develop this malignancy. Therefore, substantive confounding has been eliminated or controlled by either
the counterfactual definition must be expanded to encompass the notion the study design or the analysis. The level of confidence is ultimately
of a probabilistic rather than deterministic outcome. The formal defini- based on scientific judgment and consequently is subject to uncertainty
tion of a cause in population studies requires that a factor X be associ- and questioning.
ated with a difference in the likelihood (probability) of an outcome. For One way to increase that confidence is to repeat the study. Similar
example, if X may take on two different values, y or z: results in a series of randomized studies make it increasingly unlikely
that unmeasured confounding is accounting for the findings, as the
Condition 1: observed association process of randomization makes the mathematical probability of such
Pr(outcome | X = y) ≠ Pr(outcome | X = z) confounding progressively smaller as the sample size or number of
studies increase. In observational studies, however, increasing the num-
Properly designed studies provide a scientific basis for inferring ber of studies reduces the random component of uncertainty, but does
what the outcome of the counterfactual state would be and permit the not necessarily eliminate systematic bias from confounding. Without
related uncertainty to be quantified. In a laboratory, scientists are able randomization, there is no mathematical basis for assuming that an
to predict the outcome in this counterfactual state, generally with a imbalance of unknown confounders decreases with an increase in the
high degree of confidence, by repeating an experimental procedure number of studies. However, if observational studies are repeated in
with every factor tightly controlled, varying only the factor of interest. different settings with different persons, different eligibility criteria,
 9

Causal Inference in Cancer Epidemiology 99


and/​or different exposure opportunities, each of which might eliminate least one of these causal complexes. Individual cases result from hav-
another source of confounding from consideration, the confidence that ing the full complement of components for one or more of the causal
unmeasured confounders are not producing the findings is increased. composites. We know, for example, that cigarette smoking is a power-
There are no fixed guidelines on how many studies need to be done, ful cause of lung cancer that accounts for approximately 90% of cases
how diverse they need to be, and how relevant they are to the question in most countries. In the example shown in Figure 7–1, smoking would
at hand to guarantee the benefits of consistency. These are matters of correspond to factor B, which contributes to causal complexes I and
scientific judgment; explicit criteria cannot be offered. II but not III. Smoking is not a necessary cause of lung cancer, since
In the scientific community, including epidemiologists, there is a approximately 5%–​10% of cases in the United States occur in never-​
general move to support efforts that will enhance the reproducibility smokers (Samet et  al., 2009). Nor is smoking by itself a sufficient
of results (Begley and Ioannidis, 2015; Peng, 2011). The approaches cause of lung cancer, since not all long-​term heavy smokers develop
include active sharing of data and analytical models, including the the disease. Figure 7–1 indicates the need for additional cofactors,
code used for the models. The finding of reproducible results in analy- such as genes that influence carcinogen metabolism, DNA repair, and
ses of pooled data does not exclude bias arising from participant selec- the programmed cell death of genetically damaged cells, to complete
tion and confounding, but it does minimize false leads from chance, causal complexes I and II.
from biases that individual investigators may introduce in analyzing Rothman’s model has significant implications for considering the
and reporting data and from outright errors. burden of cancer attributable to a particular risk factor and when
Confidence that unmeasured confounding is not producing the assigning priorities to specific preventive interventions. For example, a
observed results is further increased by understanding the biologic single exposure can contribute to multiple component pathways (e.g.,
process by which the exposure might affect the outcome. This under- A and B in causes I and II), implying that the cases associated with
standing allows better identification and measurement of relevant con- these causes might be prevented by eliminating exposure to either A or
founders, making it less likely that unmeasured factors are of concern. B. This is useful when ranking the priority of various interventions,
Biologic understanding can also serve as the basis for a judgment even though it may appear confusing that the sum of the attributable
that the observed difference in outcome frequency could be produced fraction estimates exceeds 100%. The overlap in causation should not
only by an implausible degree of imbalance in the confounding factor be misinterpreted because of the expectation that the total burden of
between exposed and unexposed groups. Thus, causal inference based preventable cancers associated with all of the various causal factors
on observational studies typically requires more replication and stronger should add up to 100% (Schottenfeld et al., 2013).
biologic evidence to support plausibility and to exclude confounding
than is needed for inferring causation from randomized studies. The new
“-​omics” technologies expand the opportunities to combine mechanistic CRITERIA FOR CAUSALITY
and probabilistic evidence as the basis for causal inference.
Epidemiologists and other health researchers have needed pragmatic
definitions of causation to support the translation of evidence from
COMPONENT CAUSE MODEL etiologic research into preventive and curative interventions (Olsen,
2003; Susser, 1973). The epidemiologic literature has consequently
Causes defined in the manner described in the preceding can be placed great emphasis on identifying criteria by which evidence is
viewed as working together in many ways. Rothman (1976) proposed evaluated to determine if a factor can be considered to cause disease.
a useful framework for considering multiple-​cause diseases that has These criteria are necessarily different for chronic diseases such as
ready extension to the causation of cancer, as most cancers have sev- cancer, which have multiple causes, heterogeneous clinical features,
eral causes. Rothman proposed that a disease may have several suf- and a much longer induction period than those used for infectious dis-
ficient causes, each accounting for some proportion of the cases in the orders caused by specific microorganisms.
population, and that each cause may have several components (Figure In biomedical research, the first criteria came following the dis-
7–1) (Rothman, 1976). In this model, all of the components of any covery of bacteria during the nineteenth century. A method was then
single causal complex must be present for disease to develop. In this needed for judging if an organism caused a particular disease. The
example, causal complexes I and II would be incomplete if either A or criteria originally put forward for making this judgment are gener-
B were not present. Factor A (but not B) is also a necessary component ally attributed to Robert Koch and his mentor Jacob Henle, although
of sufficient cause III. Because factor A must be present in all of the Koch also acknowledged Eugene Klebs. Evans (1993) provided a full
causal complexes, it is a necessary cause of the disease. It is not a suffi- accounting of the elaboration of these criteria, now referred to as the
cient cause of the disease, however, since other factors must be present Henle-​Koch postulates (Box 7–1). The criteria proved valuable for
to complete any one of the causal complexes. linking infectious agents to infectious diseases, which often have spe-
Rothman’s model is useful for considering causation of the diverse cific clinical features and unique, specific causal agents (e.g., pulmo-
forms of cancer, particularly for the majority of cancers for which both nary tuberculosis and Mycobacterium tuberculosis).
inherited (genetic) and acquired (environmental) risk factors typically The criteria used for infectious diseases proved unsuitable, however,
play a role in a multistage process that transforms a normal cell into a for inferring the causation of cancer and other chronic diseases, which
malignant cell. There may be multiple ways to complete this sequence became the dominant causes of death in high-​and middle-​income
of changes, involving the actions of different complexes of factors, countries throughout the twentieth century. Unlike many infectious
analogous to composite causes I, II, and III in Figure 7–1. A particular diseases, these diseases were commonly associated with multiple fac-
exposure, such as factor A, is considered causal if it contributes to at tors, and many cases could not be clearly linked to any risk factors.
The limitations of the Henle-​Koch postulates were recognized during

Sufficient Sufficient Sufficient


Cause Cause Cause Box 7–1.  HENLE-​K OCH POSTULATES
I II III

E D H G J I
1. The parasite occurs in every case of the disease in question and
under circumstances that can account for the pathologic changes
and clinical course of the disease.
A C A F A F
2. It occurs in no other disease as a fortuitous and nonpathogenic
B B C parasite.
3. After being fully isolated from the body and repeatedly grown
in pure culture, it can induce the disease anew.
Figure 7–1.  Conceptual schemes for the causes of a hypothetical disease.
10

100 Part I:  Basic Concepts

Box 7–2.   SIR AUSTIN BRADFORD HILL’S CAUSAL different persons, places, circumstances, and times. Consistency can
CRITERIA: ASPECTS OF ASSOCIATION TO BE have two implications for causal inference. First, consistent findings
CONSIDERED BEFORE DECIDING ON CAUSATION reduce the likelihood of chance associations; if the results from several
studies are combined, this also improves the precision of the estimates.
Second, consistency makes it less likely that unmeasured confounding
Strength
accounts for the observed association. Such confounding would have
Consistency
to persist across diverse populations, conditions of exposures, and
Specificity
measurement methods. Confounding is still possible if the exposure
Temporality
of interest were strongly and universally tied to an alternative cause, as
Biologic gradient
was claimed in the form of the “constitutional hypothesis” put forward
Plausibility
in the early days of the smoking-​disease debate (US Department of
Coherence
Health, Education and Welfare, 1964). This hypothesis theorized that
Experiment
there was a constitutional (i.e., genetic) factor that made people more
Analogy
likely to smoke and, independent of smoking, to develop cancer. No
such factor has ever been identified. Nevertheless, it is true that con-
sistency serves mainly to exclude the possibility that the association is
the first wave of epidemiologic studies on chronic diseases in the mid-​ produced by an ancillary factor that differs across studies, but this does
twentieth century. In 1959, Yerushalmy and Palmer proposed criteria not eliminate the possibility that an extraneous factor could affect all
for evaluating the etiology of chronic diseases that acknowledged the or most of the studies (Rothman and Greenland, 1998).
need for evidence of increased risk in exposed persons and for hand- Consistency does not include the qualitative strength of such stud-
ling the lack of specificity compared to infectious diseases. Lilienfeld ies, which Susser subsumed under his subsidiary concept of “surviv-
(1959) and Sartwell (1960), discussing the article, added the consider- ability,” relating to the rigor and severity of tests of association (1991).
ation of dose–​response, the strength of the association, its consistency,
and its biologic plausibility.
The most widely cited criteria in epidemiology and public health were Strength of Association
set forth by Sir Austin Bradford Hill in 1965 (Box 7–2) (Hill, 1965). Five
of the nine criteria he listed were also put forward in the 1964 Surgeon Strength of association includes two dimensions: the magnitude of the
General’s report (US Department of Health, Education and Welfare, association and its statistical strength. An association strong in both
1964)  as the criteria for causal judgment:  consistency, strength, speci- aspects makes the alternative explanations of chance and confounding
ficity, temporality, and coherence of an observed association. Recently, unlikely. The larger the measured association, the less likely it is that
these criteria were attributed to Reuel Stallones, an epidemiologist and an unmeasured or poorly controlled confounder could account for it
consultant to the Advisory Committee that authored the 1964 report completely. Associations that have a small magnitude or weak statisti-
(Stallones, 2015). Hill also listed biologic gradient (dose–​response), cal strength are more likely to reflect artifactual correlations caused by
plausibility, experiment (or natural experiment), and analogy. Many of chance, a modest degree of bias, or unmeasured weak confounding.
these criteria had been cited in earlier epidemiologic writings (Lilienfeld, However, the magnitude of association is reflective of underlying bio-
1959; Sartwell, 1960; Yerushalmy and Palmer, 1959); and Susser and logic processes and should be consistent with understanding the role
others have refined them extensively by exploring their justification, mer- of the risk factor in these processes. Either a strong or a weak effect
its, and interpretations (Kaufman and Poole, 2000; Susser, 1973, 1977). might be considered plausible, based on knowledge of the underlying
Hill (1965) clearly stated that these criteria were not intended to processes.
serve as a checklist. In the example of active smoking and lung cancer, the relative risks
listed in the first Surgeon General’s Report (US Department of Health,
Here are then nine different viewpoints from all of which we Education and Welfare, 1964)  were notably elevated in men, reach-
should study association before we cry causation. What I do not ing as high as 10 or more. At that time, other causes of lung cancer,
believe … is that we can usefully lay down some hard-​and-​fast particularly occupational agents, had been identified. Exposure to
rules of evidence that must be obeyed before we accept cause occupational carcinogens would have been much lower in the general
and effect. None of my nine viewpoints can bring indisputable population than in occupational subgroups, making these implausible
evidence for or against the cause-​and-​effect hypothesis, and none as potential confounders in the general population.
can be required as a sine qua non. What they can do, with greater Passive smoking, by contrast, has a far smaller effect on lung cancer
or less strength, is to help us to make up our minds on the funda- risk. Based on studies that compared persons with greater and lesser
mental question—​is there any other way of explaining the facts exposures (e.g., never-​smoking women married to smokers vs. those
before us, is there any other answer equally, or more, likely than married to never-​smokers), the 1986 report of the US Surgeon General
cause and effect? (Hill, 1965) (US Department of Health and Human Services, 1986)  concluded
that passive smoking caused lung cancer. The magnitude of the effect
All of these criteria pertained to the evaluation of statistical associa- was much smaller than for active smoking in most studies, as would
tions observed in one or more study; if no association was observed, be anticipated, but within a plausible range. The relative risk associ-
the criteria are not relevant. Hill explained how if a given criterion ated with marriage to a smoker has been estimated to be around 1.2 in
were satisfied, it strengthened a causal claim. Each of these nine cri- various meta-​analyses (International Agency for Research on Cancer,
teria served one of two purposes: as evidence against competing non- 2004; US Department of Health and Human Services, 2006).
causal explanations, or as positive support for causal ones. Noncausal
explanations for associations include chance; residual or unmeasured
confounding; model mis-​specification; selection bias; errors in mea- Specificity
surement of exposure, confounders, or outcome; and issues regarding Specificity has been interpreted to mean that an exposure causes only
missing data (which can also include missing studies, such as publica- one (or few) diseases, or that a disease has only one cause. There are
tion bias). The criteria are discussed in the following. some cancers caused principally by a single type of exposure, for exam-
ple, mesothelioma from asbestos exposure and angiosarcoma from
exposure to vinyl chloride monomer. These are exceptions, however.
Consistency The requirement for specificity was originally derived from the Henle-​
The consistency criterion refers to the persistent finding of an associa- Koch postulates for infectious causes of disease (Susser, 1991) and is
tion between exposure and outcome in multiple studies of adequate seldom relevant to chronic diseases. When specificity with respect to
power and carried out by different investigators in studies involving outcomes exists, it can strengthen a causal claim, but its absence does
 10

Causal Inference in Cancer Epidemiology 101


not weaken other evidence (Sartwell, 1960). For example, most can- is predicted by many cause-​and-​effect models and biologic processes
cers are known to have multifactorial etiologies; many cancer-​causing but, more importantly, because it makes most noncausal explanations
agents can cause several types of cancer, and these agents can also highly unlikely. If some factor other than that of interest explains the
have noncancerous effects. observed gradient, the unmeasured factor must change in the same
When considering specificity in relation to the smoking–​lung can- manner as the exposure of interest. Except for confounders that are
cer association, the 1964 Surgeon General’s report (US Department closely related to a causal factor, it is extremely difficult for such a
of Health, Education and Welfare, 1964) provides a rich discussion of pattern to be created by virtually any of the noncausal explanations for
this criterion. The committee recognized the linkage between speci- the association. The finding of a dose–​response relation has long been
ficity and the strength of association and understood the symmetrical a mainstay of causal arguments in smoking investigations; virtually all
relationship between the two. In the most extreme instance, a particu- health outcomes causally linked to smoking have shown an increase in
lar exposure always results in a specific disease, and the disease always risk and/​or severity with an increase in the lifetime smoking history.
results from the exposure. The committee acknowledged that smoking This criterion is not based on any specific shape of the dose–​response
did not match that example, since it does not always result in lung relation.
cancer and lung cancer has other causes. However, the report noted the
extremely high relative risk for lung cancer in smokers and the high
attributable risk, and concluded that the association between smoking Experiment
and lung cancer had “a high degree of specificity.” The criterion “experiment” refers to situations where natural condi-
There is, however, another interpretation of specificity that can be tions might plausibly imitate conditions of a randomized experiment
critical to the design and interpretation of observational studies. This in an otherwise observational setting, producing a “natural experi-
is that, in a given study, some outcomes are highly unlikely to be caus- ment” whose results might have the force of a true experiment. An
ally related to the exposure, based on a priori knowledge. These can be experiment is typically a situation in which the scientist manipulates
used as internal “negative controls” to detect broad nonspecific associ- the exposure in a manner independent of the subject’s characteristics.
ations. The absence of observed associations between these outcomes Sometimes nature produces similar conditions. The reduced risk after
and the exposure of interest provides some assurance that the observed smoking cessation serves as one such situation that approximates an
associations are meaningful (Schuemie et al., 2014). experiment, although it is conceivable that unmeasured causal fac-
tors associated with health are more frequent among those who stop
smoking than among those who continue. The causal interpretation is
Temporality further strengthened if risk continues to decline among former smok-
Temporality requires that exposure to a cause precede its purported ers with increasing time since quitting. Similar to the dose–​response
effect. Temporality is the sine qua non of causality, since a causal criteria, observations of risk reduction after quitting smoking have the
exposure must be present before it can affect risk. Rothman and oth- dual effects of making most noncausal explanations unlikely and sup-
ers emphasize that temporality is the only criterion that must be ful- porting the biologic model that underlies the causal claim.
filled for an association to be considered causal (Rothman, 1986; Glass
et al., 2013). Any question about a temporal sequence seriously weak-
ens a causal claim. However, establishing temporal precedence does APPLYING THE CAUSAL CRITERIA
not by itself establish strong evidence for causality.
The process of inferring causation from observational data involves
more than simply lining up the evidence as it relates to each criterion,
Coherence, Plausibility, and Analogy although there is evidence that epidemiologists tend to use the evi-
dence in neither a consistent nor comprehensive manner (Weed and
Although the original definitions of coherence, plausibility, and anal- Gorelic, 1996). Judgments about the strength of individual studies,
ogy were subtly different, in practice they have been treated essentially the appropriateness of design and analysis, adequacy of the sample
as one idea: that a proposed causal relation should not violate known size, and rigor in excluding confounding must then be assessed in light
scientific principles, and that it be consistent with experimentally of other lines of evidence. Epidemiologic evidence alone is generally
demonstrated biologic mechanisms and other relevant data, such as not regarded as sufficient for establishing causality (Last et al., 2001).
observed patterns of disease (Rothman and Greenland, 1998). In addi- In the example of tobacco smoke, the epidemiologic data must be
tion, if biologic understanding can be used to set aside explanations integrated with evidence from experimental, toxicological, and clini-
other than a causal association, it offers further support for causality. cal studies in a multidisciplinary process. A  weight-​of-​the-​evidence
Together, these criteria can serve to both support a causal claim (by approach is then used to assess how well an observed association ful-
supporting the proposed mechanism) and refute it (by showing that the fills the causal criteria. It is ultimately a matter of judgment whether
proposed mechanism is unlikely). the collective the evidence is sufficient to exclude alternative explana-
Biological understanding evolves continuously as scientific tions. The 1964 Surgeon General’s report still stands as one of the fin-
advances make possible ever deeper exploration of disease pathogen- est examples of the power of applying these criteria systematically and
esis. For example, in 1964 the Surgeon General’s committee found the comprehensively. Starting with the criterion for consistency, the com-
causal association of smoking with lung cancer to be biologically plau- mittee noted that all 29 retrospective studies (i.e., case-​control) and 7
sible based on knowledge of the presence of carcinogens in tobacco prospective studies (i.e., cohort) at the time reported highly significant
smoke and animal experiments. More than 50  years later, this asso- smoking–​lung cancer relations. They further noted that all of the stud-
ciation remains biologically plausible, but that determination rests not ies comparing long-​term smokers to nonsmokers showed high relative
only on the earlier evidence but on subsequent discoveries regarding risks for lung cancer (~10). Dose–​response effects were also observed
the genetic and molecular basis of carcinogenesis, which provide a in every prospective study and in all retrospective studies where it
level of understanding that could not have been anticipated in 1964. In could be calculated. The temporal sequence was reported to be not
fact, the 2010 report of the Surgeon General on tobacco was devoted absolutely certain, but seemed highly unlikely to be in the direction of
to the topic of the mechanisms by which smoking causes disease (US lung cancer causing smoking, as cancer typically appears many years
Department of Health and Human Services, 2010). or decades after the onset of smoking. With regard to coherence of the
association with known facts, the studies noted the ecologic increase in
lung cancer rates with increased smoking in the population; the gender
Biologic Gradient (Dose–​Response) differential in lung cancer, which at the time was consistent with lon-
The finding of a graded increase in risk in relation to increasing expo- ger term smoking by men; the urban-​rural difference, which air pollu-
sure to the possible cause provides strong positive support in favor tion could not completely explain; socioeconomic differentials in lung
of a causal hypothesis. This is not just because such an observation cancer, for which smoking seemed to be the strongest explanation;
102

102 Part I:  Basic Concepts


and the topography of cancers within the respiratory tract in relation seems possible that such approaches may soon be able to document
to the type of smoking. The studies also cited the known reduction causal connections between a subset of exposures and disease at the
in risk among former smokers, with greater risk reductions correlated individual level. If such signatures are eventually established with suf-
with more time spent not smoking. These observations, in combination ficient confidence, the societal implications would be profound, as
with histopathological evidence, basic biologic observations, and an responsibility for causation in individuals could be assigned.
in-​depth discussion of each competing non-​smoking-​related explana- One challenge that results from these developments is that our
tion (e.g., occupation, constitutional hypothesis, infections, environ- technical ability to perform mass screening using high throughput,
mental factors such as pollution) produced a case for causation that genomic, and proteomic technologies has far outstripped our ability
proved irrefutable. to understand what we find. The functions of many genes, genetic
The 1986 Report of the Surgeon General (US Department of Health alterations, and gene products are not yet understood. The enormous
and Human Services, 1986)  concluded that passive smoking causes quantities of data related to exposure and susceptibility increasingly
lung cancer, a conclusion that has proven to be momentous in its impli- overwhelm our traditional approaches to data analysis and interpreta-
cations. This report also based its evaluation of the evidence on the tion. Epidemiologists have long faced the problem of data overload,
causal criteria. A  clear distinction was made between the evidence but never on this scale. Millions of potential markers are now measured
regarding active smoking and that expected from the much lower and analyzed in all combinations, sometimes in only tens or hundreds
doses of carcinogens arising from passive smoking. Biologic plausibil- of subjects. Undoubtedly, future research will generate even larger
ity was emphasized, including the substantial evidence on lung cancer and richer data sets from full genome sequencing and “exposomics.”
risk in active smokers. This causal conclusion has been reaffirmed in Consequently, the potential for false-​positive associations caused by
all subsequent reports (International Agency for Research on Cancer, multiplicity (“data dredging”) will continue to increase (Begley, 2013;
2004; Samet and Wang, 2000; US Department of Health and Human Begley and Ioannidis, 2015).
Services, 2006, 2014). Issues concerning measurement must also be considered in this rush
toward exploring new potential causes of cancer (Little et al., 2002).
Many of the techniques and assays used to identify metabolic prod-
EMERGING ISSUES IN CAUSAL INFERENCE ucts, genes, and gene products are still relatively new and are not yet
AND CANCER fully standardized. Understanding the reliability and validity of these
techniques is an arduous process that often does not receive sufficient
Perhaps the most challenging and exciting issue facing all scientific attention, yet it is critical for distinguishing likely spurious claims
disciplines in cancer is the prospect of more fully understanding the from those that are well grounded.
processes of carcinogenesis at the molecular level. Great advances Our existing frameworks for causal inference will certainly continue
have been made that have implications for understanding etiology, to evolve with future advances in understanding the basic mecha-
early diagnosis, and treatment. The paradigm of “precision oncology” nisms of carcinogenesis and the ever-​widening array of biomarkers
is being vigorously promoted with the goal of highly individualized that reflect internal exposures, metabolic pathways, and pre-​malignant
therapy based on the tumor’s genome. Underlying this paradigm shift events. New criteria will be needed to determine whether a proposed
is the proposition that increasingly in-​depth sequencing of germ-​line mechanistic pathway is actually an important contributor to risk
and tumor DNA, along with other data, will provide genetic and epi- (Goodman and Gerson, 2013). This inferential problem is recognized,
genetic “signatures” that indicate the causal pathways and tumor char- and adaptations of the Hill guidelines have been proposed (Goodman
acteristics that should be targeted for individual patients. and Gerson, 2013). Epidemiologists also have some new tools for
With the opening of the cancer “black box,” however, comes greater facilitating causal inference. Directed acyclic graphs (DAGs) have
recognition of the extraordinary complexity of carcinogenesis at the utility for sharply specifying underlying causal, mechanistic pathways
molecular level (Smith et al., 2015). This is apparent in many ways. (Greenland et al., 1999). Over the last two decades, causal modeling
Not only are the effects of external exposures modified by metabolic approaches have advanced that are based around a causal framework
pathways, but environmental factors also can modify gene expression and that improve estimation of causal effects in comparison with ear-
(e.g., through DNA methylation) (Brien et al., 2016; Mortensen and lier approaches (Petersen and van der Laan, 2014). Marginal structural
Euling, 2013), interact with each other and with genetic factors in myr- models are widely used for this purpose.
iad ways, and perhaps most intriguingly, behavioral patterns can be We are increasingly able to identify subpopulations of individu-
influenced by inherited genetic susceptibility, as has been documented als who may experience different degrees of risk or benefit from a
for nicotine, alcohol, and drug addiction (Morozova et al., 2014; US particular exposure or interventions. This heterogeneity of risk may
Department of Health and Human Services, 2010). This complexity result from inherited or somatic alterations that affect the absorption,
poses significant challenges for population-​based risk assessments metabolism, or cellular effect of an agent or the host’s susceptibility
and causal inference because it raises, more acutely than with con- to the exposure or intervention (Links et al., 1995; National Research
ventional risk factors, questions about which genes lie in the causal Council et al., 2012). The smaller size of these subpopulations poses
pathway (those that do should not be considered covariates), which its own set of challenges to epidemiologic approaches to risk assess-
genes or biomarkers are necessary for the effects or function of oth- ment: the finer the risk stratification, the more difficult it is to measure
ers (i.e., there may be many biologically based high-​level interaction variations in risk using epidemiologic methods and to assess whether
terms), whether an observed association represents a direct or indirect associations are causal (Moore et al., 2003; Slattery, 2002).
effect on causation, and whether it is meaningful to talk about binary As we come closer to understanding cancer on the molecular level,
exposure–​outcome or gene–​outcome relations when the components many have raised the possibility of individual risk prediction, guided
of these complex networks either cannot be changed individually or by molecular data (Hussain et al., 2001; National Research Council,
cannot be changed at all (Taioli and Garte, 2002). 2011; Terry, 2015). This supposition underlines the concepts of what
It is interesting to consider the implications of this level of complex- are currently termed “precision medicine” or “precision prevention”
ity for causal inference in cancer. Obviously, we now better understand (National Research Council, 2011; Terry, 2015). Some commentators
the biologic basis of action of long-​established carcinogens, such as have suggested that mechanistic understanding may ultimately render
smoking, chemotherapy, and various chemical agents. Mechanistic population studies irrelevant. However, it is instructive to consider the
explanations that relate particular exposures to specific types of cancer case of infectious diseases. Understanding the basic mechanisms of
increase confidence that an observed association between the exposure infectious disease has not eliminated the need to study disease patterns
and the incidence of that cancer is justifiably labeled “causal.” Such and causal factors on a population level, and the same will likely be
mechanistic understanding may also advance the possibility of early true for cancer (Nevins et al., 2003). There are many reasons to believe
detection and preventive interventions. Molecular “signatures” of spe- that cohort studies will be as important in the future as they are now,
cific exposures (e.g., p53 CpG hotspots) (Greenblatt et al., 1994) are albeit perhaps focused on different kinds of questions. To understand
identifying the relevant effects of certain exposures more precisely. It a mechanism after a cancer occurs is not the same as predicting it at
 103

Causal Inference in Cancer Epidemiology 103


some early stage of the process when the disease can be prevented. or publicity generated. While such concerns are not framed in causal
Prediction will necessarily be less than 100% accurate, and defining language, if an observed correlation is highly likely to have been
optimal groups for screening and early intervention will still require observed by chance, the question of cause is moot. Standards for the
population-​based data. Risk groups must be defined using far fewer conduct and reporting of virtually all biomedical research are evolving
factors than we know are operating at the molecular level, the latter to enhance the validity of statistical and causal inference.
being almost unique for an individual.
The molecular revolution in cancer may ultimately force a merg-
ing of two “schools” of determining causation: the probabilistic, risk-​ CONCLUSION
based model that the Hill criteria addressed and the more mechanistic,
deterministic models used for infectious disease, for which the Henle-​ It seems unlikely that the need for population-​based risk estimates and
Koch criteria were devised (Fredricks and Relman, 1996). It is interest- causal inference will disappear from cancer research, just as it has not in
ing that the Henle-​Koch criteria have proven to be inadequate, even for infectious disease. However, we are entering an era in which the relative
understanding viral carcinogenesis. These criteria were based on the strength of the “twin pillars” of causal inference—​knowledge derived
nineteenth-​century understanding of bacterial disease causation and from empirical, population-​based patterns and that based on understand-
are poorly suited for viral disease mechanisms or outcomes. Efforts ing of biologic mechanisms in individuals—​will tilt farther toward the
to refashion the Henle-​Koch criteria for the new era of molecular mechanistic end, requiring less proof from populations and more from
medicine (Fredricks and Relman, 1996; Vineis and Porta, 1996) have the laboratory. Despite this, the public health importance of the findings
shown that it is extraordinarily difficult to outline a set of experimental will continue to be based on the prevalence of exposure and magnitude
conditions that apply to all known pathogens—​not to mention new of risk. One challenge for causal inference in the future will be how best
pathogens with different mechanisms—​in order to provide a universal to integrate these various forms of evidence and how to assemble groups
framework for all new infectious diseases. with the sufficient interdisciplinary expertise to assess them.
In the case of viral carcinogenesis, the situation is even more com-
plex because the final disease is only partly a direct manifestation of an References
infectious process. Numerous viral agents have been linked to cancer Begley CG. 2013. Six red flags for suspect work. Nature, 497(7450), 433–​434.
with what is designated “sufficient” evidence of carcinogenicity (see PMID: 23698428.
Chapter 24 in this volume). In some cases, such as Epstein-​Barr virus Begley CG, and Ioannidis JP. 2015. Reproducibility in science: improving the
and human papillomavirus, the virus integrates directly into the host standard for basic and preclinical research. Circ Res, 116(1), 116–​126.
DNA. The immunosuppression caused by human immunodeficiency PMID: 25552691.
virus (HIV) increases the pathogenicity of other viruses such as HPV Brien GL, Valerio DG, and Armstrong SA. 2016. Exploiting the epigenome to
and HTLV1. Other microbial organisms, such as hepatitis B and C control cancer-​promoting gene-​expression programs. Cancer Cell, 29(4),
464–​476. PMID: 27070701.
viruses, bacterial infection with helicobacter pylori, and protozoal
Bunge M. 1959. Causality: the place of the causal principle in modern science.
infections such as schistosomiasis and liver flukes, create a continu- Cambridge, MA: Harvard University Press.
ing cycle of cell necrosis and repair that is associated with increased Collins FS, and Tabak LA. 2014. Policy: NIH plans to enhance reproducibility.
cancer risk in the affected tissues. Thus, there is no single molecular Nature, 505(7485), 612–​613. PMCID: PMC4058759.
finding that definitively implicates an infectious organism as a cause Doshi P, Goodman SN, and Ioannidis JP. 2013. Raw data from clini-
of cancer. The variety and complexity of mechanisms by which infec- cal trials:  within reach? Trends Pharmacol Sci, 34(12), 645–​ 647.
tious organisms increase cancer risk seem to defy a set of causal crite- PMID: 24295825.
ria based on any specific mechanism. Evans AS. 1993. Causation and disease:  a chronological journey.
New York: Plenum Medical.
Fredricks DN, and Relman DA. 1996. Sequence-​based identification of micro-
Multiplicity, Transparency, and Reproducible bial pathogens:  a reconsideration of Koch’s postulates. Clin Microbiol
Rev, 9(1), 18–​33. PMCID: PMC172879.
Research Glass TA, Goodman SN, Hernan MA, and Samet JM. 2013. Causal inference in
There has been increasing acknowledgment and concern about the public health. Annu Rev Public Health, 34, 61–​75. PMCID: PMC4079266.
validity of claims from biomedical research in general, and obser- Goodman SN, and Gerson J. 2013. AHRQ methods for effective health care
vational research in particular (Begley and Ioannidis, 2015; Collins mechanistic evidence in evidence-​based medicine:  a conceptual frame-
work. Rockville (MD): Agency for Healthcare Research and Quality (US).
and Tabak, 2014). Problems in cancer research have been identified at
Greenblatt MS, Bennett WP, Hollstein M, and Harris CC. 1994. Mutations in
almost all levels, from identification of therapeutic targets (Prinz et al., the p53 tumor suppressor gene: clues to cancer etiology and molecular
2011)  to alleged carcinogenic effects of common foods (Schoenfeld pathogenesis. Cancer Res, 54, 4855–​4878.
and Ioannidis, 2013) to genetic risk factors for cancer (Ioannidis and Greenland S. 1990. Randomization, statistics, and causal inference.
Khoury, 2011) and findings of modest cancer risk factors from stan- Epidemiology, 1(6), 421–​429.
dard case control or cohort studies (Tai et al., 2014). Greenland S, Pearl J, and Robins JM. 1999. Causal diagrams for epidemiologic
There are many sources of these problems, but one of the most research. Epidemiology, 10(1), 37–​48. PMID: 9888278.
common is that of multiplicity, which is particularly pernicious when Hernán MA, and Robins JM. 2016. Causal inference. Available from:  http://​
hidden. Problems with multiplicity occur when multiple exposures or www.hsph.harvard.edu/​miguel-​hernan/​causal-​inference-​book/​.
Hill AB. 1965. The environment and disease: association or causation? Proc R
risk factors are considered, multiple outcomes are assessed, multiple
Soc Med, 58, 295–​300.
models fit or analyses are implemented, and multiple experiments are Hume D. 1739. A treatise of human nature. London: Oxford University Press.
conducted. When statistical significance is a determinant of whether or Hussain SP, Hofseth LJ, and Harris CC. 2001. Tumor suppressor genes: at the
where a paper is submitted, which papers are published, which models crossroads of molecular carcinogenesis, molecular epidemiology and
are reported, which exposures, experiments, or outcomes are reported, human risk assessment. Lung Cancer, 34(Suppl 2), S7–​15.
and which analyses are presented, we in effect produce a potent scien- IARC. 2004. Tobacco smoke and involuntary smoking. IARC Monogr Eval
tific false-​positive machine with results highly unlikely to reproduce. Carcinog Risks Hum, 83, 1–1438. PMCID: PMC4781536.
Moreover, media publicity about the findings is often based on novelty, Ioannidis JP, and Khoury MJ. 2011. Improving validation practices in “omics”
rather than on reproducibility or scientific credibility. research. Science, 334(6060), 1230–​1232. PMID: 22144616.
Johnson S. 2007. The ghost map: the story of London’s most terrifying epi-
The solutions to this problem are diverse (Begley, 2013; Begley
demic—​ and how it changed science, cities, and the modern world.
and Ioannidis, 2015), ranging from raising the statistical standards of London, England: Riverhead Books.
evidence (Johnson, 2013) to complete disclosure of all analyses con- Johnson VE. 2013. Revised standards for statistical evidence. Proc Natl Acad
ducted, data sharing (Doshi et al., 2013), pre-​registration of observa- Sci U S A, 110(48), 19313–​19317. PMCID: PMC3845140.
tional studies, and modification of institutional incentive structures Kaufman JS, and Poole C. 2000. Looking back causal thinking in the health
that reward the quality of papers published rather than the quantity sciences. Annu Rev Public Health, 21, 101–​119.
104

104 Part I:  Basic Concepts


Last JM, Spasoff RA, and Harris SS. 2001. A dictionary of epidemiology, Sartwell PE. 1960. On the methodology of investigations of etiologic factors in
fourth edition. Am J Epidemiol, 154(1), 93–​94. chronic diseases: further comments. J Chron Dis, 11, 61–​63.
Lewis D. 1973. Counterfactuals. Cambridge, MA: Oxford University Press. Schoenfeld JD, and Ioannidis JP. 2013. Is everything we eat associated with
Lilienfeld AM. 1959. On the methodology of investigations of etiologic factors cancer? A systematic cookbook review. Am J Clin Nutr, 97(1), 127–​134.
in chronic diseases: some comments. J Chron Dis, 10, 41–​46. PMID: 23193004.
Links JM, Kensler TW, and Groopman JD. 1995. Biomarkers and mechanistic Schottenfeld D, Beebe-​Dimmer JL, Buffler PA, and Omenn GS. 2013. Current
approaches in environmental epidemiology. Annu Rev Public Health, 16, perspective on the global and United States cancer burden attributable
83–​103. PMID: 7639885. to lifestyle and environmental risk factors. Annu Rev Public Health, 34,
Little J, Bradley L, Bray MS, et al. 2002. Reporting, appraising, and integrat- 97–​117. PMID: PMC 23514316.
ing data on genotype prevalence and gene-​disease associations. Am J Schuemie MJ, Ryan PB, DuMouchel W, Suchard MA, and Madigan D. 2014.
Epidemiol, 156(4), 300–​310. PMID: 12181099. Interpreting observational studies:  why empirical calibration is needed
Magee B. 2001. The great philosophers: an introduction to Western philosophy. to correct p-​values. Stat Med, 33(2), 209–​218. PMCID: PMC4285234.
Oxford; New York: Oxford University Press. Slattery ML. 2002. The science and art of molecular epidemiology. J Epidemiol
Moore LE, Huang WY, Chung J, and Hayes RB. 2003. Epidemiologic con- Commun Health, 56(10), 728–​729.
siderations to assess altered DNA methylation from environmental expo- Smith MT, Guyton KZ, Gibbons CF et  al. 2015. Key characteristics of car-
sures in cancer. Ann NY Acad Sci, 983, 181–​196. cinogens as a basis for organizing data on mechanisms of carcinogenesis.
Morozova TV, Mackay TF, and Anholt RR. 2014. Genetics and genom- Environ Health Perspect, 124(6), 713–​721 PMID: 26600562.
ics of alcohol sensitivity. Mol Genet Genomics, 289(3), 253–​ 269. Stallones RA. 2015. The association between tobacco smoking and coronary
PMCID: PMC4037586. heart disease. Int J Epidemiol, 44(3), 735–​743. PMID: 26174518.
Mortensen HM, and Euling SY. 2013. Integrating mechanistic and polymor- Susser M. 1973. Causal thinking in the health sciences: concepts and strategies
phism data to characterize human genetic susceptibility for environmental of epidemiology. New York: Oxford University Press.
chemical risk assessment in the 21st century. Toxicol Appl Pharmacol, Susser M. 1977. Judgement and causal inference:  criteria in epidemiologic
271(3), 395–​404. PMID: 21291902. studies. Am J Epidemiol, 105(1), 1–​15. PMID: 318797.
National Research Council. 2011. Toward precision medicine:  building a Susser M. 1991. What is a cause and how do we know one? A  gram-
knowledge network for biomedical research and a new taxonomy of dis- mar for pragmatic epidemiology. Am J Epidemiol, 133(7), 635–​648.
ease. Washington, DC: The National Academies Press. PMID: 2018019.
National Research Council, Committee on Human and Environmental Exposure, Tai V, Grey A, and Bolland MJ. 2014. Results of observational studies: analysis
Science in the 21st Century, Board on Environmental Studies and Toxicology, of findings from the Nurses’ Health Study. PLoS One, 9(10), e110403.
and Division on Earth and Life Studies. 2012. Exposure science in the 21st PMCID: PMC4201544.
century: a vision and a strategy. Washington, DC: National Academies Press. Taioli E, and Garte S. 2002. Covariates and confounding in epidemiologic stud-
Nevins JR, Huang ES, Dressman H, et  al. 2003. Towards integrated clinico-​ ies using metabolic gene polymorphisms. Int J Cancer, 100(1), 97–​100.
genomic models for personalized medicine: combining gene expression PMID: 12115594.
signatures and clinical factors in breast cancer outcomes prediction. Hum Terry SF. 2015. Obama’s precision medicine initiative. Genet Test Mol
Mol Genet, 12(Spec No 2), R153–​R157. Biomarkers, 19(3), 113–​114. PMCID: PMC4361161.
Neyman J. 1990. On the application of probability theory to agricultural experi- US Department of Health and Human Services. 1986. The health consequences
ments: essay on principles (1923). Statist Sci, 5, 463–​572. of involuntary smoking:  a report of the Surgeon General. Washington,
Olsen J. 2003. What characterises a useful concept of causation in epidemiol- DC:  US Department of Health and Human Services, Public Health
ogy? J Epidemiol Commun Health, 57(2), 86–​88. PMCID: PMC1732370. Service, Office on Smoking and Health.
Pearl J. 2000. Causality:  models, reasoning and inference. Cambridge, US Department of Health and Human Services. 2006. The health consequences
UK: Cambridge University Press. of involuntary exposure to tobacco smoke:  a report of the Surgeon
Peng RD. 2011. Reproducible research in computational science. Science, General. Atlanta, GA:  US Department of Health and Human Services,
334(6060), 1226–​1227. PMCID: PMC3383002. Centers for Disease Control and Prevention, Coordinating Center for
Petersen ML, and van der Laan MJ. 2014. Causal models and learning from Health Promotion, National Center for Chronic Disease Prevention and
data: integrating causal modeling and statistical estimation. Epidemiology, Health Promotion, Office on Smoking and Health.
25(3), 418–​426. PMCID: PMC4077670. US Department of Health and Human Services. 2010. How tobacco smoke
Prinz F, Schlange T, and Asadullah K. 2011. Believe it or not: how much can cuases disease: the biology and behavioral basis for smoking-​atrributable
we rely on published data on potential drug targets? Nat Rev Drug Discov, disease: a report of the Surgeon General. Atlanta, GA: US Department of
10(9), 712. PMID: 21892149. Health and Human Services, Centers for Disease Control and Prevention,
Robins J. 1986. A new approach to causal inference in mortality stud- National Center for Chronic Disease Prevention and Health Promotion,
ies with a sustained exposure period:  application to control of the Office on Smoking and Health.
healthy worker survivor effect. Mathemat Model, 7(9–​12), 1393–​1512. US Department of Health and Human Services. 2014. The health consequences
PMID: ISI:A1986F488000016. of smoking—​50  years of progress:  a report of the Surgeon General.
Robins J. 1987. A graphical approach to the identification and estimation of Atlanta, GA:  US Department of Health and Human Services, Centers
causal parameters in mortality studies with sustained exposure periods. J for Disease Control and Prevention, National Center for Chronic Disease
Chron Dis, 40, 139S–​161S. Prevention and Health Promotion, Office on Smoking and Health.
Rosen G. 1993. A history of public health. Baltimore, MD:  Johns Hopkins US Department of Health, Education and Welfare. 1964. Smoking and
University Press. health:  report of the Advisory Commmittee to the Surgeon General of
Rothman KJ. 1976. Causes. Am J Epidemiol, 104(6), 587–​592. the Public Health Service (p. 386). Washington, DC:  US Government
Rothman KJ. 1986. Modern epidemiology. Boston: Little, Brown. Printing Office.
Rothman KJ, and Greenland S. 1998. Modern epidemiology. Vineis P, and Porta M. 1996. Causal thinking, biomarkers, and mechanisms of
Philadelphia: Lippincott-​Raven. carcinogenesis. J Clin Epidemiol, 49(9), 951–​956. PMID: 8780601.
Rubin DB. 1974. Estimating causal effects of treatments in randomized and Weed DL, Gorelic LS. 1996. The practice of causal inference in cancer epide-
nonrandomized studies. J Educ Psychol, 65(5), 688–​701. miology. Cancer Epidemiol Biomarkers Prev, 5(4), 303–​311.
Samet JM, Avila-​Tang E, Boffetta P, et al. 2009. Lung cancer in never smok- White C. 1990. Research on smoking and lung cancer: a landmark in the his-
ers:  clinical epidemiology and environmental risk factors. Clin Cancer tory of chronic disease epidemiology. Yale J Biol Med, 63(1), 29–​46.
Res, 15(18), 5626–​5645. PMCID: PMC3170525. PMCID: PMC2589239.
Samet JM, and Wang SS. 2000. Environmental tobacco smoke. In: Lippmann Yerushalmy J, and Palmer CE. 1959. On the methodology of investigations
M (Ed.), Environmental toxicants:  human exposures and their health of etiologic factors in chronic diseases. J Chronic Dis, 10(1), 27–​40.
effects (Vol. 2, pp. 319–​375). New York: Van Nostrand Reinhold. PMID: 13664755.
 105

II
THE MAGNITUDE OF CANCER
 107

8 Patterns of Cancer Incidence, Mortality, and Survival

AHMEDIN JEMAL, D. MAXWELL PARKIN, AND FREDDIE BRAY

OVERVIEW encompass access to cancer care, quality of care, quality of life, and
cost of care (Glaser et al., 2005; Hiatt et al., 2015; Howe et al., 2003;
The global burden of cancer is expected to increase from 14.1  mil- Wingo et al., 2005).
lion newly diagnosed cases and 8.2 million cancer deaths in 2012 to Over the last 20  years, PBCRs have become the cornerstone of
22 million cases and 13 million deaths in 2030. This increase, based on cancer surveillance and control (Shin et al., 2007). Surveillance data
projected population aging and growth, will disproportionately affect are used to monitor trends, guide resource allocation, forecast future
low-​and middle-​income countries (LMICs), where large numbers of needs (Bray and Moller, 2006; Hiatt and Rimer, 1999; Storm, 1996),
young adults are now surviving to older ages, when cancer becomes and evaluate the impact of preventive and curative interventions
common. The future cancer burden is likely to be even higher than (Parkin, 2008).
predicted from demographic changes alone, because of the ongoing This chapter describes international patterns of cancer incidence,
adoption of Westernized patterns of diet, physical inactivity, delayed mortality, and survival as estimated by GLOBOCAN 2012, a com-
reproduction, and cigarette smoking. The incidence of cancers tra- prehensive profile of the risks and burden from cancer in 184 coun-
ditionally associated with Western behavioral, environmental, and tries for the year 2012 (http://​globocan.iarc.fr). It describes temporal
cultural factors (breast, colorectum, lung, and prostate) are increas- trends in cancer incidence based on the CI5 series (Ferlay et al., 2013)
ing in LMICs, whereas cancers caused at least partly by infectious and mortality data from the World Health Organization (WHO). We
agents (stomach, liver, uterine cervix) are decreasing. The timing and begin by discussing several basic concepts in cancer surveillance, par-
pace of this epidemiologic transition varies by region; however, many ticularly the distinction between risk (the average probability of being
LMICs continue to bear a large burden of infection-​related cancers. diagnosed with or dying from cancer in a designated time period), and
Furthermore, survival after a diagnosis of cancer tends to be consider- burden (the total number of newly diagnosed cases, cancer deaths,
ably lower in LMICs because of late stage of diagnosis and a lack of, prevalent cases, and/​or economic costs in the population). Other terms
or limited availability of, standard treatments. are defined as they appear in the text. The methods used to develop
In high-​income countries, the incidence rates of the most common GLOBOCAN 2012 are described only briefly here, since they are
cancers are either stabilizing at a high level or decreasing, depending documented extensively elsewhere (Ferlay et al., 2015). Unlike the
on the underlying risk factors and the extent to which early detection chapters on cancer surveillance in previous editions of this text (Parkin
and screening approaches are utilized. The over-​diagnosis of certain and Bray, 2006; Ries and Devesa, 2006), we do not discuss cancer
tumors adds to the cancer burden in affluent countries, yet mortality surveillance in the United States separately from the global picture,
rates are decreasing for many sites due to a combination of prevention nor do we present detailed information on individual cancer sites. Site-​
and improved treatment. specific information can be found in Part IV of this text, “Cancers
Population-​based cancer registries (PBCRs) are central to cancer by Tissue of Origin.” Detailed statistical data for the United States
surveillance and control. These registries now cover over 95% of the are widely available online from major cancer research organizations
population in North America, but less than 10% of the populations (Centers for Disease Control and Prevention [CDC], 2016; National
of South America and Africa. There is an urgent need to expand the Cancer Institute [NCI], 2016; the American Cancer Society [ACS],
coverage and improve the quality of cancer registration and death cer- 2016), and are updated more frequently than is possible in this text.
tification in LMICs. Equally, there is a need to attract and train young
investigators in the uses of surveillance data for cancer control.
GENERAL CONCEPTS

INTRODUCTION Distinction Between Risk and Disease Burden


Population-​based cancer registries (PBCRs) provide information on As noted in the preceding, risk represents the average probability that
newly diagnosed cancer cases, deaths, and survival in geographically individuals in a population will be diagnosed with or die from some
well-​defined populations (Parkin, 2008). Because the target popula- specified form of cancer during a defined time interval. Conceptually,
tion is defined by place of residence rather than the location of treat- risk is probabilistic; it can be defined only in a group of people, not
ment, PBCRs avoid the potential for referral biases that affect clinical a single individual. The average annual risk of developing or dying
series and, to some extent, hospital-​based registries (Ries and Devesa, from cancer can be approximated as an incidence or mortality rate per
2006). The first PBCRs were established in North America in 1935 100,000 people per year for adult cancers, and per million per year for
(Connecticut Department of Public Health [CDPH], 2016), and in childhood cancers. While the average risk in a population is inevitably
Nordic countries in the 1940s and 1950s (Wagner, 1985). Since then, a higher or lower than that of nearly all individuals, the incidence and
network of registries, the International Association of Cancer Registries mortality rates can be standardized or stratified by age to allow valid
(IACR; http://​www.iacr.com.fr) has developed as a professional orga- comparisons between populations that differ in size or age structure.
nization with over 500 members worldwide. At present, 291 registries The rates are calculated by dividing the number of events in a given
in 68 countries provide high-​quality data, as evaluated and compiled in year by the number of people at risk. The methods of standardizing or
the most recent volume of Cancer Incidence in Five Continents (CI5; otherwise controlling for the age of the population are described at the
http://​ci5.iarc.fr). Depending on their resources, PBCRs monitor can- end of this chapter.
cer occurrence by anatomic site, histologic subtype, stage at diagnosis, Unlike risk, disease burden represents the absolute numbers (or
and survival. In high-​income countries the data can be linked to other counts) of incident cases, deaths, cancer survivors, and economic
administrative databases, thereby broadening the scope of research to costs in a population, regardless of its size or age composition. The

107
108

108 Part II: The Magnitude of Cancer


number of incident cases and deaths correspond to the numerator in cancer occurring may change with economic development. The so-​
calculations of incidence and mortality rates. These absolute numbers called epidemiologic transition involves an increase in the incidence
provide a partial indication of the human costs of cancer in the popula- rate of cancers traditionally associated with Western lifestyles (breast,
tion. They do not represent risks and should not be used to estimate colorectum, lung, and prostate), and a decrease in cancers caused
risk in different populations or time periods. The American Cancer by infectious agents (stomach, liver, uterine cervix), and usually an
Society (ACS) publishes estimates of the number of newly diagnosed increase in the all-​cancer incidence rate. The transition occurs at a dif-
cancer cases and deaths anticipated in the current year in the United ferent pace in different countries, reflecting local differences in culture
States (Siegel et al., 2016). These represent estimates rather than actual and the physical and social environment.
counts, since the data for the current year do not become available at
the national level until several years later. The ACS estimates are pro- Economic Development
jections, based on statistical modeling of the observed number of cases Economic and social development has complex effects on the risks
and deaths in previous years (Chen et al., 2012; Zhu et al., 2012). For and burden from cancer. Improvements in sanitation, nutrition, vacci-
2016, the ACS estimated that about 1.7 million new cancer cases and nation, and treatment can all reduce the occurrence of cancers caused
almost 600,000 cancer deaths would occur in the United States (Siegel by infectious agents (stomach, liver, uterine cervix, Kaposi sarcoma),
et al., 2016). as mentioned earlier. Economic development can also prevent severe
nutritional deficiencies, which likely contribute to the incidence of
certain cancers, such as squamous cell carcinoma of the esophagus
Cancer Survival and Prevalence in Asia and Africa (Schneider et al., 2007). Exposure to occupational
The terms used to describe cancer survival and prevalence are in cer- and environmental pollutants, such as asbestos, arsenic, and indoor
tain respects more ambiguous than incidence and mortality rates, since air pollution, at first increase and then decrease with economic and
it is often unclear whether a cancer has been cured or is merely in social development (Bray et al., 2015b). Some forms of early detec-
remission. The NCI program of Surveillance, Epidemiology and End tion reduce the incidence and mortality rates from specific cancer sites.
Results (SEER) in the United States uses several measures of preva- Treatment improves survival. These benefits can be offset, however,
lence. Limited duration prevalence signifies the proportion of people by urbanization and increased exposure to Westernized patterns of
alive at a designated date diagnosed with cancer within a specified diet, physical inactivity, delayed reproduction, and cigarette smoking.
time period in the past (e.g., 5, 10, or 15  years). This chapter uses Like occupational exposures, cigarette smoking first increases with
the definition adopted by the International Agency for Research on economic development, but then decreases with social development.
Cancer (IARC) in GLOBOCAN, with 5-​year prevalence estimating Over-​diagnosis inflates the incidence rate of screening-​detected cancer
the number of cancer patients diagnosed with the disease within the of the prostate, breast, and thyroid, among others.
last 5 years and alive in 2012. Complete prevalence describes the pro-
portion of people alive who were ever diagnosed with cancer, regard- Screening
less of when this occurred (SEER, 2015). The effects of screening on cancer risk and burden are equally com-
GLOBOCAN 2012 does not use the term relative survival, although plex (see Chapter 63). Screening for certain cancers (e.g., cervical and
this is mentioned in many chapters of this book. Relative survival sig- colorectal) decreases both incidence and death rates by detecting and
nifies the ratio between the proportion of cancer patients who have leading to the removal of premalignant lesions and early stage can-
survived for a specified time period after diagnosis, divided by the cers. These tests reduce both risk and burden. In contrast, screening for
percentage of survivors of the same age who have not been diagnosed breast cancer and lung cancer decreases mortality from these cancers,
with cancer. This term is often referred to erroneously as a rate. but at the cost of increasing other aspects of burden (e.g., incidence
and unnecessary treatment). Screening for prostate cancer provides
marginal benefits in terms of a reduction in mortality for men at aver-
age risk, but dramatically increases incidence and overestimates sur-
Factors That Affect Cancer Risk and Burden vival. The distorting effects of prostate cancer screening on incidence
and mortality rates are discussed later in the chapter. The effects of
Population Aging screening with prostate specific antigen (PSA) on relative survival is
Attained age is a strong risk factor for cancer. The overall incidence equally large, since periodic screening advances the time of diagnosis
rate from cancer more than doubles with each 10-​year increase in while differentially detecting indolent tumors rather than cancers that
attained age. Consequently, the number of people affected by, or dying grow rapidly. This creates the appearance that screening has improved
from, cancer (disease burden) increases as the population ages. The survival, even when it has not (Chapter 63). When routine PSA screen-
influence of the age distribution of the population on incidence (and ing was first introduced in the United States and was recommended
mortality) rates must be taken into account when comparing risk in for all men over age 50 years, the 5-​year relative survival for prostate
two or more populations (see discussion of methodologic issues later cancer increased from 68% (for those diagnosed in the mid-​1970s), to
in this chapter). In general, age-​standardized and age-​specific rates are nearly 100% (for those diagnosed in 2004–​2010) (Siegel et al., 2013).
independent of the age structure and size of the population, whereas The decrease in the mortality rate was considerably smaller (Welch
the burden increases with population aging and growth. Growth and and Albertsen, 2009; Welch and Black, 2010).
aging of populations have an especially large effect on the future
cancer burden in economically developing countries, because of the Over-​diagnosis
large population of young adults who are now surviving into older age Over-​diagnosis refers to the detection of small and/​or indolent cancers
groups where cancer becomes common. during screening or diagnostic evaluation that otherwise would not
have become clinically evident during the patient’s lifetime (Parkin
Risk Factors and Moss, 2000). This tends to affect cancers that may remain indolent
Changes in the prevalence of risk factors that increase or decrease or latent for long periods of time (e.g., prostate, thyroid, some breast
tumorigenesis directly affect both the risk and disease burden from cancers, kidney, lung, and melanoma) (Cook et  al., 2009; Fontana
cancer. Many risk factors are associated with economic develop- et al., 1991; Welch and Black, 2010) and, because it is a consequence
ment, as discussed in the following subsection. People who live in of population screening, is more of a problem in high-​income coun-
low-​income countries have greater exposure to oncogenic infections tries than in LMICs. It has been estimated that over-​diagnosis from
such as Helicobacter pylori, hepatitis B virus (HBV), untreated human screening may account for as many as 40% of newly diagnosed pros-
immunodeficiency virus (HIV), parasitic disorders, and so on (see tate cancer cases and 10%–​30% of incident breast cancer cases in the
Chapter  24), but less exposure to factors associated with economic United States and several other Western countries (Bleyer and Welch,
development (manufactured cigarettes, excessive caloric intake, physi- 2012; Etzioni et  al., 2002; Loeb et  al., 2014; Vickers et  al., 2014).
cal inactivity, and delayed reproduction). In consequence, the types of Over-​diagnosis can affect trends in mortality rates as well as incidence,
 109

Patterns of Cancer Incidence, Mortality, and Survival 109


due to attribution bias (Feuer et  al., 1999). The main problem with migration parallel the changes that occur within countries undergoing
over-​diagnosis, however, is the increasing number of people who are economic development. In both settings, the changes in risk are pre-
diagnosed with cancer and who may receive unnecessary treatment sumed to reflect change in “environmental” (i.e., acquired) exposure,
(Vaccarella et al., 2016). rather than in inherited genetic susceptibility (Bray and Parkin, 2014).
Although migrant studies can be biased by selection for factors that
Under-​diagnosis enable migration (“healthy migrant effect”) or by differences in the
Under-​diagnosis of cancer occurs more often in LMICs than in afflu- completeness of cancer detection between the country of origin and
ent countries. Under-​ diagnosis occurs because of limited medical the host country (Parkin and Khlat, 1996), they provide important evi-
resources and infrastructure, lack of awareness (education), and bar- dence that factors other than inherited genes strongly affect cancer risk
riers to care. It is more of a problem for cancers that require sophisti- in human populations.
cated diagnostic tests (e.g., pancreas and ovary) than for sites that are
more easily diagnosed.
GLOBAL CANCER PATTERNS, 2012

Methodologic Issues This section discusses global and regional patterns of cancer incidence,
mortality, and survival, based on the estimates of GLOBOCAN 2012
(http://​globocan.iarc.fr) and longitudinal data from Cancer Incidence
Age Adjustment in Five Continents (http://​ci5.iarc.fr/​). It also considers cancer risk and
As the incidence and death rates of many forms of cancer increase
burden in relation to Human Development Index (HDI) designations
exponentially with attained age, it is necessary to standardize or strat-
for 172 countries. The methods used to make the GLOBOCAN 2012
ify rates according to age groups when comparing populations with
estimates are described briefly at the end of the chapter.
different age structures. The approach used most commonly in sur-
veillance data is direct age standardization. Age-​standardized rates
summarize the average rates in two or more populations by weighting Human Development Index (HDI)
each age group to the age distribution of a standard population. This is
essentially the hypothetical rate that would prevail if all of the popula- The Human Development Index (HDI) is as a composite measure of
tions being compared had the same age distribution as the standard economic and social development, based on life expectancy, educa-
population. The actual structure of the standard is of no consequence tion, and gross domestic product per head (http://​hdr.undp.org/​en/​con-
for comparative purposes (Bray et al., 2002), but of course the same tent/​human-​development-​index-​hdi). A map of countries in relation to
standard population must be used for all of the populations being com- quartiles of HDI (Figure 8–​1) shows the geographic distribution of
pared. Nevertheless, different organizations and countries use different this index in 2012. Most low HDI countries were concentrated in sub-​
standard populations when reporting incidence and mortality rates. In Saharan Africa. Medium-​HDI countries spanned much of Asia, includ-
order to allow comparisons between different populations, and over ing China and India, and parts of Africa and Latin America. High HDI
time, IARC continues to use the World Standard (Doll and Cook, countries predominated in Eastern Europe, the former Soviet Union,
1967) to compare international rates. In the United States, health agen- northern Africa, Mexico, and northern South America. Very high HDI
cies have historically used the national age distribution as the standard, countries included Japan, Australia, Argentina, Chile, several Gulf
but have shifted this in more recent years as the population has aged States, and countries in Western Europe and North America.
(Klein and Schoenborn, 2001). In most public and private health agen-
cies in the United States, the national age distribution in 1940 was used
until 1970; it then shifted to the 1970 distribution until 1980; the 1980 Geographic Distribution of Cancer Burden
distribution until 1999, and the 2000 US population thereafter (Klein
and Schoenborn, 2001).
All Sites Combined
An alternative measure that does not require choice of an arbi-
Our analyses of all cancer sites combined exclude non-​melanoma skin
trary or varying standard is the cumulative rate (or risk) of develop-
cancer. Figure 8–​2 shows the global distribution of the cancer bur-
ing (or dying from) cancer by a selected upper age limit (e.g., 65, 75,
den by geographic region for all anatomic sites combined in 2012.
85) (Day, 1992).
Asia accounted for nearly half (48.0%) of the 14.1 million newly diag-
nosed cases in that year, and more than half (55%) of the 8.2 million
Enumeration Errors cancer deaths. Asia contributed a smaller proportion of cancer cases
Errors in enumerating cases, deaths, and/​or the population at risk can
and deaths than might be expected, given that it comprises 60% of
also bias measurements of cancer risk and burden. This can occur for
the global population. Moreover, Asia accounted for only 40% of all
several reasons. Incomplete ascertainment or changes in tumor clas-
prevalent cases, reflecting the generally poor survival and case mix
sification can bias numerator data. Inconsistent approaches to the
of cancer patients in middle-​and low-​resource countries. In contrast,
classification of multiple primary tumors can be especially problem-
Europe, which accounts for only 10% of the world population, con-
atic (Chapter 60). In the United States, delayed reporting of recently
tributed 24% of incident cases, 21% of cancer deaths, and 28% of
diagnosed cases causes an undercount of cancers diagnosed in non-​
prevalent cases.
hospital settings during the previous 1–​3  years (Clegg et  al., 2002).
For example, leukemia is underestimated by about 14% in the most
Specific Anatomic Sites
recent years, and prostate cancer by about 4% (NCI, 2015). Changes in
In analyses of men and women combined, a relatively small number
a population between census counts due to migration or other factors
of anatomic sites accounted for more than half of the total number of
can cause estimates based on interpolation to over-​or underestimate
incident cases, cancer deaths, and prevalent survivors in 2012 (Figure
the population at risk (Ward et al., 2005).
8–​3). Lung and colorectal cancer were among the four most common
sites for all three measures. Lung cancer ranked first for incidence and
Migrant Studies mortality, but fourth for prevalence. Breast cancer ranked first for prev-
alence, second for incidence, but fifth for mortality.
Classic studies have measured the risk of specific types of cancer
among migrants who move from a high-​to low-​risk country, or vice Five Most Common Cancer Sites by Gender
versa, and have compared this to the risk in their country of origin and Figures 8–​4 and 8–​5 show the five most common cancer sites in men
in the host country (Haenszel, 1961). Within one or two generations, and women separately in terms of incident cases, cancer deaths, and
the risk of colorectal, stomach, or breast cancer among immigrants prevalent cases (IARC, 2016). These rankings are not stratified by HDI
approaches that of the host country. The differences in risk following level. In men, cancers of the lung, prostate, and stomach rank among
10

110 Part II: The Magnitude of Cancer

Human Development Index


Low HDI
Medium HDI
High HDI
Very High HDI

No data Not applicable


The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever
on the part of the World Health Organization concerning the legal status of any country, territory, city or area or of its authorities,
or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps represent approximate border lines
for which there may not yet be full agreement.

Figure 8–​1.  Countries according to Human Development Index (HDI), 2012. Source: World Health Organization (WHO).

the top five sites for incident and prevalent cases and cancer deaths. as 1915, Hoffman reported a 10-​fold difference in the death rate
In women, cancers of the breast, colorectum, and uterine cervix are from breast cancer between Japanese and British women (Hoffman,
consistently among the most common. For all three measures, the five 1915; Muir, 1996). The regional differences in incidence rates
most common sites account for more than half of the total. In terms of exceed 25-​fold for cancers of the esophagus, nasopharynx, and
prevalence, thyroid cancer ranked among the five most common can- prostate (IARC, 2016), and approach 10-​fold for cancers of the
cers in women, reflecting over-​diagnosis from screening and the low breast, colorectum, lung, and liver. These differences are larg-
fatality rate of screen-​detected thyroid cancers. est when examining smaller areas within the WHO regions. For
example, Burkitt lymphoma in children is rare, except in endemic
Geographic Distribution of Cancer Risk malarial regions of equatorial Africa (Chapter 40). Nasopharyngeal
Striking geographic differences have long been observed in the cancer is rare throughout most of the world, but is very common in
incidence and mortality rates of certain types of cancer (Muir, the high-​risk regions of southern China and parts of East and South
1996; Parkin and Bray, 2006; Ries and Devesa, 2006). As early Africa (Chapter 26).

Incidence Mortality Prevalence (5 years)

Africa
Africa 5.5%
6.0% North North America
Africa America 16.3%
North 7.2% 8.4%
America Latin America &
Latin America &
12.7% the Carribean
the Carribean Latin America
7.4%
7.8% & the Carribean
8.1%
Asia Europe Asia
Asia 21.4%
48.0% Europe 54.9% 40.5%
24.4%
Oceania Europe
0.7% 28.2%

Oceania
1.1%

Oceania
1.4%

14.1 million 8.2 million 32.5 million


New cases Deaths Persons

Figure 8–​2.  Regional distribution of newly diagnosed cancer cases, deaths, and survivors worldwide, 2012. Source: GLOBOCAN (2012).
 1

Incidence Mortality Prevalence (5 years)

Breast
Lung 19.2%
13.0% Other
Lung 31.0%
Other Breast Other 19.4%
35.3% 11.9% 32.0% Liver Prostate
9.1% 12.1%
Colorectum
9.7%
Cervix uteri 3.2% Stomach
Prostate 3.7% 8.8%
Bladder 3.1% Pancreas 4.0% Colorectum Thyroid Colorectum
Prostate
Esophagus 3.2% 7.9% Esophagus Breast 8.5% 3.7% 10.9%
4.9% 6.4%
Cervix uteri 3.7% Liver Stomach Corpus uteri Lung
5.6% 6.8% 4.1%
Cervix 5.8%
Bladder uteri
4.1% 4.8%
Stomach
5.8%

14.1 million 8.2 million 32.5 million


New cases New cases Persons

Figure 8–​3.  Percentage contribution to commonly diagnosed cancer cases and deaths and prevalent cases worldwide, 2012. Source: IARC.

Prevalence
Incidence Mortality (5 years)

Lung
Lung Prostate

Prostate 40.3%
42.4% Liver 40.5%
57.6% Colorectum
59.5%
59.7%
Colorectum Stomach Lung
Stomach Colorectum Stomach
Liver Prostate Bladder

7.4 million 4.7 million 15.3 million


New cases Deaths Persons

Figure 8–​4.  Five most common cancer sites worldwide in men. Source: IARC.

Prevalence
Incidence Mortality (5 years)

Breast
Breast
Breast

Lung 32.9%
44.2% 47.8%
Colorectum 55.8% 52.2% 67.1%

Colorectum Colorectum
Lung
Cervix uteri
Cervix uteri
Cervix uteri Corpus uteri
Stomach Thyroid
Stomach

6.7 million 3.5 million 17.2 million


New cases Deaths Persons

Figure 8–​5.  Five most common cancer sites worldwide in women. Source: IARC.


12

112 Part II: The Magnitude of Cancer


The large international differences in cancer risk, and changes in countries), and lung and stomach cancer, for which the number of
risk among migrants who move from one country to another, have led prevalent cases was similar.
to estimates of the preventability of cancer. The landmark study by
Doll and Peto in 1981 estimated that approximately 80% of cancer Temporal Trends in Site-​Specific Cancer Risk
deaths in the United States were preventable “in principle” (Doll and by Level of HDI
Peto, 1981). This is supported by the large temporal changes in the Figures 8–​7 through 8–​10 show the temporal trends in incidence and
incidence of specific cancer sites occurring within countries. mortality rates for four cancer sites in relation to the level of HDI.
Decreasing incidence and mortality rates are observed for cancers
Cancer Risk and Burden by Level of HDI of the stomach (Figure 8–​7) and uterine cervix in most populations
Whereas the annual risk (per 100,000) of being diagnosed with cancer (Figure 8–​8), whereas increasing rates are widely observed for cancers
is about 80% higher in countries designated as high/​very high HDI of the breast (Figure 8–​9) and colorectum (Figure 8–​10).
(240.6) than in low/​medium HDI countries (135.8), the latter account
for more than half of all cases globally. This reflects the large constit- Relation of 15 Cancer Sites to Level of HDI
uent populations of low and medium HDI countries, which in 2012 The relationships between the incidence and death rates from specific
included China and India. The burden from cancer is higher and is cancer sites and the level of HDI are also evident cross-​sectionally
increasing more rapidly in LMICs than in high-​and very-​high-​income in GLOBOCAN 2012 (Figure 8–​ 11). This figure shows the age-​
countries. Figure 8–​6 shows the anticipated shift in the cancer bur- standardized incidence rates of the 15 most common types of cancer
den from developed to developing countries. From 2012 to 2035, worldwide in men (10 sites) and women (12 sites) in relation to the
the proportion of all cancers accounted for by LMICs is projected to level of HDI. The data from China and India are shown separately
increase from 57% to 65% (IARC, 2016). The future cancer burden from other medium HDI countries because of their massive popula-
is likely to be even larger than predicted from demographic changes tions (1.35 and 1.25 billion inhabitants, respectively). Incidence rates
alone, because the incidence rates of cancers that are high in Western rise with increasing HDI for lung, prostate, and colorectal cancer in
countries and traditionally associated with factors linked to lifestyle men and for breast and colorectal cancer in women. The relationships
and environment within these countries (breast, colorectal, prostate, between HDI and the incidence of other cancer sites are inconsist-
lung) are increasing rapidly in many LMICs, as discussed later in the ent. Figure 8–​11 also illustrates that the incidence rate of lung cancer
chapter. in 2012 among Chinese women was as high as that in high and very
The cumulative risk of cancer between ages 0–​74  years is lowest high HDI countries, despite the low prevalence of cigarette smoking.
in low HDI countries in Western Africa (9.8%) and Middle Africa Factors that affect lung cancer risk in Chinese women are discussed
(10.9%) and in medium HDI countries in South Central Asia (10.7%), later in the chapter. The incidence rate of lung cancer among Chinese
and highest in very high-​income countries such as Australia and New men was about four times higher than that among men in India, and
Zealand (30.7%), North America (30.9%), Western Europe (29.1%), was higher than that of any of the HDI groupings.
and Northern Europe (27.6%) (IARC, 2016).
Stomach Cancer.  A decrease in non-​gastric cardia cancer inci-
Cancer Mortality Risk and Burden by Level of HDI dence and mortality rates has occurred in nearly all countries in the
There was less variation in overall cancer mortality rates than in inci- last 30 years (Figure 8–​7). The rate of decrease has been remarka-
dence rates in relation to HDI level. When expressed as cumulative bly similar in men and women, in high-​risk countries such as Japan
risk of death from cancer by age 74, the probabilities in GLOBOCAN and low-​risk countries such as Denmark (Ferlay et  al., 2013), and
2012 were 9.0% in economically developed countries and 8.1% in across a wide range of HDI levels. The decrease has been observed
developing countries. The generally poor survival of patients with can- in mortality rates in the United States since 1930 (Paik et al., 2001).
cer in LMICs partially offsets their lower incidence rates. The age-​standardized incidence rate has decreased by more than
For prevalent cases, countries designated as high or very high HDI 80% in parts of the United States since 1950 (Siegel et  al., 2016).
account for the majority of cases of cancers of the breast, prostate, The reasons for the decreasing rates are not known conclusively,
colorectum, urinary bladder, uterus, thyroid and kidney, as discussed but are postulated to involve improvements in sanitation, hygiene,
later. Exceptions to this were cancers of the uterine cervix (which food preservation, and antibiotics (Howson et al., 1986). The fortu-
were more common in low and medium than in high or very high HDI itous decrease in gastric cancer parallels temporal decreases in the
prevalence of Helicobacter pylori, the principal cause of non-​cardia
stomach cancer (Chapter 31), and coincides with the global spread
Estimated Cases Projected Cases of refrigeration that made curing and salting foods obsolete in most
(millions) parts of the world. The decrease in gastric cancer that occurs within
countries undergoing economic development parallels the decrease
2012 2025 2035 observed among migrants who move from high-​risk areas such as
Japan to lower risk areas such as the United States (Locke and King,
1980). Nevertheless, gastric cancer remains a common cancer site
globally, ranking among the top four incident cancers among men,
57% and fifth among women.
61% 65%
Cervical Cancer. Although less common than stomach cancer,
cancer of the uterine cervix remains the most commonly diagnosed
14.1 19.3 24.0 cancer among women in 39 of the poorest countries, and the leading
cause of cancer death among women in 19 countries in sub-​Saharan
Africa. The incidence and death rates are decreasing in most countries
Developed regions (Figure 8–​8), but the incidence rates remain approximately twice as
high and the mortality rates three times higher in countries designated
Developing regions low/​medium HDI than in those designated high/​very high HDI. The
decreases in higher-​income countries largely parallel the introduction
of effective screening programs, and have occurred despite high preva-
Figure 8–​6.  Shift in cancer burden from developed to developing c­ ountries. lence of persistent human papillomavirus (HPV) infection (Vaccarella
Source: GLOBOCAN (2012). et al., 2014). The decreases in some lower income countries are thought
 13

Patterns of Cancer Incidence, Mortality, and Survival 113


Low/Medium HDI High/Very high HDI
80 80

60 60

50 50
Japan*
Age-standardized (world) incidence rate per 100,000, males

Age-standardized (world) incidence rate per 100,000, males


40 40

30 30
China
25 Colombia*
25

20 Japan 20
China* Denmark
France Colombia
15 15
Philippines*

Australia
10 10
India* USA*:
USA*: France* Black
Uganda* White
7 Thailand* 7
Australia
USA:
5 Black 5
Philippines Denmark
4 4

3 3

USA:
White
2 2

1980 1990 2000 2010 1980 1990 2000 2010


Year

Incidence Mortality

Figure 8–​7.  Trends in stomach cancer incidence and mortality rates for select countries, 1975–​2012. Source: IARC.

to result from improved genital hygiene (Dhillon et al., 2011), delayed in those designated as low/​medium HDI. In general, the incidence
exposure to HPV, and some limited implementation of screening. rates in countries with high-​quality population-​based cancer regis-
tries increase with rising levels of HDI (Bray et al., 2012; Center and
Female Breast Cancer.  Breast cancer is the most common cancer Jemal, 2011). However, a downturn in incidence rates has occurred in
among women worldwide. It is also the most common cause of cancer the United States (in both whites and blacks) and France, and a level-
death among women in less developed regions and the second most ing off in Australia. The downturn in the United States is attributed
common cause of cancer death (after lung cancer) in women in more to the increased uptake of screening and the detection and removal of
developed regions. Figure 8–​9 shows the temporal trends in incidence premalignant colorectal lesions (Siegel et al., 2014). The stabilization
and mortality rates from breast cancer in relation to HDI. The incidence or decrease in incidence rates in younger birth cohorts in high-​income
rates are increasing in most countries, but have reached a plateau in some countries is thought to reflect changes in dietary and activity patterns
high/​very high HDI countries, where mortality rates are now decreasing. in recent decades.
The incidence rates still vary by a factor of nearly four across the world Mortality rates from colorectal cancer are substantially lower than
regions. Cumulative risk between ages 0–​74 years in 2012 was highest in incidence rates. The mortality rates are increasing in low/​medium
North America (10%) and other high-​income countries (Western Europe, HDI countries, but decreasing in all high/​very high HDI countries
Canada, and Australia). Cumulative risk was lowest in Central Africa and except for Japan (Figure 8–​10). The increase in incidence rates began
East Asia (2.7%). The variation in mortality rates across world regions later and has been larger in Japan than in other affluent countries. For
was less than the variation in incidence rates, since it reflects differences example, the incidence rate (per 100,000) of colorectal cancer among
in treatment as well as underlying occurrence. The cumulative risk of men in the Myagi prefecture in Japan increased from 19.7 in 1979 to
breast cancer death before age 75 in 2012 was lowest in East Asia (0.6%) 61.6 in 2005, surpassing that of other high/​very high HDI countries
and highest in West Africa (2.1%). (Center et al., 2009).

Colorectal Cancer. Colorectal cancer is the third most com- Lung Cancer.  The geographic and temporal patterns of lung cancer
mon cancer in men and the second most common cancer in women. are discussed in Chapter 28. In absolute terms, the variations in lung
Regional incidence rates vary by a factor of 10 in both sexes. cancer risk during the last century have been larger than for any other
Figure 8–​10 shows the increase in incidence rates in most countries anatomic site. In 2012, lung cancer was the most frequently diagnosed
worldwide, with higher rates in high/​very high HDI countries than neoplasm among men in 38 countries and the leading cause of cancer
14

114 Part II: The Magnitude of Cancer


Low/Medium HDI High/Very high HDI

50 50
Uganda*
40 40
35
Age-standardized (world) incidence rate per 100,000, females

30

Age-standardized (world) incidence rate per 100,000, females


30
25 25
Thailand*
20 Colombia* 20
Philippines*
Japan*
15 India* 15

Australia Denmark
10 10
USA: Colombia
White
7 France* 7
6 USA:
China* 6
Black
5 5

4 Denmark 4
Philippines

3 USA*: 3
China Black
Japan
2 2
USA*:
1.5 Australia White 1.5
France

1 1
1980 1990 2000 2010 1980 1990 2000 2010
Year
Incidence Mortality

Figure 8–​8.  Trends in cervical cancer incidence and mortality rates for select countries, 1975–​2012. Source: IARC.

death in 91 countries (Figures 8–​12 and 8–​13). The incidence and accounted for more than half of all incident cases of cancers of the
mortality rates were highest among men in Eastern Europe and much lung, breast, colorectum, prostate, urinary bladder, and non-​Hodgkin
of Asia, including China and Indonesia. Among women, lung cancer lymphoma, notwithstanding the much smaller populations in these
was the most commonly diagnosed cancer only in China, but was the countries. Low/​medium HDI countries accounted for the majority of
leading fatal cancer in 25 countries, predominantly in North America, cancers of the stomach, liver, uterine cervix, and esophagus.
Northern Europe, Australia and New Zealand, and China (Figures In terms of cancer deaths, the low/​medium HDI countries accounted
8–​14 and 8–​15). More than half of all cases occur in economically for more fatal cancers of the lung, liver, stomach, esophagus, and uter-
developing countries where smoking remains common, especially ine cervix than the high/​very high HDI countries. The latter accounted
among men. for more deaths from cancers of the colorectum, pancreas, and pros-
Except among women in China, lung cancer rates overwhelm- tate. These two groups were approximately equal for deaths from can-
ingly reflect lifetime and recent cigarette smoking. Smoking cessation cers of the breast and leukemia. In terms of prevalent cases, high/​very
among men in high-​income countries is causing a substantial reduc- high HDI countries accounted for the majority of cancers of the breast,
tion in lung cancer incidence and mortality rates, although the rates prostate, and colorectum, by far the most prevalent types of cancer.
continue to increase or are only now reaching a plateau among women
in these countries. In China, only 2% of women are current smok-
ers, and many former smokers used pipes rather than cigarettes. The Survival Patterns
high lung cancer rates among Chinese women are largely the result of As indicated in Figure 8–​16, the number of cancer patients surviv-
indoor air pollution from cooking and heating with unventilated stoves ing 5 years after diagnosis (prevalent cases) is higher in high/​very
that have historically used coal (Chapter 28). high HDI countries than in low/​medium HDI countries. Prevalence
is affected by the true underlying incidence rate and clinical behavior
Site-​Specific Cancer Burden by Level of HDI of the cancer and by the availability and effectiveness of treatment.
Figure 8–​16 shows the estimated number of incident cases, deaths, Survival has lengthened for cancers of the breast and prostate and
and prevalent cancers in 2012 for the 10 most common cancer sites colorectum in high-​and very-​high-​income countries (Allemani et al.,
in relation to two categories of HDI. High/​very high HDI countries 2015). For breast and prostate cancer, this is due to an earlier average
 15

Patterns of Cancer Incidence, Mortality, and Survival 115


Low/Medium HDI High/Very high HDI
France* Denmark
100 100
USA*:
80 White USA*: 80
Black

Age-standardized (world) incidence rate per 100,000, females


Age-standardized (world) incidence rate per 100,000, females

60 Philippines* 60
Australia
50 Colombia* 50
China* Japan*
40 40

30 30
Thailand* Denmark
25 India* 25

Uganda* USA:
20 Black 20

France
Australia
15 Philippines 15
USA:
White
12 12

10 Colombia 10
Japan
8 8
7 7

6 China 6

5 5

1980 1990 2000 2010 1980 1990 2000 2010


Year

Incidence Mortality

Figure 8–​9.  Trends in female breast cancer incidence and mortality rates for select countries, 1975–​2012. Source: IARC.

stage at diagnosis and greater availability of effective treatments in In North Africa and West Asia, lung cancer predominates among
affluent countries. Survival from breast, prostate, and thyroid can- men, but colorectal cancer is increasing in the Gulf States, and liver
cer, however, are artificially inflated in high/​very high HDI countries cancer and non-​Hodgkin lymphoma are major cancers elsewhere
because of the early diagnosis of non-​aggressive cancers detected by (Figure 8–​19). Among women, breast cancer ranks first in incidence
screening. Figures 8–​2 through 8–​5 show the distribution of prevalent throughout the region (Figure 8–​20). In Central and South America,
cases by WHO region and anatomic site. prostate cancer is the leading cause of cancer death among men
(Figure 8–​21), although lung and stomach cancer predominate in
some countries. Among women, breast cancer is the most common
Regional Patterns incident cancer regionally, although cervical cancer still ranks first
Figures 8–​17 through 8–​30 depict regional patterns of cancer inci- in low-​income countries in Central America, as well as in Peru and
dence, mortality, and survival among men and women in seven WHO Bolivia in South America (Figure 8–​22). In North America, lung
geographic regions. There are many distinctive features. Sub-​Saharan cancer is the leading fatal cancer in both men and women (Figures
Africa is the only region in which men have high death rates as well 8–​23 and 8–​24, respectively). South Asia includes a large area
as high incidence rates of prostate cancer (Figure 8–​17). This contrasts where incident cancers of the lip and oral cavity predominate in men
with the patterns in North America and Europe, where prostate cancer (Figure 8–​25), whereas breast cancer is the predominant incident
incidence rates are high, partly due to screening practices, while mor- cancer in women (Figure 8–​26). In East and Southeast Asia, lung
tality rates are relatively low. Even within sub-​Saharan Africa, prostate cancer is the most common site for incidence and mortality in men,
cancer mortality rates vary by 8-​fold, with higher death rates in Central and breast cancer in women (Figures 8–​27 and 8–​28, respectively).
and East Africa. Kaposi sarcoma is common is Southeast Africa due to Mongolia has the highest liver cancer incidence rate in the world
untreated HIV infection. Cervical cancer is the leading cause of can- in both sexes, due to the high prevalence of chronic infection with
cer death among women in large areas of sub-​Saharan Africa (Figure hepatitis B and/​or C viruses (Chimed et al., 2017). Thyroid cancer is
8–​18). The lifetime risk of cervical cancer is 6%–​8% in Malawi, commonly diagnosed in Korea due to screening for this cancer with
Mozambique, and Zimbabwe. Cervical and breast cancers account for ultrasound. The patterns in Europe are generally similar to those in
over half of all female incident cancers in sub-​Saharan Africa. North America for men (Figure 8–​29) and women (Figure 8–​30).
16

116 Part II: The Magnitude of Cancer


Low/Medium HDI High/Very high HDI
50 50
Australia

40 France* USA*: 40
Black
35 China* Denmark 35
Age-standardized (world) incidence rate per 100,000, males

Age-standardized (world) incidence rate per 100,000, males


30 Japan* USA*: 30
Philippines*
White
25 25
Denmark

20 20
Australia

USA:
15 Black 15
Thailand*
France

Japan Colombia*
USA:
10 White 10
China

Uganda*
India*
7 7
Colombia

5 5
Philippines

4 4

3 3

1980 1990 2000 2010 1980 1990 2000 2010


Year

Incidence Mortality

Figure 8–​10.  Trends in colorectal cancer incidence and mortality rates for select countries, 1975–​2012. Source: IARC.

Breast cancer is the leading cancer among women in the region in by cancers of the gastrointestinal and respiratory tracts and bladder
terms of incidence and mortality; the highest breast cancer rates are cancer.
observed in a number of Northern and Western European countries Armitage and Doll and others studied the mathematical relation-
(Figure 8–​30). ship between attained age and the mortality rate from various solid
tumors (Armitage and Doll, 1954). They observed a linear relation-
ship between the log of the age-​specific death rates for certain types
Demographic Patterns of cancer and the log of attained age. From this, they hypothesized
Cancer registries collect individual-​level information on core demo- that carcinogenesis was a multistage process that required six or
graphic characteristics on all cases. These data include date of birth, more discrete events for the development of certain solid tumors.
gender, race/​ethnicity, age at diagnosis, and place of residence. This Their hypothesis was advanced 10  years before elucidation of the
demographic information allows study of cancer risk in individuals structure of DNA, and over 30  years before the identification of
according to these characteristics, thereby avoiding the well-​known the first oncogenes and tumor suppression genes. Molecular biolo-
limitations of “ecologic” studies, which rely on exposure information gists have since identified many of the actual genetic and epigenetic
on the population, rather than individual cases. Historically, epidemi- events. These accumulate in tumor tissue over time and dysregu-
ologists have drawn landmark insights from the demographic informa- late normal cellular control over replication, survival, and growth
tion, particularly on the relationships between cancer incidence and (see Chapter 2) (Armitage and Doll, 2004; Hornsby et al., 2007; Wu
attained age. et al., 2016).
The shape of the relationship between attained age and the age-​
Relationships with Age specific incidence rates can also reflect physiologic changes during
The incidence and mortality rates of many types of cancer increase life. Clemmesen observed that the age-​specific incidence rate of breast
exponentially with age during adulthood, except in the oldest age cancer was not a smooth continuous curve (Clemmesen, 1948). Rather,
groups. The relationships between attained age and the age-​specific incidence rates increase rapidly between menarche and menopause,
incidence rates of various types of cancer are illustrated in Figure followed by a “point of inflection” and a more gradual increase after
8–​31. The age-​related increases in risk are approximately exponen- menopause. This observation (ascribed to the author as Clemmesen’s
tial for most solid tumors in adults below age 80 years, as evidenced hook) led him to theorize that there were two different pathways for
 17

Incidence, Men Mortality, Men

Low HDI Low HDI


Medium HDI Medium HDI
High HDI High HDI
Very high HDI Very high HDI
China China
India India

0 50 100 150 200 0 50 100 150 200


Age-standardized rate (W) per 100,000

Lung Stomach Liver Esophagus Pancreas


Prostate Colorectum Bladder Other pharynx Lip, oral cavity

Incidence, Women Mortality, Women

Low HDI Low HDI


Medium HDI Medium HDI
High HDI High HDI
Very high HDI Very high HDI
China China
India India

0 50 100 150 200 0 50 100 150 200


Age-standardized rate (W) per 100,000

Breast Cervix uteri Liver Ovary Stomach Pancreas


Colorectum Lung Corpus uteri Thyroid Esophagus Lip, oral cavity

Figure 8–​11.  Age-​standardized cancer incidence and mortality rates for specific cancer sites in relation to the level of Human Development Index (HDI).
Source: World Health Organization (WHO).

Prostate (87) Kaposi sarcoma (6)

Lung (38) Lip, oral cavity (5)

Liver (20) Leukaemia (2)

Stomach (12) Non-Hodgkin lymphoma (2)

Colorectum (11) Esophagus (1)

No data Not applicable

The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization concerning
the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps represent
approximate border lines for which there may not yet be full agreement.

Figure 8–​12.  Most commonly diagnosed cancer among men by country, 2012. The parentheses following the cancer type represents the number of coun-
tries in which that cancer is the most commonly diagnosed among men. Source: World Health Organization (WHO).
18

118 Part II: The Magnitude of Cancer

Lung (91) Leukaemia (4)


Prostate (42) Esophagus (2)
Liver (23) Colorectum (1)
Stomach (13) Lip, oral cavity (1)
Kaposi sarcoma (6) Non-Hodgkin lymphoma (1)
No data Not applicable

The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization concerning
the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps represent
approximate border lines for which there may not yet be full agreement.

Figure 8–​13.  The leading cause of cancer death among men by country, 2012. The parentheses following the cancer type represents the number of coun-
tries in which that cancer is the leading cause of death among men. Source: World Health Organization (WHO).

pre-​and postmenopausal breast cancer, and that hormonal changes Birth Cohort and Period Patterns in Age-​Specific
associated with menopause modify the relationship with age (Hakama, Incidence Rates
1969; Hill et  al., 1983). Other distinctive age relationships are The relationships between age and cancer incidence rates can vary
observed with cancer of the cervix (Chapter 48), Hodgkin lymphoma because of differences in the timing and intensity of exposure and
(Chapter  39), kidney cancer (Chapter  51), and all of the childhood because of changes in tumor detection (Bray, 2016). Consequently,
cancers (Chapter 59). These can reflect age-​related changes in the tim- the age-​related patterns are neither static nor universal. Figures 8–​32
ing of exposure, susceptibility to the exposure, and/​or the number of and 8–​33 show temporal changes in the age-​specific incidence rates of
discrete events related to carcinogenesis. lung and prostate cancer, respectively, in US men. The rise and fall of

Breast (140)

Cervix uteri (39)

Liver (2)

Lung (2)

Thyroid (1)

No data

Not applicable

The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization concerning
the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps represent
approximate border lines for which there may not yet be full agreement.

Figure 8–​14.  Most commonly diagnosed cancer among women by country, 2012. The parentheses following the cancer type represents the number of
countries in which that cancer is the most commonly diagnosed among women. Source: World Health Organization (WHO).
 19

Breast (103)

Cervix uteri (45)

Lung (25)

Stomach (5)

Liver (3)

Colorectum (2)

Esophagus (1)

No data

Not applicable

The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization
concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps
represent approximate border lines for which there may not yet be full agreement.

Figure 8–​15.  The leading cause of cancer death among women by country, 2012. The parentheses following the cancer type represents the number of
countries in which that cancer is the leading cause of death among women. Source: World Health Organization (WHO).

World cancer incidence


1034
1000 947
927897 913
New cases in 2012 (Thousands)

750
641
554 582 High/Very High HDI
500 447 Low/Medium HDI
397 368
331 310
243
250 200
164 160
124 120 143

0
ng

st

te

a
i

r
er
ve

de
gu
ac

om
ea

tu

ta
Lu

ut
Li

ad
om

ha
ec

os
Br

ph
x

Bl
or

Pr

op
vi
St

m
er
ol

Es

ly
C
C

in
gk
od
–H
on
N

World cancer mortality World cancer prevalence (5 years)


4103
816 4000
773
750 3489
Persons in 2012 (Thousands)
Deaths in 2012 (Thousands)

3000
563 2586
500 462 2152
418
2000
265 297
261 275
250 257 227 1100
204 199 958 1009 1017
183 1000
128138
793 781757 765 845 689
103 103 103 538 451 361
66 434 303 218
0 0
g

as

te

ia
er
st
ve

Pr st

te

ng

er

y
gu
n

ac

er

er
m
tu

ta

ne
ac

oi
ea

re
Lu

ut

ea

ta

tu

d
Li

Lu

ut

ut
om

ha

ae
ec

os

yr
ad
nc

om

d
os

ec
Br

Br

Th

Ki
x

s
or

op

Pr

vi

uk

Bl
St

Pa

or

vi

pu
St
er
ol

Le
Es

er
ol

or
C
C

C
C

Figure 8–​16.  Number of cancer cases and deaths for top 10 cancers according to Human Development Index (HDI), worldwide, 2012. Source: World
Health Organization (WHO).
120

120 Part II: The Magnitude of Cancer


Prevalence
Incidence Mortality (5 years)

Prostate Prostate
Prostate

46.6%
49.4% 49.4%
Liver Liver
50.6% 50.6%
Kaposi 53.4%
Kaposi sarcoma Kaposi sarcoma sarcoma
Non-Hodgkin Colorectum
Esophagus
lymphoma Lip, oral cavity
Non-Hodgkin
Colorectum lymphoma Non-Hodgkin lymphoma

256,000 201,000
453,000
New cases Deaths
Persons

Prostate
Uganda
Burundi
Congo, Democratic Republic of
Congo, Republic of
Kenya
Madagascar
South African Republic
Tanzania
Angola
Nigeria
South Sudan
Central African Republic
Rwanda
Equatorial Guinea
Cameroon
Guinea
Zimbabwe
Sierra Leone
Zimbla
Comoros
Benin
Liberia
Chad
Nambia
Senegal
Somalia
Mauritius
Mortality Mauritania
Guinea-Bissau
Prostate (19) Burkina Faso
Cote d Ivoire
Liver (14) Gabon
France, La Reunion
Kaposi sarcoma (6) Mali
Togo
Lung (3) Niger
Swaziland
Leukemia (2) Malawi
Cape Verde
Non-Hodgkin lymphoma (1) Mozambique
Ghana
Esophagus (1) Djibouti Eastern Africa
Eritrea
Lesotho Middle Africa
Stomach (12) Botswana Southern Africa
No data Ethiopia Western Africa
The Gambia
Not applicable
0 0.5 1 1.5 2 2.5 3 3.5 4 4.5
Mortality cumulative risk, male, 0–74 years (%)

The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization
concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps
represent approximate border lines for which there may not yet be full agreement.

Figure 8–​17.  The cancer burden in sub-​Saharan Africa, males, 2012. The parentheses following the cancer type represents the number of countries in
which that cancer is the leading cause of death among men in sub-Saharan Africa. Source: World Health Organization (WHO).

age-​specific lung cancer rates (Figure 8–​32) follow generational (birth States. Other examples of birth cohort effects include the elevated liver
cohort) patterns of smoking that were established many decades ear- cancer rates in US baby boomers (Altekruse et al., 2009), thought to
lier. Birth cohort patterns reflect exposures (or patterns of exposure) reflect the high prevalence of hepatitis C virus infections among intra-
that become fixed early in life, change across successive generations, venous drug users during 1960s–​1980s (Armstrong et al., 2006), and
and cause disease later in life. Figure 8–​32 plots age-​specific incidence the patterns of testicular cancer in several countries (Bergstrom et al.,
rates of lung cancer by 10-​year intervals of attained age on the y axis, 1996; Liu et al., 2000; Verhoeven et al., 2008).
versus 5-​year intervals of year of birth (birth cohorts) on the x axis. The In contrast, period effects result from changes in detection or expo-
peak in lung cancer incidence in every category of attained age corre- sures that affect cancer risk in all age groups at the same time. This
sponds to the birth cohort with the maximum lifetime smoking. This is manifested by an increase or decrease in risk at all ages during the
peak shifts toward earlier birth cohorts as age increases, because these same calendar year of diagnosis or death (Rosenberg and Anderson,
older birth cohorts preceded the period of peak smoking in the United 2011). Figure 8–​33 illustrates the “period” increase in prostate cancer
 12

Patterns of Cancer Incidence, Mortality, and Survival 121


Prevalence
Incidence Mortality (5 years)

Breast Cervix uteri


Breast

27.6%
38.1% 43.9%
61.9% 56.1%
Breast
Cervix uteri 72.4%
Cervix uteri

Liver
Liver Corpus uteri
Colorectum Colorectum
Ovary
Kaposi sarcoma Ovary Colorectum

370,000 247,000 864,000


New cases Deaths Persons

Cervix uteri
Malawi
Mozambique
Comoros
Zimbabwe
Tanzania
Zimbia
Burundi
Swaziland
Mall
Uganda
Madagascar
Senegal
Rwanda
Kenya
Guinea
Congo, Democratic Republic of
Ghana
Lesotho
Angolo
Somalia
South Sudan
Sierra Leone
Liberia
Nigeria
Cameroon
South African Republic
Congo, Republic of
Botswana
Guinea-Bissau
Mauritania
Cape Verde
Benin
Incidence Ethiopia
Equatorial Guinea
Cervix uteri (28) Burkina Faso
Central African Republic
Cote d Ivoire
Breast (19) Togo
Gabon
Chad
No data Eritrea Eastern Africa
Djibouti
Mauritius Middle Africa
Not applicable France, La Reunion
Namibia Southern Africa
The Gambia Western Africa
Niger

0 1 2 3 4 5 6 7 8
Incidence cumulative risk, female, 0–74 years (%)

The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization
concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps
represent approximate border lines for which there may not yet be full agreement.

Figure 8–​18.  The cancer burden in sub-​Saharan Africa, females, 2012. The parentheses following the cancer type represents the number of countries in
which that cancer is the most commonly diagnosed among women in sub-Saharan Africa. Source: World Health Organization (WHO).

incidence in the early 1990s in SEER areas of the United States, fol- therapy (HRT; Jemal et al., 2007; Ravdin et al., 2007). This followed
lowing the introduction of prostate specific antigen (PSA) testing in a major publication associating HRT use with breast cancer and other
late 1980s. PSA testing was recommended for men age 50 years and conditions (Rossouw et al., 2002).
older (Brawley, 2012; Hankey et al., 1999a; Potosky et al., 1995). The The age-​period-​cohort (APC) model provides a somewhat more for-
increase in detection produced a dramatic increase in the recorded inci- mal statistical approach for partitioning age, cohort, and period effects.
dence of prostate cancer, followed by a sharp decrease, reflecting the It is based on a log-​linear model of temporal trends in cancer rates.
depletion of unrecognized prevalent disease and decreases in PSA test- The APC analysis can be carried out through a freely available and
ing (Brawley, 2012; Welch and Albertsen, 2009). Period effects can be user-​friendly web tool developed by NCI (http:/​analysistools.nci.nih.
identified by plotting calendar year of diagnosis or death on the x axis. gov/​apc/​) (Rosenberg et al., 2014) and other similar tools (Carstensen,
Another example of a period effect is the sharp decrease in breast 2007; Rutherford et  al., 2010). Unfortunately, the interpretability of
cancer incidence rates in postmenopausal women between 2002 and results from this model is limited by collinearity between attained age,
2003, coinciding with the reduction in use of hormone replacement period, and birth cohort (Rosenberg and Anderson, 2011). Because
12

122 Part II: The Magnitude of Cancer


Prevalence
Incidence Mortality (5 years)

Lung Prostate
Lung

Prostate Bladder
49.0% Liver 48.4% 47.1%
51.0%
51.6% 53. 0%
Bladder
Prostate Colorectum

Colorectum Colorectum
Lung
Liver Bladder Larynx

274,000 187,000
492,000
New cases Deaths
Persons

Lung

Armenia
Turkey
Tunisia
Georgia
Lebanon
Israel
Libya
Jordan
Morocco
Syrian Arab Republic
Iraq
State of Palestine
Bahrain
Azerbaijan
Algeria
Qatar
Western Sahara
Incidence United Arab Emirartes
Lung (13) Egypt
Kuwait
Colorectum (6)
Saudi Arabia
Prostate (2)
Oman
North Africa
Leukemia (1) Yemen West Asia
Liver (1) Sudan

Non-Hodgkin lymphoma (1)


0 2 4 6 8
Not applicable Incidence cumulative risk, male, 0–74 years (%)

The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization
concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps
represent approximate border lines for which there may not yet be full agreement.

Figure 8–​19.  The cancer burden in North Africa and West Asia, males, 2012. The parentheses following the cancer type represents the number of countries
in which that cancer is the most commonly diagnosed among males in North Africa and West Asia. Source: World Health Organization (WHO).

these variables are linearly dependent, their effects cannot be uniquely comparison, the ratio of male-​to-​female incidence rates for cancer
and simultaneously determined without unverifiable assumptions sites that are more common in men ranges from 1.25 for colon cancer
(Clayton and Schifflers, 1987a, 1987b; Holford, 1983). to 11.8 for Kaposi sarcoma.
When all age groups are considered, the ratio of male to female
Sex Relationships incidence rates is 1.35 for all cancers combined. This ratio varies
For nearly all types of cancer that affect both sexes, men have higher by age, however, as shown for the United States in Figure 8–​34. At
age-​standardized incidence and death rates than women. In high-​ younger ages (approximately age 20–​55  years), the incidence rate
income countries, the few exceptions to this, other than breast can- of all cancers combined is higher in women than in men, mostly
cer, include cancers of the thyroid, gallbladder, and anus (Table 8–​1). due to premenopausal breast cancer. Above age 55  years, this pat-
For these sites, the female-​to-​male rate ratio in GLOBOCAN 2012 tern reverses. In contrast, the mortality rate is higher in men than in
ranges from 1.15 for anal cancer to over 120 for breast cancer. By women at all ages.
 123

Patterns of Cancer Incidence, Mortality, and Survival 123


Prevalence
Incidence Mortality (5 years)

Breast
Breast Breast
31.9%
48.6%
45.0%
51.4% Colorectum 55.0%
68.1%
Colorectum Lung Thyroid
Thyroid Stomach Colorectum
Non-Hodgkin
lymphoma Corpus uteri
Liver Cervix uteri
Ovary

264,000 146,000 658,000


New cases Deaths Persons

Breast

Israel
Lebanon
Armenia
Jordan
Syrian Arab Republic
Kuwait
Egypt
Qatar
Algeria
Georgia
Iraq
State of Palestine
Bahrain
United Arab Emirartes
Morocco
Turkey
Western Sahara
Tunisia
Saudi Arabia
Yemen
Sudan
Incidence
Oman
Breast (24) Azerbaijan
North Africa
West Asia
Not applicable Libya

0 2 4 6 8
Incidence cumulative risk, female, 0–74 years (%)

The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization
concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps
represent approximate border lines for which there may not yet be full agreement.

Figure 8–​20.  The cancer burden in North Africa and West Asia, females, 2012. The parentheses following the cancer type represents the number of coun-
tries in which that cancer is the most commonly diagnosed among females in North Africa and West Asia. Source: World Health Organization (WHO).

For some cancers, the gender disparity largely reflects differences ratio for lung cancer decreased from 3 in 1975 to 1.4 in 2008–​2012
in exogenous exposure to carcinogens, whereas for others it reflects (Jemal et al., 2003b). The male and female incidence rates of smoking-​
endogenous exposures or inherent biological differences (Walter et al., related oral cavity cancer also converged (Brown et al., 2012), whereas
2013; Wiren et al., 2014). For example, the higher rates of lung cancer the gender difference in HPV-​related oropharyngeal cancers widened
and other smoking-​related cancers in men than women reflect the his- because of differential changes in sexual behavior (Chaturvedi et al.,
torically higher prevalence of male smoking (Jemal et al., 2008; Thun 2008; Simard et al., 2012). In populations of European origin, the inci-
et al., 2013), whereas the elevated breast cancer risk in women com- dence rate of melanoma is higher in women than men before age 40,
pared to men reflects both endogenous and exogenous exposure to hor- but not at older ages, possibly because of the use of artificial sun beds
mones. Because the decrease in smoking began earlier and was larger by young women (Heckman et al., 2008). After age 40, the incidence
among men than women in the United States, the male-​to-​female rate rate becomes progressively higher in men (Chapters 57 and 58).
124

124 Part II: The Magnitude of Cancer

Prevalence
Incidence Mortality (5 years)

Prostate
Prostate
Prostate
Lung
44.5% 47.0% 39.5%
55.5%
Lung 53.0%
60.5%
Stomach
Colorectum
Colorectum
Colorectum Stomach
Stomach Bladder
Bladder Liver Lung

484,000 284,000 1.1 million


New cases Deaths Persons

Prostate

Guyana
Uruguay
Suriname
Belize
Paraguay
Venezuela
Ecuador
Brazil
Argentina
Costa Rica
Panama
Chile
Bolivia
Nicaragua
Colombia
Honduras
Mortality Guatemala
Prostate (13) Peru
Stomach (5) EI Salvador
South America
Lung (3) Mexico
Central America
No data French Guyana
Not applicable
0 1 2 3 4
Mortality cumulative risk, male, 0–74 years (%)

The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization
concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps
represent approximate border lines for which there may not yet be full agreement.

Figure 8–​21.  The cancer burden in Central and South America, males, 2012. The parentheses following the cancer type represents the number of countries
in which that cancer is the leading cause of death among men in Central and South America. Source: World Health Organization (WHO).

Race/​Ethnicity Australia (Indigenous vs. all others) (Cunningham et  al., 2008;
Differences in the risk of specific types of cancer in relation to Moore et al., 2010).
race or ethnicity have been widely reported (Moore et  al., 2015). One example of genetic predisposition that is indisputably associ-
In many instances, it is difficult to separate racial and ethnic dif- ated with race is skin pigmentation. Melanin deposits in the skin protect
ferences from socioeconomic disparities (discussed in Chapter 9). against both melanoma (Chapter 57) and non-​melanoma skin cancer
It is also difficult to separate inherited genetic susceptibility from (Chapter 58). The incidence rate of melanoma among Caucasians in
cultural differences that affect exposures. The examples discussed the United States are more than 10 times higher than in blacks (Jemal
here are drawn from the United States (blacks vs. whites) and et al., 2011), due to the joint factors of sunlight and lack of melanin.
 125

Patterns of Cancer Incidence, Mortality, and Survival 125


Prevalence
Incidence Mortality (5 years)

Breast
Breast Breast

Cervix uteri 29.6%


43.1%
49.4% 50.6%
Cervix uteri 57.0% Lung 70.4%
Cervix uteri

Colorectum Colorectum
Colorectum
Lung
Stomach Thyroid
Stomach Corpus uteri

520,000 266,000 1.4 million


New cases Deaths Persons

Breast

Argentina

Uruguay
Brazil

Guyana
Suriname
Costa Rica

Panama

Paraguay
Venezuela

Belize
French Guyana

Colombia
Chile
Incidence
Mexico
Breast (15)
Ecuador
Cervix uteri (6)
Peru
No data Nicaragua

Not applicable EI Salvador

Honduras
South America
Bolivia
Central America
Guatemala

0 1 2 3 4 5 6 7 8
Incidence cumulative risk, female, 0–74 years (%)

The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization
concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps
represent approximate border lines for which there may not yet be full agreement.

Figure 8–​22.  The cancer burden in Central and South America, females, 2012. The parentheses following the cancer type represents the number of coun-
tries in which that cancer is the most commonly diagnosed among women in Central and South America. Source: World Health Organization (WHO).

For most cancers, however, differences in risk between racial/​ smoking and lower frequency of cervical cancer screening among
ethnic groups are due largely to differences in exposures and access Indigenous Australians than in the general population.
to preventive services, rather than differences in genetic susceptibil- It is noteworthy that large differences in cancer risk can exist within
ity. For example, compared to the rest of the Australian population, subpopulations of the same racial or ethnic group, as well as those in
Indigenous Australians have a two-​fold higher risk of lung cancer different groups. For example, the lung cancer incidence rate among
and a seven-​fold higher risk of cervical cancer (Cunningham et al., Hispanic men in the United States is twice as high in those of Cuban
2008; Moore et  al., 2010). This reflects the higher prevalence of descent as in Mexican Americans, because of the historically high
126

126 Part II: The Magnitude of Cancer


Prevalence
Incidence Mortality (5 years)

Prostate Lung
Prostate

29.6%
38.2% 41.8%
58.2%
Lung 61.8% Prostate
70.4%
Colorectum
Colorectum Colorectum
Bladder
Bladder Pancreas Melanoma of skin
Melanoma of skin Liver Lung

920,000 363,000 2.7 million


New cases Deaths Persons

Prostate

12

10

Incidence cumulative risk, male, 0–74 years (%)


8

Incidence
Prostate (2) 0
United Canada
No data States
of America

North America

The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization
concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps
represent approximate border lines for which there may not yet be full agreement.

Figure 8–​23.  The cancer burden in North America, males, 2012. The parentheses following the cancer type represents the number of countries in which
that cancer is the most commonly diagnosed among men in North America. Source: World Health Organization (WHO).

prevalence of smoking among Cubans (Martinez-​Tyson et al., 2009; 2015). Briefly, IARC uses a hierarchical approach that draws on dif-
Pinheiro et al., 2009; Siegel et al., 2015a). ferent sources, depending on the availability and accuracy of local
data. Information on incident cases and survival are obtained from
population-​based registries where available. PBCRs routinely col-
METHODS USED FOR GLOBOCAN 2012 lect demographic data on the age, sex, place of residence, country
of origin, and date of migration (if applicable) of each case. Clinical
The purpose of GLOBOCAN 2012 is to provide a comprehensive information is collected on the anatomic site of the tumor, and its
global picture of the risks and burden from 27 cancer types and all histologic characteristics, date of diagnosis, stage at diagnosis, report-
sites combined in 184 countries for the year 2012 (http://​globocan. ing hospital or cytology/​pathology lab, first course of treatment, and
iarc.fr). The methods are described in detail elsewhere (Ferlay et al., survival. IARC publishes updated incidence rates in the database
 127

Patterns of Cancer Incidence, Mortality, and Survival 127


Prevalence
Incidence Mortality (5 years)

Lung
Breast
Breast
29.5%
37.1% 37.9%
Breast 62.1%
Lung 62.9%
Colorectum 70.5%

Colorectum Colorectum Corpus uteri


Corpus uteri Pancreas Thyroid
Thyroid Ovary Lung

865,000 329,000 2.6 million


New cases Deaths Persons

Breast

10

Incidence cumulative risk, female, 0–74 years (%)


8

Incidence
Breast (2) 0
United Canada
No data States
of America

North America

The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization
concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps
represent approximate border lines for which there may not yet be full agreement.

Figure 8–​24.  The cancer burden in North America, females, 2012. The parentheses following the cancer type represents the number of countries in which
that cancer is the most commonly diagnosed among women in North America. Source: World Health Organization (WHO).

CI5, through collaboration with the global network of PBCRs and classified by histologic type and stage of disease. In the 62 countries
the IACR (http://​www.iacr.com.fr; http://​ci5.iarc.fr/​). The number where incidence data are unavailable, the estimates of cancer inci-
of high-​quality registries included in CI5 has increased markedly dence in GLOBOCAN 2012 are made from regional data and statisti-
over the last half-​century, as shown in Table 8–​2 (Bray et al., 2015c; cal modeling, often based on neighboring countries. In countries that
Forman et  al., 2014; Parkin, 2006). The most recent (10th) volume have only mortality data, a registry-​based ratio of incidence to mortal-
presents information from 290 PCBRs in 68 countries for cancers ity is used to estimate incidence.
diagnosed during 2003–​2007. Less than a fifth of all countries collect national data on newly diag-
Incident cases are classified according to the codes of the nosed cancer cases and deaths, however (Bray et al., 2015a). In places
International Statistical Classification of Diseases and Health-​Related where no vital statistics are available, cancer-​specific mortality rates
Problems, 10th Revision (ICD-​10). Where possible, tumors are also are estimated using interpolated incidence and survival probabilities,
128

128 Part II: The Magnitude of Cancer


Prevalence
Incidence Mortality (5 years)

Lung Lung Lip, oral cavity

Lip, oral cavity Stomach Prostate


44.0% 48.4% 42.1%

Lip, oral cavity 51.6% Colorectum


Stomach 56.0% 57.9%

Colorectum Esophagus Other Pharynx

Other Pharynx Other Pharynx Larynx

675,000 506,000 1 million


New cases Deaths Persons

Lip, oral cavity

Maldives

Sri Lanka

Bangladesh

Pakistan

India

Nepal

Afghanistan
Incidence
Lip, oral cavity (7) Bhutan
Stomach (3)
South Asia
Lung (2)
Iran, Islamic Republic of
Not applicable

0 0.5 1 1.5 2
Incidence cumulative risk, male, 0–74 years (%)

The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization
concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps
represent approximate border lines for which there may not yet be full agreement.

Figure 8–​25.  The cancer burden in South Asia, males, 2012. The parentheses following the cancer type represents the number of countries in which that
cancer is the most commonly diagnosed among men in South Asia. Source: World Health Organization (WHO).

based on macroeconomic measures and scaled to the WHO mortality mortality statistics in their database (Mathers et al., 2005). Even inclu-
estimates wherever possible. sion in the WHO database is not a guarantee of data quality, however; in
A relatively small subset of registries, representing about 30% of some countries and/​or time periods, population coverage is manifestly
the world’s population, submit high-​quality mortality data to the WHO incomplete, and the mortality rates produced are implausibly low.
mortality database. Reasonably reliable national information is cur- Death certificates record information on the person dying, and the
rently available for 38 countries; these include all of the economically cause of death, as certified, usually by a medical practitioner. The
developed countries and a subset of developing countries (Parkin and International Classification of Diseases (ICD) provides a uniform sys-
Bray, 2006). The remaining countries provide either lower-​quality tem of nomenclature and coding, and a recommended format for the
cause-​of-​death data or, in 74 (mainly low-​resource) countries, no such death certificate (WHO, 1992).
data at all. The Global Health Observatory (GHO; http://​www.who. As noted earlier, the survival of patients with cancer is measured
int/​gho/​) publishes tables of estimated coverage and completeness of conventionally for the first 5 years after diagnosis. In settings where
 129

Patterns of Cancer Incidence, Mortality, and Survival 129


Prevalence
Incidence Mortality (5 years)

Breast
Breast
Breast

27.5%
38.6% 44.2%
61.4% Cervix uteri 55.8%
72.5%
Cervix uteri
Cervix uteri
Ovary
Ovary
Colorectum Ovary
Colorectum
Corpus uteri
Lip, oral cavity Stomach Lip, oral cavity

759,000 466,000 1.7 million


New cases Deaths Persons

Breast

Pakistan

Afghanistan

Sri Lanka

Maldives

Iran, Islamic Republic of

India

Incidence Bangladesh
Breast (7)
Cervix uteri (2)
Nepal
Not applicable
South Asia
Bhutan

0 1 2 3 4 5
Incidence cumulative risk, female, 0–74 years (%)

The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization
concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps
represent approximate border lines for which there may not yet be full agreement.

Figure 8–​26.  The cancer burden in South Asia, females, 2012. The parentheses following the cancer type represents the number of countries in which that
cancer is the most commonly diagnosed among women in South Asia. Source: World Health Organization (WHO).

follow-​up for vital status is not feasible, the ratios of cases to deaths priorities, to evaluate the effectiveness of programs in prevention and
expressed as percent are used to estimate survival. The 5-​year survival treatment, and to forecast future healthcare needs (Anderson, 2009;
has been increasing rapidly in high-​income countries. Glaser et al., 2005; Hankey et al., 1999b; Parkin, 2008). Surveillance
data have documented that the implementation of organized cervical
cancer screening programs, beginning in the Nordic countries in the
FUTURE DIRECTIONS early 1960s, was associated with a dramatic reduction in cervical can-
cer mortality by the late 1980s (Laara et al., 1987). A national hepa-
Population-​based surveillance data have become essential for cancer titis B infant vaccination program in Taiwan, which began in 1984,
control. They allow governments to monitor the occurrence and disease has reduced liver cancer rates by over 70% in vaccinated children
burden from cancer in their population, to assess needs, to establish and adolescents (Chiang et al., 2013) (Chapter 62.3). Thun and Jemal
130

130 Part II: The Magnitude of Cancer


Prevalence
Incidence Mortality (5 years)

Lung Stomach
Lung

Colorectum 37.5%
Stomach 31.8% 24.8%
Liver
68.2% 75.2%
Lung 62.5%
Liver
Stomach

Colorectum Prostate
Esophagus
Esophagus Colorectum Liver

2.8 million 2.0 million 4.6 million


New cases Deaths Persons

Lung

Korea, DPR

Timor–Leste

Korea, Republic of

China

Viet Nam

Japan

Singapore

Philippines

Brunei

Mongolia

Thailand

Malaysia

Indonesia

Myanmar
Incidence
Lung (7) Lao PDR
Southeast Asia
Liver (5)
Cambodia East Asia
Colorectum (2)
Stomach (2)
0 1 2 3 4 5 6 7
No data Mortality cumulative risk, male, 0–74 years (%)
Not applicable

The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization
concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps
represent approximate border lines for which there may not yet be full agreement.

Figure 8–​27.  The cancer burden in East and Southeast Asia, males, 2012. The parentheses following the cancer type represents the number of coun-
tries in which that cancer is the most commonly diagnosed among men in East and Southeast Asia. Source: World Health Organization (WHO).

estimated that tobacco control activities since the 1960s accounted for than in other states (CDC, 2000; Jemal et  al., 2008). Robbins et  al.
40% of the decrease in overall cancer death rates between the early described a shift toward earlier stage diagnosis of cervical cancer and
1990s and 2000 in the United States (Thun and Jemal, 2006). increases in receipt of fertility-​sparing treatment among young women
The value of population-​based data on cancer outcomes is enhanced aged 21–​25 years, following expansion of the Affordable Care Act to
if survey data on cancer risk factors and screening are also available. cover young adults on their parents’ health insurance plans until age
Even ecological data (pertaining to populations, not individuals) can 26 years (Robbins et al., 2015).
be highly informative for cancer control. Lung cancer incidence has The infrastructure for population-​ based cancer surveillance has
decreased more rapidly in California, where comprehensive tobacco expanded greatly since 1975, when IARC first estimated the global
control measures were implemented earlier and more rigorously burden of cancer (IARC, 1966). Despite this, the coverage and
 13

Patterns of Cancer Incidence, Mortality, and Survival 131


Prevalence
Incidence Mortality (5 years)

Breast Lung
Breast

Lung Liver 36.5%


43.9% 40.2%
59.8% Colorectum
56.1% 63.5%
Stomach
Colorectum
Cervix uteri
Breast
Stomach Corpus uteri

Liver Colorectum Thyroid

2.1 million 1.2 million 4.8 million


New cases Deaths Persons

Breast

Singapore

Brunei

Japan

Philippines

Korea, Republic of

Indonesia

Malaysia

Timor-Leste

Korea, DPR

Thailand

Viet Nam

Myanmar

China
Incidence
Breast (10) Cambodia

Liver (2) Lao PDR


Southeast Asia
Lung (2)
Mongolia East Asia
Cervix uteri (1)
Thyroid (2) 0 1 2 3 4 5 6 7
No data Incidence cumulative risk, female, 0–74 years (%)
Not applicable

The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization
concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps
represent approximate border lines for which there may not yet be full agreement.

Figure 8–​28.  The cancer burden in East and Southeast Asia, females, 2012. The parentheses following the cancer type represents the number of countries
in which that cancer is the most commonly diagnosed among women in East and Southeast Asia. Source: World Health Organization (WHO).

granularity of the data are highly uneven. Less than 10% of the popula- established that it is feasible to collect reliable local information at
tions of Africa and South America are covered, in contrast to over 95% the population level, even in the lowest income settings (Bray et al.,
of the population of North America. The Global Initiative for Cancer 2015c; Tangka et al., 2010).
Registry Development is an international partnership established to Linkage of cancer registry data with other administrative databases
develop the infrastructure for high-​quality PBCRs in 50 countries provides opportunities to study the quality and cost of care, access to
over the next decade (GICR; http://​gicr.iarc.fr). This effort supports care, and quality of life (Glaser et al., 2005; Hiatt et al., 2015; Howe
the global monitoring framework established by WHO to reduce non-​ et al., 2003; Wingo et al., 2005). A recent study in the United States
communicable diseases in LMICs by 25% by 2025. Studies have documented substantial regional variation in Medicare spending for
132

132 Part II: The Magnitude of Cancer


Prevalence
Incidence Mortality (5 years)

Prostate
Lung Prostate

32.8%
Lung 37.4% 41.0%
62.6% Colorectum 59.0%
Colorectum
67.2%
Colorectum Prostate
Bladder
Bladder Stomach Lung
Stomach Pancreas Kidney

1.8 million 976,000 4.5 million


New cases Deaths Persons

Lung

Hungary
FYR Macedonia
Serbia
Montenegro
Poland
Croatia
Belarus
Romania
Mortality Latvia
Lithuania
Lung (39) Bulgaria
Belgium
Prostate (1) Russian Federation
Estoria
Bosnia Herzegovina
No data Greece
Ukraine
Slovenia
Czech Republic
Slovakia
Spain
France
Republic of Moldova
The Netherlands
Denmark
Albaria
Luxembourg
Italy
Germany
Austria
Ireland
United Kingdom
Portugal
Norway
Malta Eastern Europe
Switzerland Western Europe
Iceland
Finland Northern Europe
Cyprus Southern Europe
Sweden

0 2 4 6 8
Mortality cumulative risk, male, 0–74 years (%)
The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization
concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps
represent approximate border lines for which there may not yet be full agreement.

Figure 8–​29.  The cancer burden in Europe, males, 2012. The parentheses following the cancer type represents the number of countries in which that cancer
is the leading cause of death among men in Europe. Source: World Health Organization (WHO).

advanced cancers without a meaningful correlation between the amount high-​income countries, which impedes efforts to evaluate quality
expended and survival (Brooks et al., 2013). Linkage of cancer registry of care (Hewitt and Simone, 2000; IOM, 2013). Numerous tumor
data with claims data and biomarkers can provide valuable information markers are already informative about the prognosis and treatment
on the prognosis of disease in relation to the quality and cost of care of various cancers. Examples include (1) oncotype DX (which pre-
(Hiatt et al., 2015; Huckman and Kelley, 2013; Institute of Medicine dicts recurrence and the effectiveness of chemotherapy after sur-
[IOM], 2013; Lipscomb and Gillespie, 2011; Spinks et al., 2011). gery for early stage, estrogen positive breast cancer); (2) mutations
In addition, the lack of information by which to classify tumors in the gene encoding epidermal growth factor receptor (EGFR)
according to molecular characteristics, rather than anatomic or (which indicates the sensitivity of advanced non-​small-​cell lung
morphologic characteristics, has been identified as a major gap in cancer to EGFR tyrosine kinase inhibitors); (3)  BRAF mutation
 13

Patterns of Cancer Incidence, Mortality, and Survival 133


Prevalence
Incidence Mortality (5 years)

Breast
Breast
Breast
32.9%
Colorectum
41.0% 45.1%
54.9%
Colorectum 59.0%
Lung Colorectum 68.7%

Lung
Pancreas Corpus uteri
Corpus uteri
Melanoma of skin
Ovary Stomach Cervix uteri

1.6 million 779,000 4.6 million


New cases Deaths Persons

Breast

Belgium
Denmark
Iceland
The Netherlands
United Kingdom
Germany
Finland
Ireland
France
Italy
Luxembourg
Switzerland
Malta
Incidence Sweden
Cyprus
Breast (40) FYR Macedonia
Norway
No data Czech Republic
Serbia
Austria
Slovenia
Croatia
Spain
Portugal
Slovakia
Bulgaria
Montenegro
Hungary
Poland
Latvia
Estoria
Romania
Lithuania
Albaria
Russian Federation
Belarus Eastern Europe
Republic of Moldova Western Europe
Ukraine Northern Europe
Greece Southern Europe
Bosnia Herzegovina

0 2 4 6 8 10 12
Incidence cumulative risk, female, 0–74 years (%)

The boundaries and names shown and the designations used on this map do not imply the expression of any opinion whatsoever on the part of the World Health Organization
concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted and dashed lines on maps
represent approximate border lines for which there may not yet be full agreement.

Figure 8–​30.  The cancer burden in Europe, females, 2012. The parentheses following the cancer type represents the number of countries in which that
cancer is the most commonly diagnosed among women in Europe. Source: World Health Organization (WHO).

(for treatment of advanced melanoma), and (4)  KRAS mutations Population-​based surveillance data on cancer are still widely unde-
(which predict the resistance of colon cancers to treatment with rused, given their potential to monitor progress or lack thereof and to
EGFR inhibitors (Chang et  al., 2015). According to American drive preventive interventions (Howe et al., 2003; Jemal et al., 2003a;
Society of Clinical Oncology, routine testing of colon cancer Siegel et al., 2015b; Wingo et al., 2005), largely because of a lack of
patients for KRAS mutations in the United States could save at skilled manpower in surveillance research. There is an urgent need to
least US$ 600 million a year from the cost of drugs, in addition to attract and train young investigators in innovative methods of surveil-
avoiding unnecessary toxicity and suffering among tens of thou- lance research. A study by Glaser and colleagues found that none of
sands of patients (Poste, 2011). the 34 accredited schools of public health in the United States offered
134

1000
Oral cavity & Esophagus Stomach Colon
100 pharynx

10

0.1

1000 Melanoma
Rectum Pancreas Lung &
bronchus
100

10

0.1

1000
Breast Cervix Uterus Ovary
Rate per 100,000

100

10

0.1

1000
Prostate Bladder Kidney Brain & ONS
100

10

0.1

1000
Hodgkin lymphoma Non-Hodgkin Multiple myeloma Leukemia
lymphoma
100

10

0.1
0 20 40 60 80 0 20 40 60 80 0 20 40 60 80 0 20 40 60 80
Age

Male Female

Figure 8–​31.  Age-​specific incidence rates for select cancers by sex, 2012. Source: SEER.
 135

Males Table 8–​1. Incidence Rate and Male-​to-​Female Incidence Ratios for All


1000 Races Combined, 2008–​2012, SEER Registries
80–84 Cancer Sites Male Female RR (M vs. F) 95% CI
75–79
70–74 All sites 578.3 428.5 1.35 1.3–1.4
65–69 Oral cavity and 16.0 6.6 2.41 2.3–2.5
pharynx
60–64 Esophagus 8.2 2.1 3.98 3.7–4.2
Stomach 11.7 5.6 2.09 2.0–2.2
100 55–59 Colon and rectum 64.1 47.3 1.36 1.3–1.4
Anus, anal canal, 1.3 1.5 0.86 0.8–0.9
and anorectum
Liver and 8.6 3.2 2.67 2.5–2.8
Rate per 100,000

50–54
intrahepatic
bile duct
Gallbladder 0.9 1.4 0.62 0.5–0.7
45–49
Other biliary 1.9 1.3 1.48 1.4–1.6
Pancreas 13.1 10.1 1.29 1.3–1.3
Larynx 6.8 1.5 4.56 4.3–4.9
10 40–44 Lung and 83.2 52.2 1.60 1.6–1.6
bronchus
Bones and joints 1.0 0.8 1.31 1.2–1.5
Soft tissue 3.7 2.4 1.50 1.4–1.6
including
heart
35–39 Melanoma of the 23.5 15.6 1.51 1.5–1.5
skin
Breast 1.1 138.8 0.01 0.0–0.0
1 Cervix uteri 8.1
Corpus and 25.2
30–34 uterus, NOS
0.5 Ovary 14.4
1865
1870

1880

1890

1900

1910

1920

1930

1940

1950

1960

1972

Prostate 181.2
Year of birth Testis 5.6
Urinary bladder 38.3 9.7 3.69 3.9–4.1
Figure  8–​32. Age-​specific lung cancer rates among men in the United Kidney and renal 17.1 8.4 2.03 2.0–2.1
States by year of birth. Source: SEER. pelvis
Brain and other 8.1 5.6 1.43 1.4–1.5
nervous
system
1800 Brain 7.7 5.2 1.47 1.4–1.5
Cranial nerves, 0.4 0.4 0.98 0.8–1.2
other nervous
1600 system
Thyroid 4.2 11.5 0.36 0.3–0.4
1400 Hodgkin 3.1 2.5 1.25 1.2–1.3
Incidence rate per 100,000 males

lymphoma
Non-​Hodgkin 24.1 16.6 1.45 1.4–1.5
1200
lymphoma
70–79
Myeloma 7.5 4.8 1.55 1.5–1.6
1000 Leukemia 17.8 10.4 1.71 1.7–1.8
80+ Mesothelioma 2.1 0.4 5.37 4.7–6.2
800 Kaposi sarcoma 1.5 0.1 11.77 9.5–14.7
60–69
Using incidence-​SEER 9 Regs Research Data, Nov 2014 Sub (1973–​2012) Katrina/​
600 Rita Population Adjustment; rates per 100,000 and age-​adjusted to the 2000 US Std
Population (19 age groups; Census P25-​1130) standard; confidence intervals (Tiwari
mod) are 95% for rates and ratios.
400 ~
Statistic could not be calculated.

200 50–59

40–49
0
1980 1983 1986 1989 1992 1995 1998

Figure 8–​33.  Prostate cancer incidence rates by calendar year of d­ iagnosis.


Source: SEER.
136

136 Part II: The Magnitude of Cancer

3000 Incidence 3000 Mortality


2750 2750
2500 Male 2500
2250 2250
Rate per 100,000

2000 2000
1750 1750 Male
1500 1500
Female
1250 1250
1000 1000
750 750
500 500 Female
250 250
0 0

01 0
05–04
10 09
15 14
20 9
25 24
30 29
35 34
40 39
45 4
50 49
55 54
60 59
65 4
70 69
75 74
80 79
4
+
01 0
05–04
10 09
15 14
20 9
25 24
30 9
35 34
40 39
45 4
50 49
55 4
60 59
65 4
70 69
75 74
80–79
4
+

–1

–4

–6

–8
85
–1

–2

–4

–5

–6

–8
85















Age (years) Age (years)

Figure 8–​34.  Age-​specific incidence and mortality rates by sex, 2012. Source: SEER.

a course in cancer surveillance in their catalogs for 2003–​ 2004. to 8.3 billion by 2030. Most of the increase will be in LMICs, where
Educators and academic institutions have not yet integrated cancer large numbers of young adults are now surviving to older ages, when
surveillance research into the teaching curriculum. The current lack cancer becomes common. The increase is likely to be even higher
of postgraduate training and fellowship in the United States will not than predicted from the demographic changes alone, because of
be adequate to train future generations of surveillance researchers the ongoing adoption of Western patterns of diet, physical inactiv-
(Glaser et al., 2005). This is also true in most parts of the world. ity, delayed reproduction, and cigarette smoking. Consequently, the
future cancer burden will increasingly fall on LMICs that can least
afford it.
THE FUTURE SCALE AND PROFILE OF CANCER 2030
References
The global burden of cancer is expected to increase from 14.1 mil- Allemani C, Weir HK, Carreira H, et al. 2015. Global surveillance of cancer
lion newly diagnosed cases and 8.2 million cancer deaths in 2012 to survival 1995–​2009: analysis of individual data for 25,676,887 patients
21.5  million cases and 13  million deaths in 2030. These projected from 279 population-​based registries in 67 countries (CONCORD-​2).
increases are based solely on demographic predictions, with esti- Lancet, 385(9972), 977–​1010. PMCID: PMC4588097.
mates that the global population will increase from 7 billion in 2012 Altekruse SF, McGlynn KA, and Reichman ME. 2009. Hepatocellular carci-
noma incidence, mortality, and survival trends in the United States from
1975 to 2005. J Clin Oncol, 27(9), 1485–​1491. PMCID: PMC2668555.
Anderson WF. 2009. Cancer surveillance research. Cancer Epidemiol
Biomarkers Prev, 18(6), 1669–​1671. PMCID: PMC2943641.
Table 8–​2. Coverage of the Eight Volumes of Cancer Incidence in Five Armitage P, and Doll R. 1954. The age distribution of cancer and a multi-​stage
Continents theory of carcinogenesis. Br J Cancer, 8(1), 1–​12. PMCID: PMC2007940.
Armitage P, and Doll R. 2004. The age distribution of cancer and a multi-​
Year of Period stage theory of carcinogenesis. 1954. Int J Epidemiol, 33(6), 1174–​1179.
Volume Publication Registries Populations Countries (approx.) PMID: 15319408.
Armstrong GL, Wasley A, Simard EP, et al. 2006. The prevalence of hepatitis C
I 1966 32 35 29 1960–1​ 962 virus infection in the United States, 1999 through 2002. Ann Intern Med,
II 1970 47 58 24 1963–1​ 967 144(10), 705–​714. PMID: 16702586.
III 1976 61 79 29 1968–1​ 972 Bergstrom R, Adami HO, Mohner M, et al. 1996. Increase in testicular cancer
incidence in six European countries: a birth cohort phenomenon. J Natl
IV 1982 79 103 32 1973–1​ 977 Cancer Inst, 88(11), 727–​733. PMID: 8637026.
V 1987 105 137 36 1978–1​ 982 Bleyer A, and Welch HG. 2012. Effect of three decades of screening mammog-
VI 1992 138 166 49 1983–1​ 987 raphy on breast-​cancer incidence. N Engl J Med, 367(21), 1998–​2005.
VII 1997 150 183 50 1988–1​ 992 PMID: 23171096.
VIII 2002 186 214 57 1993–1​ 997 Brawley OW. 2012. Trends in prostate cancer in the United States. J Natl
Cancer Inst Monogr, 2012(45), 152–​156. PMCID: PMC3540881.
Bray F. 2016. The evolving scale and profile of cancer worldwide:  much
As the specificity of data available in cancer registries such as National Cancer Institute's
Surveillance, Epidemiology, and End Results (SEER) program has increased, the ado about everything. Cancer Epidemiol Biomarkers Prev, 25(1), 3–​5.
potential for complex tumor subtype analyses has grown, substantiating the hypothesis PMID: 26667885.
that overall breast cancer incidence patterns are misleading in that they represent Bray F, Ferlay J, Laversanne M, et  al. 2015a. Cancer Incidence in Five
the combined effects of at two or more very different cancer subtypes (Anderson Continents:  inclusion criteria, highlights from Volume X and the
et al., 2014).
 137

Patterns of Cancer Incidence, Mortality, and Survival 137


global status of cancer registration. Int J Cancer, 137(9), 2060–​2071. Day NE. 1992. Cancer Incidence in Five Continents:  cumulative rate and
PMID: 26135522. cumulative risk. IARC Sci Publ(120), 862–​864. PMID: 1284607.
Bray F, Guilloux A, Sankila R, and Parkin DM. 2002. Practical implications Dhillon PK, Yeole BB, Dikshit R, Kurkure AP, and Bray F. 2011. Trends in
of imposing a new world standard population. Cancer Causes Control, breast, ovarian and cervical cancer incidence in Mumbai, India over a
13(2), 175–​182. PMID: 11936824. 30-​year period, 1976–​2005: an age-​period-​cohort analysis. Br J Cancer,
Bray F, Jemal A, Grey N, Ferlay J, and Forman D. 2012. Global cancer tran- 105(5), 723–​730. PMCID: PMC3188937.
sitions according to the Human Development Index (2008–​ 2030):  a Doll R, and Cook P. 1967. Summarizing indices for comparison of cancer inci-
population-​based study. Lancet Oncol, 13(8), 790–​801. PMID: 22658655. dence data. Int J Cancer, 2(3), 269–​279. PMID: 6041994.
Bray F, Jemal A, Torre LA, Forman D, and Vineis P. 2015b. Long-​term real- Doll R, and Peto R. 1981. The causes of cancer: quantitative estimates of avoid-
ism and cost-​effectiveness: primary prevention in combatting cancer and able risks of cancer in the United States today. J Natl Cancer Inst, 66(6),
associated inequalities worldwide. J Natl Cancer Inst, 107(12), djv273. 1191–​1308. PMID: 7017215.
PMCID: PMC4673394. Etzioni R, Penson DF, Legler JM, et al. 2002. Overdiagnosis due to prostate-​
Bray F, and Moller B. 2006. Predicting the future burden of cancer. Nat Rev specific antigen screening:  lessons from U.S.  prostate cancer incidence
Cancer, 6(1), 63–​74. PMID: 16372017. trends. J Natl Cancer Inst, 94(13), 981–​990. PMID: 12096083.
Bray F, and Parkin DM. 2014. Migrant studies: descriptive studies. In: Ahrens Ferlay J, Soerjomataram I, Dikshit R, et al. 2015. Cancer incidence and mor-
W, and Pigeot I (Eds.), Handbook of Epidemiology. New  York:  Spring tality worldwide: sources, methods and major patterns in GLOBOCAN
Science+Business Media. 2012. Int J Cancer, 136(5), E359–​E386. PMID: 25220842.
Bray F, Znaor A, Cueva P, et al. 2015c. Planning and Developing Population-​ Ferlay J, Soerjomataram I, Ervik M, et  al. 2013. GLOBOCAN 2012 v1.0,
Based Cancer Registration in Low-​and Middle-​Income Settings, IARC Cancer Incidence and Mortality Worldwide:  IARC CancerBase No. 11
Technical Publication No. 43. Lyon, France:  International Agency for [Internet]. Lyon, France:  International Agency for Research on Cancer.
Research on Cancer. Available from: http://​globocan.iarc.fr.
Brooks GA, Li L, Sharma DB, et al. 2013. Regional variation in spending and Ferlay J, Steliarova-​Foucher E, Lortet-​Tieulent J, et al. 2013. Cancer incidence
survival for older adults with advanced cancer. J Natl Cancer Inst, 105(9), and mortality patterns in Europe: estimates for 40 countries in 2012. Eur
634–​642. PMCID: PMC3691948. J Cancer, 49(6), 1374–​1403. PMID: 23485231.
Brown LM, Check DP, and Devesa SS. 2012. Oral cavity and pharynx cancer Feuer EJ, Merrill RM, and Hankey BF. 1999. Cancer surveillance series: inter-
incidence trends by subsite in the United States:  changing gender pat- preting trends in prostate cancer. Part II: Cause of death misclassification
terns. J Oncol, 2012, 649498. PMCID: PMC3345247. and the recent rise and fall in prostate cancer mortality. J Natl Cancer Inst,
Carstensen B. 2007. Age-​period-​cohort models for the Lexis diagram. Stat 91(12), 1025–​1032. PMID: 10379965.
Med, 26(15), 3018–​3045. PMID: 17177166. Fontana RS, Sanderson DR, Woolner LB, et al. 1991. Screening for lung can-
Centers for Disease Control and Prevention (CDC). 2000. Declines in lung can- cer:  a critique of the Mayo Lung Project. Cancer, 67(4 Suppl), 1155–​
cer rates: California, 1988–​1997. MMWR Morb Mortal Wkly Rep, 49(47), 1164. PMID: 1991274.
1066–​1069. PMID: 11186612. Forman D, Bray F, Brewster DH, et  al (Eds.). 2014. Cancer Incidence in
CDC. 2016. National Program of Cancer Registries (NPCR). Available Five Continents, Vol. X. Lyon:  International Agency for Research on
from: www.cdc.gov/​cancer/​npcr/​. Accessed November 14, 2016. Cancer: IARC Scientific Publication No. 164.
CDPH. 2016. Connecticut Tumor Registry. Available from: www.ct.gov/​dph/​ Glaser SL, Clarke CA, Gomez SL, et al. 2005. Cancer surveillance research: a
TumorRegistry. Accessed November 14, 2016. vital subdiscipline of cancer epidemiology. Cancer Causes Control,
Center MM, and Jemal A. 2011. International trends in liver cancer inci- 16(9), 1009–​1019. PMID: 16184466.
dence rates. Cancer Epidemiol Biomarkers Prev, 20(11), 2362–​2368. Haenszel W. 1961. Cancer mortality among the foreign-​born in the United
PMID: 21921256. States. J Natl Cancer Inst, 26, 37–​132. PMID: 13710420.
Center MM, Jemal A, and Ward E. 2009. International trends in colorectal Hakama M. 1969. The peculiar age specific incidence curve for cancer of the
cancer incidence rates. Cancer Epidemiol Biomarkers Prev, 18(6), 1688–​ breast: Clemmesen’s hook. Acta Pathol Microbiol Scand, 75(3), 370–​374.
1694. PMID: 19505900. PMID: 5801659.
Chang CQ, Tingle SR, Filipski KK, et al. 2015. An overview of recommenda- Hankey BF, Feuer EJ, Clegg LX, et al. 1999a. Cancer surveillance series: inter-
tions and translational milestones for genomic tests in cancer. Genet Med, preting trends in prostate cancer. Part I: Evidence of the effects of screen-
17(6), 431–​440. PMCID: PMC4686861. ing in recent prostate cancer incidence, mortality, and survival rates. J
Chaturvedi AK, Engels EA, Anderson WF, and Gillison ML. 2008. Incidence Natl Cancer Inst, 91(12), 1017–​1024. PMID: 10379964.
trends for human papillomavirus-​related and -​unrelated oral squamous Hankey BF, Ries LA, and Edwards BK. 1999b. The surveillance, epidemiol-
cell carcinomas in the United States. J Clin Oncol, 26(4), 612–​619. ogy, and end results program:  a national resource. Cancer Epidemiol
PMID: 18235120. Biomarkers Prev, 8(12), 1117–​1121. PMID: 10613347.
Chen HS, Portier K, Ghosh K, et  al. 2012. Predicting US-​and state-​level Heckman CJ, Coups EJ, and Manne SL. 2008. Prevalence and correlates of
cancer counts for the current calendar year:  Part I:  evaluation of tem- indoor tanning among US adults. J Am Acad Dermatol, 58(5), 769–​780.
poral projection methods for mortality. Cancer, 118(4), 1091–​ 1099. PMCID: PMC2601681.
PMCID: PMC3277448. Hewitt M, and Simone J. 2000. Enhancing Data Systems to Improve the Quality
Chiang CJ, Yang YW, You SL, Lai MS, and Chen CJ. 2013. Thirty-​year out- of Cancer Care. Washington, DC: National Academy Press.
comes of the national hepatitis B immunization program in Taiwan. Hiatt RA, and Rimer BK. 1999. A new strategy for cancer control research.
JAMA, 310(9), 974–​976. PMID: 24002285. Cancer Epidemiol Biomarkers Prev, 8(11), 957–​964. PMID: 10566549.
Chimed T, Sandagdorj T, Znaor A, et  al. 2017. Cancer incidence and cancer Hiatt RA, Tai CG, Blayney DW, et  al. 2015. Leveraging state cancer reg-
control in Mongolia: results from the National Cancer Registry 2008–​12. istries to measure and improve the quality of cancer care:  a potential
Int J Cancer, 140(2), 302–​309. PMID: 27716912. strategy for California and beyond. J Natl Cancer Inst, 107(5). PMID:
Clayton D, and Schifflers E. 1987a. Models for temporal variation in cancer 25766400.
rates. I:  Age-​period and age-​cohort models. Stat Med, 6(4), 449–​467. Hill GB, Burns PE, Koch M, Lees AW, and Starreveld AA. 1983. Trends in the
PMID: 3629047. incidence of cancer of the female breast and reproductive tract in Alberta,
Clayton D, and Schifflers E. 1987b. Models for temporal variation in can- 1953 to 1977. Prev Med, 12(2), 296–​303. PMID: 6878191.
cer rates. II:  Age-​ period-​
cohort models. Stat Med, 6(4), 469–​ 481. Hoffman FL. 1915. The Mortality from Cancer Throughout the World. Newark,
PMID: 3629048. NJ: Prudential Press.
Clegg LX, Feuer EJ, Midthune DN, Fay MP, and Hankey BF. 2002. Impact of Holford TR. 1983. The estimation of age, period and cohort effects for vital
reporting delay and reporting error on cancer incidence rates and trends. J rates. Biometrics, 39(2), 311–​324. PMID: 6626659.
Natl Cancer Inst, 94(20), 1537–​1545. PMID: 12381706. Hornsby C, Page KM, and Tomlinson IP. 2007. What can we learn from the
Clemmesen J. 1948. Carcinoma of the breast: results from statistical research. population incidence of cancer? Armitage and Doll revisited. Lancet
Br J Radiol, 21(252), 583–​590. PMID: 18099749. Oncol, 8(11), 1030–​1038. PMID: 17976613.
Cook MB, Dawsey SM, Freedman ND, et al. 2009. Sex disparities in cancer Howe HL, Edwards BK, Young JL, et al. 2003. A vision for cancer incidence
incidence by period and age. Cancer Epidemiol Biomarkers Prev, 18(4), surveillance in the United States. Cancer Causes Control, 14(7), 663–​
1174–​1182. PMCID: PMC2793271. 672. PMID: 14575364.
Cunningham J, Rumbold AR, Zhang X, and Condon JR. 2008. Incidence, aeti- Howson CP, Hiyama T, and Wynder EL. 1986. The decline in gastric can-
ology, and outcomes of cancer in Indigenous peoples in Australia. Lancet cer:  epidemiology of an unplanned triumph. Epidemiol Rev, 8, 1–​27.
Oncol, 9(6), 585–​595. PMID: 18510990. PMID: 3533579.
138

138 Part II: The Magnitude of Cancer


Huckman RS, and Kelley MA. 2013. Public reporting, consumerism, and patient Parkin DM, and Moss SM. 2000. Lung cancer screening: improved survival but
empowerment. N Engl J Med, 369(20), 1875–​1877. PMID: 24131139. no reduction in deaths: the role of “overdiagnosis.” Cancer, 89(11 Suppl),
International Agency for Research on Cancer (IARC). 1966. Cancer Incidence 2369–​2376. PMID: 11147614.
in Five Continents, Vol. I. Geneva: Union Internationale Contre le Cancer. Pinheiro PS, Sherman RL, Trapido EJ, et  al. 2009. Cancer incidence in first
IARC. 2016. GLOBOCAN 2012. Available from:  http://​globocan.iarc.fr. generation U.S. Hispanics:  Cubans, Mexicans, Puerto Ricans, and
Accessed November 14, 2016. new Latinos. Cancer Epidemiol Biomarkers Prev, 18(8), 2162–​2169.
Institute of Medicine (IOM). 2013. Delivering High-​ Quality Cancer PMID: 19661072.
Care:  Charting a New Course for a System in Crisis. Washington, Poste G. 2011. Bring on the biomarkers. Nature, 469(7329), 156–​ 157.
DC: National Academy Press. PMID: 21228852.
Jemal A, Cokkinides VE, Shafey O, and Thun MJ. 2003a. Lung cancer trends Potosky AL, Miller BA, Albertsen PC, and Kramer BS. 1995. The role of
in young adults: an early indicator of progress in tobacco control (United increasing detection in the rising incidence of prostate cancer. JAMA,
States). Cancer Causes Control, 14(6), 579–​585. PMID: 12948289. 273(7), 548–​552. PMID: 7530782.
Jemal A, Saraiya M, Patel P, et al. 2011. Recent trends in cutaneous melanoma Ravdin PM, Cronin KA, Howlader N, et al. 2007. The decrease in breast-​cancer
incidence and death rates in the United States, 1992–​2006. J Am Acad incidence in 2003 in the United States. N Engl J Med, 356(16), 1670–​
Dermatol, 65(5 Suppl 1), S17–​25 e11–​e13. PMID: 22018063. 1674. PMID: 17442911.
Jemal A, Thun MJ, Ries LA, et  al. 2008. Annual report to the nation on the Ries LAG, and Devesa SS. 2006. Cancer incidence, mortality, and patient
status of cancer, 1975–​2005, featuring trends in lung cancer, tobacco survival in the United States. In:  Schottenfeld D, and Fraumeni JF Jr
use, and tobacco control. J Natl Cancer Inst, 100(23), 1672–​ 1694. (Eds.), Cancer Epidemiology and Prevention, 3rd ed. (pp. 139–​167).
PMCID: PMC2639291. New York: Oxford University Press.
Jemal A, Travis WD, Tarone RE, Travis L, and Devesa SS. 2003b. Lung cancer Robbins AS, Han X, Ward EM, et  al. 2015. Association between the
rates convergence in young men and women in the United States: analy- Affordable Care Act dependent coverage expansion and cervical can-
sis by birth cohort and histologic type. Int J Cancer, 105(1), 101–​107. cer stage and treatment in young women. JAMA, 314(20), 2189–​2191.
PMID: 12672038. PMID: 26599188.
Jemal A, Ward E, and Thun MJ. 2007. Recent trends in breast cancer incidence Rosenberg PS, and Anderson WF. 2011. Age-​period-​cohort models in can-
rates by age and tumor characteristics among U.S. women. Breast Cancer cer surveillance research:  ready for prime time? Cancer Epidemiol
Res, 9(3), R28. PMCID: PMC1929089. Biomarkers Prev, 20(7), 1263–​1268. PMCID: PMC3132831.
Klein RJ, and Schoenborn CA. 2001. Age adjustment using the 2000 pro- Rosenberg PS, Check DP, and Anderson WF. 2014. A web tool for age-​period-​
jected U.S.  population. Healthy People 2010 Stat Notes (20), 1–​ 10. cohort analysis of cancer incidence and mortality rates. Cancer Epidemiol
PMID: 11676466. Biomarkers Prev, 23(11), 2296–​2302. PMCID: PMC4221491.
Laara E, Day NE, and Hakama M. 1987. Trends in mortality from cervical Rossouw JE, Anderson GL, Prentice RL, et  al. 2002. Risks and benefits of
cancer in the Nordic countries: association with organised screening pro- estrogen plus progestin in healthy postmenopausal women:  principal
grammes. Lancet, 1(8544), 1247–​1249. PMID: 2884378. results from the Women’s Health Initiative randomized controlled trial.
Lipscomb J, and Gillespie TW. 2011. State-​level cancer quality assessment and JAMA, 288(3), 321–​333. PMID: 12117397.
research: building and sustaining the data infrastructure. Cancer J, 17(4), Rutherford MJ, Lambert PC, and Thompson JR. 2010. Age-​period-​cohort mod-
246–​256. PMID: 21799333. eling. Stata Journal, 10(4), 606–​627.
Liu S, Semenciw R, Waters C, et al. 2000. Clues to the aetiological hetero- Schneider M, Norman R, Steyn N, Bradshaw D, and South African Comparative
geneity of testicular seminomas and non-​seminomas:  time trends and Risk Assessment Collaborating G. 2007. Estimating the burden of disease
age-​period-​cohort effects. Int J Epidemiol, 29(5), 826–​ 831. PMID: attributable to low fruit and vegetable intake in South Africa in 2000. S
11034964. Afr Med J, 97(8 Pt 2), 717–​723. PMID: 17952229.
Locke FB, and King H. 1980. Cancer mortality risk among Japanese in the SEER. 2015. Surveillance, Epidemiology, and End Results (SEER) Program
United States. J Natl Cancer Inst, 65(5), 1149–​1156. PMID: 6933246. (www.seer.cancer.gov) SEER*Stat Database: Incidence—​SEER 18 Regs
Loeb S, Bjurlin MA, Nicholson J, et al. 2014. Overdiagnosis and overtreatment Research Data + Hurricane Katrina Impacted Louisiana Cases, Nov
of prostate cancer. Eur Urol, 65(6), 1046–​1055. PMCID: PMC4113338. 2014 Sub (2000–​2012) Katrina/​Rita Population Adjustment—​Linked To
Martinez-​Tyson D, Pathak EB, Soler-​Vila H, and Flores AM. 2009. Looking County Attributes—​Total U.S., 1969–​2013 Counties, National Cancer
under the Hispanic umbrella: cancer mortality among Cubans, Mexicans, Institute, DCCPS, Surveillance Research Program, Surveillance Systems
Puerto Ricans and other Hispanics in Florida. J Immigr Minor Health, Branch, released April 2015, based on the November 2014 submission.
11(4), 249–​257. PMCID: PMC3086376. Shin H, Curado M, Ferlay J, et  al. 2007. Comparability and quality of data.
Mathers CD, Fat DM, Inoue M, Rao C, and Lopez AD. 2005. Counting In: Curado MP, Edwards B, Shin HR, Storm H, Ferlay J, Heanue M, and
the dead and what they died from:  an assessment of the global sta- Boyle P (Eds.), Cancer Incidence in Five Continents, Vol. IX. http://​www.
tus of cause of death data. Bull World Health Organ, 83(3), 171–​177. iarc.fr/​en/​publications/​pdfs-​online/​epi/​sp160/​CI5vol9-​5.pdf. Accessed
PMCID: PMC2624200. July 14, 2015.
Moore SP, Antoni S, Colquhoun A, et  al. 2015. Cancer incidence in indige- Siegel R, Desantis C, and Jemal A. 2014. Colorectal cancer statistics, 2014. CA
nous people in Australia, New Zealand, Canada, and the USA:  a com- Cancer J Clin, 64(2), 104–​117. PMID: 24639052.
parative population-​ based study. Lancet Oncol, 16(15), 1483–​ 1492. Siegel R, Naishadham D, and Jemal A. 2013. Cancer statistics, 2013. CA
PMID: 26476758. Cancer J Clin, 63(1), 11–​30. PMID: 23335087.
Moore SP, O’Rourke PK, Mallitt KA, et al. 2010. Cancer incidence and mor- Siegel RL, Fedewa SA, Miller KD, et al. 2015a. Cancer statistics for Hispanics/​
tality in Indigenous Australians in Queensland, 1997–​2006. Med J Aust, Latinos, 2015. CA Cancer J Clin, 65(6), 457–​480. PMID: 26375877.
193(10), 590–​593. PMID: 21077815. Siegel RL, Miller KD, and Jemal A. 2016. Cancer statistics, 2016. CA Cancer
Muir CS. 1996. Epidemiology of cancer in ethnic groups. Br J Cancer Suppl, J Clin, 66(1), 7–​30. PMID: 26742998.
29, S12–​S16. PMCID: PMC2149849. Siegel RL, Sahar L, Robbins A, and Jemal A. 2015b. Where can colorectal
National Cancer Institute (NCI). 2015. Cancer Query System: Historic SEER cancer screening interventions have the most impact? Cancer Epidemiol
Delay-​Adjustment Ratios. Available from: http://​surveillance.cancer.gov/​ Biomarkers Prev, 24(8), 1151–​1156. PMID: 26156973.
delay/​canques.html. Accessed July 15, 2015. Simard EP, Ward EM, Siegel R, and Jemal A. 2012. Cancers with increasing
NCI. 2016. Surveillance, Epidemiology, and End Results Program (SEER). incidence trends in the United States: 1999 through 2008. CA Cancer J
Available from: www.seer.cancer.gov. Accessed November 14, 2016. Clin, 62(2), 118–​128. PMID: 22281605.
Paik DC, Saborio DV, Oropeza R, and Freeman HP. 2001. The epidemiological Spinks TE, Walters R, Feeley TW, et al. 2011. Improving cancer care through
enigma of gastric cancer rates in the US: was grandmother’s sausage the public reporting of meaningful quality measures. Health Aff (Millwood),
cause? Int J Epidemiol, 30(1), 181–​182. PMID: 11171883. 30(4), 664–​672. PMID: 21471487.
Parkin DM. 2006. The evolution of the population-​based cancer registry. Nat Storm HH. 1996. Cancer registries in epidemiologic research. Cancer Causes
Rev Cancer, 6(8), 603–​612. PMID: 16862191. Control, 7(3), 299–​301. PMID: 8734821.
Parkin DM. 2008. The role of cancer registries in cancer control. Int J Clin Tangka F, Subramanian S, Beebe MC, Trebino D, and Michaud F. 2010.
Oncol, 13(2), 102–​111. PMID: 18463952. Economic assessment of central cancer registry operations. Part III:
Parkin DM, and Bray F. 2006. International patterns of cancer incidence Results from 5 programs. J Registry Manag, 37(4), 152–​155. PMID:
and mortality. In:  Schottenfeld D, and Fraumeni JF, Jr (Eds.), Cancer 21688745.
Epidemiology and Prevention, 3rd ed. New York: Oxford University Press. Thun MJ, Carter BD, Feskanich D, et  al. 2013. 50-​year trends in smoking-​
Parkin DM, and Khlat M. 1996. Studies of cancer in migrants: rationale and related mortality in the United States. N Engl J Med, 368(4), 351–​364.
methodology. Eur J Cancer, 32A(5), 761–​771. PMID: 9081351. PMCID: PMC3632080.
 139

Patterns of Cancer Incidence, Mortality, and Survival 139


Thun MJ, and Jemal A. 2006. How much of the decrease in cancer death rates Welch HG, and Albertsen PC. 2009. Prostate cancer diagnosis and treatment
in the United States is attributable to reductions in tobacco smoking? Tob after the introduction of prostate-​specific antigen screening: 1986–​2005.
Control, 15(5), 345–​347. PMCID: PMC2563648. J Natl Cancer Inst, 101(19), 1325–​1329. PMCID: PMC2758309.
Vaccarella S, Franceschi S, Bray F, et al. 2016. Worldwide thyroid-​cancer epi- Welch HG, and Black WC. 2010. Overdiagnosis in cancer. J Natl Cancer Inst,
demic? The increasing impact of overdiagnosis. N Engl J Med, 375(7), 102(9), 605–​613. PMID: 20413742.
614–​617. PMID: 27532827. World Health Organization (WHO). 1992. International Statistical
Vaccarella S, Franceschi S, Engholm G, et al. 2014. 50 years of screening in the Classification of Diseases and Related Health Problems, Tenth Revision.
Nordic countries: quantifying the effects on cervical cancer incidence. Br Geneva, Switzerland: WHO.
J Cancer, 111(5), 965–​969. PMCID: PMC4150271. Wingo PA, Howe HL, Thun MJ, et al. 2005. A national framework for cancer
Verhoeven R, Houterman S, Kiemeney B, Koldewijn E, and Coebergh JW. surveillance in the United States. Cancer Causes Control, 16(2), 151–​
2008. Testicular cancer: marked birth cohort effects on incidence and a 170. PMID: 15868456.
decline in mortality in southern Netherlands since 1970. Int J Cancer, Wiren S, Haggstrom C, Ulmer H, et al. 2014. Pooled cohort study on height and
122(3), 639–​642. PMID: 17764111. risk of cancer and cancer death. Cancer Causes Control, 25(2), 151–​159.
Vickers AJ, Sjoberg DD, Ulmert D, et al. 2014. Empirical estimates of prostate PMCID: PMC3929024.
cancer overdiagnosis by age and prostate-​specific antigen. BMC Med, 12, Wu S, Powers S, Zhu W, and Hannun YA. 2016. Substantial contribution of
26. PMCID: PMC3922189. extrinsic risk factors to cancer development. Nature, 529(7584), 43–​47.
Wagner G. 1985. The Role of the Registry in Cancer Control. Parkin DM, PMID: 26675728.
Wagner G, and Muir CS (Eds.). Lyon: No. 66 (IARC). Zhu L, Pickle LW, Ghosh K, et al. 2012. Predicting US-​and state-​level can-
Walter RB, Brasky TM, Buckley SA, Potter JD, and White E. 2013. Height as cer counts for the current calendar year. Part II: evaluation of spatiotem-
an explanatory factor for sex differences in human cancer. J Natl Cancer poral projection methods for incidence. Cancer, 118(4), 1100–​ 1109.
Inst, 105(12), 860–​868. PMCID: PMC3687370. PMID: 22228583.
Ward E, Jemal A, and Thun M. 2005. Regarding “Increase in breast cancer inci-
dence in middle-​aged women during the 1990s.” Ann Epidemiol, 15(6),
424–​425; author reply 426-​427. PMID: 15967388.
140
 14

9 Socioeconomic Disparities in Cancer Incidence and Mortality

CANDYCE KROENKE AND ICHIRO KAWACHI

OVERVIEW that treat people differently depending on their diverse socioeconomic


backgrounds.
The relationship between socioeconomic status (SES) and cancer is Rather than simply reflecting health disparities at a single time
complex, dynamic, and evolving. Associations depend on SES mea- point, however, this theoretical model helps to explain secular trends
sures, cancer type, sociodemographic factors including race/​ethnicity, in socioeconomic disparities, which may emerge or widen with dispa-
and historical trends. However, socioeconomic disadvantage is often rate gains in knowledge or adoption of practices across SES groups.
associated with a higher risk of cancer, particularly cancers diagnosed Disparities may narrow as new knowledge diffuses through the popula-
at a late stage, as well as worse prognosis once diagnosed. Research tion and as low-​SES individuals adopt practices that are initially more
on secular trends over the past 70  years has shown reversals of the common among those of high SES, such as screening for colorectal
socioeconomic gradient for lung and colorectal cancer consistent with cancer, or when efforts are targeted to improve outcomes among the
differential trends by SES in patterns of smoking, diet, and obesity. socioeconomically disadvantaged. However, disparities have often
Rates of these cancers are now currently higher in socioeconomi- widened because those of higher SES learn, incorporate, and benefit
cally disadvantaged groups. SES is considered to be a “fundamental” from new information about cancer prevention before those of lower
determinant of health outcomes, and this appears true throughout the SES do.
cancer spectrum—​from cancer incidence, to detection, treatment, and Exceptions to these rules do exist (e.g., low and late parity among
survival. Population-​level research on SES and cancer still predomi- women of high SES, leading to a higher risk of breast cancer)
nantly employs area-​level measures (as a proxy for individual SES), (Pudrovska & Anikputa, 2012). However, rather than high SES rep-
though investigations over the past decade have increasingly consid- resenting adversity, the absence or reversal of a socioeconomic dis-
ered the simultaneous impact of individual SES and area-​level SES (as parity often reflects lack of knowledge about a particular cause of
a contextual influence) on health outcomes. Investigations of the link cancer or mortality risk factors. Evidence suggests that once associa-
between SES and cancer will continue to benefit from more sophisti- tions are elucidated and disseminated, those of high SES are the first
cated approaches to the measurement of SES, including evaluation of to take advantage of novel findings, resulting in widening disparities
socioeconomic trajectories and modeling of cumulative disadvantage (Eckersley, Dixon, & Douglas, 2001). Thus, the relationship between
over the life course. Programs or policies designed to reduce the bur- SES and health is dynamic, and may change (or even reverse) over
den of cancer need to pay heed to reducing and ultimately eliminating time. Major risk factors for cancer, such as cigarette smoking, physical
socioeconomic disparities. inactivity, and obesity, were each more prevalent among higher SES
groups in industrialized societies during earlier periods of economic
development, but the patterns reversed during more recent history so
INTRODUCTION that the same risk factors now exhibit higher prevalence among disad-
vantaged groups (Eckersley et al., 2001).
The association between socioeconomic status (hereafter abbreviated There is an important exception to this generalization:  the direc-
as SES) and health status is so robust and consistent that epidemiolo- tion of causation does not uniformly run from SES to health. Some
gists routinely adjust for it as a potential confounding variable when evidence suggests that health status can exert influences on socioeco-
evaluating the etiological role of other risk factors for disease. The nomic attainment, such as when a diagnosis of cancer early in life
present chapter turns this logic on its head, to focus on SES as a fun- adversely impacts educational attainment and employment prospects,
damental determinant of disease, specifically cancer incidence and particularly in women (D. Lee & Jackson, 2015). In fact, socioeco-
mortality. nomic attainment and health status exert reciprocal influences on each
The association between SES and health status has been recorded other, though the current evidence more often supports the influence of
throughout history. William Farr, viewed as an intellectual founder of SES on cancer than the reverse (DiMartino, Birken, & Mayer, 2016).
epidemiology, documented the existence of socioeconomic disparities When describing the relationships between SES and cancer out-
in mortality in nineteenth-​century Britain, and concluded that “[n]‌o comes, it is important to be specific about the exact site and type of
variation in the health of the states of Europe is the result of chance; cancer, as well as the class of outcome measure (incidence versus
it is the direct result of the physical and political conditions in which survival) (Krieger, 2005). The incidence of some cancers, notably
nations live” (quoted in Beaglehole & Bonita, 1997). The persistent breast cancer and melanoma, is higher among more advantaged SES
and pervasive association between lower socioeconomic position and groups, presumably reflecting the underlying socioeconomic distri-
worse health status accounts for the belief that SES may be a “fun- bution of risk factors. For breast cancer, the higher incidence among
damental” determinant of individual and population health; no mat- higher SES women has been attributed to reproductive factors includ-
ter what the current health threats confronted in any given society, ing earlier age at menarche, later age at first birth, and lower fertility.
disadvantaged groups are worse off in terms of their achieved health However, recent secular trends show that age at menarche, declining
(Phelan, Link, & Tehranifar, 2010; Sistino & Ellis, 2011). overall, is falling at a faster rate among girls of low SES. Moreover,
Freese and Lutfey (2011) identified four meta-​mechanisms by which with the publication of research about the adverse cardiac effects of
SES is linked to health: (1) the means by which individuals use SES-​ hormone therapy, a known risk factor for breast cancer, the use of hor-
related resources to gain a health advantage; (2) spillovers, in which mone therapy declined markedly (Hersh, Stefanick, & Stafford, 2004;
people benefit from being embedded in contexts in which others (e.g., Steinkellner et al., 2012), particularly among women from advantaged
spouses, neighbors, coworkers) are attentive to health concerns; (3) a socioeconomic backgrounds, notable because initial use of hormone
health habitus, in which socially structured preferences for a “healthy therapy in low-​SES women was lower (Krieger, Chen, & Waterman,
lifestyle” shape health outcomes; and (4) the influence of institutions 2010). On the other hand, survival following the diagnosis of breast

141
142

142 Part II: The Magnitude of Cancer


cancer consistently favors higher SES women, due, among other rea- with and “navigate” medical care systems and options (Yen & Moss,
sons, to earlier detection, better access to effective treatment (Lochner 1999). Income, on the other hand, enables individuals to purchase the
& Kawachi, 2000), and beneficial social networks (C. H.  Kroenke, various goods and services that are necessary for maintaining and pro-
Kubzansky, Schernhammer, Holmes, & Kawachi, 2006; C. H. Kroenke moting health. Occupational status often determines an individual’s
et al., 2013). exposure to hazards (including carcinogens) in the workplace (Pearce
This chapter expands upon the previous version published over a & Matos, 1994), in addition to access to psychosocial resources, such
decade ago. While the overall literature has not changed markedly as prestige, respect, and security. Wealth can help to cushion eco-
(i.e., low SES continues to predict worse cancer incidence and out- nomic shocks in the case of job loss, minimize psychosocial risk (e.g.,
comes generally), particular associations or changing trends between depressive symptoms, anxiety) associated with economic insecurity,
SES and cancer reflect changing secular trends in behaviors and other or enhance social capital. In the past decade, researchers have endeav-
factors. This chapter will cover those discussions, as well as the advent ored to more carefully distinguish and clarify the mechanisms for each
of new kinds of research (e.g., epigenetics), which may help shed light variable so that researchers can be more thoughtful about the ways in
on the relationships between SES and cancer. We have also updated which they measure SES (P. A. Braveman et al., 2005) and their inter-
literature when available, particularly data incorporating secular trends related influences over the life course.
and new insights. Finally, we will briefly describe needed research in The example of cigarette smoking illustrates some potential pitfalls
the area and consider future trends. of the uncritical use of SES measures. Higher educational attainment is
The present chapter is organized into four major sections. The strongly inversely associated with cigarette smoking in most advanced
first section will define the concept of SES and describe the various industrialized societies, including the United States (Pamuk, Makuc,
approaches to its measurement. The second section will summarize Heck, Reuben, & Lochner, 1998). The inverse association between
the observations and updates on the general nature of the associations smoking and education is thought to be mediated by knowledge about
between area-​level and individual-​level SES, and cancer morbidity, the hazards of cigarette smoking. In support of this hypothesis, an
mortality, and survival. The third section will outline the general cat- association between education and smoking among young adults did
egories of explanations, both causal and non-​causal, that have been put not exist prior to 1953, when the health hazards of smoking had yet
forward to account for the association between SES and cancer. The to be established (Fuchs, 1983). Educational differentials in smoking
fourth section will provide a survey of the specific causal mechanisms emerged soon after the first (1964) US Surgeon General’s report on
underlying the relationship between SES and cancer. This section is smoking and health. Higher incomes are also correlated with a lower
organized into two subsections dealing, respectively, with early life prevalence of smoking, although less strongly than is the case with
and adult risk factors that potentially account for the observed dispari- education (Pamuk et al., 1998). Increased income should result in the
ties in cancer incidence. We conclude the chapter with a summary and enhanced ability to afford more goods, including cigarettes, all other
thoughts about needs for future research. things being equal. Historically, during the economic development
of societies, as real personal income rose, so did per capita cigarette
consumption.
WHAT IS SES? DEFINITIONS AND MEASUREMENT Also, within certain subgroups in the population, such as teenag-
ers, higher incomes (in the form of more pocket money, for example)
Sociologists and allied social scientists have studied social strati- have been shown to be associated with more cigarette consumption
fication and its consequences for over 150  years (Grusky, 1994). (Scragg, Laugesen, & Robinson, 2002). And in fact, the implementa-
Epidemiologists who study the effects of social stratification on health tion of excise taxes to raise the price of cigarettes has been shown
have, for the most part, followed the conceptual approach established to decrease cigarette consumption particularly among youth (Kostova
by Max Weber (1864–​ 1920) (Liberatos, Link, & Kelsey, 1988). et al., 2014; van Hasselt et al., 2015) and those with lower disposable
According to Weber, “socioeconomic status” is defined as groups income (Levy, Mumford, & Compton, 2006). So why is higher income
within society that are relatively homogeneous with respect to life associated with lower cigarette consumption?
chances and opportunities (Grusky, 1994). In turn, life opportunities The reason is unlikely to be due to a direct causal effect of income
(and hence, SES groups) have been measured by socioeconomic indi- on smoking. Rather, higher incomes are most likely associated with
cators, such as educational attainment (years of schooling, educational other variables that reduce the probability of smoking. For example,
credentials gained), income and wealth, as well as occupational sta- teenagers with greater discretionary dollars to spend are also more
tus or prestige (Lynch & Kaplan, 2000). SES is therefore a multidi- likely to come from higher socioeconomic backgrounds, so that the
mensional concept that is intended to capture the manifold, and often effect of higher parental education (and stronger family socialization),
subtle, differences between individuals with respect to their access to as well as social norms associated with higher education and prohibi-
life opportunities. Other terms that are commonly used in the social tions on smoking, may override the effect of having more money on
epidemiologic literature to characterize social stratification include cigarette consumption. Put simply, education and income may influ-
socioeconomic position (SEP) and social class, among others. While ence cigarette smoking through different, multiple, and even coun-
each term has a different theoretical meaning and origin (see Glymour, tervailing mechanisms. Care is therefore warranted in specifying the
Avendano, & Kawachi, 2014), we specifically use the broader term indicator of SES used when describing its association with health out-
“socioeconomic status” (SES), because it encompasses economic/​ comes. For the remainder of the chapter, the term “socioeconomic dis-
material resources (e.g., income, wealth) as well as psychosocial dif- parities” will be used as a broad descriptive term to refer to variations,
ferences (e.g., occupational prestige), each of which influence health inequalities, or differences in cancer outcomes by SES. The specific
over the life course (C. Kroenke, 2008). indicator of SES used will be described wherever appropriate.
Individual-​level measures, including education, occupation, Three additional points are worth noting with respect to the mea-
income, and wealth, are linked across the life course, so that more surement of SES. First, SES can be conceptualized and measured at
schooling determines an individual’s ability to access higher status both the individual level and at the area level (for example, the neigh-
jobs, which in turn determines the rate of monetary compensation, borhoods in which individuals reside). A great deal of recent research
and so on. It is not surprising, then, that indicators of SES are usually has made use of area-​level socioeconomic measures, including median
found to be correlated with each other. That said, epidemiologists have household income from Census tracts; average SES of residents; and
often used three of the individual indicators of SES (education, occu- neighborhood social, physical, and food environments. An individual
pation, income) in an interchangeable manner, even though they may with a given level of income or educational attainment could experi-
be in fact linked to health outcomes through different mechanisms. ence different chances of health depending upon the average SES level
For example, schooling and education are believed to influence health of his or her neighborhood (for example, as measured by the median
through the acquisition of knowledge in one or more forms—increased household income of a Census tract). Capturing area-​level SES is
knowledge about health-​promoting behaviors, enhanced ability to pro- therefore of intrinsic interest over and above measuring individual
cess and act upon that information, as well as the ability to interact SES. This is a different argument from using area SES as a proxy for
 143

Socioeconomic Disparities in Cancer Incidence and Mortality 143


individual SES when the latter measures are not available on data millennials have been shown to live with their parents at a higher rate
sets. The reason that area SES is believed to contribute independently than previous generations (Fry, 2015); parental resources may buffer
to health outcomes is based on the differential quality of neighbor- some of the socioeconomic difficulties. Future work may incorporate
hood environments according to the average SES level of its resi- new SES measures as current individual-​and neighborhood-​ level
dents, a phenomenon referred to as residential segregation (Kawachi measures fail to capture familial resources.
& Berkman, 2003). Examples of such neighborhood-​level patterning An additional point worth mentioning about the measurement
include differential access to services and amenities (e.g., mammog- of socioeconomic disparities in health is in regard to its distinction
raphy screening services, supermarkets that sell fresh fruits and veg- from racial disparities in health. This point is particularly pertinent
etables); the physical environment (e.g., air pollution, the presence of in the United States, where official statistics have often conflated
parks and recreational playgrounds for physical activity, differential racial disparities in health with socioeconomic disparities (Williams,
exposures to outdoor tobacco advertising); and the social context (e.g., 1997). Race and SES are not synonymous or interchangeable. Race is
social support among neighbors) (Kawachi & Berkman, 2003). In emphatically not a proxy for SES, despite the fact that racial minori-
turn, the differential distribution of these protective and risk factors at ties in the United States are overrepresented among lower SES groups,
the neighborhood level is thought to contribute to the disease risks of stemming from discrimination and the denial of opportunities through-
residents, including cancer risk. Since the publication of the previous out history. The finding that race is often associated with morbidity
version of this chapter, a growing number of articles have examined and mortality independent of SES emphasizes that race is more than
the independent effects of area-​level and individual-​level SES mea- SES. Accordingly, understanding the sources of racial disparities in
sures. Investigators have often, though not always, found independent health requires attention to a separate set of causes and mechanisms,
effects of these, though area-​level effects are often more weakly asso- including the potential influence of racial discrimination on health out-
ciated than individual-​level SES factors with cancer outcomes. comes. Fortunately, this has been increasingly reflected in the research
Second, in addition to the dimension of place, researchers have literature. There is also the growing realization that elucidating pat-
increasingly emphasized the importance of the dimension of time in terns of health by socioeconomic status may be insufficient to under-
conceptualizing and measuring SES and its effects on health. SES sel- stand and describe those patterns and that ultimately, research may be
dom remains static across the life course. Individuals exhibit upward needed to understand how the combination of SES and race, as well as
(or downward) social mobility, for example, when they quit jobs to other social factors (e.g., urban/​rural status, immigrant status/​nativity),
return for more schooling, when they are involuntarily laid off, when influence patterns of associations.
they are promoted to higher positions, and so on. Accordingly, the
measurement of SES at any single point in time is unlikely to capture
the dynamic, as well as cumulative, effects of SES on health. Income The SES Gradient in Cancer Incidence and Mortality
dynamics, in the form of accumulated spells of poverty, have been
shown to predict mortality and other health outcomes (McDonough, Individuals from lower SES backgrounds, whether measured by edu-
Duncan, Williams, & House, 1997). Childhood socioeconomic cir- cational attainment, income and wealth, or occupational status, gen-
cumstances have been shown to predict health outcomes in later life erally experience worse health outcomes than those higher on the
(Davey-​ Smith, Gunnell, & Ben-​ Shlomo, 2001), including cancer socioeconomic hierarchy (Adler et  al., 1993). This widely observed
(Vohra, Marmot, Bauld, & Hiatt, 2016), independently of, or in com- pattern of the rise in health status with each level of SES has been
bination with, SES attained in adulthood. referred to as the SES “gradient” in health. It is a “gradient” because
Beyond the fact that SES can change over time, there are other there is no apparent threshold or cut-​point in the relationship between
important life course considerations. First, there are cumulative effects SES and health, that is, the excess morbidity and mortality risks of
of low SES on health outcomes. The concept of “allostatic load” cap- disadvantaged groups are not solely confined to those who are poor by
tures the accumulated “wear and tear” on the body’s physiologic sys- officially defined criteria. Instead, at each level of the SES hierarchy,
tems as a result of lifelong exposure to adverse social circumstances people experience better health compared to those immediately below
(Kubzansky, Seeman, & Glymour, 2014). Second, there are believed them, even among groups that are considered middle class and above.
to be “critical periods” in development during which socioeconomic
disadvantage may have particularly adverse impacts on the trajecto-
ries of subsequent health (C. Kroenke, 2008). Though socioeconomic SOCIOECONOMIC STATUS AND CANCER: 
disadvantage at any point in the life course can result in health insults, GENERAL REMARKS
low SES during infancy and childhood may have profound effects
on health. For example, spells of income poverty during the prenatal Socioeconomic disadvantage, whether measured by low income, low
and infancy period have been linked to increased risk of adult obesity, educational attainment, low occupational status, or area-​level socio-
more so than exposure during subsequent periods of childhood (Ziol-​ economic measures, has been linked with both higher overall cancer
Guest, Duncan, & Kalil, 2009). incidence and mortality (Doubeni, Laiyemo, et  al., 2012; Enewold,
There are, additionally, powerful cohort effects that influence socio- Horner, Shriver, & Zhu, 2014; Harper et al., 2009; Hastert, Beresford,
economic trajectories over the life course with possible ramifications Sheppard, & White, 2015; Krieger et al., 2006; Ma, Xu, Anderson, &
for cancer and other health outcomes. In work by Elder, military con- Jemal, 2012). However, in contrast to the SES gradient reported for
scription during World War II of men in their twenties and thirties as other major health outcomes, such as cardiovascular disease (CVD)
they were establishing themselves in their careers profoundly altered or infectious diseases, the SES gradient across cancer incidence and
their life chances as compared with those who were conscripted in mortality is often not as strong or consistent (Hastert et  al., 2015;
their late teens (G. H. Elder, Jr., Shanahan, & Clipp, 1994). They had Steenland, Henley, & Thun, 2002).
lower lifetime earnings and attainment of wealth and a higher divorce Three major reasons for the weaker associations between SES
rate. Late entrants were shown to have higher mortality than early and overall cancer incidence and mortality are (a)  the heterogeneity
entrants due to these factors (G. H.  Elder, Clipp, Brown, Martin, & of associations between SES and specific cancer sites; (b) the length
Friedman, 2009). Though the consequences for cancer were not spe- of the induction period between exposure to low SES conditions and
cifically addressed, a major mechanism to adverse health outcomes cancer onset; and (c) differences that reflect the current state of knowl-
included alcohol consumption, a known predictor of several types of edge about CVD and cancer (i.e., we know more about CVD preven-
cancer. tion and treatment than we do about cancer prevention and treatment).
In more recent history, crushing levels of educational debt and com- With regard to the first point, cancer (unlike CVD) involves a highly
promised job prospects, particularly among “millennials” who gradu- heterogeneous mix of diseases that are caused by different sets of risk
ated around the 2008 downturn, threaten to limit their earning and factors, many of which may be unrelated to, or related in opposite
occupational potential compared with earlier cohorts. It is yet uncer- directions to, SES. Thus, an association between overall cancer inci-
tain how such differences will influence health outcomes since adult dence and/​or mortality with SES at any given point in time reflects
14

144 Part II: The Magnitude of Cancer


the weighted contribution of the component sites, each of which may information on income. Individual-​level socioeconomic data are not
have a different relationship with SES. For example, as the contribu- routinely available in patient hospital records.
tion of lung cancer deaths outgrows the contribution of breast cancer In recent years, there have been increasing calls—​as from the
in overall cancer mortality rates for US women, the SES gradient in Institute of Medicine’s Committee on Public Health Strategies to
total cancer deaths can be expected to grow stronger (G. K.  Singh, Improve Health (Institute of Medicine, 2011)—​for more compre-
Miller, Hankey, Feuer, & Pickle, 2002). The analysis of SES gradi- hensive approaches to collecting individual and community SES
ents in overall cancer incidence and mortality is therefore of limited measures to improve public health surveillance (P. Braveman, 2011;
value in understanding etiological relationships, though it may serve a P. A. Braveman et al., 2005; Gottlieb, Sandel, & Adler, 2013; Koh,
broader purpose in monitoring population trends in health disparities. 2011; Sadana & Harper, 2011). Work has also been done to link
With regard to the second point, most cancers (unlike CVD) are large cohort data sets with data available on individual SES mea-
associated with lengthy induction periods between exposure (in this sures to population-​ level Surveillance, Epidemiology, and End
case, to the living conditions associated with low SES) and the onset Results (SEER) data with area-​level measures (Clegg et al., 2009;
of disease. Induction periods for cancers are typically on the order Doubeni, Laiyemo, et  al., 2012; Du, Lin, Johnson, & Altekruse,
of decades, whereas they may be just a few years for heart disease 2011; Hastert et  al., 2015). Insights from this work suggest that
or infectious diseases, or other health outcomes that exhibit strong area-​and individual-​level socioeconomic variables exert indepen-
SES gradients. The length of the induction period for most cancers dent effects on outcomes, though specific effects differ by cancer,
produces measurement error in exposure status, as individuals move study, and country (C. M.  Chang et  al., 2012). In the absence of
out of poverty, or experience upward job mobility, and so forth, with individual-​level SES information, US researchers have contin-
the result that the association between SES and cancer outcomes may ued to examine area-​level (e.g., county, zip code, census tract, or
be biased in the direction of the null. Also, it may take decades for block group) SES disparities in cancer incidence as a proxy for
a socioeconomic gradient in cancer mortality (e.g., lung cancer) to individual SES (Harper et  al., 2009; Hastert et  al., 2015; Krieger
emerge, even after SES gradients in health behaviors (e.g., smoking) et al., 2006; L. Liu, Cozen, Bernstein, Ross, & Deapen, 2001; Tao,
have become well established—​such was the case for the educational Ladabaum, Gomez, & Cheng, 2014). More recent studies are begin-
gradient in lung cancer mortality among women in the United States ning to examine the independent or combined effects of area-​and
(Steenland et al., 2002). Changes in behaviors impact CVD event rates individual-​level SES on cancer incidence (Doubeni, Schootman,
considerably faster than they do cancer rates. et al., 2012; Hastert et al., 2015) and cancer mortality (C. M. Chang
However, it is important to note that trends in socioeconomic et  al., 2012; Enewold et  al., 2014)  or individual-​level SES factors
disparities in cardiovascular factors could portend future trends in and cancer outcomes (Clegg et al., 2009; Kinsey, Jemal, Liff, Ward,
socioeconomic disparities for certain types of cancer. Kanjilal and & Thun, 2008; Ma et al., 2012).
colleagues (Kanjilal et al., 2006) found between 1971 and 2002 that
while CVD risk factors, including high blood pressure, high choles-
terol, and smoking, declined at all socioeconomic levels, socioeco- SES AND CANCER INCIDENCE
nomic disparities widened (on a difference scale) for smoking and
high cholesterol because of greater improvements among those of Cancer in Adults
high SES. Socioeconomic disparities also widened for type 2 diabe-
tes, but this was due to larger increases among those of low SES. With regard to incidence rates, the cancer sites in adults exhibiting
Smoking and diabetes are each risk factors for cancer, portending the strongest and most consistent associations with low SES are
widening future cancer disparities for lung cancer and gastrointesti- cancers of the lung, upper aerodigestive tract (e.g., oral, esopha-
nal and other cancers influenced by metabolic dysregulation, even if geal, laryngeal), stomach, and cervix (Boscoe, Henry, Sherman, &
overall cancer incidence declines. Researchers should nonetheless be Johnson, 2016; Clegg et  al., 2009; Faggiano, Partanen, Kogevinas,
cognizant that trends in disparities on relative versus absolute scales & Boffetta, 1997; Kogevinas, Pearce, Susser, & Boffetta, 1997;
can yield diverging conclusions about whether disparities are increas- Kogevinas & Porta, 1997; Lochner & Kawachi, 2000; Merletti,
ing or decreasing. Galassi, & Spadea, 2011). Low SES also appears to be related to
With regard to the third point, there is a great deal we do not know a higher incidence of liver cancer (E. T. Chang et al., 2010; Shebl,
about the causes of, or how to screen for, certain types of cancer, as Capo-​Ramos, Graubard, McGlynn, & Altekruse, 2012). By contrast,
reflected by the very high mortality rates for certain kinds of cancer, hormonally related cancers, such as prostate and breast cancer, and
including pancreatic and ovarian cancers. As knowledge around risk melanoma are cancers in which incidence rates appear to be higher
factors, treatment, and methods of screening have improved, there is in individuals of higher SES (Boscoe et al., 2016; Clegg et al., 2009;
evidence that socioeconomic disparities favoring those of higher SES Faggiano et al., 1997). Direct associations of SES with colon cancer
have increased. Those cancers for which this statement is true include and ovarian cancer were previously reported (Kogevinas & Porta,
cancers more amenable to improvements among those of lower SES. 1997). However, inverse relationships of SES with colon and ovar-
ian cancer have been reported in more recent years (Praestegaard
et al., 2016). Gaps in prostate and breast cancer incidence have been
Notes on Area-​Level and Individual-​Level shrinking (Krieger, Chen, Kosheleva, & Waterman, 2012), but low
SES is associated with higher risks of late-​stage breast and prostate
SES and Cancer
cancers. Patterns of association between SES and cancer incidence
In the previous edition of this book, we reported a dearth of population-​ are summarized in work by Boscoe and colleagues (Boscoe et  al.,
level cancer incidence studies in the United States that had data on 2016) (Figure 9–​1). In this diagram, lower SES is related to a higher
SES at the individual level. This continues to be true, due in large risk of several cancers, but most particularly, late-​stage cancers.
part to the lack of socioeconomic information available within official Higher SES is related to a higher risk of several types of cancer, and
sources of data, including cancer registries. In the 1990s, Krieger and these are generally local tumors.
colleagues (Krieger, Chen, & Ebel, 1997)  conducted a survey of all
cancer registries in the United States to ascertain the availability of Area-​Level SES and Cancer Incidence
SES information. Of the 45 state cancer registries that responded to In the previous version of the chapter, we reported on area-​level find-
their mailed survey, 36 (80%) collected some information on occu- ings and incident cancer in a study by Krieger and colleagues (Krieger
pation, but only 4 routinely reported occupational data, reflecting the et  al., 1999). They examined associations between cancer incidence
constraints posed by the accuracy and reliability of socioeconomic and SES, measured at the Census block group level in California’s San
data available from patients’ medical records (the principal source Francisco Bay area. A Census block group is a division of a Census
of cancer registry data). Only two of the state cancer registries col- tract, typically containing 1000 residents, with boundaries drawn to
lected information on education, and none of the registries collected maximize social homogeneity. The results of this study, which were
 145

Socioeconomic Disparities in Cancer Incidence and Mortality 145


0.5 1.0 1.5 2.0 2.5 3.0
Larynx 3.2
Cervix
HPV-related
Oral
Liver
Lung
Tobacco-related
Esophagus
Prostate
Kidney
Bladder
Colorectal
Female breast
Stomach
All stageable cancers
Testis
Brain Distant
Uterus Local

Melanoma
HL
Pancreas
Thyroid
Ovary
NHL
0.5 1.0 1.5 2.0 2.5 3.0

Figure 9–​1.  Relative risk of cancer incidence between highest (over 20%) and lowest (<5%) poverty category, based on census tract, by site and stage,
United States, 2005–​2009. Source: Boscoe (2015).

also stratified by race/​ethnicity, indicated a strong area-​SES gradient Individual-​Level SES and Cancer Incidence
for lung cancer and cervix cancer with lower SES related to higher Few studies have evaluated individual-​level socioeconomic factors and
cancer risks. However, and counter to other race/​ethnic groups, there surveillance data because SES data are not available in patient hospital
was a positive association between higher area SES and excess risk of records and are therefore not routinely reported by cancer registries
lung cancer in Hispanic women and men. The authors speculated that in the United States. However, in 1999, the National Cancer Institute
this Hispanic “paradox” may reflect the persistently higher smoking (NCI) initiated a study resulting in the linkage of population-​based
prevalence among professional classes in immigrant populations from SEER cancer registry data to data from the US representative National
Latin America (Krieger et al., 1999). Longitudinal Mortality Study (NLMS) study with self-​ reported
In a more recent study using data from the California Cancer socioeconomic data from the Social and Economic Supplement to
Registry, Yin and colleagues (Yin et  al., 2010)  corroborated the the Census Bureau’s Current Population Survey (CPS). Using these
apparent paradox in Hispanics and also reported other complexi- data, Clegg and colleagues (2009) evaluated associations of education,
ties. Using the relative index of inequality (RII), the authors found, income, poverty status, and employment status to cancer incidence in
consistent with previous findings, that women of low SES had a 11,464 people from 26 NLMS cohorts. Education was most strongly
lower risk of breast cancer and a higher risk of cervical cancer com- associated with cancer; lower education levels were related to a higher
pared with women of high SES; these patterns held for all race/​ risk of colorectal cancer and lower risks of prostate and breast can-
ethnic groups. Parallel associations were seen in men for prostate cer but higher risks of late-​stage prostate and breast cancer. Parallel
cancer, as for breast cancer. The consistencies ended there. Similar patterns of association for income, poverty status, and employment
to findings in Krieger, women and men of low SES had higher risks status and cancer were reported, but associations were less often sta-
of lung cancer in whites, blacks, and Asians and Pacific Islanders tistically significant (Table 9–​2). Educational attainment is more stable
(APIs), whereas they had a lower risk of lung cancer in Hispanics. than income, poverty status, and employment status, which may help
Showing even greater complexity, men and women of low SES had explain the stronger findings for education, though it is important to
higher risks of colorectal cancer among non-​Hispanic whites, no consider that different socioeconomic variables could be differently
difference in risk of colorectal cancer in blacks and Hispanic men, predictive by type of cancer. Future research incorporating both area-​
and a lower risk of colorectal cancer in Hispanic women and API and individual-​level SES measures will enable multilevel evaluations
men and women, compared with those of higher SES (Table 9–​1). of socioeconomic effects on cancer.
The authors surmised that the opposite SES gradient in the Hispanic
population may be partly explained by relationships between SES Area-​and Individual-​Level SES and Cancer
and cancer risk factors including diet, obesity, and smoking, that run In addition to individual-​level analyses, researchers have begun to
counter to relationships in other racial/​ethnic groups. Few studies examine the independent effects of area-​and individual-​level SES
have explored the influence of SES on cancer incidence in multi- on cancer incidence. In the ongoing prospective NIH-​AARP Diet
racial populations, but studies have shown variation in the patterns and Health Study of 506,488 residents in six US states and two met-
of association between SES and cancer incidence by type of cancer ropolitan areas, Doubeni and colleagues (Doubeni, Laiyemo, et al.,
and by race/​ethnicity. 2012)  reported significant independent associations of area-​level
146
Table 9–​1. Relative Index of Socioeconomic Inequality and Cancer Incidence by Race

Breast Cancer Prostate Cancer Cervical Cancer Colorectal Cancer Lung Cancer

Female Male Female Male Female Male Female

RII 95% CI RII 95% CI RII 95% CI RII 95% CI RII 95% CI RII 95% CI RII 95% CI

Non-​Hispanic White 0.78 (0.76, 0.80) 0.80 (0.78, 0.83) 2.98 (2.61, 3.40) 1.25 (1.19, 1.31) 1.19 (1.12, 1.25) 2.40 (2.31, 2.50) 1.71 (1.63, 1.79)

Black 0.85 (0.78, 0.93) 0.86 (0.81, 0.91) 2.77 (2.01, 3.81) 1.03 (0.91, 1.17) 1.11 (0.97, 1.27) 1.54 (1.41. 1.68) 1.26 (1.12, 1.43)

Hispanic 0.50 (0.47, 0.52) 0.58 (0.56, 0.60) 2.18 (1.93, 2.46) 0.60 (0.56, 0.64) 0.62 (0.57, 0.67) 0.82 (0.77, 0.87) 0.60 (0.55, 0.65)

Asian/​Pacific Islander 0.66 (0.61, 0.77) 0.76 (0.70, 0.81) 2.26 (1.77, 2.89) 0.99 (0.89, 1.10) 0.88 (0.78, 0.99) 1.46 (1.33, 1.60) 1.16 (1.02, 1.33)

The relative index of inequality (RII) represents the ratio of cancer incidence in the lowest to the incidence in the highest socioeconomic group for socioeconomic status and 95% confidence intervals (CI) of cancer incidence rates by sex
and race/​ethnicity (consistent with the census 2000, race/​ethnicity categories were not mutually exclusive): selected cancer sites, California, 1998–​2002.
 147
Table 9–2. Age-​Adjusted Incidence Ratesa and Covariate-​Adjusted Rate Ratios (RR)b by Selected Socioeconomic Characteristics: Colorectal, Prostate, and Female Breast Cancer

Colorectal Cancer
(Both Sexes Combined) Prostate Cancer Female Breast Cancer

Characteristic No. Rate SE RR 95% CI No. Rate SE RR 95% CI No. Rate SE RR 95% CI

Total population 1467 68.39 1.69 –​ –​ –​ 1995 218.11 4.64 –​ –​ –​ 1739 149.10 3.54 –​ –​ –​

Educational attainment (years of education)

< HS graduates (≤11) 512 71.94 3.26 1.45 1.31 1.61 622 203.50 7.91 0.79 0.70 0.90 407 124.93 6.87 0.74 0.63 0.86

HS graduates (12) 527 69.50 2.90 1.22 1.04 1.44 592 211.14 8.43 0.83 0.74 0.94 708 151.23 5.65 0.88 0.77 1.01

Some post HS (13–​15) 217 64.39 4.14 1.13 0.93 1.36 308 221.75 12.19 0.89 0.77 1.03 333 164.23 8.87 0.96 0.82 1.13

College education or beyond (16+) 211 66.31 4.07 1.00 Reference 471 253.34 11.64 1.00 Reference 290 167.82 9.95 1.00 Reference

Family income (1990 dollars)

< $12,500 286 69.55 4.33 1.20 1.02 1.43 245 201.15 12.81 0.84 0.72 0.98 304 136.35 8.82 0.90 0.77 1.05

$12,500–​$24,999 353 69.63 3.62 1.20 1.02 1.43 430 207.05 9.40 0.87 0.77 0.99 397 152.73 7.89 0.98 0.85 1.12

$25,000–​$34,999 217 72.85 4.78 1.21 1.02 1.43 268 207.64 12.14 0.86 0.74 0.99 225 139.22 9.29 0.87 0.74 1.02

$35,000–​$49,999 208 66.53 4.39 1.05 0.88 1.25 332 220.30 12.11 0.92 0.81 1.05 268 151.15 9.27 0.94 0.81 1.09

$50,000+ 347 64.09 3.51 1.00 Reference 655 232.47 9.58 1.00 Reference 502 158.60 7.50 1.00 Reference

Poverty status (ratio of family income to poverty threshold)

At or below 100% 157 69.87 5.47 1.24 1.30 1.60 136 209.09 17.06 0.87 0.72 1.00 185 135.48 10.13 0.89 0.73 1.07

100%–​200% 280 64.53 3.79 1.11 0.93 1.34 285 177.27 10.04 0.71 0.61 1.06 314 136.93 7.94 0.90 0.76 1.06

200%–​400% 525 73.21 3.02 1.21 1.03 1.42 711 230.86 8.09 0.93 0.82 1.05 586 148.28 6.08 0.94 0.88 1.09

400%–​600% 291 69.67 3.91 1.10 0.92 1.31 435 212.20 10.10 0.87 0.76 1.00 381 160.76 8.21 1.03 0.88 1.20

Above 600% 214 63.29 4.09 1.00 Reference 428 236.44 11.31 1.00 Reference 273 157.61 9.66 1.00 Reference

HS: High School
Source: SEER-​NLMS Record Linkage Study. Based on the 1979–​1998 follow-​up of residents of 11 SEER Registries (Iowa, Hawaii, Seattle, Connecticut, Detroit, Utah, Los Angeles, San Francisco/​Oakland/​San Jose/​Monterey,
Greater California, Louisiana, and Kentucky) who were 25 years of age or older on their CPS survey date.
a
Rates are per 100,000 population and are age-​adjusted to the 2000 US standard population by the direct method.
b
Rate ratios were estimated from Cox regression models that stratified for age at survey and CPS cohort and controlled for sex when relevant.
148

148 Part II: The Magnitude of Cancer


SES and education with colorectal cancer incidence. Neighborhood SES AND CANCER SURVIVAL AND MORTALITY
deprivation was associated with a 16% increased risk (95% CI: 5%,
28%; p-​trend  =  0.002) of colorectal cancer compared with the
least deprived neighborhoods, and those with < 12 years of formal Cancer in Adults
schooling had a 19% increased risk (95% CI:  7%, 31%; p-​trend Even though lower SES is not universally associated with increased
< 0.001) of colorectal cancer compared to those with a post-​ risks of cancer incidence across different sites, lower SES is consis-
graduate education. tently related to worse prognosis and survival following the diagnosis
Though area-​level measures have enabled researchers to evaluate of cancer, and several large studies have demonstrated associations
associations between SES and cancer, a potentially important goal is between SES, whether measured at the individual or area level, and
to be able to distinguish contextual (e.g., environmental, neighborhood, mortality from cancer (Byers et al., 2008; Hastert et al., 2015; Kish,
policy) from compositional (i.e., people living in low-​SES areas are Yu, Percy-​Laurry, & Altekruse, 2014; Parise & Caggiano, 2013; Tao
themselves of lower SES) effects. We previously cautioned against over- et al., 2014). Even for those cancers with a positive SES gradient with
interpretation of area-​level SES data for this reason. This concern also incidence, people of lower SES have a greater risk of mortality follow-
motivated recent work in the VITamins and Lifestyle (VITAL) study. ing the diagnosis of cancer. For example, the positive SES gradient for
In this study, which included 52,186 participants, the authors (Hastert breast cancer incidence but reversal in the SES gradient for breast can-
et al., 2015) found that the lowest quintile of area-​level SES, measured cer mortality has been frequently documented in the literature. Though
as the 2000 Census block group-​level index of social disadvantage, was women of higher SES have higher breast cancer rates, mortality rates
not significantly associated with total cancer, regardless of adjustment are lower and survival times better with increasing SES among those
for education and household income (Hastert et  al., 2015). A  direct diagnosed.
association of area SES and prostate cancer was explained by individ-
ual SES factors. By contrast, area-​level SES was significantly associ- Area-​Level SES Measures and Secular Trends
ated with lung cancer and, of borderline significance, associated with In the absence of individual-​level SES information, investigators have
colorectal cancer after adjustment for individual SES variables (Hastert continued to document the long-​term trends in the relationship between
et al., 2015). The area-​level measure was unrelated to breast cancer in area-​level SES measures and cancer mortality (Du, Fang, & Meyer,
any analysis. Area-​level effects may be more relevant for those cancers, 2008; Hastert et al., 2015; Kish et al., 2014; G. K. Singh, Miller, &
including lung and colorectal cancer, that are influenced by lifestyle risk Hankey, 2002; G. K. Singh, Miller, Hankey, et al., 2002). As a proxy
factors such as diet, smoking, and obesity. Additional multilevel studies for individual SES, the main drawback of the use of area-​level (e.g.,
are needed to understand the independent effects of area-​and individual-​ zipcode) information is measurement error; SES based on zipcode
level SES on cancer. does not necessarily correlate with individual-​level SES. Additionally,
there is the risk of committing an ecological fallacy whenever we use
Secular Trends aggregated data (on both the exposure and outcome side) to make
Breast cancer incidence continues to be higher among women of inferences about individual-​ level associations. With these caveats,
higher SES. The Women’s Health Initiative (WHI) Trial findings, area-​level SES measures have been useful in predicting cancer mortal-
released in 2003, showed that hormone therapy in the form of the ity and in demonstrating changes in patterns of associations between
combination of estrogen plus progestin increased the risk of cor- SES and cancer outcomes that have occurred over the past 65 years.
onary heart disease (Manson et  al., 2003), prompting an abrupt Singh and colleagues (G. K. Singh, Miller, & Hankey, 2002) devel-
decline in the use and prescription of hormone therapy. There was oped an area SES index at the level of the 3097 counties of the United
a corresponding decline in the rate of breast cancer incidence, with States, in order to track the evolution of the SES gradient in cancer
possible implications for socioeconomic disparities in this out- mortality between 1950 and 1998. The SES index was composed of
come, though there are mixed findings:  some researchers found a 11 variables available on the US Census, including aggregate indi-
steeper decline in the use of hormone therapy after the WHI report, cators that assessed the domains of education (percent of the county
using area-​level measures (Krieger et  al., 2010), and others found population with less than 9 years of education; percent with at least
proportional reductions by SES, using individual-​level data from a high school education), occupation (percent of employed persons
221,378 women from five health maintenance organizations (F. Wei in white collar occupations; the unemployment rate), and income/​
et al., 2005). wealth (median county-​ level family income; county-​ level income
In summary, the patterns of SES gradients with cancer are com- inequality; median home value; median gross rent; percent of families
plex, and can vary according to the setting (i.e., region or country of below the poverty level; percent occupied housing units without tele-
the world), time period, and population. Patterns “defy easy general- phone access; and percent of occupied housing units without complete
ization” (Krieger et al., 1999) and warrant careful attention to meth- plumbing). The authors found a dramatic change in the area SES pat-
odological issues, such as the level at which SES is being measured tern of cancer mortality during the 40-​year study period (Figures 9–​2
(area-​level or individual-​level or both) and stratification by race/​ethnic and 9–​3). Singh et al. (2002) showed reversals of the socioeconomic
group or other sociodemographic factors. gradient in men and younger women and a narrowing of the gradient
in older women.
Throughout the 1950s and 1960s, there was a positive SES gradient
Cancer in Children and Youth (i.e., higher cancer mortality rates in areas of higher SES than areas of
A large part of the literature on SES and cancer focuses on adult lower SES), which was true for both men and women. For example, in
cancers, in part because of the higher rates of cancer with increas- 1950–​1952, cancer mortality was 49% higher in the highest SES areas
ing age. Many known causes of adult cancer are risk factors asso- compared to the lowest (95% CI: 41%, 59%). The positive SES gradi-
ciated with socioeconomic status that influence the development ent narrowed in the 1970s for men, and by the late 1980s, the gradient
of cancer over many years. The causes of cancer in children have began to reverse and then widen. Between 1997 and 1998, male cancer
not been historically related to SES and were attributed instead to mortality rates were 19% higher (95% CI: 11%, 28%) in the lowest
random genetic mutations. However, increasing research has evalu- versus the highest SES areas. Among women, an interaction was found
ated the impact of risk factors, particularly environmental toxins, with age group. For women over 65 years, cancer mortality rates were
on the risk of childhood cancer early in life, discussed later in this higher in higher SES areas throughout the study period, though the gra-
chapter. Given that families and children of low SES are known to dient narrowed over time. In younger women (aged 25–​64), the SES
be exposed to higher levels of environmental toxins compared to gradient was reversed in the early 1990s. By 1998, the cancer mortal-
children from high SES backgrounds, this research has important ity rate in younger women was 13% higher (95% CI: 9%, 16%) in the
implications for understanding socioeconomic disparities on cancer lowest versus the highest SES areas (G. K. Singh, Miller, & Hankey,
outcomes. 2002). Similar reversals of the SES gradient in cancer mortality were
 149

Socioeconomic Disparities in Cancer Incidence and Mortality 149


All Ages Age 25–64 Years Age 65+ Years
240 190 1600

180 1500

220

170 1400
Age-Adjusted Death Rate per 100,000 Population

Age-Adjusted Death Rate per 100,000 Population


Age-Adjusted Death Rate per 100,000 Population
1970 US Population Used as Standard

1970 US Population Used as Standard


1970 US Population Used as Standard
200 160 1300

150 1200

180

140 1100

160 1000
130

120 900

140 1st Quintile (Low SES) 1st Quintile (Low SES) 1st Quintile (Low SES)
2nd Quintile 2nd Quintile 2nd Quintile
3rd Quintile 110 3rd Quintile 800 3rd Quintile
4th Quintile 4th Quintile 4th Quintile
5th Quintile (High SES) 5th Quintile (High SES) 5th Quintile (High SES)
120 700
100
1950
1954
1958
1962
1966
1970
1974
1978
1982
1986
1990
1994
1998

1950
1954
1958
1962
1966
1970
1974
1978
1982
1986
1990
1994
1998
1950
1954
1958
1962
1966
1970
1974
1978
1982
1986
1990
1994
1998
Figure 9–​2.  Cancer mortality rates for US men by age and socioeconomic status (SES) index, 1950–​1998. Source: Singh et al. (2002a).

reported in Britain, Canada, and Australia (G. K.  Singh, Miller, & time period were smaller than those for lung and colorectal cancer. By
Hankey, 2002; G. K. Singh, Miller, Hankey, et al., 2002). contrast, low SES was associated with higher cervical cancer mortality
In a more recent analysis, Krieger (Krieger et  al., 2012)  reported throughout the study period.
secular trends of area-​level SES between 1960 and 2006 and incident Similar to previous studies showing different patterns of associa-
cancer by type of cancer. These data also showed reversals in associa- tion by race/​ethnicity, Tao and colleagues (Tao et al., 2014) found that
tions between SES and lung, prostate, colorectal, breast, and stomach associations between area-​level socioeconomic measures and colorec-
cancer, consistent with the pattern seen in Singh (G. K. Singh, Miller, & tal cancer mortality in the Hispanic population differed by nativity. An
Hankey, 2002; G.  K. Singh, Miller, Hankey, et  al., 2002). Of note, inverse association between area-​level SES and colorectal cancer mor-
disparities for prostate, breast, and stomach cancer by the end of the tality was seen in US-​born, but not foreign-​born, Hispanics, possibly

All Ages Age 25–64 Years


Age 65+ Years
160 165
900
1st Quintile (Low SES)
1st Quintile (Low SES) 1st Quintile (Low SES) 2nd Quintile
155 160
2nd Quintile 2nd Quintile
3rd Quintile
3rd Quintile 3rd Quintile 850 4th Quintile
4th Quintile 155 4th Quintile
Age-Adjusted Death Rate per 100,000 Population

150
Age-Adjusted Death Rate per 100,000 Population
Age-Adjusted Death Rate per 100,000 Population

5th Quintile (High SES)


5th Quintile (High SES) 5th Quintile (High SES)
1970 US Population Used as Standard
1970 US Population Used as Standard
1970 US Population Used as Standard

150
145 800

145
140
750
140
135
135
700
130
130
125
125 650

120
120
600
115
115

110 110 550


1950
1954
1958
1962
1966
1970
1974
1978
1982
1986
1990
1994
1998

1950
1954
1958
1962
1966
1970
1974
1978
1982
1986
1990
1994
1998
1950
1954
1958
1962
1966
1970
1974
1978
1982
1986
1990
1994
1998

Figure 9–​3.  Cancer mortality rates for US women by age and socioeconomic status (SES) index, 1950–​1998. Source: Singh et al. (2002a).
150

150 Part II: The Magnitude of Cancer


related to dietary differences in immigrants compared with those born lung cancer mortality rates increased in white women with < 12 years
in the United States (Tao et al., 2014). of education (annual percent change = +2.4%, p < 0.001), and declining
mortality rates among women college graduates were less pronounced
Individual-​Level SES Measures and Secular Trends than in men (annual percent change (APC) = –​2.2% in black women
Still among the largest cohort studies to be conducted of individual-​ and  –​2.9% in white women vs.  –​4.9% in white men and  –​6.8% in
level SES measures and cancer mortality are the two American Cancer black men). The authors also noted larger declines in colorectal cancer
Society cohorts—​the Cancer Prevention Study I (CPS-​I) and Cancer mortality with greater education, particularly in black men and white
Prevention Study II (CPS-​II). Steenland, Henley, and Thun (2002) men and women, though the rate of colorectal cancer mortality among
examined the association between educational attainment and can- those with < 12 years of education increased significantly in black men
cer mortality rates in the two cohorts. The CPS-​I cohort comprised (+2.7%, p < 0.001). There were also larger declines in breast cancer
1,051,038 men and women enlisted by American Cancer Society vol- mortality and prostate cancer mortality among those with higher levels
unteers in 1959, whose vital status was followed until 1972. The CPS-​ of education in both blacks and whites. Deaths rates and rate ratios at
II cohort consisted of 1,184,657 men and women enlisted by American the two time periods, by level of educational attainment, are repro-
Cancer Society volunteers in 1982 and followed until 1996. duced for the cancer sites examined (Table 9–​4).
Lung cancer mortality rates increased between the two study peri-
ods for both men and women. Among men, there was a significant Independent Effects of Individual and Area-​Level
educational gradient in both periods, but the SES gradient grew stron- SES Measures
ger in the second period (CPS-​II). By contrast, among women, there In addition to individual-​level analyses, researchers have begun to
was no statistically significant educational gradient during the 1960s examine the independent effects of area-​and individual-​level SES
(CPS-​I)—​only women with the highest educational attainment (col- on cancer mortality outcomes (Hastert et  al., 2015; Shariff-​Marco
lege graduates) had a lower mortality rate from lung cancer compared et al., 2014; Steenland, Henley, Calle, & Thun, 2004). In the VITAL
to all other women. However, during the second period (1982–​1996, study in western Washington, area-​level SES was consistently asso-
CPS-​ II), a statistically significant educational gradient emerged. ciated with overall cancer mortality; residents in the lowest quintile
Adjusting the rate ratios for smoking, diet, and alcohol intake (among of area SES had a 40% elevated risk of mortality (95% CI:  16%,
other things) attenuated, but did not remove, the excess risks among 69%, p-​trend < 0.001) (Hastert et al., 2015). In 179,383 persons from
lower educational groups, suggesting that these factors alone did not the American Cancer Society Nutrition Cohort, however, Steenland
explain the educational gradient. (Steenland, Henley, et al., 2004) found independent effects of area-​
In the CPS cohorts, Steenland and colleagues (2002) also found no level SES (census block composite score) and education and cancer
consistent educational gradient in colorectal cancer mortality for the mortality in men, but no significant associations of either area-​or
earlier period, but 20%–​30% lower mortality rates for the most edu- individual-​level SES and cancer mortality in women (Steenland, Hu,
cated men and women in the later cohort (CPS-​II). A  weak inverse & Walker, 2004).
gradient was also found for prostate cancer death rates (lower educa- Steenland (Steenland, Hu, et  al., 2004)  further evaluated secular
tion → higher death rates), which was stronger among the 0.6% of men trends. As noted, men and women from higher SES backgrounds
with prevalent disease at baseline. showed higher cancer mortality rates for certain types of cancer; how-
Although the American Cancer Society cohorts provide intriguing ever, that pattern has shifted, with higher mortality rates observed in
data for the 1960s–​1980s, caution is warranted in interpreting the find- those from lower SES backgrounds, a finding that has been repeat-
ings, as the CPS cohort members were not representative of the general edly corroborated. In a population-​based representative sample of US
US population. More recent research has incorporated individual-​level employed adults ages 35–​64 years, residing in 27 states spanning the
SES variables into more population-​representative studies of cancer period 1984–​1997, Steenland, Hu, and Walker (2004) reported higher
mortality. Investigators (Clegg et al., 2009; Du et al., 2011) linked data rates of mortality among low-​SES men for lung and colorectal cancer,
from the NLMS to SEER registry data. In the NLMS-​SEER data, Du and among low-​SES women for lung cancer. These differences grew
and colleagues (2011) evaluated associations between individual-​level throughout the 1990s and 2000s. Patterns in women differed by type
socioeconomic factors (education, income, poverty status, and health of cancer. They found a direct association of SES and breast cancer
insurance) on treatment and survival. With simultaneous adjustment mortality, but this was due to mixed effects of SES with incidence and
for the individual-​level socioeconomic variables, the authors found mortality since the association was reversed in those with prevalent
that lower levels of education, income, and government insurance disease. They found no association for colorectal cancer in women
were significantly related to both higher overall and cancer-​specific (Steenland, Hu, et  al., 2004). These data thus provide evidence of
mortality (Table 9–​3). persistent or growing socioeconomic disparities over the past several
In other work, investigators evaluated education and cancer mortal- decades, when SES is measured as an individual-​level factor, paral-
ity, obtaining education data from death certificates from the National lel to that seen in research on area-​level SES factors, and in a pat-
Center for Health Statistics (NCHS) and population denominator data tern that generally reflects the theory of fundamental cause (Phelan
from the US Bureau of the Census for 47 US states and Washington, et al., 2010).
DC (Albano et al., 2007; Kinsey et al., 2008). Examining by black and
white race, comparing those with ≤ 12 years versus > 12 years of educa-
tion, Albano and colleagues (2007) found disparities in cancer mortality
Cancer in Children and Youth
in black men (RR = 2.38; 95% CI: 2.33, 2.43), white men (RR = 2.24;
95% CI:  2.23, 2.26), black women (RR  =  1.43; 95% CI:  1.41, 1.46) Advances in treatment have led to dramatic improvements in survival
and white women (RR = 1.76; 95% CI: 1.75, 1.78). Associations were of childhood cancer over the past few decades. At the same time, soci-
strongly influenced by lung and colorectal cancer–​specific associations. oeconomic disparities in cancer survival in children have emerged as
Expanding on Albano’s work (Albano et  al., 2007), Kinsey and well. Gupta and colleagues (Gupta, Wilejto, Pole, Guttmann, & Sung,
colleagues (2008) evaluated trends over time in associations between 2014)  conducted a systematic review of SES and survival of child-
individual SES and cancer mortality. Specifically, they compared rates hood cancer in low-​, middle-​, and high-​income countries and found
of lung, colorectal, breast, and prostate cancers in 1993 and 2001 by consistent socioeconomic gradients with cancer survival among child-
race (black and white) and level of education, obtaining education ren (Gupta et al., 2014). A more recent meta-​analysis found socioeco-
level from death certificates and population data from the US Bureau nomic disparities in survival after childhood leukemia (Petridou et al.,
of Census Current Population Survey. In men, rates of lung cancer 2015)  and concluded that the benefits of life-​saving treatment have
declined in all educational groups, but declines were significantly accrued disproportionately to children from more affluent SES back-
larger in those with greater education in both white (p-​trend = 0.001) grounds. Findings, however, have not been totally consistent. Kent
and black (p-​trend = 0.004) men. A similar gradient was seen in white and colleagues (2015) showed a narrowing of SES survival disparities
women (p-​trend = 0.009) and black women (p-​trend = 0.059), though in lymphoma in adolescents and young adults. When socioeconomic
 15

Socioeconomic Disparities in Cancer Incidence and Mortality 151


Table 9–​3. Hazard Ratios of Mortality According to Individual-​Level Socioeconomic Factors Among Patients with Invasive Tumors

Hazard Ratio (95% CI) of


Hazard Ratio (95% CI) of All-​Cause Mortality Cancer-​Specific Mortality

Attributes No. Model-​1 No. Model-​2 Model-​1 Model-​2

Health Insurance

Employer/​Medicare/​Private  –​ –​ 5595 1.0 (Referent) –​ 1.0 (Referent)


Government  –​ –​ 78 1.5 (1.1, 2.1) –​ 1.6 (1.1, 2.3)
Medicaid/​Not-​insured  –​ –​ 694 1.4 (1.2, 1.5) –​ 1.3 (1.1, 1.4)
Years of Education

< 12 3607 1.3 (1.2. 1.4) 1605 1.2 (1.1, 1.4) 1.4 (1.3, 1.5) 1.3 (1.1, 1.4)
12  4814 1.2 (1.1, 1.2) 2306 1.1 (1.0, 1.2) 1.2 (1.1, 1.3) 1.2 (1.1, 1.3)
≥ 13  4810 1.0 (Referent) 2456 1.0 (Referent) 1.0 (Referent) 1.0 (Referent)
Annual Family Income

< $10,000 1288 1.3 (1.2, 1.5) 646 1.3 (1.1, 1.6) 1.2 (1.1, 1.4) 1.2 (1.0, 1.5)
$10,000–​$34,999  4921 1.2 (1.1, 1.3) 2404 1.2 (1.0, 1.3) 1.1 (1.0, 1.2) 1.1 (1.0, 1.3)
≥ $35,000  6314 1.0 (Referent) 3024 1.0 (Referent) 1.0 (Referent) 1.0 (Referent)
Unknown  711 1.1 (1.0, 1.2) 293 0.2 (0.0, 2.2) 1.0 (0.9, 1.2) 0.3 (0.0, 2.5)
Family Poverty Status (1990 threshold)

≤ 100%  1007 1.0 (0.9, 1.2) 473 0.9 (0.7, 1.1) 1.0 (0.8, 1.2) 0.9 (0.7, 1.1)
100%–​400%  6090 1.0 (1.0, 1.1) 1248 1.0 (0.9, 1.2) 1.1 (1.0, 1.2) 1.1 (0.9, 1.2)
≥ 400%  5982 1.0 (Referent) 1721 1.0 (Referent) 1.0 (Referent) 1.0 (Referent)
Unknown  155 1.1 (1.0, 1.2) 2925 4.9 (0.5, 43.7) 1.0 (0.9, 1.2) 3.8 (0.4, 34.5)

Model 1: Hazard ratios simultaneously adjusted for age, sex, tumor stage, education, poverty, family income, surgery, and radiotherapy.
Model 2: Adjusted additionally for health insurance in those with these data.

disparities have been documented, lack of health insurance and other effects. The analysis of such birth cohorts has documented that health
barriers to care appear to be major sources of survival differences. status can (and does) affect later social mobility. However, the general
magnitude of the drift effect is too small to explain away the observed
EXPLANATIONS FOR THE ASSOCIATION BETWEEN SES gradient in health. That said, almost all of the studies of the drift
SES AND CANCER OUTCOMES phenomenon have been carried out in studies of SES differentials in
all-​cause mortality. Whether reverse causation accounts for some of
Having described the general patterns of association between various the SES differential in cancer outcomes is unresolved. One of the
indicators of SES and cancer outcomes, we now consider the general more convincing examples of this phenomenon is the evidence show-
categories of explanation for the observed gradients. Generally speak- ing that cancer in childhood may jeopardize educational and occupa-
ing, there are three types of explanations for the repeatedly observed tional attainment, particularly in women, through adverse effects on
correlation between SES and cancer. First, the association may arise development, cardiovascular risk, lower attainment of adult height,
through reverse causation, that is, the diagnosis of cancer may result and higher levels of obesity (Ater, 2015; Barrera, Shaw, Speechley,
in loss of employment or loss of income (or both). Alternatively, lower Maunsell, & Pogany, 2005; Kuehni et al., 2012; Maule et al., 2016;
socioeconomic position may be causally linked to cancer incidence Wilson et al., 2014).
and/​or survival, through mechanisms described in detail in the fol- Investigators may minimize the influence of reverse causation by
lowing. Finally, the correlation between SES and cancer may be the carefully selecting the indicator of SES. For example, in adulthood,
spurious artifact of a third, unobserved variable that affects both SES educational attainment is less susceptible to drift effects, compared
and cancer risk, for example, inherited personality characteristics that to other indicators of SES such as income or occupational status.
cause people to strive harder and succeed at upward social mobility The reason is because educational attainment is usually completed
and at the same time invest in their personal future health (including in early adulthood before individuals succumb to most forms of can-
steps to prevent cancer). These three explanations are not necessarily cer. However, even educational attainment might be plausibly subject
mutually exclusive (Glymour, Avendano, & Kawachi, 2014). to reverse causation (i.e., effects running from health status to edu-
The reverse causation hypothesis is sometimes referred to as the cational attainment). For example, the 1958 British National Child
“drift” hypothesis, meaning that the onset of illness (such as cancer) Development Study suggested that “health shocks” in the form of low
can act as a potent trigger for individuals to drift downward on the birthweight resulted in a persistent deleterious effect on O-​level exam-
socioeconomic hierarchy. Conversely, healthier people are more likely ination performance (the O-​levels were national examinations taken
to succeed in moving up the socioeconomic hierarchy. The implica- by British students at about age 16 prior to 1988, the results of which
tion is that health disparities are due to selective social mobility, which determine whether or not one can proceed with further schooling)
sorts healthy and unhealthy people into different socioeconomic posi- (Currie & Hyson, 1999). Low birthweight might thus predict lower
tions (Pais, 2014; Wilkinson, 1996). So-​called birth cohorts, in which educational attainment (Litt et al., 2012), as well as reduced risks of
individuals are followed up from birth, with repeated assessments prostate and breast cancer (Davey-​Smith et  al., 2001). According to
of health status and SES, are ideally suited to test for these kinds of this scenario, it is not lower educational attainment that causes lower
152

152 Part II: The Magnitude of Cancer


Table 9–​4. Trends in Age-​Adjusted Rates* of Death from Four Major Cancers by Educational Attainment, Ages 25–​64 Years, 43 States and the District
of Columbia, 1993–​2001

White Non-​Hispanic Black Non-​Hispanic

1993 Rate 2001 Rate APC‡ P value§ 1993 Rate 2001 Rate APC‡ P value§

Lung and bronchus, Male    


Education < 12 years 88.1 87.3 −0.1 .905 98.3 90.4 −0.2 .705
12 years             59.5 53.2 −1.5 .001 98.6 73.7 −3.2 .009
13–​15 years             32.7 24.2 −3.5 < .001 45.6 32.2 −4.7 < .001
≥ 16 years             20.7 13.7 −4.9 < .001 38.3 21.0 −6.8 < .001
Rate ratio¶ 4.2 (4.1–​4.4) 6.4 (6.2–​6.6) < .001 2.6 (2.5–​2.8) 4.3 (3.9–​4.8) < .001
     Ptrend# .001 .004
Lung and bronchus, Female    
Education < 12 years 45.5 55.4 2.4 < .001 32.7 30.4 0.8 .476
12 years             32.1 33.1 0.1 .657 37.3 35.7 −0.7 .191
13–​15 years             19.8 16.6 −1.7 .003 20.1 19.3 −1.3 .282
≥ 16 years             13.9 11.6 −2.9 < .001 14.8 16.7 −2.2 .246
Rate ratio¶ 3.3 (3.1–​3.5) 4.8 (4.5–​5.0) < .001 2.2 (1.8–​2.7) 1.8 (1.6–​2.1) .090
     Ptrend# .009 .059
Colon and rectum, Male
Education < 12 years 14.1 16.0 0.9 .237 17.4 20.9 2.7 < .001
12 years             14.6 13.9 −0.9 .130 21.9 23.9 1.0 .341
13–​15 years             9.2 8.1 −1.1 .006 15.4 11.7 −2.7 .161
≥ 16 years             9.3 7.9 −2.4 < .001 16.3 11.5 −4.8 .011
Rate ratio¶ 1.5 (1.4–​1.6) 2.0 (1.9–​2.2) < .001 1.1 (0.9–​1.3) 1.8 (1.5–​2.2) < .001
     Ptrend# .039 .010
Colon and rectum, Female    
Education < 12 years 9.5 10.4 1.4 .073 11.0 10.3 −0.3 .745
12 years             9.7 9.2 −1.0 .010 16.1 17.8 −0.3 .730
13–​15 years             6.4 5.5 −1.6 .006 9.4 10.0 0.7 .659
≥ 16 years             6.8 5.4 −3.0 < .001 15.6 12.2 −2.6 .030
Rate ratio¶ 1.4 (1.3–​1.6) 1.9 (1.7–​2.1) < .001 0.7 (0.6–​0.9) 0.8 (0.7–​1.0) .232
     Ptrend# .063 .288
Breast (Female)   
Education < 12 years 27.4 24.1 −1.4 .029 30.0 28.7 0.1 .855
12 years             30.6 25.4 −2.9 < .001 45.3 43.4 −1.5 .078
13–​15 years             23.2 17.3 −3.6 < .001 35.3 30.0 −0.9 .447
≥ 16 years             27.4 20.1 −4.3 < .001 45.7 35.8 −3.8 < .001
Rate ratio¶ 1.0 (1.0–​1.1) 1.2 (1.1–​1.3) < .001 0.7 (0.6–​0.7) 0.8 (0.7–​0.9) .004
     Ptrend# .025 .046
Prostate
Education < 12 years 4.0 3.4 −1.6 .054 10.4 9.6 −1.6 .133
12 years             4.3 3.3 −3.5 .003 16.2 12.7 −1.6 .171
13–​15 years             3.4 2.3 −5.5 < .001 10.3 5.3 −7.4 .003
≥ 16 years             3.8 2.3 −6.3 < .001 7.6 4.8 −5.9 .071
Rate ratio¶ 1.1 (0.9–​1.2) 1.5 (1.3–​1.7) .001 1.4 (1.0–​1.8) 2.0 (1.5–​2.7) .054
     Ptrend# .022 .211

*
Single-​year rates per 100 000 people were age adjusted to the 2000 US standard population for ages 25–​64 years.

Annual percent change (APC) estimated from weighted least squares linear regression models fit to the log-​transformed age-​adjusted death rates for years 1993–​2001.
§
Value adjacent to APC indicates test for APC statistical difference from 0, two-​sided. Value adjacent to rate ratios indicates test for statistical difference between rate ratios for single
years 1993 and 2001.

Rate ratio (95% confidence interval) comparing age-​adjusted rate for less than 12 years education to 16 or more years education for the indicated year.
#
Test for trend in APC over levels of education using inverse variance weighted linear regression.

risks of breast and prostate cancer; rather, low birthweight may be the observed at age 17, when all subjects were still in the same grade. In
factor underlying both. other words, the amount of formal schooling that a person will even-
The argument that the correlation between SES and health may be tually achieve predicts their smoking behavior before their schooling
due to (unobserved) third variable bias is often made by economists is actually completed. Achieving the additional years of schooling
(Farrell & Fuchs, 1982; Fuchs, 1983), and has been increasingly con- beyond age 17 had no additional effect on smoking behavior. It is pos-
sidered by epidemiologists. The argument is based upon observations sible that schooling is effective in preventing smoking uptake only up
such as the timing of the onset of educational differentials in cigarette to the 12th grade, and not thereafter. However, the authors rejected
smoking. In a cohort of young persons with 12–​18 years of completed this unlikely explanation in favor of underlying “third variables” that
schooling, Farrell and Fuchs (1982) found that the strong negative are related to both schooling and health maintenance. The problem of
relationship between schooling and smoking observed at age 24 was omitted variable bias can be generalized to the entirety of empirical
almost completely accounted for by differences in smoking behavior evidence linking SES to diverse health outcomes, including cancer. In
 153

Socioeconomic Disparities in Cancer Incidence and Mortality 153


other words, the challenge of research in this field is to move beyond problematic from the point of view of prevention that many risk fac-
documenting associations between SES and health, toward testing tors tend to cluster together or “crystallize” around low SES. As the
causal connections. The key issue is that the same factors that prompt chapter emphasized at the outset, SES is a multidimensional construct,
people to seek more education and better jobs, such as higher abilities, with complex pathways linking it to cancer outcomes. Accordingly,
higher family socioeconomic background, more patience, and less reducing or eliminating socioeconomic disparities in cancer is unlikely
impulsiveness (i.e., what economists refer to as higher time discount—​ to be accomplished by focusing on and acting to remove one risk fac-
or the willingness to incur current costs for future benefits)—​are also tor at a time.
likely to be determinants of health behaviors and future health. The remainder of this section will summarize the known risk factors
In an attempt to overcome the problem of unobserved heterogeneity for cancer that also exhibit SES patterning. The section is subdivided
and omitted variable bias, researchers have begun to consider instru- into early life risk factors and adult risk factors.
mental variable (IV) analysis to identify the causal relationship of SES
and health outcomes (Greenland, 2000). These methods are not with-
out their own problems, however, and the most convincing approach Early Life Exposures
(short of experimentation) to establish the causal link between SES
and health remains to measure and control for the array of potential
“third variables” that are related to both socioeconomic position and Prenatal and Early Postnatal Exposures
health (Rehkopf, Glymour, & Osypuk, 2016). Ultimately, the clearest
understanding of how SES influences cancer incidence and mortality Still relatively little is known about the effects of prenatal exposures
may come about as the result of examining the multiplicity of contex- linking SES to childhood cancer. Early work explored the link between
tual and individual socioeconomic variables over the life course (or certain occupational exposures or lifestyle exposures (such as cigarette
over multiple generations) on these outcomes. smoking or diet) in relation to specific childhood cancer, as well as
Even after a causal relationship between SES and cancer has been the impacts of viruses and body size on adult cancer. However, there
established, many unresolved questions remain about the exact mech- has been substantial growing interest in understanding how environ-
anisms by which the link occurs. Broadly speaking, there are two sets mental prenatal exposures influence childhood cancer, as evidenced
of pathways by which higher SES may improve cancer outcomes. by the growth of the research literature in this area over the past few
One direct pathway is through the ability of higher SES individu- years (Chuang et al., 2011; Malecki et al., 2006; Spycher et al., 2015).
als to gain access to various resources that help to prevent cancer, or Current evidence is still very limited for most exposures, but the evi-
improve outcomes following the onset of cancer. For example, more dence is generally consistent in suggesting that toxic environmental
years of schooling may result in greater ability to translate health exposures, which predominantly affect children of lower SES, increase
knowledge into health-​ promoting behaviors (avoiding smoking, the risk of childhood cancer.
maintaining regular physical activity, presenting for regular screening
check-​ups) (Pampel, Krueger, & Denney, 2010). Health literacy may Infection and Viruses.  There is substantial evidence on several
also enable individuals to “navigate” medical treatment options, and viruses that may be transmitted vertically from mother to child in
to maximize the effectiveness of cancer therapy (Institute of Medicine utero or during childbirth or breastfeeding, including human lympho-
Committee on Health Literacy & Development, 2004). In the words tropic virus-​type I (HTLV-​1) and hepatitis B virus (HBV), which are
of Grossman (1975), additional years of schooling make an individual both patterned by SES and which can lead to cancer in adulthood (De
a more efficient producer of his or her own health. Income may also Flora & Bonanni, 2011; De Flora et al., 2015; Schottenfeld & Beebe-​
operate directly to reduce an individual’s cancer risk by determin- Dimmer, 2015). Both of these viruses, which are endemic in some
ing his or her level of access to material resources—​for example, the countries, are far rarer in the United States.
ability to afford nicotine replacement therapy, or the ability to afford Unlike HTLV-​1 and HBV, hepatitis C (HCV), a major cause of liver
a health plan that offers regular screening services for the detection cancer, is far more common among underserved, low SES popula-
of cancer. Higher incomes may also enable individuals to move to tions in the United States (Denniston et  al., 2014)  and is primarily
higher quality residential neighborhoods, with access to safe recre- transmitted through injection drug use (IDU). In 2014, the US Centers
ational spaces (for maintaining exercise), or supermarkets and grocer- for Disease Control and Prevention reported that 68% of all HCV
ies with ready availability of fresh produce. Finally, the labor market cases nationwide occurred as a result of IDU. Low SES rural com-
tends to sort individuals into workplaces and jobs with differential munities in the southern United States have been particularly hit, with
exposures to carcinogens (e.g., chemical exposures, indoor tobacco IDU accounting for 73% of all HCV cases in Kentucky, Tennessee,
smoke pollution). Virginia, and West Virginia between 2006 and 2012 (J. Zibbell, 2015).
An alternative pathway from SES to health (including, potentially, However, the risk of perinatal transmission of hepatitis C has been
cancer outcomes) is through differential exposure to psychosocial suggested to be low.
mediators, such as stress, a sense of control, social support, and so on
(Ross & Wu, 1995). In turn, psychosocial factors such as stress and Birth Size.  There has also been some exploration of birth weight
social support may influence health outcomes through health-​related and future cancer risk (Cook, Gamborg, Aarestrup, Sorensen, &
behaviors, as well as through direct effects on physiological mecha- Baker, 2013; Lope et  al., 2016). Trichopoulos (1990) hypothesized
nisms, such as altered immune and neuroendocrine function. that prenatal exposure to high concentrations of pregnancy estrogens,
In the final section, we shall survey and summarize the known risk related to fetal growth rate, may influence subsequent risk of breast
(and protective) factors for cancer incidence and mortality that are cancer. Studies have found positive or J-​shaped associations between
believed to be patterned according to SES, and hence offer potential birth weight, birth length, or head circumference and breast cancer
clues about the mediating causal mechanisms linking the two. (McCormack et al., 2003; Vatten et al., 2002). Since low SES is associ-
ated with a higher prevalence of low birth weight (Parker, Schoendorf,
& Kiely, 1994), this may be one mechanism through which higher SES
MECHANISMS LINKING SOCIOECONOMIC is related to an elevated risk of breast cancer.
STATUS TO CANCER Fewer studies have examined birth size and other cancer outcomes.
Sandhu and colleagues (Sandhu, Luben, Day, & Khaw, 2002) found a
Cigarette smoking is a risk factor par excellence that is linked to low J-​shaped association of birth weight and colorectal cancer—​a positive
SES (in industrialized societies like the US) as well as to cancer out- association corroborated by Smith and colleagues (N. R. Smith et al.,
comes. The relationship between low SES and smoking-​related can- 2016). These results are consistent with findings on birth weight and
cers is well established. However, as the previous section implied, adult obesity (Elks et  al., 2012; Leong et  al., 2003). However, in a
socioeconomic position is also associated with a host of other risk retrospective analysis of birth weight and prostate cancer in the Health
(and protective) factors beyond cigarette smoking. Moreover, it is Professionals Follow-​up Study, Platz and colleagues (1998) found no
154

154 Part II: The Magnitude of Cancer


association between birth weight and prostate cancer, though a more EBV is involved in about 35%–​50% of cases of Hodgkin disease.
recent study found an association between pre-​term birth and risk for Though most adults have had an EBV infection, and are thus carriers
non-​epithelial ovarian cancer in 1.5  million Swedish women (Sieh, of these viral genes (Evans & Mueller, 1997), higher SES groups tend
Rothstein, McGuire, & Whittemore, 2014). to be infected at later ages than those of lower SES. EBV infection at
a later age is associated with more severe clinical sequelae, namely
Environmental Pollutants. As indicated earlier, there has infectious mononucleosis and Hodgkin lymphoma, in higher SES
been considerable growth in the literature on environmental risk fac- populations (Gutensohn, 1982).
tors experienced prenatally or early in life. Between 1990 and 2007, Findings strongly indicate that chronic Helicobacter pylori infec-
based on the California Cancer Registry, air toxin exposures during tion is involved in the development of non-​cardia gastric adenocar-
pregnancy/​infancy from industrial and road traffic sources—​including cinoma and mucosal-​ associated B-​ cell lymphoma (International
acetaldehyde 1,3-​butadiene, benzene, tolune, ortho-​dichlorobenzene, Agency for Research on Cancer Ad Hoc Working Group, 1994;
and polycyclic aromatic hydrocarbons—​have been associated with Nightingale & Gruber, 1994). About two-​thirds of the world’s pop-
higher risks of brain tumors before 6  years of age (von Ehrenstein ulation and half of all adults over age 50 in the United States are
et al., 2015). Intra-​uterine exposure to pesticides, either through occu- infected with H. pylori (Correa & Piazuelo, 2011). Since the 1930s,
pational or home exposures, have been related to an increased risk modernization, leading to clean water, fewer children sharing a bed,
of brain tumors (Kunkle, Bae, Singh, & Roy, 2014), testicular germ smaller families, and possibly the increasing use of antibiotics in
cell tumors (Le Cornet et al., 2015), and childhood leukemia (Bailey children (Blaser, 1999), has contributed to reductions in H.  pylori
et al., 2015). Three meta-​analyses (Van Maele-​Fabry, Hoet, & Lison, transmission and prevalence (Parsonnet, 1995). The infection is
2013; Van Maele-​Fabry, Lantin, Hoet, & Lison, 2010; Wigle, Turner, typically acquired in childhood and there are fewer children in the
& Krewski, 2009), summarizing studies occurring over decades, pro- United States today with H. pylori infection than in previous periods.
vided consistent support for an association between pesticide exposure H. pylori is found more often in lower socioeconomic groups in the
and childhood leukemia. Another study showed associations between United States, many of whom report one or more risk factors for
parental exposure to solvents such as benzene and childhood brain infection, including a history of crowding in childhood, a mother
tumors (Peters et al., 2014). Yet another showed support for an asso- who carries H. pylori, a large number of siblings, presence of older
ciation between prenatal air toxin exposure and risk of Wilms’s tumor siblings (< 4-​year age difference), and unclean water sources. In turn,
in children 0–​5 years of age (Shrestha et al., 2014). Children growing several of these factors are associated with lower SES (Goodman &
up in disadvantaged circumstances are disproportionately affected by Correa, 1995).
such exposures (Adler & Rehkopf, 2008; Hogue, 2011). In one of the
only papers of its kind, in a case-​control study, investigators evalu- Lifestyle Behaviors and Obesity.  The US Centers for Disease
ated the impact of a well-​documented, distinct period of high arsenic Control and Prevention reported that the number of overweight chil-
concentrations in drinking water in Chile during 1958–​1970 on cancer dren more than doubled in the last three decades of the twentieth cen-
risk and found that high early life exposure (> 800 vs. ≤ 110 μg/​L) tury (Centers for Disease Control and Prevention, 1997). More recent
predicted 5-​fold higher rates of lung and 8-​fold higher rates of bladder evidence suggests that rates of obesity have stabilized in the past cou-
cancer decades later (Steinmaus et al., 2014). ple of decades (Ogden, Carroll, Fryar, & Flegal, 2015; Ogden, Carroll,
Kit, & Flegal, 2012; Ogden et  al., 2016; Ogden, Lamb, Carroll, &
Postnatal and Later Exposures Flegal, 2010), but overweight and obesity are more prevalent among
children and adolescents from lower SES backgrounds, and 12.7 mil-
Major mechanisms for postnatal exposures have included childhood lion children and teens ages 2–​18 (17% of the entire population) are
infection, lifestyle behaviors, and secondhand exposure to tobacco currently overweight or obese (Ogden, Carroll, Kit, & Flegal, 2014).
smoke. Population-​ level dietary trends, combined with reductions Maternal obesity, also predominant in adults from lower SES back-
in physical activity, have led to an unprecedented rise in childhood grounds, is strongly linked with childhood obesity (Strauss & Knight,
obesity, which has disproportionately affected children from low SES 1999). Low SES children often live in areas with large numbers of fast
households. The childhood obesity epidemic in the United States (and food restaurants and stores that sell primarily low-​quality, nutritionally
in many other parts of the globe) has dire implications for future can- poor, and calorie-​dense foods. Studies also have shown that children
cer risk and disparities in cancer incidence. The latency period for can- in lower SES families are more likely to be exposed to advertisements
cer incidence is typically decades, but these factors may lead to an that promote eating large portions of obesity-​promoting foods, includ-
accelerated risk of the development of cancer early in life, particularly ing sugar-​sweetened beverages, fast food, and processed foods (Harris,
cancers (e.g., colon cancer) that share many of the same risk factors as Pomeranz, Lobstein, & Brownell, 2009; Piernas & Popkin, 2011;
CVD and diabetes. Powell, Szczypka, & Chaloupka, 2010; Powell, Wada, & Kumanyika,
2014). Childhood overweight and obesity in turn may track into adult-
Childhood and Adolescent Infection.  There is substantial hood overweight and obesity, which are major causes of cancer in
evidence that infections are likely causal factors in several types later life.
of cancer, including lymphoma, cancers of the liver, nasophar- Dietary habits, which begin in childhood, are also predictive of
ynx, cervix, and stomach. Earlier studies showed that these types future dietary patterns. Lack of local availability of fresh produce,
of cancers accounted for up to 20% of cancer worldwide (Eckhart, cultural patterns of food consumption (R. E.  Lee & Cubbin, 2002),
1998), though recent studies indicate that cancers caused by infec- norms accepting of overweight (Becker, Yanek, Koffman, & Bronner,
tions are more than three times more prevalent in developing (26%) 1999; Crawford, Story, Wang, Ritchie, & Sabry, 2001), and sedentary
than in developed (8%) countries (Thun, DeLancey, Center, Jemal, behaviors are each related to lower SES and overweight. In addition to
& Ward, 2010). habits that are developed in the context of a lack of resources, current
Viruses linked to human tumors include Epstein-​Barr virus (EBV) overweight predicts future overweight. Girls who are overweight have
(B-​cell lymphomas, Burkitt’s lymphoma, nasopharyngeal cancer, earlier maturational timing, which is associated with later overweight
Hodgkin lymphoma, T-​cell lymphomas, and gastric cancer); hepati- (Adair & Gordon-​Larsen, 2001), and is also an independent risk fac-
tis B virus (hepatocellular carcinoma); papillomavirus types including tor for breast cancer (Cancer, 2012). Adolescents who are obese are
16, 18, 31, 33, 35, 39, 45, 52, 56, 58 (cervical and anogenital cancer); far more likely to be obese as adults, with long-​term, multiple conse-
and HTLV-​1 (adult T-​cell leukemia) (Chelimo, Wouldes, Cameron, quences for morbidity (Dietz, 1998a, 1998b; Micic, 2001). Patterns
& Elwood, 2013; F.  Liu et  al., 2016; G.  S. Taylor, Long, Brooks, established in childhood and adolescence therefore predispose people
Rickinson, & Hislop, 2015; Zane & Jeang, 2014). Transmission and from lower SES backgrounds to worse health outcomes, including
timing of infection have been linked to SES. cancer, in later adulthood.
 15

Socioeconomic Disparities in Cancer Incidence and Mortality 155


Smoking and Secondhand Exposure to  Cigarette were smokers (Pesch et  al., 2012). Reducing or eliminating smok-
Smoke.  Children from lower SES backgrounds are more likely to ing would prevent the bulk of lung cancer cases, as well as drasti-
be exposed to environmental tobacco smoke (ETS) in their homes (K. cally reduce socioeconomic disparities in lung cancer incidence. In
M. Emmons et al., 2001; Schuster, Franke, & Pham, 2002). Though a response to declining cigarette consumption in the United States, par-
meta-​analysis found no increased association of parental smoking and ticularly among higher SES groups, the tobacco industry substantially
lung cancer of the grown child (Boffetta, Tredaniel, & Greco, 2000), increased expenditures on marketing and advertising promotions,
ETS has been associated with an increased risk of lung cancer in such as “Marlboro Miles” and Camel Cash,” and tobacco promotional
spouses (adult non-​smoking women) (Kreuzer et al., 2002; R. Taylor, items (Federal Trade Commission, 1999). After the Master Settlement
Cumming, Woodward, & Black, 2001). Agreement of 1998 banned the distribution of branded merchandise,
Low-​SES youth are subsequently more prone to take up smoking tobacco companies invested in discounts and point-​of-​sale advertis-
compared to youth from more affluent backgrounds. Factors that may ing, which are disproportionately targeted to people—​and especially
be related to the higher initiation of smoking among low-​SES youth youth—​ from lower SES backgrounds (Robert, Cheney, & Azad,
include parental smoking, though especially for white adolescents 2009). Within the past decade, electronic cigarettes (e-​cigarettes) have
(Griesler & Kandel, 1998; Tyas & Pederson, 1998), greater exposure also come into use. Several studies have reported on disparities in
to peer norms of smoking (Alexander, Piazza, Mekos, & Valente, 2001; awareness and use of e-​cigarettes by income levels (Carroll Chapman
Tyas & Pederson, 1998), as well as greater targeting by the tobacco & Wu, 2014) but the links to SES and to health are as yet unclear.
industry, a strong predictor of adolescent onset of smoking (Altman,
Levine, Coeytaux, Slade, & Jaffe, 1996; Pierce, Choi, Gilpin, Farkas, Diet.  Higher SES is positively correlated with higher quality diet,
& Berry, 1998). Low-​SES youth are also more likely to start smoking including greater consumption of fruit, vegetables, and legumes
at an earlier age, which has been linked to greater difficulty in quitting (Centers for Disease Control and Prevention, 2000; Pechey &
later on (Pampel, Mollborn, & Lawrence, 2014). Furthermore, it has Monsivais, 2016), and lower consumption of foods with high salt/​
been hypothesized that early smoking occurring during adolescence, a fat/​sugar content (Pechey & Monsivais, 2016). Factors such as fat
“critical period” in lung development, may be particularly hazardous, intake, red meat consumption, inadequate vegetable consumption,
in that tobacco carcinogens may induce genetic alterations that make high caloric intake, physical inactivity, and heavy alcohol consump-
the early smoker more susceptible to the damaging effects of contin- tion have been suggested as important risk factors for colorectal cancer
ued smoking (Wiencke & Kelsey, 2002). (Vargas & Thompson, 2012). Each of these factors has been related to
lower SES, both at the individual level and the area (neighborhood)
Environmental Exposures. A meta-​analysis and a registry-​ level (Centers for Disease Control and Prevention, 2000; Diez-​Roux
based case control study showed that postnatal exposure to traffic et  al., 1999; Kamimoto, Easton, Maurice, Husten, & Macera, 1999;
pollution and air toxics, particularly benzene, was related to a higher Li et al., 2000).
risk of childhood leukemia (Amigou et  al., 2011; Filippini, Heck, However, the apparent correlation between low SES and unhealthy
Malagoli, Del Giovane, & Vinceti, 2015). Recently, there has also been diet has not always been direct. A study of dietary trends by Popkin
a growing research emphasis on the potential cancer-​causing effects and colleagues (Popkin, Siega-​Riz, & Haines, 1996) found large dif-
of plasticizers, chemicals, and flame retardants. Though exposure in ferences in dietary quality in 1965, with whites of high SES eating the
daily life is nearly ubiquitous, there is evidence that children from low-​ least, and blacks of low SES eating the most healthful diet as meas-
SES households have higher exposures to flame retardants because of ured by a diet quality index. By the 1989–​1991 survey, the diets of all
exposure to household furniture in poor condition (Schreder, Uding, & groups studied were relatively similar. Of particular note, grain and
La Guardia, 2016). These chemicals have been shown to cause cancer legume consumption declined among low SES blacks since the 1960s
in animal studies, though researchers are only beginning to understand but increased among higher SES whites. The study also indicated that
the potential impact of these unregulated chemicals on the develop- people from both lower and higher SES backgrounds showed improve-
ment of cancer and on the implications for the SES-​cancer gradi- ments in diet, based on increases in fruit and vegetable intake and then-​
ent (Costa, Pellacani, Dao, Kavanagh, & Roque, 2015). Research in current recommendations for reductions in fat intake. Higher SES
humans is not well established. In addition to direct effects of chemi- groups initially had more improvements to make, but dietary quality
cals on risk of cancer, plasticizers have been hypothesized to be related was similar across SES groups by the early 1990s after the advent of
to higher estrogens in children, leading to earlier menarche in girls, recommendations to reduce fat (≤ 30%), saturated fat (< 10%), and
and potentially an increasing risk of breast cancer in girls of low SES cholesterol (< 300 mg/​day) intake; to increase consumption of fruits
(M. Lee, 2012). and vegetables (5+ per day) and complex starches (6+ servings/​day);
to limit sodium intake (≤ 2400 mg/​day); and to maintain proper cal-
cium (RDA) and protein (< 2 times RDA) intake (Popkin et al., 1996).
In 1998, Shi noted that changing patterns in diet may have reflected
Adult Exposures a greater receptivity of higher SES people to public health messages
about consuming foods of high nutritional quality (L. Shi, 1998).
Lifestyle and Related Factors Although the authors did not discuss the consumption of junk food,
studies throughout the 2000s have shown that the potential benefits of
Major lifestyle factors in adult life linked to cancer incidence include relatively healthier diets in blacks compared with whites in the 1960s
smoking, poor diet, alcohol consumption, physical inactivity, and were reversed in the following decades, related to increased intakes
overweight or obesity. Many of these factors are associated with of low-​quality foods (Bahr, 2007; Darmon & Drewnowski, 2008,
each other and tend to cluster together. Several health-​related behav- 2015; Hiza, Casavale, Guenther, & Davis, 2013; Siega-​Riz & Popkin,
iors (e.g., smoking, heavy alcohol consumption, overeating) are 2001; Thompson et al., 2009; Vargas & Thompson, 2012; C. Y. Wang
used by individuals to cope with stress, a fact that has not escaped et al., 2014).
the notice of manufacturers and advertisers who target vulnerable Thus the rise in obesity prevalence among lower SES groups has
audiences. In combination with physical inactivity, an imprudent been influenced by increased consumption of low-​quality foods and
diet can increase the risk of being overweight or obese. The chang- overall caloric consumption, even while efforts were made toward
ing temporal socioeconomic patterns in US lung and colorectal can- meeting dietary recommendations (Harnack, Jeffery, & Boutelle, 2000;
cer mortality may be attributable in part to changing socioeconomic Heini & Weinsier, 1997; Siega-​Riz & Popkin, 2001). Trends toward
patterns in these factors. increased consumption may be due to increases in availability and rel-
ative affordability of food items, increased marketing of low-​quality
Smoking.  Smoking continues to be among the most important fac- food items (Chandon & Wansink, 2012; C. Y. Wang et al., 2014), and
tors responsible for the SES gradient in cancer. Cigarette smoking is increases in portion sizes (Nielsen & Popkin, 2003). Associated with
the leading determinant of lung cancer; 90% of those with lung cancer these, changes may be attributed to changes in agricultural practices,
156

156 Part II: The Magnitude of Cancer


subsidizing of corn production, and food technologies leading to cheap Sexual Behavior.  Aspects of sexual behavior and sexually trans-
production of low-​quality foods (Franck, Grandi, & Eisenberg, 2013; mitted infections (STIs) have been linked to reproductive cancers such
Rickard, Okrent, & Alston, 2013). as cervical and prostate cancers (Castellsague et al., 2002). There are
limited data available for assessing rates of STIs by SES, and when
Alcohol.  Though wealthier people are more likely to engage in data exist, associations are generally weaker than those seen for race/​
moderate alcohol consumption, which has been linked to lower lev- ethnicity and STIs, though some studies do show elevated rates of STIs
els of insulin, body weight, and risk of diabetes (Gepner et al., 2015; among disadvantaged groups (Harling, Subramanian, Barnighausen,
Wakabayashi, 2014), heavy alcohol consumption is more predomi- & Kawachi, 2013).
nant among lower SES groups (Esser et al., 2014). Considerable evi- Cervical cancer is more common among women from lower SES
dence suggests a connection between heavy alcohol consumption and backgrounds, who are less likely to receive regular screening and early
increased risk for cancer, with an estimated 5.8% of total cancer deaths treatment (Churilla et al., 2016). Other important factors related to the
attributable directly or indirectly to alcohol (Rothman, 1980). Alcohol development of cervical cancer include a woman’s history of sexual
consumption is an established cause of cancers of the mouth, pharynx, exposure, her partners’ history of sexual exposure, use of barrier
larynx, esophagus, liver, and, in women, breast (Boffetta, Hashibe, La methods of contraception (protective), and the age at which a woman
Vecchia, Zatonski, & Rehm, 2006). For each of these cancers, risk becomes sexually active (Chelimo et al., 2013). Risk of cervical can-
increases substantially with intake of more than 2 drinks per day, cer is also increased when a woman is exposed to the virus before full
though regular consumption of even a few drinks per week has been maturation of the cervix. In turn, several of these factors have been
associated with an increased risk of breast cancer in women (Shield, shown to be related to SES, especially level of education.
Soerjomataram, & Rehm, 2016). In a study by Hogan, Sun, and Cornwell (2000), adolescent females
Other research suggests that other lifestyle factors associated with whose parents were better educated (greater than high school educa-
low SES, in combination with heavy alcohol consumption, such as tion) were 28% less likely to initiate sexual intercourse and 52% more
malnutrition (Giskes, Turrell, Patterson, & Newman, 2002; Su & Arab, likely to use a contraceptive at first intercourse. Similar findings in
2001) or smoking, may interact to further increase risk of certain types other studies also suggest that females with lower income or parental
of cancer (Jones, Bates, McCoy, & Bellis, 2015). Alcohol consump- educational attainment are more likely to have an early age at first inter-
tion combined with low folate consumption appears to predict higher course (Blum et al., 2000; Lammers, Ireland, Resnick, & Blum, 2000;
levels of colon cancer (Vargas & Thompson, 2012), and combined Santelli, Lowry, Brener, & Robin, 2000; S. Singh, Darroch, & Frost,
with tobacco use, increases risk of cancers of the mouth, larynx, and 2001), and subsequently they have more and/​or concurrent sexual part-
esophagus more than the independent effect of either drinking or ners (Coker et al., 1994; O’Donnell, O’Donnell, & Stueve, 2001) and
smoking (Hashibe et al., 2009). are less likely to use barrier contraception (Bankole, Darroch, &
Singh, 1999). Roura and colleagues (2014) noted that compared to
Obesity and Physical Activity. Overweight and obesity are women who never smoked, women who currently smoked were 1.9
associated with several cancers, including postmenopausal breast times more likely to develop cervical cancer, due to the damage ciga-
cancer, colon cancer, endometrial cancer, prostate cancer, renal cell rette smoking causes to cervical cells.
carcinoma, esophageal adenocarcinoma, ovarian, cervical, and thyroid
cancers. Reproductive Factors
In addition to changing diets, the disproportionate prevalence of Reproductive factors associated with SES and with cancer incidence
obesity among people from lower SES backgrounds may help to include age at menarche, parity, age at first birth, age at menopause,
account for the changing SES gradient for colorectal cancer. As indi- and use of exogenous hormones. Higher SES women have been shown
cated, obesity has increased dramatically over the past several decades, consistently to have higher rates of breast cancer. They are more likely
though the prevalence has stabilized over the past decade or so (Flegal, to have first children at a later age and to have fewer children. They have
Carroll, Kit, & Ogden, 2012; Flegal, Carroll, Ogden, & Curtin, 2010; been more likely to take exogenous hormones after menopause. These
Yanovski & Yanovski, 2011), but lower SES groups, especially factors have in turn been associated with an increased breast cancer risk.
women, are much more likely to be overweight than people from Early age at menarche is also associated with higher risk of breast
higher SES backgrounds (Robbins, Vaccarino, Zhang, & Kasl, 2000; cancer. Higher SES has been historically associated with earlier age
Sharpe, Whitaker, Alia, Wilcox, & Hutto, 2016). As socioeconomic at menarche, which has been observed both within countries, as well
disparities in overweight have widened, the SES gradient in colorec- as in comparisons of developed versus less developed countries. On
tal cancer incidence and mortality is likely to increase. Similarly, the the other hand, with the obesity epidemic growing disproportionately
gradient will likely change as well for other cancers related to obesity. among low-​SES children, early menarche may increasingly weigh
Higher SES is generally positively correlated with physical activity, against lower SES women in the United States, thereby leading to a
which in turn has been associated with lower levels of obesity-​related further narrowing of the SES gap in breast cancer incidence in the
cancers. As an independent risk factor, the evidence for decreased risk future.
with higher physical activity is classified as convincing for breast, As previously discussed, declines in the use of exogenous hormones
colon, and endometrial cancers, probable for prostate cancer, and after 2003 led to declines in breast cancer incidence, but particularly
possible for lung cancers (American Cancer Society, 2016; Clague in women of higher SES who were taking hormone therapy at a higher
& Bernstein, 2012; Friedenreich & Orenstein, 2002). In recent years, rate than women of low SES.
research has been extended to examine the potential effects of physical
activity on other types of cancer.
The hypothesized biological mechanisms for the association Occupational Exposures
between physical activity and cancer include changes in endogenous Blue-​collar and manual workers experience greater exposure than
sex hormone levels and growth factors, decreased obesity and central white-​collar workers to chemicals, diesel fumes, dyes, and other agents
adiposity, and possibly changes in immune function. Central adiposity in the workplace, including inorganic gases, organic compounds, sol-
has been implicated in metabolic conditions that promote carcinogen- vents, mineral and wood dusts, silica, metals, and bioaerosols, which
esis. Evidence is also increasing that exercise influences other aspects are established carcinogens (Boffetta et al., 2000; Kauppinen, Teschke,
of the cancer process, including cancer detection, coping, rehabilita- Savela, Kogevinas, & Boffetta, 1997; Kogevinas & Porta, 1997; US
tion, and survival (Friedenreich & Orenstein, 2002). Based on existing Department of Health and Human Services, 2002; Weston, Aronson,
evidence, the American Cancer Society has issued physical activity Siemiatycki, Howe, & Nadon, 2000).
guidelines for cancer prevention, generally recommending at least 30 Blue-​collar workers are also more likely to be exposed to environ-
minutes of moderate-​to-​vigorous intensity physical activity on 5 or mental tobacco smoke (ETS) (Curtin, Morabia, & Bernstein, 1998).
more days per week, or vigorous activity for 75 minutes or more per Percent of white-​collar workers was a predictor of more restrictive
week (American Cancer Society, 2016). smoking policies in a national sample of worksites (K.M. Emmons,
 157

Socioeconomic Disparities in Cancer Incidence and Mortality 157


2000). Furthermore, the interactive effects of smoking and chemical “contagion” (K. Smith & Christakis, 2008; Valente, 2009), through
exposures may put workers of low SES at additional increased risk of norms (Berkman & Glass, 2000; K. Smith & Christakis, 2008), peer
cancer. The combination of asbestos exposure and cigarette smoking, modeling (Bandura, 1986), and via access to “social capital”—​defined
both correlated with low SES, has been associated with a synergisti- as the informational and material resources exchanged through social
cally increased risk of lung cancer (Gustavsson et al., 2002). ties (Moore, Daniel, Paquet, Dube, & Gauvin, 2009). Depending on
Though not all shift workers occupy lower SES positions (e.g., network structure (Granovetter, 1973), cancer patients may have dif-
healthcare professionals), shift work is more common among persons ferent levels of access to resources, referrals, advice, opportunities
from lower SES backgrounds (e.g., jobs in the service industry, trans- to participate in clinical trials, and knowledge about the side effects
port, security work, manufacturing, etc.) (Boggild, Suadicani, Hein, and outcomes of treatment. Identification with social network mem-
& Gyntelberg, 1999). Longer durations (> 20 years) of rotating night-​ bers may increase the likelihood of adopting behaviors (Christakis &
shift work have been linked to immune dysfunction (Nakata, 2012) and Fowler, 2007; Pachucki, Jacques, & Christakis, 2011) through influ-
rotating night-​shift work has been linked to increased risks of breast ences on shared behaviors and norms.
and colon cancer (Schernhammer et  al., 2001, 2003a). In particular, Social networks may also bring costs or burdens related to social roles
working 30 or more years on the night shift was associated with a that can adversely influence health (C. H. Kroenke et al., 2012). Larger
moderate elevation in risk of breast cancer (RR = 1.36; 95% CI: 1.04, social networks can increase caregiving obligations, disproportionately
1.78) and working 15 or more years on rotating night shifts was associ- true among women of lower SES. While potentially rewarding, caregiving
ated with an increase in colon cancer (RR = 1.35; 95% CI: 1.03, 1.77). can be physically and emotionally demanding, leading to poorer self-​care
The mechanism is believed to be through the suppression of melatonin and worse health outcomes (Cannuscio et al., 2002; Kiecolt-​Glaser et al.,
production, caused by prolonged exposure to light during night-​time 1987; S. Lee, Colditz, Berkman, & Kawachi, 2003; S. Lee, Kawachi, &
(melatonin, in turn, is believed to have oncostatic action). Grodstein, 2004; Schulz & Beach, 1999). The quality of social relation-
ships also appears to differ by SES; high-​strain relationships, more com-
The Social Environment mon in those of low SES, can lead to higher levels of stress.
People of different SES also appear to depend differently on social
Neighborhood Environments.  Much empirical work on SES networks (August & Sorkin, 2011; Bureau, 2011). Women with
and cancer to date has focused on individual-​level exposures and rela- greater education have larger social networks but a greater propor-
tionships. However, increasing attention has been paid more recently tion of distal members. In contrast, women of lower SES are more
to the independent contribution of area-​level socioeconomic factors likely to have smaller but denser networks of people who live in close
on health outcomes (Kawachi & Berkman, 2003)  including cancer proximity and have more intense, frequent contact with greater inter-
(Gomez et  al., 2015), and research to identify contextual effects on dependency. In men, their spouses are often the most critical mem-
health outcomes has expanded markedly over the past decade. A study ber of their social network influencing their health; marital status has
by Merkin and colleagues (Merkin, Stevenson, & Powe, 2002) found been reliably associated with cancer outcomes (Hanske et al., 2016;
that living in areas with lower levels of education and income Jin et al., 2016; R. L. Shi et al., 2016; van Jaarsveld, Miles, Edwards,
increased the odds of presenting with advanced-​stage breast cancer & Wardle, 2006; L.  Wang, Wilson, Stewart, & Hollenbeak, 2011).
by 50% for black women and by 75% for white women. One study However, men of lower SES are less likely to be married (Ajrouch,
found that those in severe poverty or near poverty were, respectively, Blandon, & Antonucci, 2005; Choi & Marks, 2011; Gomez et  al.,
3.0 and 1.6 times more likely to live in areas of higher-​than-​expected 2016; Villingshoj, Ross, Thomsen, & Johansen, 2006).
incidence of late-​stage breast cancer when compared with women liv- Larger social networks predict more optimal cancer mortality risk
ing in non-​poverty (MacKinnon et al., 2007). However, a recent study factors (C. H. Kroenke et al., 2016) and lower cancer mortality gener-
in Michigan in 1992–​2009 found that women with breast cancer living ally, but associations may depend substantially on the quality of, and
in low-​SES areas showed greater declines in mortality rates compared obligations entailed by, those relationships, as well as sociodemo-
to those living in high-​SES areas, resulting in a narrowing of the SES-​ graphic factors.
mortality gap (Akinyemiju et al., 2013).
Community and neighborhood level factors, including the avail- Psychosocial Factors
ability of green space (for physical activity), access to stores and
grocers, as well as social norms and social cohesion, may have an Several mechanisms have been hypothesized through which psycho-
influence on health above and beyond individual factors, including social factors (such as stress, depression, and social support) may
individual SES (Kawachi & Berkman, 2003). Special study designs influence cancer outcomes. First, stress and depression are related to
and analytical techniques (multilevel analysis) are required to detect health behaviors that are linked to cancer, including cigarette smoking,
the presence of contextual effects. Obstacles abound in drawing excessive alcohol intake, and low levels of physical activity. Cigarettes
causal inferences from such data. Nevertheless, redirecting the focus are a relatively affordable and accessible means of stress reduction,
of interventions from individuals toward improving the quality of particularly in the context of poverty (K. M. Emmons, 2000). A recent
the places where they reside may result in new potential solutions to meta-​analysis also found that levels of job strain and social support
improve outcomes. The potential existence of contextual influences predicted physical activity (Griep et al., 2015). However, psychosocial
on cancer risk offers a promising avenue for cancer prevention. factors may also have direct effects on cancer mortality through a vari-
ety of biological mechanisms (Lutgendorf & Sood, 2011).
Social Networks.  One major way through which SES may have
differential effects on cancer mortality is through effects on social net- Stress and Depression. Psychosocial factors have been also
works. Those with larger personal social networks, defined as the web hypothesized to directly influence immune function (Spiegel, Sephton,
of social relationships that surround an individual (Berkman & Glass, Terr, & Stites, 1998). Acting via the hypothalamic-​pituitary-​adrenal
2000), have lower cancer mortality (Beasley et  al., 2010; Ellwardt, axis in a complex feedback loop between the central nervous (CNS)
van Tilburg, Aartsen, Wittek, & Steverink, 2015; C. H. Kroenke et al., and immune systems, stress increases cortisol levels, which adversely
2006, 2013; Rottenberg et  al., 2014)  and specifically breast cancer affects immune function (Segerstrom & Miller, 2004). Frequent corti-
mortality, since there is almost no work on social ties and cancer sol release that occurs with chronic stress may lead to persistently high
outcomes for cancers other than breast cancer. Persons with larger cortisol levels (Kirschbaum et al., 1995), leading to immune suppres-
social networks are said to be more socially integrated, and those with sion. Because the immune system is involved in the body’s immune
smaller networks are more socially isolated. surveillance mechanism (e.g., eliminating mutated cells), immune
Social isolation may be plausibly hypothesized to influence dysfunction may lead to more rapid development of cancer. Stress may
outcomes through several mechanisms. First, social relation- also promote cancer through DNA damage, faulty DNA repair, inhibi-
ships and interactions influence behaviors and health outcomes by tion of apoptosis, effects on endocrine parameters, or somatic mutation
158

158 Part II: The Magnitude of Cancer


(Forlenza & Baum, 2000; Jenkins, Van Houten, & Bovbjerg, 2014). 2007). There is little work evaluating other types of social interven-
These may be precursors to certain types of cancer such as hormonal tions on cancer mortality.
(Riley, 1981; Rowse, Weinberg, Bellward, & Emerman, 1992)  and
lymphatic cancers (Fox, Goldblatt, & Jones, 1985; Levav et al., 2000). Access to Healthcare and Screening
Despite plausible mechanisms linking psychosocial factors to SES is inversely related to the likelihood of obtaining cancer screening
cancer outcomes, the empirical evidence has been decidedly mixed. (Katz & Hofer, 1994; Katz, Zemencuk, & Hofer, 2000). In their study,
For example, certain types of job stress have been hypothesized to Challenges in Meeting Healthy People 2020 Objectives for Cancer-​
be linked to increased disease risk, including cancer. According to Related Preventive Services, National Health Interview Survey, 2008
Karasek (Karasek & Theorell, 1990), jobs that are both high in psy- and 2010, Brown and colleagues noted that only women in the highest
chological demands and low in decision-​making authority (or control) income group (with incomes ≥ 400% of the federal poverty level or
are hypothesized to result in job strain. High-​strain jobs are typically FPL) surpassed the 2020 Healthy People’s goal of having 81.1% of
over-​represented in lower SES occupations, such as assembly-​line jobs all women engage in breast cancer screening. Among women whose
and certain kinds of jobs in the clerical and service sector. In turn, job incomes less than 200% FPL, less than 62% had screened for breast
strain has been shown to raise the likelihood of deleterious behavioral cancer (US Department of Health and Human Services, 2000). As a
coping responses (such as cigarette smoking, alcohol abuse), thereby consequence, the poor often present with cancer at later, less treat-
increasing the risk of disease outcomes. While the majority of epide- able stages. Bradley and colleagues found that persons < 65 years who
miological studies of job strain and CVD have found support for such were insured by Medicaid had a greater risk of late-​stage diagnosis
an association, the link to cancer still remains elusive. For example, and death for breast, cervical, colon, lung, and prostate cancer than
in the Harvard Nurses’ Health Study, Achat and colleagues (Achat, those not insured by Medicaid (Brewster et  al., 2001). People from
Kawachi, Byrne, Hankinson, & Colditz, 2000)  found no evidence lower SES backgrounds generally present and are diagnosed at later
of an association between job strain and breast cancer incidence in stages, which explains, in part, their higher mortality rates after cancer
a cohort of nurses. In 2007, in a study with data from The Women’s (Boscoe et al., 2016; F. Wang, Luo, & McLafferty, 2010).
Lifestyle and Health Cohort Study (n = 36,332), Kuper and colleagues Even holding cancer stage constant, the poor have been shown to
(Kuper, Yang, Theorell, & Weiderpass, 2007) found that women with have worse survival (Bradley, Given, & Roberts, 2002). A higher prev-
low job control and high job demands (“job strain”) had only a slightly alence of comorbid conditions may complicate recovery. They have
higher risk of breast cancer than women with high job control and low historically had more limited access to healthcare (Bindman et al.,
demands (“low strain”) (HR = 1.2; CI: 0.9, 1.6). Though the number 1995; Hargraves, 2002) and have often received less aggressive treat-
of breast cancer cases was relatively small in both studies (n  =  219 ment (Bristow et al., 2013; Mahal et al., 2014; N. Wang et al., 2015).
in Achat et  al., and n  =  767 in Kuper et  al.), a follow-​up study by The Affordable Care Act (ACA), signed into law in 2010, expanded
Schernhammer and colleagues (Schernhammer et  al., 2003b), with health care to 20 million previously uninsured people (Obama, 2016)
1,030 breast cancer cases, found an inverse, rather than the expected and held promise in reducing SES disparities in preventive services
positive, association between job strain and breast cancer. and treatment (O’Keefe, Meltzer, & Bethea, 2015) particularly
Consistent with the finding in the Schernhammer study, Kroenke through its expansion of affordable Medicaid coverage. However,
and colleagues also found that high stress from caregiving was related these advances may be reversed with the repeal of the ACA and phase
to a possible lower risk of breast cancer (Kroenke et al., 2004). In a out of Medicaid financing intended by the current administration and
meta-​analysis of 116,056 European men and women, job stress was majority Republican congress (Kaplan, 2017).
not significantly associated with colorectal, breast, or prostate cancer. The socioeconomic pattern of colorectal cancer mortality began to
It was positively associated with lung cancer in age-​and sex-​adjusted cross over (from higher rates in persons of higher SES up to the 1980s,
analyses, but was no longer significantly associated after adjustment
for cigarette smoking. There was a suggestion of an inverse associa-
tion of stress with breast and prostate cancer, though these associations 1.05
were not significant (Heikkila et  al., 2013). Gradus and colleagues
found no association between post-​traumatic stress disorder and can- 1
cer (Gradus et al., 2015). Equivocal effects of stress on cancer may be
due to the result of the combination of behavioral, hormonal, and other 0.95
biological effects and the mixed result on cancer incidence, or the pos-
sible methodological issues inherent with the lack of evaluation of
0.9
coping in combination with stress, and their influences on outcomes.
Mortality Rate Ratios (MRR)

0.85
Social Support.  Social support—​for example, tangible, emotional,
informational, and appraisal support (Berkman & Glass, 2000)—​is
one means through which social relationships may influence can- 0.8
cer survival. In addition, Sherbourne and Stewart (1991) identified
affectionate support and “positive social interaction” in patients with 0.75
chronic illness. Tangible support could include rides to the hospital,
trips to the pharmacy, or provision of healthy meals (Hirschman & 0.7
Bourjolly, 2005; Woloshin et al., 1997). Network members may pro-
vide informational support through referrals to physicians and clinics
0.65
or alternative types of treatment, or they may buffer stress (Cohen &
Wills, 1985) through provision of emotional support.
Observational studies of women with breast cancer have typically, 0.6

though not always, found increased mortality rates among those with
5

8
–7

–8

–8

–9

–9

–0

–0

–0
71

76

81

86

91

96

01

06

low levels of social support, adjusted for stage of disease and treatment
19

19

19

19

19

19

20

20

factors (Chou, Stewart, Wild, & Bloom, 2012; Goodwin et al., 1996; 5-year groups
Maunsell, Brisson, & Deschenes, 1995; Reynolds et al., 1994).
Average Diffusion Diffusion 1SD above average Diffusion 1SD below average
However, promising results from early trials (Spiegel, Bloom,
Kraemer, & Gottheil, 1989) suggesting that peer support may improve
cancer survival in metastatic breast cancer patients have not been Figure  9–​4. The modified impact of SES (MMRs) with three different
borne out by more recent studies (Goodwin et al., 2001; Spiegel et al., state-​level diffusion speeds, 1971–​2008.
 159

Socioeconomic Disparities in Cancer Incidence and Mortality 159


then lower rates thereafter) prior to the dissemination of colorectal Svenson, Oberg, Stenling, Palmqvist, & Roos, 2016; Weischer et al.,
screening guidelines (G. K. Singh, Miller, & Hankey, 2002), suggest- 2013; Willeit et al., 2010, 2011; Zhang et al., 2015).
ing that other factors besides screening and health insurance status
are responsible for the reversal in the SES gradient for this disease
(Niu, Roche, Pawlish, & Henry, 2013). Screening for colorectal can- A Note on SES versus Race
cer has increased since the previous version of this chapter (Meissner, SES is correlated with race. African-​Americans and Hispanics are dis-
Breen, Klabunde, & Vernon, 2006), moreso among those of higher proportionately represented in low SES groups, whether measured by
SES, though the SES differential is smaller in states with rapid dif- education, income, or occupation. A great deal of work has explored
fusion of information. Figure  9-​4 shows that states with a higher differential cancer outcomes by race, predominantly focusing on dif-
propensity for diffusion show smaller socioeconomic disparities for ferences between blacks and whites. In the absence of SES data in
colorectal cancer mortality over the 40-​year study period, while those routine sources of data, there has been a tendency to use race as a
states with a propensity toward slow and moderate rates of diffusion proxy for SES (Williams, 1997). Though the practice of conflating
show wider disparities, suggesting that this mechanism may moderate SES with race/​ethnicity has historically posed a barrier to understand-
socioeconomic disparities (A. Wang, Clouston, Rubin, Colen, & Link, ing the etiological relationships of each to cancer outcomes, research-
2012) (Figure 9–​4). ers have more carefully distinguished SES from race in more recent
research.
Biological Influences Related to Aging For some cancers, SES may entirely “explain” the association of
race and cancer. In a study of Detroit women, Bradley and colleagues
While there are many biological mechanisms through which socioeco- found, after accounting for SES, that race was not related to stage at
nomic disadvantage may influence cancer incidence and mortality, we presentation or survival after breast cancer diagnosis (Bradley et al.,
briefly focus on two areas of research that have mushroomed since the 2002). Also, after controlling for income, black women were as likely,
last version of this chapter—​epigenetics and telomeres. Epigenetics is or more likely, than white women to report having had a recent mam-
the study of heritable changes in gene expression, without changes to mogram. Black women from higher SES backgrounds experienced
the underlying DNA, that lead to phenotypic changes in living organ- similar rates of breast cancer compared to affluent white women.
isms; DNA methylation is believed to provide a stable and comprehen- In other cases, SES has explained part but not all of the racial dif-
sive record of epigenetic influence (Hochberg et al., 2011). Telomeres ference in risk (Bristow et al., 2013; Robbins et al., 2000). Tellingly,
are nucleoprotein structures and the protective caps at the ends of lin- race and SES do not always vary in the same direction with respect to
ear eukaryotic chromosomes that are required for genomic stability cancer incidence and mortality. When incidence rates were stratified
(Chiodi & Mondello, 2016; Meena, Rudolph, & Gunes, 2015). We by race and census block SES, Krieger and colleagues (1999) found
address these areas in particular because of their potential relevance to marked heterogeneity in the relationships of SES and race to individ-
cumulative socioeconomic disadvantage or “weathering” (Geronimus, ual cancer sites. This was also true in a recent analysis in the California
1992) and their potential in future research. Cancer Registry (Yin et al., 2010). The strength of association between
SES and cancer may also vary by race. In a study by Yost et al. (Yost,
Epigenetics and DNA Methylation. Socioeconomic dis- Perkins, Cohen, Morris, & Wright, 2001), SES was positively related
advantage may be expressed through epigenetic modifications—​ to breast cancer incidence, but the effect was stronger for Hispanic and
variations in genetic expression, triggered by changes in DNA Asian women than it was for whites and blacks.
methylation, resulting from external and/​ or environmental influ- This chapter has focused specifically on the influence of SES on
ences (Bhattacharjee, Shenoy, & Bairy, 2016; Sapienza & Issa, 2016; cancer outcomes, and fortunately, since the previous edition of this
Trimarchi, Mouangsavanh, & Huang, 2011). Researchers in Canada chapter, there has been a greater tendency, and a greater recognition
and the United Kingdom found that socioeconomic position in child- of the need, to evaluate the independent and joint effects of SES and
hood is associated with DNA methylation differences many years later. race/​ethnicity on cancer outcomes. Research that evaluates the cross-​
Socioeconomic disadvantage in childhood appears to have a greater classification of SES and race, as well as other factors including urban/​
influence on DNA methylation patterns than socioeconomic disad- rural status, nativity, and residence in ethnic enclaves (Schupp, Press,
vantage in adulthood (Borghol et al., 2012). Thus, having a disadvan- & Gomez, 2014), may be the most helpful in trying to understand
taged (or advantaged) background may have profound ramifications health disparities.
for health over the life course, even if socioeconomic circumstances
improve in adulthood. Socioeconomic disadvantage has been related to
DNA methylation of genes involved in inflammation (Needham et al., NEEDED RESEARCH
2015; Stringhini et al., 2015), a major predictor of tumor progression.
Abnormal DNA methylation patterns have been associated with devel- Vice President Joseph Biden called for and President Barack Obama
opment of various cancers, including head and neck cancers, breast launched a “moonshot” initiative to cure cancer (Conrads & Petricoin,
cancer, and prostate cancer (Montenegro et  al., 2016; Nelson et  al., 2016) that would focus on expediting collaborative clinical and labora-
2007; Shaw et al., 2008; Veeck & Esteller, 2010). tory research and encourage information sharing. However, given that
lifestyle and environmental factors have a substantial impact on global
Telomeres.  Low SES in adulthood has been weakly associated cancer incidence and mortality, and that most of these risk factors
with telomere length (Robertson et al., 2013), though social disadvan- are related to socioeconomic factors, national efforts in cancer pre-
tage has been linked to shorter telomeres in children (Mitchell et al., vention could be facilitated by focusing on socioeconomic disparities
2014). Chronic stress, common among those of low SES, has also (Horwitz, 2016) and by evaluating the effects of policies on cancer-​
been related to telomere attrition (Epel et  al., 2004). Aberrations in related health behaviors.
telomere maintenance contribute to cancer development (Basu et al., Understanding the effects of SES on cancer over the life course
2013) and have predicted cancers of the breast, gastrointestinal tract, will necessitate research on the cumulative impact of SES on out-
larynx, bladder, and ovary, as well as chronic lymphocytic leukemia comes using more complex analytic methodological tools (S. Liu,
and cutaneous melanoma and non-​ melanoma skin cancers (Caini Jones, & Glymour, 2010). There is a need for research that considers
et al., 2015; Kotsopoulos et al., 2014; Shen et al., 2012; Valls-​Bautista, other SES measures such as familial wealth. Furthermore, there is a
Pinol-​Felis, Rene-​Espinet, Buenestado-​Garcia, & Vinas-​Salas, 2015; need to explore more fully the combined effects of multiple sociode-
R. Wei, DeVilbiss, & Liu, 2015; Willeit, Willeit, Kloss-​Brandstatter, mographic factors including SES, urban/​rural status (G. K.  Singh,
Kronenberg, & Kiechl, 2011; Willeit et  al., 2010). Telomere short- Williams, Siahpush, & Mulhollen, 2011), race/​ethnicity, and immigra-
ening has been related to cancer progression and earlier mortality tion history on outcomes. To understand the ways in which area-​and
(Duggan et  al., 2014; Kammori et  al., 2015; Svenson et  al., 2008; neighborhood-​level environments affect cancer, more specific research
160

160 Part II: The Magnitude of Cancer


will be needed on social and built-​in environments (Hernandez & ESCALE study (SFCE). Environ Health Perspect, 119(4), 566–​572.
Blazer, 2006). Study of environmental factors and cancer outcomes doi:10.1289/​ehp.1002429
pertinent to SES disparities may also need to begin to consider the Ater, J. (2015). Adult survivorship of pediatric cancers. In L. Foxhall & M.
implications of the progression of climate change on cancer (Portier Rodriguez (Eds.), Advances in cancer survivorship management, MD
Anderson cancer care series (pp. 41–​56). New York: Springer.
et al., 2010). August, K. J., & Sorkin, D. H. (2011). Support and influence in the context
of diabetes management:  Do racial/​ethnic differences exist? J Health
Psychol, 16(5), 711–​ 721. doi:1359105310388320 [pii] 10.1177/​
CONCLUSION 1359105310388320
Bahr, P. R. (2007). Race and nutrition: an investigation of Black-​White differ-
The relationship of SES to cancer is complex and dynamic, and con- ences in health-​related nutritional behaviours. Sociol Health Illn, 29(6),
tinues to evolve. It can be difficult to make generalizations about the 831–​856. doi:10.1111/​j.1467-​9566.2007.01049.x
relationship, because of the heterogeneity of cancer types and outcome Bailey, H. D., Infante-​Rivard, C., Metayer, C., Clavel, J., Lightfoot, T., Kaatsch,
measures, as well as the differences in approaches to measuring SES P., . . . Schuz, J. (2015). Home pesticide exposures and risk of childhood
itself (different indicators, different levels of analysis). Whether the leukemia: findings from the childhood leukemia international consortium.
Int J Cancer, 137(11), 2644–​2663. doi:10.1002/​ijc.29631
association is causal remains controversial in some cases, even though Bandura, A. (1986). Social foundations of thought and action: a social cogni-
several plausible pathways have been identified by which SES could tive theory. Englewood Cliffs, NJ: Prentice-​Hall.
causally affect the onset and prognostic course of specific cancers. Bankole, A., Darroch, J. E., & Singh, S. (1999). Determinants of trends in con-
However, socioeconomic disadvantage is often associated with higher dom use in the United States, 1988–​1995. Fam Plann Perspect, 31(6),
cancer incidence and worse prognosis once diagnosed. SES may be a 264–​271.
“fundamental” determinant of health outcomes, and this appears true Barrera, M., Shaw, A. K., Speechley, K. N., Maunsell, E., & Pogany, L. (2005).
throughout the cancer spectrum—​from cancer incidence, to detection, Educational and social late effects of childhood cancer and related clini-
treatment, and survival. cal, personal, and familial characteristics. Cancer, 104(8), 1751–​1760.
Importantly, there is evidence that the diffusion of knowledge can doi:10.1002/​cncr.21390
Basu, N., Skinner, H. G., Litzelman, K., Vanderboom, R., Baichoo, E., &
mitigate socioeconomic disparities. Investigations of the link between Boardman, L. A. (2013). Telomeres and telomere dynamics: relevance to
SES and cancer will continue to benefit from more sophisticated cancers of the GI tract. Expert Rev Gastroenterol Hepatol, 7(8), 733–​748.
approaches to the measurement of SES, including multidimensional doi:10.1586/​17474124.2013.848790
measures, measures of familial resources, and more sophisticated Beaglehole, R., & Bonita, R. (1997). Public health at the cross-
evaluation of socioeconomic trajectories over the life course, as well roads: achievements and prospects. Cambridge; New York: Cambridge
as better specification and tests of etiological mechanisms. Such an University Press.
exercise is of more than passing academic interest, since any program Beasley, J. M., Newcomb, P. A., Trentham-​Dietz, A., Hampton, J. M., Ceballos,
or policy to reduce the burden of cancer must also pay attention to R. M., Titus-​Ernstoff, L., . . . Holmes, M. D. (2010). Social networks and
mitigating the existing socioeconomic disparities in cancer outcomes. survival after breast cancer diagnosis. J Cancer Surviv, 4(4), 372–​380.
doi:10.1007/​s11764-​010-​0139-​5
Becker, D. M., Yanek, L. R., Koffman, D. M., & Bronner, Y. C. (1999). Body
image preferences among urban African Americans and whites from low
ACKNOWLEDGMENTS income communities. Ethn Dis, 9(3), 377–​386.
Berkman, L., & Glass, T. A. (2000). Social integration, social networks,
We would like to thank Circe Le Compte and Jon Pfefferle for their social support, and health. In L. Berkman & I. Kawachi (Eds.), Social
assistance in updating references. epidemiology (pp. 137–​173). New York: Oxford University Press.
Bhattacharjee, D., Shenoy, S., & Bairy, K. L. (2016). DNA methylation and
References chromatin remodeling:  the blueprint of cancer epigenetics. Scientifica
Achat, H., Kawachi, I., Byrne, C., Hankinson, S., & Colditz, G. (2000). A pro- (Cairo), 2016, 6072357. doi:10.1155/​2016/​6072357
spective study of job strain and risk of breast cancer. Int J Epidemiol, Bindman, A. B., Grumbach, K., Osmond, D., Komaromy, M., Vranizan, K.,
29(4), 622–​628. Lurie, N., . . . Stewart, A. (1995). Preventable hospitalizations and access
Adair, L. S., & Gordon-​Larsen, P. (2001). Maturational timing and overweight to health care. JAMA, 274(4), 305–​311.
prevalence in US adolescent girls. Am J Public Health, 91(4), 642–​644. Blaser, M. J. (1999). Where does Helicobacter pylori come from and why is it
Adler, N. E., Boyce, T., Chesney, M. A., Cohen, S., Folkman, S., Kahn, R. L., going away? JAMA, 282(23), 2260–​2262.
& Syme, S. L. (1993). Socioeconomic status and health: the challenge of Blum, R. W., Beuhring, T., Shew, M. L., Bearinger, L. H., Sieving, R. E., & Resnick,
the gradient. Am Psychol, 49, 15–​24. M. D. (2000). The effects of race/​ethnicity, income, and family structure on
Adler, N. E., & Rehkopf, D. H. (2008). U.S.  disparities in health:  descrip- adolescent risk behaviors. Am J Public Health, 90(12), 1879–​1884.
tions, causes, and mechanisms. Annu Rev Public Health, 29, 235–​252. Boffetta, P., Hashibe, M., La Vecchia, C., Zatonski, W., & Rehm, J. (2006). The
doi:10.1146/​annurev.publhealth.29.020907.090852 burden of cancer attributable to alcohol drinking. Int J Cancer, 119(4),
Ajrouch, K. J., Blandon, A. Y., & Antonucci, T. C. (2005). Social networks 884–​887. doi:10.1002/​ijc.21903
among men and women: the effects of age and socioeconomic status. J Boffetta, P., Tredaniel, J., & Greco, A. (2000). Risk of childhood cancer and
Gerontol B Psychol Sci Soc Sci, 60(6), S311–​S317. adult lung cancer after childhood exposure to passive smoke:  a meta-​
Akinyemiju, T. F., Soliman, A. S., Copeland, G., Banerjee, M., Schwartz, K., analysis. Environ Health Perspect, 108(1), 73–​82.
& Merajver, S. D. (2013). Trends in breast cancer stage and mortality in Boggild, H., Suadicani, P., Hein, H. O., & Gyntelberg, F. (1999). Shift work,
Michigan (1992–​2009) by race, socioeconomic status, and area healthcare social class, and ischaemic heart disease in middle aged and elderly men;
resources. PLoS One, 8(4), e61879. doi:10.1371/​journal.pone.0061879 a 22 year follow up in the Copenhagen Male Study. Occup Environ Med,
Albano, J. D., Ward, E., Jemal, A., Anderson, R., Cokkinides, V. E., Murray, 56(9), 640–​645.
T., . . . Thun, M. J. (2007). Cancer mortality in the United States by educa- Borghol, N., Suderman, M., McArdle, W., Racine, A., Hallett, M., Pembrey,
tion level and race. J Natl Cancer Inst, 99(18), 1384–​1394. doi:10.1093/​ M.,  .  .  .  Szyf, M. (2012). Associations with early-​life socio-​economic
jnci/​djm127 position in adult DNA methylation. Int J Epidemiol, 41(1), 62–​74.
Alexander, C., Piazza, M., Mekos, D., & Valente, T. (2001). Peers, schools, and doi:10.1093/​ije/​dyr147
adolescent cigarette smoking. J Adolesc Health, 29(1), 22–​30. Boscoe, F. P., Henry, K. A., Sherman, R. L., & Johnson, C. J. (2016). The
Altman, D. G., Levine, D. W., Coeytaux, R., Slade, J., & Jaffe, R. (1996). relationship between cancer incidence, stage, and poverty in the United
Tobacco promotion and susceptibility to tobacco use among adolescents States. Int J Cancer, 139(3), 607–​612. doi:10.1002/​ijc.30087
aged 12 through 17  years in a nationally representative sample. Am J Bradley, C. J., Given, C. W., & Roberts, C. (2002). Race, socioeconomic status,
Public Health, 86(11), 1590–​1593. and breast cancer treatment and survival. J Natl Cancer Inst, 94(7), 490–​496.
American Cancer Society. (2016). Physical activity and cancer. Retrieved Braveman, P. (2011). Accumulating knowledge on the social determinants of
from:  www.cancer.org/​NUPA:  http://​www.cancer.org/​acs/​groups/​con- health and infectious disease. Public Health Rep, 126(Suppl 3), 28–​30.
tent/​@nho/​documents/​document/​physicalactivityandcancerpdf.pdf. Braveman, P. A., Cubbin, C., Egerter, S., Chideya, S., Marchi, K. S., Metzler,
Amigou, A., Sermage-​Faure, C., Orsi, L., Leverger, G., Baruchel, A., Bertrand, M., & Posner, S. (2005). Socioeconomic status in health research:  one
Y.,  .  .  .  Clavel, J. (2011). Road traffic and childhood leukemia:  the size does not fit all. JAMA, 294(22), 2879–​2888.
 16

Socioeconomic Disparities in Cancer Incidence and Mortality 161


Brewster, D. H., Thomson, C. S., Hole, D. J., Black, R. J., Stroner, P. L., & Clague, J., & Bernstein, L. (2012). Physical activity and cancer. Curr Oncol
Gillis, C. R. (2001). Relation between socioeconomic status and tumour Rep, 14(6), 550–​558. doi:10.1007/​s11912-​012-​0265-​5
stage in patients with breast, colorectal, ovarian, and lung cancer: results Clegg, L. X., Reichman, M. E., Miller, B. A., Hankey, B. F., Singh, G. K.,
from four national, population based studies. BMJ, 322(7290), 830–​831. Lin, Y. D.,  .  .  .  Edwards, B. K. (2009). Impact of socioeconomic sta-
Bristow, R. E., Powell, M. A., Al-​Hammadi, N., Chen, L., Miller, J. P., Roland, tus on cancer incidence and stage at diagnosis:  selected findings from
P. Y., . . . Cliby, W. A. (2013). Disparities in ovarian cancer care quality the surveillance, epidemiology, and end results:  National Longitudinal
and survival according to race and socioeconomic status. J Natl Cancer Mortality Study. Cancer Causes Control, 20(4), 417–​435. doi:10.1007/​
Inst, 105(11), 823–​832. doi:10.1093/​jnci/​djt065 s10552-​008-​9256-​0
Bureau, U. S. C. (2011). US Census Bureau, 2005–​2009 American Community Cohen, S., & Wills, T. A. (1985). Stress, social support, and the buffering
Survey. Retrieved from: http://​factfinder.census.gov/​servlet/​STTable?_​ hypothesis. Psychol Bull, 98(2), 310–​357.
bm=y&-​geo_​id=01000US&-​qr_​name=ACS_​2009_​5YR_​G00_​S1201&-​ Coker, A. L., Richter, D. L., Valois, R. F., McKeown, R. E., Garrison, C. Z., &
ds_​name=ACS_​2009_​5YR_​G00_​. Vincent, M. L. (1994). Correlates and consequences of early initiation of
Byers, T. E., Wolf, H. J., Bauer, K. R., Bolick-​Aldrich, S., Chen, V. W., Finch, sexual intercourse. J Sch Health, 64(9), 372–​377.
J. L., . . . Patterns of Care Study, G. (2008). The impact of socioeconomic Collaborative Group on Hormonal Factors in Breast Cancer (2012). Menarche,
status on survival after cancer in the United States:  findings from the menopause, and breast cancer risk:  individual participant meta-​
National Program of Cancer Registries Patterns of Care Study. Cancer, analysis, including 118 964 women with breast cancer from 117 epi-
113(3), 582–​591. doi:10.1002/​cncr.23567 demiological studies. Lancet Oncol, 13(11), 1141–​1151. doi:10.1016/​
Caini, S., Raimondi, S., Johansson, H., De Giorgi, V., Zanna, I., Palli, S1470-​2045(12)70425-​4
D., & Gandini, S. (2015). Telomere length and the risk of cutane- Conrads, T. P., & Petricoin, E. F. (2016). The Obama administration’s cancer
ous melanoma and non-​melanoma skin cancer:  a review of the litera- moonshot:  a call for proteomics. Clin Cancer Res. doi:10.1158/​1078-​
ture and meta-​analysis. J Dermatol Sci, 80(3), 168–​174. doi:10.1016/​ 0432.CCR-​16-​0688
j.jdermsci.2015.08.003 Cook, M. B., Gamborg, M., Aarestrup, J., Sorensen, T. I., & Baker, J. L. (2013).
Cannuscio, C. C., Jones, C., Kawachi, I., Colditz, G. A., Berkman, L., & Rimm, Childhood height and birth weight in relation to future prostate cancer
E. (2002). Reverberations of family illness: a longitudinal assessment of risk: a cohort study based on the copenhagen school health records regis-
informal caregiving and mental health status in the Nurses’ Health Study. ter. Cancer Epidemiol Biomarkers Prev, 22(12), 2232–​2240. doi:10.1158/​
Am J Public Health, 92(8), 1305–​1311. 1055-​9965.EPI-​13-​0712
Carroll Chapman, S. L., & Wu, L. T. (2014). E-​cigarette prevalence and cor- Correa, P., & Piazuelo, M. B. (2011). Helicobacter pylori infection and gastric
relates of use among adolescents versus adults: a review and comparison. adenocarcinoma. US Gastroenterol Hepatol Rev, 7(1), 59–​64.
J Psychiatr Res, 54, 43–​54. doi:10.1016/​j.jpsychires.2014.03.005 Costa, L. G., Pellacani, C., Dao, K., Kavanagh, T. J., & Roque, P. J. (2015). The
Castellsague, X., Bosch, F. X., Munoz, N., Meijer, C. J., Shah, K. V., de brominated flame retardant BDE-​47 causes oxidative stress and apop-
Sanjose, S., . . . Franceschi, S. (2002). Male circumcision, penile human totic cell death in vitro and in vivo in mice. Neurotoxicology, 48, 68–​76.
papillomavirus infection, and cervical cancer in female partners. N Engl J doi:10.1016/​j.neuro.2015.03.008
Med, 346(15), 1105–​1112. Crawford, P. B., Story, M., Wang, M. C., Ritchie, L. D., & Sabry, Z. I. (2001).
Centers for Disease Control and Prevention. (1997). Update:  prevalence of Ethnic issues in the epidemiology of childhood obesity. Pediatr Clin
overweight among children, adolescents, and adults—​ United States, North Am, 48(4), 855–​878.
1988–​1994. MMWR Morb Mortal Wkly Rep, 46, 199–​202. Currie, J., & Hyson, R. (1999). Is the impact of health shocks cushioned by
Centers for Disease Control and Prevention. (2000). Behavioral Risk Factor socioeconomic status? The case of low birthweight. Am Econ Rev, 89,
Surveillance System:  Prevalence data:  Nationwide—​ 2000 nutrition. 245–​250.
Retrieved from: http://​apps.nccd.cdc.gov/​brfss/​income.asp?cat=NU&yr= Curtin, F., Morabia, A., & Bernstein, M. (1998). Lifetime exposure to envi-
2000&qkey=4340&state=US. ronmental tobacco smoke among urban women: differences by socioeco-
Chandon, P., & Wansink, B. (2012). Does food marketing need to make us nomic class. Am J Epidemiol, 148(11), 1040–​1047.
fat? A  review and solutions. Nutr Rev, 70(10), 571–​593. doi:10.1111/​ Darmon, N., & Drewnowski, A. (2008). Does social class predict diet quality?
j.1753-​4887.2012.00518.x Am J Clin Nutr, 87(5), 1107–​1117.
Chang, C. M., Su, Y. C., Lai, N. S., Huang, K. Y., Chien, S. H., Chang, Y. Darmon, N., & Drewnowski, A. (2015). Contribution of food prices and diet
H., . . . Lee, C. C. (2012). The combined effect of individual and neigh- cost to socioeconomic disparities in diet quality and health:  a system-
borhood socioeconomic status on cancer survival rates. PLoS One, 7(8), atic review and analysis. Nutr Rev, 73(10), 643–​660. doi:10.1093/​nutrit/​
e44325. doi:10.1371/​journal.pone.0044325 PONE-​D-​12-​15961 [pii] nuv027
Chang, E. T., Yang, J., Alfaro-​Velcamp, T., So, S. K., Glaser, S. L., & Gomez, S. Davey-​ Smith, G., Gunnell, D., & Ben-​ Shlomo, Y. (2001). Life-​ course
L. (2010). Disparities in liver cancer incidence by nativity, acculturation, approaches to socio-​economic differentials in cause-​specific adult mor-
and socioeconomic status in California Hispanics and Asians. Cancer tality. In D. Leon & G. Walt (Eds.), Poverty, inequality and health:  an
Epidemiol Biomarkers Prev, 19(12), 3106–​3118. doi:10.1158/​1055-​9965. international perspective (pp. 88–​124). Oxford: Oxford University Press.
EPI-​10-​0863 De Flora, S., & Bonanni, P. (2011). The prevention of infection-​associated can-
Chelimo, C., Wouldes, T. A., Cameron, L. D., & Elwood, J. M. (2013). Risk cers. Carcinogenesis, 32(6), 787–​795. doi:10.1093/​carcin/​bgr054
factors for and prevention of human papillomaviruses (HPV), geni- De Flora, S., Crocetti, E., Bonanni, P., Ferro, A., Vitale, F., Vaccines, . . . Public,
tal warts and cervical cancer. J Infect, 66(3), 207–​217. doi:10.1016/​ H. (2015). Incidence of infection-​associated cancers in Italy and preven-
j.jinf.2012.10.024 tion strategies. Epidemiol Prev, 39(4 Suppl 1), 14–​20.
Chiodi, I., & Mondello, C. (2016). Telomere and telomerase stability in human Denniston, M. M., Jiles, R. B., Drobeniuc, J., Klevens, R. M., Ward, J. W.,
diseases and cancer. Front Biosci (Landmark Ed), 21, 203–​224. McQuillan, G. M., & Holmberg, S. D. (2014). Chronic hepatitis C virus
Choi, H., & Marks, N. F. (2011). Socioeconomic status, marital status conti- infection in the United States, National Health and Nutrition Examination
nuity and change, marital conflict, and mortality. J Aging Health, 23(4), Survey 2003 to 2010. Ann Intern Med, 160(5), 293–​300. doi:10.7326/​
714–​742. doi:10.1177/​0898264310393339 M13-​1133
Chou, A. F., Stewart, S. L., Wild, R. C., & Bloom, J. R. (2012). Social support Dietz, W. H. (1998a). Childhood weight affects adult morbidity and mortality.
and survival in young women with breast carcinoma. Psychooncology, J Nutr, 128(2 Suppl), 411S–​414S.
21(2), 125–​133. doi:10.1002/​pon.1863 Dietz, W. H. (1998b). Health consequences of obesity in youth: childhood pre-
Christakis, N. A., & Fowler, J. H. (2007). The spread of obesity in a large dictors of adult disease. Pediatrics, 101(3 Pt 2), 518–​525.
social network over 32  years. N Engl J Med, 357(4), 370–​379. Diez-​Roux, A. V., Nieto, F. J., Caulfield, L., Tyroler, H. A., Watson, R. L., &
doi:NEJMsa066082 [pii] 10.1056/​NEJMsa066082 Szklo, M. (1999). Neighbourhood differences in diet: the Atherosclerosis
Chuang, S. C., Gallo, V., Michaud, D., Overvad, K., Tjonneland, A., Clavel-​ Risk in Communities (ARIC) Study. J Epidemiol Community Health,
Chapelon, F., . . . Vineis, P. (2011). Exposure to environmental tobacco 53(1), 55–​63.
smoke in childhood and incidence of cancer in adulthood in never smok- DiMartino, L. D., Birken, S. A., & Mayer, D. K. (2016). The relationship
ers in the European Prospective Investigation into Cancer and Nutrition. between cancer survivors’ socioeconomic status and reports of follow-​
Cancer Causes Control, 22(3), 487–​494. doi:10.1007/​s10552-​010-​9723-​2 up care discussions with providers. J Cancer Educ. doi:10.1007/​
Churilla, T., Egleston, B., Dong, Y., Shaikh, T., Murphy, C., Mantia-​Smaldone, s13187-​016-​1024-​3
G., . . . Anderson, P. (2016). Disparities in the management and outcome Doubeni, C. A., Laiyemo, A. O., Major, J. M., Schootman, M., Lian, M., Park,
of cervical cancer in the United States according to health insurance sta- Y., . . . Sinha, R. (2012). Socioeconomic status and the risk of colorec-
tus. Gynecol Oncol, 141(3), 516–​523. doi:10.1016/​j.ygyno.2016.03.025 tal cancer: an analysis of more than a half million adults in the National
162

162 Part II: The Magnitude of Cancer


Institutes of Health-​ AARP Diet and Health Study. Cancer, 118(14), Forlenza, M. J., & Baum, A. (2000). Psychosocial influences on cancer pro-
3636–​3644. doi:10.1002/​cncr.26677 gression:  alternative cellular and molecular mechanisms. Curr Opin
Doubeni, C. A., Schootman, M., Major, J. M., Stone, R. A., Laiyemo, A. O., Psychiatry, 13(6), 639–​645.
Park, Y.,  .  .  .  Schatzkin, A. (2012). Health status, neighborhood socio- Fox, A. J., Goldblatt, P. O., & Jones, D. R. (1985). Social class mortality differ-
economic context, and premature mortality in the United States:  The entials: artefact, selection or life circumstances? J Epidemiol Community
National Institutes of Health-​AARP Diet and Health Study. Am J Public Health, 39(1), 1–​8.
Health, 102(4), 680–​688. doi:10.2105/​AJPH.2011.300158 Franck, C., Grandi, S. M., & Eisenberg, M. J. (2013). Agricultural subsidies
Du, X. L., Fang, S., & Meyer, T. E. (2008). Impact of treatment and socio- and the American obesity epidemic. Am J Prev Med, 45(3), 327–​333.
economic status on racial disparities in survival among older women doi:10.1016/​j.amepre.2013.04.010
with breast cancer. Am J Clin Oncol, 31(2), 125–​ 132. doi:10.1097/​ Freese, J., & Lutfey, K. (2011). Fundamental causality: challenges of an ani-
COC.0b013e3181587890 mating concept for medical sociology. In B. A. Pescosolido, J. Martin,
Du, X. L., Lin, C. C., Johnson, N. J., & Altekruse, S. (2011). Effects of J. McLeod, & A. Rogers (Eds.), Handbook of the sociology of health,
individual-​level socioeconomic factors on racial disparities in cancer illness, and healing:  a blueprint for the 21st cenutry (pp. 67–​ 81).
treatment and survival: findings from the National Longitudinal Mortality New York: Springer.
Study, 1979–​2003. Cancer, 117(14), 3242–​3251. doi:10.1002/​cncr.25854 Friedenreich, C. M., & Orenstein, M. R. (2002). Physical activity and cancer
Duggan, C., Risques, R., Alfano, C., Prunkard, D., Imayama, I., Holte, prevention: etiologic evidence and biological mechanisms. J Nutr, 132(11
S., . . . McTiernan, A. (2014). Change in peripheral blood leukocyte telo- Suppl), 3456S–​3464S.
mere length and mortality in breast cancer survivors. J Natl Cancer Inst, Fry, R. (2015). More millennials living with parents despite improved job
106(4), dju035. doi:10.1093/​jnci/​dju035 market. Retrieved from http://​www.pewsocialtrends.org/​2015/​07/​29/​
Eckersley, R., Dixon, J., & Douglas, B. (2001). The social origins of health and more-​millennials-​living-​with-​family-​despite-​improved-​job-​market/​
well-​being. Cambridge: Cambridge University Press. Fuchs, V. R. (1983). How we live: an economic perspective on Americans from
Eckhart, W. (1998). Viruses and human cancer. Sci Prog, 81(Pt 4), 315–​328. birth to death. Cambridge, MA: Harvard University Press.
Elder, G. H., Clipp, E. C., Brown, J. S., Martin, L. R., & Friedman, H. W. Gepner, Y., Golan, R., Harman-​ Boehm, I., Henkin, Y., Schwarzfuchs, D.,
(2009). The life-​long mortality risks of World War II experiences. Res Shelef, I., . . . Shai, I. (2015). Effects of initiating moderate alcohol intake
Aging, 31(4), 391–​412. doi:10.1177/​0164027509333447 on cardiometabolic risk in adults with type 2 diabetes: a 2-​year random-
Elder, G. H., Jr., Shanahan, M. J., & Clipp, E. C. (1994). When war comes ized, controlled trial. Ann Intern Med, 163(8), 569–​579. doi:10.7326/​
to men’s lives: life-​course patterns in family, work, and health. Psychol M14-​1650
Aging, 9(1), 5–​16. Geronimus, A. T. (1992). The weathering hypothesis and the health of African-​
Elks, C. E., Loos, R. J., Hardy, R., Wills, A. K., Wong, A., Wareham, N. American women and infants: evidence and speculations. Ethn Dis, 2(3),
J., . . . Ong, K. K. (2012). Adult obesity susceptibility variants are associ- 207–​221.
ated with greater childhood weight gain and a faster tempo of growth: the Giskes, K., Turrell, G., Patterson, C., & Newman, B. (2002). Socioeconomic
1946 British Birth Cohort Study. Am J Clin Nutr, 95(5), 1150–​1156. differences among Australian adults in consumption of fruit and vege-
doi:10.3945/​ajcn.111.027870 tables and intakes of vitamins A, C and folate. J Hum Nutr Diet, 15(5),
Ellwardt, L., van Tilburg, T., Aartsen, M., Wittek, R., & Steverink, N. (2015). 375–​385; discussion 387–​390.
Personal networks and mortality risk in older adults: a twenty-​year longitu- Glymour, M., Avendano, M., & Kawachi, I. (2014). Socioeconomic status and
dinal study. PLoS One, 10(3), e0116731. doi:10.1371/​journal.pone.0116731 health. In L. Berkman, I. Kawachi, & M. Glymour (Eds.), Social epide-
Emmons, K. M. (2000). Health behaviors in a social context. In L. F. Berkman & miology, 2nd ed. Oxford: Oxford University Press.
I. Kawachi (Eds.), Social epidemiology (pp. 242–​266). New York: Oxford Gomez, S. L., Hurley, S., Canchola, A. J., Keegan, T. H., Cheng, I., Murphy,
University Press. J. D., . . . Martinez, M. E. (2016). Effects of marital status and economic
Emmons, K. M., Wong, M., Hammond, S. K., Velicer, W. F., Fava, J. L., resources on survival after cancer:  a population-​based study. Cancer,
Monroe, A. D., & Evans, J. L. (2001). Intervention and policy issues 122(10), 1618–​1625. doi:10.1002/​cncr.29885
related to children’s exposure to environmental tobacco smoke. Prev Med, Gomez, S. L., Shariff-​Marco, S., DeRouen, M., Keegan, T. H., Yen, I. H.,
32(4), 321–​331. doi:10.1006/​pmed.2000.0822 Mujahid, M.,  .  .  .  Glaser, S. L. (2015). The impact of neighborhood
Enewold, L., Horner, M. J., Shriver, C. D., & Zhu, K. (2014). Socioeconomic social and built environment factors across the cancer continuum: current
disparities in colorectal cancer mortality in the United States, 1990–​2007. research, methodological considerations, and future directions. Cancer,
J Community Health, 39(4), 760–​766. doi:10.1007/​s10900-​014-​9824-​z 121(14), 2314–​2330. doi:10.1002/​cncr.29345
Epel, E. S., Blackburn, E. H., Lin, J., Dhabhar, F. S., Adler, N. E., Morrow, Goodman, K. J., & Correa, P. (1995). The transmission of Helicobacter
J. D., & Cawthon, R. M. (2004). Accelerated telomere shortening in pylori:  a critical review of the evidence. Int J Epidemiol, 24(5),
response to life stress. Proc Natl Acad Sci U S A, 101(49), 17312–​17315. 875–​887.
doi:10.1073/​pnas.0407162101 Goodwin, P. J., Leszcz, M., Ennis, M., Koopmans, J., Vincent, L., Guther,
Esser, M. B., Hedden, S. L., Kanny, D., Brewer, R. D., Gfroerer, J. C., & Naimi, H.,  .  .  .  Hunter, J. (2001). The effect of group psychosocial support on
T. S. (2014). Prevalence of alcohol dependence among US adult drinkers, survival in metastatic breast cancer. N Engl J Med, 345(24), 1719–​1726.
2009–​2011. Prev Chronic Dis, 11, E206. doi:10.5888/​pcd11.140329 doi:10.1056/​NEJMoa011871
Evans, A. S., & Mueller, N. E. (1997). Malignant lymphomas. In A. S. Evans & Goodwin, P. J., Leszcz, M., Koopmans, J., Arnold, A., Doll, R., Chochinov,
R. Kaslow (Eds.), Viral infections of humans: epidemiology and control H.,  .  .  .  Pritchard, K. I. (1996). Randomized trial of group psycho-
(4th ed.). New York: Plenum Publishers. social support in metastatic breast cancer:  the BEST study. Breast-​
Faggiano, F., Partanen, T., Kogevinas, M., & Boffetta, P. (1997). Socioeconomic Expressive Supportive Therapy study. Cancer Treat Rev, 22(Suppl
differences in cancer incidence and mortality. IARC Sci Publ, 138, A), 91–​96.
65–​176. Gottlieb, L., Sandel, M., & Adler, N. E. (2013). Collecting and applying data
Farrell, P., & Fuchs, V. R. (1982). Schooling and health: the cigarette connec- on social determinants of health in health care settings. JAMA Intern Med,
tion. J Health Econ, 1(3), 217–​230. 173(11), 1017–​1020. doi:10.1001/​jamainternmed.2013.560
Federal Trade Commission. (1999). Report to Congress for 1997 Pursuant to the Gradus, J. L., Farkas, D. K., Svensson, E., Ehrenstein, V., Lash, T. L., Milstein,
Federal Cigarette Labeling and Advertising Act. Retrieved from: https://​ A.,  .  .  .  Sorensen, H. T. (2015). Posttraumatic stress disorder and can-
www.ftc.gov/​sites/​default/​files/​documents/​reports/​1999-​report-​cigarette-​ cer risk:  a nationwide cohort study. Eur J Epidemiol, 30(7), 563–​568.
sales-​advertising-​and-​promotion-​covering-​1997-​section-​iii.recommenda- doi:10.1007/​s10654-​015-​0032-​7
tion-​regarding-​ftc-​cigarette-​test-​method/​1997cigarettereport.pdf. Granovetter, M. (1973). The strength of weak ties. Am J Sociol, 78(6),
Filippini, T., Heck, J. E., Malagoli, C., Del Giovane, C., & Vinceti, M. (2015). 1360–​1380.
A review and meta-​analysis of outdoor air pollution and risk of child- Greenland, S. (2000). An introduction to instrumental variables for epidemiolo-
hood leukemia. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev, gists. Int J Epidemiol, 29(4), 722–​729.
33(1), 36–​66. doi:10.1080/​10590501.2015.1002999 Griep, R. H., Nobre, A. A., Alves, M. G., da Fonseca Mde, J., Cardoso Lde, O.,
Flegal, K. M., Carroll, M. D., Kit, B. K., & Ogden, C. L. (2012). Prevalence Giatti, L., . . . Chor, D. (2015). Job strain and unhealthy lifestyle: results
of obesity and trends in the distribution of body mass index among US from the baseline cohort study, Brazilian Longitudinal Study of Adult
adults, 1999–​2010. JAMA, 307(5), 491–​497. doi:10.1001/​jama.2012.39 Health (ELSA-​Brasil). BMC Public Health, 15, 309. doi:10.1186/​
Flegal, K. M., Carroll, M. D., Ogden, C. L., & Curtin, L. R. (2010). Prevalence s12889-​015-​1626-​4
and trends in obesity among US adults, 1999–​2008. JAMA, 303(3), 235–​ Griesler, P. C., & Kandel, D. B. (1998). Ethnic differences in correlates of ado-
241. doi:10.1001/​jama.2009.2014 lescent cigarette smoking. J Adolesc Health, 23(3), 167–​180.
 163

Socioeconomic Disparities in Cancer Incidence and Mortality 163


Grossman, M. (1975). The correlation between health and schooling. In N. E. Hogan, D. P., Sun, R., & Cornwell, G. T. (2000). Sexual and fertility behav-
Terleckyj (Ed.), Household production and consumption (pp. 147–​223). iors of American females aged 15–​19 years: 1985, 1990, and 1995. Am J
New York: Columbia University Press. Public Health, 90(9), 1421–​1425.
Grusky, D. B. (1994). Social stratification in sociological perspective. Boulder, Hogue, C. J. (2011). Environmental health disparities in children. Rev Environ
CO: Westview Press. Health, 26(3), 139–​140.
Gupta, S., Wilejto, M., Pole, J. D., Guttmann, A., & Sung, L. (2014). Low Horwitz, R. I. (2016). Equity in cancer care and outcomes of treatment: a differ-
socioeconomic status is associated with worse survival in children with ent type of cancer moonshot. JAMA, 315(12), 1231–​1232. doi:10.1001/​
cancer: a systematic review. PLoS One, 9(2), e89482. doi:10.1371/​jour- jama.2016.2242
nal.pone.0089482 PONE-​D-​13-​42635 [pii] Institute of Medicine. (2011). Institute of Medicine Committee on Public
Gustavsson, P., Nyberg, F., Pershagen, G., Scheele, P., Jakobsson, R., & Health Strategies to Improve Health (9780309161275 9780309161282).
Plato, N. (2002). Low-​dose exposure to asbestos and lung cancer: dose-​ Retrieved from: http://​www.ncbi.nlm.nih.gov/​pubmed/​24983050.
response relations and interaction with smoking in a population-​based Kindig, D. A., Panzer, A. M., & Nielsen-​Bohlman, L. (2004). Health literacy:
case-​referent study in Stockholm, Sweden. Am J Epidemiol, 155(11), a prescription to end confusion. National Academies Press. Washington,
1016–​1022. D.C. Retrieved from: https://​www.nap.edu/​read/​10883/​chapter/​1
Gutensohn, N. M. (1982). Social class and age at diagnosis of Hodgkin’s International Agency for Research on Cancer Ad Hoc Working Group. (1994).
disease:  new epidemiologic evidence for the “two-​disease hypothesis.” Schistosomes, liver flukes and Helicobacter pylori. IARC Monogr Eval
Cancer Treat Rep, 66(4), 689–​695. Carcinog Risks Humans, 61.
Hanske, J., Meyer, C. P., Sammon, J. D., Choueiri, T. K., Menon, M., Lipsitz, Jenkins, F. J., Van Houten, B., & Bovbjerg, D. H. (2014). Effects on DNA dam-
S. R., . . . Trinh, Q. D. (2016). The influence of marital status on the use of age and/​or repair processes as biological mechanisms linking psychologi-
breast, cervical, and colorectal cancer screening. Prev Med, 89, 140–​145. cal stress to cancer risk. J Appl Biobehav Res, 19(1), 3–​23. doi:10.1111/​
doi:10.1016/​j.ypmed.2016.05.017 jabr.12019
Hargraves, J. L. (2002). The Insurance Gap and Minority Health Care, Jin, J. J., Wang, W., Dai, F. X., Long, Z. W., Cai, H., Liu, X. W., . . . Wang, Y. N.
1997–​2001: Center for Studying Health Systems Change Data Bulletin (2016). Marital status and survival in patients with gastric cancer. Cancer
(Tracking report #2). Retrieved from:  http://​www.hschange.org/​ Med, 5(8), 1821–​1829. doi:10.1002/​cam4.758
CONTENT/​443/​443.pdf. Jones, L., Bates, G., McCoy, E., & Bellis, M. A. (2015). Relationship between
Harling, G., Subramanian, S., Barnighausen, T., & Kawachi, I. (2013). alcohol-​attributable disease and socioeconomic status, and the role of
Socioeconomic disparities in sexually transmitted infections among alcohol consumption in this relationship: a systematic review and meta-​
young adults in the United States:  examining the interaction between analysis. BMC Public Health, 15, 400. doi:10.1186/​s12889-​015-​1720-​7
income and race/​ethnicity. Sex Transm Dis, 40(7), 575–​581. doi:10.1097/​ Kamimoto, L. A., Easton, A. N., Maurice, E., Husten, C. G., & Macera, C.
OLQ.0b013e31829529cf A. (1999). Surveillance for five health risks among older adults: United
Harnack, L. J., Jeffery, R. W., & Boutelle, K. N. (2000). Temporal trends in States, 1993–​1997. MMWR CDC Surveill Summ, 48(8), 89–​130.
energy intake in the United States:  an ecologic perspective. Am J Clin Kammori, M., Sugishita, Y., Okamoto, T., Kobayashi, M., Yamazaki, K.,
Nutr, 71(6), 1478–​1484. Yamada, E., & Yamada, T. (2015). Telomere shortening in breast cancer
Harper, S., Lynch, J., Meersman, S. C., Breen, N., Davis, W. W., & Reichman, correlates with the pathological features of tumor progression. Oncol Rep,
M. C. (2009). Trends in area-​socioeconomic and race-​ethnic disparities 34(2), 627–​632. doi:10.3892/​or.2015.4063
in breast cancer incidence, stage at diagnosis, screening, mortality, and Kanjilal, S., Gregg, E. W., Cheng, Y. J., Zhang, P., Nelson, D. E., Mensah,
survival among women ages 50  years and over (1987–​2005). Cancer G., & Beckles, G. L. (2006). Socioeconomic status and trends in dis-
Epidemiol Biomarkers Prev, 18(1), 121–​131. doi:10.1158/​1055-​9965. parities in 4 major risk factors for cardiovascular disease among US
epi-​08-​0679 adults, 1971–​2002. Arch Intern Med, 166(21), 2348–​2355. doi:10.1001/​
Harris, J. L., Pomeranz, J. L., Lobstein, T., & Brownell, K. D. (2009). A cri- archinte.166.21.2348
sis in the marketplace:  how food marketing contributes to childhood Kaplan, T. (2017). Abby Goodnough and Jennifer Steinhauer. March 9, 2017.
obesity and what can be done. Annu Rev Public Health, 30, 211–​225. G.O.P. Health Care Bill Moves Closer After Vote at 4 A.M. New  York
doi:10.1146/​annurev.publhealth.031308.100304 Times https://​www.nytimes.com/​2017/​03/​09/​us/​politics/​health-​bill-​
Hashibe, M., Brennan, P., Chuang, S. C., Boccia, S., Castellsague, X., clears-​ h ouse- ​p anel-​ i n-​ p re- ​ d awn-​ h ours.html?hp&action=click&pgty
Chen, C.,  .  .  .  Boffetta, P. (2009). Interaction between tobacco and pe=Homepage&clickSource=story- ​ h eading&module=first-​ c olumn-​
alcohol use and the risk of head and neck cancer:  pooled analysis in region&region=top-​news&WT.nav=top-​news
the International Head and Neck Cancer Epidemiology Consortium. Karasek, R., & Theorell, T. (1990). Healthy work. New York: Basic Books.
Cancer Epidemiol Biomarkers Prev, 18(2), 541–​ 550. doi:10.1158/​ Katz, S. J., & Hofer, T. P. (1994). Socioeconomic disparities in preventive care
1055-​9965.epi-​08-​0347 persist despite universal coverage: breast and cervical cancer screening in
Hastert, T. A., Beresford, S. A., Sheppard, L., & White, E. (2015). Disparities Ontario and the United States. JAMA, 272(7), 530–​534.
in cancer incidence and mortality by area-​level socioeconomic status: a Katz, S. J., Zemencuk, J. K., & Hofer, T. P. (2000). Breast cancer screening in
multilevel analysis. J Epidemiol Community Health, 69(2), 168–​176. the United States and Canada, 1994: socioeconomic gradients persist. Am
doi:jech-​2014-​204417 [pii] 10.1136/​jech-​2014-​204417 J Public Health, 90(5), 799–​803.
Heikkila, K., Nyberg, S. T., Theorell, T., Fransson, E. I., Alfredsson, L., Kauppinen, T., Teschke, K., Savela, A., Kogevinas, M., & Boffetta, P. (1997).
Bjorner, J. B., . . . Consortium, I. P.-​W. (2013). Work stress and risk of International data base of exposure measurements in the pulp, paper and
cancer: meta-​analysis of 5700 incident cancer events in 116,000 European paper product industries. Int Arch Occup Environ Health, 70(2), 119–​127.
men and women. BMJ, 346, f165. doi:10.1136/​bmj.f165 Kawachi, I., & Berkman, L. F. (2003). Neighborhoods and health.
Heini, A. F., & Weinsier, R. L. (1997). Divergent trends in obesity and fat intake New York: Oxford University Press.
patterns: the American paradox. Am J Med, 102(3), 259–​264. Kent, E. E., Breen, N., Lewis, D. R., de Moor, J. S., Smith, A. W., & Seibel, N.
Hernandez, L. M., & Blazer, D. G. (2006). The impact of social and cultural L. (2015). US trends in survival disparities among adolescents and young
environment on health. In Genes, behavior, and the social environ- adults with non-​ Hodgkin lymphoma. Cancer Causes Control, 26(8),
ment:  moving beyond the nature/​nurture debate (p. xv). Washington, 1153–​1162. doi:10.1007/​s10552-​015-​0609-​1
DC: National Academies Press. Kiecolt-​Glaser, J. K., Glaser, R., Shuttleworth, E. C., Dyer, C. S., Ogrocki, P., &
Hersh, A. L., Stefanick, M. L., & Stafford, R. S. (2004). National use of post- Speicher, C. E. (1987). Chronic stress and immunity in family caregivers
menopausal hormone therapy: annual trends and response to recent evi- of Alzheimer’s disease victims. Psychosom Med, 49(5), 523–​535.
dence. JAMA, 291(1), 47–​53. doi:10.1001/​jama.291.1.47 Kinsey, T., Jemal, A., Liff, J., Ward, E., & Thun, M. (2008). Secular trends
Hirschman, K. B., & Bourjolly, J. N. (2005). How do tangible supports impact in mortality from common cancers in the United States by educational
the breast cancer experience? Soc Work Health Care, 41(1), 17–​32. attainment, 1993–​2001. J Natl Cancer Inst, 100(14), 1003–​1012.
doi:10.1300/​J010v41n01_​02 doi:10.1093/​jnci/​djn207
Hiza, H. A., Casavale, K. O., Guenther, P. M., & Davis, C. A. (2013). Diet qual- Kirschbaum, C., Prussner, J. C., Stone, A. A., Federenko, I., Gaab, J., Lintz,
ity of Americans differs by age, sex, race/​ethnicity, income, and education D.,  .  .  .  Hellhammer, D. H. (1995). Persistent high cortisol responses
level. J Acad Nutr Diet, 113(2), 297–​306. doi:10.1016/​j.jand.2012.08.011 to repeated psychological stress in a subpopulation of healthy men.
Hochberg, Z., Feil, R., Constancia, M., Fraga, M., Junien, C., Carel, J. Psychosom Med, 57(5), 468–​474.
C.,  .  .  .  Albertsson-​Wikland, K. (2011). Child health, developmental Kish, J. K., Yu, M., Percy-​Laurry, A., & Altekruse, S. F. (2014). Racial and eth-
plasticity, and epigenetic programming. Endocr Rev, 32(2), 159–​224. nic disparities in cancer survival by neighborhood socioeconomic status
doi:10.1210/​er.2009-​0039 in Surveillance, Epidemiology, and End Results (SEER) Registries. J Natl
164

164 Part II: The Magnitude of Cancer


Cancer Inst Monogr, 2014(49), 236–​243. doi:10.1093/​jncimonographs/​ Educational achievement in Swiss childhood cancer survivors compared with
lgu020 the general population. Cancer, 118(5), 1439–​1449. doi:10.1002/​cncr.26418
Kogevinas, M., Pearce, N., Susser, M., & Boffetta, P. (1997). Social inequalities Kunkle, B., Bae, S., Singh, K. P., & Roy, D. (2014). Increased risk of childhood
and cancer. IARC Scientific Publications(138). brain tumors among children whose parents had farm-​related pesticide
Kogevinas, M., & Porta, M. (1997). Socioeconomic differences in cancer sur- exposures during pregnancy. JP J Biostat, 11(2), 89–​101.
vival: a review of the evidence. IARC Sci Publ(138), 177–​206. Kuper, H., Yang, L., Theorell, T., & Weiderpass, E. (2007). Job strain and
Koh, H. K. (2011). The ultimate measures of health. Public Health Rep, risk of breast cancer. Epidemiology, 18(6), 764–​ 768. doi:10.1097/​
126(Suppl 3), 14–​15. EDE.0b013e318142c534
Kostova, D., Andes, L., Erguder, T., Yurekli, A., Keskinkılıç, B., Polat, S., . . . Lammers, C., Ireland, M., Resnick, M., & Blum, R. (2000). Influences on
Asma S; Office on Smoking and Health, National Center for Chronic adolescents’ decision to postpone onset of sexual intercourse: a survival
Disease Prevention and Health Promotion, CDC. Cigarette prices and analysis of virginity among youths aged 13 to 18 years. J Adolesc Health,
smoking prevalence after a tobacco tax increase—Turkey, 2008 and 26(1), 42–​48.
2012. MMWR Morb Mortal Wkly Rep. 2014 May 30, 63(21), 457–461. Le Cornet, C., Fervers, B., Dalton, S. O., Feychting, M., Pukkala, E., Tynes,
PubMed PMID: 24871250. T., . . . Schuz, J. (2015). Testicular germ cell tumours and parental occu-
Kotsopoulos, J., Prescott, J., De Vivo, I., Fan, I., McLaughlin, J., Rosen, pational exposure to pesticides:  a register-​based case-​control study in
B., . . . Narod, S. A. (2014). Telomere length and mortality following a the Nordic countries (NORD-​TEST study). Occup Environ Med, 72(11),
diagnosis of ovarian cancer. Cancer Epidemiol Biomarkers Prev, 23(11), 805–​811. doi:10.1136/​oemed-​2015-​102860
2603–​2606. doi:10.1158/​1055-​9965.EPI-​14-​0885 Lee, D., & Jackson, M. (2015). The relationship between lifetime health tra-
Kreuzer, M., Heinrich, J., Kreienbrock, L., Rosario, A. S., Gerken, M., & jectories and socioeconomic attainment in middle age. Soc Sci Res, 54,
Wichmann, H. E. (2002). Risk factors for lung cancer among nonsmok- 96–​112. doi:10.1016/​j.ssresearch.2015.06.023
ing women. Int J Cancer, 100(6), 706–​713. Lee, M. (2012). Endocrine-​ disrupting compounds and pubertal develop-
Krieger, N. (2005). Defining and investigating social disparities in can- ment: the effects of environmental compounds on the timing of puberty in
cer:  critical issues. Cancer Causes Control, 16(1), 5–​14. doi:10.1007/​ humans. In V. R. Preedy (Ed.), Handbook of growth and growth monitor-
s10552-​004-​1251-​5 ing in health and disease. New York: Springer.
Krieger, N., Chen, J. T., & Ebel, G. (1997). Can we monitor socioeconomic Lee, R. E., & Cubbin, C. (2002). Neighborhood context and youth cardiovascu-
inequalities in health? A survey of U.S. health departments’ data collec- lar health behaviors. Am J Public Health, 92(3), 428–​436.
tion and reporting practices. Public Health Reports, 112, 481–​491. Lee, S., Colditz, G. A., Berkman, L. F., & Kawachi, I. (2003). Caregiving and
Krieger, N., Chen, J. T., Kosheleva, A., & Waterman, P. D. (2012). Shrinking, risk of coronary heart disease in U.S. women: a prospective study. Am J
widening, reversing, and stagnating trends in US socioeconomic Prev Med, 24(2), 113–​119. doi:S0749379702005822 [pii]
inequities in cancer mortality for the total, black, and white popula- Lee, S., Kawachi, I., & Grodstein, F. (2004). Does caregiving stress affect
tions: 1960–​2006. Cancer Causes Control, 23(2), 297–​319. doi:10.1007/​ cognitive function in older women? J Nerv Ment Dis, 192(1), 51–​57.
s10552-​011-​9879-​4 doi:10.1097/​01.nmd.0000106000.02232.30
Krieger, N., Chen, J. T., & Waterman, P. D. (2010). Decline in US breast cancer Leong, N. M., Mignone, L. I., Newcomb, P. A., Titus-​Ernstoff, L., Baron, J.
rates after the Women’s Health Initiative: socioeconomic and racial/​ethnic A., Trentham-​Dietz, A., . . . Egan, K. M. (2003). Early life risk factors in
differentials. Am J Public Health, 100(Suppl 1), S132–​139. doi:10.2105/​ cancer: the relation of birth weight to adult obesity. Int J Cancer, 103(6),
AJPH.2009.181628 789–​791. doi:10.1002/​ijc.10886
Krieger, N., Chen, J. T., Waterman, P. D., Rehkopf, D. H., Yin, R., & Coull, Levav, I., Kohn, R., Iscovich, J., Abramson, J. H., Tsai, W. Y., & Vigdorovich,
B. A. (2006). Race/​ethnicity and changing US socioeconomic gradients D. (2000). Cancer incidence and survival following bereavement. Am J
in breast cancer incidence:  California and Massachusetts, 1978–​2002 Public Health, 90(10), 1601–​1607.
(United States). Cancer Causes Control, 17(2), 217–​226. doi:10.1007/​ Levy, D. T., Mumford, E. A., & Compton, C. (2006). Tobacco control poli-
s10552-​005-​0408-​1 cies and smoking in a population of low education women, 1992–​2002. J
Krieger, N., Quesenberry, C., Jr., Peng, T., Horn-​Ross, P., Stewart, S., Brown, Epidemiol Community Health, 60(Suppl 2), 20–​26.
S.,  .  .  .  Ward, F. (1999). Social class, race/​ethnicity, and incidence of Li, R., Serdula, M., Bland, S., Mokdad, A., Bowman, B., & Nelson, D. (2000).
breast, cervix, colon, lung, and prostate cancer among Asian, Black, Trends in fruit and vegetable consumption among adults in 16 US
Hispanic, and White residents of the San Francisco Bay Area, 1988–​92 states:  Behavioral Risk Factor Surveillance System, 1990–​1996. Am J
(United States). Cancer Causes Control, 10(6), 525–​537. Public Health, 90(5), 777–​781.
Kroenke, C. (2008). Socioeconomic status and health: youth development and Liberatos, P., Link, B. G., & Kelsey, J. L. (1988). The measurement of social
neomaterialist and psychosocial mechanisms. Soc Sci Med, 66(1), 31–​42. class in epidemiology. Epidemiol Rev, 10, 87–​121.
doi:10.1016/​j.socscimed.2007.07.018 Litt, J. S., Gerry Taylor, H., Margevicius, S., Schluchter, M., Andreias,
Kroenke, C. H., Hankinson, S. E., Schernhammer, E. S., Colditz, G. A., L., & Hack, M. (2012). Academic achievement of adolescents born
Kawachi, I., & Holmes, M. D. (2004). Caregiving stress, endogenous with extremely low birth weight. Acta Paediatr, 101(12), 1240–​1245.
sex steroid hormone levels, and breast cancer incidence. Am J Epidemiol, doi:10.1111/​j.1651-​2227.2012.02790.x
159(11), 1019–​1027. doi:10.1093/​aje/​kwh148 Liu, F., Wang, X. W., Chen, L., Hu, P., Ren, H., & Hu, H. D. (2016). Systematic
Kroenke, C. H., Kubzansky, L. D., Schernhammer, E. S., Holmes, M. D., & review with meta-​analysis: development of hepatocellular carcinoma in
Kawachi, I. (2006). Social networks, social support, and survival after chronic hepatitis B patients with hepatitis B surface antigen seroclearance.
breast cancer diagnosis. J Clin Oncol, 24(7), 1105–​1111. Aliment Pharmacol Ther, 43(12), 1253–​1261. doi:10.1111/​apt.13634
Kroenke, C. H., Michael, Y., Tindle, H., Gage, E., Chlebowski, R., Garcia, Liu, L., Cozen, W., Bernstein, L., Ross, R. K., & Deapen, D. (2001). Changing
L., . . . Caan, B. J. (2012). Social networks, social support and burden in relationship between socioeconomic status and prostate cancer incidence.
relationships, and mortality after breast cancer diagnosis. Breast Cancer J Natl Cancer Inst, 93(9), 705–​709.
Res Treat, 133(1), 375–​385. doi:10.1007/​s10549-​012-​1962-​3 Liu, S., Jones, R. N., & Glymour, M. M. (2010). Implications of lifecourse epi-
Kroenke, C. H., Michael, Y. L., Shu, X. O., Poole, E. M., Kwan, M. L., demiology for research on determinants of adult disease. Public Health
Nechuta, S., . . . Chen W. Y. (2017). Post-diagnosis social networks, and Rev, 32(2), 489–​511.
lifestyle and treatment factors in the After Breast Cancer Pooling Project. Lochner, K., & Kawachi, I. (2000). Socioeconomic status. In D. Hunter & G. A.
Psychooncology, 26(4), 544-552. doi: 10.1002/pon.4059. Epub 2016 Jan Colditz (Eds.), Cancer prevention: the causes and prevention of cancer
8. PubMed PMID: 26749519; PubMed Central PMCID: PMC4938778. (Vol. 1). Dordrecht, The Netherlands: Kluwer Academic Publishers.
Kroenke, C. H., Quesenberry, C., Kwan, M. L., Sweeney, C., Castillo, A., & Lope, V., Garcia-​ Esquinas, E., Perez-​ Gomez, B., Altzibar, J. M., Gracia-​
Caan, B. J. (2013). Social networks, social support, and burden in rela- Lavedan, E., Ederra, M., . . . Pollan, M. (2016). Perinatal and childhood
tionships, and mortality after breast cancer diagnosis in the Life After factors and risk of breast cancer subtypes in adulthood. Cancer Epidemiol,
Breast Cancer Epidemiology (LACE) study. Breast Cancer Res Treat, 40, 22–​30. doi:10.1016/​j.canep.2015.11.004
137(1), 261–​271. doi:10.1007/​s10549-​012-​2253-​8 Lutgendorf, S. K., & Sood, A. K. (2011). Biobehavioral factors and cancer
Kubzansky, L. D., Seeman, T. E., & Glymour, M. M. (2014). Biological path- progression: physiological pathways and mechanisms. Psychosom Med,
ways linking social conditions and health:  plausible mechanisms and 73(9), 724–​730. doi:10.1097/​PSY.0b013e318235be76
emerging puzzles. In L. F. Berkman, I. Kawachi, & M. M. Glymour Lynch, J., & Kaplan, G. A. (2000). Socioeconomic position. In L. F. Berkman
(Eds.), Social epidemiology (2nd ed., pp. 512–​561). New York: Oxford & I. Kawachi (Eds.), Social epidemiology (pp. 13–​35). New York: Oxford
University Press. University Press.
Kuehni, C. E., Strippoli, M. P., Rueegg, C. S., Rebholz, C. E., Bergstraesser, Ma, J., Xu, J., Anderson, R. N., & Jemal, A. (2012). Widening educational
E., Grotzer, M.,  .  .  .  Swiss Pediatric Oncology Group (SPOG). (2012). disparities in premature death rates in twenty six states in the United
 165

Socioeconomic Disparities in Cancer Incidence and Mortality 165


States, 1993–​2007. PLoS One, 7(7), e41560. doi:10.1371/​ journal. O’Keefe, E. B., Meltzer, J. P., & Bethea, T. N. (2015). Health disparities and
pone.0041560 cancer: racial disparities in cancer mortality in the United States, 2000–​
MacKinnon, J. A., Duncan, R. C., Huang, Y., Lee, D. J., Fleming, L. E., Voti, 2010. Front Public Health, 3, 51. doi:10.3389/​fpubh.2015.00051
L., . . . Wilkinson, J. D. (2007). Detecting an association between socio- Obama, B. (2016). United States health care reform: progress to date and next
economic status and late stage breast cancer using spatial analysis and steps. JAMA, 316(5), 525–​532. doi:10.1001/​jama.2016.9797
area-​based measures. Cancer Epidemiol Biomarkers Prev, 16(4), 756–​ Ogden, C. L., Carroll, M. D., Fryar, C. D., & Flegal, K. M. (2015). Prevalence
762. doi:10.1158/​1055-​9965.EPI-​06-​0392 of obesity among adults and youth:  United States, 2011–​2014. NCHS
Mahal, B. A., Aizer, A. A., Ziehr, D. R., Hyatt, A. S., Lago-​Hernandez, C., Chen, Data Brief, 219, 1–​8.
Y. W., . . . Nguyen, P. L. (2014). The association between insurance status Ogden, C. L., Carroll, M. D., Kit, B. K., & Flegal, K. M. (2012). Prevalence
and prostate cancer outcomes: implications for the Affordable Care Act. of obesity and trends in body mass index among US children and
Prostate Cancer Prostatic Dis, 17(3), 273–​279. doi:10.1038/​pcan.2014.23 adolescents, 1999–​2010. JAMA, 307(5), 483–​ 490. doi:10.1001/​
Malecki, K. C., Bekkedal, M., Hanrahan, L., Stephenson, L., Werner, M., & jama.2012.40
Anderson, H. A. (2006). Linking childhood cancer with potential environ- Ogden, C. L., Carroll, M. D., Kit, B. K., & Flegal, K. M. (2014). Prevalence
mental exposure determinants. WMJ, 105(2), 32–​35. of childhood and adult obesity in the United States, 2011–​2012. JAMA,
Manson, J. E., Hsia, J., Johnson, K. C., Rossouw, J. E., Assaf, A. R., Lasser, N. 311(8), 806–​814. doi:10.1001/​jama.2014.732
L., . . . Women’s Health Initiative Investigators. (2003). Estrogen plus proges- Ogden, C. L., Carroll, M. D., Lawman, H. G., Fryar, C. D., Kruszon-​Moran, D.,
tin and the risk of coronary heart disease. N Engl J Med., 349(6), 523–​534. Kit, B. K., & Flegal, K. M. (2016). Trends in obesity prevalence among
Maule, M., Zugna, D., Migliore, E., Alessi, D., Merletti, F., Onorati, children and adolescents in the United States, 1988–​1994 through 2013–​
R.,  .  .  .  Spadea, T. (2016). Surviving a childhood cancer:  impact 2014. JAMA, 315(21), 2292–​2299. doi:10.1001/​jama.2016.6361
on education and employment. Eur J Cancer Prev. doi:10.1097/​ Ogden, C. L., Lamb, M. M., Carroll, M. D., & Flegal, K. M. (2010). Obesity
CEJ.0000000000000258. [Epub ahead of print] and socioeconomic status in children and adolescents:  United States,
Maunsell, E., Brisson, J., & Deschenes, L. (1995). Social support and survival 2005–​2008. NCHS Data Brief, 51, 1–​8.
among women with breast cancer. Cancer, 76(4), 631–​637. Pachucki, M. A., Jacques, P. F., & Christakis, N. A. (2011). Social network
McCormack, V. A., dos Santos Silva, I., De Stavola, B. L., Mohsen, R., Leon, D. concordance in food choice among spouses, friends, and siblings. Am J
A., & Lithell, H. O. (2003). Fetal growth and subsequent risk of breast can- Public Health, 101(11), 2170–​2177. doi:10.2105/​AJPH.2011.300282
cer: results from long term follow up of Swedish cohort. BMJ, 326(7383), 248. Pais, J. (2014). Cumulative structural disadvantage and racial health dispari-
McDonough, P., Duncan, G. J., Williams, D., & House, J. (1997). Income ties: the pathways of childhood socioeconomic influence. Demography,
dynamics and adult mortality in the United States, 1972 through 1989. 51(5), 1729–​1753. doi:10.1007/​s13524-​014-​0330-​9
Am J Public Health, 87(9), 1476–​1483. Pampel, F. C., Krueger, P. M., & Denney, J. T. (2010). Socioeconomic dis-
Meena, J., Rudolph, K. L., & Gunes, C. (2015). Telomere dysfunction, chro- parities in health behaviors. Annu Rev Sociol, 36, 349–​370. doi:10.1146/​
mosomal instability and cancer. Recent Results Cancer Res, 200, 61–​79. annurev.soc.012809.102529
doi:10.1007/​978-​3-​319-​20291-​4_​3 Pampel, F. C., Mollborn, S., & Lawrence, E. M. (2014). Life course transitions
Meissner, H. I., Breen, N., Klabunde, C. N., & Vernon, S. W. (2006). Patterns of in early adulthood and SES disparities in tobacco use. Soc Sci Res, 43,
colorectal cancer screening uptake among men and women in the United 45–​59. doi:10.1016/​j.ssresearch.2013.08.005
States. Cancer Epidemiol Biomarkers Prev, 15(2), 389–​394. doi:10.1158/​ Pamuk, E., Makuc, D., Heck, K., Reuben, C., & Lochner, K. (1998).
1055-​9965.EPI-​05-​0678 Socioeconomic status and health chartbook: health, United States, 1998.
Merkin, S. S., Stevenson, L., & Powe, N. (2002). Geographic socioeconomic Hyattsville, MD: National Center for Health Statistics.
status, race, and advanced-​stage breast cancer in New  York City. Am J Parise, C. A., & Caggiano, V. (2013). Disparities in race/​ethnicity and socioeco-
Public Health, 92(1), 64–​70. nomic status: risk of mortality of breast cancer patients in the California
Merletti, F., Galassi, C., & Spadea, T. (2011). The socioeconomic deter- Cancer Registry, 2000–​ 2010. BMC Cancer, 13, 449. doi:10.1186/​
minants of cancer. Environ Health, 10(Suppl 1), S7. doi:10.1186/​ 1471-​2407-​13-​449
1476-​069X-​10-​S1-​S7 Parker, J. D., Schoendorf, K. C., & Kiely, J. L. (1994). Associations between
Micic, D. (2001). Obesity in children and adolescents:  a new epidemic? measures of socioeconomic status and low birth weight, small for gesta-
Consequences in adult life. J Pediatr Endocrinol Metab, 14(Suppl 5), tional age, and premature delivery in the United States. Ann Epidemiol,
1345–​1352; discussion 1365. 4(4), 271–​278.
Mitchell, C., Hobcraft, J., McLanahan, S. S., Siegel, S. R., Berg, A., Brooks-​ Parsonnet, J. (1995). The incidence of Helicobacter pylori infection. Aliment
Gunn, J., . . . Notterman, D. (2014). Social disadvantage, genetic sensitiv- Pharmacol Ther, 9(Suppl 2), 45–​51.
ity, and children’s telomere length. Proc Natl Acad Sci U S A, 111(16), Pearce, N., & Matos, E. (1994). Occupational cancer in developing coun-
5944–​5949. doi:10.1073/​pnas.1404293111 tries: introduction. IARC Sci Publ, 129, 1–​3.
Montenegro, M. F., Gonzalez-​Guerrero, R., Sanchez-​Del-​Campo, L., Pinero-​ Pechey, R., & Monsivais, P. (2016). Socioeconomic inequalities in the healthi-
Madrona, A., Cabezas-​Herrera, J., & Rodriguez-​Lopez, J. N. (2016). ness of food choices: Exploring the contributions of food expenditures.
Targeting the epigenetics of the DNA damage response in breast cancer. Prev Med, 88, 203–​209. doi:10.1016/​j.ypmed.2016.04.012
Cell Death Dis, 7, e2180. doi:10.1038/​cddis.2016.85 Pesch, B., Kendzia, B., Gustavsson, P., Jockel, K. H., Johnen, G., Pohlabeln,
Moore, S., Daniel, M., Paquet, C., Dube, L., & Gauvin, L. (2009). Association H., . . . Bruning, T. (2012). Cigarette smoking and lung cancer: relative
of individual network social capital with abdominal adiposity, overweight risk estimates for the major histological types from a pooled analysis
and obesity. J Public Health (Oxf), 31(1), 175–​183. doi:fdn104 [pii] of case-​control studies. Int J Cancer, 131(5), 1210–​1219. doi:10.1002/​
10.1093/​pubmed/​fdn104 ijc.27339
Nakata, A. (2012). Psychosocial job stress and immunity: a systematic review. Peters, S., Glass, D. C., Greenop, K. R., Armstrong, B. K., Kirby, M., Milne, E.,
Methods Mol Biol, 934, 39–​75. doi:10.1007/​978-​1-​62703-​071-​7_​3 & Fritschi, L. (2014). Childhood brain tumours: associations with paren-
Needham, B. L., Smith, J. A., Zhao, W., Wang, X., Mukherjee, B., Kardia, tal occupational exposure to solvents. Br J Cancer, 111(5), 998–​1003.
S. L.,  .  .  .  Diez Roux, A. V. (2015). Life course socioeconomic status doi:10.1038/​bjc.2014.358
and DNA methylation in genes related to stress reactivity and inflamma- Petridou, E. T., Sergentanis, T. N., Perlepe, C., Papathoma, P., Tsilimidos, G.,
tion: the multi-​ethnic study of atherosclerosis. Epigenetics, 10(10), 958–​ Kontogeorgi, E.,  .  .  .  Adami, H. O. (2015). Socioeconomic disparities
969. doi:10.1080/​15592294.2015.1085139 in survival from childhood leukemia in the United States and globally: a
Nelson, W. G., Yegnasubramanian, S., Agoston, A. T., Bastian, P. J., Lee, B. H., meta-​analysis. Ann Oncol, 26(3), 589-​597. doi:10.1093/​annonc/​mdu572
Nakayama, M., & De Marzo, A. M. (2007). Abnormal DNA methylation, Phelan, J. C., Link, B. G., & Tehranifar, P. (2010). Social conditions as fun-
epigenetics, and prostate cancer. Front Biosci, 12, 4254–​4266. damental causes of health inequalities:  theory, evidence, and policy
Nielsen, S. J., & Popkin, B. M. (2003). Patterns and trends in food portion sizes, implications. J Health Soc Behav, 51(Suppl), S28–​S40. doi:10.1177/​
1977–​1998. JAMA, 289(4), 450–​453. 0022146510383498
Nightingale, T. E., & Gruber, J. (1994). Helicobacter and human cancer. J Natl Pierce, J. P., Choi, W. S., Gilpin, E. A., Farkas, A. J., & Berry, C. C. (1998).
Cancer Inst, 86(20), 1505–​1509. Tobacco industry promotion of cigarettes and adolescent smoking. JAMA,
Niu, X., Roche, L. M., Pawlish, K. S., & Henry, K. A. (2013). Cancer sur- 279(7), 511-​515.
vival disparities by health insurance status. Cancer Med, 2(3), 403–​411. Piernas, C., & Popkin, B. M. (2011). Increased portion sizes from energy-​dense
doi:10.1002/​cam4.84 foods affect total energy intake at eating occasions in US children and
O’Donnell, B. L., O’Donnell, C. R., & Stueve, A. (2001). Early sexual initia- adolescents: patterns and trends by age group and sociodemographic char-
tion and subsequent sex-​related risks among urban minority youth:  the acteristics, 1977–​2006. Am J Clin Nutr, 94(5), 1324–​1332. doi:10.3945/​
reach for health study. Fam Plann Perspect, 33(6), 268–​275. ajcn.110.008466
16

166 Part II: The Magnitude of Cancer


Platz, E. A., Giovannucci, E., Rimm, E. B., Curhan, G. C., Spiegelman, D., Sapienza, C., & Issa, J. P. (2016). Diet, nutrition, and cancer epigenetics. Annu
Colditz, G. A., & Willett, W. C. (1998). Retrospective analysis of birth Rev Nutr, 36, 665–​681. doi:10.1146/​annurev-​nutr-​121415-​112634
weight and prostate cancer in the Health Professionals Follow-​up Study. Schernhammer, E. S., Laden, F., Speizer, F. E., Willett, W. C., Hunter, D. J.,
Am J Epidemiol, 147(12), 1140–​1144. Kawachi, I., & Colditz, G. A. (2001). Rotating night shifts and risk of
Popkin, B. M., Siega-​Riz, A. M., & Haines, P. S. (1996). A comparison of breast cancer in women participating in the nurses’ health study. J Natl
dietary trends among racial and socioeconomic groups in the United Cancer Inst, 93(20), 1563–​1568.
States. N Engl J Med, 335(10), 716–​720. Schernhammer, E. S., Laden, F., Speizer, F. E., Willett, W. C., Hunter, D. J.,
Portier, C. J., K., T. T., Carter, S. R., Dilworth, C. H., Grambsch, A. E., Gohlke, Kawachi, I.,  .  .  .  Colditz, G. A. (2003a). Night-​shift work and risk of
J.,  .  .  .  Whung, P.-​Y. (2010). A human health perspective on climate colorectal cancer in the nurses’ health study. J Natl Cancer Inst, 95(11),
change: a report outlining the research needs on the human health effects 825–​828.
of climate change. Retrieved from: www.niehs.nih.gov/​climatereport. Schernhammer, E. S., Hankinson, S., Rosner, B., Kroenke, C. H., Speizer, F.
Powell, L. M., Szczypka, G., & Chaloupka, F. J. (2010). Trends in exposure E., Willett, W. C., . . . Kawachi, I. (2003b). Job stress and breast cancer
to television food advertisements among children and adolescents in the risk: the Nurses’ Health Study. Unpublished data.
United States. Arch Pediatr Adolesc Med, 164(9), 794–​802. doi:10.1001/​ Schottenfeld, D., & Beebe-​Dimmer, J. (2015). The cancer burden attribut-
archpediatrics.2010.139 able to biologic agents. Ann Epidemiol, 25(3), 183–​187. doi:10.1016/​
Powell, L. M., Wada, R., & Kumanyika, S. K. (2014). Racial/​ethnic and income j.annepidem.2014.11.016
disparities in child and adolescent exposure to food and beverage tele- Schreder, E. D., Uding, N., & La Guardia, M. J. (2016). Inhalation a signifi-
vision ads across the U.S.  media markets. Health Place, 29, 124–​131. cant exposure route for chlorinated organophosphate flame retardants.
doi:10.1016/​j.healthplace.2014.06.006 Chemosphere, 150, 499–​504. doi:10.1016/​j.chemosphere.2015.11.084
Praestegaard, C., Kjaer, S. K., Nielsen, T. S., Jensen, S. M., Webb, P. M., Schulz, R., & Beach, S. R. (1999). Caregiving as a risk factor for mortal-
Nagle, C. M., . . . Ovarian Cancer Association, C. (2016). The association ity:  the Caregiver Health Effects Study. JAMA, 282(23), 2215–​2219.
between socioeconomic status and tumour stage at diagnosis of ovarian doi:joc91040 [pii]
cancer: a pooled analysis of 18 case-​control studies. Cancer Epidemiol, Schupp, C. W., Press, D. J., & Gomez, S. L. (2014). Immigration factors and
41, 71–​79. doi:10.1016/​j.canep.2016.01.012 prostate cancer survival among Hispanic men in California: does neigh-
Pudrovska, T., & Anikputa, B. (2012). The role of early-​life socioeconomic sta- borhood matter? Cancer, 120(9), 1401–​1408. doi:10.1002/​cncr.28587
tus in breast cancer incidence and mortality: unraveling life course mecha- Schuster, M. A., Franke, T., & Pham, C. B. (2002). Smoking patterns of house-
nisms. J Aging Health, 24(2), 323–​344. doi:10.1177/​0898264311422744 hold members and visitors in homes with children in the United States.
Rehkopf, D., Glymour, M., & Osypuk, T. (2016). The consistency assumption Arch Pediatr Adolesc Med, 156(11), 1094–​1100.
for causal inference in social epidemiology:  when a rose is not a rose. Scragg, R., Laugesen, M., & Robinson, E. (2002). Cigarette smoking, pocket
Curr Epidemiol Rep, 3(1), 63–​71. money and socioeconomic status:  results from a national survey of 4th
Reynolds, P., Boyd, P. T., Blacklow, R. S., Jackson, J. S., Greenberg, R. S., form students in 2000. N Z Med J, 115(1158), U108.
Austin, D. F., . . . Edwards, B. K. (1994). The relationship between social Segerstrom, S. C., & Miller, G. E. (2004). Psychological stress and the human
ties and survival among black and white breast cancer patients. National immune system:  a meta-​analytic study of 30  years of inquiry. Psychol
Cancer Institute Black/​ White Cancer Survival Study Group. Cancer Bull, 130(4), 601–​630. doi:10.1037/​0033–​2909.130.4.601
Epidemiol Biomarkers Prev, 3(3), 253–​259. Shariff-​Marco, S., Yang, J., John, E. M., Sangaramoorthy, M., Hertz, A., Koo,
Rickard, B. J., Okrent, A. M., & Alston, J. M. (2013). How have agricultural J.,  .  .  .  Gomez, S. L. (2014). Impact of neighborhood and individual
policies influenced caloric consumption in the United States? Health socioeconomic status on survival after breast cancer varies by race/​eth-
Econ, 22(3), 316–​339. doi:10.1002/​hec.2799 nicity:  the Neighborhood and Breast Cancer Study. Cancer Epidemiol
Riley, V. (1981). Psychoneuroendocrine influences on immunocompetence and Biomarkers Prev, 23(5), 793–​811. doi:10.1158/​1055-​9965.EPI-​13-​0924
neoplasia. Science, 212(4499), 1100–​1109. Sharpe, P. A., Whitaker, K., Alia, K. A., Wilcox, S., & Hutto, B. (2016). Dietary
Robbins, J. M., Vaccarino, V., Zhang, H., & Kasl, S. V. (2000). Excess intake, behaviors and psychosocial factors among women from food-​
type 2 diabetes in African-​American women and men aged 40–​74 secure and food-​insecure households in the United States. Ethn Dis,
and socioeconomic status:  evidence from the Third National Health 26(2), 139–​146. doi:10.18865/​ed.26.2.139
and Nutrition Examination Survey. J Epidemiol Community Health, Shaw, R. J., Hall, G. L., Lowe, D., Liloglou, T., Field, J. K., Sloan, P., & Risk,
54(11), 839–​845. J. M. (2008). The role of pyrosequencing in head and neck cancer epi-
Robert, J., Cheney, M. K., & Azad, M. R. (2009). Point-​of-​sale marketing genetics: correlation of quantitative methylation data with gene expres-
of tobacco products:  taking advantage of the socially disadvantaged? J sion. Arch Otolaryngol Head Neck Surg, 134(3), 251–​256. doi:10.1001/​
Health Care Poor Underserved, 20(2), 489–​506. doi:10.1353/​hpu.0.0147 archoto.2007.50
Robertson, T., Batty, G. D., Der, G., Fenton, C., Shiels, P. G., & Benzeval, Shebl, F. M., Capo-​Ramos, D. E., Graubard, B. I., McGlynn, K. A., & Altekruse,
M. (2013). Is socioeconomic status associated with biological aging as S. F. (2012). Socioeconomic status and hepatocellular carcinoma in the
measured by telomere length? Epidemiol Rev, 35, 98–​111. doi:10.1093/​ United States. Cancer Epidemiol Biomarkers Prev, 21(8), 1330–​1335.
epirev/​mxs001 doi:10.1158/​1055-​9965.EPI-​12-​0124
Ross, C. E., & Wu, C. (1995). The links between education and health. Am Shen, J., Gammon, M. D., Terry, M. B., Bradshaw, P. T., Wang, Q., Teitelbaum,
Sociol Rev, 60, 719–​745. S. L.,  .  .  .  Santella, R. M. (2012). Genetic polymorphisms in telomere
Rothman, K. J. (1980). The proportion of cancer attributable to alcohol con- pathway genes, telomere length, and breast cancer survival. Breast
sumption. Prev Med, 9(2), 174–​179. Cancer Res Treat, 134(1), 393–​400. doi:10.1007/​s10549-​012-​2058-​9
Rottenberg, Y., Litwin, H., Manor, O., Paltiel, A., Barchana, M., & Paltiel, O. Sherbourne, C. D., & Stewart, A. L. (1991). The MOS social support survey.
(2014). Prediagnostic self-​assessed health and extent of social networks Soc Sci Med, 32(6), 705–​714.
predict survival in older individuals with cancer: a population based cohort Shi, L. (1998). Sociodemographic characteristics and individual health behav-
study. J Geriatr Oncol, 5(4), 400–​407. doi:10.1016/​j.jgo.2014.08.001 iors. South Med J, 91(10), 933–​941.
Roura, E., Castellsague, X., Pawlita, M., Travier, N., Waterboer, T., Margall, Shi, R. L., Qu, N., Lu, Z. W., Liao, T., Gao, Y., & Ji, Q. H. (2016). The impact
N.,  .  .  .  Riboli, E. (2014). Smoking as a major risk factor for cervical of marital status at diagnosis on cancer survival in patients with differ-
cancer and pre-​cancer: results from the EPIC cohort. Int J Cancer, 135(2), entiated thyroid cancer. Cancer Med, 5(8), 2145–​ 2154. doi:10.1002/​
453–​466. doi:10.1002/​ijc.28666 cam4.778
Rowse, G. J., Weinberg, J., Bellward, G. D., & Emerman, J. T. (1992). Shield, K. D., Soerjomataram, I., & Rehm, J. (2016). Alcohol use and breast
Endocrine mediation of psychosocial stressor effects on mouse mammary cancer:  a critical review. Alcohol Clin Exp Res, 40(6), 1166–​1181.
tumor growth. Cancer Lett, 65(1), 85–​93. doi:10.1111/​acer.13071
Sadana, R., & Harper, S. (2011). Data systems linking social determinants of Shrestha, A., Ritz, B., Wilhelm, M., Qiu, J., Cockburn, M., & Heck, J. E.
health with health outcomes: advancing public goods to support research (2014). Prenatal exposure to air toxics and risk of Wilms’ tumor in 0-​to
and evidence-​based policy and programs. Public Health Rep, 126(Suppl 5-​year-​old children. J Occup Environ Med, 56(6), 573–​578. doi:10.1097/​
3), 6–​13. JOM.0000000000000167
Sandhu, M. S., Luben, R., Day, N. E., & Khaw, K. T. (2002). Self-​reported Siega-​Riz, A. M., & Popkin, B. M. (2001). Dietary trends among low socioec-
birth weight and subsequent risk of colorectal cancer. Cancer Epidemiol onomic status women of childbearing age in the United States from 1977
Biomarkers Prev, 11(9), 935–​938. to 1996: a comparison among ethnic groups. J Am Med Womens Assoc,
Santelli, J. S., Lowry, R., Brener, N. D., & Robin, L. (2000). The association of 56(2), 44–​48, 72.
sexual behaviors with socioeconomic status, family structure, and race/​ Sieh, W., Rothstein, J. H., McGuire, V., & Whittemore, A. S. (2014). The role
ethnicity among US adolescents. Am J Public Health, 90(10), 1582–​1588. of genome sequencing in personalized breast cancer prevention. Cancer
 167

Socioeconomic Disparities in Cancer Incidence and Mortality 167


Epidemiol Biomarkers Prev, 23(11), 2322–​2327. doi:10.1158/​1055-​9965. Tao, L., Ladabaum, U., Gomez, S. L., & Cheng, I. (2014). Colorectal can-
EPI-​14-​0559 cer mortality among Hispanics in California:  differences by neighbor-
Singh, G. K., Miller, B. A., & Hankey, B. F. (2002). Changing area socioeco- hood socioeconomic status and nativity. Cancer, 120(22), 3510–​3518.
nomic patterns in U.S.  cancer mortality, 1950–​1998. Part II:  Lung and doi:10.1002/​cncr.28837
colorectal cancers. J Natl Cancer Inst, 94(12), 916–​925. Taylor, G. S., Long, H. M., Brooks, J. M., Rickinson, A. B., &
Singh, G. K., Miller, B. A., Hankey, B. F., Feuer, E. J., & Pickle, L. W. Hislop, A. D. (2015). The immunology of Epstein-​ Barr virus-​
(2002). Changing area socioeconomic patterns in U.S. cancer mortality, induced disease. Annu Rev Immunol, 33, 787–​ 821. doi:10.1146/​
1950–​1998. Part I: All cancers among men. J Natl Cancer Inst, 94(12), annurev-​immunol-​032414-​112326
904–​915. Taylor, R., Cumming, R., Woodward, A., & Black, M. (2001). Passive smoking
Singh, G. K., Williams, S. D., Siahpush, M., & Mulhollen, A. (2011). and lung cancer: a cumulative meta-​analysis. Aust N Z J Public Health,
Socioeconomic, rural-​urban, and racial inequalities in US cancer mor- 25(3), 203–​211.
tality: Part I: All cancers and lung cancer and Part II: Colorectal, pros- Thompson, F. E., McNeel, T. S., Dowling, E. C., Midthune, D., Morrissette, M.,
tate, breast, and cervical cancers. J Cancer Epidemiol, 2011, 107497. & Zeruto, C. A. (2009). Interrelationships of added sugars intake, socio-
doi:10.1155/​2011/​107497 economic status, and race/​ethnicity in adults in the United States: National
Singh, S., Darroch, J. E., & Frost, J. J. (2001). Socioeconomic disadvantage and Health Interview Survey, 2005. J Am Diet Assoc, 109(8), 1376–​1383.
adolescent women’s sexual and reproductive behavior:  the case of five doi:10.1016/​j.jada.2009.05.002
developed countries. Fam Plann Perspect, 33(6), 251–​258, 289. Thun, M. J., DeLancey, J. O., Center, M. M., Jemal, A., & Ward, E. M. (2010).
Sistino, J. J., & Ellis, C., Jr. (2011). Effects of health disparities on survival The global burden of cancer:  priorities for prevention. Carcinogenesis,
after neonatal heart surgery: why should racial, ethnic, gender, and socio- 31(1), 100–​110. doi:10.1093/​carcin/​bgp263
economic status be included in the risk analysis? J Extra Corpor Technol, Trichopoulos, D. (1990). Hypothesis:  does breast cancer originate in utero?
43(4), 232–​235. Lancet, 335(8695), 939–​940.
Smith, K., & Christakis, N. (2008). Social networks and health. Ann Rev Sociol, Trimarchi, M. P., Mouangsavanh, M., & Huang, T. H. (2011). Cancer epi-
34, 405–​429. genetics:  a perspective on the role of DNA methylation in acquired
Smith, N. R., Jensen, B. W., Zimmermann, E., Gamborg, M., Sorensen, T. endocrine resistance. Chin J Cancer, 30(11), 749–​ 756. doi:10.5732/​
I., & Baker, J. L. (2016). Associations between birth weight and colon cjc.011.10128
and rectal cancer risk in adulthood. Cancer Epidemiol, 42, 181–​185. Tyas, S. L., & Pederson, L. L. (1998). Psychosocial factors related to adolescent
doi:10.1016/​j.canep.2016.05.003 smoking: a critical review of the literature. Tob Control, 7(4), 409–​420.
Spiegel, D., Bloom, J. R., Kraemer, H. C., & Gottheil, E. (1989). Effect of psy- US Department of Health and Human Services. (2000). Healthy people
chosocial treatment on survival of patients with metastatic breast cancer. 2010: understanding and improving health. 2nd ed. Retrieved from: http://​
Lancet, 2(8668), 888–​891. www.health.gov/​healthypeople/​document/​html/​volume1/​03cancer.htm.
Spiegel, D., Butler, L. D., Giese-​Davis, J., Koopman, C., Miller, E., DiMiceli, NTP (National Toxicology Program). 2002. Report on Carcinogens, 10th Ed.
S., . . . Kraemer, H. C. (2007). Effects of supportive-​expressive group ther- U.S. Department of Health and Human Services, Public Health Service,
apy on survival of patients with metastatic breast cancer: a randomized National Toxicology Program, Research Triangle Park, NC.
prospective trial. Cancer, 110(5), 1130–​1138. doi:10.1002/​cncr.22890 Valente, T. W. (2009). Social networks and health: models, methods, and appli-
Spiegel, D., Sephton, S. E., Terr, A. I., & Stites, D. P. (1998). Effects of psy- cations. New York: Oxford University Press.
chosocial treatment in prolonging cancer survival may be mediated by Valls-​Bautista, C., Pinol-​Felis, C., Rene-​Espinet, J. M., Buenestado-​Garcia,
neuroimmune pathways. Ann N Y Acad Sci, 840, 674–​683. J., & Vinas-​Salas, J. (2015). In colon cancer, normal colon tissue and
Spycher, B. D., Feller, M., Roosli, M., Ammann, R. A., Diezi, M., Egger, M., & blood cells have altered telomere lengths. J Surg Oncol, 111(7), 899–​904.
Kuehni, C. E. (2015). Childhood cancer and residential exposure to high- doi:10.1002/​jso.23894
ways: a nationwide cohort study. Eur J Epidemiol, 30(12), 1263–​1275. van Hasselt, M., Kruger, J., Han, B., Caraballo, R. S., Penne, M. A., Loomis, B.,
doi:10.1007/​s10654-​015-​0091-​9 & Gfroerer, J. C. (2015). The relation between tobacco taxes and youth
Steenland, K., Henley, J., Calle, E., & Thun, M. (2004). Individual-​and area-​ and young adult smoking: what happened following the 2009 U.S. fed-
level socioeconomic status variables as predictors of mortality in a cohort eral tax increase on cigarettes? Addict Behav, 45, 104–​109. doi:10.1016/​
of 179,383 persons. Am J Epidemiol, 159(11), 1047–​1056. doi:10.1093/​ j.addbeh.2015.01.023
aje/​kwh129 van Jaarsveld, C. H., Miles, A., Edwards, R., & Wardle, J. (2006). Marriage and
Steenland, K., Henley, J., & Thun, M. (2002). All-​cause and cause-​specific cancer prevention: does marital status and inviting both spouses together
death rates by educational status for two million people in two American influence colorectal cancer screening participation? J Med Screen, 13(4),
Cancer Society cohorts, 1959–​1996. Am J Epidemiol, 156(1), 11–​21. 172–​176.
Steenland, K., Hu, S., & Walker, J. (2004). All-​cause and cause-​specific mortal- Van Maele-​Fabry, G., Hoet, P., & Lison, D. (2013). Parental occupational expo-
ity by socioeconomic status among employed persons in 27 US states, sure to pesticides as risk factor for brain tumors in children and young
1984–​1997. Am J Public Health, 94(6), 1037–​1042. adults:  a systematic review and meta-​analysis. Environ Int, 56, 19–​31.
Steinkellner, A. R., Denison, S. E., Eldridge, S. L., Lenzi, L. L., Chen, doi:10.1016/​j.envint.2013.02.011
W., & Bowlin, S. J. (2012). A decade of postmenopausal hormone Van Maele-​Fabry, G., Lantin, A. C., Hoet, P., & Lison, D. (2010). Childhood
therapy prescribing in the United States:  long-​ term effects of the leukaemia and parental occupational exposure to pesticides:  a system-
Women’s Health Initiative. Menopause, 19(6), 616–​621. doi:10.1097/​ atic review and meta-​analysis. Cancer Causes Control, 21(6), 787–​809.
gme.0b013e31824bb039 doi:10.1007/​s10552-​010-​9516-​7
Steinmaus, C., Ferreccio, C., Acevedo, J., Yuan, Y., Liaw, J., Duran, V., . . . Smith, Vargas, A. J., & Thompson, P. A. (2012). Diet and nutrient factors in
A. H. (2014). Increased lung and bladder cancer incidence in adults after colorectal cancer risk. Nutr Clin Pract, 27(5), 613–​623. doi:10.1177/​
in utero and early-​life arsenic exposure. Cancer Epidemiol Biomarkers 0884533612454885
Prev, 23(8), 1529–​1538. doi:10.1158/​1055-​9965.EPI-​14-​0059 Vatten, L. J., Maehle, B. O., Lund Nilsen, T. I., Tretli, S., Hsieh, C. C.,
Strauss, R. S., & Knight, J. (1999). Influence of the home environment on the Trichopoulos, D., & Stuver, S. O. (2002). Birth weight as a predic-
development of obesity in children. Pediatrics, 103(6), e85. tor of breast cancer:  a case-​control study in Norway. Br J Cancer,
Stringhini, S., Polidoro, S., Sacerdote, C., Kelly, R. S., van Veldhoven, K., 86(1), 89–​91.
Agnoli, C., . . . Vineis, P. (2015). Life-​course socioeconomic status and Veeck, J., & Esteller, M. (2010). Breast cancer epigenetics: from DNA meth-
DNA methylation of genes regulating inflammation. Int J Epidemiol, ylation to microRNAs. J Mammary Gland Biol Neoplasia, 15(1), 5–​17.
44(4), 1320–​1330. doi:10.1093/​ije/​dyv060 doi:10.1007/​s10911-​010-​9165-​1
Su, L. J., & Arab, L. (2001). Nutritional status of folate and colon cancer Villingshoj, M., Ross, L., Thomsen, B. L., & Johansen, C. (2006). Does mar-
risk:  evidence from NHANES I  epidemiologic follow-​up study. Ann ital status and altered contact with the social network predict colorec-
Epidemiol, 11(1), 65–​72. tal cancer survival? Eur J Cancer, 42(17), 3022–​ 3027. doi:10.1016/​
Svenson, U., Nordfjall, K., Stegmayr, B., Manjer, J., Nilsson, P., Tavelin, j.ejca.2006.04.027
B., . . . Roos, G. (2008). Breast cancer survival is associated with telo- Vohra, J., Marmot, M. G., Bauld, L., & Hiatt, R. A. (2016). Socioeconomic pos-
mere length in peripheral blood cells. Cancer Res, 68(10), 3618–​3623. ition in childhood and cancer in adulthood: a rapid-​review. J Epidemiol
doi:10.1158/​0008-​5472.CAN-​07-​6497 Community Health, 70(6), 629–​634. doi:10.1136/​jech-​2015-​206274
Svenson, U., Oberg, A., Stenling, R., Palmqvist, R., & Roos, G. (2016). von Ehrenstein, O. S., Heck, J. E., Park, A., Cockburn, M., Escobedo, L., &
Telomere length in peripheral leukocytes is associated with immune cell Ritz, B. (2015). In utero and early-​life exposure to ambient air toxics
tumor infiltration and prognosis in colorectal cancer patients. Tumour and childhood brain tumors:  a population-​based case-​control study in
Biol, 37(8), 10877–​10882. doi:10.1007/​s13277-​016-​4987-​0 California, USA. Environ Health Perspect, 124(7), 1093–​1099.
168

168 Part II: The Magnitude of Cancer


Wakabayashi, I. (2014). Light-​to-​moderate alcohol drinking reduces the impact Willeit, P., Willeit, J., Kloss-​ Brandstatter, A., Kronenberg, F., & Kiechl,
of obesity on the risk of diabetes mellitus. J Stud Alcohol Drugs, 75(6), S. (2011). Fifteen-​ year follow-​
up of association between telomere
1032–​1038. length and incident cancer and cancer mortality. JAMA, 306(1), 42–​44.
Wang, A., Clouston, S. A., Rubin, M. S., Colen, C. G., & Link, B. G. (2012). doi:10.1001/​jama.2011.901
Fundamental causes of colorectal cancer mortality:  the implications Willeit, P., Willeit, J., Mayr, A., Weger, S., Oberhollenzer, F., Brandstatter,
of informational diffusion. Milbank Q, 90(3), 592–​ 618. doi:10.1111/​ A.,  .  .  .  Kiechl, S. (2010). Telomere length and risk of incident cancer
j.1468-​0009.2012.00675.x and cancer mortality. JAMA, 304(1), 69–​75. doi:10.1001/​jama.2010.897
Wang, C. Y., de Dieu Tapsoba, J., Anderson, M. L., Vernon, S. W., Chubak, J., Williams, D. R. (1997). Commentary on monitoring socioeconomic status.
Fuller, S., & Green, B. B. (2014). Time to screening in the systems of Public Health Reports, 112, 492–​494.
support to increase colorectal cancer screening trial. Cancer Epidemiol Wilson, C. L., Stratton, K., Leisenring, W. L., Oeffinger, K. C., Nathan, P.
Biomarkers Prev, 23(8), 1683–​1688. doi:10.1158/​1055-​9965.EPI-​14-​0270 C., Wasilewski-​Masker, K.,  .  .  .  Ness, K. K. (2014). Decline in physi-
Wang, F., Luo, L., & McLafferty, S. (2010). Healthcare access, socioeconomic cal activity level in the Childhood Cancer Survivor Study cohort. Cancer
factors and late-​stage cancer diagnosis: an exploratory spatial analysis and Epidemiol Biomarkers Prev, 23(8), 1619–​1627. doi:10.1158/​1055-​9965.
public policy implication. Int J Public Pol, 5(2–​3), 237–​258. doi:10.1504/​ EPI-​14-​0213
IJPP.2010.030606 Woloshin, S., Schwartz, L. M., Tosteson, A. N., Chang, C. H., Wright, B.,
Wang, L., Wilson, S. E., Stewart, D. B., & Hollenbeak, C. S. (2011). Marital Plohman, J., & Fisher, E. S. (1997). Perceived adequacy of tangible social
status and colon cancer outcomes in US Surveillance, Epidemiology support and health outcomes in patients with coronary artery disease. J
and End Results registries:  does marriage affect cancer survival by Gen Intern Med, 12(10), 613–​618.
gender and stage? Cancer Epidemiol, 35(5), 417–​ 422. doi:10.1016/​ Yanovski, S. Z., & Yanovski, J. A. (2011). Obesity prevalence in the United
j.canep.2011.02.004 States:  up, down, or sideways? N Engl J Med, 364(11), 987–​989.
Wang, N., Cao, F., Liu, F., Jia, Y., Wang, J., Bao, C., . . . Cheng, Y. (2015). The doi:10.1056/​NEJMp1009229
effect of socioeconomic status on health-​care delay and treatment of esoph- Yen, I. H., & Moss, N. (1999). Unbundling education: a critical discussion of
ageal cancer. J Transl Med, 13, 241. doi:10.1186/​s12967-​015-​0579-​9 what education confers and how it lowers risk for disease and death. Ann
Wei, F., Miglioretti, D. L., Connelly, M. T., Andrade, S. E., Newton, K. M., N Y Acad Sci, 896, 350–​351.
Hartsfield, C. L.,  .  .  .  Buist, D. S. (2005). Changes in women’s use of Yin, D., Morris, C., Allen, M., Cress, R., Bates, J., & Liu, L. (2010). Does
hormones after the Women’s Health Initiative estrogen and progestin trial socioeconomic disparity in cancer incidence vary across racial/​ eth-
by race, education, and income. J Natl Cancer Inst Monogr, 35, 106–​112. nic groups? Cancer Causes Control, 21(10), 1721–​1730. doi:10.1007/​
doi:10.1093/​jncimonographs/​lgi047 s10552-​010-​9601-​y
Wei, R., DeVilbiss, F. T., & Liu, W. (2015). Genetic polymorphism, telomere Yost, K., Perkins, C., Cohen, R., Morris, C., & Wright, W. (2001).
biology and non-​small lung cancer risk. J Genet Genomics, 42(10), 549–​ Socioeconomic status and breast cancer incidence in California for differ-
561. doi:10.1016/​j.jgg.2015.08.005 ent race/​ethnic groups. Cancer Causes Control, 12(8), 703–​711.
Weischer, M., Nordestgaard, B. G., Cawthon, R. M., Freiberg, J. J., Tybjaerg-​ Zane, L., & Jeang, K. T. (2014). HTLV-​1 and leukemogenesis:  virus-​cell
Hansen, A., & Bojesen, S. E. (2013). Short telomere length, cancer sur- interactions in the development of adult T-​cell leukemia. Recent Results
vival, and cancer risk in 47102 individuals. J Natl Cancer Inst, 105(7), Cancer Res, 193, 191–​210. doi:10.1007/​978-​3-​642-​38965-​8_​11
459–​468. doi:10.1093/​jnci/​djt016 Zhang, C., Chen, X., Li, L., Zhou, Y., Wang, C., & Hou, S. (2015). The asso-
Weston, T. L., Aronson, K. J., Siemiatycki, J., Howe, G. R., & Nadon, L. ciation between telomere length and cancer prognosis:  evidence from
(2000). Cancer mortality among males in relation to exposures assessed a meta-​analysis. PLoS One, 10(7), e0133174. doi:10.1371/​ journal.
through a job-​exposure matrix. Int J Occup Environ Health, 6(3), 194–​ pone.0133174
202. doi:10.1179/​oeh.2000.6.3.194 Zibbell, J. E., Iqbal, K., Patel, R. C., Suryaprasad, A., Sanders, K. J., Moore-​
Wiencke, J. K., & Kelsey, K. T. (2002). Teen smoking, field cancerization, and a Moravian, L., . . . ; Centers for Disease Control and Prevention (CDC).
“critical period” hypothesis for lung cancer susceptibility. Environ Health (2015). Increases in hepatitis C virus infection related to injection drug
Perspect, 110(6), 555–​558. use among persons aged ≤30  years—​ Kentucky, Tennessee, Virginia,
Wigle, D. T., Turner, M. C., & Krewski, D. (2009). A systematic review and meta-​ and West Virginia, 2006–​2012. MMWR Morb Mortal Wkly Rep, 64(17),
analysis of childhood leukemia and parental occupational pesticide exposure. 453–​458.
Environ Health Perspect, 117(10), 1505–​1513. doi:10.1289/​ehp.0900582 Ziol-​Guest, K. M., Duncan, G. J., & Kalil, A. (2009). Early childhood pov-
Wilkinson, R. G. (1996). Unhealthy societies:  the afflictions of inequality. erty and adult body mass index. Am J Public Health, 99(3), 527–​532.
London: Routledge. doi:10.2105/​ajph.2007.130575
 169

10 The Economic Burden of Cancer in the United States

K. ROBIN YABROFF, GERY P. GUY JR., MATTHEW P. BANEGAS,


AND DONATUS U. EKWUEME

OVERVIEW are considered, expenditures in 2020 are projected to be even greater


(Mariotto et al., 2011). Thus, estimating and projecting the economic
With an aging and growing population and improved early detection burden of cancer, including healthcare expenditures and productivity
and survival following cancer diagnosis in the United States, the preva- losses for patients and their families, are increasingly important issues
lence of cancer survivorship is expected to increase in the future. Based for healthcare policymakers, healthcare systems, physicians, employ-
only on population trends, national expenditures for cancer care are ers, and society overall. In this chapter, we describe measures of the
projected to increase as well, from $124.6 billion in 2010 to $157.8 economic burden of cancer and approaches for estimating economic
billion in 2020 (both in 2010 US dollars). When trends toward greater burden, and we present recent national estimates. We also describe
intensity of healthcare service use and more expensive treatments are publicly available data sources and discuss methodologic issues
considered, expenditures are projected to be even greater. Thus, esti- with measuring economic burden, and identify key areas for future
mating and projecting the economic burden of cancer, including health- research.
care expenditures and productivity losses for patients and their families,
are increasingly important issues for healthcare policymakers, health-
care systems, physicians, employers, and society overall. In this chap- WHY MEASURE THE ECONOMIC BURDEN
ter, we describe measures of the economic burden of cancer, including OF CANCER?
(1) direct costs, resulting from the use of resources for medical care for
cancer; (2) indirect costs, resulting from the loss of economic resources Measurement of the economic burden of cancer is important at many
and opportunities associated with morbidity and mortality due to cancer levels for medical and non-​medical resource allocation, reimbursement
and its treatment; and (3) psychosocial or intangible costs, such as pain decisions, and the evaluation of specific programs throughout the course
and suffering. Consistent with the intensity of treatment for initial care, of cancer care, from early detection and diagnosis to treatment, survivor-
recurrence, and end-​of-​life care, costs are highest in the initial period ship, and end-​of-​life care. Descriptive studies of the various components
following diagnosis and, among patients who die from their disease, at of the economic burden of cancer also serve as an initial step in elucidat-
the end of life, following a U-​shaped curve. We describe how this tem- ing relevant domains in cost-​effectiveness analysis of cancer prevention
poral cost pattern influences differences in per-​person and aggregate and control interventions and identifying priorities and resource needs in
cost estimates for cancers with excellent prognosis and longer survival the planning of cancer prevention and control strategies and programs.
(e.g., breast and prostate) and cancers with poorer prognosis (e.g., pan-
creas and lung) and we present recent per-​person and national esti-
mates of direct and indirect costs overall and by cancer site. Finally, we HOW IS THE ECONOMIC BURDEN
describe publicly available data sources and methodologic issues with OF CANCER MEASURED?
measuring economic burden and identify key areas for future research.
Illness and disease create an economic burden for patients, family and
friends, employers, the healthcare system, and society. Cancer and its
INTRODUCTION treatment may result in pain and suffering, morbidity and reduced qual-
ity of life, financial losses for the patient and family, and in some cases,
In 2014, an estimated 14.5 million Americans were alive with a his- premature mortality. The economic burden of cancer also includes the
tory of cancer (American Cancer Society, 2016; Howlander et  al., loss of economic resources and opportunities from the perspective of
2016). The absolute number of cancer survivors is expected to be sub- employers and society more broadly.
stantially larger in the future because cancer incidence increases with The economic burden of disease is measured by cost, the monetary
age, and the US population is both aging and growing (de Moor et al., valuation of resources used to treat disease and the loss of economic
2013; Mariotto et al., 2011). In addition, advances in screening, detec- opportunities related to disease occurrence and treatment. Three cat-
tion, and treatment are associated with improved survival following egories of cost domains are typically identified: (1) direct costs, result-
cancer diagnosis. These developments will also result in increased ing from the use of resources for medical care for cancer as well as
cancer prevalence. It is projected that the number of cancer survi- care for lasting and late effects of cancer; (2) indirect costs, resulting
vors will increase to 19 million by 2024 (American Cancer Society, from the loss of economic resources and opportunities associated with
2016). This increasing number of cancer survivors will receive morbidity and mortality due to cancer and its treatment; and (3) psy-
medical care throughout the trajectory of their cancer experiences, chosocial (intangible) costs, such as pain and suffering. Examples of
including short-​and long-​term effects of disease and its treatment. As these categories of costs are listed in Table 10–1 and are described in
shown in Figure 10–1, based only on population aging and growth, greater detail in the following subsections.
national expenditures for cancer care are projected to increase from
$124.6 billion in 2010 to $157.8 billion by 2020 (both in 2010 US
dollars), a 27% increase (Mariotto et al., 2011). When recent trends
Direct Costs
toward greater intensity of healthcare service use (Warren et al., 2008;
Dinan et  al., 2010; Conti et  al., 2014) and increasing costs of can- Direct costs include the monetary value of resources used for medi-
cer care (Bach 2009; Conti et  al., 2014; Dinan et  al., 2010; Elkin cal care in the early detection, diagnosis, and treatment of disease, as
and Bach, 2010; Howard et  al., 2010; Schrag, 2004; Tangka et  al., well as for rehabilitation, surveillance, and end-​of-​life care. The dis-
2010; Warren et al., 2008; Woodward et al., 2007; Wong et al., 2009) tinguishing characteristic of a direct cost is that it represents resource

169
170

170 PART II:  THE MAGNITUDE OF CANCER

Breast
Colorectal
Lymphoma
Lung
Prostate
Leukemia
Ovary
Brain
Bladder
Kidney
Head_Neck
Uterus
Melanoma
Pancreas
Stomach
Cervix
Esophagus

0 5 10 15 20 25
Billion US$

Expenditures in 2010 Additional Expenditures in 2020

Figure 10–1.  Projected increase in national expenditures in 2020 by cancer site (in billion US$): Based on population changes only. Source: Mariotto AB,
Yabroff KR, Shao Y, Feuer EJ, Brown ML. 2011. Projections of the costs of cancer care in the United States: 2010–​2020. J Natl Cancer Inst, 103, 117–​128.

utilization associated with a direct monetary payment. These payments costs. The majority of studies of the economic burden of cancer report
are usually recorded in the financial records of a healthcare insurance direct medical costs (Yabroff et al., 2012), in part, because these data
system, healthcare providers, or an individual household. Such costs are more readily available. Studies of direct medical costs are typically
include medical resources provided under public or private insurance based on billing system data from insurers, hospital discharge data,
systems, as well as individual out-​of-​pocket expenditures. Direct costs and large nationally representative surveys.
for medical care include hospitalizations, emergency room care, out- In addition to costs for the provision of medical care, the direct
patient care, nursing home and home healthcare, and the services of non-​medical costs are those costs borne by patients, caregivers, and
primary care physicians and specialists as well as other health practi- families. These costs include those associated with transportation to
tioners (Table 10–1). Cancer treatments, such as chemotherapy, immu- and from health providers, child and dependent care costs, and costs
notherapy, and radiation therapy, supportive agents, rehabilitation of relocating temporarily or permanently to improve access to specific
counseling, and other rehabilitation costs, are also included in direct treatments or facilities. Illness can force a family to incur expenses as
part of caring for a cancer patient, including those for routine house-
hold tasks, special diets or clothing, items for rehabilitation, alterations
Table 10–1.  Cost Domains of property, and vocational, social, and family counseling services.
Expenditures for retraining or re-​education and interest lost on with-
DIRECT COSTS Medical Hospitalizations drawal of savings or interest charges on funds borrowed to pay illness-​
Physician visits related expenses are also considered direct non-​medical costs. These
Home healthcare data are not systematically collected as part of billing systems, and as
Hospice care
a result, few studies report direct non-​medical costs.
Chemotherapy
Radiation
Rehabilitation
Supportive agents Indirect Costs
Non-​Medical Transportation to and from medical care Indirect costs are the time and economic output lost or forgone due
Housekeeping services to disease and its treatment and any late and lasting effects of treat-
Costs of relocating ment on the usual activities of patients, family, and friends, including
Alterations to property employment, housekeeping, volunteer activities, and leisure. These
INDIRECT Morbidity Time lost from work/​lost productivity costs are not reflected by direct monetary transactions but do reflect
COSTS Time spent seeking medical care the use of economic resources in response to disease and its treatment
Caregiver time or changes in caregiver that could be used for other purposes in the absence of disease. Indirect
productivity costs are measured as the value of losses of economic output due to
Mortality Economic productivity lost due to morbidity or premature mortality from disease. Cancer survivors may
premature death not be able to fully participate in work or other usual activities because
of morbidity associated with the illness. Family members and others
INTANGIBLE/​PSYCHOSOCIAL Pain
COSTS Suffering
may spend time caring for the patient rather than pursuing other activi-
Grief ties, may make unwanted job changes, or may miss opportunities for
promotion and education.
 17

The Economic Burden of Cancer in the United States 171


Estimated morbidity costs of cancer can include losses measured $18,000
by the value of forgone earnings and the imputed value of lost house- $16,000
keeping services (measured by wages that would have to be paid to
replace those services). A related type of indirect cost is the time $14,000
the patient and/​or family spend receiving medical care, known as $12,000 36 Month Survival
patient or caregiver “time costs.” Longitudinal morbidity data and
$10,000
patient time data are not collected systematically as part of bill- 48 Month Survival
ing systems, and as a result, few studies have reported these costs. $8,000
60 Month Survival
However, several studies have reported productivity losses associ- $6,000
ated with morbidity costs in a given year using nationally represen- 72 Month Survival
tative cross-​sectional data (Ekwueme et al., 2014; Guy et al., 2013, $4,000
2014). Patient time costs have been estimated from medical service $2,000 84 Month Survival
frequencies (e.g., hospitalizations, physician visits, chemotherapy)
$0
combined with time estimates of medical service duration and values 0 12 24 36 48 60 72 84 96
96 Month Survival
for that time (e.g., median wages) (Yabroff et al., 2007, 2014).
Month After Diagnosis
Mortality costs are the present value of future output lost because of
premature death due to cancer. One of the main components of mor-
tality costs is the remaining life expectancy in the absence of cancer Figure 10–2.  Monthly Medicare payments for colorectal cancer patients
death—​the person-​years of life lost (PYLL). The other main compo- by length of survival. Source: Yabroff KR, Warren JL, Schrag D, Mariotto
nent of mortality costs is the time value of the PYLL. All measures of M, Meekins A, Topor M, Brown ML. Comparison of approaches for esti-
indirect costs require a method for estimating the value of time associ- mating incidence costs of care for colorectal cancer patients. Med Care.
ated with morbidity, patient time, or the PYLL due to early death from 2009 Jul;47(7 Suppl 1):S56-​63. doi: 10.1097/​MLR.0b013e3181a4f482.
cancer. Two general approaches are commonly used to value time: the PubMed PMID: 19536010.
human-​capital approach and the willingness-​to-​pay approach. In the
human-​capital approach, sex-​and age-​specific earnings are combined
with expected productivity trends and time lost to estimate unreal- intensity of treatment for initial care, recurrence, and end-​of-​life
ized lifetime earnings. This approach explicitly values the time of care, cancer costs are highest in the initial period following diagnosis
individuals with higher earning potential as greater than those with and, among patients who die from their disease, at the end of life
less earning potential, which is one of its limitations. The willingness-​ (Brown et al., 2002; Fireman et al., 1997; Riley et al., 1995; Taplin
to-​pay approach, in contrast, incorporates both lost productivity and et al., 1996; Yabroff et al., 2007, 2008b, 2009). Costs are lowest in
the intrinsic value of life by estimating the amount an average indi- the period between the initial and end-​of-​life periods, following a U-​
vidual would be willing to pay for an additional year of life. Because shaped curve. As shown in Figure 10–2, this U-​shaped medical care
incidence and mortality rates for most cancer sites are highest in the cost pattern is consistent across cohorts of cancer patients with very
elderly, a population that is less likely to be in the workforce than their different survival times following diagnosis (Yabroff et  al., 2009).
younger counterparts, comparing results from these two approaches The width and height of this U-​shaped cost curve vary by cancer site,
for valuing time is particularly relevant for estimating the burden of stage at diagnosis, and patient age (Brown et al., 2002; Riley et al.,
cancer (Ramsey, 2008). 1995; Taplin et al., 1996; Yabroff et al., 2007, 2008b, 2009). All of
Few studies have systematically measured indirect costs associated these factors affect summary measures of costs and inform the meth-
with cancer care. However, studies that have attempted to measure ods used to estimate these costs.
components of these costs have reported that these frequently unmea-
sured costs may be substantial (Bradley et al., 2008; Ekwueme et al., INCIDENCE AND PREVALENCE CANCER COSTS
2008; Li et al., 2010; Yabroff et al., 2007, 2008a).
Direct, indirect, and intangible cancer costs are typically reported
starting at diagnosis for a group of cancer patients defined by clini-
Psychosocial or Intangible Costs cal characteristics (incidence costs), or for all cancer survivors alive
Disease may also impact the quality of life of the patient, family, and in a specific year (prevalence costs). An incidence cost estimate in
friends in ways that are not reflected in the categories of direct or indi- 2015 will include only those patients diagnosed within this year, and
rect costs. These are referred to as psychosocial or intangible costs. costs will begin accumulating at the high-​cost diagnosis period for all
As the result of a cancer diagnosis, its treatment, and late or lasting patients. Incidence cost estimates can range from periods of less than
effects of treatment, patients may suffer from reduced sexual function- 1 year to a lifetime (for patients diagnosed in 2015 in this example).
ing, disfigurement, disability, pain, or fears of life-​threatening disease. A  prevalence cost estimate in 2015 will include all cancer patients
Patients and their families may make changes in life plans, which can alive at any point during the year, but will only include the portion
induce anxiety, reduce self-​esteem and feelings of well-​being, or cre- of the cost trajectory that occurs during 2015. Prevalence cost esti-
ate resentment and family conflict. The combination of financial hard- mates are typically reported as annual estimates or for a particular
ship and psychosocial problems can be especially devastating. For the year. Importantly, the high-​and low-​cost portions of the U-​shaped
purposes of cost-​effectiveness analysis, psychosocial cost is conceptu- curve included in the prevalence cost estimate will vary by cancer
alized as a quality of life outcome and is measured in terms of decre- site. For example, because long-​term survival following breast and
ments of quality-​adjusted life-​years or in terms of utility. Developing prostate cancer diagnosis is high, in any given year, the majority of
appropriate concepts and measures of the quality of life ramifications breast and prostate cancer survivors will be in the low-​cost bottom of
of cancer and cancer treatment is currently an active area of cancer the U-​shaped curve. In contrast, because survival following pancre-
research. atic cancer diagnosis is generally poor, in any given year the majority
of pancreatic cancer survivors will be in the high-​cost period follow-
ing diagnosis or at the end-​of-​life, the arms of the “U.” As with other
WHAT ARE THE TEMPORAL PATTERNS OF measures of disease burden, per capita incidence and prevalence cost
THE ECONOMIC BURDEN OF CANCER? estimates in a given year will be relatively similar when survival is
short, because the vast majority of individuals will have high costs
When measured longitudinally starting from cancer diagnosis, the in either arm of the “U,” with few individuals with low costs at the
monthly patterns of medical care use and the associated costs of bottom of the “U” (e.g., pancreatic cancer). For cancers with long sur-
cancer change over time. As noted earlier, and consistent with the vival (e.g., breast and prostate cancers), per-​capita prevalence costs
172

172 PART II:  THE MAGNITUDE OF CANCER


in a given year will be significantly lower than per-​capita incidence per person for breast and prostate cancer in women and men, respec-
costs in that year, because the prevalence cost estimate will be pre- tively, to more than $25,000 per person for colorectal, lung, and pan-
dominantly composed of survivors at the bottom of the “U,” where creas cancers (in 2004 dollars; Yabroff et  al., 2008b; Figures 10–3a
costs are lowest. Both incidence and prevalence cost measures can and 10–3b). Differences in 1-​year incidence costs by cancer site reflect
be useful for resource allocation and policy and program planning. differences in the distribution of stage of disease at diagnosis, survival
Incidence costs are commonly used in cost-​effectiveness models, for following diagnosis, and treatment intensity, especially for hospital-
decisions about specific therapies or understanding patient and treat- izations, by cancer site. The majority of breast and prostate cancer
ment trajectories, whereas prevalence costs are most commonly used patients are diagnosed with early stage disease, have long survival,
in understanding the overall impact of disease on local, federal, or and do not require extensive hospitalization, whereas the majority
health plan budgets. of lung and pancreatic cancer patients are diagnosed with advanced
disease and have relatively short survival, often requiring extensive
hospitalization.
APPROACHES FOR ESTIMATING CANCER COSTS These per-​person incidence estimates can be applied to national
estimates of the number of newly diagnosed elderly cancer patients
Several approaches are commonly used for estimating incidence and to generate a national incidence cost estimate for a specific year.
prevalence cancer costs. As with other measures of disease burden, Even though the per-​ person 1-​ year estimates of the incidence
cohort studies, both retrospective and prospective, can be used for esti- costs for pancreas cancer are among the highest ($29,814 in men
mating incidence costs. Cross-​sectional data can be used for estimating and $30,357 in women), approximately 21,000 elderly patients
prevalence costs, although in some situations, such as the use of billing were diagnosed with pancreatic cancer in 2004, yielding a national
records (without linkage to cancer registry data), the prevalence esti- 1-​year incidence cost estimate of $642 million. On the other hand,
mate will reflect “treated prevalence,” rather than prevalence cost mea- the per-​person costs associated with female breast cancer and pros-
sured from all cancer survivors alive in a specific year. Only patients tate cancer are relatively low ($11,748 and $10,626), but more than
receiving treatment in a specific year can be identified from claims for 77,000 elderly female breast cancer patients and 118,000 elderly
cancer treatment. Other approaches for estimating incidence and prev- prostate cancer patients were diagnosed in 2004, yielding much
alence costs include modeling or microsimulation. This approach is larger national 1-​year incidence cost estimates of $905 million and
typically based on well-​defined hypothetical cohorts of cancer patients $1,254 million, respectively.
in a natural history of disease model where the probability of medical
events (e.g., hospitalizations for a side effect of treatment) is combined Prevalence Cost Estimates
with cost estimates for each of the medical events (e.g., average hospi- Several studies have reported prevalence costs for cancer survivors
talization cost for patients with that side effect of treatment). (Bernard et  al., 2011; Guy et  al., 2013, 2014; Howard et  al., 2004;
Another approach, the phase of care approach, uses the U-​shaped Short et al., 2011). Mean annual direct medical costs have been con-
cost pattern described in Figure 10–2 to divide healthcare services, sistently reported to be greater for cancer survivors than for individu-
costs, and observation time for cancer patients into clinically rel- als without a history of cancer (Bernard et al., 2011; Guy et al., 2013,
evant periods or phases in relation to the date of diagnosis and the 2014; Howard et al., 2004; Short et al., 2011). In addition, as shown
date of death. Three phases, corresponding to the arms of the “U” in Figure 10–4, mean annual prevalence costs for the recently diag-
and the bottom of the “U” (i.e., initial, last year of life, and continu- nosed are greater than for the previously diagnosed and individuals
ing phases, respectively), have been commonly used. Phase-​specific without a cancer history in those aged 18–​64 years and 65+ years (Guy
estimates can then be used as an input to estimate either incidence or et al., 2013). Mean annual prevalence costs are higher for the older age
prevalence costs. Phase-​specific cost estimates can be combined with group than in the younger age group, reflecting greater comorbidity
survival probabilities following diagnosis to yield modeled incidence burden and healthcare utilization in those ages 65+, for individuals
costs (Yabroff et al., 2009) or with phase-​specific cancer prevalence with and without cancer.
estimates in a given year to produce prevalence costs for that year The distribution of costs by type of medical care service also var-
(Mariotto et  al., 2011). The phase-​of-​care approach is a more effi- ies between cancer survivors and individuals without a cancer his-
cient use of data than a cohort approach because patients can con- tory (Guy et al., 2013). Among those aged 18–​64 years, ambulatory
tribute to multiple phases and this can lead to more robust estimates, care and inpatient care are the most common services for recently
particularly for less common cancer sites. diagnosed cancer survivors (46.2% and 40.5%, respectively), with
prescription medications contributing only a small portion of costs
(8.1%). For previously diagnosed cancer survivors aged 18–​64, pre-
WHAT ARE SOME RECENT ESTIMATES OF scription medications contribute a larger portion of costs (18.9%);
THE COSTS OF CANCER CARE? and for individuals without a cancer history, the contribution of ambu-
latory care (33.9%), inpatient care (27.4%), and prescription medica-
In this section, we present recent estimates of the direct medical costs tions (22.6%) are more similar. The distribution of costs by service
of cancer and report both incidence and prevalence cost estimates. We type is similar for the older age group, with ambulatory and inpa-
also present estimates of indirect costs of cancer, including annual pro- tient care the most common service category in recently diagnosed
ductivity losses due to employment disability, missed workdays, and cancer survivors (41.2% and 41.9%, respectively); prescriptions are a
lost household productivity, and mortality costs associated with early larger portion for the previously diagnosed (18.9%), and ambulatory
death from cancer. care (25.9%), inpatient care (34.3%), and prescription medications
(24.3%) have similar distributions for those without a cancer history.
Estimates of Direct Medical Costs The distribution of direct medical costs by payer type is quite
different between individuals aged 18–​ 64  years and 65+ years
A number of studies have estimated the direct medical costs associ- (Figures 10–5a and 10–5b). In the younger group with and without
ated with cancer (Bernard et  al., 2011; Ekwueme et  al., 2011; Guy cancer, private health insurance is the predominant payer for medi-
et al., 2013, 2014; Howard et al., 2004; Mariotto et al., 2011; Short cal care, whereas in the older group, the Medicare program is the
et al., 2011). These studies include estimates of incidence costs among predominant payer.
the newly diagnosed and annual prevalence costs among all cancer A study using the phase-​of-​care approach to estimate national prev-
patients alive in a given year. alence costs in the United States reported costs associated with cancer
of $124.6 billion dollars (in 2010 dollars) (Mariotto et al., 2011). The
Incidence Cost Estimates highest costs were for female breast ($16.5 billion), colorectal ($14.1
Costs associated with cancer for newly diagnosed elderly patients in billion), lymphoma ($12.1 billion), lung ($12.1 billion) and pros-
the first year following diagnosis range from a little more than $10,000 tate ($11.9 billion) cancers, reflecting the absolute number of cancer
 173

The Economic Burden of Cancer in the United States 173


(a)
Bladder $14,313

Breast $11,748

Colorectal $25,903

Corpus uteri $16,485

Head and Neck $22,633

Kidney $23,836

Liver $25,398

Lung $26,764

Pancreas $30,557

$0 $5,000 $10,000 $15,000 $20,000 $25,000 $30,000 $35,000

(b)
Bladder $13,466

Colorectal $25,711

Head and Neck $21,052

Kidney $23,920

Figure 10–3.  (a) Direct medical costs in year


Liver $24,086
following diagnosis, by cancer site in elderly
female patients. (b) Direct medical costs in
Lung $26,449 year following diagnosis, by cancer site in
elderly male patients. Note: Calculations
made using the linked SEER-​Medicare data,
Pancreas $29,814 1999–​2003. Estimates are the net estimates
based on controls. Source:  Yabroff KR,
Lamont EB, Mariotto A, Warren JL, Meekins
Prostate $10,626 A, Topor M, Brown ML. 2008. Cost of care
for elderly cancer patients in the United
$0 $5,000 $10,000 $15,000 $20,000 $25,000 $30,000 $35,000 States. J Natl Cancer Inst, 100, 630–​641.

survivors by phases of care and phase-​specific cost estimates by cancer ($3,719 and $4,033 in 2011 US dollars, respectively) than for males
site. A larger proportion of prevalence costs occurred in the continu- and females without a cancer history ($2,260 and $2,703 in 2011 US
ing phase of care for cancers with longer survival, such as breast and dollars, respectively) (Ekwueme et al., 2014). For all groups, employ-
prostate cancers, than for those with short survival, such as lung and ment disability is the largest component of productivity loss, followed
pancreas cancers. by missed workdays and lost household productivity.
A number of studies have estimated mortality costs and the pro-
ductivity losses associated with premature death due to cancer
Indirect Costs (Bradley et al., 2008; Ekwueme et al., 2008; Li et al., 2010; Yabroff
To date, the majority of studies estimating indirect costs associated et al., 2008a). Table 10–2 lists the number of cancer deaths in 2005,
with cancer have reported prevalence estimates. These studies have person-​years of life lost (PYLL), and present value of lifetime earn-
mainly reported productivity losses from employment disability ings by cancer site, using the human capital approach for valuing the
(being unable to work due to health), missed worked days among the PYLLs (Bradley et  al., 2008). Because this approach uses gender-​
employed, and lost household productivity (Guy et  al., 2013, 2014) and age-​specific earnings to value the years with unrealized earnings,
or mortality costs associated with premature death due to cancer cancers that affect individuals with lower earnings potential will have
(Bradley et al., 2008; Ekwueme et al., 2008; Li et al., 2010; Yabroff less productivity loss than cancers that affect individuals with higher
et  al., 2008a). As shown in Figures 10–6a and 10–6b, total per-​per- earnings potential. For example, the majority of men who die from
son productivity losses are highest among recently diagnosed cancer prostate cancer are ages 65 and older, an age when most are retired
survivors aged 18–​64 ($4,694 in 2010 US dollars) and 65+ ($6,133 in and would not otherwise be earning wages from work in the future.
2010 US dollars), compared with the previously diagnosed, and indi- Although the number of prostate cancer deaths is higher than the
viduals without a cancer history. As shown in Figure 10–7, per-​person number of pancreas cancer deaths, the PYLL is lower, and the pres-
productivity losses are higher for male and female cancer survivors ent value of lifetime earnings is lower ($3.3 billion vs. $6.6 billion).
174

Recently diagnosed $17,170

Ages 18–64
Previously diagnosed $6,485

No cancer history $3,611

Recently diagnosed $23,441

Ages 65+ Previously diagnosed $12,357

No cancer history $8,724

$0 $5,000 $10,000 $15,000 $20,000 $25,000

Figure 10–4.  Mean annual direct medical costs in cancer survivors and individuals without a cancer history. Notes: Calculations using the Medical Expenditure
Panel Survey–​Household Component, 2008–​2010. Estimates are predicted margins from a generalized linear regression model with a gamma distribution
and a log link controlling for age, sex, race/​ethnicity, and number of comorbid conditions. All monetary amounts are in 2010 dollars. Source: Guy GP Jr,
Ekwueme DU, Yabroff KR, et al. 2013. The economic burden of cancer survivorship among adults in the United States. J Clin Oncol, 31(30), 3749–3757. PMCID:
PMC3795887.

(a) Recently diagnosed cancer survivors No cancer history

7.3% 6.1% 9.8%


16.9%
17.1% Out-of-pocket 10.6%

Private health insurance


Medicare 8.2%
2.8%
Medicaid
Other
66.7% 54.6%

$17,170 (95% Cl: $13,433, $20,907) $3,611 (95% Cl: $3,486, $3,736)

(b) Recently diagnosed cancer survivors No cancer history

1.1% 10% 6.9% 8.7%


13.9%
4.4%
11.3%
Out-of-pocket
Private health insurance 13.1%
Medicare
Medicaid
Other
70.7%
59.9%

$23,441 (95% Cl: $18,367, $28,514) $8,724 (95% Cl: $8,302, $9,147)

Figure 10–5.  (a) Annual medical expenditures by payer type, for individuals aged 18–​64 years. (b) Annual medical expenditures by payer type, for individu-
als aged 65+ years. Notes: Calculations using the Medical Expenditure Panel Survey–​Household Component, 2008–​2010. Estimates are predicted margins
from a generalized linear regression model with a gamma distribution and a log link controlling for age, sex, race/​ethnicity, and number of comorbid con-
ditions. All monetary amounts are in 2010 dollars. Source: Guy GP Jr, Ekwueme DU, Yabroff KR, Dowling EC, Li C, Rodriguez J, DeMoor J, Virgo K.
2013. The economic burden of cancer survivorship in the United States. J Clin Oncol, 31(30), 3749–​3757.
 175

The Economic Burden of Cancer in the United States 175


(a) $7,000 $5,000 Employment disability
Employment disability
Missed work days
$6,000 Missed work days
Lost household productivity
Lost household productivity $4,000
$5,000
US dollars

$4,000 $3,000

US dollars
$2,664 $2,961
$3,000 $2,831
$2,000
$2,833 $2,109
$2,000
$1,862
$1,644 $1,585
$1,000 $1,000
$447 $686
$321 $597
$386 $313 $134 $933 $267
$0 $386 $291
$0 $201 $131
Recently Previously No cancer
diagnosed diagnosed history Cancer No cancer Cancer No cancer
cancer survivors cancer survivors history history history history
Women Men

Employment disability
(b) $7,000 Figure 10–7. Annual lost productivity among cancer survivors and indi-
Missed work days
viduals without a cancer history. Notes: Calculations using the Medical
$6,000 Lost household productivity Expenditure Panel Survey–​Household Component, 2008–​ 2011. All mon-
etary amounts are in 2010 dollars. Source: Ekwueme DU, Yabroff KR, Guy
$5,000 GP Jr, et  al. 2014. Medical costs and produc­tivity losses of cancer survivors:
United States, 2008–2011. MMWR Morb Mortal Wkly Rep, 63 (23), 505–510.
US dollars

$4,000 $4,350 PMID: 24918485.

$3,000 $4,485 of cancer are listed in Table 10–3, including the Healthcare Costs and
$3,777 Utilization Program (H​CUP) discharge data, the linked Surveillance
$2,000 Epidemiology and End Results cancer registry–​ Medicare insurance
claims data (SEER-​Medicare), the Medical Expenditure Panel Survey
$1,000 $1,109 (MEPS), and the Medicare Current Beneficiary Survey (MCBS).
$428 $331 Features of these data sources are described in the following, including
$676 $382 $301
$0 population coverage, patient characteristics, information about cancer
Recently Previously No cancer diagnosis, and the availability of information about the burden of cancer,
diagnosed diagnosed history including direct, indirect, and intangible costs. We also discuss whether
cancer survivors cancer survivors these data sources can be used to estimate incidence and prevalence
costs associated with cancer.
Figure 10–6. (a) Annual lost productivity among cancer survivors and
individuals without a cancer history, aged 18–​64 years. (b) Annual lost
productivity among cancer survivors and individuals without a cancer his- Table 10–2.  Lost Productivity Due to Cancer Deaths in the US Among
tory, aged 65+ years. Notes: Calculations using the Medical Expenditure Adults Aged 20 Years and Older by Cancer Site (2005)
Panel Survey–​House­hold Component, 2008–​2010. All monetary amounts are
Number Person-​Years Present Value of
in 2010 dollars. Source: Guy GP Jr, Ekwueme DU, Yabroff KR, Dowling of Deaths of Life Lost Lifetime Earnings
EC, Li C, Rodriguez J, DeMoor J, Virgo K. 2013. The economic burden of in 2005 (in thousands) (in billions of dollars)
cancer survivorship in the United States. J Clin Oncol, 31(30), 3749–​3757.
Lung and bronchus 166,755 2569.8 $36.131
Other approaches to value the PYLL, such as the willingness-​to-​pay Female breast 44,546 874.0 $12.096
approach, incorporate both lost productivity and the intrinsic value Colon and rectum 61,240 861.4 $10.652
of life and place an equivalent value on all years of life. Estimates Pancreas 32,054 472.5 $6.610
of the value of life lost for prostate cancer are substantially higher Brain and other nervous 13,607 314.2 $5.743
system
when the willingness-​to-​pay approach is used to value the PYLL
Leukemia 22,945 390.1 $5.722
(Yabroff et al., 2008a) than a human capital approach because older
Non-​Hodgkin’s 23,850 362.3 $5.511
men dying of prostate cancer have lower earnings potential. lymphoma
The direct medical cost and indirect cost estimates presented in this Liver and intrahepatic 14,502 242.8 $4.420
section were calculated from a variety of existing data sources. In the bile duct
next section, we describe common data sources for estimating eco- Kidney and renal pelvis 12,819 206.2 $3.424
nomic burden of cancer in the United States in more detail. Head and neck 10,830 181.4 $3.413
Prostate 33,642 300.5 $3.301
WHAT DATA SOURCES ARE USED FOR ESTIMATING Stomach 13,351 201.3 $3.225
THE ECONOMIC BURDEN OF CANCER? Melanoma of the skin 8062 155.1 $3.194
Ovary 15,253 274.5 $2.824
In this section, we discuss different types of data commonly used for Cervix uteri 4338 114.9 $1.827
estimating the economic burden of cancer in the United States, includ- Urinary bladder 13,275 150.5 $1.826
ing hospital discharge data, health insurance claims, and household sur- Hodgkin lymphoma 1414 36.4 $0.833
veys (Lund et al., 2009; Yabroff et al., 2011). Selected publicly available
data sources that are commonly used to estimate aspects of the burden Source: Bradley et al., 2008.
176

176 PART II:  THE MAGNITUDE OF CANCER


Table 10–3.  Characteristics of Selected Publicly Available Data Sources/​Research Resources in the US for Estimating Economic Burden of Cancer

SEER-​Medicare MEPS H​CUP MCBS

Nationally Representative
SEER Tumor Registries Linked In-​Person Survey with Inpatient Discharge Data National Survey Linked
Description to Medicare Claims Provider Data Collection from Sampled Hospitals to Medicare Claims

DATA CHARACTERISTICS
National or nationally representative Geographically defined √ √ √
Individual-​level longitudinal data √ 5 panels over 2 years √
Approximate number of cancer > 1,000,000 < 2000 > 1,000,000 < 2000
survivors in 2015
Duration of information Medicare eligibility through 2 years Hospital admission through
death discharge
Health insurance type Medicare fee-​for-​service only All payers All payers All payers
PATIENT INFORMATION
Age distribution Aged 65+ or disabled (any age) Aged 18+ All ages Aged 65+ or disabled
(any age)
Information about patients without cancer In cancer registry regions √ √ √
CANCER INFORMATION
Cancer diagnosis Registry, procedure or diagnosis Self-​report, procedure or Procedure or diagnosis codes Self-​report, procedure or
codes diagnosis codes diagnosis codes
Stage at diagnosis √
Treatment √ √ Inpatient hospital only √
TYPE OF COST ESTIMATE
Incidence √
Prevalence in a specific year √ √ √ √
COST DOMAINS
Direct Medical Cost Components
Hospital √ √ √ √
Physician and other outpatient services √ √ √
Outpatient pharmacy √* √ √*
Out of pocket √ √
Indirect Cost Components
Productivity loss (e.g., days lost √
from work)
Patient time √
Caregiver time
Intangible costs √ √

*Data on Medicare Part D prescription drug services are available starting in 2006. Before 2006, drugs administered parenterally, and their administration was covered by Medicare Part B,
as were Prodrugs, the oral drug equivalent of drugs administered parenterally.
SEER-​Medicare = Surveillance Epidemiology and End Results–​Medicare; MEPS = Medical Expenditure Panel Survey; H-​CUP = Healthcare Costs and Utilization Program;
MCBS = Medicare Current Beneficiary Survey.

Healthcare Costs and Utilization Project (HCUP) the NIS. The NIS has been mainly used to estimate the use and costs of
surgery among hospitalized cancer survivors (Newton and Ewer, 2010;
The HCUP is a family of national and state healthcare databases and Seifeldin and Hantsch, 1999). Because information about the date of
tools sponsored by the Agency for Healthcare Quality and Research, and cancer diagnosis is not available, the estimates from the NIS are prev-
includes the National (Nationwide) Inpatient Sample (NIS) and State alence estimates. Although the number of hospital days can be used to
Inpatient Databases (SID), Nationwide Emergency Department Sample identify patient time in the hospital, these data have rarely been used for
and State Emergency Department Databases, and State Ambulatory Care estimating any types of costs other than direct medical costs.
Databases. The NIS and SID include medical care use and associated
costs for individuals of all ages with all types of health insurance, but
Linked SEER-​Medicare Data
for only one aspect of medical care, inpatient hospitalizations (Agency
for Healthcare Research and Quality, 2016). These NIS data have been The linked SEER-​Medicare data are made available by the National
available every year since 1988. The unit of observation is the hospitali- Cancer Institute and contain longitudinal information about medical
zation rather than the individual patient, so multiple hospitalizations for care and associated payments received before, during, and after cancer
the same person are unique observations in the NIS and cannot be linked diagnosis for Medicare beneficiaries (Warren et al., 2002). Information
at the individual level to provide longitudinal information about care. about cancer diagnoses is available for the years that each geographi-
In some states, linkage for multiple hospitalizations and across HCUP cally defined registry has been part of the SEER program, starting in
databases is available, although these data have not been commonly 1973. Medicare claims since 1991 are available for two cohorts of ben-
used to estimate cancer burden. Although millions of cancer survivors eficiaries included in the SEER-​Medicare data: cancer survivors and a
can be identified in the NIS by diagnosis and procedure codes, informa- sample of Medicare beneficiaries residing in the SEER areas who do
tion about date of diagnosis, stage, additional cancers, pre-​hospitaliza- not have cancer. Because more than 95% of individuals over the age
tion comorbidity, and other factors that influence patient eligibility for of 65 in the United States are enrolled in the Medicare program, and
specific treatments is not available. Further, information about cancer SEER registries currently cover approximately 26% of the US popu-
survivors treated outside the hospital inpatient setting is unavailable in lation, these data contain information about cancer care for a large
 17

The Economic Burden of Cancer in the United States 177


portion of the elderly in the United States. More than 3 million cancer 2014). Because date of diagnosis is not available, cost estimates from
survivors can be identified from SEER-​Medicare with detailed infor- the MEPS are prevalence cost estimates.
mation about cancer stage, grade, and histology for each diagnosis. In 2011, the MEPS Experiences with Cancer survey was a self-​
Identification of incident cancer patients and patients with multiple administered questionnaire of adult cancer survivors, containing ques-
cancers are additional strengths of the linkage of health insurance pro- tions on financial hardship related to cancer, its treatment, and lasting
gram data with cancer registry data. and late effects of treatment, such as having to borrow money or go
As with other health insurance program data (also known as admin- into debt, being unable to cover medical care costs, worry about pay-
istrative data), Medicare claims do not contain any information about ing medical bills, and other financial sacrifices (Yabroff et al., 2012). It
individuals without health insurance or the utilization and costs of spe- also included a number of questions related to productivity for cancer
cific services covered by other insurance programs or those services paid survivors and their caregivers. An updated MEPS Experiences with
entirely out of pocket. Medicare payments represent approximately 51%–​ Cancer survey of adult cancer survivors is being fielded in 2016.
65% of all direct medical care costs in the elderly (Crystal et al., 2000;
Guy et  al., 2013), with the remaining costs covered by other insurers
Medicare Current Beneficiary Survey
or by out-​of-​pocket payments. These linked SEER-​Medicare data have
been used extensively to estimate the incidence and prevalence costs of The Medicare Current Beneficiary Survey (MCBS) is a longitudinal,
medical care associated with cancer in the elderly (Mariotto et al., 2011; nationally representative survey of Medicare beneficiaries in the United
Warren et al., 2008). Additionally, the costs associated with patient time States conducted by the Centers for Medicare & Medicaid Services.
have been estimated from patients’ medical service frequencies (e.g., The MCBS collects information about expenditures and sources of
hospital length of stay, physician visits, chemotherapy), combined with payment for all services used by Medicare beneficiaries, including
average service-​specific time estimates (Yabroff et al., 2007). Notably, copayments, deductibles, and non-​covered services. In addition, demo-
the linked SEER-​Medicare data cannot be used to estimate costs associ- graphics, health status, access to medical care, and physical functioning
ated with cancer care in the working-​age population under the age of 65 are collected about individuals with all types of health insurance. The
(who are only eligible for Medicare in limited situations), and informa- survey is conducted over a 4-​year period, with multiple interviews a
tion about employment and health status is unavailable. year. The MCBS Cost and Use files link Medicare claims from the pop-
ulation with fee-​for-​service coverage to the survey. Cancer survivors
can be identified based on their response to the question “Have you ever
Medical Expenditure Panel Survey been told by a doctor or other health professional that you had cancer or
The Medical Expenditure Panel Survey (MEPS) is an annual nation- a malignancy of any kind?” or with claims algorithms using diagnosis
ally representative household survey conducted by the Agency for codes. The survey can be used to supplement the claims data to explore
Healthcare Research and Quality (AHRQ) (Cohen et al., 2009). The out-​of-​pocket spending or use of strategies to reduce the costs of medi-
MEPS was first fielded in 1996 and consists of three surveys:  the cations (Davidoff et al., 2013; Nekhlyudov et al., 2011). Cost estimates
Household Component (HC), the Medical Provider Component, and from the MCBS are prevalence cost estimates.
the Insurance Component. The MEPS-​ HC collects demographic, Nationally representative surveys such as the MEPS or the MCBS
health status, access to medical care, employment, and healthcare uti- are limited by small numbers of patients who are newly diagnosed or
lization and expenditure data for adults of all ages, with all insurance diagnosed with cancers associated with short survival (e.g., lung) or
types (Table 10–3). The MEPS also collects these data for individuals low incidence (e.g., testicular). Details about cancer diagnosis, includ-
without health insurance. ing stage, clinical characteristics and other prognostic information,
The MEPS-​HC uses an overlapping panel design, in which each and treatment are generally not available. Because questions are not
panel consists of individuals who are interviewed in person five times specific to burden of illness related to cancer (except in the MEPS
(or rounds) over approximately 2.5  years. Data from two panels are Experiences with Cancer survey), these MEPS and MCBS data have
combined to produce estimates for each calendar year, and each panel been previously used to assess employment, insurance coverage, access
separately produces longitudinal estimates. Through panel 11 in 2007, to care, and health status in cancer survivors compared with individu-
cancer survivors were only identified in the MEPS if they received care als without a history of cancer (Davidoff et al., 2013; Dowling et al.,
for cancer, or missed work or school or spent a day in bed because of 2010; Guy et al., 2013; Nekhlyudov et al., 2011; Yabroff et al., 2004).
cancer, during the survey period (“treated prevalence”). Only very small As described earlier and as illustrated in Table 10.3, commonly used
numbers of cancer survivors were identified in a given year during this publicly available data sources have different strengths and limitations
period, although multiple years of the MEPS have been combined to for estimating the burden of cancer in the United States. No single
assess economic burden (Howard et al., 2004; Short et al., 2011). data source contains comprehensive longitudinal information for large
Starting in panel 12 and in 2008, a new question was added for numbers of cancer survivors about multiple healthcare services across
adults aged 18 years and older about whether a doctor or other health insurance type (including the uninsured) in the elderly or in younger
professional ever told the person that he or she had cancer or a malig- populations. Further, few data sources contain information about indi-
nancy of any kind. Rather than only identifying the subset of cancer rect costs, including patient and caregiver time costs and lost produc-
survivors undergoing cancer treatment at the time of the survey, this tivity due to morbidity (Lund et al., 2009).
change now results in the identification of all individuals with a history
of cancer, consistent with definitions of cancer survivorship used to
estimate cancer prevalence in the United States (de Moor et al., 2013; WHAT ARE KEY FACTORS RELATED TO THE
Mariotto et al., 2011). Notably, the question about a cancer diagnosis MEASUREMENT OF HEALTHCARE COSTS?
is asked for all adults in the family, making the MEPS one of the only
data sources to identify cancer history for the family, and these data There are several key factors that are critical to the measurement of the
can be used to assess the effects of cancer on a spouse (Litzelman and economic burden of cancer, including the perspective of the economic
Yabroff, 2015). Details about cancer diagnosis, including stage, tumor analyses, consideration of current and future costs, and measurement
characteristics, and other prognostic information, are unavailable. of cancer-​attributable costs. These issues are defined and discussed in
Several studies have combined multiple years of the MEPS to describe following sections.
healthcare utilization and expenditures in cancer survivors receiving
cancer care compared with individuals without cancer (Bernard et al.,
The Frame of Reference or Perspective
2011; Finkelstein et al., 2009; Guy et al., 2013, 2014; Howard et al.,
2004; Short et al., 2011). These MEPS data have also been used to esti- The frame of reference of an economic study is the viewpoint from
mate the costs associated with patient time from medical service fre- which the analyses are conducted. As shown in Table 10–4, examples
quencies (e.g., hospital length of stay, physician visits, chemotherapy), of perspectives are those of the patient and family, employers, health-
combined with average service-​specific time estimates (Yabroff et al., care insurers or payer, including federal and state programs such as
178

178 PART II:  THE MAGNITUDE OF CANCER


Table 10–4.  Perspectives and Relevant Cost Categories

Perspective

Cost Category Patient and Family Employer Health Insurer/​Payer Society

DIRECT COSTS Medical √ Premiums, copayments √ Premiums √ Payments for services √


and other out-​of-​pocket
Non-​Medical √ Transportation, housekeeping √
INDIRECT COSTS Morbidity √ Reduced productivity, time √ Reduced productivity, presentism, √
seeking medical care absenteeism, disability payments,
replacement costs
Mortality √ Lost productivity √
INTANGIBLE/​PSYCHOSOCIAL COSTS √ √

Medicare and Medicaid, and society. Depending on the perspective is necessary to use a price index specific to this sector of the economy.
of the analysis, the types of costs included, sources of data, and study Direct medical cost estimates using multiple years of data (e.g., 2008–​
design will vary. While the societal perspective is preferred for pur- 2011) are typically inflated using a price index to the latest year of data
poses of economic analysis for general policy analysis, for specific and reported for that year (e.g., in 2011 US dollars). The healthcare
purposes, such as the impact of new healthcare legislation, it is often component of the Consumer Price Index (CPI) is often used for this
useful to define a frame of reference that is narrower than all of society purpose. This index, however, refers only to the components of health-
(Gold et al., 1996). For example, while the societal perspective may be care expenditures paid for directly by consumers. Other prices indices
the relevant perspective for policy analysis involving approval of an are used in other settings, such as the Personal Health Care Expenditure
innovative cancer control intervention, the perspective of the Medicare Price Index or indices that are designed to reflect healthcare price infla-
program or a health maintenance organization may be relevant for tion as experienced by the Medicare program (Agency for Healthcare
assessing the financial impact of such a decision on resource require- Research and Quality, 2015a). Note that adjustments for inflation and
ments required to fund these operations. the discounting of future costs and benefits are distinct and unrelated
Not all social costs will be recognized as costs at various organiza- issues.
tional levels. Time lost from paid work for unpaid caregiving by the rel-
atives of the cancer patient is a cost to society in terms of lost economic
productivity and a cost to the family in terms of lost household income, Charges, Reimbursements, Expenditures, and Costs
but it might represent a savings to the Medicare program in terms of a The economic concept of cost relates to the use of specific resources
shorter length of hospital stay or less provision of formal home-​care ser- in an efficient manner. Direct observation of resource use is rarely
vices. As noted previously, the time spent by cancer patients and family available, however. Reflections of cost are available, such as the
members related to cancer and its treatment is rarely fully accounted for amount billed, charged, or reimbursed for the provision of healthcare
among existing studies because these data are not collected systemati- services. The amount billed or charged may not reflect the actual cost
cally. Excluding patient time costs from analyses comparing alternative associated with providing healthcare services, however, and may have
interventions (e.g., cost-​effectiveness analyses) may result in favoring little relationship to underlying costs (Finkler, 1982). For example,
interventions with large patient-​time cost burdens (Russell, 2009). some hospitals or other providers may negotiate with health plans
for reduced payments for specific patients. Here, the actual “cost”
of the service may be unrelated to the amount charged to any of
Discounting the purchasers. When using claims data, researchers frequently use
When costs are distributed over time and the purpose of analysis is payments to reflect costs, rather than charges (Yabroff et al., 2013).
to assess these costs relative to an initial investment, such as a can- Charges may also be converted into costs using “cost-​to-​charge”
cer screening program to promote early detection, economists use an ratios (Agency for Healthcare Research and Quality, 2015b).
adjustment known as discounting. The purpose of discounting is to
adjust the value of costs and benefits (savings) incurred in the future
to their present market value because the same resources, if avail- Measurement of Costs Attributable to Cancer
able and invested today, would yield a return if placed in a productive There are two general approaches to estimating the costs attributable
activity. In the United States, the most commonly used value in cur- to cancer:  estimating costs for specific events assumed to be cancer-​
rent health economics literature is 3% (in real terms—​that is, adjusted related, and comparing costs of all care for cancer patients to other
for inflation). Other industrialized countries have used different dis- individuals without a cancer history. In the first approach, research-
count rates, and there is some discussion about what the appropri- ers classify specific costs as being due to cancer or not. This approach
ate discount rate should be from a social viewpoint. The choice of a can be used in observational studies and is more frequently used in
discount rate reflects a value judgment: a low discount rate indicates a microsimulation modeling, where treatment scenarios are developed as
long time horizon, and a high discount rate indicates a short time hori- a series of probabilistic events for a well-​defined, hypothetical popula-
zon. All else being equal, the use of a high discount rate would tend tion of cancer patients. The cost of medical services and procedures
to favor investment in a treatment program where benefits would be defined by the scenario can then be estimated using a cost estimate from
realized more rapidly over investment in a prevention program where another data source. This approach may underestimate the frequency of
ongoing costs are incurred well in advance of the realization of any services that cancer patients receive for seemingly unrelated care.
benefits. In the second approach, researchers measure attributable costs of
cancer care by comparing the costs of all care for cancer patients or
survivors to those of non-​cancer patients or patients without a cancer
Adjusting for Inflation history. The difference is a net cost. The comparison can match cancer
It is often useful to express the cost of cancer in current-​year dollars. patients and individuals without a cancer history by relevant character-
Because the rate of inflation for the price of healthcare services has istics, such as age, gender, geographic region, and comorbidity status,
consistently been higher than the rate for the economy as a whole, it or can control for these characteristics with multivariable analyses.
 179

The Economic Burden of Cancer in the United States 179


This approach is particularly advantageous when using administra- well as psychological aspects of financial hardship, including stress,
tive data because it minimizes reliance on diagnostic and procedure anxiety, and worry about their financial situation (Meneses et  al.,
codes for identifying services as cancer-​related or not. In some situ- 2012; Yabroff et al., 2016). Among working-​age cancer survivors, one
ations, such as the estimation of costs associated with lung cancer, in four report ever having any material financial hardship (i.e., medical
this approach may overstate services as attributable to cancer that are debt, bankruptcy, trouble paying medical bills) and one in three report
more common among lung cancer patients. For example, emphysema psychological financial hardship (Yabroff et al., 2016).
is more common among smokers who are also more likely to develop Informal caregiving may also influence employment opportunities for
lung cancer. Costs associated with emphysema care would also be caregivers, potentially limiting a caregiver’s ability to hold a full-​time
included in any net cost of lung cancer. position or resulting in higher rates of absenteeism, particularly when
the patient travels long distances for specialized care. Alternatively,
maintaining health insurance coverage may lead cancer survivors or
WHAT ARE EMERGING ISSUES IN THE ECONOMIC their employed family caregivers to work longer hours (Bradley et al.,
BURDEN OF CANCER IN THE UNITED STATES? 2002; Hollenbeak et al., 2012) or to continue working (Bradley et al.,
2007)  and delay retirement. Evaluation of financial hardship and
In this section, we discuss several emerging issues related to the eco- employment and health insurance trajectories in cancer survivors and
nomic burden of cancer in the United States, including precision medi- their families will be an important area for additional research, particu-
cine and financial hardship associated with cancer care. larly in relation to changes in health insurance status that may occur
with ongoing implementation of the Affordable Care Act.
As the prevalence of cancer survivorship increases in the United
Precision Medicine States, the economic burden of cancer will increase as well. Thus,
estimating and projecting the economic burden of cancer, including
The average cost of newly developed cancer treatments, primarily tar- healthcare expenditures and productivity losses for patients and their
geted therapies used in precision medicine, has risen dramatically over families, are increasingly important issues for healthcare policymak-
the past decades (Bach, 2009; Schrag, 2004), and many have price tags of ers, healthcare systems, physicians, employers, and society overall.
more than $10,000 per month. Advances in the identification of genetic Efforts to ensure the comprehensiveness and quality of data resources
mutations associated with treatment response allow the tailoring of treat- for estimating the economic burden of cancer, particularly indirect
ment to individual patients. Limiting the use of expensive therapies to costs, are ongoing. Emerging areas for future research include evalu-
patients with genetic profiles associated with treatment response has the ating the use, effectiveness, and consequences of targeted therapies,
potential to reduce the program costs of these treatments. For example, and financial hardship for patients and their families. This work
colorectal cancer patients carrying KRAS mutations do not respond to will inform efforts by healthcare policymakers, healthcare systems,
treatment with cetuximab or panitumumab (Amado et al., 2008), and by and employers to improve the cancer survivorship experience in the
restricting the use of cetuximab to patients without the KRAS mutations, United States.
the incremental cost-​effectiveness ratio is more favorable (Mittmann
et al., 2009). However, because the goal of precision medicine is to iden- References
tify patients most likely to respond to targeted therapies, all potentially Agency for Healthcare Research and Quality. 2015a. Using the appropriate
eligible patients must be tested, including those for whom treatment price indices for analyses of health care expenditures or income across
will not ultimately be indicated. Finally, the increase in median survival multiple years. Available from: http://​meps.ahrq.gov/​about_​meps/​Price_​
attributed to treatment with many of these targeted therapies, even among Index.shtml. Accessed October 18, 2015.
those most likely to respond, is typically only a few additional months Agency for Healthcare Research and Quality. 2015b. Cost-​to-​charge ratio files.
(Fojo et  al., 2014). Currently, little is known about the utilization and Available from:  https://​www.hcup-​us.ahrq.gov/​db/​state/​costtocharge.jsp.
effectiveness of precision medicine therapies in community practice. Accessed October 9, 2015.
Important areas for future research include estimating and projecting Agency for Healthcare Research and Quality. 2016. Healthcare Cost and Utili­
zation Project (HCUP). Available from: http://​www.ahrq.gov/​research/​data/​
their uptake, patterns of care, and cost; the appropriate use of genetic tests
hcup/​index.html. Accessed April 15, 2016.
to personalize therapy; and the impact on patient morbidity and survival. Amado RG, Wolf M, Peeters M, et al. 2008. Wild-​type KRAS is required for
panitumumab efficacy in patients with metastatic colorectal cancer. J Clin
Oncol, 26(10), 1626–​1634. PMID: 18040272.
Financial Hardship Associated with Cancer American Cancer Society. 2016. Cancer treatment and survivorship facts &
­figures  2014–​2015. Available from:  http://​www.cancer.org/​acs/​groups/​
The use of precision medicine highlights an increasingly important
content/​@research/​documents/​document/​acspc-​042801.pdf Accessed
issue—​out-​of-​pocket costs and financial hardship to patients and their April 19, 2016.
families. Because most health insurance plans require some form Bach PB. 2009. Limits on Medicare’s ability to control rising spending on can-
of cost-​sharing for oral drug therapy (20% co-​insurance is typical), cer drugs. N Engl J Med, 360(6), 626–​633. PMID: 9176475.
patients and their families with health insurance may face bills of Banegas MP, Guy GP Jr, de Moor JS, et al. 2016. For working-​age cancer sur-
tens of thousands of dollars for a full course of treatment. The costs vivors, medical debt and bankruptcy create financial hardships. Health
of cancer care will present additional challenges to cancer survivors Aff, 35(1), 54–​61. PMID: 26733701.
without health insurance because they may responsible for the entire Bernard DS, Farr SL, Fang Z. 2011. National estimates of out-​of-​pocket health
treatment cost. Patients may delay treatment or may fail to seek care care expenditure burdens among nonelderly adults with cancer: 2001 to
2008. J Clin Oncol, 29(20), 2821–​2826. PMCID: PMC3139395.
because of high patient cost-​sharing or financial hardship (Kent et al.,
Bradley CJ, Bednarek HL, Neumark D. 2002. Breast cancer and women’s labor
2013; Weaver et al., 2010). Treatment adherence is also poorer among supply. Health Serv Res, 37(5), 1309–​1328. PMCID: PMC1464031.
cancer survivors with financial hardship or higher out-​of-​pocket costs Bradley CJ, Neumark D, Luo Z, Bednarek HL. 2007. Employment-​contingent
(Dusetzina et al., 2014; Neugut et al., 2011). A national study estimated health insurance, illness, and labor supply of women: evidence from married
that more than 2 million cancer survivors in the United States did not women with breast cancer. Health Econ, 16(7), 19–​37. PMID: 17177273.
receive needed medical services because of cost (Weaver et al., 2010). Bradley CJ, Yabroff KR, Dahman B, Feuer EJ, Mariotto A, Brown ML. 2008.
Because health insurance in the United States is predominantly Productivity costs of cancer mortality in the United States: 2000–​2020.
employment-​ based in the working-​ age population, its relationship J Natl Cancer Inst, 100(24), 1763–​1770. PMCID: PMC2720777.
with the economic burden of cancer survivorship is complex. A can- Brown ML, Riley GF, Schussler N, Etzioni RD. 2002. Estimating health care
costs related to cancer treatment from SEER-​Medicare data. Med Care,
cer diagnosis may limit employment opportunities, which in turn may
40(8 Suppl), IV-​104–​117. PMID: 12187175.
lead to a loss of health insurance and fewer resources for paying for Cohen JW, Cohen SB, Banthin JS. 2009. The Medical Expenditure Panel
medical care, further magnifying financial hardships associated with Survey:  a national information resource to support healthcare cost
cancer. Cancer survivors are at risk of medical debt and bankruptcy research and inform policy and practice. Med Care, 47(7 Suppl 1),
(Banegas et  al., 2016; Ramsey et  al., 2013; Yabroff et  al., 2016), as S44–​50. PMID: 19536015.
180

180 PART II:  THE MAGNITUDE OF CANCER


Conti RM, Fein AJ, Bhatta SS. 2014. National trends in spending on and use Li C, Ekwueme DU, Rim SH, Tangka FK. 2010. Years of potential life lost and
of oral oncologics, first quarter 2006 through third quarter. Health Aff, productivity losses from male urogential cancer deaths:  United States,
33(10), 1721–​1727. PMCID: PMC4594844. 2004. Urology, 76(3), 528–​535. PMID: 20573389.
Crystal S, Johnson RW, Harman J, Sambamoorthi U, Kumar R. 2000. Out-​of-​ Litzelman K, Yabroff KR. 2015. How are spousal depressed mood, distress,
pocket health care costs among older Americans. J Gerontol B Psychol and quality of life associated with risk of depressed mood in cancer sur-
Sci Soc Sci, 55(1), S51–​S62. PMID: 10728130. vivors? Longitudinal findings from a national sample. Cancer Epidemiol
Davidoff AJ, Erten M, Shaffer T, et al. 2013. Out-​of-​pocket health care expen- Biomarkers Prev, 24(6), 969–​977. PMCID: PMC4453017.
diture burden for Medicare beneficiaries with cancer. Cancer, 119(6), Lund JL, Yabroff KR, Ibuka Y, et al. 2009. Inventory of data sources for esti-
1257–​1265. PMID: 23225522. mating health care costs in the United States. Med Care, 47 (7 Suppl 1),
de Moor JS, Mariotto AB, Parry C, et al. 2013. Cancer survivors in the United S127–​S142. PMCID: PMC309738.
States:  prevalence across the survivorship trajectory and implica- Mariotto AB, Yabroff KR, Shao Y, Feuer EJ, Brown ML. 2011. Projections of
tions for care. Cancer Epidemiol Biomarkers Prev, 22(4), 561–​ 570. the cost of cancer care in the United States: 2010–​2020. J Natl Cancer
PMCID: PMC3654837. Inst, 103(2), 117–​128. PMCID: PMC3107566.
Dinan MA, Curtis LH, Hammill BG, et al. 2010. Changes in the use and costs Meneses K, Azuero A, Hassey L, McNees P, Pisu M. 2012. Does economic bur-
of diagnostic imaging among Medicare beneficiaries with cancer, 1999–​ den influence quality of life in breast cancer survivors? Gynecol Oncol,
2006. JAMA, 303(16), 1625–​1631. PMID: 20424253. 124(3), 437–​443. PMCID: PMC3278545.
Dowling E, Yabroff KR, Mariotto AB, McNeel T, Zeruto C, Buckman D. Mittmann N, Au H-​J, Tu D, et al. 2009. Prospective cost-​effectiveness analy-
2010. Burden of illness in adult survivors of childhood cancers: findings sis of cetuximab in metastatic colorectal cancer: economic evaluation of
from a population-​based national sample. Cancer, 116(15), 3712–​3721. National Cancer Institute of Canada Clinical Trials Group CO.17 trial.
PMCID: PMC2913863. J Natl Cancer Inst, 101(17), 1182–​1192. PMID: 19666851.
Dusetzina SB, Winn AN, Abel GA, Huskamp HA, Keating NL. 2014. Cost shar- Nekhlyudov L, Madden J, Graves AJ, Zhang F, Soumerai SB, Ross-​Degnan D.
ing and adherence to tyrosine kinase inhibitors for patients with chronic 2011. Cost-​related medication nonadherence and cost-​ saving strategies
myeloid leukemia. J Clin Oncol, 32(4), 306–​311. PMID: 24366936. used by elderly Medicare cancer survivors. J Cancer Surviv, 5(4), 395–​404.
Ekwueme DU, Chesson HW, Zhang KB, Balamurugan A. 2008. Years of poten- PMCID: PMC3767465.
tial life lost and productivity costs because of cancer mortality and for Neugut AI, Subar M, Wilde ET, et al. 2011. Association between prescription
specific cancer sites where human papillomavirus may be a risk factor for co-​payment amount and compliance with adjuvant hormonal therapy in
carcinogenisis: United States, 2003. Cancer, 113(10 Suppl), 2936–​2945. women with early-​stage breast cancer. J Clin Oncol, 29(18), 2534–​2542.
PMID: 18980277. PMCID: PMC3138633.
Ekwueme DU, Guy GP Jr, Li C, Rim SH, Parelkar P, Chen SC. 2011. The Newton AN, Ewer SR. 2010. Inpatient cancer treatment: an analysis of finan-
health burden and economic costs of cutaneous melanoma mortality by cial and nonfinancial performance measures by hospital-​ownership type.
race/​ethnicity-​United States, 2000 to 2006. J Am Acad Dermatol, 65(5 J Health Care Finance, 37(2), 56–​80. PMID: 21294439.
Suppl 1), S133–​S143. PMID: 22018062. Ramsey SD. 2008. How should we value lives lost to cancer? J Natl Cancer,
Ekwueme DU, Yabroff KR, Guy GP Jr, et al. 2014. Medical costs and produc- 100(24), 1742–​1743. PMID: 19066279.
tivity losses of cancer survivors: United States, 2008–​2011. MMWR Morb Ramsey S, Blough D, Kirchhoff A, et al. 2013. Washington state cancer patients
Mortal Wkly Rep, 63 (23), 505–​510. PMID: 24918485. found to be at greater risk for bankruptcy than people without a cancer
Elkin EB, Bach PB. 2010. Cancer’s next frontier: addressing high and increas- diagnosis. Health Aff, 32(6), 1143–​1152. PMCID: PMC4240626.
ing costs. JAMA, 303(11), 1086–​1087. PMCID: PMC3647336. Riley GF, Potosky AL, Lubitz JD, Kessler LG. 1995. Medicare payments from
Finkelstein EA, Tangka FK, Trogdon JG, Sabatino SA, Richardson LC. 2009. diagnosis to death for elderly cancer patients by stage and diagnosis. Med
The personal financial burden of cancer for the working age population. Care, 33(8), 828–​841. PMID: 7637404.
Am J Manag Care, 15(11), 801–​806. PMID: 19895184. Russell L. 2009. Completing costs: patients’ time. Med Care, 47 (7 Suppl 1),
Finkler SA. 1982. The distinction between cost and charges. Ann Intern Med, S89–​S93. PMID: 19536025.
96(1), 102–​109. PMID: 7053682. Sabatino SA, Coates RJ, Uhler RJ, Alley LG, Pollack LA. 2006. Health insur-
Fireman BH, Quesenberry CP, Somkin CP, et al. 1997. Cost of care for cancer ance coverage and cost barriers to needed medical care among US adult
in a health maintenance organization. Health Care Financ Rev, 18(4), 51–​ cancer survivors age <65  years. Cancer, 106(11), 2466–​2475. PMID:
76. PMCID: PMC4194474. 16639732.
Fojo T, Mailankody S, Lo A. 2014. Unintended consequences of expensive can- Schrag D. 2004. The price tag on progress: chemotherapy for colorectal cancer.
cer therapeutics: the pursuit of marginal indications and a me-​too mental- N Engl J Med, 351(4), 317–​319. PMID: 15269308.
ity that stifles innovation and creativity: the John Conley Lecture. JAMA Seifeldin R, Hantsch JJ. 1999. The economic burden associated with
Otolaryngol Head Neck Surg, 140(12), 1225–​1236. PMID: 25068501. colon cancer in the United States. Clin Ther, 21(8), 1370–​ 1379.
Gold MR, Siegel JE, Russell LB, Weinstein MC (Eds.). 1996. Cost-​Effectiveness PMID: 10485508.
in Health and Medicine. New York: Oxford University Press. Short PF, Moran JR, Punekar R. 2011. Medical expenditures of adult cancer
Guy GP Jr, Ekwueme DU, Yabroff KR, et al. 2013. The economic burden of survivors aged <65 years in the United States. Cancer, 117(12), 2791–​
cancer survivorship among adults in the United States. J Clin Oncol, 2800. PMCID: PMC4124459.
31(30), 3749–​3757. PMCID: PMC3795887. Tangka FK, Trogdon JG, Richardson LC, Howard D, Sabatino SA, Finkelstein
Guy GP Jr, Yabroff KR, Ekwueme DU, et al. 2014. Estimating the health and EA. 2010. Cancer treatment cost in the United States. Cancer, 116(14),
economic burden of cancer among those diagnosed as adolescents and 3477–​3484. PMID: 20564103.
young adults. Health Aff, 33(6), 1024–​1031. PMCID: PMC4582764. Taplin SH, Barlow W, Urban N et al. 1995. Stage, age, comorbidity, and direct
Hollenbeak CS, Short PF, Moran J. 2011. The implications of cancer sur- costs of colon, prostate, and breast cancer care. J Natl Cancer Inst, 87(6),
vivorship for spousal employment. J Cancer Surviv, 5(3), 226–​ 234. 417–​426. PMID: 7861461.
PMCID: PMC4176691. Warren JL, Klabunde CN, Schrag D, Bach PB, Riley GF. 2002. Overview of the
Howard DH, Molinari N-​A, Thorpe KE. 2004. National estimates of medical SEER-​Medicare data: content, research applications, and generalizabilty
costs incurred by nonelderly cancer patients. Cancer, 100(5), 883–​891. to the United States elderly population. Med Care, 40 (8 Suppl). IV-​3–​18.
PMID: 14983481. PMID: 12187163.
Howard DH, Kauh J, Lipscomb J. 2010. The value of new chemotherapeutic Warren JL, Yabroff KR, Meekins A, Topor M, Lamont EB, Brown ML. 2008.
agents for metastatic colorectal cancer. Arch Intern Med, 170(6), 537–​542. Evaluation of trends in the cost of initial cancer treatment. J Natl Cancer
PMID: 20233802. Inst, 100(12), 888–​897. PMCID: PMC3298963.
Howlander N, Noone AM, Krapcho M, et  al. 2016. SEER Cancer Statistics Weaver KE, Rowland JH, Bellizzi KM, Aziz NM. 2010. Forgoing medical care
Review, 1975–​2013. Bethesda, MD: National Cancer Institute. http://​seer. because of cost:  assessing disparities in healthcare access among can-
cancer.gov/​csr/​1975_​2013/​, based on November 2015 SEER data submis- cer survivors living in the United States. Cancer, 116(14), 3493–​3504.
sion, posted to the SEER website, April 2016. PMCID: PMC3018838.
Kent EE, Forsythe LP, Yabroff KR, et al. 2013. Are survivors who report can- Wong Y-​N, Meropol NJ, Speier W, Sargent D, Goldberg RM, Beck JR. 2009.
cer-​related financial problems more likely to forgo or delay medical care? Cost implication of new treatments for advanced colorectal cancer.
Cancer, 119(20), 3710–​3717. PMCID: PMC4552354. Cancer, 115(10), 2081–​2091. PMCID: PMC2875773.
 18

The Economic Burden of Cancer in the United States 181


Woodward RM, Brown ML, Stewart ST, Cronin KA, Cutler DM. 2007. colo­rectal cancer patients. Med Care, 47 (7 Suppl 1), S56–​63. PMID:
The value of medical interventions for lung cancer in the elderly: 19536010.
results from SEER-​ CMHSF. Cancer, 110(11), 2511–​ 2518. PMID: Yabroff KR, Lund J, Kepka D, Mariotto AB. 2011. Economic burden of can-
17955504. cer in the US: estimates, projections, future research. Cancer Epidemiol
Yabroff KR, Lawrence WF, Clauser S, Davis WW, Brown ML. 2004. Burden Biomarkers Prev, 20(10), 2006–​2014. PMCID: PMC3191884.
of illness in cancer survivors: findings from a population-​based national Yabroff KR, Dowling E, Rodriguez J, et  al. 2012. The Medical Expenditure
sample. J Natl Cancer Inst, 96(17), 1322–​1330. PMID: 15339970. Panel Survey (MEPS) Experiences with Cancer survivorship supplement.
Yabroff KR, Davis WW, Lamont EB, et al. 2007. Patient time costs associated J Cancer Surviv, 6(4), 407–​419. PMID: 23011572.
with cancer care. J Natl Cancer Inst, 99(1), 14–​23. PMID: 17202109. Yabroff KR, Borowski L, Lipscomb J. 2013. Economic studies in colorectal
Yabroff KR, Bradley CJ, Mariotto AB, Brown ML, Feuer EJ. 2008a cancer: challenges in measuring and comparing costs. J Natl Cancer Inst,
Estimates and projections of value of life lost from cancer deaths in 2013(46), 62–​78. PMCID: PMC3859288.
the United States. J Natl Cancer Inst, 100(24), 1755–​1762. PMCID: Yabroff KR, Guy GP, Ekwueme DU, et  al. 2014. Annual patient time costs
PMC2720776. associated with medical care among cancer survivors in the United States.
Yabroff KR, Lamont EB, Mariotto A, et  al. 2008b. Cost of care for elderly Med Care, 52(7), 594–​601. PMCID: PMC4058637.
cancer patients in the United States. J Natl Cancer Inst, 100(9), 630–​641. Yabroff KR, Dowling EC, Guy GP Jr, et al. 2016. Financial hardship associ-
PMID: 18445825. ated with cancer in the United States: findings from a population-​based
Yabroff KR, Warren JL, Schrag D, Meekins A, Topor M, Brown ML. 2009. sample of adult cancer survivors. J Clin Oncol, 4(3), 259–​67. PMID:
Comparison of approaches for estimating incidence costs of care for 26644532.
182
 183

III
THE CAUSES OF CANCER
 185

11 Tobacco

MICHAEL J. THUN AND NEAL D. FREEDMAN

OVERVIEW 2004; US Department of Health and Human Services, 2014). A major


contributor to this transformation was the rise of manufactured ciga-
Tobacco is the leading preventable cause of cancer and other non-​com- rettes, first in high-​and later in middle-​and low-​income countries dur-
municable diseases worldwide. The International Agency for Research ing the twentieth century. The term “tobacco epidemic” refers to the
on Cancer (IARC) and the US Surgeon General designate over 20 can- global dissemination and disease consequences of manufactured ciga-
cer sites or subsites as causally related to active cigarette smoking, rettes and cigarette-​like products (Lopez et  al., 1994); these greatly
including lung, oral cavity, nasal cavity, and accessory sinuses, naso-​, amplify the endemic problems from other forms of tobacco use.
oro-​, and hypopharynx, larynx, esophagus (squamous cell carcinoma Patterns of tobacco use have changed markedly over time. The
and adenocarcinoma), stomach, pancreas, colorectum, liver, kidney uptake of manufactured cigarettes began among affluent and middle-​
(adeno-​and urothelial carcinomas), ureter, urinary bladder, uterine class men in high-​income countries in the early twentieth century
cervix, ovary (mucinous), and acute myeloid leukemia. Even this list (Doll, 1998). Successive generations of first men and then women
may be incomplete, as it does not include sites for which the evidence initiated cigarette smoking at progressively earlier ages and smoked
is still considered limited, such as advanced prostate cancer and breast more intensively, creating strong birth cohort patterns in both smok-
cancer. In addition to cigarettes, all other forms of smoked and conven- ing and lung cancer, as discussed further later in the chapter. By the
tional smokeless tobacco products, as well as involuntary exposure to mid-​twentieth century, manufactured cigarettes had largely displaced
tobacco smoke, cause cancer. the use of other tobacco products in English-​speaking countries, as
Trends in tobacco use and tobacco-​related diseases are complex. shown for the United States in Figure 11–​1 (US Department of Health
Despite progress in decreasing smoking prevalence in many countries, and Human Services, 2014). The prevalence of cigarette smoking and
the number of smokers worldwide is projected to increase from 1.3 per capita consumption began to decrease in the United States and the
billion in 2010 to 1.7 billion by 2025, with much of the growth occur- United Kingdom by the mid-​1960s, but continued to increase else-
ring in low-​to middle-​income countries. Population-​based surveys where as economies recovered after World War II. In high-​income
have only recently begun to monitor patterns of tobacco use in many countries, smokers who did not quit and who represented birth cohorts
countries. In high-​income countries, lung cancer risk has continued to with the highest lifetime smoking histories have dominated age-​
increase among cigarette smokers for decades after the initial decline standardized lung cancer rates for decades (Pirie et  al., 2013; Thun
in smoking prevalence, due to birth cohort patterns in age at initiation et al., 2013).
of smoking as well as intensity. The histologic distribution of lung The design and composition of tobacco products has also changed
cancers has also changed in many countries, because of changes in over time (US Department of Health and Human Services, 2014). The
the design and composition of cigarettes. The use of multiple tobacco introduction of blended and reconstituted tobacco in high-​ income
products continues to complicate tobacco control, as does the increas- countries in the mid-​twentieth century changed the bioavailability of
ing use of novel products such as e-​cigarettes that have created chal- nicotine and the relative concentration of different classes of carcino-
lenges for regulators and have divided the public health community. gens in tobacco smoke. The introduction of filters (initially plain cellu-
lose acetate filters and later “ventilated” filters that diluted the smoke)
did not produce the anticipated linear reduction in exposure to carcino-
INTRODUCTION gens (US Department of Health and Human Services, 2010). Tobacco
products other than cigarettes continue to be widely used, even in high-​
Tobacco use is the largest single recognized cause of cancer and other income countries (Table 11–​1). While manufactured cigarettes are by
non-​communicable diseases worldwide. Prolonged cigarette smoking far the most commonly used tobacco product worldwide, 15%–​35% of
is estimated to cause premature death in one of every two smokers (Peto global production involves other products (Tobacco Atlas, 2015). The
et al., 1992). Cigarette smoking alone has been estimated to account emergence of novel nicotine-​delivery products, such as e-​cigarettes, is
for about 30% of all cancer deaths in the United States since the early further complicating patterns of consumer behavior.
1980s (Doll and Peto, 1981; Jacobs et  al., 2015; US Department of Epidemiologic studies continue to be central in identifying the
Health and Human Services, 2004); smoking causes even more deaths adverse health effects of tobacco use and in monitoring the progres-
from non-​ malignant cardiovascular and respiratory diseases than sion of the tobacco epidemic. Humans, unlike other mammals, use
from cancer (US Department of Health and Human Services, 2014). tobacco voluntarily (Thun and Henley, 2006). Whereas tobacco smoke
Despite reductions in smoking prevalence and consumption in many is irritating and highly toxic to other species exposed experimentally in
countries, the global burden of cancer and other diseases caused by laboratory studies, non-​smokers who experiment with tobacco persist
tobacco use is increasing rapidly because massive numbers of young through the initial noxious effects until they develop physical and psy-
smokers in highly populous low-​and middle-​income countries are chological dependence on tobacco. Observational (non-​randomized)
now surviving into the ages where cancer becomes common. Tobacco studies of smoking continue to be ethical and feasible, whereas ran-
use caused an estimated 100 million deaths in the twentieth century domized clinical trials are not. Epidemiologic studies are better suited
(Peto et  al., 2015). If current smoking patterns persist, the growing than clinical observations to surmount the long delay between the
toll is expected to exceed one billion deaths in the twenty-​first century initiation of exposure and the onset of tobacco-​related diseases. The
(Jha et al., 2013). association between active smoking and many diseases is sufficiently
The history of how tobacco use was transformed from a ceremo- strong that epidemiologic studies provide convincing evidence of
nial practice in precolonial North and South America into the world’s causality despite imprecise quantification of lifetime tobacco expo-
largest preventable cause of non-​communicable diseases has been sure (Thun and Henley, 2006). For all of these reasons, epidemio-
reviewed extensively elsewhere (Brandt, 2007; Doll, 1998; IARC, logic studies have been essential in identifying the long-​term adverse

185
186

186 PART III:  THE CAUSES OF CANCER


14

12
Pounds of tobacco per capita
10

Snuff
6
Chewing
Pipe/roll your own
4
Cigars
2 Cigarettes

0
1880 1890 1900 1910 1920 1930 1940 1950 1960 1970 1980 1990 2000 2010
Year

Figure 11–​1.  Per capita sale of tobacco products in the United States, 1880–​2011. Source: Surgeon General Report (2014).

effects of tobacco use and the progression of the tobacco epidemic in countries (Centers for Disease Control and Prevention [CDC], 2016;
populations. Warren et  al., 2009). The two largest surveillance initiatives are the
This chapter will discuss (a)  the epidemiology of cigarette smok- Global Adult Tobacco Survey (GATS) and the Global Youth Tobacco
ing and use of other tobacco products; (b)  the composition of ciga- Survey (GYTS). GATS is a nationally representative, face-​to-​face
rette smoke; (c)  aspects of product design and usage that influence household interview of persons 15 years of age and older, designated
exposure; (d)  tobacco products other than manufactured cigarettes; as “adults.” GATS has collected cross-​sectional data on all forms of
(e)  cancers caused by various forms of tobacco use; (f)  the benefits tobacco use and cessation on more than three billion individuals in 16
of cessation; (g) the adverse health effects of involuntary exposure to countries (Giovino et al., 2012). The parallel survey of youth, GYTS,
tobacco smoke; (h)  emerging tobacco products such as e-​cigarettes is a school-​based survey designed to provide primarily cross-​sectional,
and other products; and (i)  opportunities for prevention and future nationally representative estimates of tobacco use among students, age
research. Readers are referred to the chapter on tobacco in the previ- 13–​15 years, in 168 countries (Warren et al., 2008). The area covered
ous edition of this text for additional background information (Thun can be a country, province, city, or any other geographic area. Other
and Henley, 2006). data sources, such as the Demographic and Health Survey, the World
Health Organization (WHO) STEPS instrument, the Canadian Youth
Smoking Survey, and the US National Tobacco Survey are described
GLOBAL PATTERNS OF TOBACCO USE elsewhere (National Cancer Institute and Centers for Disease Control
and Prevention, 2014).
Geographic Distribution
Figure 11–​ 2 illustrates that, among men, the prevalence of daily Temporal Variation
cigarette smoking in 2012 was highest in Eastern Europe, the former
Soviet Union, and Eastern Asia (Ng et al., 2014). In women, the preva- Per Capita Sales
lence was highest in North America, Northern Europe, and Australia. As indicated in Figure 11–​1, most tobacco use in the United States in
The Global Tobacco Surveillance System has greatly expanded col- the early twentieth century involved chewing tobacco, pipes, cigars,
lection of nationally representative data in middle-​and low-​income and snuff, rather than cigarettes (US Department of Health and Human
Services, 2014). The invention during the 1880s of portable safety
matches and cigarette rolling machines (which could mass-​produce
Table 11–​1. Commonly Used Tobacco Products cigarettes that were considerably less expensive than hand-​rolled prod-
ucts) made it possible to smoke tobacco frequently throughout the day
Smoked/​Combustible
in diverse settings. These technologies were coupled with novel and
Cigarettes (Manufactured and hand-​rolled)
aggressive advertising campaigns that glamorized smoking of par-
Cigars (Large and small)
ticular brands of cigarettes, beginning with Camel cigarettes in 1913
Pipes
(Slade, 1993). Free cigarettes were distributed to soldiers during World
Bidis
Wars I and II and the Korean War.
Kreteks
The data on per capita sales in Figure 11–​1 are based on tax infor-
Waterpipes (hookah, shisha, narghile)
mation on total sales of tobacco by weight. If these were expressed as
Chutta
the number of cigarettes sold per capita, they would show a more than
Non-​combusted 80-​fold increase in the United States from 1900 (when the average was
Chewing tobacco 54 cigarettes per person per year) to 1963 (when the average peaked at
Snuff (moist and dry, high and low nitrosamine content) 4345 cigarettes per person per year) (CDC, 1999).
Betel quid (Pan masala) Figure 11–​3 shows that the global sale of manufactured cigarettes
Novel tobacco products increased nearly 500-​fold from 1880 to 2009 (Asma et al., 2014).
E-​cigarettes and other electronic nicotine delivery products (ENDS) Almost six trillion cigarettes were sold worldwide in 2009. While the
Dissolvable products rate of increase has slowed, the number of cigarettes sold continues
to increase because of population growth and the persistently high
 187

(a) Age–standardized smoking prevalence among men, 2012

Smoking prevalence deciles, %


0.5 to < 2.6 16.5 to < 19.7
2.6 to < 5.6 19.7 to < 23.4
5.6 to < 9.8 23.4 to < 27.5
9.8 to < 13.3 27.5 to < 34.7
13.3 to < 16.5 34.7 to < 61.1
Deciles of prevalence were calculated
for men and women combined.

(b) Age–standardized smoking prevalence among women, 2012

Smoking prevalence deciles, %


0.5 to < 2.6 16.5 to < 19.7
2.6 to < 5.6 19.7 to < 23.4
5.6 to < 9.8 23.4 to < 27.5
9.8 to < 13.3 27.5 to < 34.7
13.3 to < 16.5 34.7 to < 61.1
Deciles of prevalence were calculated
for men and women combined.

Figure 11–​2.  Global prevalence of daily cigarette smoking among men (a) and women (b), estimated for 2012. Modified from Ng et al., JAMA (2014).

6000

5000

4000
Billions of cigarettes

3000

2000

1000

0
1880 1890 1900 1910 1920 1930 1940 1950 1960 1970 1980 1990 2000 2009
Year

Figure 11–​3.  Trends in global sale of manufactured cigarettes (billions of sticks). Source: Asma et al. (2014).
18

188 PART III:  THE CAUSES OF CANCER


prevalence of cigarette smoking, especially among men in highly pop- countries collected for the WHO Global Monitoring Framework indi-
ulous middle-​and low-​income countries. The largest market for manu- cate that male smoking is decreasing in 125 (72%) countries and that
factured cigarettes is in China, where more than 300 million smokers female smoking is decreasing in 156 (88%) (Bilano et  al., 2015).
consume 37% of all cigarettes sold (Tobacco Atlas, 2015). However, only one in five countries is on track to achieve the 2025
Sales data derived from tax records provide the earliest objective targets specified by WHO. Furthermore, the prevalence of cigarette
record of the uptake of cigarette smoking in a given country. They smoking was increasing rapidly among men in parts of sub-​Saharan
indicate that the increase in smoking manufactured cigarettes occurred Africa and among both sexes in the Eastern Mediterranean. The abso-
first in North America and Northern Europe and then proceeded to lute number of smokers is also increasing worldwide because of popu-
Central, Southern, and Eastern Europe, South America, and Asia. lation growth (Ng et al., 2014).
Harsh economic conditions during and after World War II delayed
the increase in cigarette smoking by men in China until the 1960s, Birth Cohort Patterns
with much of the initial increase involving light smoking by adult In high-​income countries, the uptake of cigarette smoking has followed
men (Chen et al., 2015). The sustained decrease in per capita cigarette strong birth cohort patterns in terms of prevalence and consumption.
sales that began later in the twentieth century has followed approxi- Birth cohort patterns typically reflect exposures or exposure patterns
mately the same progression. The downturn in the United States fol- that are established in early life and then affect behavior and/​or dis-
lowed publication of the inaugural 1964 Surgeon General Report ease occurrence later in life. The initiation of tobacco use is influenced
on Smoking and Health (US Department of Health Education and by social norms as each birth cohort passes through critical periods
Welfare, 1964). Between 1964 and 2012, per capita sales decreased by of life, especially adolescence. The trajectory of continued smoking
72%, from 4345 cigarettes per adult per year in 1964 to 1196 in 2012. within a birth cohort is influenced by peer behavior, tobacco market-
By comparison, smoking prevalence decreased by 58% in men and ing, and other social and economic conditions that individuals encoun-
women combined, as discussed below (US Department of Health and ter as they age through different periods of calendar time.
Human Services, 2014). Birth cohort trends in the uptake of cigarette smoking in the United
Sales data provide a useful ecological indicator of the average inten- States have been reconstructed from serial National Health Interview
sity of smoking at various points in time. Their main limitation is that Surveys (NHIS) (Burns et al., 1997; Harris, 1983; Holford et al., 2014).
they do not indicate who is smoking the cigarettes. Descriptive infor- Figure 11–​5 shows the birth cohort trends in smoking prevalence for
mation on differences in smoking behavior between women and men men and women in the United States born between 1890 and 2000 and
by age, birth cohort, and other characteristics can be derived only from followed through 2010 (Holford et al., 2014). These data are adjusted
population-​based surveys. for the differential mortality of smokers. Prevalence at first increased
and then decreased in a distinctive wave-​like progression across suc-
Smoking Prevalence cessive 5-​year birth cohorts. The progression in men preceded that in
The first national survey of smoking prevalence among adults in the women by about 15 years. The highest prevalence of daily cigarette
United States was conducted in 1955, when 57% of men and 28% smoking in men occurred around 1950 in the birth cohort 1920–​1925;
of women age 18 years and older reported current cigarette smoking the corresponding peak in women occurred in about 1965 in the birth
(Haenszel and Shimkin, 1956). Prevalence was reportedly even higher cohort 1935–​1940.
in Britain between 1948 and 1952, when nearly 70% of men and more The birth cohort patterns in Figure 11–​5 are asymmetrical in two
than 40% of women between the ages of 25 and 59 smoked cigarettes respects. First, the downturn in smoking prevalence begins at progres-
(Peto et al., 2000). Figure 11–​4 shows the 58% decrease in smoking sively younger ages in the birth cohorts from 1890–​1894 to 1950–​
prevalence among men and women in the United States from 1965 1954 in men and after 1960–​1964 in women. This downturn reflects
(42.7%) to 2012 (18.1%). By 2015, the proportion of US adults, age increased cessation, due to growing public awareness about the
> 18 years, who currently smoked cigarettes had decreased further to adverse health effects of smoking, that occurred first among men in the
15.1% (Jamal et al., 2016). 1950s and later among women (US Department of Health and Human
Smoking prevalence and/​or consumption are now decreasing in Services, 2014). The second asymmetry is the partial resurgence in
the majority of countries worldwide (Ng et  al., 2014). Data on 172 smoking prevalence that began in the 1970s for women and 1990s for

90

80

70
Current smoking prevalence (%)

Males
Females
60

50

40

30

20

10

0
1965 1970 1975 1980 1985 1990 1995 2000 2005 2010 2015
Year

Figure 11–4.  Annual global sale of manufactured cigarettes (billions of sticks), 1880–​2009. Source: Surgeon General Report (2014).
 189

Tobacco 189
(a) Males
90

80

70
Current smoker prevalence (%)

60

50

40

30

20

1930

1950
1940
1920
1910
1900

1960

0
1890

198

0
10

199
70
19
0
1890 1900 1910 1920 1930 1940 1950 1960 1970 1980 1990 2000 2010
Year

(b) Females
90

80

70
Current smoker prevalence (%)

60

50

40

30

20
1940
1930
1920

1950

1960

10 00
0

0
10

19
197

198

199
19

0
189
0
1890 1900 1910 1920 1930 1940 1950 1960 1970 1980 1990 2000 2010
Year

Figure 11–​5.  Prevalence of current smoking by gender, calendar year, and birth cohort in the United States. Source: Holford TR et al. (2014).

men. The resurgence among women coincided with the introduction continued to increase until it peaked in the 1935–​1939 birth cohort of
of the Virginia Slims brand of cigarettes and increased initiation of men and 1940–​1944 birth cohort of women (Holford et al., 2014) (data
smoking among young women (O’Keefe and Pollay, 1996; Toll and not shown). Daily cigarette consumption among smokers continued
Ling, 2005). The upturn among men likely reflects a decrease in ces- to increase for up to 20 years beyond the initial population decrease
sation rates after 1970, when ventilated cigarettes with progressively in smoking prevalence. The complex birth cohort patterns in smoking
lower “tar” ratings, as measured by machine smoking, were marketed prevalence, intensity, and age at starting give rise to the birth cohort
as having putatively lower health risks (US Department of Health and patterns in lung cancer incidence and mortality observed 20–​50 years
Human Services, 2014). later in the general population, as discussed in Chapter 28.
Not all indices of smoking behavior follow exactly the same Not all countries have followed the same temporal sequence in
birth cohort patterns. Whereas smoking prevalence peaked in birth which the uptake of smoking in women follows 15–​20 years after the
cohorts 1915–​ 1919 and 1920–​ 1924 among men and 1935–​ 1938 uptake in men. The opposite pattern occurred among Chinese women,
among women (Figure 11–​5), cigarette consumption among smokers in whom the prevalence of ever smoking decreased over successive
190

190 PART III:  THE CAUSES OF CANCER

30 2 study areas (1 urban, 1 rural) in


28 Northeast or Southwest China

24

Ever-smoking prevalence (%)


20

12

10

8 remaining 6
5 study areas
3
2
1 0 0
0
1930 1940 1950 1960 1970
Mean birth year

Figure 11–​6.  Prevalence of ever-​smoking among Chinese women in Kadoori Study, by year of birth and locality. Source: Chen Z et al. (2015).

birth cohorts as smoking by women became unfashionable. Figure These include at least 70 chemicals designated by the IARC as having
11–​6 shows that nearly 30% of Chinese women born in the mid-​1930s sufficient evidence of carcinogenicity, based on studies of laboratory
and participating in the Kadoori study smoked at some point in their animals and/​or humans (IARC, 2012a; US Department of Health and
life (Chen et al., 2015). This decreased to 2% among those born in Human Services, 2014). Comprehensive reviews of the chemical com-
the late 1960s. In contrast, the prevalence of ever smoking increased position of tobacco smoke are available elsewhere (IARC, 2004).
among Chinese men, peaking at nearly 80% in rural areas in those At least seven classes of carcinogens are formed by the combustion
born during the 1950s and early 1960s, before decreasing in later of tobacco (Table 11–​2). These include polycyclic aromatic hydro-
birth cohorts (Chen et al., 2015). carbons (PAHs), heterocyclic compounds, N-​nitrosamines, aromatic
Patterns of tobacco use continue to change worldwide. For example, amines, formaldehyde, phenolic compounds, and a variety of free
in the United States the prevalence of light and intermittent smoking radicals (IARC, 2004). Others, such as arsenic, cadmium, chromium,
has become more common. Over the last 10 years, the proportion of nickel, and polonium 210, are incorporated into the tobacco plant from
non-​daily cigarette smokers has increased from 19.2% to 24.3% of soil or phosphate fertilizers. The concentrations of carcinogens such as
smokers; the proportion of smokers who smoke less than 10 cigarettes tobacco-​specific nitrosamines are increased by fermentation and some
per day increased from 16% to 25%. A growing proportion of smok- forms of curing. The concentration of nicotine and the rapidity with
ers, particularly young people, also use cigarettes in combination with which it can be absorbed depends on the selection of tobacco strains
other tobacco products, such as e-​cigarettes or water pipes. and the pH of the smoke (IARC, 2004; US Department of Health and
Figure 11–​7 shows the dramatic changes in the tobacco products Human Services, 2010). The chemical characteristics of the smoke
being smoked by middle and high school students in the United States have not generally been considered in epidemiologic studies of cancer,
from 2011 to 2015 (Singh T et  al., 2016). The use of e-​cigarettes largely because the presence and concentration of various constituents
increased rapidly throughout the period, and e-​cigarettes became the have changed over time and are difficult to reconstruct.
most commonly used tobacco product by 2014. The increased use of e-​
cigarettes was accompanied by decreases in all other forms of tobacco
use except waterpipes. About half of the students who reported any Mainstream Versus Sidestream Smoke
tobacco use used > 2 products.
Active smoking generates both mainstream smoke (MS) and side-
stream smoke (SS):  MS is drawn directly from the burning tobacco
NATURE OF THE EXPOSURE into the mouth; SS is released from the smoldering tobacco into the
ambient air, where it mixes with exhaled MS to make up environmental
tobacco smoke (ETS). Involuntary exposure to ETS, also called pas-
Smoked Tobacco Products sive or secondhand smoking, involves similar chemical constituents,
Manufactured cigarettes comprise an estimated 84% of global tobacco although the concentrations may differ by several orders of magnitude
consumption. Commonly used smoked and non-​combustible tobacco (IARC, 2004; US Department of Health and Human Services, 2014).
products are listed in Table 11–​1 and are discussed later in this chapter.

Nicotine
Composition of Tobacco Smoke Nicotine is the principal alkaloid present in tobacco and accounts for
Tobacco smoke is a complex, heterogeneous mixture that contains at 0.05%–​4.00% (by weight) of the tobacco leaf (US Department of
least 5300 (and by some estimates as many as 7000) identified chem- Health and Human Services, 1988). While nicotine itself is not car-
icals (US Department of Health and Human Services, 2010, 2014). cinogenic, physical dependence on nicotine is the main factor that
 19

Tobacco 191
100

2011
2012
2013
25
2014
2015

20
Percentage of students

15

10

0
Any ≥ 2 types E-cigarettes Cigarettes Cigars Hookahs Smokeless Pipe tobacco Bidis
tobacco
Tobacco product

Figure 11–​7.  Tobacco use in middle and high school students, United States, 2011–​2015. Source: Singh T et al. (2016).

sustains tobacco use (US Department of Health and Human Services, neurotransmitters such as dopamine, serotonin, and γ-​aminobutyric
2014). Nicotine from tobacco binds with the nicotinic receptors for acid. Exposure to exogenous nicotine stimulates the production of
acetylcholine in the central and peripheral nervous systems. In the additional nicotine receptors (Benowitz, 1996).
central nervous system (CNS), the receptors regulate the release of For most users, nicotine addiction from tobacco use represents true
drug dependence (US Department of Health and Human Services,
2010). Withdrawal symptoms among cigarette smokers who attempt
Table 11–​2. Tobacco Smoke Carcinogens Evaluated in the IARC
to quit include anxiety, irritability, weariness, constipation/​diarrhea,
Monographs
insomnia, intense craving, and difficulty concentrating (Balfour and
Number of Fagerstrom, 1996). The severity of these symptoms may equal with-
Chemical Class Carcinogens Representative Carcinogens drawal from opiates, amphetamines, and cocaine. The strength of the
addiction is illustrated by the high failure rate among smokers who
Polycyclic aromatic 15 Benzo(a)pyrene (BaP) attempt to quit. Approximately 70% of current smokers express a
hydrocarbons Dibenz(a,h)anthracene desire to quit, yet fewer than 50% try to stop each year (Centers for
(PAHs) and their Disease Control and Prevention, 2000). Unassisted, about 2.5% of
heterocyclic smokers succeed in quitting permanently on a single quit attempt. The
analogues success rate approximately doubles with appropriate pharmacological
N-​Nitrosamines 8 4-​(Methylnitrosamino)-​1-​(3-​ and/​or behavioral treatment.
pyridyl)-​1-​butanone (NNK)
N'-​Nitrosonornicotine (NNN)
Aromatic amines 12 4-​Aminobiphenyl Factors That Influence Exposure
2-​Naphthylamine
Aldehydes 2 Formaldehyde Cumulative exposure to the carcinogens in tobacco depends on fac-
Acetaldehyde tors that affect the design and manufacture of the product, and social,
Phenols 2 Catechol economic, and biological determinants of usage.
Caffeic acid
Volatile 3 Benzene Bioavailability of Nicotine
hydrocarbons 1,3-​Butadiene Tobacco products vary in their delivery of nicotine in a form that can
Isoprene be rapidly absorbed. Differences in the bioavailability of nicotine
Other organics 12 Ethylene oxide influence both the addictiveness of various products and the surface
Acrylonitrile
area of epithelium or number of cells exposed to harmful constituents
Inorganic 8 Cadmium
compounds Polonium-​210
in tobacco smoke. Cigarettes and moist snuff increase plasma nicotine
concentration almost immediately (Figure 11–​8) and are most addic-
tive, whereas nicotine replacement products generally provide much
There are many other carcinogens in cigarette smoke that have not been evaluated in an
IARC Monograph.
slower nicotine uptake (Benowitz, 1996; US Department of Health
Source: IARC (2004a). and Human Services, 2010). Inhalation of cigarette smoke increases
192

192 PART III:  THE CAUSES OF CANCER


Tobacco
20
Cigarette (nicotine Oral snuff Nasal spray (1 mg)
delivery 1–2 mg)
15

Plasma nicotine concentration (ng/mL)


10

20
Nicotine gum (Polacrilex 4 mg) Nico Derm CQ patch (21 mg) Nicotrol patch (21 mg)

15

10

0
0 30 60 90 120 0 30 60 90 120 0 30 60 90 120
Time post administration (minutes)

Figure 11–​8.  Plasma nicotine uptake from various tobacco products.

plasma nicotine and produces discernible CNS effects in as little as 7 et  al., 2003). Even when measured by machine smoking, the “tar”
seconds owing to the large surface area of the lungs. and nicotine yields of cigarettes vary by a factor of 3 across WHO
The uptake of nicotine from various tobacco products depends on regions (Calafat et  al., 2004). Although epidemiologic studies have
the pH of the product. Commercial brands of moist snuff produce a pH not generally been able to consider measurements of the chemical
range in saliva from 5.39 (at which less than 3% of the nicotine is free composition of smoke in different countries when comparing risk,
or un-ionized) to 8.19 (where at least half of the nicotine is un-ionized the observed variations in TSNA levels provide a promising target
and can be rapidly absorbed) (Djordjevic et al., 1994). Nicotine in an for regulation. As described later in the chapter, urinary markers of
alkaline environment, as produced by pipes, cigars, smokeless tobacco TSNA exposure have been associated with esophageal and lung can-
products, and cigarettes in the first half of the twentieth century is cer (Yuan et al., 2014).
absorbed through the oral mucosa. Alkaline smoke is too irritating for
most people to inhale. In contrast, protonated nicotine, as delivered by Ventilated Cigarettes
contemporary cigarettes, is absorbed more rapidly through respiratory Cigarettes with ventilated filters were introduced in the United States
epithelium. Deeper inhalation was made palatable by the development, in the late 1960s. Unlike plain cellulose acetate filters, these were
selection, and mixing of special tobacco blends and other modifica- designed with ventilation holes that allowed air to enter and dilute the
tions. Inhalation not only accelerates the absorption of nicotine, but smoke when the cigarettes were smoked by machines (US Department
also exposes a much larger surface area to the toxic and carcinogenic of Health and Human Services, 2014). Machine-​measured “tar” and
compounds in smoke. nicotine ratings from cigarettes with ventilated filters were substan-
tially lower than machine ratings from a similar cigarette with an
Other Aspects of Product Design and Composition unventilated filter or from an unfiltered cigarette. Smokers who used
As mentioned, the concentration of toxic and carcinogenic chemicals these products could block the ventilation holes to obtain their accus-
in tobacco smoke is influenced by methods of curing and manufactur- tomed level of intake. Nevertheless, they perceived the smoke to be
ing, as well as by agricultural practices. For example, the introduction less harsh than that from unfiltered or unventilated cigarettes. Cigarette
of reconstituted tobacco in the 1950s allowed manufacturers to use the companies marketed these products to health-​conscious smokers as an
ribs and stems from tobacco as well as the leaves (US Department of alternative to quitting, using the terms “light,” “ultralight,” “mild,” and
Health and Human Services, 2010). This reduced waste and produc- “low tar” to imply lower health risks.
tion costs and somewhat lowered the concentration of polycyclic aro- The method of machine smoking was developed by the tobacco
matic hydrocarbons (PAHs), but greatly increased the concentration industry during the 1930s and was adopted officially by the Federal
of tobacco-​specific nitrosamines (TSNAs) (Hoffmann and Hoffmann, Trade Commission (FTC) in 1969. It became known as the “FTC pro-
1997; US Department of Health and Human Services, 2014). Tobacco tocol” (Institute of Medicine, 2001). Settings were fixed so that the
that is flue-​cured and has direct contact with the combustion byprod- machine took one 2-​second (35 ml) puff per minute until the ciga-
ucts from propane gas heaters also has higher concentrations of rette was consumed. Tar and nicotine were extracted from a special
TSNAs than air-​cured tobacco (Collishaw, 2016). Fermentation of filter through which the machine “smoked” the cigarette. Tar repre-
tobacco for use in cigars and moist snuff greatly increases the con- sents the total particulate matter after removing the nicotine and water
centration of TSNAs (US Department of Health and Human Services, (Institute of Medicine, 2001). As shown in Figure 11–​9, the average
2010). Brands of moist snuff commonly used in the United States have sales-​weighted tar and nicotine yields of US cigarettes decreased dra-
TSNA levels far above the limit allowed in other consumer products matically according to machine measurements. The average “tar”
(Hoffmann and Hoffmann, 1997). yield per cigarette decreased from 38 mg in 1954 to 12 mg in 1998,
Cigarettes from different countries differ in their concentration while the average nicotine rating decreased from 2.3 mg to 0.9 mg (US
of nitrosamines, nitrates, nicotine, and other compounds (Ashley Department of Health and Human Services, 1981, 1989; Kozlowski
et  al., 2003). Comparative studies of the levels of TSNAs in ciga- et al., 2001). Initially, the reduction was achieved by adding cellulose
rette tobacco documented that Marlboro cigarettes purchased in 11 acetate filters and using more porous wrapping paper (Hoffmann and
of 13 foreign countries had significantly higher TSNA levels than Hoffmann, 1997). Further reductions were achieved after 1970 by the
the locally popular non-​U.S. brands from the same country (Ashley use of ventilated filters, “puffed tobacco” that reduced the amount
 193

Tobacco 193
4.0 3.0

3.5 Tar
2.5
Nicotine
3.0
2.0
2.5

Nicotine (mg)
Tar (mg)
2.0 1.5

1.5
1.0
1.0
0.5
0.5

0.0 0.0
1950 1955 1960 1965 1970 1975 1980 1985 1990 1995
Year

Figure 11–​9.  Sales-​weighted tar and nicotine values for US cigarettes as measured by a smoking machine using the Federal Trade Commission (FTC) method,
1954–​1998. Note: Values before 1968 are estimated from available data. Courtesy of D. Hoffmann, personal communication. Source: National Cancer Institute
Smoking and Tobacco Control Monograph 13 (2002), p. 2.

per cigarette, and modified wrapping paper that burned more rap- States, since they are exempted from the excise taxes on manufactured
idly so that the testing machine could take fewer puffs (Kozlowski cigarettes. For tax purposes, cigars are defined as shredded tobacco
et al., 2001). wrapped in tobacco leaf or paper (Department of the Treasury, 2006).
Epidemiologists who studied the cancer risks from “reduced-​yield” They vary in size from cigarette-​sized cigarillos to cheroots and dou-
cigarettes were largely unaware of the changing technologies that ble coronas (Eriksen et  al., 2012). Small filter-​tip cigars are usually
affected the machine measurements. Studies conducted before the smoked like cigarettes but are less heavily taxed and can be purchased
year 2000 equated “high” and “low” tar cigarettes with unfiltered and individually. Their use is increasing in the United States, whereas the
filter-​tip cigarettes respectively. It was assumed that a reduction in risk previously widespread use of pipes is decreasing worldwide (Shafey
associated with the addition of a filter would correspond to further et al., 2009). Waterpipes (shisha or sheecha) have been used tradition-
reductions in risk across much lower levels of machine-​measured “tar” ally in Middle-​Eastern countries and in Russia and Vietnam (Giovino
(Harris et  al., 2004). The critical distinction between ventilated and et  al., 2012; IARC, 2004), but are increasingly marketed to young
unventilated filters had not yet been disclosed to the public. people in Western countries using flavors of molasses, apple, banana,
The limitations of the FTC protocol in measuring the risk of venti- vanilla, and other fruits or candies (Tobacco Atlas, 2016).
lated cigarettes became apparent by the late twentieth century, when The most common form of smoked tobacco in India involves bidis,
age-​standardized lung cancer rates continued to increase in smokers, traditionally hand-​rolled in dried temburni leaf and tied with a string
despite large reductions in machine-​measured yield (Thun et al., 1997; (Shafey et  al., 2009). Kreteks are clove-​and cocoa-​flavored small
US Department of Health and Human Services, 1989). Studies of sali- cigars that are produced and sold in Indonesia but also marketed
vary cotinine found little relationship between machine-​measured rat- worldwide (Gandini et al., 2008).
ings below 1.0 mg nicotine and actual nicotine uptake (Figure 11–​10)
(Benowitz, 2001; Jarvis et al., 2001). Moreover, an analysis by Harris Non-​Combusted Products
and colleagues at the American Cancer Society found no reduction An historical overview of the use of smokeless or non-​combusted
in lung cancer risk from “light” or “ultralight” cigarettes compared tobacco products is provided in IARC Monograph 89 (IARC,
to “regular” cigarettes with unventilated cellulose acetate filters, but 2007)  and more recently in a National Cancer Institute Monograph
higher risk from smoking unfiltered cigarettes (Figure 11–​11) (Harris (National Cancer Institute and Centers for Disease Control and
et al., 2004). Prevention, 2014). Traditional forms of smokeless or “spit” tobacco
Based on these findings, the FTC rescinded its approval of machine used in Western countries include snuff (moist and dry) and chewing
smoking in 2008 and the Food and Drug Administration (FDA) banned tobacco. In the United States alone, several million people use smoke-
the use of descriptors such as “light,” “mild,” and “low tar” in the less products (Agaku et  al., 2014; Lee et  al., 2014; Mazurek et  al.,
United States. The tobacco industry has subsequently circumvented 2014). Moist snuff is the most commonly used product in the United
this restriction by introducing color codes that smokers recognize as States (Agaku and Alpert, 2015); snus, a lower nitrosamine form of
equivalent to the banned terms (Connolly and Alpert, 2014). moist snuff, is widely used in Nordic countries. Moist snuff consists
of finely ground tobacco with 20%–​55% moisture content, often fla-
vored with mint, wintergreen, or raspberry (Borgerding et al., 2012).
Tobacco Products Other Than Cigarettes A pinch (called a “dip” or “rub”) is placed between the gum and the
cheek or under the tongue (Shafey et al., 2009). Chewing tobacco is
Combustible popular among baseball players, male adolescents, and in both sexes
As mentioned, about 15%–​ 35% of global tobacco consumption in some rural populations in the United States. Products may be fla-
involves tobacco products other than manufactured cigarettes (Eriksen vored with sugar, molasses, or licorice, and in the United States are
et  al., 2012). These are listed in Table 11–​1. Other smoked tobacco sold with a range of concentrations of nicotine and other toxicants
products include hand-​ rolled cigarettes, cigars, pipes, and a vari- that increase as users become habituated. Dual use of cigarettes plus
ety of products smoked widely in Southeast Asia. Hand-​rolled ciga- smokeless tobacco products is most common among young unmar-
rettes made from loose tobacco are used increasingly in the United ried men (Lee et al., 2014). This form of poly-​tobacco use increased
194

194 PART III:  THE CAUSES OF CANCER


900

800

700

Saliva cotinine (ng/ml)


600

500

400

300

200

100

0
0.0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 1.0 1.1
Nicotine yield (mg)

Figure 11–​10.  Range of salivary cotinine among smokers smoking cigarettes within a given FTC rating for nicotine. Source: Jarvis et al. (2001).

among users under age 26 in the United States between 2002 and 2011 countries (Palipudi KM et  al., 2015). They were invented by the
(Fix et al., 2014). The predominant form of tobacco used in Sweden Chinese pharmacist Hon Lik in the first decade of the 2000s and are
is moist snuff or snus with much lower concentration of nitrosamines principally manufactured in China. According to tobacco industry
than most products in the United States (Nilsson, 1998). documents, cigarette manufacturers were working to develop simi-
Betel quid or pan is widely used in Asia and the Western Pacific, lar products since the early 1990s to serve as alternatives to nicotine-​
especially by women (Shafey et al., 2009). Betel leaves (Piper betle) replacement therapy and for use in smoke-​free environments, but only
are mixed with tobacco, Areca nut (Areca catechu), lime, wood ash, recently decided to bring these products to market (Dutra et al., 2016).
or other substances to form a “quid,” “pan,” or “nass” (IARC, 2004). Users obtain nicotine through vaporization of a liquid solution that
The mixture is then chewed and/​or retained in the mouth. The use of typically includes a carrier compound such as propylene glycol, as
smokeless tobacco is estimated to cause more than 100,000 deaths well as various flavorings (Grana et al., 2014). Many of these flavors,
annually from cancer, with most deaths occurring in Southeast Asia such as “gummy bear” and “jolly rancher” are attractive to youth (US
(Ezzati et al., 2002; Sinha et al., 2016). Department of Health and Human Services, 2016). The products come
in many shapes and sizes, including both disposable and reusable
Novel Tobacco Products models, as illustrated in Figure 11–​12. Cross-​sectional studies indicate
that ENDS users are exposed to a number of carcinogens, including
Electronic Nicotine Delivery Devices (ENDS). Novel tobacco specific–​nitrosamines and acrolein (Goniewicz et al., 2014),
devices that heat tobacco or nicotine but do not produce smoke have although at concentrations that are lower and involve fewer carcino-
been introduced in recent decades. E-​cigarettes are the most widely gens than tobacco smoke.
used of the electronic nicotine delivery systems (ENDS). The use The ultimate impact of e-​cigarettes and other putative harm reduc-
of e-​cigarettes is greatest in the United States and Europe (Arrazola tion products on health is unknown. Although use of these products
et al., 2015), but uptake is also occurring in middle-​and low-​income produces lower exposures to toxic and carcinogenic compounds than

2.5 2.5

2.0 2.0
Hazard ratio (95% CI)

Hazard ratio (95% CI)

1.5 1.5

1.0 1.0

0.5 0.5

≤ 35 35–54 ≥ 55 ≤ 7 8–14 15–21 ≥ 21 ≤ 35 35–54 ≥ 55 ≤ 7 8–14 15–21 ≥ 21

Never smoked Quit smoking Tar level (mg) Never smoked Quit smoking Tar level (mg)
regularly at age (years) current smoker regularly at age (years) current smoker

Figure 11–​11.  Hazard ratios for lung cancer in men and women from the CPS-​II cohort by level of machine-​measured “tar” in cigarettes smoked for cur-
rent smokers or by age at cessation for former smokers. Source: Harris et al. (2004).
 195

Tobacco 195

ENDS type Product characteristics

Disposable e-cigarette • For short time use


• Cheap; widely available from gas
stations or super markets
• Convenient, as no refill solution or
cartridge is needed
• Might be a starting point for many users

Rechargeable e-cigarette • Various sizes from cigarette size


to compact flashlight size
• Various colors and style
• Requires battery with a charger
• Requires replacing or refilling
nicotine cartridges

Tank system e-cigarette • High capacity


• Battery operated
• Lasts longer than typical
rechargeable models
• Requires refilling the tank with
nicotine solution
• Often recommended for heavy
smokers by retailers

Personal Vaporizer • Large size and heavy weight


• Battery power output control allows
the user to adjust the output of the
voltage going to the heating element,
which has a direct effect on the
temperature of the heating element
• Usually not recommended by
retailers for beginning users

E-cigar • Looks like an authentic large ciger


• Usually disposable

Figure 11–​12.  E-​cigarettes and other ENDs. Source: ASCO, AACR policy statement: Brandon et al., JCO (2015).

active smoking, marketing practices may detrimentally affect cessa- effective smoking-​cessation devices. Observational studies are needed
tion and initiation patterns in the population. Smokers who continue to determine the effects of switching from cigarettes to e-​cigarettes on
to smoke in addition to using e-​cigarettes often use them to delay ces- disease risk.
sation by circumventing smoke-​free laws. Ultimately, it is unknown As discussed earlier, a substantial proportion of younger e-​cigarette
whether e-​cigarettes will be used predominantly by smokers or by users have never tried cigarettes or other tobacco products. Over
adolescents and young adults who do not otherwise use tobacco, and the last few years, longitudinal studies of tobacco-​use patterns have
whether e-​cigarettes are contributing to the decline in other forms of been initiated. Published studies so far suggest that youths who use
tobacco smoking among youth in the United States (Figure 11–​7). e-​cigarettes may be more likely to use cigarettes subsequently than
Accurate prevalence data on the use of e-​cigarettes and other ENDs is never e-​cigarette users (Barrington-​Trimis et al., 2016; US Department
only now emerging from the National Health Interview Survey (Figure of Health and Human Services, 2016). Whether these associations per-
11–​13) (Arrazola et al., 2015; QuickStats, 2016). The majority of e-​ sist in future studies and ultimately affect the prevalence of cigarette
cigarette smokers reported continuing to smoke cigarettes, especially smoking in the United States and other countries will be a critical
at ages > 25 years. A substantial proportion of e-​cigarette users in the gauge of the impact of e-​cigarettes on health.
youngest age group (18–​24  years) had never smoked cigarettes, but
may have used other tobacco products. The percentage who had never
smoked cigarettes became much smaller with age. There is consider- MEASUREMENT OF EXPOSURE
able variation among countries in awareness and use of e-​cigarettes
(Gravely et al., 2014).
Only a few studies have examined the effects of e-​cigarette use on Questionnaire Measures
cessation of cigarette smoking (Orellana-​Barrios et  al., 2016), with The best and most thoroughly validated external measures of active
data from randomized trials being particularly scarce. As described cigarette smoking derive from self-​ reports (Shields, 2002). Most
in the 2016 Surgeon General’s report (US Department of Health and adults can report whether they have smoked 100 or more cigarettes
Human Services, 2016), current knowledge suggests health benefits in their lifetime and whether they now smoke every day or on some
for current adult cigarette smokers who use e-​cigarettes as a way to days, the definition of current smoking (Centers for Disease Control
quit tobacco use completely or perhaps even as a long-​term replace- and Prevention, 1994). A meta-​analysis of 26 studies that evaluated the
ment for cigarette smoking. Much larger randomized clinical trials validity of self-​reported data on tobacco use found that self-​reported
are urgently needed, however, to determine whether e-​cigarettes are smoking status predicted biochemical evidence of active smoking
196

196 PART III:  THE CAUSES OF CANCER


100

90
Current cigarette smokers
80 Former cigarette smokers
Never cigarette smokers
70

60
Percentage

50

40

30

20

10

0
Total 18–24 years 25–44 years ≥45 years
Age group

Figure 11–​13. Cigarette smoking status among current adult e-​cigarette users, by age group. Sources: National Health Interview Survey (2016) in
QuickStats (2016).

with 87% sensitivity and 89% specificity (Patrick et  al., 1994; US a single tank of e-​juice over the course of a day or days. Waterpipe
Department of Health and Human Services, 2001). The sensitiv- users often share puffs with others over the course of several hours.
ity and specificity of self-​reported smoking were higher in studies Future studies of non-​cigarette tobacco products should extensively
of adults than in those of children. Less than 1% of respondents in query usage patterns in order to further delineate the health risks of
the National Health and Nutrition Examination Survey (NHANES) these products.
who denied the use of a nicotine-​containing product had an elevated The FDA Center for Tobacco Products (FDA-​ CTP) recently
serum cotinine level, after adjustment for passive smoking (Yeager and released a list of 93 harmful and potentially harmful constituents
Krosnick, 2010). Information on tobacco use of comparable quality (HPHC) in tobacco products and tobacco smoke that must be reported
can be collected online, by paper questionnaire, or by structured inter- by tobacco manufacturers and importers for their products (Food and
view (Steffen et al., 2014). Smoking history is considered a more sen- Drug Administration, 2012). This information has not yet been used
sitive measure of intermittent smoking than are biochemical indices, in epidemiologic studies, but could conceivably be coupled with bio-
as the half-​life of cotinine is only about 17 hours (Benowitz, 1996). markers of exposure to estimate exposure to specific chemicals.
However, self-​reported data on the number of cigarettes consumed per Exposure to secondhand smoke can be estimated from qualitative
day are thought to underestimate actual consumption by at least 20%. information on questionnaires, supplemented by quantitative assess-
Estimates of per capita consumption based on questionnaire surveys ments based on biomarkers or air measurements (Apelberg et  al.,
consistently underestimate consumption based on cigarette sales data 2013; Avila-​Tang et al., 2013a, 2013b) Study participants can describe
by 20%–​30% (Todd, 1978). the smoking status of their spouse, the number of smokers at home,
Despite their quantitative limitations, self-​reported data on num- and the extent of exposure at work or in other settings in the recent past
ber of cigarettes smoked per day, years of smoking, and ages at (see Chapter  17). Cotinine and other biomarkers of tobacco smoke
initiation and cessation have been sufficient to document etiologic provide objective evidence of exposure within the past 3–​7 days. Self-​
relationships between smoking and numerous disease endpoints reported information on exposures in childhood and at various periods
(IARC, 2004). They may not be sufficient to document relatively throughout life cannot be validated directly, but can provide qualitative
small differences in the pathogenicity of cigarettes, however, given information about past exposures.
potentially large unmeasured variations in smoking behavior. Other
measures that might refine estimates of cumulative exposure over a
Measures of Addiction
lifetime could consider the intensity of smoking at different ages, as
well as daily and non-​daily use, birth cohorts, indices of addiction, A parameter that has not often been included in epidemiologic stud-
the number and duration of failed cessation attempts, and the design ies but that appears informative is the level of nicotine addiction of
characteristics of the product being smoked. Behavioral character- individual smokers. The Fagerstrom index is a widely used index of
istics such as puff volume, the average number of puffs taken per addiction in behavioral studies (Fagerstrom, 1978; Fagerstrom et al.,
cigarette, depth of inhalation, and retention time in the lung cannot 1996) that measures six correlates of physical dependence. It includes
be reliably measured by questionnaire. Self-​reported data on depth questions such as: “How soon after you wake up do you smoke your
of inhalation have not proven to be reliable, and personal monitor- first cigarette? Do you find it difficult to refrain from smoking in places
ing has not yet been widely used. where it is forbidden? Do you smoke if you are so ill that you are in
Exposure assessment is less well developed for non-​ cigarette bed most of the day?” Questions in the Fagerstrom index may corre-
tobacco products. For example, few epidemiological studies have late with parameters of tobacco exposure that are difficult to measure
assessed risk in relation to age at initiation and cessation of use of by questionnaire, such as greater puff volume, depth of inhalation, and
products other than cigarettes. It is particularly difficult to assess expo- retention time in the lung. Including one of more of these questions in
sures from e-​cigarettes and waterpipes. E-​cigarette users may puff on epidemiologic studies might prove to be a useful adjunct to the current
 197

Tobacco 197
assessment of lifetime exposure. For example, a recent study found so far. Further studies in other populations with a range of tobacco use
that time to first cigarette was associated with lung cancer in and above patterns are merited.
standard smoking measures such as smoking duration and cigarettes
smoked per day (Gu et al., 2014). Biomarkers of Genotoxicity
The common practice of combining information on the intensity DNA adducts provide direct evidence that carcinogenic constituents
and duration of smoking into a single variable of cumulative exposure of tobacco smoke interact with circulating lymphocytes and with tis-
(pack-​years or cigarette-​years) is contraindicated. Analyses of both the sues affected by smoking-​related cancers (lung, oral cavity, urinary
British Doctors’ Study (Doll and Peto, 1978) and the American Cancer bladder, and uterine cervix). Cigarette smoking increases the muta-
Society Cohorts (Flanders et al., 2003) have shown that the duration genicity of urine, the activation of certain enzymes in body tissue, and
of smoking is a much stronger predictor of lung cancer risk than is the presence of DNA adducts from tobacco-​specific nitrosamines or
the number of cigarettes smoked per day. Lung cancer risk increases 4-​aminobiphenyl in lymphocytes, hemoglobin, and tumor tissue (US
with the fourth or fifth power of years of smoking but only the second Department of Health and Human Services, 2010). Adducts bound to
power of cigarettes per day. Researchers increasingly recommend that cellular macromolecules persist longer than nicotine metabolites after
pack-​years no longer be used as an exposure variable (Leffondre et al., abstinence from tobacco use. The number of chromosomal aberra-
2002), just as “ever” smoking is no longer considered an optimal cat- tions in cultured lymphocytes and the extent of lipid peroxidation have
egory of exposure because of changes in risk after cessation. been shown to correlate with the number of cigarettes smoked per day
(Shields, 2002).
Smoking produces multiple distinctive mutational signatures in
Biomarkers tumors that are correlated with aspects of cigarette smoking behav-
ior (Alexandrov et al., 2016). The most widely recognized are the
Various biomarkers have been used in epidemiologic studies of tobacco G→T transversions in TP53 found in adenocarcinomas and squamous
to assess absorption, metabolism, excretion, and biologic activity of cell carcinomas of the lung in smokers (Alexandrov et al., 2016; US
constituents in smoke (IARC, 2004; US Department of Health and Department of Health and Human Services, 2010). Thirty percent of
Human Services, 2010). the TP53 mutations in lung tumors of smokers involve G→T transver-
sions, whereas only 10% of the mutations in lung tumors obtained
Biomarkers of Exposure from non-​smokers are of this type. TP53 mutations in smokers occur
The most commonly used biomarkers of exposure are those that preferentially at hotspots where DNA adducts from exposure to poly-
reflect nicotine absorption. Cotinine is the main proximate metabo- cyclic aromatic hydrocarbons (PAH) are formed and incompletely
lite of nicotine and is considered the biomarker of choice for indicat- repaired (US Department of Health and Human Services, 2010). They
ing exposure to tobacco during the last 2–​7 days (Avila-​Tang et al., are found in pre-​neoplastic lesions as well as lung cancer in smokers,
2013a; Benowitz, 1996). The concentration of cotinine in plasma, indicating that they represent early somatic events (US Department of
saliva, or urine can reliably differentiate active smoking from ETS Health and Human Services, 2010).
exposure (IARC, 2004). Other biomarkers, such as thiocyanate Other studies have shown clear associations between cigarette smok-
in plasma or saliva, carbon monoxide in exhaled alveolar air, and ing and methylation status at numerous genomic regions (Joehanes
blood carboxyhemoglobin concentrations, are less sensitive and/​or et al., 2016). These and future molecular studies may provide further
less specific as markers of tobacco exposure than cotinine (Institute insights into the mechanisms by which tobacco causes various types of
of Medicine, 2001). Biomarkers of nicotine metabolism are highly cancer, may refine associations with particular subtypes of cancer, and
informative in studies of secondhand smoke, but less useful for quan- may suggest new treatment modalities.
tifying the lifetime history of active smoking because of their short
half-​life.
Other biomarkers reflect the uptake, activation, and excretion of CANCERS CAUSED BY ACTIVE CIGARETTE SMOKING
carcinogens in tobacco smoke. These are discussed extensively else-
where (IARC, 2012a; US Department of Health and Human Services,
2010). Studies that measure metabolites of tobacco-​specific nitrosa- Cancer Sites with “Sufficient Evidence”
mines in urine and other bodily fluids (Hecht et al., 2016) document Active cigarette smoking causes more than 20 different cancer sites
the absorption and metabolic activation of two powerful carcino- or subsites, according to designations by the IARC and the US
gens, 4-​(methylnitrosamino)-​1-​(3-​pyridyl)-​1-​butanone (NNK) and Surgeon General (IARC, 2012a; US Department of Health and Human
N'-​nitrosonornicotine (NNN) following active or passive exposure to Services, 2014). These include cancers of the lung (all histologic sub-
tobacco smoke or smokeless products (Hecht et al., 2008). types), oral cavity, nasal cavity and accessory sinuses, naso-​, oro-​, and
All of these biomarkers of exposure reflect recent time periods hypopharynx, larynx, esophagus (including both squamous cell car-
rather than lifetime exposure. They also cannot typically distinguish cinoma and adenocarcinoma), stomach, pancreas, colorectum, liver,
between forms of tobacco, challenging interpretation in populations kidney (including adeno-​and urothelial carcinomas), ureter, urinary
where dual or poly use is common. Biomarkers are useful, however, bladder, uterine cervix, ovary (mucinous), and acute myeloid leuke-
for validating questionnaire information on recent smoking, for docu- mia. Even this list may be incomplete, as it does not include sites such
menting the absorption of carcinogens from active and passive smok- as breast cancer or advanced prostate cancer for which the Surgeon
ing, and for excluding active smokers from studies of secondhand General has designated the evidence as “suggestive but not conclu-
smoke. However, they do not improve measurably on questionnaire sive” (US Department of Health and Human Services, 2014).
data in epidemiologic studies of smoking and cancer. Serum cotinine Table 11–​3 shows the relative risk (RR) estimates for the associ-
has been shown to predict lung cancer risk among smokers (Boffetta ation between current cigarette smoking and cancer sites that are
et al., 2006), but there is no evidence that it is more predictive than formally designated as smoking-​related by both the IARC and the
questionnaire-​based measures of smoking. Cotinine and carbon mon- Surgeon General (IARC, 2012a; US Department of Health and Human
oxide levels reflect smoke exposure within the last few days, and thio- Services, 2014). It also shows the year when each site was classified
cyanate (from hydrogen cyanide) within the past few weeks (Jarvis as causally related to smoking, and the estimated attributable fraction
et al., 1987). (AF) of deaths from active cigarette smoking in the United States.
Several studies suggest that measurement of urinary levels of the The number of cancer sites and/​or subsites classified as causally
tobacco-​specific carcinogens NNN and NNK and their metabolites related to smoking has increased since the earliest studies of smok-
may improve on estimates of carcinogen exposure from active or pas- ing and cancer (Doll, 1998; US Department of Health and Human
sive smoking when combined with cotinine and self-​reported smok- Services, 2014). Larger studies with longer follow-​up have identi-
ing (Goniewitz et al., 2011; Vardavas et al., 2013; Yuan et al., 2014). fied relationships with cancers that are less common or less strongly
However, these studies have been conducted in only a few populations associated with smoking than cancers of the lung, larynx, oral cavity,
198

198 PART III:  THE CAUSES OF CANCER


and esophagus. The identification of oncogenic infectious agents has (Chapter 27). Smoking was designated “a significant factor in cau-
allowed studies to control for confounding by these pathogens with sation” of laryngeal cancer among men in 1964 (US Department of
regard to cancers of the liver, stomach, and uterine cervix (IARC, Health Education and Welfare, 1964) and among women in 1980 (US
2004; US Department of Health and Human Services, 2014). Changes Department of Health and Human Services, 1980). The association
in cigarette design have strengthened the association between smok- between active cigarette smoking and death from laryngeal cancer
ing and adenocarcinoma of the lung (US Department of Health and appears to have strengthened in women but not men in the United
Human Services, 2014). Studies of tumor subsites have revealed causal States since the 1960s. The relative risk estimates for current ver-
relationships with mucinous ovarian cancer and with both adenocarci- sus never cigarette smoking were 14.6 in men and 13.0 in women in
noma and squamous cell carcinoma of the esophagus. The recognition Cancer Prevention Study I, a large American Cancer Society cohort
of additional smoking-​related cancers may also reflect the aging of followed from 1959 to 1966 (Hammond, 1966). The corresponding
birth cohorts with the heaviest lifetime exposure, especially in women relative risk estimates from a pooled analysis of five contemporary US
(Carter et al., 2015; Pirie et al., 2013; Thun et al., 2013). Undiscovered cohorts was 13.9 (CI: 8.3, 23.3) in men and 103.8 (CI: 24.2, 445.5) in
causal relationships may still exist with rare cancers, uncommon sub- women (Carter et al., 2015). The wide confidence interval for women
types, or molecularly defined subgroups (Carter et al., 2015). in the contemporary cohorts reflects the low risk of death from laryn-
geal cancer among female never smokers (only two deaths from this
cancer observed in female never smokers). It is important to note that,
Cigarette Smoking and Respiratory Tract Cancers although the relative risks of cigarette smoking for laryngeal cancer
tend to be higher in women than in men, the absolute number of smok-
The respiratory epithelium is heavily exposed to cigarette smoke. ing-​attributable deaths from laryngeal cancer annually in the United
Cancers of the lung and larynx were the first to be designated as States is far higher in men (2125) than in women (730), reflecting the
causally related to active smoking in men (US Department of Health particularly low incidence of laryngeal cancer in non-​smoking women
Education and Welfare, 1964). The associations between active ciga- (Siegel et al., 2015). Variations in the strength of the association by
rette smoking and cancers of the lung and larynx are the strongest of tumor subsite, gender, age, type of tobacco, and time since cessation
all sites—​an estimated 77% of laryngeal cancer and 80% of lung can- are discussed further in Chapter 27. Siegel et al. estimate that 77% of
cers in the United States are attributed smoking (Siegel et al., 2015). deaths from laryngeal cancer in the United States are attributable to
cigarette smoking (Siegel et al., 2015). The combination of tobacco
Nasal Cavity and Paranasal Sinuses.  The evidence linking smoking and heavy alcohol consumption creates much higher risk of
active cigarette smoking to cancers of the nasal cavity and paranasal laryngeal as well as other aerodigestive tract cancers than does smok-
sinuses was designated as causal by the IARC in 2004 (IARC, 2004). ing alone (Hashibe et al., 2009), as discussed below.
Sinonasal cancers are rare; thus the evidence comes mainly from case-​
control studies. The nasal cavity and paranasal sinuses are exposed to Trachea, Bronchus, and Lung. Cigarette smoking is more
mainstream tobacco smoke only during exhalation and only for a sub- strongly associated with lung cancer than with any other cancer site
set of smokers. Evidence for causality includes the presence of dose–​ (Table 11–​3). All histologic subtypes of lung cancer are now strongly
response relationships in most studies, the decrease in relative risk associated with cigarette smoking, whereas during the 1950s, adeno-
observed with time since quitting, the absence of likely confounders, carcinoma was reported to be minimally associated (Chapter 28) (Doll
and the stronger relationship observed for squamous cell cancers than et al., 1957; Kreyberg, 1962; Wynder and Hoffmann, 1994). A recent
for adenocarcinomas (IARC, 2004). Active cigarette smoking approxi- US Surgeon General Report attributed this increase to changes in the
mately doubles the incidence of nasal cavity squamous cell carcino- design and composition of cigarettes (US Department of Health and
mas in case-​control studies conducted in North America, Europe, Human Services, 2014). The Report identified the two most plausible
Japan, and China (IARC, 2004; US Department of Health and Human explanations for the increased association with adenocarcinoma of the
Services, 2004). Prospective studies of these sites are few, although lung as the increase in TSNA concentrations in smoke from recon-
consistent associations were observed in the large NIH-​AARP cohort stituted tobacco and the shift in inhalation patterns due to ventilated
(Freedman et al., 2016). cigarettes. The evidence for any specific explanation was characterized
as suggestive rather than definite.
Larynx.  Cancer of the larynx is more strongly associated with active The association between active cigarette smoking and death from
cigarette smoking than any other cancer except lung (Table 11–​3) lung cancer has strengthened over the last 50 years in the United States

Table 11–​3. Smoking-​Attributable Mortality from Cancer, 2010–​2014

Attributable Deaths
Relative Risk for Current
vs. Never Smoking % No.
Year Formally
Cancer Type Classified Men Women Men Women Men Women

Lip, oral cavity, pharynx 1964/​1971* 5.7 5.6 48.7 43.0 2955 1077
Esophagus 1982 3.9 5.1 52.3 44.4 6011 1296
Stomach 2004 1.9 1.7 25.6 10.8 1656 476
Colorectum 2014 1.4 1.6 11.2 8.0 2976 1992
Liver 2014 2.3 1.8 28.1 14.1 4085 975
Pancreas 1982 1.6 1.9 9.9 14.2 1870 2626
Larynx 1964 13.9 103.8 72.1 93.4 2125 730
Trachea, lung, bronchus 1964/​1968* 25.3 22.9 83.2 76.4 72,164 53,635
Cervix uteri 2004 N.A.-​ 3.5 N.A.-​ 22.2 -​-​-​ 862
Urinary bladder 1979 3.9 3.9 46.5 40.9 4920 1804
Kidney, other urinary tract 1982 1.8 1.2 22.3 7.2 1904 350
Acute myeloid leukemia 2004 1.9 1.1 23.2 3.4 1181 136

Source: Modified from US Surgeon General report 2014; Carter et al., NEJM 2015; Siegel et al. (JAMA Internal Medicine, 2015).
*Lip cancer was classified as causal in 1964, other oropharyngeal cancers in 1971. Lung cancer was classified as causal in men in 1964 and in women in 1968.
 19

Tobacco 199
and the United Kingdom, following birth cohort patterns in smoking oropharyngeal cancers should separate them into etiologically relevant
(Pirie et al., 2013; Thun et al., 2013). In men, the relative risk for death subsites.
from lung cancer in current versus never smokers increased from
approximately 12 in the 1960s to about 24 in the 1980s and then pla- Nasopharynx.  Active smoking was first classified as causally
teaued (Thun et al., 2013). The corresponding relative risk for female related to nasopharyngeal cancer by the IARC in 2004 (IARC, 2004).
smokers increased from less than 3 in the 1960s to almost 26 in the A meta-​analysis of 28 case-​control studies and four prospective cohort
contemporary cohorts (2000–​2010). In both sexes, the relative risk studies published between 1979 and 2011 reported a meta-​relative risk
estimates for long-​term smokers of 40+ cigarettes per day approached estimate of 1.60 (95% CI: 1.38, 1.87) for ever versus never smoking,
50. Projections based on age and birth cohort patterns suggest that with higher risks associated with greater intensity and longer duration
the average risk will continue to increase among older female smok- of smoking (Xue et al., 2013). Other studies and heterogeneity by cell
ers in the United States for several decades (Thun et al., 2013; Pirie type and geographic population are discussed further in Chapter 26.
et al., 2013).
Active cigarette smoking is estimated to account for approxi- Esophagus.  Active cigarette smoking was designated as a major
mately 80% (CI:  79.2, 81.1) percent of lung cancer deaths in the cause of esophageal cancer by the US Surgeon General in 1982
US general population (Siegel et al., 2015), and 96% among current and by the IARC in 1986 (IARC, 1986; US Department of Health
smokers, based on updated hazard ratio estimates (Thun et al., 2013). and Human Services, 1982). Historically, only cancers of the lung
Incidence and mortality rates and the relative risk estimates associ- and larynx were more strongly associated with active smoking than
ated with smoking have been shown to vary by ethnicity, with the esophageal cancer (Vineis et al., 2004). Active cigarette smoking is
highest rates in the United States found among blacks (Torre et al., causally related to both esophageal squamous cell carcinoma and
2016). Several factors are thought to contribute to these dispari- adenocarcinoma, the two main histologic subtypes (IARC, 2004).
ties, as reviewed in Thun and Henley (2006). Menthol cigarettes are The association with squamous cell carcinoma is stronger (RR 5.63;
smoked more commonly by black smokers than other racial or ethnic 95% CI: 2.7, 11.7) than that with adenocarcinoma (RR 2.77; 95%
groups, but epidemiological studies have not found that these confer CI: 1.4, 5.6) in a pooled analysis of smokers with > 60 pack-​years
higher risk for lung cancer than non-​menthol cigarettes (Blot et al., in the Beacon Consortium (Lubin et al., 2012). Alcohol consump-
2011; Brooks et al., 2003; Rostron, 2012). The association between tion strongly potentiates the relationship of smoking to esophageal
current cigarette smoking and lung cancer is considerably weaker squamous cell carcinoma but not adenocarcinoma (see Chapter 30).
in Japan and China; it is not yet clear whether this reflects the more The overall association between cigarette smoking and esophageal
recent uptake of manufactured cigarettes in these countries than in cancer appears to have decreased in high-​income countries, coinci-
the West, less intensive smoking, or, in the case of Japan, the use of dent with the shift in cell type toward a larger proportion of adeno-
charcoal filters. carcinoma. Whereas the relative risk estimates in Cancer Prevention
Study I  during follow-​up from 1959 to 1966 were 6.76 for men
Cigarette Smoking and Gastrointestinal Cancers and 7.75 for women (Hammond, 1966), they had decreased to 3.9
(CI: 3.0, 5.0) and 5.1 (CI: 3.5, 7.4) in men and women, respectively,
Cigarette smoking is causally associated with cancer at all sites in the in a pooled analysis of five large contemporary cohorts followed
gastrointestinal tract except the salivary glands and possibly the small from 2000 to 2010 (Carter et al., 2015). Active cigarette smoking is
intestine (IARC, 2012a). It is also causally associated with cancer of estimated to account for about half (50.7%, range 44.8% to 56.5%)
the pancreas and liver. Among the gastrointestinal tract cancers, the of deaths from esophageal cancer in the United States (Siegel et al.,
association with smoking becomes progressively weaker from the oral 2015). Greater detail about the association between cigarette smok-
cavity to the colorectum (Table 11–​3). The interaction between smok- ing and esophageal cancer is provided in Chapter 30.
ing and alcohol is also strongest for squamous cell cancers of the upper
aerodigestive tract (Hashibe, 2010). Stomach.  The IARC and the US Surgeon General first desig-
nated the evidence linking active smoking to stomach cancer as
Oral Cavity, Oropharynx, Lip, and Salivary Glands.  Active causal in 2004 (IARC, 2004; US Department of Health and Human
cigarette smoking was first designated as causally related to cancers of Services, 2014). The relative risk estimates for current versus never
the oral cavity and pharynx by the US Surgeon General in 1982 (US smokers average approximately 1.6 for men and women combined
Department of Health and Human Services, 1982) and by the IARC in more than 20 cohort studies and nearly 40 case-​control studies
in 1986 (IARC, 1986). Historically, many studies have combined can- (IARC, 2004). As described in Chapter  31, the relationship is not
cers of the oral cavity with those of the oropharynx, even though these confounded by infection with Helicobacter pylori. However, case-​
are now recognized to have distinctive etiologic and clinical features. control studies stratified on H.  pylori seropositivity have reported
More recently, a pooled analysis of over 13,000 cases and 18,000 con- stronger associations in persons who are seropositive for H. pylori
trols reported similar relative risk estimates for ever smoking with oral than in uninfected individuals (Brenner et al., 2002; Jedrychowski
cavity cancer (RR 2.87; 95% CI: 2.60, 3.18) and oropharyngeal cancer et al., 1993, 1999; Siman et al., 2001; Wu et al., 2003; Zaridze et al.,
(RR 3.01; 95% CI: 2.71, 3.35) (Wyss et al., 2013). Unlike other sites, 2000). Levels of DNA methylation also increase with pack-​years
cancers of the salivary gland have not been shown to be associated of smoking only among H.  pylori–​infected individuals (Shimazu
with cigarette smoking. et  al., 2015). Cigarette smoking increases the risk of cancers
The etiology of cancers of the oral cavity and oropharynx differ throughout the stomach, including cardia and non-​cardia (IARC,
substantially with regard to human papillomavirus (HPV) infection. 2004; Ladeiras-​Lopes et  al., 2008). No studies have yet examined
Whereas HPV is a strong risk factor for cancers of the oropharynx and associations between smoking and four recently identified molec-
base of the tongue, it is minimally associated with cancers of the oral ularly defined subtypes of gastric cancer (Cancer Genome Atlas
cavity. Studies have not yet fully elucidated whether cigarette smok- Research, 2014a). Siegel et al. estimate that 19.6% (13.8%–​26.8%)
ing interacts with HPV infection for cancers of the oropharynx. Active of deaths from stomach cancer among men and women combined
cigarette smoking is estimated to account for almost half (47.0%; in the United States are attributable to cigarette smoking (Siegel
CI: 38.6, 55.5) of deaths from cancers of the oral cavity and pharynx et al., 2015).
in the United States (Siegel et al., 2015). This attributable fraction esti-
mate has not decreased, despite decreases in smoking prevalence and a Colorectum.  Smoking was not associated with colorectal cancer
large increase in HPV-​related oropharyngeal cancers (see Chapter 29) in large cohort mortality studies conducted in the mid-​twentieth cen-
(Chaturvedi et al., 2008, 2013). The high fraction of cases attributed to tury (Doll and Hill, 1964; Hammond, 1966; Kahn, 1966; Weir and
smoking may increasingly represent an interaction between smoking Dunn, 1970). Beginning in the 1980s, studies consistently reported
and HPV, rather than smoking alone. Future studies of oral cavity and associations between smoking and colorectal adenomatous polyps
20

200 PART III:  THE CAUSES OF CANCER


(IARC, 2004). In the mid-​1990s, Giovannucci and colleagues reported Kuper HE et al., 2000; Marrero et al., 2005), raising concerns about
increased frequency of adenomatous polyps and colorectal can- the future burden of liver cancer in low-​and middle-​income countries
cer among smokers in the Harvard Nurses and Health Professionals with continued high prevalence of viral hepatitis and increasing smok-
cohorts (Giovannucci et  al., 1994a, 1994b). They hypothesized that ing prevalence.
smoking affected an early stage of colorectal neoplasia, and that a long
induction period might be needed to observe an association with inci- Cigarette Smoking and Urinary Tract Cancers
dence or mortality rates.
The relationship between active smoking and large bowel cancer Active cigarette smoking is a well-​established risk factor for cancers
was designated causal by the IARC in 2012 (IARC, 2012a) and by the throughout the urinary tract, including the kidney (parenchyma and
US Surgeon General in 2014 (US Department of Health and Human pelvis), ureter, and bladder. The association of these sites with cig-
Services, 2014). The association between cigarette smoking and colo- arette smoking has long been classified as causal (IARC, 1986; US
rectal cancer is weaker than that for most other smoking-​related can- Department of Health and Human Services, 2004). Current smoking
cers (Table 11–​3); it is also weaker for cancer of the colon (RR = 1.2; is more strongly associated with urothelial (formerly known as tran-
95% CI: 1.1, 1.3) than rectum (RR = 1.6; 95% CI:1.3, 1.8) (Botteri sitional) carcinomas of the renal pelvis (RR > 3) than with tumors of
et  al., 2008). The IARC based its conclusion on the consistent rela- the renal parenchyma (RR about 1.3), as discussed in the following
tionship between smoking and adenomatous colorectal polyps and the and in Chapter 51.
results of four meta-​analyses that controlled for multiple covariates
(Botteri et al., 2008; Huxley et al., 2009; Liang et al., 2009; Tsoi et al., Renal Adenocarcinoma. Active cigarette smoking has been
2009). The 2014 US Surgeon General report reviewed 19 high-​quality designated a cause of renal cell carcinoma by both the IARC and the
prospective studies of smoking in relation to incident colorectal cancer, US Surgeon General (IARC, 2012a; US Department of Health and
9 prospective cohort studies of cancer mortality, and 16 case-​control Human Services, 2014). Relative risk estimates from a recent meta-​
studies, published during 2002–​2009, plus the meta-​analyses men- analysis indicate that, compared to never smokers, risk is approxi-
tioned earlier (US Department of Health and Human Services, 2014). mately 36% higher in current smokers and 16% higher among former
Nearly all studies reported the strongest associations with adenoma- smokers (Cumberbatch et al., 2016).
tous polyps and colorectal cancer in current smokers and intermediate The classification of renal cell tumors has evolved substantially over
associations in former smokers. The associations are stronger among the last 30 years to incorporate genetic and molecular tumor charac-
smokers with early age at initiation and longer duration of smoking. teristics (Chapter 51). The current classification scheme recognizes 15
Intriguingly, several studies have suggested stronger associations distinct entities and 4 subtypes (Srigley et al., 2013). Most of the avail-
between cigarette smoking and the subset of colorectal tumors with able data on cigarette smoking pertain to clear cell tumors, which com-
microsatellite instability (MSI) and CIMP-​or BRAF-​positive tumors prise over 75% of renal cell carcinomas. Relatively few studies have
(Campbell et al., 2009; Chia et al., 2006; Limsui et al., 2010; Poynter examined associations by histological subtype. These report associa-
et al., 2009). These findings may point to the carcinogenic mechanism tions with clear cell and papillary type, but not with the rarer chromo-
underlying the association. Such relationships, as well as the impor- phobe type (Patel et al., 2015; Purdue et al., 2013). Larger studies are
tance of early life exposure, are discussed in Chapter 36. needed to characterize variations in the association between smoking
and histologic and molecular subtypes of renal cell cancer.
Pancreas.  As described in Chapter  32, the association between
active cigarette smoking and pancreatic cancer was designated as Renal Pelvis and Ureter. Active cigarette smoking is firmly
causal by the US Surgeon General in 1982 (US Department of Health established as the major cause of urothelial cancers of the renal pel-
and Human Services, 1982) and by the IARC in 1986 (IARC, 1986). vis and ureter. Relative risk estimates exceed 3.0 in most studies
A recent pooled analysis of five cohorts in the United States reported (McLaughlin et al., 1992), including the large prospective NIH-​AARP
relative risk estimates of 1.6 in men and 1.9 in women for current cohort (Freedman et al., 2016). The estimates of attributable fraction
versus never cigarette smokers (Carter et  al., 2015). Relative risk is are 50% or higher in many parts of the world.
lower for former than for current smokers and decreases with earlier
age at cessation (Iodice et al., 2008; Lynch et al., 2009). As noted in Urinary Bladder.  The association between active cigarette smok-
Chapter 32, the decrease in pancreatic incidence and mortality rates ing and cancer of the urinary bladder was first designated as causal by
that occurred among US whites in the late twentieth century, following the IARC in 1986 (IARC, 1986) and by the US Surgeon General in
reductions in smoking, reversed after the year 2000, following large 1979 (US Department of Health and Human Services, 1979). Cigarette
increases in the prevalence of obesity and type 2 diabetes. Recent esti- smoking is the leading cause of bladder cancer in most countries (see
mates indicate that active smoking accounts for 10%–​14% of pancre- Chapter 52). As is the case for lung and laryngeal cancers, the asso-
atic cancer in the United States (Siegel et al., 2015; US Department ciation between smoking and bladder cancer has increased over the
of Health and Human Services, 2014). Further information about the last few decades. The relative risk in current compared to never smok-
association of active smoking with pancreatic cancer, including poten- ers increased from about 3 during the 1990s to approximately 4–​5 in
tial molecular mechanisms, can be found in Chapter 32. the last decade (Baris et al., 2009; Freedman et al., 2011; Purdue and
Silverman, 2016). The relative risk and attributable fraction estimates
Liver.  The IARC designated the relationship between active smok- have historically been higher among men than women (IARC, 2004),
ing and liver cancer as causal in 2004 (IARC, 2004), as did the US but have converged over time in the United States. The attributable
Surgeon General in 2014 (US Department of Health and Human fraction was estimated to be approximately 50% in both men and
Services, 2014). The latter reviewed 113 studies published through women in the NIH-​AARP cohort (Freedman et al., 2011).
2012, with particular attention to potential confounding by viral hepa- Metabolites of heterocyclic aromatic amines, polycyclic aromatic
titis and alcohol consumption (US Department of Health and Human hydrocarbons, and other carcinogens in tobacco are excreted in urine
Services, 2014). Meta-​analyses found comparable associations in 31 (IARC, 2004; US Department of Health and Human Services, 2010).
studies that allowed comparisons between current and never smok- Studies have investigated interactions between cigarette smoking and
ers (mRR = 1.7; CI: 1.5, 1.9) and in a subset of nine case-​control and inherited variants in metabolizing genes and have documented differ-
four cohort studies in which subjects had no evidence of viral hepatitis ences in risk associated with loci that affect N-​acetyltransferase (NAT2)
(mRR = 1.8; CI: 1.2, 2.7). Other supporting evidence for causality is activity (Garcia-​Closas et al., 2013; Kilfoy et al., 2010). These stud-
discussed in Chapter 33. ies, in combination with functional genomic analyses, have provided
Active smoking has been suggested to interact with other liver can- important clues to bladder carcinogenesis (Chapter 52). Emerging data
cer risk factors, including viral hepatitis and consumption of alco- on the somatic mutations in bladder cancer from The Cancer Genome
hol (Chuang et al., 2010; Hassan et al., 2008; Kuper H et al., 2000; Atlas (TCGA) and similar projects (Cancer Genome Atlas Research,
 201

Tobacco 201
2014b) indicate that there are distinct molecularly defined subtypes of Fewer studies have examined associations with other myeloid neo-
bladder cancer. Future studies that examine associations between ciga- plasms, and the results have been mixed (Chapter 38). Existing stud-
rette smoking and molecularly defined subtypes will likely provide ies have been limited, however, by the uncommon nature of many
further insights into carcinogenic mechanisms. types and subtypes of myeloid neoplasms and changing classification
schemes over time.
Cigarette Smoking and Reproductive Tract Cancers

The evidence that cigarette smoking causes cervical cancer and muci- Cancer Sites with Suggestive or Limited Evidence
nous carcinoma of the ovary is designated as “sufficient” and is dis-
cussed here. The evidence that active smoking causes cancers of the Breast
breast, vulva and vagina, and aggressive prostate cancer, or reduces The question of whether a causal relationship exists between breast
the risk of endometrial cancer, are considered “suggestive” and are cancer and exposure to tobacco smoke has been controversial for
discussed later. over 20 years (Chapter 45). The 2014 US Surgeon General Report
comprehensively reviewed 15 cohort and 34 case-​control studies pub-
Uterine Cervix.  Cancer of the uterine cervix is consistently associ- lished between 2000 and 2012 (US Department of Health and Human
ated with cigarette smoking. Not until HPV infection was identified as a Services, 2014), as well as studies previously considered by the IARC
necessary cause of cervical cancer, however, were concerns about con- (IARC, 2004, 2012a) and the US Surgeon General (US Department of
founding by sexually transmitted diseases addressed directly (IARC, Health and Human Services, 2004). The 2014 Report devoted more
1995)  (Chapter  48). Later studies investigated cigarette smoking as than 160 pages of tables and text to this issue. Based on the data then
a cofactor for cervical cancer in HPV-​positive women and observed available, it concluded that the evidence regarding active smoking as
consistent associations with both invasive squamous cell cancer and a cause of breast cancer was “suggestive but not sufficient to infer a
carcinoma in situ (Castellsague and Munoz, 2003; International causal relationship” (US Department of Health and Human Services,
Collaboration of Epidemiological Studies of Cervical Cancer et  al., 2014). There was concern about the potential for residual confound-
2006). The relative risk for squamous cell carcinoma increases in cur- ing by alcohol consumption and screening and inconsistencies regard-
rent compared to never smokers with the number of cigarettes smoked ing the timing of exposure. An additional eight large cohort studies
per day and with younger age at starting smoking. No association is and meta-​analyses have been published on smoking and breast can-
observed between smoking and adenocarcinoma of the cervix, how- cer since the Surgeon General Report (Bjerkaas et al., 2013; Catsburg
ever. The IARC and the US Surgeon General designated the overall et al., 2015; Dossus et al., 2014; Gaudet et al., 2013; Gram et al., 2015,
relationship between active cigarette smoking and cervical cancer as 2016; Nyante et al., 2014; Rosenberg et al., 2013). These consistently
causal in 2004 (IARC, 2004; US Department of Health and Human showed a 10%–​20% higher risk of breast cancer in women who are
Services, 2004). current compared to never smokers and a somewhat stronger associa-
tion in women who initiated smoking before first birth. Given the sub-
Ovary.  Well over 30 epidemiologic studies have investigated the stantial public health importance of the issue, the evidence should be
association of active cigarette smoking with ovarian cancer (IARC, periodically reviewed by expert panels to identify and resolve residual
2012a). Most reported no overall association, although at least four controversies.
studies have reported a positive association between active cigarette
smoking and mucinous ovarian cancer (Collaborative Group on Advanced Stage Prostate Cancer
Epidemiological Studies of Ovarian Cancer et al., 2012; Faber et al., Many studies have investigated associations between active cigarette
2013; Gram et al., 2012; Wentzensen et al., 2016). Smoking has not smoking and prostate cancer (Chapter 53). The relationship is compli-
been associated with serous or endometrioid subtypes of ovarian can- cated by potential differences in PSA screening and healthcare utilization
cer; an inverse association has been reported for clear cell tumors among smokers and non-​smokers. For incident cancer, a large meta-​
(Wentzensen et al, 2016). Etiologic heterogeneity among subtypes of analysis of more than 50,000 incident cases found an inverse association
ovarian cancer has also been observed for other established risk factors comparing current to never smokers (RR = 0.90; 95% CI: 0.85, 0.96).
besides smoking (Wentzensen et al., 2016). This association was limited to studies completed after 1995, however,
when PSA screening became widely adopted (Islami et al., 2014).
Cigarette Smoking and Hematopoietic Cancers In contrast, studies of advanced stage or aggressive prostate can-
cer have tended to find positive associations with smoking (Zu and
As part of the 2001 WHO classification of hematopoietic and lym- Giovannucci, 2009), as have studies of prostate cancer mortality.
phoid neoplasms, “the leukemias” were classified into two major Current cigarette smoking was associated with increased mortality
groupings:  myeloid neoplasms (including acute myeloid leukemia, from prostate cancer in a meta-​analysis of nearly 12,000 prostate
myelodysplastic syndromes, and myeloproliferative neoplasms) cancer deaths (RR:  1.24; 95% CI:  1.18, 1.31), with evidence for
(Chapter 38) and lymphoid neoplasms, including lymphoid leukemias a dose-​response relationship (Islami et  al., 2014). There remains
(circulating phase) and lymphomas (solid phase) (Chapter 40). uncertainly about whether this association is causal, however. In
Active cigarette smoking has consistently been associated with 2014, the US Surgeon General comprehensively reviewed the evi-
increased risk of acute myeloid leukemia (AML), as discussed here, dence and concluded that the available data were suggestive of no
but not other hematological or lymphoid malignancies, discussed later causal relationship with incident prostate cancer but higher risk
in the chapter. of prostate cancer death (US Department of Health and Human
Services, 2014). Future epidemiologic and mechanistic research is
Myeloid Leukemia. The association between active cigarette certainly warranted.
smoking and increased risk of AML was designated as causal by
both the US Surgeon General and the IARC in 2004. The relative Vulva and Vagina
risk for current versus never smoking was 1.40 (95% CI: 1.22,1.60) Cancers of the vulva and vagina are rare, but active cigarette smoking
in a recent meta-​analysis of 23 studies and 7746 cases (Fircanis et al., is associated with increased risk of both sites in the limited number
2014). Accumulating evidence also suggests that there is also a posi- of studies on this issue (Chen et al., 1999; Daling et al., 1992, 2002;
tive association between smoking and the preleukemic myelodysplas- Hussain et  al., 2008; Madeleine et  al., 1997). A  strong interaction
tic syndromes. For example, the relative risk for current versus never between cigarette smoking and HPV 16 on the risk of vulvar cancer
smoking was 1.81 (95% CI: 1.24, 2.66) in a 2013 meta-​analysis of 14 was reported by Madeleine et al. (1997). More work is needed to char-
studies and 2588 cases, with evidence for a dose-​response relationship acterize the combined effect of smoking and HPV infection on these
(Tong et al., 2013). cancers with respect to the timing of exposure (Chapter 49).
20

202 PART III:  THE CAUSES OF CANCER

Anus Cancer Sites with Limited Evidence or No Association


Cancer of the anus, a malignancy with squamous or transitional cell
histology, has been repeatedly associated with cigarette smoking, Small Intestine
although confounding by HPV has not been excluded (Chapter  37). Relatively few studies have examined active smoking in relation to
Future studies with careful control of confounding are needed to cancers of the small intestine, due to the rarity and heterogeneity of
resolve the issue. these cancers. However, recent studies do not suggest an association
with either adenocarcinoma or carcinoid tumors, which make up the
Biliary Tract two main histological types of small intestinal cancers (Chapter 35).
Studies have investigated associations between active smoking and
cancers of the biliary tract, including gallbladder, extrahepatic bile Testes
duct, and ampulla of Vater (Chapter  34). Neither the IARC nor the There is limited published evidence on cigarette smoking and cancer
US Surgeon General has classified biliary tract cancer as being caus- of the testes (Brown et  al., 1987; Gallagher et  al., 1995; Henderson
ally related to tobacco smoking. However, a recent meta-​analysis of et al., 1979), although three relatively recent studies have found asso-
11 studies reported an association between ever smoking and gallblad- ciations with marijuana smoking (Daling et  al., 2009; Lacson et  al.,
der cancer (summary RR = 1.45; 95% CI: 1.11, 1.89) (Wenbin et al., 2012; Trabert et al., 2011) (Chapter 54).
2013). A separate meta-​analysis also found evidence for an associa-
tion with extrahepatic bile duct cancers (summary RR  =  1.23; 95% Penis
CI: 1.01, 1.50) (Ye et al., 2013). Several case-​control studies have examined the association of active
cigarette smoking with penile cancer (Chapter 55); however, the results
Keratinocyte Carcinomas to date are inconsistent, and confounding by HPV status is a concern.
Keratinocyte cancers are the most common malignancies in humans.
These tumors, which include basal and squamous cell carcinomas, Non-​Hodgkin Lymphoma
arise from keratinocytes or their precursor cells (Chapter 58). Results Numerous studies have investigated associations of active smoking
from case-​control and cohort studies generally suggest an associa- with non-​Hodgkin lymphomas (NHL), a heterogeneous group of over
tion between active cigarette smoking and squamous cell carcinomas. 40 lymphoid neoplasms (Chapter 40). Overall, there appears to be no
A meta-​analysis in 2012 observed a relative risk of 1.54 (95% CI: 1.03, association, although associations with certain subtypes have been
2.31) comparing current to never smokers (Leonardi-​Bee et al., 2012). noted (Morton et al., 2014).
In contrast, active smoking does not appear to increase risk of basal
cell carcinoma (0.89; 95% CI: 0.77, 1.03). Hodgkin Lymphoma
Active cigarette smoking is not generally considered to be a risk fac-
tor for Hodgkin lymphoma. However, results from a pooled analysis
Cancer Sites Inversely Associated with Cigarette from 12 case-​control studies in the InterLymph consortium (Kamper-​
Smoking Jorgensen et  al., 2013), as well as separate meta-​analyses, provide
evidence for a positive association (Castillo et al., 2011; Sergentanis
Endometrium et al., 2013), at least for some subtypes (Chapter 39).
The incidence of endometrial cancer is inversely associated with
active cigarette smoking (IARC, 2004; US Department of Health Multiple Myeloma
and Human Services, 2004). The relationship varies by smoking There is no evidence that smoking causes multiple myeloma
status; risk was 30%–​40% lower in current smokers and 9%–​12% (Chapter 41).
lower among former compared to never smokers in an analysis of
the National Institutes of Health-​AARP Diet and Health Study (Felix Nervous System
et al., 2014). The association is also stronger in postmenopausal than Most studies have found no association with smoking and glioma or
premenopausal women (Zhou et al., 2008), but has not been exam- other tumors of the nervous system (Chapter 56).
ined in relation to tumor subtypes or molecular markers. A number of
mechanisms have been hypothesized, including effects of smoking on Soft Tissue Sarcomas
circulating hormone levels (discussed in Chapter 47 for endometrial The data regarding cigarette smoking and soft-​tissue sarcoma are lim-
cancer and in Chapter  45 for breast cancer, as well as in the 2014 ited and mixed (Franceschi and Serraino, 1992; Serraino et al., 1991;
US Surgeon General’s report) (US Department of Health and Human Zahm et al., 1992) (Chapter 43).
Services, 2014).

Thyroid Fraction of Cancer Deaths Attributed


Data from both case-​control and cohort studies suggest an inverse to Cigarette Smoking
association between active cigarette smoking and thyroid cancer
(Chapter 44). For example, the relative risks for current versus never Estimates of the fraction of all cancer deaths attributable to tobacco
smoking was 0.68 (95% CI: 0.55, 0.85) in a pooled analysis of five use have changed little over the past 30–​40  years. In 1981, Doll
prospective US cohort studies (Wiersinga, 2013). The mechanisms and Peto estimated that 30% (acceptable range 25%–​40%) of cancer
that underlie this association are unclear. deaths in the United States could be attributed to tobacco smok-
ing (Doll and Peto, 1981). This estimate has persisted, despite
Melanoma reductions in smoking prevalence and consumption, partly because
Both case-​control and cohort studies have examined the association additional cancer sites have been designated as causally related to
between active cigarette smoking and melanoma (Chapter 57). Most smoking (US Department of Health and Human Services, 2014),
studies have reported inverse associations. A  2015 meta-​analysis of and partly because of the delayed effect of birth cohort patterns of
23 studies observed pooled relative risks of 0.70 (95% CI: 0.63, 0.78) smoking on cancer risk. Jacobs et  al. recently estimated that the
for current smoking and 0.90 (95% CI: 0.85, 0.95) for former smok- population attributable fraction (PAF) for deaths from all cancers
ing (Li et al., 2015). The mechanisms underlying this association are combined was 28.7% when estimated conservatively, including only
unclear. Non-​causal explanations, including confounding by sunlight deaths from the 12 cancers currently formally designated as caus-
and other risk factors, selection bias, competing risks, and publica- ally related to smoking by the US Surgeon General, but that the PAF
tion bias, have been suggested (Freedman et al., 2003; Li et al., 2015; was 31.7% when estimated more comprehensively, including excess
Thompson et al., 2013). deaths from all cancers (Jacobs et al., 2015). The PAF also varies by
 203

Tobacco 203
state and region, reflecting the levels of tobacco use and control. In cancer were reviewed by the IARC (IARC, 2012a). Additional cohort
a recent state-​wide analysis, the PAF estimates for active smoking and case control studies from India have reported associations between
ranged from 16.6% in Utah to 34% in Kentucky (Lortet-​Tieulent bidi smoking and cancers of the oral cavity (Jayalekshmi et al., 2011;
et  al., 2016). Although substantial, these estimates do not include Pednekar et  al., 2011), hypopharynx and larynx (Jayalekshmi et  al.,
involuntary exposure to tobacco smoke or other types of tobacco 2013; Pednekar et al., 2011), lung (Pednekar et al., 2011), esophagus,
use such as cigars, pipes, or smokeless tobacco (Jacobs et  al., and stomach (Jayalekshmi et al., 2015).
2015). The attributable fraction is smaller for incident cancers than
for deaths from cancer, due to the large contribution of breast and
prostate cancer to incident cases. Parkin has estimated that smoking Kreteks, Waterpipes, and Chuttas
caused 60,000 new cancer cases in the United Kingdom in 2010 Kreteks, waterpipes, and chuttas are other smoked tobacco products
(19.4% of all new cases) (Parkin, 2011). The only global estimate for which the long-​term effects on health are not well characterized.
of smoking-​attributable cancer is by Jha, who estimated that 31% of Kreteks are clove-​flavored cigarettes, manufactured and widely con-
cancer deaths worldwide in men, and 6% in women, were attribut- sumed in Indonesia (Palipudi K et al., 2015). They contain eugenol,
able to smoking (Jha, 2009). a natural compound found in high concentration in clove buds, which
has an anesthetic effect (Tobacco Atlas, 2015). Waterpipes, also known
as “hookah,” “narghile,” and “shisha,” are commonly smoked in North
CANCER RISKS FROM OTHER TOBACCO PRODUCTS Africa, the Mediterranean, and parts of Asia. They have recently
been popularized among students in high-​income countries. A meta-​
Both the IARC and the US Surgeon General have concluded that all
analyses of 13 case-​control studies suggest increased risks for water-
forms of tobacco use are carcinogenic. However, the evidence based
pipe use with cancers of the lung, esophagus, and possibly other sites
on combustible products other than cigarettes and on non-​combustible
(Montazeri et al., 2017). However, most existing studies have method-
tobacco products in relation to cancer is less extensive than that for
ological limitations, and high-​quality studies with standardized expo-
cigarettes (IARC, 2012b).
sure measurements are needed.
Chuttas are coarse cheroots, infrequently smoked in South India
Other Combustible Products with the lighted end inside the mouth (reverse smoking). They have
been associated with carcinomas of the hard palate in a case series
(Reddy, 1974).
Pipes and Cigars
Pipes and cigars are the main non-​cigarette smoked tobacco products
in high-​and many middle-​income countries. Pipe smoking was desig- SMOKELESS TOBACCO PRODUCTS
nated causally related to lip cancer in 1964 (US Department of Health
Education and Welfare, 1964). Exclusive pipe smoking was associ- More than 300 million adults in 70 countries use smokeless tobacco
ated with increased mortality rates from four cancer sites among men (National Cancer Institute and Centers for Disease Control and
in an analysis of the CPS-​II cohort by Henley et al. (2004). Compared Prevention, 2014). The IARC has determined that smokeless
to never smokers, men who currently smoked pipes but reported no tobacco is causally related to cancers of the esophagus, oral cav-
history of using other tobacco products had increased mortality from ity, and pancreas (IARC, 2012a). The majority of consumers (89%)
cancer of the oral cavity/​pharynx (RR = 3.90; 95% CI: 2.15, 7.08), are in Southeast Asia, where these products are inexpensive, socially
larynx (RR = 13.1; 95% CI: 5.2, 33.1, lung (RR = 5.00; 95% CI: 4.16, acceptable, and readily available. In most countries, usage is more
6.01), and esophagus (RR  =  2.44; 95% CI:  1.51, 3.95), as well as common in men than women. In some countries, however (e.g.,
coronary heart disease, stroke, and chronic obstructive pulmonary Bangladesh, Thailand, Cambodia, Malaysia, Vietnam, and some
disease (Henley et al., 2004). The risks were generally smaller than African countries), use by adult women is similar to or greater than
those associated with cigarette smoking and similar to or larger than that by adult men.
those associated with cigar smoking. The chemical composition and levels of free nicotine vary widely
A separate analysis of men who currently and exclusively smoked among these products. The concentrations of tobacco-​specific carcino-
cigars in CPS-​II reported increased death rates from cancers of the gens (TSNAs) such as NNN and NNK can vary by several orders of
lung (RR = 5.1; 95% CI: 4.0, 6.6), oral cavity/​pharynx (RR = 4.0; 95% magnitude (National Cancer Institute and Centers for Disease Control
CI: 1.5, 10.3), larynx (RR = 10.3; 95% CI: 2.6,41.0), and esophagus and Prevention, 2014). Products that are widely used in low-​and
(RR  =  1.8; 95% CI:  0.9, 3.7) compared to never smokers (Shapiro middle-​income countries are usually handmade or produced by cottage
et al., 2000). industries, and are consequently less standardized than manufactured
Both cigar and pipe smokers had increased incidence of head smoked or smokeless tobacco products. Studies of these products in
and neck cancer in the International Head and Neck Cancer relation to cancer are complicated by their diversity and the frequent
Epidemiology (INHANCE) Consortium (Wyss et  al., 2013). The use of more than one product. They involve exposure to complex and
risk of head and neck cancer was elevated for those who reported varying mixtures of ingredients that may include other plant materi-
exclusive cigar smoking (odds ratio  =  3.49; 95% CI:  2.58, 4.73) als, such as areca nut and tonka bean, in addition to tobacco (National
or exclusive pipe smoking (odds ratio = 3.71; 95% CI: 2.59, 5.33) Cancer Institute and Centers for Disease Control and Prevention, 2014).
compared to never smokers. The associations with cancers of the
head and neck and esophagus are predominantly with squamous cell
carcinoma rather than adenocarcinoma (Chang et al., 2015; National Betel Quid
Cancer Institute, 1998).
The INHANCE study separately examined the association of pipe Betel quid is commonly chewed in India and throughout much of
and cigar smoking with oral cavity cancer, based on approximately Southeast Asia and the Western Pacific. As mentioned earlier, it con-
4100 cases. The odds ratio (OR) estimates for oral cavity cancer were tains a mixture of areca nut, betel leaf (Piper betle), and other ingredi-
2.51 (95% CI: 1.68, 3.75) for exclusive pipe smoking and 2.83 (95% ents, and can be made with or without tobacco. IARC Working Groups
CI: 1.91, 4.17) for cigar smoking, compared to the risk of never smok- have classified betel quid with tobacco as a Group 1 human carcino-
ers (Wyss et al., 2013). gen, based on cancers of the oral cavity (IARC, 2004) and squamous
carcinoma of the esophagus (IARC, 2012a). Areca nut is also classi-
Bidis fied as carcinogenic to humans. A  Taiwanese cohort study reported
As mentioned earlier, bidis are local tobacco products composed of associations between betel-​quid chewing and laryngeal cancer (Lee
coarse and uncured tobacco, generally smoked without filters by men et al., 2011), with consumption of > 20 quids daily associated with an
in India (IARC, 2012a). Epidemiologic studies of bidi smoking and adjusted hazard ratio of 1.7 (CI: 1.2, 2.6).
204

204 PART III:  THE CAUSES OF CANCER

Moist Snuff and Chewing Tobacco agents may foster transformation of premalignant abnormalities into
invasive cancer of the liver, stomach, uterine cervix, and/​or lung. The
In high-​income countries, the most commonly used traditional product evidence currently available regarding possible interactions between
is moist snuff, followed by chewing tobacco. Fermentation and aging tobacco use and diet is limited.
are used to modify the taste of both moist and dry snuff, but these
increase the concentration of TSNA. The IARC has designated moist
snuff as carcinogenic to humans, based on cancers of the oral cavity Occupational Exposures
and pancreas (IARC, 2007). In high-​income countries, moist snuff is
increasingly sold in teabag-​like sachets that reduce the need to spit. There are well established interactions between tobacco smoking and
A  moist snuff, low-​nitrosamine product called snus is manufactured several occupational exposures with respect to lung cancer. These
and widely used in Scandinavia. The levels of TSNA are much higher include asbestos and radon (National Research Council Committee
in commercial brands of moist snuff sold in the United States and in on Health Risks of Exposure to Radon, 1998). In general, the statis-
products sold in low-​and middle-​income countries than in snus. Until tical interaction between smoking and radon appears submultiplica-
recently, tobacco control and regulatory efforts have focused primarily tive, but without strong evidence against multiplicative interaction
on cigarettes, and have paid less attention to the marketing practices (IARC, 2004).
and chemical composition of smokeless products. The situation is fur-
ther complicated in low-​and middle-​income countries by the unorga-
nized nature of a large business sector, which impedes product control INVOLUNTARY EXPOSURE TO TOBACCO SMOKE
and regulation (National Cancer Institute and Centers for Disease
Control and Prevention, 2014). Lung Cancer
Many authoritative scientific groups have concluded that involuntary
Novel Smokeless Tobacco Products exposure to tobacco smoke causes lung cancer and coronary heart
disease in humans (Australian National Health and Medical Research
As mentioned previously, tobacco manufacturers have introduced Council, 1987; IARC, 1986, 2004, 2012a; National Research Council,
novel smokeless tobacco products using innovations such as por- 1986; US Environmental Protection Agency, 1992; US Department of
tion pouches, dissolvable tobacco, unique flavorings (such as fruit Health and Human Services, 1986, 2006, 2014). The association has
and candy flavors), electronic nicotine delivery systems (ENDS) and long been judged to be causal based on its biologic plausibility, replica-
varying nicotine levels, which may make products more attractive to tion in multiple different settings by different investigators using a vari-
consumers, including those who have not previously used tobacco ety of study designs, and supportive clinical information. Biomarker
products (National Cancer Institute and Centers for Disease Control studies indicate that non-​ smokers exposed to secondhand smoke
and Prevention, 2014). Among these, e-​cigarettes are by far the most absorb, metabolize, and excrete toxic constituents of tobacco smoke
popular. E-​cigarettes are overtly marketed to smokers as a source of (US Department of Health and Human Services, 2006). In fact, passive
nicotine in settings where they cannot smoke, and are informally posi- smokers appear to have greater excretion of a metabolite of a tobacco-​
tioned as cessation devices. They also are sold in a remarkable variety specific carcinogen than active smokers (Vogel et al., 2011). Studies of
of flavors, many of which appeal to youth. genotoxicity document a greater prevalence of DNA adducts and strand
The health effects of e-​cigarettes are at this point unclear. With their breaks in non-​ smokers exposed to secondhand smoke (Husgafvel-​
recent introduction to the marketplace, e-​cigarettes may have long-​ Pursiainen, 2004). The most recent comprehensive review by IARC
term effects that will not be fully apparent for many years. In addition (2012a) cited more than 50 epidemiologic studies and meta-​analyses
to directly affecting health, they can indirectly affect disease risks by published internationally since the first studies in Japan and Greece
modifying the use of other tobacco products. For example, adolescents reported increased lung cancer risk in non-​smoking women married to
may choose e-​cigarettes as an alternative to cigarette smoking, or they cigarette smokers (Hirayama, 1981; Trichopoulos et al., 1981). Despite
may become addicted to nicotine from e-​cigarettes and then prog- this evidence, the tobacco industry has long sought to maintain the
ress to cigarette smoking. Cigarette smokers may use e-​cigarettes to appearance of controversy, as discussed in Chapter 17.
facilitate quitting, or alternatively may use ENDS to supplement their
nicotine intake in settings where they cannot smoke. Any product that
facilitates continued smoking instead of quitting will increase disease Breast Cancer
risks. Even smokers who continue to smoke just a few cigarettes a day
have measurably higher risks of cancer and other chronic diseases than In contrast to the evidence on secondhand smoke and lung cancer,
those who quit (Inoue-​Choi et al., 2016). reviews of breast cancer in relation to involuntary exposure have
reached different conclusions (California Environmental Protection
Agency, 1997; IARC, 2004, 2012a; Johnson, 2005; Miller et al., 2007;
US Department of Health and Human Services, 2006). The IARC
INTERACTIONS WITH OTHER EXPOSURES and the US Surgeon General have variously described the evidence
as “inconsistent” (IARC, 2004)  or as “suggestive but not sufficient”
Alcohol to infer causality, whereas reviews by the California Environmental
Protection Agency in (California Environmental Protection Agency,
The combined effect of tobacco use and alcohol consumption has 2005) and by a panel of researchers convened in Canada (Collishaw
been examined extensively for cancers of the oral cavity, pharynx, lar- NE et  al., 2009)  designated the evidence for secondhand tobacco
ynx, and esophagus and to a lesser extent for cancers of the liver and smoke as “consistent with a causal association in younger primarily
pancreas (IARC, 2004). In the larger studies, cancer risk consistently premenopausal women.”
increased more rapidly with the combination of smoking and heavy Much of the supportive evidence comes from case-​control studies
drinking than with either exposure alone. Case-​control studies that that have attempted to collect full “lifetime exposure histories” of sec-
evaluated statistical interaction formally demonstrated a greater than ondhand smoke (Collishaw NE et al., 2009). These studies observed
multiplicative relationship with joint exposure. the strongest associations with breast cancer and were considered by
Collishaw et  al. to have the most complete information on lifetime
exposure to secondhand tobacco smoke from all sources. An important
Infectious Agents limitation of these studies, however, is that they are also most suscep-
Tobacco use is an established cofactor with human papillomavirus tible to recall bias, especially for exposures many years in the past,
infection for anogenital cancers (Chapter 24). There is intriguing, albeit during an era when exposure to secondhand smoke was ubiquitous
limited, evidence that tobacco use combined with certain infectious (IARC, 2012a).
 205

Tobacco 205
According to the IARC, the strongest support for the hypothesis comes of cessation appear more quickly than does the increase in cancer risk
from a cohort study in Japan (Hanaoka et al., 2005), which reported sig- after initiation, presumably because quitting removes late-​stage pro-
nificantly increased risk (RR 2.6; 95% CI: 1.3, 5.2) of premenopausal moting effects of smoking on carcinogenesis. The benefits of stopping
breast cancer in women who previously reported having ever lived with a smoking or other forms of tobacco use are best seen by comparing
regular smoker or ever being exposed to secondhand tobacco smoke for risk among smokers who quit at various ages with the risk of those
at least 1 hour per day in settings outside the home. The referent group who continue to smoke (Figure 11–​11). Smoking cessation at any age
in this analysis included only nine unexposed cases, however. A weak avoids much of the future risk seen with continued smoking. The rel-
association between secondhand tobacco smoke exposure and premeno- ative and absolute benefits are greatest when cessation occurs at an
pausal breast cancer was also reported in the California Teachers cohort, early age, but are substantial even when stopping occurs by age 50
when menopausal status was defined by age at diagnosis rather than age or 60 (Peto et al., 2000). The relative risks among persons who quit
at entry into the study (Reynolds et al., 2006). smoking compared with those who continue are progressively smaller
The absence of a strong and convincing relationship between breast the earlier the age of cessation and the longer the time that has elapsed
cancer and active smoking weakens the case for a causal relationship since cessation. Only in smokers who quit at younger ages does the
with secondhand smoke, given that the level of exposure is orders of relative risk of death from these cancers approach unity when com-
magnitude lower. Theories have been advanced to explain why sec- pared with that of persons who have never smoked.
ondhand tobacco smoke might have a stronger effect on breast cancer Analyses of cessation are more informative when based on the age
than active smoking (California Environmental Protection Agency, at cessation than time since quitting, since the relative and absolute
1997; Collishaw NE et  al., 2009; Johnson, 2005), these assume the benefits vary by age, and individual smokers can more easily relate
existence of a bidirectional effect of tobacco smoke on breast cancer their personal situation to age at quitting. It is also more informative to
risk, for which there is no direct evidence (IARC, 2012a). compare smokers who have quit smoking to those who continue than
to compare them with lifelong non-​smokers. Smokers have the option
to quit smoking but not to become lifelong non-​smokers.
Other Cancer Sites
The IARC 2012 review also comprehensively considered the epide-
FUTURE RESEARCH DIRECTIONS
miologic evidence regarding involuntary smoking and cancers of the
upper aerodigestive tract, gastrointestinal and genitourinary systems,
Despite progress in reducing cigarette smoking in many high-​and
brain, leukemias, lymphomas, and childhood cancers. The evidence
middle-​income countries and the immense literature that already exists
regarding these sites remains inadequate.
on the harmful health effects of tobacco use, continuing research is
needed to strengthen global tobacco control efforts and address unre-
OPPORTUNITIES FOR PREVENTION solved etiologic and mechanistic questions. The significant expansion
of efforts to collect nationally representative surveillance data (CDC,
Well-​established “best practices” in tobacco control are discussed 2016), especially in low-​and middle-​income countries, creates opportu-
in Chapter 62.1. The implementation of these can effectively reduce nities to monitor patterns of tobacco use in countries where the number
tobacco use and prevent the devastating effects of tobacco on health of smokers is expected to increase most rapidly over the next decade.
(US Department of Health and Human Services, 2014). Long-​term Studies that track the implementation of “best practices” for compre-
progress depends on the systematic application of primary prevention hensive tobacco control, as mandated by the Framework Convention on
measures that can reduce the initiation of tobacco use by young people Tobacco Control (FCTC) (WHO, 2003), are essential in countries at all
and end the pandemic during the second half of the twenty-​first cen- levels of economic development. Other types of application research,
tury. In the near term, substantial reductions in smoking-​attributable such as comparative effectiveness studies, can be used to tailor inter-
cancers and other diseases can be achieved by providing counseling ventions to specific populations and social contexts. Descriptive studies
and treatment to facilitate cessation among the 36.5 million Americans can characterize the temporal and geographic relationships between the
and others who currently smoke (Jamal et al., 2016). implementation of tobacco control measures and changes in knowl-
edge, attitudes, beliefs, and behaviors in populations. Assessment of
key indicators, such as tobacco use by medical providers and average
Increasing the Age at Initiation age at initiation, are especially informative in this regard.
The introduction of novel tobacco products, such as e-​cigarettes,
A growing public health emphasis has been placed on increasing the creates both challenges and opportunities for research. For example,
minimum age of tobacco purchase to age 21 (Winickoff et al., 2014). much larger randomized clinical trials are urgently needed to deter-
As described earlier, most cigarette smokers in economically devel- mine whether e-​cigarettes are effective for smoking cessation, and, if
oped countries begin smoking in their teenage years. Earlier age at so, to compare their efficacy to that of established cessation treatments.
initiation has been associated with increased addiction to nicotine and More studies are needed to characterize the impact of e-​cigarettes on
greater risk of nearly all smoking-​related diseases. Furthermore, the nicotine dependence, intermediate endpoints, and patterns of tobacco
main source of cigarettes for minors is from others under the age of usage in populations. Longitudinal studies are needed to determine
21 (DiFranza and Coleman, 2001). Adolescent brains have also been whether e-​cigarettes as currently marketed deter the uptake of ciga-
shown to be particularly sensitive to the addictive properties of nico- rette smoking in young people or simply addict them to nicotine.
tine (US Department of Health and Human Services, 2012). For these Continued surveillance research is needed to monitor the shifting
and other reasons, modeling studies, including those in a compre- patterns of tobacco use, trends in dual or poly use, and increased con-
hensive 2015 report by the Institute of Medicine, estimate that rais- sumption of roll-​your-​own cigarettes and small filter-​tip cigars to cir-
ing the cigarette smoking age to 21 years could substantially decrease cumvent the excise taxes on cigarettes. Research on marketing must
the prevalence of cigarette smoking in the population (Institute of increasingly monitor point-​of-​sale promotions (discounts, single ciga-
Medicine, 2015). At the time of this writing, at least 200 local and rettes) designed to counter tobacco control policies.
two state governments have implemented “tobacco 21” policies, and Important etiologic questions remain unresolved as well, such as
emerging data suggest that these may indeed affect teenage smoking whether active cigarette smoking is causally related to breast cancer
prevalence (Kessel Schneider et al., 2016). and aggressive prostate cancer. Larger studies that incorporate detailed
information on known risk factors for these cancers and screening
are needed to resolve these controversies. It is hypothesized, but not
Tobacco Cessation firmly established, that the initiation of smoking between menarche
Many of the adverse effects of tobacco use can be prevented or and the time of first childbirth confers increased susceptibility to pre-
reversed by cessation (IARC, 2004, 2012a). Paradoxically, the benefits menopausal breast cancer, and that tobacco smoke has a bidirectional
206

206 PART III:  THE CAUSES OF CANCER


effect on the metabolism of steroidal sex hormones (US Department of WHO Comprehensive Information Systems for Tobacco Control. Lancet,
Health and Human Services, 2014). The availability of more accurate 385(9972), 966–​976. PMID: 25784347.
high-​throughput measurement of sex hormones and their metabolites Bjerkaas E, Parajuli R, Weiderpass E, et  al. 2013. Smoking duration before
(Chapter 22) could be used to test the latter hypothesis. Similarly, mea- first childbirth: an emerging risk factor for breast cancer? Results from
302,865 Norwegian women. Cancer Causes Control, 24(7), 1347–​1356.
surements of harmful and potentially harmful constituents (HPHCs) PMCID: PMC3675272.
among users of different classes of tobacco products have the potential Blot WJ, Cohen SS, Aldrich M, et  al. 2011. Lung cancer risk among smok-
to refine our understanding of tobacco carcinogenesis and may con- ers of menthol cigarettes. J Natl Cancer Inst, 103(10), 810–​ 816.
tribute to regulation. PMCID: PMC3096798.
Studies based on histologic or molecular characteristics have already Boffetta P, Clark S, Shen M, et al. 2006. Serum cotinine level as predictor of
demonstrated associations with cigarette smoking for some tumor sub- lung cancer risk. Cancer Epidemiol Biomarkers Prev, 15(6), 1184–​1188.
types but not others. Broader efforts are needed to identify molecular PMID: 16775179.
subtypes associated with tobacco use across a range of cancer sites. Borgerding MF, Bodnar JA, Curtin GM, and Swauger JE. 2012. The chemi-
Advances in molecular and genomic technologies create opportunities cal composition of smokeless tobacco: a survey of products sold in the
United States in 2006 and 2007. Regul Toxicol Pharmacol, 64(3), 367–​
to study the effects of tobacco exposure on the genome and transcrip- 387. PMID: 23000415.
tome (methylation, mutational patterns, mosaicism, mRNA expres- Botteri E, Iodice S, Bagnardi V, et al. 2008. Smoking and colorectal cancer: a
sion). Already cigarette smoking has been demonstrated to cause meta-​analysis. JAMA, 300(23), 2765–​2778. PMID: 19088354.
wholesale genomic and epigenetic changes in users. Studies to assess Brandt AM. 2007. The Cigarette Century: The Rise, Fall, and Deadly Persistence
whether smoking affects later stage molecular events in prostate can- of the Product That Defined America. New York: Basic Books.
cer could provide mechanistic information to assess the causality of its Brandon TH, Goniewicz ML, Hanna NH, et al. 2015. Electronic nicotine
relationship with advanced disease. delivery systems: a policy statement from the American Association for
Finally, there is a continuing need to assess the usage and disease Cancer Research and the American Society of Clinical Oncology. J Clin
consequences of multiple types of tobacco products, including bio- Oncol, 33(8), 952–963.
Brenner H, Arndt V, Bode G, et al. 2002. Risk of gastric cancer among smok-
markers of exposure, measures of addictiveness, and transition to ers infected with Helicobacter pylori. Int J Cancer, 98(3), 446–​449.
manufactured cigarettes. PMID: 11920598.
Brooks DR, Palmer JR, Strom BL, and Rosenberg L. 2003. Menthol cigarettes
References and risk of lung cancer. Am J Epidemiol, 158(7), 609–​616; discussion
Agaku IT, and Alpert HR. 2015. Trends in annual sales and current use of 617–​620. PMID: 14507595.
cigarettes, cigars, roll-​your-​own tobacco, pipes, and smokeless tobacco Brown LM, Pottern LM, and Hoover RN. 1987. Testicular cancer in young
among US adults, 2002–​2012. Tob Control. PMID: 25899447. men:  the search for causes of the epidemic increase in the United
Agaku IT, King BA, Husten CG, et  al. 2014. Tobacco product use among States. J Epidemiol Community Health, 41(4), 349–​ 354. PMCID:
adults:  United States, 2012–​ 2013. MMWR Morb Mortal Wkly Rep, PMC1052660.
63(25), 542–​547. PMID: 24964880. Burns D, Lee L, Shen LZ, et  al. 1997. Cigarette smoking behavior in the
Alexandrov LB, Ju YS, Haase K, et al. 2016. Mutational signatures associated United States. In: Shopland D, Burns D, Garfinkel L, and Samet J (Eds.),
with tobacco smoking in human cancer. Science, 354(6312), 618–​622. Changes in Cigarette Related Disease Risks and Theit Implication for
PMID: 27811275. Prevention and Control, Smoking and Tobacco Control Monograph 8 (pp.
Apelberg BJ, Hepp LM, Avila-​Tang E, et  al. 2013. Environmental monitor- 13–​112). Washington, DC: National Cancer Institute.
ing of secondhand smoke exposure. Tob Control, 22(3), 147–​ 155. Calafat AM, Polzin GM, Saylor J, et al. 2004. Determination of tar, nicotine,
PMCID: PMC3639351. and carbon monoxide yields in the mainstream smoke of selected inter-
Arrazola RA, Singh T, Corey CG, et al. 2015. Tobacco use among middle and national cigarettes. Tob Control, 13(1), 45–​51. PMCID: PMC1747810.
high school students:  United States, 2011–​2014. MMWR Morb Mortal California Environmental Protection Agency. (2005). Health Effects of Exposure
Wkly Rep, 64(14), 381–​385. PMID: 25879896. to Environmental Tobacco Smoke:  Final Report. Sacramento, CA.
Ashley DL, Beeson MD, Johnson DR, et  al. 2003. Tobacco-​specific nitrosa- https://​oehha.ca.gov/​air/​report/​health-​effects-​exposure-​environmental-​
mines in tobacco from U.S. brand and non-​U.S. brand cigarettes. Nicotine tobacco-​smoke-​final-​report
Tob Res, 5(3), 323–​331. PMID: 12791527. Campbell PT, Curtin K, Ulrich CM, et al. 2009. Mismatch repair polymorphisms
Asma S, Song Y, Cohen J, et al. 2014. CDC Grand Rounds: Global Tobacco and risk of colon cancer, tumour microsatellite instability and interactions
Control. MMWR Morb Mortal Wkly Rep, 63(13), 277–280. PMID: with lifestyle factors. Gut, 58(5), 661–​667. PMCID: PMC2903215.
24699763. Cancer Genome Atlas Research Network. 2014a. Comprehensive molecular
Australian National Health and Medical Research Council. (1987). Effects characterization of gastric adenocarcinoma. Nature, 513(7517), 202–​209.
of Passive Smoking on Health. Canbera, AU:  Government Publishing PMCID: PMC4170219.
Service. Cancer Genome Atlas Research Network. 2014b. Comprehensive molecular
Avila-​Tang E, Al-​Delaimy WK, Ashley DL, et  al. 2013a. Assessing second- characterization of urothelial bladder carcinoma. Nature, 507(7492),
hand smoke using biological markers. Tob Control, 22(3), 164–​171. 315–​322. PMCID: PMC3962515.
PMCID: PMC3639350. Carter BD, Abnet CC, Feskanich D, et al. 2015. Smoking and mortality: beyond
Avila-​Tang E, Elf JL, Cummings KM, et  al. 2013b. Assessing secondhand established causes. N Engl J Med, 372(7), 631–​640. PMID: 25671255.
smoke exposure with reported measures. Tob Control, 22(3), 156–​163. Castellsague X, and Munoz N. 2003. Chapter  3:  Cofactors in human papil-
PMCID: PMC3639349. lomavirus carcinogenesis: role of parity, oral contraceptives, and tobacco
Balfour DJ, and Fagerstrom KO. 1996. Pharmacology of nicotine and its smoking. J Natl Cancer Inst Monogr, 31, 20–​28. PMID: 12807941.
therapeutic use in smoking cessation and neurodegenerative disorders. Castillo JJ, Dalia S, and Shum H. 2011. Meta-​analysis of the association
Pharmacol Ther, 72(1), 51–​81. PMID: 8981571. between cigarette smoking and incidence of Hodgkin’s lymphoma. J Clin
Baris D, Karagas MR, Verrill C, et al. 2009. A case-​control study of smoking Oncol, 29(29), 3900–​3906. PMID: 21911724.
and bladder cancer risk: emergent patterns over time. J Natl Cancer Inst, Catsburg C, Miller AB, and Rohan TE. 2015. Active cigarette smoking
101(22), 1553–​1561. PMCID: PMC2778671. and risk of breast cancer. Int J Cancer, 136(9), 2204–​2209. PMID:
Barrington-​Trimis JL, Urman R, Berhane K, et  al. 2016. E-​Cigarettes and 25307527.
future cigarette use. Pediatrics, 138(1). PMCID: PMC4925085. Centers for Disease Control and Prevention (CDC). 1994. Cigarette smok-
Benowitz N. 2001. Compensatory smoking of low-​yield cigarettes. In: Shopland ing among adults-​-​United States, 1992, and changes in the definition
D, Burns D, Benowitz N, and Amacher R (Eds.), Risks Associated with of current cigarette smoking. MMWR Morb Mortal Wkly Rep, 43(19),
Smoking Cigarettes with Low Machine-​ Measured Levels of Tar and 342–​346.
Nicotine, Smoking and Tobacco Control Monograph 13 (Vol. NIH Publ. CDC. 1999. Achievements in public health, 1900–​1999: tobacco use—​United
No. 02-​5074, pp. 39–​63). Bethesda, MD: US Department of Health and States, 1900–​1999. MMWR Morb Mortal Wkly, 48(43), 986–​993. https://​
Human Services, National Institutes of Health, National Cancer Institute. www.cdc.gov/​mmwr/​preview/​mmwrhtml/​mm4843a2.htm
Benowitz NL. 1996. Pharmacology of nicotine:  addiction and therapeutics. CDC. 2016. Global Tobacco Surveillance System. www.cdc.gov/​tobacco/​
Annu Rev Pharmacol Toxicol, 36, 597–​613. PMID: 8725403. global/​gtss/​index.htm
Bilano V, Gilmour S, Moffiet T, et al. 2015. Global trends and projections for CDC. 2000. Use of FDA-​ approved pharmacologic treatments for tobacco
tobacco use, 1990–​2025:  an analysis of smoking indicators from the dependence: United States, 1984–​1998. Morb Mortal Wkly Rep, 49, 4.
 207

Tobacco 207
Chang CM, Corey CG, Rostron BL, and Apelberg BJ. 2015. Systematic review pii:  tobaccocontrol-​2016-​053406. doi:  10.1136/​tobaccocontrol-​2016-​
of cigar smoking and all cause and smoking related mortality. BMC 053406. [Epub ahead of print]. PMID: 27852893.
Public Health, 15, 390. PMCID: PMC4408600. Djordjevic MJ, Hoffmann D, Glynn T, Connally GN. US commercial brands of
Chaturvedi AK, Anderson WF, Lortet-​Tieulent J, et al. 2013. Worldwide trends moist snuff, 1994. I assessment of nicotine, moisture, and pH. NCBI - NIH,
in incidence rates for oral cavity and oropharyngeal cancers. J Clin Oncol, https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1759381/pdf/v004p00062.
31(36), 4550–​4559. PMCID: PMC3865341. pdf. Accessed June 2017.
Chaturvedi AK, Engels EA, Anderson WF, and Gillison ML. 2008. Incidence Eriksen M, Mackay J, and Ross H. 2012. The Tobacco Atlas, 4th ed. Atlanta,
trends for human papillomavirus-​related and -​unrelated oral squamous GA; New York: American Cancer Society, World Lung Foundation.
cell carcinomas in the United States. J Clin Oncol, 26(4), 612–​619. Ezzati M, Lopez AD, Rodgers A, et al. 2002. Selected major risk factors and
PMID: 18235120. global and regional burden of disease. Lancet, 360(9343), 1347–​1360.
Chen C, Cook LS, Li XY, et al. 1999. CYP2D6 genotype and the incidence of PMID: 12423980.
anal and vulvar cancer. Cancer Epidemiol Biomarkers Prev, 8(4 Pt 1), Faber MT, Kjaer SK, Dehlendorff C, et al. 2013. Cigarette smoking and risk
317–​321. PMID: 10207635. of ovarian cancer: a pooled analysis of 21 case-​control studies. Cancer
Chen Z, Peto R, Zhou M, et al. 2015. Contrasting male and female trends in Causes Control, 24(5), 989–​1004. PMCID: PMC3818570.
tobacco-​attributed mortality in China: evidence from successive nation- Fagerstrom KO. 1978. Measuring degree of physical dependence to tobacco
wide prospective cohort studies. Lancet, 386(10002), 1447–​ 1456. smoking with reference to individualization of treatment. Addict Behav,
PMCID: PMC4691901. 3(3–​4), 235–​241. PMID: 735910.
Chia VM, Newcomb PA, Bigler J, et al. 2006. Risk of microsatellite-​unstable Fagerstrom KO, Kunze M, Schoberberger R, et al. 1996. Nicotine dependence
colorectal cancer is associated jointly with smoking and nonsteroi- versus smoking prevalence: comparisons among countries and categories
dal anti-​ inflammatory drug use. Cancer Res, 66(13), 6877–​ 6883. of smokers. Tob Control, 5(1), 52–​56. PMCID: PMC1759482.
PMID: 16818666. Felix AS, Yang HP, Gierach GL, Park Y, and Brinton LA. 2014. Cigarette
Chuang SC, Lee YC, Hashibe M, et  al. 2010. Interaction between cigarette smoking and endometrial carcinoma risk:  the role of effect modifica-
smoking and hepatitis B and C virus infection on the risk of liver cancer: a tion and tumor heterogeneity. Cancer Causes Control, 25(4), 479–​489.
meta-​analysis. Cancer Epidemiol Biomarkers Prev, 19(5), 1261–​1268. PMCID: PMC4151521.
PMCID: PMC4170071. Fircanis S, Merriam P, Khan N, and Castillo JJ. 2014. The relation between
Collaborative Group on Epidemiological Studies of Ovarian Cancer, Beral cigarette smoking and risk of acute myeloid leukemia: an updated meta-​
V, Gaitskell K, et  al. 2012. Ovarian cancer and smoking:  individual analysis of epidemiological studies. Am J Hematol, 89(8), E125–​E132.
participant meta-​analysis including 28,114 women with ovarian can- PMID: 24753145.
cer from 51 epidemiological studies. Lancet Oncol, 13(9), 946–​956. Fix BV, O’Connor RJ, Vogl L, et al. 2014. Patterns and correlates of polyto-
PMCID: PMC3431503. bacco use in the United States over a decade: NSDUH 2002–​2011. Addict
Collishaw N. 2016. Blowing smoke:  the history of tobacco-​specific nitrosa- Behav, 39(4), 768–​781. PMCID: PMC3984759.
mines in Canadian tobacco. Tob Control Published Online First: 07 June Flanders WD, Lally CA, Zhu BP, Henley SJ, and Thun MJ. 2003. Lung cancer
2016. doi: 10.1136/​tobaccocontrol-​2015-​PMID: 27272915. mortality in relation to age, duration of smoking, and daily cigarette con-
Collishaw NE, Boyd NF, Cantor KP, et  al. 2009. Canadian Expert Panel on sumption: results from Cancer Prevention Study II. Cancer Res, 63(19),
Tobacco Smoke and Breast Cancer Risk. OTRU Special Report Series, 6556–​6562. PMID: 14559851.
April 2009. Toronto: Ontario Tobacco Research Unit. Food and Drug Administration. (2012). Harmful and Potentially Harmful
Connolly GN, and Alpert HR. 2014. Has the tobacco industry evaded the Constituents in Tobacco Products and Tobacco Smoke: Established List.
FDA’s ban on “Light” cigarette descriptors? Tob Control, 23(2), 140–​145. Washington, DC: Federal Register.
PMCID: PMC3932763. Franceschi S, and Serraino D. 1992. Risk factors for adult soft tissue sarcoma in
Cumberbatch MG, Rota M, Catto JW, La Vecchina C. The Role of Tobacco northern Italy. Ann Oncol, 3 Suppl 2, S85–​S88. PMID: 1622876.
Smoke in Bladder and Kidney Carcinogenesis:  A  Comparison of Freedman DM, Sigurdson A, Doody MM, Rao RS, and Linet MS. 2003. Risk
Exposures and Meta-​analysis of Incidence and Mortality Risks. Eur Urol, of melanoma in relation to smoking, alcohol intake, and other factors in
70(3), 458–​466. a large occupational cohort. Cancer Causes Control, 14(9), 847–​857.
Daling JR, Doody DR, Sun X, et al. 2009. Association of marijuana use and PMID: 14682442.
the incidence of testicular germ cell tumors. Cancer, 115(6), 1215–​1223. Freedman ND, Abnet CC, Caporaso NE, et al. 2016. Impact of changing US
PMCID: PMC2759698. cigarette smoking patterns on incident cancer:  risks of 20 smoking-​
Daling JR, Madeleine MM, Schwartz SM, et  al. 2002. A population-​based related cancers among the women and men of the NIH-​AARP cohort. Int
study of squamous cell vaginal cancer:  HPV and cofactors. Gynecol J Epidemiol, 45(3), 846–​856. PMCID: PMC5005940.
Oncol, 84(2), 263–​270. PMID: 11812085. Freedman ND, Silverman DT, Hollenbeck AR, Schatzkin A, and Abnet CC.
Daling JR, Sherman KJ, Hislop TG, et al. 1992. Cigarette smoking and the risk 2011. Association between smoking and risk of bladder cancer among
of anogenital cancer. Am J Epidemiol, 135(2), 180–​189. PMID: 1311142. men and women. JAMA, 306(7), 737–​745. PMCID: PMC3441175.
Department of the Treasury. 2006. Tax Classification of Cigars and Cigarettes Gallagher RP, Huchcroft S, Phillips N, et  al. 1995. Physical activity, medi-
(2006R–​ 276P). Retrieved from Tax Classification of Cigars and cal history, and risk of testicular cancer (Alberta and British Columbia,
Cigarettes website:  https://​www.federalregister.gov/​documents/​2006/​10/​ Canada). Cancer Causes Control, 6(5), 398–​406. PMID: 8547537.
25/​06-​8835/​tax-​classification-​of-​cigars-​and-​cigarettes-​2006r-​276p Gandini S, Botteri E, Iodice S, et  al. 2008. Tobacco smoking and cancer:  a
DiFranza JR, and Coleman M. 2001. Sources of tobacco for youths in commu- meta-​analysis. Int J Cancer, 122(1), 155–​164. PMID: 17893872.
nities with strong enforcement of youth access laws. Tob Control, 10(4), Garcia-​Closas M, Rothman N, Figueroa JD, et al. 2013. Common genetic poly-
323–​328. PMCID: PMC1747607. morphisms modify the effect of smoking on absolute risk of bladder can-
Doll R. 1998. Uncovering the effects of smoking: historical perspective. Stat cer. Cancer Res, 73(7), 2211–​2220. PMCID: PMC3688270.
Methods Med Res, 7(2), 87–​117. PMID: 9654637. Gaudet MM, Gapstur SM, Sun J, et al. 2013. Active smoking and breast cancer
Doll R, and Hill AB. 1964. Mortality in relation to smoking:  ten years’ risk: original cohort data and meta-​analysis. J Natl Cancer Inst, 105(8),
observations of British doctors. Br Med J, 1(5395), 1399–​ 1410. 515–​525. PMID: 23449445.
PMCID: PMC1814562. Giovannucci E, Colditz GA, Stampfer MJ, et al. 1994a. A prospective study of
Doll R, Hill AB, and Kreyberg L. 1957. The significance of cell type in cigarette smoking and risk of colorectal adenoma and colorectal cancer in
relation to the aetiology of lung cancer. Br J Cancer, 11(1), 43–​48. U.S. women. J Natl Cancer Inst, 86(3), 192–​199. PMID: 8283491.
PMCID: PMC2073933. Giovannucci E, Rimm EB, Stampfer MJ et al. 1994b. A prospective study of
Doll R, and Peto R. 1978. Cigarette smoking and bronchial carcinoma: dose cigarette smoking and risk of colorectal adenoma and colorectal cancer in
and time relationships among regular smokers and lifelong non-​smokers. U.S. men. J Natl Cancer Inst, 86(3), 183–​191. PMID: 8283490.
J Epidemiol Community Health, 32(4), 303–​313. PMCID: PMC1060963. Giovino GA, Mirza SA, Samet JM, et  al. 2012. Tobacco use in 3 billion
Doll R, and Peto R. 1981. The causes of cancer: quantitative estimates of avoid- individuals from 16 countries:  an analysis of nationally representa-
able risks of cancer in the United States today. J Natl Cancer Inst, 66(6), tive cross-​sectional household surveys. Lancet, 380(9842), 668–​ 679.
1191–​1308. PMID: 7017215. PMID: 22901888.
Dossus L, Boutron-​Ruault MC, Kaaks R, et al. 2014. Active and passive ciga- Goniewicz ML, Eisner MD, Lazcano-​Ponce E, et  al. 2011. Comparison of
rette smoking and breast cancer risk: results from the EPIC cohort. Int J urine cotinine and the tobacco-​specific nitrosamine metabolite 4-​(methyl-
Cancer, 134(8), 1871–​1888. PMID: 24590452. nitrosamino)-​1-​(3-​pyridyl)-​1-​butanol (NNAL) and their ratio to discrim-
Dutra LM, Grana R, and Glantz SA. 2016. Philip Morris research on inate active from passive smoking. Nicotine Tob Res, 13(3), 202–​208.
precursors to the modern e-​ cigarette since 1990. Tob Control. PMCID: 3045466.
208

208 PART III:  THE CAUSES OF CANCER


Goniewicz ML, Knysak J, Gawron M, et al. 2014. Levels of selected carcin- variation in interleukin 2. Cancer Epidemiol Biomarkers Prev, 17(7),
ogens and toxicants in vapour from electronic cigarettes. Tob Control, 1790–​1799. PMCID: PMC2497438.
23(2), 133–​139. PMCID: PMC4154473. Huxley RR, Ansary-​Moghaddam A, Clifton P, et al. 2009. The impact of dietary
Gram IT, Little MA, Lund E, and Braaten T. 2016. The fraction of breast cancer and lifestyle risk factors on risk of colorectal cancer: a quantitative over-
attributable to smoking: The Norwegian Women and Cancer Study 1991–​ view of the epidemiological evidence. Int J Cancer, 125(1), 171–​180.
2012. Br J Cancer, 115(5), 616–​623. PMCID: PMC4997535. PMID: 19350627.
Gram IT, Lukanova A, Brill I, et al. 2012. Cigarette smoking and risk of histo- International Agency for Research on Cancer (IARC). 1986. Tobacco smoke.
logical subtypes of epithelial ovarian cancer in the EPIC cohort study. Int IARC Monogr Eval Carcinogenic Risks Hum 83. PMID: 6642935.
J Cancer, 130(9), 2204–​2210. PMID: 21678398. IARC. 1995. Human papilloma viruses. IARC Monogr Eval Carcinogenic
Gram IT, Park SY, Kolonel LN, et al. 2015. Smoking and risk of breast can- Risks Hum 64. PMID: 6642935.
cer in a racially/​ethnically diverse population of mainly women who do IARC. 2004. Tobacco smoke and involuntary smoking. IARC Monogr Eval
not drink alcohol: The MEC Study. Am J Epidemiol, 182(11), 917–​925. Carcinogenic Risks Hum 83. PMID: 6642935.
PMCID: PMC4836396. IARC. 2007. Smokeless tobacco and some tobacco-​specific N-​nitrosamines.
Grana R, Benowitz N, and Glantz SA. 2014. E-​cigarettes: a scientific review. IARC Monogr Eval Carcinogenic Risks Hum 89. Retrieved from http://​
Circulation, 129(19), 1972–​1986. PMCID: PMC4018182. site.ebrary.com/​lib/​yale/​Doc?id=10252497
Gravely S, Fong GT, Cummings KM, et al. 2014. Awareness, trial, and current use IARC. 2012a. A Review of human carcinogens. E. Personal habits and indoor
of electronic cigarettes in 10 countries: findings from the ITC project. Int J combustions. IARC Monogr Eval Carcinogenic Risks Hum 100E.
Environ Res Public Health, 11(11), 11691–​11704. PMCID: PMC4245638. IARC. 2012b. A review of human carcinogens. F. Chemical agents and related
Gu F, Wacholder S, Kovalchik S, et al. 2014. Time to smoke first morning ciga- occupations. IARC Monogr Eval Carcinogenic Risks Hum 100E.
rette and lung cancer in a case-​control study. J Natl Cancer Inst, 106(6), Inoue-​Choi M, Liao LM, Reyes-​Guzman C, et al. 2017. Association of long-​
dju118. PMCID: PMC4072901. term, low-​intensity smoking with all-​cause and cause-​specific mortality
Haenszel W, and Shimkin MB. 1956. Smoking patterns and epidemiology of in the National Institutes of Health-​AARP Diet and Health Study. JAMA
lung cancer in the United States: are they compatible? J Natl Cancer Inst, Intern Med. 177(1), 87–​ 95. doi:  10.1001/​jamainternmed.2016.7511.
16(6), 1417–​1441. PMID: 13320121. PMID: 27918784.
Hammond E. 1966. Smoking in relation to the death rates of one million men Institute of Medicine. 2001. Clearing the Smoke: Assessing the Science Base
and women. In:  Haenszel W (Ed.), Epidemiologic Approaches to the for Tobacco Harm Reduction. Washington, DC: National Academy Press.
Study of Cancer and Other Chronic Diseases (National Cancer Institute Institute of Medicine. 2015. Public Health Implications of Raising the Minimum
Monograph 19, pp. 127–​204). Rockville, MD: US Department of Health, Age of Legal Access to Tobacco Products. Washington, DC:  National
Education, and Welfare. Academies Press.
Hanaoka T, Yamamoto S, Sobue T, et al. 2005. Active and passive smoking and International Collaboration of Epidemiological Studies of Cervical Cancer,
breast cancer risk in middle-​aged Japanese women. Int J Cancer, 114(2), Appleby P, Beral V, et  al. 2006. Carcinoma of the cervix and tobacco
317–​322. PMID: 15540214. smoking:  collaborative reanalysis of individual data on 13,541 women
Harris JE. 1983. Cigarette smoking among successive birth cohorts of men and with carcinoma of the cervix and 23,017 women without carcinoma of the
women in the United States during 1900–​80. J Natl Cancer Inst, 71(3), cervix from 23 epidemiological studies. Int J Cancer, 118(6), 1481–​1495.
473–​479. PMID: 6577223. PMID: 16206285.
Harris JE, Thun MJ, Mondul AM, and Calle EE. 2004. Cigarette tar Iodice S, Gandini S, Maisonneuve P, and Lowenfels AB. 2008. Tobacco and the
yields in relation to mortality from lung cancer in the Cancer risk of pancreatic cancer: a review and meta-​analysis. Langenbecks Arch
Prevention Study II prospective cohort, 1982–​8. BMJ, 328(7431), 72. Surg, 393(4), 535–​545. PMID: 18193270.
PMCID: PMC314045. Islami F, Moreira DM, Boffetta P, and Freedland SJ. 2014. A systematic
Hashibe M. 2010. Risk factors:  tobacco and alcohol. In:  Olshan AF (Ed.), review and meta-​analysis of tobacco use and prostate cancer mortality
Epidemiology, Pathogenesis, and Prevention of Head and Neck Cancer and incidence in prospective cohort studies. Eur Urol, 66(6), 1054–​1064.
(pp. 65–​86). New York: Springer. PMCID: PMC4566150.
Hashibe M, Brennan P, Chuang SC, et al. 2009. Interaction between tobacco and Jacobs EJ, Newton CC, Carter BD, et  al. 2015. What proportion of cancer
alcohol use and the risk of head and neck cancer: pooled analysis in the deaths in the contemporary United States is attributable to cigarette smok-
International Head and Neck Cancer Epidemiology Consortium. Cancer ing? Ann Epidemiol, 25(3), 179–​182 e171. PMID: 25487970.
Epidemiol Biomarkers Prev, 18(2), 541–​550. PMCID: PMC3051410. Jamal A, King B, Neff L, et  al. 2016. Current cigarette smoking among
Hassan MM, Spitz MR, Thomas MB, et  al. 2008. Effect of different types adults:  United States, 2005–​2015. MMWR Morb Mortal Wkly 65(44),
of smoking and synergism with hepatitis C virus on risk of hepatocel- 1205–​1211.
lular carcinoma in American men and women: case-​control study. Int J Jarvis MJ, Boreham R, Primatesta P, Feyerabend C, and Bryant A. 2001.
Cancer, 123(8), 1883–​1891. PMCID: PMC2673571. Nicotine yield from machine-​smoked cigarettes and nicotine intakes in
Hecht SS, Carmella SG, Edmonds A, et al. 2008. Exposure to nicotine and a smokers: evidence from a representative population survey. J Natl Cancer
tobacco-​specific carcinogen increase with duration of use of smokeless Inst, 93(2), 134–​138. PMID: 11208883.
tobacco. Tob Control, 17(2), 128–​131. PMCID: PMC3889131. Jarvis MJ, Tunstall-​Pedoe H, Feyerabend C, Vesey C, and Saloojee Y. 1987.
Hecht SS, Stepanov I, and Carmella SG. 2016. Exposure and metabolic activa- Comparison of tests used to distinguish smokers from nonsmokers. Am J
tion biomarkers of carcinogenic tobacco-​specific nitrosamines. Acc Chem Public Health, 77(11), 1435–​1438. PMCID: PMC1647100.
Res, 49(1), 106–​114. PMID: 26678241. Jayalekshmi PA, Gangadharan P, Akiba S, Koriyama C, and Nair RR. 2011.
Henderson BE, Benton B, Jing J, Yu MC, and Pike MC. 1979. Risk factors Oral cavity cancer risk in relation to tobacco chewing and bidi smok-
for cancer of the testis in young men. Int J Cancer, 23(5), 598–​602. ing among men in Karunagappally, Kerala, India: Karunagappally cohort
PMID: 37169. study. Cancer Sci, 102(2), 460–​467. PMID: 21129124.
Henley SJ, Thun MJ, Chao A, and Calle EE. 2004. Association between exclu- Jayalekshmi PA, Hassani S, Nandakumar A, et  al. 2015. Gastric can-
sive pipe smoking and mortality from cancer and other diseases. J Natl cer risk in relation to tobacco use and alcohol drinking in Kerala,
Cancer Inst, 96(11), 853–​861. PMID: 15173269. India:  Karunagappally cohort study. World J Gastroenterol, 21(44),
Hirayama T. 1981. Non-​smoking wives of heavy smokers have a higher risk 12676–​12685. PMCID: PMC4658623.
of lung cancer: a study from Japan. Br Med J (Clin Res Ed), 282(6259), Jayalekshmi PA, Nandakumar A, Akiba S, Gangadharan P, and Koriyama
183–​185. PMCID: PMC1503989. C. 2013. Associations of tobacco use and alcohol drinking with laryn-
Hoffmann D, and Hoffmann I. 1997. The changing cigarette, 1950–​1995. J geal and hypopharyngeal cancer risks among men in Karunagappally,
Toxicol Environ Health, 50(4), 307–​364. PMID: 9120872. Kerala, India:Karunagappally cohort study. PLoS One, 8(8), e73716.
Holford TR, Meza R, Warner KE, et al. 2014. Tobacco control and the reduc- PMCID: PMC3756048.
tion in smoking-​related premature deaths in the United States, 1964–​ Jedrychowski W, Boeing H, Wahrendorf J, et  al. 1993. Vodka consumption,
2012. JAMA, 311(2), 164–​171. PMCID: PMC4056770. tobacco smoking and risk of gastric cancer in Poland. Int J Epidemiol,
Holford TR, Levy DT, McKay LA, et  al. 2014. Patterns of birth cohort-​ 22(4), 606–​613. PMID: 8225732.
specific smoking histories, 1965–​2009. Am J Prev Med, 46(2), e31–​e37. Jedrychowski W, Popiela T, Drews M, et al. 1999. Effect of Helicobacter pylori
PMCID: 3951759. infection, smoking and dietary habits on the occurrence of antrum intes-
Husgafvel-​Pursiainen K. 2004. Genotoxicity of environmental tobacco tinal metaplasia: clinico-​epidemiological study in Poland. Pol J Pathol,
smoke: a review. Mutat Res, 567(2–​3), 427–​445. PMID: 15572289. 50(4), 289–​295. PMID: 10721269.
Hussain SK, Madeleine MM, Johnson LG, et  al. 2008. Cervical and vulvar Jha P. 2009. Avoidable global cancer deaths and total deaths from smoking. Nat
cancer risk in relation to the joint effects of cigarette smoking and genetic Rev Cancer, 9(9), 655–​664. PMID: 19693096.
 209

Tobacco 209
Jha P, Ramasundarahettige C, Landsman V, et al. 2013. 21st-​century hazards Lynch SM, Vrieling A, Lubin JH, et al. 2009. Cigarette smoking and pancreatic
of smoking and benefits of cessation in the United States. N Engl J Med, cancer: a pooled analysis from the pancreatic cancer cohort consortium.
368(4), 341–​350. PMID: 23343063. Am J Epidemiol, 170(4), 403–​413. PMCID: PMC2733861.
Joehanes R, Just AC, Marioni RE, et al. 2016. Epigenetic signatures of cigarette Madeleine MM, Daling JR, Carter JJ, et al. 1997. Cofactors with human papil-
smoking. Circ Cardiovasc Genet, 9(5), 436–​447. PMID: 27651444. lomavirus in a population-​based study of vulvar cancer. J Natl Cancer
Johnson KC. 2005. Accumulating evidence on passive and active smoking and Inst, 89(20), 1516–​1523. PMID: 9337348.
breast cancer risk. Int J Cancer, 117(4), 619–​628. PMID: 15929073. Marrero JA, Fontana RJ, Fu S, et al. 2005. Alcohol, tobacco and obesity are
Kahn HA. 1966. The Dorn study of smoking and mortality among U.S. veter- synergistic risk factors for hepatocellular carcinoma. J Hepatol, 42(2),
ans: report on eight and one-​half years of observation. Natl Cancer Inst 218–​224. PMID: 15664247.
Monogr, 19, 1–​125. PMID: 5905668. Mazurek JM, Syamlal G, King BA, et al. 2014. Smokeless tobacco use among
Kamper-​Jorgensen M, Rostgaard K, Glaser SL, et al. 2013. Cigarette smoking working adults—​United States, 2005 and 2010. MMWR Morb Mortal
and risk of Hodgkin lymphoma and its subtypes: a pooled analysis from Wkly Rep, 63(22), 477–​482. PMID: 24898164.
the International Lymphoma Epidemiology Consortium (InterLymph). McLaughlin JK, Silverman DT, Hsing AW, et  al. 1992. Cigarette smoking
Ann Oncol, 24(9), 2245–​2255. PMCID: PMC3755332. and cancers of the renal pelvis and ureter. Cancer Res, 52(2), 254–​257.
Kessel Schneider S, Buka SL, Dash K, Winickoff JP, and O’Donnell L. 2016. PMID: 1728398.
Community reductions in youth smoking after raising the minimum Miller MD, Marty MA, Broadwin R, et  al. 2007. The association between
tobacco sales age to 21. Tob Control, 25(3), 355–​359. PMID: 26071428. exposure to environmental tobacco smoke and breast cancer: a review by
Kilfoy BA, Zheng T, Lan Q, et al. 2010. Genetic variation in N-​acetyltransferases the California Environmental Protection Agency. Prev Med, 44(2), 93–​
1 and 2, cigarette smoking, and risk of non-​Hodgkin lymphoma. Cancer 106. PMID: 17027075.
Causes Control, 21(1), 127–​133. PMCID: PMC2972187. Montazeri Z, Nyiraneza C, El-​Katerji H, and Little J. 2017. Waterpipe smok-
Kozlowski L, O’Connor RJ, Sweeney CT. 2001. Cigarette design. In: Shopland ing and cancer: systematic review and meta-​analysis. Tob Control, 26(1),
D, Burns D, Benowitz N, Amacher R (Eds.), Risks Associated With Smoking 92–​97. PMID: 27165994.
Cigarettes With Low Machine-​ Measured Yields of Tar and Nicotine, Morton LM, Slager SL, Cerhan JR, et  al. 2014. Etiologic heterogeneity
Smoking and Tobacco Control Monograph 13 (Vol. NIH Publ. No. 02-​5074, among non-​Hodgkin lymphoma subtypes: the InterLymph Non-​Hodgkin
pp. 13–​38). Bethesda, MD: U.S. Department of Health and Human Services, Lymphoma Subtypes Project. J Natl Cancer Inst Monogr, 2014(48), 130–​
National Institutes of Health, National Cancer Institute, pp. 13–​38. 144. PMCID: PMC4155467.
Kreyberg L. 1962. Histological lung cancer types:  a morphological and bio- National Cancer Institute. 1998. Cigar Smoking and Cancer. Tobacco Control
logical correlation. Acta Pathol Microbiol Scand Suppl, Suppl 157, 1–​92. Monograph 9.  Bethesda, MD:  US Department of Health and Human
PMID: 14035775. Services, National Cancer Insitute, National Insitutes of Health.
Kuper H, Tzonou A, Kaklamani E, et al. 2000. Tobacco smoking, alcohol con- National Cancer Institute and Centers for Disease Control and Prevention.
sumption and their interaction in the causation of hepatocellular carci- 2014. Smokeless Tobacco and Public Health:  A  Global Perspective.
noma. Int J Cancer, 85(4), 498–​502. PMID: 10699921. Bethesda, MD: US Department of Health and Human Services, Centers
Kuper HE, Tzonou A, Kaklamani E, et  al. 2000. Hepatitis B and C viruses for Disease Control and Prevention, and National Institutes of Health,
in the etiology of hepatocellular carcinoma; a study in Greece using National Cancer Institute.
third-​generation assays. Cancer Causes Control, 11(2), 171–​ 175. National Research Council. 1986. Environmental Tobacco Smoke: Measuring
PMID: 10710202. Exposures and Assessing Health Effects. Washington, DC:  National
Lacson JC, Carroll JD, Tuazon E, et al. 2012. Population-​based case-​control Academy Press.
study of recreational drug use and testis cancer risk confirms an asso- National Research Council Committee on Health Risks of Exposure to Radon
ciation between marijuana use and nonseminoma risk. Cancer, 118(21), (BIER VI). 1998. Health Effects of Exposure to Radon. Biological Effects
5374–​5383. PMCID: PMC3775603. of Ionizing Radiation. Washington, DC: National Academy Press.
Ladeiras-​Lopes R, Pereira AK, Nogueira A, et al. 2008. Smoking and gastric Ng M, Freeman MK, Fleming TD, et al. 2014. Smoking prevalence and ciga-
cancer:  systematic review and meta-​analysis of cohort studies. Cancer rette consumption in 187 countries, 1980–​2012. JAMA, 311(2), 183–​192.
Causes Control, 19(7), 689–​701. PMID: 18293090. PMID: 24399557.
Lee CH, Lee KW, Fang FM, et al. 2011. The use of tobacco-​free betel-​quid in Nilsson R. 1998. A qualitative and quantitative risk assessment of snuff dip-
conjunction with alcohol/​tobacco impacts early-​onset age and carcinoma ping. Regul Toxicol Pharmacol, 28(1), 1–​16. PMID: 9784428.
distribution for upper aerodigestive tract cancer. J Oral Pathol Med, 40, Nyante SJ, Gierach GL, Dallal CM, et al. 2014. Cigarette smoking and post-
684–​692. PMID: 21385213. menopausal breast cancer risk in a prospective cohort. Br J Cancer,
Lee YO, Hebert CJ, Nonnemaker JM, and Kim AE. 2014. Multiple tobacco 110(9), 2339–​2347. PMCID: PMC4007228.
product use among adults in the United States:  cigarettes, cigars, elec- O’Keefe AM, and Pollay RW. 1996. Deadly targeting of women in promoting
tronic cigarettes, hookah, smokeless tobacco, and snus. Prev Med, 62, cigarettes. J Am Med Womens Assoc, 51(1–​2), 67–​69. PMID: 8868553.
14–​19. PMID: 24440684. Orellana-​Barrios MA, Payne D, Medrano-​Juarez RM, Yang S, and Nugent K.
Leffondre K, Abrahamowicz M, Siemiatycki J, and Rachet B. 2002. Modeling 2016. Electronic cigarettes for smoking cessation. Am J Med Sci, 352(4),
smoking history: a comparison of different approaches. Am J Epidemiol, 420–​426. PMID: 27776725.
156(9), 813–​823. PMID: 12396999. Palipudi K, Mbulo L, Kosen S, et al. 2015. A cross sectional study of kretek
Leonardi-​Bee J, Ellison T, and Bath-​Hextall F. 2012. Smoking and the risk smoking in Indonesia as a major risk to public health. Asian Pac J Cancer
of nonmelanoma skin cancer: systematic review and meta-​analysis. Arch Prev, 16(16), 6883–​6888. PMID: 26514461.
Dermatol, 148(8), 939–​946. PMID: 22711192. Palipudi KM, Mbulo L, Morton J, et al. 2015. Awareness and current use of
Li Z, Wang Z, Yu Y, Zhang H, and Chen L. 2015. Smoking is inversely related electronic cigarettes in Indonesia, Malaysia, Qatar, and Greece: findings
to cutaneous malignant melanoma:  results of a meta-​ analysis. Br J from 2011–​2013 Global Adult Tobacco Surveys. Nicotine Tob Res, 18,
Dermatol, 173(6), 1540–​1543. PMID: 26134854. 501–​507. PMID: 25895951.
Liang PS, Chen TY, and Giovannucci E. 2009. Cigarette smoking and colorec- Parkin DM. 2011. The fraction of cancer attributable to lifestyle and environ-
tal cancer incidence and mortality: systematic review and meta-​analysis. mental factors in the UK in 2010. Br J Cancer, 105 Suppl 2, S2–​S5.
Int J Cancer, 124(10), 2406–​2415. PMID: 19142968. PMCID: PMC3252063.
Limsui D, Vierkant RA, Tillmans LS, et  al. 2010. Cigarette smoking and Patel NH, Attwood KM, Hanzly M, et al. 2015. Comparative analysis of smok-
colorectal cancer risk by molecularly defined subtypes. J Natl Cancer ing as a risk factor among renal cell carcinoma histological subtypes. J
Inst, 102(14), 1012–​1022. PMCID: PMC2915616. Urol, 194, 640–​646. PMID: 25896558.
Lopez AD, Collishaw N, and Piha T. 1994. A descriptive model og the cigarette Patrick DL, Cheadle A, Thompson DC, et al. 1994. The validity of self-​reported
epidemic in developed countries. Tob Control, 3, 242–​247. smoking: a review and meta-​analysis. Am J Public Health, 84(7), 1086–​
Lortet-​Tieulent J, Goding Sauer A, Siegel RL, et al. 2016. State-​level cancer 1093. PMCID: PMC1614767.
mortality attributable to cigarette smoking in the United States. JAMA Pednekar MS, Gupta PC, Yeole BB, and Hebert JR. 2011. Association of
Intern Med, 176, 1792–​1798. PMID: 27775761. tobacco habits, including bidi smoking, with overall and site-​specific
Lubin JH, Cook MB, Pandeya N, et al. 2012. The importance of exposure rate on cancer incidence: results from the Mumbai cohort study. Cancer Causes
odds ratios by cigarette smoking and alcohol consumption for esophageal Control, 22(6), 859–​868. PMCID: PMC3756904.
adenocarcinoma and squamous cell carcinoma in the Barrett’s Esophagus Peto R, Darby S, Deo H, et al. 2000. Smoking, smoking cessation, and lung
and Esophageal Adenocarcinoma Consortium. Cancer Epidemiol, 36(3), cancer in the UK since 1950: combination of national statistics with two
306–​316. PMCID: PMC3489030. case-​control studies. BMJ, 321(7257), 323–​329. PMCID: PMC27446.
210

210 PART III:  THE CAUSES OF CANCER


Peto R, Lopez AD, Boreham J, Thun M, and Heath C, Jr. 1992. Mortality from Thun MJ, Carter BD, Feskanich D, et  al. 2013. 50-​year trends in smoking-​
tobacco in developed countries:  indirect estimation from national vital related mortality in the United States. N Engl J Med, 368(4), 351–​364.
statistics. Lancet, 339(8804), 1268–​1278. PMID: 1349675. PMCID: PMC3632080.
Peto R, Lopez AD, Pan H, Boreham J, Thun M. 2017. Mortality from smok- Thun M, Day-​Lally C, Myers D, et  al. 1997. Trends in tobacco smoking
ing in developed countries, 1950–​2020. Available from: https://​www.ctsu. and mortality from cigarette use in Cancer Prevention Studies I (1959–​
ox.ac.uk/​.../​mortality-​from-​smoking. Accessed April 24, 2015. 1965) and II (1982–​1988). In:  Shopland D, Burns D, Garfinkel L, and
Pirie K, Peto R, Reeves GK, et al. 2013. The 21st century hazards of smoking Samet J (Eds.), Changes in Cigarette-​Related Disease Risks and Their
and benefits of stopping: a prospective study of one million women in the Implications for Prevention and Control. National Cancer Institute,
UK. Lancet, 381(9861), 133–​141. PMCID: PMC3547248. Smoking and Tobacco Control, Monograph 8 (Vol. NIH Publication
Poynter JN, Haile RW, Siegmund KD, et al. 2009. Associations between smok- No. 97-​4213, pp. 305–​382). Washington, DC: US Department of Health
ing, alcohol consumption, and colorectal cancer, overall and by tumor and Human Services, National Institutes of Health, National Cancer
microsatellite instability status. Cancer Epidemiol Biomarkers Prev, Institute.
18(10), 2745–​2750. PMCID: PMC2759847. Thun MJ, and Henley SJ. 2006. Tobacco. In:  Schottenfeld D, and Fraumeni
Purdue MP, Moore LE, Merino MJ, et al. 2013. An investigation of risk factors JF (Eds.), Cancer Epidemiology and Prevention (3rd ed., pp. 217–​242).
for renal cell carcinoma by histologic subtype in two case-​control studies. Oxford; New York: Oxford University Press.
Int J Cancer, 132, 2640–​2647. PMCID: PMC3717609. Tobacco Atlas. 2015. The Tobacco Atlas, 5th edition. Eriksen M, Mackay J,
Purdue MP, and Silverman DT. 2016. Clearing the Air:  Summarizing the Schluger N, Gomeshtapeh F I, and Drapoe J (Eds.). Atlanta, GA: American
smoking-​related relative risks of bladder and kidney cancer. Eur Urol, Cancer Society, American Lung Association.
70(3), 467–​468. PMID: 27130147. Tobacco Atlas. 2016. Water pipes. www.tobaccoatlas.org/​topic/​water-​pipes
QuickStats. 2016. Cigarette smoking status among current adult e-​cigarette Todd GF. 1978. Cigarette consumption per adult of each sex in various countries.
users, by age group:  National Health Interview Survey, United States, J Epidemiol Community Health, 32(4), 289–​293. PMCID: PMC1060961.
2015. MMWR Morb Mortal Wkly Rep, 65, 1177. Toll BA, and Ling PM. 2005. The Virginia Slims identity crisis: an inside look
Reddy CR. 1974. Carcinoma of hard palate in India in relation to reverse smok- at tobacco industry marketing to women. Tob Control, 14(3), 172–​180.
ing of chuttas. J Natl Cancer Inst, 53(3), 615–​619. PMID: 4412322. PMCID: PMC1748044.
Reynolds P, Hurley S, Goldberg D, and California Teachers Study Steering Tong H, Hu C, Yin X et al. 2013. A Meta-​analysis of the relationship between
Committee. 2006. Accumulating evidence on passive and active smoking cigarette smoking and incidence of myelodysplastic syndromes. PLoS
and breast cancer risk. Int J Cancer, 119(1), 239; author reply 240–​231. One, 8(6), e67537. PMCID: PMC3689714.
PMID: 16432834. Torre LA, Siegel RL, and Jemal A. 2016. Lung cancer statistics. Adv Exp Med
Rosenberg L, Boggs DA, Bethea TN, et al. 2013. A prospective study of smok- Biol, 893, 1–​19. PMID: 26667336.
ing and breast cancer risk among African-​American women. Cancer Trabert B, Sigurdson AJ, Sweeney AM, Strom SS, and McGlynn KA. 2011.
Causes Control, 24(12), 2207–​2215. PMCID: PMC3855285. Marijuana use and testicular germ cell tumors. Cancer, 117(4), 848–​853.
Rostron B. 2012. Lung cancer mortality risk for U.S. menthol cigarette smok- PMCID: PMC3017734.
ers. Nicotine Tob Res, 14(10), 1140–​1144. PMID: 22387991. Trichopoulos D, Kalandidi A, Sparros L, and MacMahon B. 1981. Lung cancer
Sergentanis TN, Kanavidis P, Michelakos T, and Petridou ET. 2013. Cigarette and passive smoking. Int J Cancer, 27(1), 1–​4. PMID: 7251227.
smoking and risk of lymphoma in adults: a comprehensive meta-​analysis Tsoi KK, Pau CY, Wu WK, et  al. 2009. Cigarette smoking and the risk of
on Hodgkin and non-​Hodgkin disease. Eur J Cancer Prev, 22(2), 131–​ colorectal cancer:  a meta-​analysis of prospective cohort studies. Clin
150. PMID: 22759975. Gastroenterol Hepatol, 7(6), 682–​688 e681–​685. PMID: 19245853.
Serraino D, Franceschi S, Talamini R, Frustaci S, and La Vecchia C. 1991. Non-​ US Department of Health and Human Services. 1979. Smoking and
occupational risk factors for adult soft-​tissue sarcoma in northern Italy. Health: A Report of the Surgeon General. Vol. DHEW Publ No (PHS) 79-​
Cancer Causes Control, 2(3), 157–​164. PMID: 1873445. 50066. Washington, DC: US Department of Health and Human Services,
Shafey O, Eriksen M, and Ross H. 2009. The Tobacco Atlas, 3rd edition. Public Health Service, Office of the Assistant Secretary for Health, Office
Atlanta, GA: American Cancer Society, World Lung Foundation. on Smoking and Health.
Shapiro JA, Jacobs EJ, and Thun MJ. 2000. Cigar smoking in men and risk of US Department of Health and Human Services. 1980. The Health Consequences
death from tobacco-​related cancers. J Natl Cancer Inst, 92(4), 333–​337. of Smoking for Women: A Report of the Surgeon General. Washington,
PMID: 10675383. DC:  US Department of Health and Human Services, Public Health
Shields PG. 2002. Tobacco smoking, harm reduction, and biomarkers. J Natl Service, Office of the Assistant Secretary for Health, Office on Smoking
Cancer Inst, 94(19), 1435–​1444. PMID: 12359853. and Health.
Shimazu T, Asada K, Charvat H, et  al. 2015. Association of gastric cancer U.S. Department of Health and Human Services. 1981. The Health
risk factors with DNA methylation levels in gastric mucosa of healthy Consequences of Smoking:  The Changing Cigarette. A  Report of the
Japanese:  a cross-​sectional study. Carcinogenesis, 36(11), 1291–​1298. Surgeon General. Rockville, MD: US Department of Health and Human
PMID: 26354778. Services, Public Health Service, Office on Smoking and Health.
Siegel RL, Jacobs EJ, Newton CC, et al. 2015. Deaths due to cigarette smoking US Department of Health and Human Services. 1982. The Health Consequences
for 12 smoking-​related cancers in the United States. JAMA Intern Med, of Smoking:  Cancer:  A  Report of the Surgeon General. Washington,
175(9), 1574–​1576. PMID: 26076120. DC:  US Department of Health and Human Services, Public Health
Siman JH, Forsgren A, Berglund G, and Floren CH. 2001. Tobacco smok- Service, Office on Smoking and Health.
ing increases the risk for gastric adenocarcinoma among Helicobacter US Department of Health and Human Services. 1986. The Health Consequences
pylori-​infected individuals. Scand J Gastroenterol, 36(2), 208–​ 213. of Involuntary Smoking: A Report of the Surgeon General. Washington,
PMID: 11252415. DC:  US Department of Health and Human Services, Public Health
Singh T, Arrazola RA, Corey CG, and et al. 2016. Tobacco use among mid- Service, Office on Smoking and Health.
dle and high school students: United States, 2011–​2015. MMWR Morb US Department of Health and Human Services. 1988. The Health Consequences
Mortal Wkly Rep, 65, 361–​367. of Smoking:  Nicotine Addiction. Rockville, MD:  US Department of
Sinha DN, Abdulkader RS, and Gupta PC. 2016. Smokeless tobacco-​associated Health and Human Services, Public Health Service, Centers for Disease
cancers:  a systematic review and meta-​analysis of Indian studies. Int J Control, Center for Chronic Disease Prevention and Health Promotion,
Cancer, 138(6), 1368–​1379. PMID: 26443187. Office on Smoking and Health.
Slade J. 1993. Nicotine delivery devices. In:  Orleans C, and Slade J US Department of Health and Human Services. 1989. Reducing the Health
(Eds.), Nicotine Addiction:  Principles and Management (pp. 3–​ 23). Consequences of Smoking:  25 Years of Progress. Rockville, MD:  US
New York: Oxford University Press. Department of Health and Human Services, Public Health Service,
Srigley JR, Delahunt B, Eble JN, et  al. 2013. The International Society of Centers for Disease Control, Center for Chronic Disease Prevention and
Urological Pathology (ISUP) Vancouver classification of renal neoplasia. Health Promotion, Office on Smoking and Health.
Am J Surg Pathol, 37, 1469–​1489. PMID: 24025519. US Department of Health and Human Services. 2001. Women and
Steffen MW, Murad MH, Hays JT, et al. 2014. Self-​report of tobacco use sta- Smoking:  A  Report of the Surgeon General. Rockville, MD:  US
tus: comparison of paper-​based questionnaire, online questionnaire, and Department of Health and Human Services, Centers for Disease Control
direct face-​ to-​
face interview:  implications for meaningful use. Popul and Prevention, National Center for Chronic Disease Prevention and
Health Manag, 17(3), 185–​189. PMCID: PMC4442565. Health Promotion, Office on Smoking and Health.
Thompson CA, Zhang ZF, and Arah OA. 2013. Competing risk bias to explain US Department of Health and Human Services. 2004. The Health Consequences
the inverse relationship between smoking and malignant melanoma. Eur of Smoking:  A  Report of the Surgeon General. Rockville, MD:  US
J Epidemiol, 28(7), 557–​567. PMCID: PMC3864891. Department of Health and Human Services, Centers for Disease Control
 21

Tobacco 211
and Prevention, National Center for Chronic Disease Prevention and Warren CW, Asma S, Lee J, Lea V, Mackay J. 2009. Global Tobacco Surveillance
Health Promotion, Office on Smoking and Health. System: The GTSS Atlas. Brighton, UK: CDC, CDC Foundation, World
US Department of Health and Human Services. 2006. The Health Consequences Lung Foundation.
of Involuntary Exposure to Tobacco Smoke:  A  Report of the Surgeon Weir JM, and Dunn JE, Jr. 1970. Smoking and mortality: a prospective study.
General. Rockville, MD: US Department of Health and Human Services, Cancer, 25(1), 105–​112. PMID: 5410301.
Centers for Disease Control and Prevention, National Center for Chronic Wenbin D, Zhuo C, Zhibing M, et al. 2013. The effect of smoking on the risk
Disease Prevention and Health Promotion, Office on Smoking and Health. of gallbladder cancer:  a meta-​analysis of observational studies. Eur J
US Department of Health and Human Services. 2010. How Tobacco Smoke Gastroenterol Hepatol, 25(3), 373–​379. PMID: 23085578.
Causes Disease:  The Biology and Behavioral Basis for Smoking-​ Wentzensen N, Poole EM, Trabert B, et  al. 2016. Ovarian cancer risk fac-
Attributable Disease. A  Report of the Surgeon General. Rockville, tors by histologic subtype: an analysis from the Ovarian Cancer Cohort
MD: US Department of Health and Human Services, Centers for Disease Consortium. J Clin Oncol. 34, 2888–​2898. PMID: 27325851.
Control and Prevention, National Center for Chronic Disease Prevention Wiersinga WM. 2013. Smoking and thyroid. Clin Endocrinol (Oxf), 79(2),
and Health Promotion, Office on Smoking and Health. 145–​151. PMID: 23581474.
US Department of Health and Human Services. 2012. Preventing Tobacco Winickoff JP, Gottlieb M, and Mello MM. 2014. Tobacco 21: an idea whose
Use among Youth and Young Adults. A  Report of the Surgeon General. time has come. N Engl J Med, 370, 295–​297.
Rockville, MD: US Department of Health and Human Services, Centers World Health Organization (WHO). (2003). WHO Framework Convention
for Disease Control and Prevention, National Center for Chronic Disease on Tobacco Control. Geneva:  World Health Organization. www.who.
Prevention and Health Promotion, Office on Smoking and Health. int/​tobacco/​framework/​WHO_​FCTC_​english.pdf. Accessed April 24,
US Department of Health and Human Services. 2014. The Health Consequences 2017.
of Smoking:  50 Years of Progress. A  Report of the Surgeon General. Wu AH, Crabtree JE, Bernstein L, et  al. 2003. Role of Helicobacter pylori
Rockville, MD: US Department of Health and Human Services, Centers CagA + strains and risk of adenocarcinoma of the stomach and esopha-
for Disease Control and Prevention, National Center for Chronic Disease gus. Int J Cancer, 103(6), 815–​821. PMID: 12516104.
Prevention and Health Promotion, Office on Smoking and Health. Wynder EL, and Hoffmann D. 1994. Smoking and lung cancer:  scien-
US Department of Health and Human Services. 2016. E-​Cigarette Use Among tific challenges and opportunities. Cancer Res, 54(20), 5284–​ 5295.
Youth and Young Adults. Rockville, MD: US Department of Health and PMID: 7923155.
Human Services, Centers for Disease Control and Prevention, National Wyss A, Hashibe M, Chuang SC, et al. 2013. Cigarette, cigar, and pipe smoking
Center for Chronic Disease Prevention and Health Promotion, Office on and the risk of head and neck cancers: pooled analysis in the International
Smoking and Health. Head and Neck Cancer Epidemiology Consortium. Am J Epidemiol,
US Department of Health Education and Welfare. 1964. Smoking and 178(5), 679–​690. PMCID: PMC3755640.
Health:  Report of the Advisory Committee to the Surgeon General of Xue WQ, Qin HD, Ruan HL, Shugart YY, and Jia WH. 2013. Quantitative asso-
the Public Health Service. Washington, DC: US Department of Health, ciation of tobacco smoking with the risk of nasopharyngeal carcinoma: a
Education and Welfare, Public Health Service, Centers for Disease comprehensive meta-​analysis of studies conducted between 1979 and
Control. 2011. Am J Epidemiol, 178(3), 325–​338. PMCID: PMC3727336.
US Environmental Protection Agency. (1992). Respiratory Health Effects of Yeager DS, and Krosnick JA. 2010. The validity of self-​reported nicotine prod-
Passive Smoking:  Lung Cancer and Other Disorders (EPA/​600/​6-​90/​ uct use in the 2001–​2008 National Health and Nutrition Examination
006F). Washington, DC: Office of Health and Environmental Assessment, Survey. Med Care, 48(12), 1128–​1132. PMID: 20940652.
Office of Research and Development, US Environmental Protection Yuan JM, Butler LM, Stepanov I, and Hecht SS. 2014. Urinary tobacco smoke-​
Agency. constituent biomarkers for assessing risk of lung cancer. Cancer Res,
Vardavas CI, Fthenou E, Patelarou E, et al. 2013. Exposure to different sources 74(2), 401–​411. PMCID: PMC4066207.
of second-​hand smoke during pregnancy and its effect on urinary cotinine Zahm SH, Heineman EF, and Vaught JB. 1992. Soft tissue sarcoma and tobacco
and tobacco-​specific nitrosamine (NNAL) concentrations. Tob Control, use:  data from a prospective cohort study of United States veterans.
22(3), 194–​200. PMID: 22253001. Cancer Causes Control, 3(4), 371–​376. PMID: 1617125.
Vineis P, Alavanja M, Buffler P, et  al. 2004. Tobacco and cancer:  recent Zaridze D, Borisova E, Maximovitch D, and Chkhikvadze V. 2000. Alcohol
epidemiological evidence. J Natl Cancer Inst, 96(2), 99–​106. PMID: consumption, smoking and risk of gastric cancer:  case-​control study
14734699. from Moscow, Russia. Cancer Causes Control, 11(4), 363–​ 371.
Vogel RI, Carmella SG, Stepanov I, Hatsukami DK, and Hecht SS. 2011. The PMID: 10843447.
ratio of a urinary tobacco-​specific lung carcinogen metabolite to cotinine Zhou B, Yang L, Sun Q, et al. 2008. Cigarette smoking and the risk of endo-
is significantly higher in passive than in active smokers. Biomarkers, metrial cancer:  a meta-​ analysis. Am J Med, 121(6), 501–​ 508 e503.
16(6), 491–​497. PMCID: PMC3159775. PMID: 18501231.
Warren CW, Jones NR, Peruga A, et  al. 2008. Global youth tobacco sur- Zu K, and Giovannucci E. 2009. Smoking and aggressive prostate cancer:  a
veillance, 2000–​2007. MMWR Surveill Summ, 57(1), 1–​ 28. PMID: review of the epidemiologic evidence. Cancer Causes Control, 20(10),
18219269. 1799–​1810. PMID: 19562492.
21
 213

12 Alcohol and Cancer Risk

SUSAN M. GAPSTUR AND PHILIP JOHN BROOKS

OVERVIEW and death. The WHO estimates that 5.9% of all deaths (7.4% of males
and 4% of females) and 5.1% of disease worldwide is attributed to
Alcoholic beverages have been consumed by humans for thousands of alcohol, and has identified 60 common alcohol-​related conditions and
years. Currently, the alcoholic beverages most frequently consumed are more than 200 conditions in which alcohol consumption is recog-
spirits, beer, and wine, respectively. Worldwide, an estimated 47.7% of nized as a component cause (World Health Organization, 2014). The
men and 28.9% of women aged 15 years or older report drinking alco- adverse effects of harmful drinking include, for example, intentional
hol, and approximately 16.0% of drinkers engage in heavy, episodic and unintentional injuries, violence, acute alcohol poisoning, liver cir-
(binge) drinking. In 2010, alcoholic beverage consumption caused an rhosis, social disruption, impoverishment, neuropsychiatric disorders,
estimated 3.3  million deaths worldwide, and contributed to injuries, gastrointestinal and cardiovascular problems, fetal alcohol syndrome,
violence, liver cirrhosis, social disruption, and at least seven differ- preterm complications, diabetes mellitus, exacerbation of certain infec-
ent types of cancer. The International Agency for Research on Cancer tious diseases, and seven types of cancer (IARC, 2012).
(IARC) classifies exposure to both ethanol in alcoholic beverages and Acetaldehyde is the primary metabolite of ethanol from alcoholic
acetaldehyde, the primary metabolite of ethanol, as carcinogenic to beverage consumption; preformed acetaldehyde can be measured
humans (Group 1) based on “sufficient” evidence that alcoholic bev- in alcoholic beverages. The cytotoxic, mutagenic, and carcinogenic
erage consumption is causally related to cancers of the oral cavity, effects of acetaldehyde, including formation of DNA adducts and inhi-
pharynx, larynx, esophagus, liver, colorectum, and female breast. The bition of DNA repair, have been shown in eukaryotic cells and animal
World Cancer Research Fund/​American Institute of Cancer Research models (Seitz and Stickel, 2010). Acetaldehyde related to alcohol con-
Continuous Update Project (WCRF/​AICR CUP) characterized the sumption has been classified as carcinogenic to humans (IARC, 2012).
evidence as “limited” that heavy alcohol consumption increases pan-
creatic cancer risk. The IARC also notes lack of carcinogenicity of
alcohol consumption with non-​ Hodgkin lymphoma and renal cell METABOLISM OF ALCOHOL AND ACETALDEHYDE
carcinoma, and for other cancers the evidence is inconclusive. The
biologic mechanisms by which alcohol and its primary metabolite After ingestion, alcohol undergoes “first pass metabolism” (FPM) in the
acetaldehyde affect cancer risk appear to vary across anatomic sites. upper aerodigestive tract (UADT) and liver, which reduces the amount
Broadly, these mechanisms involve DNA and protein damage from of alcohol in circulation relative to the amount consumed. FPM begins
acetaldehyde and oxidative stress, nutritional malabsorption, and met- in the oral cavity, where both human cells and microorganisms in saliva
abolic effects, and for breast cancer, increased estrogen levels. In addi- oxidize alcohol to acetaldehyde. An estimated 5%–​14% of the oral dose
tion, carcinogenic contaminants can be introduced during alcoholic of alcohol undergoes FPM (Ammon et al., 1996), with a slightly lower
beverage production. While there are known harmful effects of alcohol percent when gastric emptying is rapid or the amount consumed is high.
consumption, relative to no consumption, light to moderate consump- Alcohol that does not undergo FPM is distributed throughout the body;
tion has been associated with reduced risk of coronary heart disease, the majority (about 90%) of absorbed alcohol is metabolized in the
although recent Mendelian randomization studies have challenged this liver. The enzymes involved in the metabolism of alcohol are expressed
finding. The World Health Organization (WHO) has increased global in many tissues throughout the human body, albeit at varying levels in
surveillance of alcohol consumption and encourages national efforts to different cell types (IARC, 2010), and thus play a role in the toxicity
apply evidence-​based policies to reduce consumption. and/​or carcinogenicity of alcohol in specific tissues.
In humans, the metabolism of alcohol primarily involves a two-​step
process in which ethanol is first oxidized to acetaldehyde by the enzyme
INTRODUCTION alcohol dehydrogenase (ADH), and then acetaldehyde is oxidized to
acetate (acetic acid) by the enzyme aldehyde dehydrogenase (ALDH).
Alcoholic beverages have been consumed for religious, social, and The activity of these enzymes profoundly influences the ability of a
cultural reasons for thousands of years. Archaeological evidence indi- drinker to metabolize alcohol and detoxify acetaldehyde. The human
cates that honey, rice, and hawthorn fruit or grapes were fermented genome contains multiple ADH and ALDH genes that are expressed in
to produce alcohol as early as 7000–​6600 bc in China’s Yellow River different tissues. The ADH1A, ADH1B, and ADH1C genes encode the
Valley (McGovern et  al., 2004). Today, the most common commer- ADH1 α, β, and γ protein subunits, respectively. These different sub-
cially produced alcoholic beverages are spirits distilled from grains, unit proteins form dimers that are the active form of the ADH enzyme,
sugars, fruits or vegetables, beer from barley, and wine from grapes and are expressed at high levels in the liver. In contrast, the ADH7
(World Health Organization, 2014). In some developing countries, protein is expressed in the stomach and the gastrointestinal tract, and
locally or home-​produced alcoholic beverages from, for example, fer- is thought to play a role in FPM in these tissues (Crabb et al., 2004).
mented apples (cider) or honey-​water (mead) are important contribu- The ADH and ALDH genes encode proteins that combine to form
tors to daily consumption (World Health Organization, 2014). the functional enzymes and are polymorphic in humans; the preva-
The principal form of alcohol found in alcoholic beverages is ethanol lence of functional variants varies among different geographic popula-
(ethyl alcohol), which is produced by the fermentation of sugars and tions (Figure 12–1). Mendelian randomization studies have examined
starches by yeast. Ethanol is a central nervous system depressant that the role of ADH and ALDH gene polymorphisms on alcohol avoidance
motivates recreational use of alcohol, and engenders a sense of excite- and consumption, and have informed studies of alcohol-​associated
ment, sociability, pleasure, and intoxication. At progressively higher chronic diseases (Holmes et al., 2014). As discussed later in the chap-
blood levels, it impairs sensory and motor function, cognition, and judg- ter, the *2 allele of ADH1B and the *2 allele of ALDH2 are particularly
ment; severe intoxication can produce stupefaction, unconsciousness, relevant to alcohol-​related carcinogenesis.

213
214

214 PART III:  THE CAUSES OF CANCER


The ADH1B*2 allele, which is common in Asian and Ashkenazi with significantly elevated risk of alcohol-​related esophageal cancer
Jewish populations, encodes the β2 subunit that forms an enzyme (Yokoyama et al., 1996).
with roughly 40-​ fold greater activity at generating acetaldehyde In addition to the enzymes in human cells, other important contribu-
from ethanol than the β1 subunit, which is encoded by the ADH1B*1 tors to alcohol metabolism are microbes (particularly bacteria) resi-
allele. ADH1B*1 is commonly found in European and some African dent in the human body. Some of these bacteria express ADHs. The
populations. The ADH1B*3 allele, which commonly occurs in Native microbial metabolism of alcohol to acetaldehyde can result in locally
American and other African populations, encodes a subunit of the high concentrations of extracellular acetaldehyde in the oral cavity and
enzyme with low affinity for ethanol (Crabb et al., 2004). intestine (Homann et  al., 2000), which are thought to contribute to
In humans, ALDH enzymes are coded by 19 different genes alcohol-​related carcinogenesis in those tissues.
(Koppaka et  al., 2012). Based on their affinity for acetaldehyde, the Aside from ADH and ALDH, another enzyme that metabolizes alco-
ALDH isoforms that are thought to play a role in ethanol-​derived acet- hol and affects alcohol-​related carcinogenesis is the ethanol-​inducible
aldehyde metabolism include ALDH1A1, ALDH1B1 and ALDH2. cytochrome P4502E1 (CYP2E1) (Cederbaum, 2012). While both ADH
Other ALDH enzymes have low affinity for acetaldehyde and are and CYP2E1 convert alcohol to acetaldehyde, there are several important
unlikely to play a significant role in acetaldehyde metabolism under differences between these two enzymes. Whereas the level of CYP2E1
normal physiologic conditions. protein increases in the presence of high concentrations of circulating
A polymorphism of ALDH2 (rs671) is of particular relevance for alcohol, the level of ADH is generally not affected. This alcohol-​related
understanding the relationship of alcohol consumption to acetaldehyde increase in CYP2E1 occurs because ethanol is both a substrate for the
production and cancer risk. Whereas the ALDH2*1 allele produces a enzyme and, when present, stabilizes the enzyme against degradation.
protein with normal catalytic activity, the ALDH2*2 allele encodes the Alcohol metabolism by CYP2E1 significantly increases at alcohol con-
amino acid LYS instead of GLU at position 487, resulting in a cata- centrations above ≈ 20 mM (Asai et al., 1996; Salaspuro and Lieber,
lytically inactive enzyme subunit. The ALDH2*2 allele is found pre- 1978). For comparison, the legal blood alcohol limit for driving in many
dominantly in East Asians (Japanese, Chinese, and Koreans) (Goedde countries is 0.08%, which corresponds to an alcohol concentration of
and Agarwal, 1987). When individuals carrying the ALDH2*2 allele approximately 17 mM. CYP2E1 oxidation of ethanol can generate
drink alcohol, they experience a variety of aversive effects, including reactive oxygen species that react with cellular lipids to form highly
nausea, headache, and facial flushing, resulting from elevated acetal- mutagenic DNA adducts (Linhart et al., 2014). Thus, at high blood con-
dehyde concentrations in the body. Individuals who are homozygous centrations, a qualitatively different type of alcohol metabolism takes
for the ALDH2*2 allele have essentially undetectable ALDH2 activity, places in human cells, resulting in an additional mechanism of genotox-
and they generally cannot tolerate alcohol due to severe acetaldehyde icity separate from that involving acetaldehyde (Figure 12–2).
toxicity. Paradoxically, heterozygotes for the ALDH2*2 allele have an
enzyme activity roughly 1% of that of ALDH2*1 homozygotes, far
less than the 50% value typically expected. The ALDH2 enzyme is EXPOSURE ASSESSMENT
a functional tetramer, and the low enzyme activity among heterozy-
gotes suggests that the presence of even one inactive subunit may be Ethanol Content in Alcoholic Beverages
sufficient to prevent enzyme activity (Crabb et  al., 2004). However,
individuals who are heterozygous for the ALDH2*2 allele can become The content (concentration) of pure ethanol in alcoholic beverages
tolerant to the side effects of alcohol and can become heavy drinkers, is usually expressed either as percent of volume (% vol.), or “proof”

Ethanol Acetaldehyde Acetate


ADH ALDH

Cell death/hyper-regeneration
Mutagenesis
carcinogenesis
Acetaldehyde-DNA adducts

O O O OH

N NH N NH N N

N N N
N N CH3 N NH N N CH3
HO HO HO
O O O H
H3C
O
OH OH OH

N2-ethylidenedeoxyguanosine Crotonaldehyde-propanodeoxyguanosine adducts

Figure 12–​1.  Alcohol and acetaldehyde metabolism, acetaldehyde–​DNA adducts. Top: schematic diagram of enzymatic pathway of alcohol and acet-
aldehyde metabolism. As noted in the text, ALDH2 is the primary ALDH involved in the metabolism of acetaldehyde derived from alcohol metabo-
lism. Other ALDHs may be involved in the acetaldehyde metabolism in certain tissues. Bottom panel: Structures of acetaldehyde-​related DNA adducts.
Crotonaldehyde-​derived adducts can exist in either of two forms, ring-​opened (left) and ring-​closed (right). The ring-​opened forms can result in DNA
intrastrand crosslinks. ADH: alcohol dehydrogenase; ALDH: aldehyde dehydrogenase.
 215

Alcohol and Cancer Risk 215


Ethanol Acetaldehyde
CYP2E1

O2
Reactive oxygen H2O
species

Lipid peroxidation Ethenobase-DNA Mutagenesis


products adducts carcinogenesis

N N O

N N N N N
N N
N N O N N
HO HO
O HO O
O

OH OH
OH

1,N6 etheno-deoxyadenosine 3,N4-etheno-deoxycytidine 1,N2-ethenodeoxyguanosine

Figure 12–​2.  Alcohol metabolism by CYP2E1 and ethenobase DNA adducts. Top: schematic diagram of alcohol metabolism by cytochrome P450 2E1
(CYP2E1). CYP2E1 utilizes molecular oxygen to oxidize ethanol to acetaldehyde, generating H2O in the process. CYP2E1 can also generate reactive
oxygen species, including superoxide radical and hydrogen peroxide, which can further react with cellular lipids, resulting in lipid peroxidation and etheno-​
base DNA adducts. The bottom panel shows the molecular structures of three ethenobase DNA adducts.

(which in the United States is twice the percent of alcohol by volume Surveillance and Epidemiologic Assessment
[e.g., 80 proof is 40% vol.]). The ethanol content is typically lowest of Alcohol Consumption
(4%–​5% vol.) in commercially produced beer, intermediate (about
12% vol.) in wine, and highest (about 40% vol.) in distilled spirits. For both surveillance and epidemiologic studies, the assessment of
However, the concentration of ethanol in each beverage type varies alcohol intake takes into account the volume and ethanol content of
widely (IARC, 2010). In beer, the ethanol content ranges from 2.3% beverage(s) consumed. Intake is expressed as the average amount
vol. to over 10% vol., with home-​or locally produced beverages such of pure ethanol (in g) consumed per person per unit time.
as sorghum beer having lower content. The ethanol content in wine At the population level, surveillance data of alcohol consumption typi-
ranges from 8% to 15% vol., and in spirits ranges from 20% vol. cally reflect annual consumption of pure ethanol per capita, including
(aperitifs) to well over 40% vol. (e.g., 80% vol. in certain kinds of both recorded data and estimates of unrecorded data. WHO estimates
absinthe). average annual per capita consumption for ages 15+ years to include
The absolute amount of pure ethanol contained in a serving of older adolescents as well as adults (World Health Organization, 2014).
alcoholic beverage is estimated by multiplying the standard volume Estimates of recorded consumption derive from official statistics on
of an alcoholic drink by its alcohol content. The amount is expressed production, trade, taxes, and/​or sales. However, estimates of unrecorded
either as mL or as grams of pure ethanol (1 mL of ethanol = 0.79 g). consumption, which comprises nearly 25% of all alcohol consumed, may
The volume of a standard alcohol drink in the United States is gen- include formal or informal accounts of domestic production, illegal sales,
erally 12 ounces of beer, 5 ounces of wine, or 1.5 ounces of distilled smuggling and cross-​border shopping, and in some parts of the world a
spirits (Centers for Disease Control and Prevention, 2016). Thus, substantial fraction of alcohol consumption is unrecorded (World Health
for example, a 12 oz. serving of beer with an ethanol content of Organization, 2014). This is particularly important when estimating
5% vol. contains about 14 grams of ethanol (i.e., 355 mL × 0.05 × consumption in the poorest regions of Africa, Asia, and South America,
0.79 g/​mL). and where drinking is prohibited by religion. For example, although the
total level of consumption is very low in Islamic countries in the Eastern
Mediterranean Region, the proportion contributed by unrecorded con-
sumption is high (i.e., more than 50% of consumption). Consumption by
Acetaldehyde Content in Alcoholic Beverages
tourists is excluded from these estimates where possible.
Although most exposure to acetaldehyde from consuming alcoholic In large epidemiologic studies of cancer risk, individual alcoholic
beverages results from ethanol metabolism, high concentrations beverage consumption is often self-​reported on food frequency ques-
of preformed acetaldehyde are present in alcoholic beverages that tionnaires (FFQ). An FFQ queries study participants on the average
are commonly consumed in many countries (IARC, 2012). Average frequency of consumption (number of days per week or month), the
levels vary from 9 mg/​L in beer, 34 mg/​L in wine, 66 mg/​L in spir- usual quantity consumed (number of drinks), and the types of alco-
its, and 90 mg/​L in unrecorded alcohol (Lachenmeier et  al., 2012). holic beverages (e.g., spirits, can/​bottle of beer, glass of red or white
Concentrations are especially high in distilled beverages from Brazil, wine) consumed during a specified time period (usually the previous
the People’s Republic of China, Guatemala, Mexico, and the Russian year). Intake is then characterized in terms of either average consump-
Federation, as well as in fortified wines such as calvados and spirits tion of ethanol by weight (expressed as grams [g]‌per day) or the aver-
distilled from fruit in Europe. age number of alcoholic beverages consumed per day or per week.
216

216 PART III:  THE CAUSES OF CANCER


The validity of self-​reported information on alcohol consumption AMR (61.5%). Interestingly, only 14.7% of the world’s population
has been questioned because of poor recall, inaccurate estimation of the aged 15+ years lives in the EUR, but residents of this region account
number of drinks consumed on drinking days, inability to report aver- for more than 25.7% of global consumption.
age intake according to standard drink sizes (Stockwell et al., 2004), Males are 1.6 times more likely to be current alcohol drinkers than
and variable drinking patterns (Boniface et al., 2014). To address some females (47.7% vs. 28.9%, respectively). This gender difference varies
of these concerns, several large epidemiologic studies compared alco- regionally, and is greater in the SEAR (i.e., 21.7% of males vs. 5% of
hol intake data estimated from daily diaries or multiple 24-​hour recalls females) than in the EUR or AMR, where the ratio of male to female
to alcohol intake data obtained from FFQs, and found the correlations current drinkers is 1.2 and 1.3, respectively. In the EUR, 73.4% of
to be high (range from 0.7 to 0.9) (Giovannucci et  al., 1991; Kaaks males and 59.9% of females are current drinkers.
and Riboli, 1997). These findings suggest that the self-​reported data on Worldwide, 61.7% of the population aged 15+ years did not drink
usual alcohol consumption are reasonably consistent across different alcohol in the past 12 months; about half of the population report life-
methods of collection. However, this comparison does not address all long abstinence. There is wide variation in the prevalence of absten-
issues of underreporting, such as differences between actual drink size tion across WHO Regions, but females are more likely to be abstainers
consumed and standard drink sizes. While misreporting does not influ- than males.
ence the conclusion that alcohol is a human carcinogen, it can affect the
quantitative estimates of the cancer risk associated with a given amount
of consumption. Therefore, methods have been developed to correct Per Capita Amount of Alcohol Consumed
estimates of relative risk (Rosner, 1995), and population-​attributable In 2010, the global average annual consumption per capita of pure
risk for errors in the measurement of alcohol consumption (Shield ethanol among persons aged 15+ years was estimated to be about 6.2
et al., 2013). liters, or about 13.5 g/​day per capita. The average annual per capita
Another source of error occurs when weekly alcohol consumption consumption was highest in the EUR with 10.9 liters and the lowest in
is divided by seven to estimate the number of drinks consumed per the EMR with 0.7 liters.
day. As discussed elsewhere (Brooks and Zakhari, 2013), consum- Since only a subset of the population consumes alcohol, the
ing seven drinks per week might not correspond to consuming one worldwide average annual alcohol consumption per capita is
drink per day. An individual who drinks three to four drinks on each considerably higher when the estimate is restricted to drinkers.
weekend night, but nothing during the rest of the week, might report The average for those who drink alcohol is 17.2 liters per year,
seven drinks per week. However, this pattern of consumption could with a range of 11.3 liters in the EMR to 23.1 liters in the SEAR.
produce a blood alcohol level sufficient to induce CYP2E1, which Worldwide, men who drink alcohol consume more than twice as
would not be induced by actual consumption of one drink per day. much than women; the average annual per capita consumption
As noted earlier, the induction of CYP2E1 has distinct biological and among male drinkers (aged 15+ years) was 21.2 liters and among
genotoxic consequences, with mechanistic implications for alcohol-​ female drinkers was 8.9 liters of pure ethanol. The sex difference
related carcinogenesis. in average annual alcohol consumption among drinkers varies con-
siderably across WHO regions; in the SEAR annual consumption is
GLOBAL PATTERNS OF ALCOHOL CONSUMPTION 3.2 times greater among male than among female drinkers, and in
the AFR annual consumption is 1.7 times greater among male than
The Global Information System on Alcohol and Health provides infor- among female drinkers.
mation on 180 alcohol-​related indicators for 194 WHO member states
and two associate members. This information is used for the WHO
Global Status Report on Alcohol and Health and is a valuable resource Prevalence of Heavy Episodic Drinking
for worldwide, regional, and country-​specific estimates of total alco- Globally, approximately 7.5% of the total population aged 15+ years
hol consumption (both recorded and unrecorded) (World Health and 16.0% of drinkers aged 15+ years engage in heavy episodic drink-
Organization, 2014). Summaries of international statistics on alcohol ing (i.e., defined as 60 or more g of pure ethanol on at least one occa-
consumption have been reported for the six WHO regions:  Region sion at least monthly).
for the Americas (AMR), South-​East Asia Region (SEAR), European
Region (EUR), Eastern Mediterranean Region (EMR), African Region
(AFR) and Western Pacific Region (WPR). These region-​specific sum- Trends in Consumption
maries provide a useful comparison of alcohol consumption across
the world. In some parts of the world, alcohol consumption increased from 2005
to 2010, whereas in other parts of the world, consumption decreased or
remained stable. For example, the prevalence of current alcohol drink-
Types of Alcoholic Beverages Consumed ing in those aged 15+ years increased from 58.3% to 61.5% in the
AMR, from 3.5% to 5.4% in the EMR, and from 10.7% to 13.5% in
Globally, distilled spirits comprise 50.1% of total recorded alcohol
the SEAR. Conversely, from 2005 to 2010 the prevalence of current
consumption, beer accounts for 38.8%, wine for 8.0%, and other
drinking decreased in the EUR from 68.8% to 66.4%, and in the WPR
commercially produced alcoholic beverages for the remainder.
from 56.3% to 45.8%, but remained stable in the AFR at approximately
Consumption patterns vary regionally, however. Wine consumption
29%. Worldwide the total average annual per capita alcohol consump-
accounts for one-​fourth (25.7%) of total consumption in the WHO
tion remained stable between 2005 and 2010 at approximately 6.1 and
EUR and one-​ninth (11.7%) of total consumption in the WHO AMR.
6.2 liters, respectively. However, there was a slight decrease in con-
Beer accounts for over-​half (55.3%) of the consumption of alcoholic
sumption in the EUR from 12.2 to 10.9 liters and an increase in con-
beverages in the WHO AMR.
sumption in the SEAR from 2.2 to 3.4 liters. Some of the differences
in the prevalence of current drinkers and in the average annual amount
Prevalence of Current Alcohol Drinkers and consumed per capita are due to changes over time in the available data
and methods to estimate consumption, and therefore these differences
Abstainers should be interpreted cautiously.
Worldwide, an estimated 38.3% of the population aged 15+ years Per capita alcohol consumption among those aged 15+ years is
report current alcohol consumption. However, there are substantial expected to increase slightly through 2025. In particular, without
differences in the prevalence of current drinkers by WHO region. The effective policies to prevent an increase, the highest increase in aver-
lowest prevalence of current drinkers is in the EMR (5.4%) and the age annual per capita consumption is expected to occur in the WPR
highest prevalence is in the EUR (66.5%), followed closely by the (i.e., from 6.8 liters to 8.3 liters).
 217

Alcohol and Cancer Risk 217


Patterns of Consumption by the Economic Wealth carcinomas (IARC, 2012). Compared to alcohol non-​drinkers, people
of a Country who consume about 50 g of ethanol per day have about a 2-​to 3-​
fold higher risk of UADT cancers (IARC, 2010). It is worth noting
The economic wealth of a country correlates with the prevalence of that acetaldehyde, which can be formed during FPM in the oral cavity,
current drinking, the amount consumed, and the prevalence of heavy has a boiling point of 25ºC, which is well below body temperature.
episodic drinking. The prevalence of current drinking is 18.3% in This raises the possibility that acetaldehyde vapor may play a role in
low-​income countries, 19.6% in middle-​income countries, 45% in alcohol-​related UADT cancers. Experimental studies in animals have
upper middle-​income countries, and 69.5% in high-​income countries. shown that chronic exposure to acetaldehyde vapor increases UADT
Similarly, average annual per capita alcohol consumption in the total metaplasia (Woutersen et al., 1986).
population aged 15+ years ranged from 3.1 liters in low-​income coun- A significant confounder or effect modifier of the association
tries to 9.6 liters in high-​income countries. Furthermore, the preva- between alcoholic beverage consumption and UADT cancer risk is
lence of heavy episodic drinking among those who drink alcohol is smoking. Smoking causes multiple types of cancer, and alcohol drink-
11.6% in low-​income countries to 22.3% in high-​income countries. ers, particularly heavy drinkers, are more likely to be smokers than
non-​drinkers. Controlling for smoking in statistical models only partly
separates the risk due to alcohol consumption from that due to smoking.
TYPES OF CANCER CAUSED BY ALCOHOL A more thorough approach is to study the relationship between alcohol
CONSUMPTION and cancer in lifelong non-​smokers. Several early studies showed a
significant positive association between alcohol intake and risk, even
The consumption of alcoholic beverages was first classified as carci- in never smokers (IARC, 2010). A meta-​analysis of 15 studies found
nogenic to humans in 1987 by the International Agency for Research alcoholic beverage consumption (4 or more drinks per day) was most
on Cancer (IARC, 1988). At that time, the evidence for causality was strongly associated with increased risk of pharynx cancer, but was also
deemed to be “sufficient” for oral cavity, pharynx, larynx, esophagus, associated with oropharynx, larynx, and esophageal squamous cell
and liver cancer. Although results of some studies had shown that carcinoma among those who did not use tobacco (Zeka et al., 2003).
alcohol consumption was also positively associated with the risk of In a pooled analysis of case-​control studies, heavy alcohol consump-
colorectal and female breast cancer, the evidence for these sites was tion (3 or more drinks per day) was associated with an increased risk
considered inconclusive. Importantly, no conclusions were made of cancers of the oropharynx/​hypopharynx and larynx in never users of
about the carcinogenic agent in alcoholic beverages (e.g., ethanol or tobacco (Hashibe et al., 2007). There is extensive evidence of a strong
other constituents of alcoholic beverages) at that time. synergistic effect of tobacco smoking and alcohol consumption on the
Another expert Working Group (which included both authors of this risk of UADT cancer, even at moderate amounts of consumption. In
chapter) convened by the IARC in 2007 reviewed the epidemiologic most epidemiologic studies, the interaction is greater than multipli-
and other evidence on the associations of alcoholic beverage con- cative (IARC, 2012). In a large prospective study of British women,
sumption and cancer at 27 anatomic sites (IARC, 2010). This Working alcohol consumption was most strongly associated with increased risk
Group concluded that “ethanol in alcoholic beverages” is carcinogenic of UADT cancers among current smokers, less strongly among former
to humans (Group  1). The Group found no consistent evidence that smokers, and not at all among never smokers (Allen et al., 2009). One
the effects of alcoholic beverage consumption on cancer differed by of the two proposed mechanisms by which alcohol consumption might
beverage type, and deemed the evidence that ethanol causes cancer potentiate the carcinogenicity of tobacco smoking depends on dose.
in experimental animals to be sufficient. They reaffirmed that alcohol CYP2E1, which is induced by heavy alcohol consumption, can acti-
consumption causes cancers of the liver and UADT, including the oral vate procarcinogens in cigarette smoke (Cederbaum, 2012). However,
cavity, pharynx, larynx, and esophagus. Moreover, they expanded the since the interaction between smoking and alcohol consumption has
list of cancers caused by alcohol consumption to include colorectal been observed even at low levels of alcohol consumption, where the
and female breast. induction of CYP2E1 is unlikely, alcohol might also act as a “solvent,”
The 2007 Working Group also concluded that both non-​Hodgkin enhancing the hydrophobic diffusion of carcinogens present in ciga-
lymphoma and kidney cancer were not caused by alcohol consump- rette smoke into epithelial tissues (Squier et al., 1986).
tion. However, the evidence linking alcohol consumption to other can- Much of the evidence that acetaldehyde associated with alcoholic
cer sites was considered inconclusive. For example, no conclusions beverage consumption is a cause of UADT cancer comes from a
were drawn concerning lung or stomach cancer due to potential con- series of epidemiologic studies by Yokoyama and colleagues showing
founding by tobacco smoking and diet. increased risk of esophageal squamous cell carcinoma (ESCC) among
In 2009, a Working Group convened by the IARC again reviewed ALDH2*2 heterozygous-​alcoholics (Yokoyama et al., 2006). As noted
the evidence regarding alcohol consumption and updated and con- earlier, the adverse effects of alcohol in ALDH2*2 homozygous indi-
firmed the conclusions of previous Working Groups (IARC, 2012). viduals are so severe that they are generally unable to consume signifi-
The evidence that heavy alcohol intake was associated with a small cant amounts of alcohol. However, some ALDH2*2 heterozygotes are
increase in pancreatic cancer risk was considered limited. For the first able to overcome the adverse effects of acetaldehyde related to alcohol
time, the 2009 Working Group concluded that both ethanol and acetal- drinking, and become heavy drinkers. When matched for alcohol con-
dehyde associated with the consumption of alcoholic beverages were sumption and smoking, ALDH2*2 heterozygotes have a significantly
carcinogenic to humans for cancers of the UADT. elevated risk of ESCC compared to those with fully active ALDH2
As described later in the chapter, the carcinogenic effects of ethanol (reviewed by Brooks et al., 2009).
found in alcoholic beverages and the primary metabolite of ethanol, Using a Mendelian randomization approach, a meta-​analysis of
acetaldehyde, vary by cancer type. Broadly, these mechanisms involve studies examining ALDH2 genotype and esophageal cancer risk dem-
DNA and protein damage from acetaldehyde and oxidative stress, onstrated a reduced risk for ALDH2*2 homozygous individuals (odds
alterations in DNA repair, cell death and hyper-​regeneration, nutri- ratio [OR]  =  0.36; 95% confidence interval [CI]:  0.16, 0.80) and a
tional malabsorption, changes in DNA methylation, and metabolic higher risk among ALDH2*2 heterozygotes (OR = 3.19; 95% CI: 1.86,
effects, and for breast cancer, increased estrogen levels (Seitz and 5.47), compared with ALDH2*1 fully active homozygotes (Lewis and
Stickel, 2007). In addition, carcinogenic contaminants can be intro- Smith, 2005). The lower risk of esophageal cancer in ALDH2*2 homo-
duced during alcoholic beverage production. zygotes likely reflects their avoidance of alcohol. When stratified by
alcohol drinking status, ALDH2*2 heterozygosity was not associated
with risk of esophageal cancer in non-​drinkers, but there was a 2.5-​fold
Upper Aerodigestive Tract higher risk among moderate drinkers and a 7-​fold higher risk among
Alcoholic beverage consumption is a known cause of cancers of the heavy drinkers. Similarly, in a meta-​analysis of studies of ALDH2
UADT, including the oral cavity, pharynx, larynx, and esophagus genotype and head and neck cancer risk, there was a reduced risk for
(IARC, 1988). The causal relationship is strongest for squamous cell ALDH2*2 homozygous individuals (OR = 0.53; 95% CI: 0.28, 1.00)
218

218 PART III:  THE CAUSES OF CANCER


and a higher risk among ALDH2*2 heterozygotes (OR = 1.83; 95% risk of colorectal cancer compared to non-​ drinkers; however, there
CI: 1.21, 2.77) as compared with fully active ALDH2*1 homozygotes; was uncertainty regarding the nature of the dose–​response relation-
the association with ALDH2*2 heterozygosity also varied accord- ship (IARC, 2010). A systematic literature review by the WCRF/​AICR
ing to the level of alcohol consumption, with no association among CUP designated the evidence for a causal association between ethanol
never drinkers and 3.6-​fold higher risk among heavy drinkers (Boccia from alcoholic consumption and colorectal cancer risk as convincing in
et al., 2009). Taken together, these studies support the evidence that men and probably convincing in women (World Cancer Research Fund
alcoholic beverage consumption can increase risk of UADT cancers International, 2011). More specifically, the CUP meta-​analyses of pro-
through the carcinogenic effects of acetaldehyde. spective studies showed that a 10 g per day increase in ethanol was asso-
ciated with a 10% increased risk of colorectal and rectal cancers, and an
8% increased risk of colon cancer. The association was stronger among
Liver men than women for both colorectal and colon cancers. The risk of
The association between alcohol consumption, particularly heavy alco- colorectal cancer increased by 11% among men and 7% among women
hol consumption, and liver cancer (i.e., hepatocellular carcinoma) is well per 10 g per day increase in ethanol. Reasons for the gender difference
established (Turati et al., 2014). This association has been observed in are unclear, but may relate to differences in the gut microbiome, or hor-
studies of individuals infected with hepatitis viruses (HV) B or C, and in mone-​related differences in alcohol metabolism (Park et al., 2006).
unaffected individuals without cirrhosis. A meta-​analysis of 19 prospec- It is unclear whether tobacco use—​an established cause of colorec-
tive studies that excluded cohorts of alcoholics, patients with cirrhosis, tal cancer—​modifies the association between alcohol consumption
and individuals with HBV or HCV infection showed that consumption and colorectal cancer risk. In the pooled analysis of eight prospective
of 3 or more drinks per day was associated with a 16% increased risk studies mentioned in the preceding paragraph (Cho et al., 2004), the
compared to non-​drinkers, although at this level there was considerable association between alcohol consumption and colorectal cancer risk
heterogeneity among studies (p = 0.002) (Turati et al., 2014). The pooled was statistically significant among past and current smokers, but not
relative risk for heavy consumption of 6 or more drinks per day was 1.22 among never smokers; however, the test for interaction was not sta-
(95% CI: 1.10, 1.35) with no evidence of heterogeneity among studies tistically significant (p > 0.2). Similarly, in the European Prospective
(p = 0.8). However, because most liver cancers occur in the context of Investigation into Cancer and Nutrition, alcohol consumption was
cirrhosis, which can motivate individuals to reduce their alcohol con- more strongly associated with colorectal cancer risk among smokers
sumption, estimates of the dose–​response relationship between alcohol than among non-​smokers, but again there was no evidence of a statisti-
consumption and liver cancer should be interpreted cautiously. Findings cally significant interaction (p = 0.41) (Ferrari et al., 2007). Another
from several studies suggest possible synergistic effects of alcohol with study showed a marginally significant interaction between smoking
other established risk factors for hepatocellular carcinoma, including and alcohol consumption for colon cancer (p = 0.051) but not for rectal
HBV and HCV infection (Hassan et al., 2002; Yuan et al., 2004), obesity cancer (p > 0.19) (Tsong et al., 2007). It is plausible that the combined
(Loomba et al., 2010, 2013; Marrero et al., 2005), and smoking (Kuper exposure to smoking and alcohol could modify their separate effects
et al., 2000; Marrero et al., 2005). More research from prospective stud- on colorectal cancer risk. As noted previously for UADT cancers, alco-
ies is needed to confirm these findings. hol might function as a solvent, enhancing the penetration of other
Most liver cancers occur in the context of viral hepatitis, fibrosis, carcinogenic molecules into mucosal epithelium (Squier et al., 1986).
and/​or cirrhosis (El-​Serag and Rudolph, 2007). Chronic heavy alco- There is compelling evidence for a causal relationship between
hol consumption often leads to fibrosis and ultimately cirrhosis, and acetaldehyde derived from alcohol metabolism and colorectal cancer.
alcohol-​related liver cancer in the absence of these conditions is rare Experiments in animals have shown that alcohol metabolism by colonic
(Seitz and Stickel, 2007). Alcohol-​related liver disease not only causes bacteria leads to very high (up to mM) concentrations of acetaldehyde
cell death and regeneration, but also results in chronic inflammation (Jokelainen et  al., 1996; Visapaa et  al., 1998). Although no similar
(Wang et al., 2012), which can itself generate mutagens and damage measurements have been made in humans, it is not unreasonable to
DNA (Son et  al., 2008)  beyond the effects of alcohol metabolism. assume that acetaldehyde levels in the human colon could reach the
More specifically, inflammation and oxidative stress resulting from same concentrations after alcohol consumption, at least in some indi-
elevated levels of CYP2E1 can increase levels of etheno DNA-​base viduals. Variation between individuals might be substantial, depending
adducts, some of which are highly mutagenic (Pandya and Moriya, upon the composition of the colonic microbiome. The mechanisms by
1996). Elevated levels of etheno DNA-​base adducts have been docu- which acetaldehyde affects the risk of colorectal cancer are not com-
mented in tissue biopsies from patients with alcoholic liver disease pletely understood. Plausible mechanisms could include DNA adducts
(Frank et al., 2004; Wang et al., 2009). from acetaldehyde, and increased cell division in regenerating tissue.
Although the liver is the major site of alcohol metabolism, acetalde- Other mechanisms have also been considered (Seitz and Stickel, 2007).
hyde has not been implicated in liver carcinogenesis. Alcoholics defi- While there is some evidence that ALDH2-​deficient individuals are
cient in ALDH2 activity have little or no increase in liver cancer risk at increased risk of alcohol-​associated colorectal cancer, more data are
(IARC, 2010), despite their dramatically higher risk of UADT cancers. needed to confirm this finding (IARC, 2010). It is notable that ALDH2
This difference in the effect of acetaldehyde on the liver compared is maximally active at low acetaldehyde concentrations. If the levels of
to the UADT may be due, at least in part, to the much lower prolif- colonic acetaldehyde after alcohol consumption are roughly comparable
eration rates of hepatocytes compared to epithelial cells in the aerodi- in humans and animals, the role of ALDH2 compared to other ALDHs
gestive (and gastrointestinal) tract. In the absence of DNA synthesis expressed in the colon would be limited. Thus, the lack of a large difference
and cell replication, mutations resulting from DNA damage are not in colon cancer risk between ALDH2-​proficient and ALDH2-​deficient
transmitted to daughter cells, and there is more time for DNA repair alcohol drinkers does not exclude the possibility that acetaldehyde might
to occur. Proliferating cells also have higher levels of polyamines, contribute to alcohol-​related colorectal carcinogenesis.
which can react with acetaldehyde and lead to mutagenic DNA dam- Other mechanisms that could potentially mediate the effects of
age (Theruvathu et al., 2005). alcohol on colorectal carcinogenesis include oxidative stress, altered
DNA methylation, and nutritional deficiencies, such as reduced lev-
els of folic and retinoic acid as a result of heavy drinking (Seitz
Colorectum et al., 2005).

The IARC first concluded that alcoholic beverage consumption caused


colorectal cancer in 2007 (IARC, 2010). The evidence at that time was Breast
based partly on a pooled analysis of eight cohort studies (Cho et  al.,
2004) and a meta-​analysis of 16 prospective studies (Moskal et  al., Based on a large body of evidence, the 2007 IARC Working Group
2007). These analyses showed that regular consumption of about 50 g concluded for the first time that alcohol consumption is a cause of
of ethanol per day was associated with an approximately 40% higher breast cancer in humans (IARC, 2010). Included in the IARC review
 219

Alcohol and Cancer Risk 219


was a large pooled analysis of 53 epidemiologic studies with more than associated with higher risk of benign breast disease, another estab-
58,000 women with breast cancer. In that analysis, there was a linear lished breast cancer risk factor.
dose–​response association with a 7.1% (95% CI: 5.5, 8.7) increase in Alcohol consumption also increases circulating estrogen levels,
breast cancer risk per 10 g per day increase in ethanol consumption which are positively associated with breast cancer risk in both pre-
(Hamajima et al., 2002). Subsequently, at least 13 additional prospec- menopausal (Endogenous et al., 2013) and postmenopausal women
tive studies were published. Based on the total evidence, a 2009 IARC (Key et  al., 2002). In a controlled feeding study among premeno-
Working Group confirmed the causal relationship between alcohol and pausal women, consumption of 30 g/​day of ethanol for three men-
breast cancer risk (IARC, 2012). strual cycles increased plasma estradiol and estrone levels by 28%
In general, most factors do not appear to modify the relationship and 21%, respectively (Reichman et al., 1993). Similarly, in a ran-
between alcohol and breast cancer risk. In the aforementioned pooled domly assigned cross-​over alcohol feeding study among postmeno-
analysis of 53 case-​control and prospective studies, there was no evi- pausal women, both 15 and 30 g/​day of ethanol increased serum
dence of effect modification by age at diagnosis, parity, age at first estrone sulfate and dehydroepiandrosterone concentrations com-
birth, breastfeeding, race, country, education, mother or sister with pared to placebo, but did not affect other sex hormones, including
breast cancer, age at menarche, height, weight, body mass index, ever estradiol and testosterone (Dorgan et al., 2001). In a pooled analysis
use of hormonal contraceptives, ever use of hormone replacement of 13 prospective studies of postmenopausal women, alcohol con-
therapy, or menopausal status (Hamajima et al., 2002). The possible sumption was positively associated with both estrogens and andro-
exception to these null findings is smoking. A  recently published gens, and inversely associated with sex hormone–​binding globulin
pooled analysis of data from 14 prospective studies with over 934,000 (Endogenous et  al., 2011). Further evidence that alcohol might
participants and 36,060 invasive breast cancer cases showed a statisti- act through a hormonal mechanism is provided by epidemiologic
cally significant interaction between alcohol and smoking status (p-​ studies showing a stronger association between alcohol consump-
heterogeneity = 0.03) (Gaudet et al., 2016). tion and risk of hormone-​receptor-​positive breast cancer than with
The mechanism(s) underlying the association between alcohol and hormone-​receptor-​negative breast cancer. In a 2008 meta-​analysis
breast cancer risk are not completely understood. Alcohol consump- of three prospective cohort studies and 16 case-​control studies, the
tion typically begins in late adolescence and continues throughout life, summary relative risk for the highest versus the lowest categories of
and thus it is difficult to isolate the biologically relevant time period of alcohol consumption were 1.27 (95% CI: 1.17, 1.38) for estrogen-​
exposure (Brooks and Zakhari, 2013). receptor-​positive (ER+) breast cancer and 1.14 (95% CI: 1.03, 1.26)
One proposed mechanism involves the genotoxicity of acetalde- for estrogen-​receptor-​negative (ER–​) breast cancer (Suzuki et  al.,
hyde in breast cells. ADH enzymes produced by human breast epi- 2008). When further stratified on ER and progesterone-​receptor
thelial cells metabolize alcohol, even at low concentrations (Triano (PR) status, high versus low alcohol consumption was associated
et al., 2003), suggesting that acetaldehyde is generated at relatively with statistically significantly elevated risks of both ER+/​PR+ (RR
low levels of consumption. An important unanswered question is = 1.22; 95% CI: 1.11, 1.34) and ER+/​PR–​(RR = 1.28; 95% CI: 1.07,
whether those cells also express ALDH2, which would metabolize 1.53) breast cancer but not with ER–​/​PR–​breast cancer (RR = 1.10;
the resulting acetaldehyde. ALDH2 protein is detectable in human 95% CI: 0.98, 1.24). Overall, the 2010 IARC Working Group could
breast tissue (Sutton et al., 2010), but the cellular localization has not conclude that the association between alcohol consumption and
not been reported. breast cancer risk differed by hormonal status (IARC, 2012).
Binge drinking can raise blood alcohol levels into the range at which An argument against a significant role for estrogen in alcohol-​
CYP2E1 is induced, resulting in mutagenic DNA damage from free related breast cancer is the absence of an association between alcohol
radicals and lipid peroxidation. As noted earlier, this mechanism does consumption and risk of endometrial (Friberg et  al., 2010)  or ovar-
not involve acetaldehyde. Relatively few epidemiologic studies have ian cancer (Genkinger et  al., 2006), two other cancer sites strongly
examined binge drinking in relation to breast cancer risk. In the Danish related to hormones. Based on evidence from WCRF/​AICR CUP sys-
Nurse Cohort Study, binge drinking during the weekend was associ- tematic literature reviews, the relative risk per 10 g per day increase
ated with increased breast cancer risk (relative risk [RR] = 1.49; 95% in ethanol consumption was 1.01 (95% CI: 0.97, 1.06) for endometrial
CI: 1.04, 2.13 for 10–​15 drinks; and RR = 2.51; 95% CI: 1.37, 4.59 cancer (World Cancer Research Fund International, 2013), and 1.01
for 16–​21 drinks) compared to drinking 1–​3 drinks during the latest (95% CI: 0.96, 1.06) for ovarian cancer (World Cancer Research Fund
weekend (Morch et al., 2007). In the US Nurses’ Health Study, binge International, 2014a).
drinking, but not the frequency of drinking, was positively associated Finally, moderate alcohol consumption has been hypothesized
with breast cancer risk after adjustment for cumulative alcohol intake to increase risk of breast cancer by increasing breast density. Breast
(Chen et al., 2011). density reflects fibroglandular breast tissue that appears radiologically
Binge drinking may be particularly deleterious for younger dense on mammograms, and is consistently, strongly, and positively
women, who are most likely to binge drink. Breast epithelium is associated with breast cancer risk (Boyd et  al., 1998). However, as
thought to be most susceptible to possible carcinogens between reviewed by Liu et  al. (2015), the association between alcohol con-
menarche and first full-​term pregnancy, a period of time when breast sumption and breast density is inconsistent among epidemiologic
cells proliferate rapidly but have not yet terminally differentiated. studies.
It is well established that a longer duration between menarche and
age at first full-​term pregnancy is associated with a higher risk of
breast cancer (Li et  al., 2008). The association between alcohol
Pancreas
consumption between menarche and first pregnancy was examined
in the Nurses’ Health Study (Liu et al., 2013). In analyses adjust- Although the 2009 IARC Working Group cited accumulating evi-
ing for consumption after first pregnancy, a 10 g per day increase dence that high alcohol intake (i.e., ≥ 30 g per day) was associated
in cumulative average ethanol consumption between menarche with a small increased risk for cancer of the pancreas, the evidence
and first pregnancy was associated with an 11% increase in risk of was considered “limited.” The available studies had insufficient
breast cancer (RR = 1.11; 95% CI: 1.00, 1.23). This association was statistical power to examine the association for heavy consump-
comparable to the association with cumulative average consumption tion (e.g., 3 or more drinks per day), or to eliminate potential con-
after first pregnancy (RR = 1.09; 95% CI: 0.96, 1.23 per 10 g per day founding by cigarette smoking (IARC, 2012). Subsequently, the
increase). Notably, the association between alcohol consumption association between alcohol intake and pancreatic cancer mortality
between menarche and first pregnancy and risk of breast cancer was was examined in the American Cancer Society’s Cancer Prevention
limited to women with 10 or more years’ duration of consumption Study-​II (CPS-​II), a large prospective study of over 1  million US
(RR = 1.21; 95% CI: 1.08, 1.36 per 10 g per day increase). Similarly, adults. Based on 24 years of follow-​up and almost 7,000 pancreatic
cumulative average ethanol consumption between menarche and cancer deaths, alcohol consumption of 3 or more drinks per day was
first pregnancy, but not consumption after full-​term pregnancy, was associated with an increased risk of death from pancreatic cancer in
20

220 PART III:  THE CAUSES OF CANCER


lifelong never smokers (RR  =  1.36; 95% CI:  1.13, 1.62) (Gapstur In a large meta-​analysis of 15 prospective studies, moderate, but
et  al., 2011). These results were included in a 2012 WCRF/​AICR not heavy, alcohol consumption was associated with a reduced risk of
CUP systematic literature review, which characterized the evidence type 2 diabetes (Koppes et al., 2005). This finding might be relevant
for a causal association between heavy alcohol consumption and to the inverse relationship between alcohol consumption and kidney
risk of pancreatic cancer as limited (World Cancer Research Fund cancer. Type 2 diabetes was associated with higher risk of kidney
International, 2012). cancer in a large well-​designed prospective study that updates dia-
A causal relationship between heavy alcohol consumption and betes status during follow-​up (Joh et al., 2011). Alternatively, alco-
pancreatic cancer is biologically plausible, since alcohol causes hol has diuretic effects that could reduce the exposure of renal cells
chronic alcoholic pancreatitis (Dufour and Adamson, 2003), an to carcinogens.
established risk factor for pancreatic cancer. Go et al. (2005) hypoth-
esized that oxidative and non-​oxidative metabolism of alcohol in the
pancreas causes inflammation and promotes carcinogenesis through ESTIMATES OF ATTRIBUTABLE FRACTIONS
activation of nuclear transcription factors, increased production of
reactive oxygen species, and dysregulation of cell proliferation and Estimates of the percent of cancer deaths attributable to alco-
apoptosis. hol consumption have been remarkably stable over the past four
decades. Rothman et al. estimated that 3% of all cancer deaths in the
United States in 1974 could be attributed to alcohol consumption
Non-​Hodgkin Lymphoma (Rothman, 1980); Doll and Peto estimated that between 2% and 4%
of US cancer deaths in the late 1970s could be attributed to alcohol
The 2007 IARC Working Group concluded that for non-​Hodgkin
consumption (Doll and Peto, 1981). A recent comprehensive analy-
lymphoma (NHL), there was evidence for lack of carcinogenicity
sis by Nelson and colleagues (Nelson et  al., 2013)  estimated the
for alcohol consumption (IARC, 2010). This conclusion is sup-
population attributable fractions (PAFs) of alcohol-​related deaths
ported by a substantial body of evidence from more recent prospec-
for all cancers and for seven cancer sites (oral cavity, pharynx, lar-
tive studies showing statistically significant inverse, dose–​response
ynx, esophagus, liver, colorectum, and female breast) designated as
relationships with all NHL subtypes combined (Allen et al., 2009;
causally related to alcohol consumption by the IARC (IARC, 2010,
Chang et al., 2010; Chiu et al., 1999; Gapstur et al., 2012; Kanda
2012). These estimates were based on two different methods to esti-
et al., 2010; Klatsky et al., 2009; Lim et al., 2006, 2007; Troy et al.,
mate the prevalence of alcohol consumption and on data from two
2010). Notably, these prospective studies show no clear differences
different population-​based surveys (i.e., the 2009 Behavioral Risk
in the associations of alcohol consumption with specific histologic
Factor Surveillance System survey and the 2009–​2010 National
subtypes of NHL.
Alcohol Survey). They also considered alcohol sales data. Overall,
The inverse association between alcohol consumption and NHL
the different methods showed similar estimates of the attributable
might be an artifact of reverse causation, in which the disease influ-
fraction ranging from 3.2% to 3.7% (i.e., 18,178 to 21,284) for all
ences the exposure. Common symptoms of lymphoma include
US cancer deaths in 2009. In sex-​specific analyses, the proportion
enlarged lymph nodes, extreme fatigue, unexplained fever, weight
of all US cancer deaths due to alcohol ranged from 2.4% (6970)
loss, and night sweats. These symptoms, as well as several chronic
to 4.0% (11,820) among US men, and from 2.7% (7266) to 4.8%
diseases associated with lymphoma, might lead to pre-​diagnostic
(13,094) among women.
reductions in alcohol consumption, which could result in an appar-
Rehm and Shield have estimated that worldwide about 4.2% (i.e.,
ent inverse association with NHL risk. The possibility of reverse
337,400) of all cancer deaths could be attributed to alcohol consump-
causation is supported by findings from one prospective study that
tion in 2010 (Rehm and Shield, 2014). Deaths from cancers of the
found no difference in risk for current drinking but a higher risk for
nasopharynx, oropharynx, oral cavity, larynx, and esophagus are at
former drinking when compared to consistent non-​drinkers (Chang
least twice as likely to be alcohol-​related than deaths from cancers of
et  al., 2010). However, two other prospective studies found no
the liver, colorectum, or female breast. For all cancers combined, the
association with former alcohol consumption (Gapstur et al., 2012;
estimates of attributable proportion are higher among men (5.4%) than
Klatsky et al., 2009).
among women (2.7%). This also is true for most individual cancer
Limited experimental data suggest that alcohol consumption
sites, and especially for UADT cancers. Among men, 23.5% of deaths
might inhibit the development of NHL. Alcohol drinking has mul-
from laryngeal cancer and 37.0% of deaths from oral cavity cancer
tiple effects on the human immune system, both positive and nega-
were attributed to alcohol consumption, whereas among women only
tive (Hagner et  al., 2009; Romeo et  al., 2007). Empirical evidence
7.9% of laryngeal cancer deaths and 12.6% of nasopharynx cancer
that alcohol might inhibit the development of human lymphomas
deaths were attributed to alcohol consumption. Rehm and Shield also
comes from a human lymphoma xenograft mouse model, in which
reported rates of alcohol-​attributed cancer deaths by WHO Global
chronic exposure to 5%–​10% alcohol in water (an amount shown to
Burden of Disease regions. The highest alcohol-​ attributed cancer
lead to 0.05 to 0.15 g/​dL alcohol in circulation) decreased lymphoma
death rate (per 100,000) was in Eastern Europe (8.7), followed closely
growth; this effect was partially due to an inhibition of mTOR sig-
by East Asia (7.5) and Central Europe (7.2). These rates largely affect
naling (Hagner et  al., 2009). Similarly, ethanol was also shown to
men in those regions. The lowest alcohol-​attributed cancer death rates
inhibit mTOR signaling in another human lymphoma cell line in vitro
(per 100,000) were in North Africa/​Middle East (0.6), Andean Latin
(Mazan-​Mamczarz et  al., 2015). Multiple lines of evidence support
America (1.7) and parts of Sub-​Saharan Africa. Interestingly, men in
the development of mTOR pathway inhibitors for the treatment of
Andean Latin America have lower alcohol-​attributed deaths rates than
lymphoma (Arita et al., 2013).
women (1.4 vs. 2.0, respectively).

Kidney ALCOHOL CONSUMPTION AND PROGNOSIS AMONG


The 2007 IARC Working Group found a lack of evidence that CANCER SURVIVORS
alcohol was carcinogenic for kidney cancer (IARC, 2010). Indeed,
there is some evidence that consumption of alcoholic drinks may be In 2012, there were over 32.6  million people worldwide alive who
inversely associated with kidney cancer. A 2015 WCRF/​AICR CUP have lived at least 5 years after being diagnosed with cancer, other than
systematic literature review showed that a 10 g per day increase non-​melanoma skin cancer (Jemal et  al., 2014). Many cancer survi-
of ethanol was associated with an 8% reduction in risk of kid- vors are highly motivated to learn about and make healthful behavioral
ney cancer (RR  =  0.92; 95% CI:  0.86, 0.97); this association was changes to reduce their risk of recurrence and/​or disease progression.
observed up to 30 g per day of ethanol (World Cancer Research The most recent American Cancer Society Nutrition and Physical
Fund International, 2015). Activity Guidelines for Cancer Survivors advise cancer patients
 21

Alcohol and Cancer Risk 221


undergoing chemotherapy or radiation therapy to avoid or minimize chemoprevention trial, there was a statistically significant 30% (95%
alcohol consumption during treatment, because alcohol might affect CI: 1.0, 1.7) higher risk of second primary cancer among those who
chemotherapeutic clearance and worsen toxicity, compromise healing, continued to drink alcohol after diagnosis (Do et al., 2003). In another
or exacerbate treatment associated side effects such as oral mucositis chemoprevention trial of 264 patients with early-​stage cancer of the
(Rock et  al., 2012). However, whether alcohol consumption affects oral cavity, pharynx, and larynx randomized to β-​carotene, there was a
long-​term cancer survival is not completely understood. Issues to be dose–​response increase in total mortality for both pre-​diagnostic alco-
considered include the following: (1) the correlation between alcohol hol consumption and continuous alcohol consumption after diagno-
consumption before and after diagnosis; (2)  the stage of disease at sis; the RR of death from any cause during follow-​up was 2.72 (95%
diagnosis (because consumption is more likely to influence early-​than CI: 1.20, 6.14) times higher in those who continued to drink after diag-
late-​stage disease); (3) confounding by smoking (which is highly cor- nosis compared to non-​drinkers after diagnosis (Mayne et al., 2009).
related with alcohol consumption); and (4) assessment of all-​cause ver-
sus site-​specific cancer mortality (since alcohol consumption has been
associated with a reduced risk of cardiovascular disease mortality). OPPORTUNITIES FOR ALCOHOL PREVENTION
A WCRF/​ AICR CUP systematic literature review of 18 total AND CONTROL
studies published through June 2012 examined association of both
pre-​and post-​diagnostic alcohol consumption with total-​and breast Strategies to reduce the harmful use of alcoholic beverages include
cancer–​specific mortality, as well as risk of second primary breast measures to increase awareness and reduce consumption at both
cancer among breast cancer survivors (World Cancer Research Fund the population and individual level. In 2010, the 63rd World Health
International, 2014b). Results showed no associations of pre-​diagnos- Assembly of the WHO endorsed a Global Strategy to Reduce the
tic alcohol intake with total mortality (RR = 1.00; 95% CI: 0.99, 1.00 Harmful Use of Alcohol (World Health Organization, 2010). This
per 1 drink/​week increase), or breast cancer–​specific mortality (RR strategy was designed to support and complement public health poli-
= 1.00; 95% CI: 0.97, 1.02 per 1 drink/​week). Similarly, there was cies in WHO member states, to provide guidance for local, regional,
no evidence of associations between post-​diagnosis alcohol consump- and global action, and to set priorities for global action. The five objec-
tion assessed less than 12 months after diagnosis, or 12 months or tives of the WHO Global strategy are shown in Box 12–1. These objec-
more after diagnosis and total-​or breast cancer–​specific mortality. No tives parallel population-​level interventions to reduce tobacco use; in
study examined the association between pre-​diagnostic alcohol con- particular, increases in excise taxes to decreases sales are among the
sumption and risk of second primary breast cancer. In the two stud- most effective strategies (Eriksen et al., 2015). There is substantial evi-
ies that examined the association between consumption less than 12 dence that increasing the price of alcohol through excise taxes reduces
months after diagnosis and subsequent risk of second primary breast both consumption (Wagenaar et  al., 2009)  and adverse health out-
cancer, one reported no association and the other reported increased comes caused by alcohol, including motor vehicle crashes, violence,
risk (RR = 1.7). In the CUP meta-​analysis, five studies that investi- and cirrhosis (Elder et al., 2010; Wagenaar et al., 2010).
gated the association between alcohol consumption during the period Despite the evidence that alcohol causes multiple types of cancer,
12 or more months after diagnosis and risk of second primary breast fewer than half of the US Comprehensive Cancer Control Plans devel-
cancer showed a nearly statistically significant increased risk (RR = oped by the states and funded by the US Centers for Disease Control
1.19; 95% CI: 0.96, 1.47); however, the association was not dose-​ and Prevention (CDC) specify goals, objectives, or strategies for
related. Since that review, a large pooled analysis including more than alcohol control (Henley et al., 2014). In particular, efforts are needed
9300 stage I–​III breast cancer survivors also showed no associations of to increase awareness of the long-​term impact of alcohol drinking,
post-​diagnostic regular alcohol intake with risk of recurrence or with including binge drinking, on breast cancer risk in women. A  survey
total mortality overall (Kwan et al., 2013). However, postmenopausal of drinking patterns in the United States found that the prevalence of
women who regularly consumed at least 6 g/​day of ethanol had a statis- binge drinking increased more than twice as rapidly among women
tically significant higher risk of recurrence (RR = 1.19; 95% CI: 1.01, than men from 2005 to 2012 (Dwyer-​Lindgren et al., 2015). Effective
1.40) compared to non-​drinkers; no association was observed among population-​ wide education campaigns are needed to target young
premenopausal women (RR = 0.91; 95% CI: 0.72, 1.16). Overall, there women who drink alcohol and encourage them to limit consumption
is no clear evidence that alcohol consumption influences breast cancer and avoid binge drinking.
prognosis. At the individual level, the treatment of alcohol-​related disorders
At least nine epidemiologic studies have examined the influence includes mutual support groups, treatments with medications, behav-
of alcoholic beverage consumption on outcomes after a diagnosis of ioral therapies, and/​or combinations of these strategies. In the United
colorectal cancer (Asghari-​Jafarabadi et al., 2009; Fung et al., 2014; States, the National Institute on Alcohol Abuse and Alcoholism list
Park et al., 2006; Patel et al., 2014; Pelser et al., 2014; Phipps et al.,
2011, 2016; Walter et  al., 2016; Zell et  al., 2007). The results were
inconsistent in studies that examined pre-​diagnostic consumption in Box 12–​1  OBJECTIVES OF THE WHO GLOBAL STRATEGY
relation to mortality; some studies found that light-​to-​moderate, but
not heavy, alcohol consumption was associated with lower risk of
mortality from all causes and colorectal cancer (Pelser et  al., 2014; 1. Raise global awareness of the magnitude and nature of the health,
Phipps et al., 2011, 2016; Walter et al., 2016; Zell et al., 2007). Others social, and economic problems caused by harmful use of alcohol,
studies showed no association (Asghari-​Jafarabadi et al., 2009; Park and increase commitment by governments to act to address the
et al., 2006). In a study of 175 stage I rectal cancer patients, alcohol harmful alcohol use.
drinking at the time of diagnosis was associated with a 2.4-​fold higher 2. Strengthen knowledge base on the magnitude and determinants of
risk (p = 0.01) of local recurrence compared to never/​former drinkers alcohol-​related harm and on effective interventions to reduce and
(Patel et al., 2014). However, in the Nurses’ Health Study there was no prevent such harm.
association between post-​diagnostic alcohol consumption and colorec- 3. Increase technical support to, and enhance capacity of, member
tal cancer mortality (Fung et al., 2014). In some studies, stage I–​IV states for preventing the harmful use of alcohol and managing
colon and rectal cancer patients were included, potentially obscuring alcohol-​use disorders and associated health conditions.
an effect on early-​stage cancers. 4. Strengthen partnerships and better coordination among
There is a high prevalence of tobacco and heavy alcohol use among stakeholders and increase mobilization of resources required for
head and neck cancer patients (Hashibe et  al., 2009). Research has appropriate and concerted action to prevent the harmful use of
shown that continued alcohol consumption after diagnosis is associ- alcohol.
ated with higher risk of both second primary cancers and mortality 5. Improve systems for monitoring and surveillance at different levels,
from head and neck cancers. In a study of nearly 1200 patients with and more effective dissemination and application of information
early-​stage head and neck squamous cell carcinoma enrolled in a for advocacy, policy development, and evaluation.
2

222 PART III:  THE CAUSES OF CANCER


numerous educational resources for healthcare professionals for use in Arita A, McFarland DC, Myklebust JH, et al. 2013. Signaling pathways in lym-
diagnosing and treating excessive alcohol intake (National Institute on phoma: pathogenesis and therapeutic targets. Future Oncol, 9(10), 1549–​
Alcohol Abuse and Alcoholism, 2016). 1571. PMID: 24106904.
Despite the known adverse effects of alcohol consumption in the Asai H, Imaoka S, Kuroki T, Monna T, and Funae Y. 1996. Microsomal eth-
anol oxidizing system activity by human hepatic cytochrome P450s. J
etiology of cancer, several organizations and consensus panels (AHA, Pharmacol Exp Ther, 277(2), 1004–​1009. PMID: 8627510.
2015; AICR, 2011; Kushi et  al., 2012)  have concluded that low to Asghari-​Jafarabadi M, Hajizadeh E, Kazemnejad A, and Fatemi SR. 2009. Site-​
moderate alcohol consumption can be part of a healthful dietary pat- specific evaluation of prognostic factors on survival in Iranian colorectal
tern because of the apparent benefits of moderate alcohol consumption cancer patients: a competing risks survival analysis. Asian Pac J Cancer
on coronary heart disease (Rimm et al., 1999). However, this conclu- Prev, 10(5), 815–​821. PMID: 20104971.
sion was recently challenged by the results of a recent Mendelian Boccia S, Hashibe M, Galli P, et al. 2009. Aldehyde dehydrogenase 2 and head
randomization study of a non-​synonymous single nucleotide polymor- and neck cancer:  a meta-​analysis implementing a Mendelian random-
phism in ADH1B in relation to cardiovascular disease risk factors, and ization approach. Cancer Epidemiol Biomarkers Prev, 18(1), 248–​254.
risk of coronary heart disease and stroke (Holmes et al., 2014). In that PMID: 19124505.
Boniface S, Kneale J, and Shelton N. 2014. Drinking pattern is more strongly
study, carriers of the genetic variant associated with non-​drinking and associated with under-​ reporting of alcohol consumption than socio-​
lower self-​reported alcohol consumption also had lower systolic blood demographic factors: evidence from a mixed-​methods study. BMC Public
pressure, circulating interleukin-​6 concentration, waist circumference, Health, 14, 1297. PMCID: PMC4320509.
body mass index, risk of coronary heart disease (OR  =  0.90; 95% Boyd NF, Lockwood GA, Byng JW, Tritchler DL, and Yaffe MJ. 1998.
CI: 0.84, 0.96), and risk of ischemic stroke (OR = 0.83; 95% CI: 0.72, Mammographic densities and breast cancer risk. Cancer Epidemiol
0.95). The authors concluded that reductions of alcohol consumption, Biomarkers Prev, 7(12), 1133–​1144. PMID: 9865433.
even for light to moderate drinkers, may be beneficial for cardiovas- Brooks PJ, Enoch MA, Goldman D, Li TK, and Yokoyama A. 2009. The
cular health. alcohol flushing response:  an unrecognized risk factor for esopha-
geal cancer from alcohol consumption. PLoS Med, 6(3), e50.
PMCID: PMC2659709.
Brooks PJ, and Zakhari S. 2013. Moderate alcohol consumption and breast
FUTURE RESEARCH cancer in women: from epidemiology to mechanisms and interventions.
Alcohol Clin Exp Res, 37(1), 23–​30. PMCID: PMC4551426.
Extensive evidence demonstrates the causal relationships of ethanol Cederbaum AI. 2012. Alcohol metabolism. Clin Liver Dis, 16(4), 667–​685.
in alcoholic beverages and its primary metabolite, acetaldehyde, to at PMCID: PMC3484320.
least seven different cancer sites in humans. More research is needed, Centers for Disease Control and Prevention. 2016. Frequently asked ques-
however, to better understand the associations with other cancer sites tions. Available from: http://​www.cdc.gov/​alcohol/​faqs.htm#standDrink.
for which the evidence in considered insufficient, for example gastric Accessed June 30, 2016.
and lung cancer. For gastric cancer this might require studying the Chang ET, Clarke CA, Canchola AJ, et  al. 2010. Alcohol consumption over
separate and joint effects of Helicobacter pylori infection and alcohol time and risk of lymphoid malignancies in the California Teachers Study
cohort. Am J Epidemiol, 172(12), 1373–​1383. PMID: 20952595.
consumption. For lung cancer, large studies of lifelong non-​smokers Chen WY, Rosner B, Hankinson SE, Colditz GA, and Willett WC.
will be necessary. 2011. Moderate alcohol consumption during adult life, drink-
In addition, a better understanding of the mechanistic basis of alco- ing patterns, and breast cancer risk. JAMA, 306(17), 1884–​ 1890.
hol-​related cancers in different target tissues is needed to help clarify PMCID: PMC3292347.
the causal relationship, particularly for cancers where the current evi- Chiu BC, Cerhan JR, Gapstur SM, et al. 1999. Alcohol consumption and non-​
dence is considered inconclusive by the IARC. Technologic advance- Hodgkin lymphoma in a cohort of older women. Br J Cancer, 80(9),
ments in whole-​ genome or exome sequencing, which can detect 1476–​1482. PMCID: 2363074.
signatures of carcinogens and carcinogenic mechanisms (Alexandrov Cho E, Smith-​Warner SA, Ritz J, et  al. 2004. Alcohol intake and colorectal
and Stratton, 2014), will support these research efforts. Similarly, cancer:  a pooled analysis of 8 cohort studies. Ann Intern Med, 140(8),
603–​613. PMID: 15096331.
exploring etiologic heterogeneity based on the molecular phenotype Crabb DW, Matsumoto M, Chang D, and You M. 2004. Overview of the role
of tumors—​beyond traditional markers such as ER status—​will help to of alcohol dehydrogenase and aldehyde dehydrogenase and their variants
clarify the causal role of alcoholic beverage consumption in the etiol- in the genesis of alcohol-​related pathology. Proc Nutr Soc, 63(1), 49–​63.
ogy of many cancer sites (Ogino et al., 2013). PMID: 15099407.
Further research also is needed to examine whether quitting drink- Do KA, Johnson MM, Doherty DA, et  al. 2003. Second primary tumors in
ing can reverse the increased risk associated with alcohol consumption patients with upper aerodigestive tract cancers: joint effects of smoking
for alcohol-​related cancers. There is some evidence that quitting drink- and alcohol (United States). Cancer Causes Control, 14(2), 131–​138.
ing appears to reduce the elevated risk of oral cavity and pharynx can- PMID: 12749718.
cer (IARC, 2010), but less is known for other cancer sites. In addition, Doll R, and Peto R. 1981. The causes of cancer: quantitative estimates of avoid-
able risks of cancer in the United States today. J Natl Cancer Inst, 66(6),
testing interventions and messages that discourage drinking, especially 1191–​1308. PMID: 7017215.
binge drinking among young women, could be useful for developing Dorgan JF, Baer DJ, Albert PS, et al. 2001. Serum hormones and the alcohol-​
individual and population strategies to reduce the harmful use of alcohol. breast cancer association in postmenopausal women. J Natl Cancer Inst,
93(9), 710–​715. PMID: 11333294.
References Dufour MC, and Adamson MD. 2003. The epidemiology of alcohol-​induced
AHA. 2015. Alcohol and Heart Health. Available from:  http://​www.heart. pancreatitis. Pancreas, 27(4), 286–​290. PMID: 14576488.
org/​HEARTORG/​HealthyLiving/​HealthyEating/​Nutrition/​Alcohol-​and-​ Dwyer-​Lindgren L, Flaxman AD, Ng M, et al. 2015. Drinking patterns in US
Heart-​Health_​UCM_​305173_​Article.jsp. Accessed July 25, 2016. counties from 2002 to 2012. Am J Public Health, 105(6), 1120–​1127.
AICR. 2011. Recommendations for Cancer Prevention. Available from: http://​ PMID: 25905846.
www.aicr.org/​reduce-​your-​cancer-​risk/​recommendations-​for-​cancer-​ El-​Serag HB, and Rudolph KL. 2007. Hepatocellular carcinoma:  epidemiol-
prevention/​recommendations_​06_​alcohol.html. Accessed July 25, 2016. ogy and molecular carcinogenesis. Gastroenterology, 132(7), 2557–​2576.
Alexandrov LB, and Stratton MR. 2014. Mutational signatures: the patterns of PMID: 17570226.
somatic mutations hidden in cancer genomes. Curr Opin Genet Dev, 24, Elder RW, Lawrence B, Ferguson A, et al. 2010. The effectiveness of tax policy
52–​60. PMCID: PMC3990474. interventions for reducing excessive alcohol consumption and related
Allen NE, Beral V, Casabonne D, et  al. 2009. Moderate alcohol intake and harms. Am J Prev Med, 38(2), 217–​229. PMCID: PMC3735171.
cancer incidence in women. J Natl Cancer Inst, 101(5), 296–​ 305. Collaborative Group on Hormonal Factors in Breast Cancer. 2002. Alcohol,
PMID: 19244173. tobacco and breast cancer—​collaborative reanalysis of individual data
Ammon E, Schafer C, Hofmann U, and Klotz U. 1996. Disposition and from 53 epidemiological studies, including 58 515 women with breast can-
first-​
pass metabolism of ethanol in humans:  is it gastric or hepatic cer and 95 067 women without the disease. Br J Cancer, 87, 1234–​1245.
and does it depend on gender? Clin Pharmacol Ther, 59(5), 503–​513. Collaborative Group on Hormonal Factors in Breast Cancer. 2011. Circulating
PMID: 8646821. sex hormones and breast cancer risk factors in postmenopausal
 23

Alcohol and Cancer Risk 223


women:  reanalysis of 13 studies. Br J Cancer, 105(5), 709–​ 722. IARC. 2012. Consumption of Alcoholic Beverages. IARC Working Group
PMCID: PMC3188939. (Ed.). IARC Monographs on the Evaluation of Carcinogenic Risks to
Collaborative Group on Hormonal Factors in Breast Cancer. 2013. Sex hor- Humans (Vol. 100e, pp. 373–​499). Lyon: IARC.
mones and risk of breast cancer in premenopausal women: a collaborative Jemal A, Vineis P, Bray F, Torre L, and Forman D. 2014. The Cancer Atlas. 2nd
reanalysis of individual participant data from seven prospective studies. ed. Atlanta: American Cancer Society.
Lancet Oncol, 14(10), 1009–​1019. PMCID: PMC4056766. Joh HK, Willett WC, and Cho E. 2011. Type 2 diabetes and the risk of
Eriksen M, Mackay J, Schluger N, Gomeshtapeh FI, and Drop J. 2015. The renal cell cancer in women. Diabetes Care, 34(7), 1552–​ 1556.
Tobacco Atlas. 5th ed. Atlanta: American Cancer Society. PMCID: PMC3120193.
Ferrari P, Jenab M, Norat T, et al. 2007. Lifetime and baseline alcohol intake Jokelainen K, Matysiak-​Budnik T, Makisalo H, Hockerstedt K, and Salaspuro
and risk of colon and rectal cancers in the European prospective investiga- M. 1996. High intracolonic acetaldehyde values produced by a bacterio-
tion into cancer and nutrition (EPIC). Int J Cancer, 121(9), 2065–​2072. colonic pathway for ethanol oxidation in piglets. Gut, 39(1), 100–​104.
PMID: 17640039. PMCID: PMC1383240.
Frank A, Seitz HK, Bartsch H, Frank N, and Nair J. 2004. Immunohistochemical Kaaks R, and Riboli E. 1997. Validation and calibration of dietary intake mea-
detection of 1,N6-​ethenodeoxyadenosine in nuclei of human liver affected surements in the EPIC project: methodological considerations. European
by diseases predisposing to hepato-​ carcinogenesis. Carcinogenesis, Prospective Investigation into Cancer and Nutrition. Int J Epidemiol,
25(6), 1027–​1031. PMID: 14742317. 26(Suppl 1), S15–​S25. PMID: 9126530.
Friberg E, Orsini N, Mantzoros CS, and Wolk A. 2010. Alcohol intake and Kanda J, Matsuo K, Inoue M, et al. 2010. Association of alcohol intake with
endometrial cancer risk:  a meta-​analysis of prospective studies. Br J the risk of malignant lymphoma and plasma cell myeloma in Japanese: a
Cancer, 103(1), 127–​131. PMCID: PMC2905297. population-​ based cohort study (Japan Public Health Center-​ based
Fung TT, Kashambwa R, Sato K, et  al. 2014. Post diagnosis diet quality Prospective Study). Cancer Epidemiol Biomarkers Prev, 19(2), 429–​434.
and colorectal cancer survival in women. PLoS One, 9(12), e115377. PMID: 20086115.
PMCID: PMC4266679. Key T, Appleby P, Barnes I, et al. 2002. Endogenous sex hormones and breast
Gapstur SM, Diver WR, McCullough ML, et  al. 2012. Alcohol intake and cancer in postmenopausal women: reanalysis of nine prospective studies.
the incidence of non-​hodgkin lymphoid neoplasms in the cancer pre- J Natl Cancer Inst, 94(8), 606–​616. PMID: 11959894.
vention study II nutrition cohort. Am J Epidemiol, 176(1), 60–​ 69. Klatsky AL, Li Y, Baer D, et al. 2009. Alcohol consumption and risk of hemato-
PMID: 22562662. logic malignancies. Ann Epidemiol, 19(10), 746–​753. PMID: 19394864.
Gapstur SM, Jacobs EJ, Deka A, et al. 2011. Association of alcohol intake with Koppaka V, Thompson DC, Chen Y, et  al. 2012. Aldehyde dehydrogenase
pancreatic cancer mortality in never smokers. Arch Intern Med, 171(5), inhibitors:  a comprehensive review of the pharmacology, mechanism
444–​451. PMID: 21403041. of action, substrate specificity, and clinical application. Pharmacol Rev,
Gaudet MM, Carter BD, Brinton LA, et  al. 2016. Pooled analysis of active 64(3), 520–​539. PMCID: PMC3400832.
cigarette smoking and invasive breast cancer risk in 14 cohort studies. Int Koppes LL, Dekker JM, Hendriks HF, Bouter LM, and Heine RJ. 2005.
J Epidemiol, 2016; doi: 10.1093/​ije/​dyw288. Moderate alcohol consumption lowers the risk of type 2 diabetes: a meta-​
Genkinger JM, Hunter DJ, Spiegelman D, et al. 2006. Alcohol intake and ovar- analysis of prospective observational studies. Diabetes Care, 28(3), 719–​
ian cancer risk: a pooled analysis of 10 cohort studies. Br J Cancer, 94(5), 725. PMID: 15735217.
757–​762. PMCID: PMC2361197. Kuper H, Tzonou A, Kaklamani E, et al. 2000. Tobacco smoking, alcohol con-
Giovannucci E, Colditz G, Stampfer MJ, et al. 1991. The assessment of alco- sumption and their interaction in the causation of hepatocellular carci-
hol consumption by a simple self-​ administered questionnaire. Am J noma. Int J Cancer, 85(4), 498–​502. PMID: 10699921.
Epidemiol, 133(8), 810–​817. PMID: 2021148. Kushi LH, Doyle C, McCullough M, et  al. 2012. American Cancer Society
Go VL, Gukovskaya A, and Pandol SJ. 2005. Alcohol and pancreatic cancer. Guidelines on nutrition and physical activity for cancer prevention: reduc-
Alcohol, 35(3), 205–​211. PMID: 16054982. ing the risk of cancer with healthy food choices and physical activity. CA
Goedde HW, and Agarwal DP. 1987. Polymorphism of aldehyde dehydroge- Cancer J Clin, 62(1), 30–​67. PMID: 22237782.
nase and alcohol sensitivity. Enzyme, 37(1–​2), 29–​44. PMID: 3106030. Kwan ML, Chen WY, Flatt SW, et  al. 2013. Postdiagnosis alcohol con-
Hagner PR, Mazan-​Mamczarz K, Dai B, et al. 2009. Alcohol consumption and sumption and breast cancer prognosis in the after breast cancer pool-
decreased risk of non-​Hodgkin lymphoma:  role of mTOR dysfunction. ing project. Cancer Epidemiol Biomarkers Prev, 22(1), 32–​ 41.
Blood, 113(22), 5526–​5535. PMID: 19293424. PMCID: PMC3538884.
Hamajima N, Hirose K, Tajima K, et al. 2002. Alcohol, tobacco and breast cancer-​ Lachenmeier DW, Przybylski MC, and Rehm J. 2012. Comparative risk assess-
-​collaborative reanalysis of individual data from 53 epidemiological stud- ment of carcinogens in alcoholic beverages using the margin of exposure
ies, including 58,515 women with breast cancer and 95,067 women without approach. Int J Cancer, 131(6), E995–E1003. PMID: 22447328.
the disease. Br J Cancer, 87(11), 1234–​1245. PMCID: PMC2562507. Lewis SJ, and Smith GD. 2005. Alcohol, ALDH2, and esophageal cancer:  a
Hashibe M, Brennan P, Benhamou S, et  al. 2007. Alcohol drinking in never meta-​ analysis which illustrates the potentials and limitations of a
users of tobacco, cigarette smoking in never drinkers, and the risk of head Mendelian randomization approach. Cancer Epidemiol Biomarkers Prev,
and neck cancer:  pooled analysis in the International Head and Neck 14(8), 1967–​1971. PMID: 16103445.
Cancer Epidemiology Consortium. J Natl Cancer Inst, 99(10), 777–​789. Li CI, Malone KE, Daling JR, et al. 2008. Timing of menarche and first full-​
PMID: 17505073. term birth in relation to breast cancer risk. Am J Epidemiol, 167(2), 230–​
Hashibe M, Brennan P, Chuang SC, et  al. 2009. Interaction between 239. PMCID: PMC3804121.
tobacco and alcohol use and the risk of head and neck cancer: pooled Lim U, Morton LM, Subar AF, et al. 2007. Alcohol, smoking, and body size in
analysis in the International Head and Neck Cancer Epidemiology relation to incident Hodgkin’s and non-​Hodgkin’s lymphoma risk. Am J
Consortium. Cancer Epidemiol Biomarkers Prev, 18(2), 541–​ 550. Epidemiol, 166(6), 697–​708. PMID: 17596266.
PMCID: PMC3051410. Lim U, Weinstein S, Albanes D, et  al. 2006. Dietary factors of one-​carbon
Hassan MM, Hwang LY, Hatten CJ, et al. 2002. Risk factors for hepatocellular metabolism in relation to non-​Hodgkin lymphoma and multiple myeloma
carcinoma:  synergism of alcohol with viral hepatitis and diabetes mel- in a cohort of male smokers. Cancer Epidemiol Biomarkers Prev, 15(6),
litus. Hepatology, 36(5), 1206–​1213. PMID: 12395331. 1109–​1114. PMID: 16775167.
Henley SJ, Kanny D, Roland KB, et al. 2014. Alcohol control efforts in com- Linhart K, Bartsch H, and Seitz HK. 2014. The role of reactive oxygen spe-
prehensive cancer control plans and alcohol use among adults in the USA. cies (ROS) and cytochrome P-​450 2E1 in the generation of carcinogenic
Alcohol Alcohol, 49(6), 661–​667. PMCID: PMC4380007. etheno-​DNA adducts. Redox Biol, 3, 56–​62. PMCID: PMC4297928.
Holmes MV, Dale CE, Zuccolo L, et al. 2014. Association between alcohol and Liu Y, Colditz GA, Rosner B, et al. 2013. Alcohol intake between menarche and
cardiovascular disease: Mendelian randomisation analysis based on indi- first pregnancy: a prospective study of breast cancer risk. J Natl Cancer
vidual participant data. BMJ, 349, g4164. PMCID: PMC4091648. Inst, 105(20), 1571–​1578. PMCID: PMC3797023.
Homann N, Tillonen J, Meurman JH, et  al. 2000. Increased salivary acet- Liu Y, Nguyen N, and Colditz GA. 2015. Links between alcohol consumption
aldehyde levels in heavy drinkers and smokers:  a microbiologi- and breast cancer: a look at the evidence. Womens Health (Lond Engl),
cal approach to oral cavity cancer. Carcinogenesis, 21(4), 663–​668. 11(1), 65–​77. PMCID: PMC4299758.
PMID: 10753201. Loomba R, Yang HI, Su J, et al. 2013. Synergism between obesity and alcohol
IARC. 1988. Alcohol Drinking. IARC Monographs on the Evaluation of in increasing the risk of hepatocellular carcinoma: a prospective cohort
Carcinogenic Risks to Humans (Vol. 44, pp. 15–​378). Lyon: IARC. study. Am J Epidemiol, 177(4), 333–​342. PMCID: 3626057.
IARC. 2010. Alcohol Consumption and Ethyl Carbamate. IARC Monographs Loomba R, Yang HI, Su J, et al. 2010. Obesity and alcohol synergize to increase
on the Evaluation of Carcinogenic Risks to Humans (Vol. 96, pp. 7–​ the risk of incident hepatocellular carcinoma in men. Clin Gastroenterol
1280). Lyon: IARC. Hepatol, 8(10), 891–​898. PMID: 20621202.
24

224 PART III:  THE CAUSES OF CANCER


Marrero JA, Fontana RJ, Fu S, et al. 2005. Alcohol, tobacco and obesity are Seitz HK, and Stickel F. 2010. Acetaldehyde as an underestimated risk factor
synergistic risk factors for hepatocellular carcinoma. J Hepatol, 42(2), for cancer development:  role of genetics in ethanol metabolism. Genes
218–​224. PMID: 15664247. Nutr, 5(2), 121–​128. PMCID: PMC2885165.
Mayne ST, Cartmel B, Kirsh V, and Goodwin WJ, Jr. 2009. Alcohol and tobacco Shield KD, Gmel G, Kehoe-​Chan T, et al. 2013. Mortality and potential years
use prediagnosis and postdiagnosis, and survival in a cohort of patients of life lost attributable to alcohol consumption by race and sex in the
with early stage cancers of the oral cavity, pharynx, and larynx. Cancer United States in 2005. PLoS One, 8(1), e51923. PMCID: PMC3534703.
Epidemiol Biomarkers Prev, 18(12), 3368–​3374. PMCID: PMC2789339. Son J, Pang B, McFaline JL, Taghizadeh K, and Dedon PC. 2008. Surveying
Mazan-​Mamczarz K, Peroutka RJ, Steinhardt JJ, et al. 2015. Distinct inhibitory the damage:  the challenges of developing nucleic acid biomarkers of
effects on mTOR signaling by ethanol and INK128 in diffuse large B-​cell inflammation. Mol Biosyst, 4(9), 902–​908. PMID: 18704228.
lymphoma. Cell Commun Signal, 13, 15. PMCID: PMC4350884. Squier CA, Cox P, and Hall BK. 1986. Enhanced penetration of nitrosonornico-
McGovern PE, Zhang J, Tang J, et al. 2004. Fermented beverages of pre-​and tine across oral mucosa in the presence of ethanol. J Oral Pathol, 15(5),
proto-​historic China. Proc Natl Acad Sci U S A, 101(51), 17593–​17598. 276–​279. PMID: 3091795.
PMCID: PMC539767. Stockwell T, Donath S, Cooper-​Stanbury M, et  al. 2004. Under-​reporting of
Morch LS, Johansen D, Thygesen LC, et al. 2007. Alcohol drinking, consump- alcohol consumption in household surveys:  a comparison of quantity-​
tion patterns and breast cancer among Danish nurses: a cohort study. Eur frequency, graduated-​ frequency and recent recall. Addiction, 99(8),
J Public Health, 17(6), 624–​629. PMID: 17442702. 1024–​1033. PMID: 15265099.
Moskal A, Norat T, Ferrari P, and Riboli E. 2007. Alcohol intake and colorectal Sutton CW, Rustogi N, Gurkan C, et al. 2010. Quantitative proteomic profiling
cancer risk:  a dose-​response meta-​analysis of published cohort studies. of matched normal and tumor breast tissues. J Proteome Res, 9(8), 3891–​
Int J Cancer, 120(3), 664–​671. PMID: 17096321. 3902. PMID: 20560667.
National Institute on Alcohol Abuse and Alcoholism. 2016. Professional Suzuki R, Orsini N, Mignone L, Saji S, and Wolk A. 2008. Alcohol intake
Education Materials. Available at https://​www.niaaa.nih.gov/​publica- and risk of breast cancer defined by estrogen and progesterone receptor
tions/​clinical-​guides-​and-​manuals. Accessed July 21, 2016. status: a meta-​analysis of epidemiological studies. Int J Cancer, 122(8),
Nelson DE, Jarman DW, Rehm J, et  al. 2013. Alcohol-​attributable cancer 1832–​1841. PMID: 18067133.
deaths and years of potential life lost in the United States. Am J Public Theruvathu JA, Jaruga P, Nath RG, Dizdaroglu M, and Brooks PJ.
Health, 103(4), 641–​648. PMCID: PMC3673233. 2005. Polyamines stimulate the formation of mutagenic 1,N2-​
Ogino S, Lochhead P, Chan AT, et al. 2013. Molecular pathological epidemiol- propanodeoxyguanosine adducts from acetaldehyde. Nucleic Acids Res,
ogy of epigenetics: emerging integrative science to analyze environment, 33(11), 3513–​3520. PMCID: 1156964.
host, and disease. Mod Pathol, 26(4), 465–​484. PMCID: PMC3637979. Triano EA, Slusher LB, Atkins TA, et  al. 2003. Class  I  alcohol dehydroge-
Pandya GA, and Moriya M. 1996. 1,N6-​ethenodeoxyadenosine, a DNA adduct nase is highly expressed in normal human mammary epithelium but not
highly mutagenic in mammalian cells. Biochemistry, 35(35), 11487–​ in invasive breast cancer: implications for breast carcinogenesis. Cancer
11492. PMID: 8784204. Res, 63(12), 3092–​3100. PMID: 12810634.
Park SM, Lim MK, Shin SA, and Yun YH. 2006. Impact of prediagnosis smok- Troy JD, Hartge P, Weissfeld JL, et al. 2010. Associations between anthropom-
ing, alcohol, obesity, and insulin resistance on survival in male cancer etry, cigarette smoking, alcohol consumption, and non-​Hodgkin lym-
patients:  National Health Insurance Corporation Study. J Clin Oncol, phoma in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening
24(31), 5017–​5024. PMID: 17075121. Trial. Am J Epidemiol, 171(12), 1270–​1281. PMID: 20494998.
Patel SA, Chen YH, Hornick JL, et al. 2014. Early-​stage rectal cancer: clini- Tsong WH, Koh WP, Yuan JM, et al. 2007. Cigarettes and alcohol in relation
cal and pathologic prognostic markers of time to local recurrence and to colorectal cancer: the Singapore Chinese Health Study. Br J Cancer,
overall survival after resection. Dis Colon Rectum, 57(4), 449–​ 459. 96(5), 821–​827. PMCID: PMC2360085.
PMCID: PMC3954982. Turati F, Galeone C, Rota M, et al. 2014. Alcohol and liver cancer: a systematic
Pelser C, Arem H, Pfeiffer RM, et al. 2014. Prediagnostic lifestyle factors and review and meta-​analysis of prospective studies. Ann Oncol, 25(8), 1526–​
survival after colon and rectal cancer diagnosis in the National Institutes 1535. PMID: 24631946.
of Health (NIH)-​AARP Diet and Health Study. Cancer, 120(10), 1540–​ Visapaa JP, Jokelainen K, Nosova T, and Salaspuro M. 1998. Inhibition of
1547. PMCID: PMC4151292. intracolonic acetaldehyde production and alcoholic fermentation
Phipps AI, Baron J, and Newcomb PA. 2011. Prediagnostic smoking his- in rats by ciprofloxacin. Alcohol Clin Exp Res, 22(5), 1161–​ 1164.
tory, alcohol consumption, and colorectal cancer survival:  the PMID: 9726290.
Seattle Colon Cancer Family Registry. Cancer, 117(21), 4948–​4957. Wagenaar AC, Salois MJ, and Komro KA. 2009. Effects of beverage alcohol
PMCID: PMC3138819. price and tax levels on drinking: a meta-​analysis of 1003 estimates from
Phipps AI, Shi Q, Limburg PJ, et  al. 2016. Alcohol consumption and colon 112 studies. Addiction, 104(2), 179–​190. PMID: 19149811.
cancer prognosis among participants in north central cancer treat- Wagenaar AC, Tobler AL, and Komro KA. 2010. Effects of alcohol tax and
ment group phase III trial N0147. Int J Cancer, 139(5), 986–​ 995. price policies on morbidity and mortality:  a systematic review. Am J
PMCID: PMC4911257. Public Health, 100(11), 2270–​2278. PMCID: PMC2951962.
Rehm J, and Shield KD. 2014. Alcohol consumption. Steward B, and Wild C Walter V, Jansen L, Ulrich A, et al. 2016. Alcohol consumption and survival of
(Eds.). World Cancer Report 2014. Lyon: IARC. colorectal cancer patients: a population-​based study from Germany. Am J
Reichman ME, Judd JT, Longcope C, et al. 1993. Effects of alcohol consump- Clin Nutr, 103(6), 1497–​1506. PMID: 27146651.
tion on plasma and urinary hormone concentrations in premenopausal Wang HJ, Gao B, Zakhari S, and Nagy LE. 2012. Inflammation in alcoholic
women. J Natl Cancer Inst, 85(9), 722–​727. PMID: 8478958. liver disease. Annu Rev Nutr, 32, 343–​368. PMCID: PMC3670145.
Rimm EB, Williams P, Fosher K, Criqui M, and Stampfer MJ. 1999. Moderate Wang Y, Millonig G, Nair J, et al. 2009. Ethanol-​induced cytochrome P4502E1
alcohol intake and lower risk of coronary heart disease: meta-​analysis of causes carcinogenic etheno-​ DNA lesions in alcoholic liver disease.
effects on lipids and haemostatic factors. BMJ, 319(7224), 1523–​1528. Hepatology, 50(2), 453–​461. PMID: 19489076.
PMCID: PMC28294. World Cancer Research Fund International. 2011. Food, Nutrition, Physical
Rock CL, Doyle C, Demark-​Wahnefried W, et al. 2012. Nutrition and physical Activity, and the Prevention of Colorectal Cancer. London: WCRF.
activity guidelines for cancer survivors. CA Cancer J Clin, 62(4), 243–​ World Cancer Research Fund International. 2012. Food, Nutrition, Physical
274. PMID: 22539238. Activity, and the Prevention of Pancreatic Cancer. London: WCRF.
Romeo J, Warnberg J, Nova E, et  al. 2007. Moderate alcohol consumption World Cancer Research Fund International. 2013. Food, Nutrition, Physical
and the immune system: a review. Br J Nutr, 98(Suppl 1), S111–​S115. Activity, and the Prevention of Endometrial Cancer. London: WCRF.
PMID: 17922947. World Cancer Research Fund International. 2014a. Food, Nutrition, Physical
Rosner B. 1995. Fundamental of Biostatistics. 4th ed. Belmont, Activity, and the Prevention of Ovarian Cancer. London: WCRF.
CA: Duxbury Press. World Cancer Research Fund International. 2014b. Systematic Review on
Rothman KJ. 1980. The proportion of cancer attributable to alcohol consump- Diet, Nutrition, Physical Activity and Survival and Second Cancers in
tion. Prev Med, 9(2), 174–​179. PMID: 7383982. Breast Cancer Survivors. Continuous Update Project. London: Amperial
Salaspuro MP, and Lieber CS. 1978. Non-​uniformity of blood ethanol elimina- College.
tion:  its exaggeration after chronic consumption. Ann Clin Res, 10(5), World Cancer Research Fund International. 2015. Food, Nutrition, Physical
294–​297. PMID: 104651. Activity, and the Prevention of Kidney Cancer. London: WCRF.
Seitz HK, Maurer B, and Stickel F. 2005. Alcohol consumption and cancer of World Health Organization. 2010. Global Strategy to Reduce the Harmful Use
the gastrointestinal tract. Dig Dis, 23(3-​4), 297–​303. PMID: 16508294. of Alcohol. Geneva: WHO.
Seitz HK, and Stickel F. 2007. Molecular mechanisms of alcohol-​mediated car- World Health Organization. 2014. Global Status Report on Alcohol and Health
cinogenesis. Nat Rev Cancer, 7(8), 599–​612. PMID: 17646865. 2014. World Health Organization (Ed.). Geneva: WHO.
 25

Alcohol and Cancer Risk 225


Woutersen RA, Appelman LM, Van Garderen-​Hoetmer A, and Feron VJ. 1986. Yuan JM, Govindarajan S, Arakawa K, and Yu MC. 2004. Synergism of alcohol,
Inhalation toxicity of acetaldehyde in rats. III. Carcinogenicity study. diabetes, and viral hepatitis on the risk of hepatocellular carcinoma in blacks
Toxicology, 41(2), 213–​231. PMID: 3764943. and whites in the U.S. Cancer, 101(5), 1009–​1017. PMID: 15329910.
Yokoyama A, Muramatsu T, Ohmori T, et al. 1996. Esophageal cancer and alde- Zeka A, Gore R, and Kriebel D. 2003. Effects of alcohol and tobacco on aerodi-
hyde dehydrogenase-2 genotypes in Japanese males. Cancer Epidemiol gestive cancer risks: a meta-​regression analysis. Cancer Causes Control,
Biomarkers Prev, 5(2), 99–102. PMID: 880269. 14(9), 897–​906. PMID: 14682447.
Yokoyama A, Omori T, Yokoyama T et al. 2006. Risk of squamous cell carci- Zell JA, McEligot AJ, Ziogas A, Holcombe RF, and Anton-​Culver H. 2007.
noma of the upper aerodigestive tract in cancer-​free alcoholic Japanese Differential effects of wine consumption on colorectal cancer outcomes
men: an endoscopic follow-​up study. Cancer Epidemiol Biomarkers Prev, based on family history of the disease. Nutr Cancer, 59(1), 36–​ 45.
15(11), 2209–​2215. PMID: 17119048. PMID: 17927500.
26
 27

13 Ionizing Radiation

AMY BERRINGTON DE GONZÁLEZ, ANDRÉ BOUVILLE, PREETHA RAJARAMAN,


AND MARY SCHUBAUER-​BERIGAN

OVERVIEW has focused on the LNT question, primarily assessing whether there
are excess cancer risks in populations who received very low doses,
Ionizing radiation is classified as a universal carcinogen due to its abil- and quantifying the magnitude of the risk. Another outstanding issue
ity to induce cancer in most organs following exposure at any age, is whether the risk from protracted exposure to ionizing radiation
including in utero. Several organs are especially radiosensitive, partic- is lower than the risk from the same dose received acutely, due to
ularly when exposure occurs in childhood, including the female breast, a greater probability of DNA repair. This is particularly relevant
thyroid, brain, and red bone marrow. Very few cancers, notably cervi- for radiation protection standards for occupational groups, as their
cal and Hodgkin lymphoma, do not seem to be related to ionizing radi- radiation exposure is often received over many decades. As can-
ation, but the reasons are unknown. For most cancers (lung may be an cer survival continues to improve, there has been an expansion of
exception) the relative risk decreases with age at exposure or attained effort to improve and understand the late effects of cancer treatment.
age, and time since exposure. Radiotherapy results in exposure to very high (> 5Gy) fractionated
Currently the main sources of radiation exposure to the general pop- doses to the healthy organs surrounding the target organ, and this is
ulation are from natural background radiation (e.g., residential radon) associated with an increased risk of developing a second cancer. To
and medical (e.g., computed tomography [CT] scans). Natural back- balance these potential risks against the benefits of radiotherapy, we
ground exposure varies by location but is generally stable over time. need to have good quantitative risk estimates of cancer risks from
Medical exposure has been increasing in many countries due to the high doses.
expansion of advanced imaging technologies. Risk projection models Because of the wealth of epidemiological data on the cancer risks
suggest that currently 1%–​3% of cancers could be related to medical from moderate doses, the established carcinogenicity, and the ability to
radiation exposure in countries like the United Kingdom and the United measure biological dose, epidemiological studies of ionizing radiation
States, and about 1% of cancers could be related to natural background provide a rich setting for studying interactions with other carcinogens,
radiation. Occupational radiation exposures have decreased over the including gene–​ environment interactions, or genetic susceptibility.
last several decades. One exception is the medical workers who are These investigations have also been a focus of recent research efforts.
exposed to radiation from performing increasing numbers of interven- The advent of tumor sequencing has provided an exciting opportunity
tional fluoroscopy and nuclear medicine procedures. Historically there to search for potential “radiation signatures” in populations with very
have been important exposures to large populations due to nuclear well characterized radiation exposures and high attributable risks, such
weapons detonation and testing and nuclear accidents, particularly as the thyroid cancers diagnosed after childhood exposure to iodine-​
Chernobyl. 131 (I-​131) from the Chernobyl accident.
Although ionizing radiation is an established carcinogen, the mag- As there are many types of ionizing radiation exposure, modes of
nitude of the risk from very low-​dose acute (< 50 mGy) and protracted exposure, methods for measuring it, and several different dose units,
exposures (< 5 mGy per hour) is still uncertain, and difficult to resolve we start with a review of these basic concepts before reviewing the
via epidemiological studies. In the last decade there have been new new epidemiological studies.
studies based on large-​scale electronic record linkages of pediatric
CT scans, natural background radiation, environmental exposures,
and nuclear workers that have provided additional evidence support- NATURE OF THE EXPOSURE
ing excess cancer risks from low-​dose and/​or low-​dose rate expo-
sures. Developments in molecular epidemiology, particularly tumor
sequencing to find a potential “radiation signature,” could also be a Types of Ionizing Radiation
novel approach to address this important public health question in the Ionizing radiation consists of waves or particles (gamma, X-​rays, neu-
next decade. trons, alpha particles, etc.), usually produced during the radioactive
decay of unstable nuclei, that have sufficient energy to ionize atoms
in the human body, thus inducing chemical changes that may be bio-
INTRODUCTION logically important for the functioning of cells. The various forms of
radiation are emitted with different energies and penetrating power
Most ionizing radiation exposures to the general population today through materials and potentially different carcinogenic potential. The
are from very low doses such as CT scans, or residential radon expo- most common types of ionizing radiation are the following (each of
sure. The so-​called linear no-​threshold (LNT) assumption forms the which can cause cancer):
basis for radiation protection standards for these very low doses—​the
assumption being that there is no safe level of radiation exposure • Alpha particles, consisting of helium nuclei, which can be halted by
(i.e., no threshold) and that the cancer risks from very low doses a sheet of paper and thus can hardly penetrate the dead outer layers
can be estimated by linear extrapolation of observed risks from stud- of the skin;
ies of populations exposed to higher doses (primarily the atomic • Beta particles, consisting of electrons, which can penetrate up to
bomb survivors). This model has been challenged, however, by both 2 cm of soft tissue; and
experimental and some epidemiological data, and remains somewhat • Gamma radiation, consisting of photons, which traverse the entire
contentious. Much of the epidemiological research in the last decade human body.

227
28

228 PART III:  THE CAUSES OF CANCER

Modes and Patterns of Irradiation Radiological Protection (ICRP) are 20 for alpha particles and 1 for
electrons and photons (ICRP, 2007). These values are based primarily
There are many different modes of irradiation that can impact cancer on in vitro experiments, and hence contain a number of uncertainties,
risk and are therefore important to document and take into account especially in their transfer to humans. The unit of equivalent dose is
in epidemiological studies. The variety of well-​documented exposure the sievert (Sv).
patterns provides an important opportunity to evaluate critical expo- For regulatory purposes, the primary dosimetry quantity is the effec-
sure windows for carcinogenesis. tive dose, which is obtained as the sum, including all radiosensitive
organs and tissues of the body, of the equivalent doses weighted by a
Internal Versus External Exposure tissue-​weighting factor, reflecting the radiosensitivity of the tissue or
External exposures occur when individuals are in proximity to a organ. The current values of the tissue-​weighting factors that are rec-
photon-​emitting radiation source. Internal exposure occurs from ommended by the ICRP are 0.12 for red bone marrow, lung, stomach,
intake of radionuclides, usually by inhalation or ingestion, or some- and breasts; 0.08 for gonads; 0.04 for bladder, liver, esophagus, and
times via absorption through intact or damaged skin or from injection thyroid; 0.01 for skin, bone surface, salivary glands, and brain; 0.12 for
for medical reasons. the remainder of body (ICRP, 2007). Because the effective dose takes
into account the absorbed doses received by all radiosensitive organs
Acute, Fractionated, or Protracted Exposure and tissues of the body, weighted according to their radiosensitivity
External exposure stops as soon as the source is turned off or removed, and to the radiation quality, it can be considered to be representative of
or when the individuals move away from the radiation source. External the radiation impact caused by exposure to a given source of radiation.
exposures, therefore, can be acute (i.e., delivered within seconds or The effective dose is a regulatory quantity that is directly amenable to
less), fractionated (consisting of a series of acute or short-​term expo- the comparison of the radiation impacts of widely different sources of
sures delivered at relatively long time intervals), or protracted (i.e., exposure; its unit is also the sievert (Sv). Generally, it is not appropri-
occurring over a long time at a relatively constant rate). In contrast, ate to use estimates of effective dose in epidemiological studies.
internal exposures, which result from the incorporation of a radionu-
clide, last as long as the radionuclide has not completely decayed or
been excreted from the body, depending on the physical half-​life and
biokinetics of the incorporated radionuclide. Internal exposures, there- SOURCES OF EXPOSURE
fore, cannot be acute.
There are three broad sources of ionizing radiation exposure: environ-
Full-​Versus Partial-​Body Exposures mental, medical, and occupational. Currently the general population
External exposures are usually due to photons, which can irradiate the is primarily exposed to very low doses of radiation from environmen-
entire body in a relatively uniform manner, referred to as full-​body tal sources from natural background radiation, and medical exposures
exposure; for example the Japanese atomic bombs resulted in relatively from diagnostic tests like CT scans (Figure 13–​1). There are also an
uniform full-​body exposure. Medical exposures to external sources, estimated 23 million radiation workers in the world who receive occu-
such as diagnostic X-​rays, however, only irradiate the organs in the X-​ pational exposures (UNSCEAR, 2010b). Historically there have been
ray field and are referred to as partial-​body exposures. Internal expo- a number of important events that resulted in much higher doses of
sures can result in the irradiation of a specific organ of the body (for radiation exposure to the general population and workers, including
example, the thyroid following exposure to I-​131, with other organs the atomic bombs dropped in Japan, the Chernobyl accident, and uses
receiving some exposure, e.g., stomach) or in the uniform irradiation of high doses of radiation for the treatment of benign medical condi-
of the entire body (e.g., from exposure to cesium-​137). tions, such as ringworm (Tinea capitis). The epidemiological literature
includes populations exposed to current sources of exposure as well as
these historical sources, and therefore we review both here.
Units of Exposure
The principal radiation quantities used to express dose are presented in Environmental Exposures
Table 13–​1. The most important quantity of radiation dosimetry that is
used in epidemiologic studies is the absorbed dose, which is defined as
the radiation energy absorbed per unit mass of organ or tissue. The unit Natural Environmental Exposures
of absorbed dose is the gray (Gy), with 1 Gy equal to 1 joule per kg. Natural background radiation exposure makes up at least half of all
The absorbed dose is a physical quantity that does not take the type of radiation exposure to the general population in most countries (Figure
particle or the radiosensitivity of the organ or tissue into consideration. 13–​1). Exposure comes from radon, cosmic, terrestital gamma, and
Different types of radiation have different biological effectiveness internal sources (United Nations Scientific Committee on the Effects
because they transfer their energy in different ways that can cause of Atomic Radiation [UNSCEAR], 2010b) (Table 13–​ 2). Radon,
or less more damage. The equivalent dose takes this into account including thoron, is generally the largest contributor (about 50%, on
by weighting the absorbed dose in an organ or tissue by a radiation average, of the world population of natural background exposure) but
weighting factor, which reflects the relative biological effectiveness is also highly variable, depending on where people live and the con-
(RBE) of the radiation type. The radiation weighting factors that struction of their houses. There are several documented areas of much
are currently recommended by the International Commission on higher natural background radiation in Brazil, India, Iran, and China,
where annual doses can exceed 10 mSv per year (UNSCEAR, 2010b).
Cosmic rays that reach the Earth’s surface originate either from
outer space (galactic and extra-​galactic cosmic rays) or from the sun
Table 13–​1.  Principal Radiation Quantities Used to Express Dose (solar wind) and cause exposures predominantly via external irradia-
Quantity Unit Comment
tion. Cosmic radiation exposure to the general population varies with
latitude and altitude, and represents about 20% of the natural back-
Absorbed dose Gray (Gy) Does not account for the type of ground exposure. Enhanced exposures are due to long-​haul air travel,
radiation exposure which is at high altitude; a round-​trip flight from London to Sydney
Equivalent dose Sievert (Sv) Accounts for biological effectiveness of results in an estimated effective dose of 0.16 mSv.
different types of radiation Naturally occurring radionuclides in the earth result in effective
Effective dose Sievert (Sv) Summary dose often used for partial-​ doses of external gamma radiation that represent about 20% of the
body exposures that accounts for natural background exposure. Natural radionuclides in foods such as
different radiosensitivity of organs dairy, cereals, and vegetables contribute the remaining 10% of natural
and biological effectiveness background exposure. Some foods can contain relatively high levels
 29

Ionizing Radiation 229


7 The atomic bombs were detonated over Hiroshima and Nagasaki
at altitudes of more than 500 meters so that the radiation doses were
Other essentially due to acute whole-​body external irradiation, immediately
6 Medical after the explosion, from photons and neutrons; there was very little
Environmental radioactive fallout because the explosions occurred at relatively high
altitudes, so that the fireballs had little interaction with the ground
5
(Cullings et al., 2006). Radiation dose levels depended primarily on
location in the city at the time of detonation and shielding. Those close
4 to the hypocenter who survived the initial blast had whole-​body doses
of > 5 Gy, resulting in acute radiation sickness and death within several
weeks. Survivors far from the hypocenter (e.g., > 2 km) had very low
3 whole-​body doses (< 5 mGy).
In contrast, the nuclear weapons tests were conducted in unpop-
ulated areas at altitudes that were generally less than 100 meters.
2 Consequently, people were not exposed to radiation occurring dur-
ing or immediately after the explosion. However, radioactive fallout,
including a large number of radionuclides, were spread over the entire
1 world (Bennett, 2002). These radionuclides led to external irradia-
tion from the photons produced during their radioactive decay, and
0 internal irradiation when they contaminated food products. Most of
USA UK Germany World
the doses were delivered within a few months after the test, with resid-
ual radiation, due to long-​lived radionuclides like 90Sr and 137Cs, with
radioactive half-​lives of about 30 years, protracted over a century or
Figure 13–​1.  Comparison of current estimated annual per capita radiation so (Bouville et  al., 2002). Even longer-​lived radionuclides, like 14C
exposure (mSv) for countries with a similar health-​care level. and 239Pu, will remain in the environment for more than 10,000 years.
Current estimates for the general population from these nuclear tests
are very low, however, at 0.005 mSv per year (Table 13–​2).
of radionuclides (although still resulting in small doses), especially The manufacture of nuclear weapons was associated with the pro-
mussels and Brazil nuts. duction of uranium or plutonium in nuclear reactors and the storage of
Also associated with natural background is a category of exposures high-​level radioactive waste. At the beginning of the nuclear era, dur-
called TENORM (technologically enhanced naturally occurring radio- ing the 1940s, little attention was paid to the environmental discharges
active materials) resulting from natural radionuclides or radiation, of radioactive material. Large amounts of 131I were released into the
exposing a small part of the population because of human activities, atmosphere at Hanford, Washington, in the United States (Shipler
such as flying, manufacturing, mineral extraction, combustion of fos- et al., 1996) and at Mayak (Eslinger et al., 2014) in the Soviet Union
sil fuels, cigarette smoking, and so on. The resulting exposures are (about 30 PBq at each facility), and substantial amounts of 90Sr, 137Cs,
usually < 0.05mSv (National Council on Radiation Protection and and other short-​lived radionuclides were discharged into the Techa
Measurements [NCRP], 2009). River near Mayak (Degteva et al., 2006).

Man-​made Environmental Exposures Nuclear Accidents.  Since 1957, nuclear power stations have been
used to produce electricity. There are currently 441 nuclear power sta-
Atomic Bombs and Nuclear Weapons Tests. The first tions operable in the entire world, including 99 in the United States
nuclear weapons test took place in July 1945 in New Mexico; it was (International Atomic Energy Agency [IAEA], 2016). Under routine
followed by dropping of atomic bombs on the Japanese cities of operation, environmental discharges of radioactive material have been
Hiroshima and Nagasaki in August 1945 and by the detonation of constantly decreasing since the 1950s and are presently very low, so
nuclear weapons tests in the atmosphere, mainly by the United States that the effective doses received by the populations of the vicinity
and the Soviet Union between 1946 and 1962 (Beck and Bennett, of these plants are very low as well (0.002 mSv per year) (National
2002). The radiation exposures caused by the atomic bombs and the Reasearch Council [NRC], 2012; UNSCEAR, 2010b). Environmental
nuclear weapons tests presented different characteristics and very dif- discharges of radioactive material and resulting doses are also low for
ferent levels of radiation exposure. the other facilities in the nuclear fuel cycle, which include the extrac-
tion of uranium, the preparation of nuclear fuel, its reprocessing (only
Table 13–​2.  Worldwide Averages and Typical Ranges of Annual in a few countries), and the storage of radioactive wastes (UNSCEAR,
Effective Doses (mSv) from Environmental Radiation 2010b).
There have been four reactor accidents that resulted in irreparable
Annual Effective Dose damage to the power plant and in substantial radiation exposures as a
(mSv) consequence of the releases of radioactive materials into the environ-
ment (Bouville et  al., 2014). The first of those accidents took place
Main Type of Worldwide Typical in 1957 at Windscale in the United Kingdom and was caused by a
Source of Radiation Exposure Average Range fire in the reactor core. The second accident took place at the Three
Mile Island (TMI) reactor in the United States in 1979 and was due to
Natural both mechanical and human errors. The third, and most severe, reac-
Cosmic External (μ, e-​, n) 0.39 0.3–​1 tor accident was at the Chernobyl nuclear power plant in the former
Terrestrial External (γ) 0.48 0.3–​1 Soviet Union in 1986, and resulted from a series of human errors dur-
Radon in air Internal (α) 1.26 0.2–​10 ing the conduct of a reactor experiment. Finally, the Fukushima acci-
Radionuclides in food Internal (α, β) 0.29 0.2–​1 dent, which occurred in northern Japan in 2011, was the consequence
Man-​made of an earthquake-​triggered tsunami that damaged the reactor cooling
Atmospheric nuclear tests External (γ) and 0.005 0–​0.05 system. The relative importance of these accidents can be assessed to
internal (β) some extent from the atmospheric discharges of 131I, which were 1 PBq
Nuclear fuel cycle External (γ) 0.002 0–​0.02 at Windscale (Clarke, 1989), 0.0006 PBq at Three Mile Island, 1,800
PBq at Chernobyl (UNSCEAR, 2011), and 120 PBq at Fukushima
Source: UNSCEAR (2010b). (Terada et al., 2012). The best-​documented doses are those related to
230

230 PART III:  THE CAUSES OF CANCER


the Chernobyl accident: in a cohort of about 12,000 children, the mean Occupational Exposures
thyroid dose from intakes of I-​131 was estimated to be 580 mGy, while
the mean whole-​body doses was 14 mGy. There are about 23 million workers who are monitored currently for
their occupational radiation exposure (UNSCEAR, 2010b). The larg-
est group is those in mining (12 million), followed by medical workers
Other Man-​made Sources of Environmental Exposure.
(7 million). In general, occupational doses have been decreasing for
Many industrial operations, security inspection systems (such as some
many decades, and are now very low (effective dose of 1–​5 mSv per
airport screening machines), medical facilities, and educational and
year), which is similar to cumulative annual exposure from natural
research institutions use radiation sources or radioactive material.
background radiation. There are a few exceptions where exposures
Members of the public may be exposed to low levels of radiation when
are increasing, particularly for physicians who perform large num-
they are in proximity to such sources, or when they are near a patient
bers of fluoroscopy-​guided interventional procedures, and radiologic
who has been administered a radiopharmaceutical. Widespread envi-
technologists who administer radionuclides and perform many nuclear
ronmental contamination and exposure of large numbers of people
medicine scans.
have occurred in a few instances when industrial and medical sources
were misplaced or stolen (UNSCEAR, 2011). These include events
that occurred in Goiania, Brazil, in 1987 (IAEA, 1998), in Juarez, ASSESSMENT OF THE EXPOSURE IN ANALYTIC
Mexico, in 1983 (Molina, 1990), and in Taipei, Taiwan, in 1982–​1983
(Chang et al., 1997).
EPIDEMIOLOGIC STUDIES

The ability to estimate a biological measure of dose provides radia-


Medical Exposures tion epidemiologists a great advantage in cancer epidemiology. Dose
reconstruction methods and the associated uncertainties vary consider-
There have been increases in ionizing radiation exposure to the gen- ably, however, according to the source of exposure. In some situations
eral population in the last few decades due to increased medical it is not feasible to estimate doses, in which case exposure is based on
radiation exposure, primarily from CT scans. In the United States the proxy measures such as number of years of employment, or number of
estimated annual per capita effective dose doubled between 1980 and CT scans. Many studies are retrospective because of the long latency
2006 from 3 to 6 mSv (NCRP, 2009). The increase was due to the period for radiation-​related cancer (5+ years for solid tumors) and this
nearly 30-​fold increase in number of CT scans over the period from brings particular challenges due to the use of historical records. The
3 to 80 million per year. Smaller increases in other higher dose diag- degree to which dose estimates are individualized is important, as is
nostic exposures, nuclear medicine procedures, and fluoroscopically the assessment of uncertainties and their impact on risk.
guided interventional procedures also contributed. CT scan use has
also increased in other countries also, although less dramatically than
in the United States (Berrington de Gonzalez et  al., 2009). Current Environmental Studies
levels of CT scan use per capita, for example, are three times lower
Radiation doses from environmental exposures are some of the most
in the United Kingdom than in the United States. Nuclear medicine
challenging and time consuming to estimate. Individual exposure
tests are 30 times less frequent in the United Kingdom than in the
assessment has been conducted for studies of the Japanese atomic
United States. Estimated per capita effective dose from medical radia-
bomb survivors (Cullings et al., 2006), and of local populations around
tion in the United Kingdom has also increased, but from 0.3 in 1988
nuclear weapons sites in Nevada (Till et al., 2014) and at Semipalatinsk
to only 0.4 mSv in 2008 (Hart et al., 2010). The total estimated per
(Gronwald et  al., 2008), and sites of high radioactive discharges at
capita dose from ionizing radiation exposure now varies considerably
Hanford (Kopecky et al., 2004), the Techa River (Degteva et al., 2006),
across countries depending on levels of diagnostic medical exposures
and around the site of the Chernobyl accident (Drozdovitch et  al.,
(Figure 13–​1).
2013). All of the radiation exposures of the study subjects occurred
Although the number of procedures has increased, efforts have been
predominantly in the 1940s and 1950s, with the exception of the
made to reduce the dose per procedure without compromising the
Chernobyl accident, which took place in 1986. In order to evaluate the
medical benefits. The range of average effective doses to the patient
impact of these events, large numbers of radiation measurements on
per procedure varies from 2 to 15 mSv for CT, from 5 to 70 mSv
both the environment and population had been conducted or collected
for fluoroscopically guided interventional procedures, and from 0.2
in most of the geographic domains of interest prior to the decision to
to 40 mSv for nuclear medicine, according to the type of examina-
conduct an epidemiologic study. The approaches for estimating doses
tion (Mettler et  al., 2009). Before 2000, CT scan doses were rarely
to study subjects varied according to the type, quality, and amount
optimized for the smaller body size and weight of children, resulting
of information that was available. No two dose reconstructions were
in unnecessarily high exposures of up to 30 mGy to the lungs/​breast
alike, but there was a general strategy to estimate doses and uncer-
from a chest CT scan and 40 mGy to the brain from a head CT (Kim
tainties that includes (Bouville et al., 2014): (1) collection of as many
et al., 2012). In the United Kingdom, organ doses have been halved for
individual-​based radiation measurements as possible for subjects in
children by optimized protocols (Lee et al., 2016).
the target population, (2) collection of questionnaire-​based individual
External-​beam radiotherapy is the use of external irradiation for
personal and lifestyle information that can influence dose, (3) collec-
the treatment of disease. The therapeutic absorbed dose to the target
tion of information on the spatial and temporal patterns and variations
volume in the patient is orders of magnitude greater than the aver-
of the radiation field, (4) calculation of realistic radiation doses with
age diagnostic dose due to conventional radiography (e.g., 40+ Gy)
efforts to minimize sources of bias, (5)  validation of the dose esti-
(Thierry-​Chef et  al., 2012). Some very high-​dose exposure to sur-
mates by independent measurements or strategies, and (6) qualitative
rounding normal tissue is also unavoidable, but technology is con-
and quantitative evaluation of the uncertainties associated with dose
tinuously evolving to try to minimize these exposures. Currently,
estimates.
radiotherapy is used mostly to treat patients with life-​ threatening
diseases such as cancer, and a limited range of benign diseases like
hyperthyroidism (McKeown et  al., 2015). During the earlier part of Medical Exposures of Patients
the twentieth century, radiotherapy was used to treat a wide variety
of benign conditions including Tinea capitis, tonsillitis, peptic ulcers, Because exposures to patients are given under controlled conditions
and hemangiomas. Once the hazards of ionizing radiation were better that are generally well documented, patients exposed to radiation
recognized, these treatments were stopped, but not before thousands of during treatment for malignant and benign diseases and for diag-
patients had been exposed. Many long-​term epidemiological studies of nostic purposes are well suited to be participants in epidemiologic
these patient populations have documented the excess cancer risks that studies (Stovall et al., 2006). For diagnostic exposures, historically
the patients suffered (Ron, 2003). the documentation was only a written description of the type of
 231

Ionizing Radiation 231


examination, with no details of the specific machines or machine BIODOSIMETRY
parameters. Sometimes information on protocols and machines can
be obtained from current staff or institutional archives in the diag- In addition to the physical methods of dosimetry, biological markers
nostic radiology department. In the absence of specific information, can theoretically be incorporated into epidemiologic studies. The main
organ dose is estimated on the basis of typical values reported in the methods that have been used are the fluorescence in situ hybridiza-
literature for a given examination and time (Stovall et al., 2006), or tion (FISH) technique for analysis of stable chromosome transloca-
via reconstruction of typical values using protocols or survey data tions, and electron paramagnetic resonance (EPR) of tooth enamel and
(Kim et  al., 2012). In the specific case of patients undergoing CT bones. However, these methods are expensive, require extensive effort
scans, computational methods to readily estimate organ doses for in collecting, processing, and analyzing the biological specimens, and,
adults and children of various ages were incorporated into a com- currently, cannot capture exposures below 0.1 Gy. Other techniques
puter program called NCICT (Lee C et al., 2015). Since the wide- for lower doses, such as measurement of gamma-​H2AX foci, are also
spread introduction of electronic records for examinations like available. None of the approaches currently available is both highly
CT, dose reconstruction can now be conducted using the technical sensitive and highly specific to ionizing radiation exposure, and these
parameters directly from these records. As the exposures are usually methods have not been widely employed in epidemiological studies
partial rather than whole body, information on the specific patient (Pernot et al., 2015). For further discussion of radiation biomarkers,
anatomy is needed to help determine which organs were likely to be see the section “Cellular Assays to Measure Biological Changes in
in the exposure field. If patient anatomy is not available, then age-​ Humans” later in this chapter.
and sex-​specific phantoms (models of the human body) can be used,
but this will introduce some error.
For patients treated with external-​ beam radiation therapy, the Uncertainties
quality of the reconstructed dose depends on the degree of complete-
Methods for quantifying uncertainties and incorporating them into
ness and accuracy of the treatment records, which include radiation
the risk analysis are more advanced in radiation than in many other
energy, field size, anatomic location of fields, treatment-​planning
fields, but are still at the development stage (Little et al., 2014; Simon
details, and daily treatment logs and information on patient anatomy.
et al., 2015; Stram et al., 2015). A single ideal approach to evaluate and
To estimate the absorbed doses outside the treatment beam, three
account for all dosimetric uncertainties is not available but is an area
techniques are used: (1) calculation using a three-​dimensional math-
of active research (Kwon et al., 2016; Land et al., 2015; Li et al., 2007;
ematical computer model based on measurements made in a water
Simon et  al., 2015; Stayner et  al., 2007; Stram et  al., 2015). Until
or polystyrene phantom, (2)  direct measurements in an anthropo-
recent years, the evaluation of the uncertainties consisted of numeri-
morphic phantom constructed of tissue-​ equivalent material, and
cal simulations in which variability and lack-​of-​knowledge uncer-
(3) calculation of dose using a treatment-​planning computer program
tainties were combined in Monte-​Carlo simulations. In that method,
(Stovall et  al., 2006). As with diagnostic exposures, if CT images
probability density distributions are assigned to the parameter values
from the treatment-​planning process and patient height and weight
that are deemed to have a substantial influence on the dose estimate,
are available, this will greatly improve the accuracy of the organ
and multiple realizations of individual doses are estimated (NCRP,
dose estimates. Without these, age-​and sex-​specific phantoms can
2007). Consideration of whether shared errors and intra-​individual
be used, but this will reduce dose accuracy, and uncertainty assess-
correlations could be large is required. More sophisticated Monte-​
ments should be undertaken.
Carlo procedures are now being developed to separate and distinguish
between the shared and the unshared components. However, there are
Occupational Studies always concerns that all sources of uncertainty have not been taken
into account. For example, in environmental studies, there may be
Since the 1960s it has been common or required for occupational
“unknown” exposure pathways or unsuspected relevant radionuclides,
radiation workers to wear personal dosimeters, which measure exter-
among other factors.
nal radiation exposures. The dose measurements are monitored and
As a rule, the quality of the individual doses required in analytical
recorded for all workers. This provides an ideal scenario for epidemio-
radiation epidemiologic studies is highest when human-​based mea-
logical studies, particularly if the records are electronic, and for many
surements are reliable, available for all subjects, and representative of
nuclear workers personal dosimeters were available from the start
the organ dose of interest. This is, for example, the case for patients
of their employment. There are still challenges in estimating organ
with therapeutic treatment or workers in nuclear plants. At the other
doses from these badge measurements, however, particularly for medi-
end of the spectrum, the quality of the individual doses is lowest when
cal workers, because of the potential heterogeneity of exposures over
human-​based measurements are not available and the doses have to
the body, differences in where the personal dosimeter is worn on the
be reconstructed on the basis of sparse data and personal interviews
body (e.g., whether on the collar, belt, pocket, outside a lead apron or
conducted long after the exposure occurred. This is the case for some
inside for a medical worker), and whether or not individual protective
environmental studies. However, it is important to note that, despite
devices (e.g., lead aprons) were used by medical workers (Bouville
the absence of human-​based measurements, the individual doses to the
et  al., 2015). Administration of detailed questionnaires to the study
A-​bomb survivors could be reliably reconstructed, but this required
subjects can be used in conjunction with badge doses to address these
several decades of effort and very large resources.
factors (Bouville et al., 2014; Simon et al., 2014). Other considerations
are unmonitored dose when a dosimeter was not worn, doses below
the limit of detection, and doses received at multiple facilities and the
ability to link these records. TYPES OF CANCER CAUSED BY THE EXPOSURE
For exposures predating the 1960s when workers did not have
access to personal dosimeters or when the recorded doses cannot be From the vast array of epidemiological studies of ionizing radiation
retrieved (e.g., for the earliest medical workers), proxy measures of exposure, there is evidence that most types of cancer can be caused
dose are generally used based on surveys and periods of employment by radiation, hence, its classification as a universal carcinogen
(Simon et  al., 2015). Occupational doses resulting from intakes of (UNSCEAR, 2006; BEIR VII). Based on standard laboratory tests of
radionuclides usually are derived from bioassay measurements. These carcinogenicity, ionizing radiation is frequently referred to as a weak
were available for about 40% of the Mayak workers who received carcinogen compared to many chemicals (NCRP, 1997). However, this
internal plutonium exposure, but are not available at many other comparison and generalization are misleading. First, many chemicals
nuclear power facilities (Liu et al., 2016). It may be possible to esti- have tissue-​specific effects and only cause cancer in certain organs
mate doses for these workers using a job exposure matrix if some (NCRP, 1989). Also there is direct evidence from human studies of
measurements as well as a detailed employment history are available high risks in certain organs following radiation exposure in childhood.
(Liu et al., 2016). For example, in the Japanese Life Span Study (LSS), the estimated
23

232 PART III:  THE CAUSES OF CANCER


excess relative risk (ERR)/​Gy for exposure < age 1 year for leukemia radiation exposure, especially from childhood exposure, as about
is 50 (at attained age 20), 9 for thyroid, and 4.5 for breast cancer (Hsu 80% of those exposed before age 10 are still alive (Grant et al., 2017).
et al., 2013; Preston et al., 2002a; Veiga et al., 2016). The most recent analysis of all solid cancer incidence with follow-​up
Several national and international radiation protection agencies extended to 2009, which includes 22,538 cancers, incorporates adjust-
review the epidemiological data periodically and summarize the evi- ment for smoking for the first time and has updated dose estimates. The
dence regarding cancer risks for each cancer site. Here we provide dose–​response for females was linear (ERR/​Gy = 0.64; 95% CI: 0.52,
a meta-​summary of the most recent findings from BEIR VII (2006), 0.77), and similar in magnitude to the previous analysis (Preston et al.,
UNSCEAR (2006), and the Advisory Group on Ionising Radiation 2007). For males, there was evidence of significant upward curvature
(2011) to categorize each cancer site as related, probably related, or described by a linear-​quadratic model with an ERR at 1 Gy of 0.2 (0.12,
uncertain relation to ionizing radiation. We have also incorporated evi- 0.30), which is significantly lower than for females. The EAR estimate
dence from recent studies that were published after these reports. The for females was 54.7 (95%CI:45, 65), while for males the estimate was
majority of cancer sites can be classified as radiation-​related, generally 42.9 (95%CI:27, 58) cases per 10,000 PY at 1 Gy. When restricted to
based on evidence of a significant dose–​response relationship from doses < 0.1Gy, there was still a statistically significant dose–​response
several rigorous epidemiological studies without major biases. relationship using a sex-​averaged linear model (p = 0.038). The addi-
The Life Span Study (LSS) of the Japanese atomic bomb survivors tional follow-​up confirmed that the solid cancer risks remain signifi-
remains the gold-​standard radiation epidemiology study and the pri- cantly increased for more than 60 years after radiation exposure. The
mary source of data for the reports described in the preceding paragraph risk decreased with increasing age at exposure or attained age. There
and the basis for the classification of cancer sites here (Table13-​3). was evidence that a previous finding of an increase in risk for exposure
The cohort includes about 105,000 individuals and 22,500 incident after age 60 (Preston et  al., 2007)  was an artifact due to cases only
cancers during the seven decades of follow-​up (Grant et al., 2017. Key reported on autopsy (Grant et al., 2017). No simple explanation for the
features of the study include the whole body exposure, wide dose-​ introduction of curvature in the dose–​response relationship for cancer
range to healthy individuals of all ages, accurate dose reconstruction, incidence in the males could be identified. The revised dosimetry and
and follow-​up via cancer registries and national death records. For a increased follow-​up time tended to increase the evidence for curvature,
detailed description of the study methods and dosimetry, see Preston while smoking adjustments and sex-​specific cancers were ruled out.
et  al. (2007) and Cullings et  al. (2016). For some cancer sites such There will be a series of updated site-​specific cancer incidence papers
as sarcomas (Henderson et al., 2012), pancreatic cancer (Dores et al., over the next few years that will assess evidence for curvature for each
2014) and rectal cancer (Sakata et al., 2012), higher-​dose therapeutic site in more detail, and that will incorporate additional lifestyle factors
exposures (> 5Gy) are the primary basis for our classification. Absence as potential effect modifiers.
of significantly increased risks from lower-​dose studies for these can- The updated analysis of hematopoetic malignancies with cancer
cers likely reflects lack of power rather than evidence of a threshold for incidence data to 2001 reported that elevated leukemia risks, partic-
these sites. Cancers where there is some evidence of a dose–​response ularly acute myeloid leukemia, persist for more than 50  years after
relationship, but uncertainty about potential confounders or biases, are exposure (Hsu et  al., 2013). The dose–​ response relationship was
described as possibly related to radiation (e.g., chronic lymphocytic linear-​quadratic for acute myeloid leukemia (AML), but linear for
leukemia [CLL], multiple myeloma, and endometrial cancer). Those in acute lymphoblastic leukemia (ALL) and chronic myeloid leukemia
the “unclear” category are sites where there are no high-​quality studies (CML). There is emerging evidence of an increased risk of CLL and
with reasonable power that support a dose–​response relationship with NHL in males, but still no evidence that multiple myeloma or Hodgkin
radiation exposure or sites for which there are inconsistent findings lymphoma are related to radiation exposure. For thyroid cancer, addi-
across studies: non-​Hodgkin lymphoma (NHL); Hodgkin lymphoma tional follow-​up suggests that the excess risk persists for more than
(HL); kidney, cervical, and gallbladder cancers; and melanoma. These 50 years after exposure, but is still largely restricted to exposure before
classifications are difficult due to conflicting results and the rarity of age 20 (Furukawa et al., 2013). An updated analysis of the joint effect
some of these cancers, and the distinction between possibly related of smoking and radiation on lung cancer risk suggested a complex
and unclear should be interpreted cautiously. relationship with smoking intensity where the joint effect was mul-
To illustrate the range in the magnitude of the risks across cancer tiplicative for lower smoking intensities (< 10 cigarettes/​day) and
sites, we present estimates of the ERR/​Gy for cancer incidence from additive or sub-​additive for higher intensities (Furukawa et al., 2010).
the LSS of the Japanese atomic bomb survivors for an individual For uroethelial cancer, the joint effect of smoking and radiation was
exposed at age 30 with an attained age of 70 (Table 13–​3) (Preston best described by a multiplicative model, although the additive model
et al., 2007). Why there are large differences in radiosensitivity of dif- could not be rejected (Grant et al., 2012).
ferent organs is an interesting question that could provide insights into
radiation carcinogenesis. The weighting factors used to estimate the Natural Background Radiation
effective dose (described in the section “Units of Exposure” earlier in Most studies of high natural background radiation have used an eco-
this chapter) aim to capture these differences in sensitivity for radia- logical design, which is susceptible to many biases (Hendry et  al.,
tion protection standards (ICRP, 2007). 2009). The major exception is the case-​control studies of residential
radon exposure, which have provided convincing evidence of a link to
lung cancer due to relatively high doses, careful dose reconstruction,
MAJOR NEW STUDIES AND UPDATES and pooling of multiple studies (Darby et  al., 2005; Krewski et  al.,
2006). The risk estimates from these pooled analyses of residential
We reviewed studies of environmental, medical, and occupational exposure are compatible with the high-​dose studies of radon-​exposed
exposures published since 2006. For studies that have not reported underground miners (Lubin et al., 1997). The joint effect of smoking
significant updates, see the previous edition of this text (Boice, 2006). and radon was multiplicative, which means that the absolute hazard
Summary characteristics of the studies are shown in Table 13–​4. from radon exposure is much higher for smokers. They estimated that
the lifetime lung cancer risk for smokers with high residential radon
exposure (e.g., 800 Bq/​m2 usual exposure) could be increased from
Environmental Exposures 10% to 22%, whereas in non-​smokers the lifetime risk is increased
from 0.4% to 0.9% (Darby et al., 2005).
Japanese Atomic Bomb Survivors There also have been several case-​ control studies of natural
As described in the preceding section, the LSS study has demonstrated background radiation and childhood cancer. A  recent record-​based
that most types of cancer can be caused by ionizing radiation exposure, case-​control study in the United Kingdom (n  =  27,447 cases) using
and has provided quantification of the risks per unit dose and variation radiation exposure from mother’s residence at the child’s birth from
with gender, attained age, age at and time since exposure. The cohort national databases found a significant dose–​ response relationship
continues to provide new information about the long-​term effects of (ERR/​mGy = 0.12, 95%CI:0.03–​0.22) for leukemia in relation to red
 23

Ionizing Radiation 233


Table 13–​3. Cancers Related to Ionizing Radiation and Estimated Excess Relative Risk (ERR) at 1 Gy from the Life Span Study (LSS) of Atomic Bomb
Survivors for Age at Exposure 30 and Attained Age 70

Cancer Radiation Related ERR at 1 Gy* LSS (90% CI) N Cases in LSS Comment

Bladder Yes 1.23 (0.59, 2.10) 469


Breast Yes 0.87 (0.55, 1.30) 1073
Lung Yes 0.81 (0.56, 1.10) 1759 ERR increases with age at exposure.
Leukemia (non-​CLL) Yes 0.79 (0.03, 1.93) 416 Linear term of linear-​quadratic model provided
here.
Brain/​CNS Yes 0.62 (0.21, 1.20) 281
Ovary Yes 0.61 (0.00, 1.5) 245
Thyroid Yes 0.57 (0.24, 1.1) 471 ERR decreases with age at exposure.
Colon Yes 0.54 (0.30, 0.81) 1516
Oesophagus Yes 0.52 (0.15, 1.00) 352
Oral Yes 0.39 (0.11, 0.76) 277 Increase driven by salivary gland, confirmed in
high-​dose radiotherapy studies (Boukheris
et al., 2013).
Stomach Yes 0.34 (0.22, 0.47) 4730
Liver Yes 0.30 (0.11, 0.55) 1494
Non-​melanoma skin Yes 0.17 (0.003, 0.55) 330 ERR decreases with age at exposure.
Bone Yes na 20 Dose–​response relationship reported after high-​
dose radiotherapy (e.g., Henderson et al.,
2012; Rubino et al., 2005).
Soft tissue Yes na 33 Dose–​response relationship reported after high-​
dose radiotherapy (e.g., Henderson et al.,
2012; Rubino et al., 2005).
Pancreas Yes 0.26 (< 0.07, 0.68) 512 Dose–​response relationship reported after
high-​dose radiotherapy (Dores et al., 2014;
Hauptmann et al., 2016).
Rectum Yes 0.19 (−0.04, 0.47) 838 Dose–​response relationship reported after
high-​dose radiotherapy (e.g., Sakata et al.,
2012) and in UK nuclear workers (Muirhead
et al., 2009)
Endometrial Possibly 0.29 (−0.14, 0.95) 184 Possibly increased for exposure < age 20 in
LSS and after high-​dose radiotherapy (e.g.,
Sakata et al., 2012).
Chronic lymphocytic Possibly na 12 Dose–​response relationship reported in
leukemia Chernobyl cleanup workers (Zablotska et al.,
2013) but not in nuclear workers (Leraud
et al., 2015).
Multiple myeloma Possibly 0.38 (−0.23 to 1.36) 136 Dose–​response relationship for cancer incidence
in UK nuclear workers (Muirhead et al.,
2009) but not for mortality in INWORKS
(Leuraud et al., 2015).
Non-​Hodgkin lymphoma Unclear See comment 402 ERR at 1 GY = 0.46 (−0.08, 1.29) for males and
0.02 (< −0.44, 0.64) for females.
Prostate Unclear 0.11 (−0.10, 0.54) 387 No evidence of increased risks in occupational
cohorts (Muirhead et al., 2009).
Renal cell Unclear 0.13 (−0.25-​0.75) 247
Cervix Unclear 0.06 (−0.14, 0.31) 859 No evidence of increased risks after high-​dose
radiotherapy (e.g., Sakata et al., 2012).
Gallbladder Unclear −0.05 (< −0.3, 0.3) 549
Melanoma Unclear na 17

*For age at exposure 30, attained age 70 (solid cancers from Preston et al., 2007, and hematopoietic from Hsu et al., 2013).
na = not available.

bone marrow dose from gamma radiation (mean RBM dose = 4mSv; (Raaschou-​Nielsen, 2008). The UK study is currently being further
range  =  0–​31mSv) (Kendall et  al., 2013). This was also consistent expanded and dose estimates refined.
with the dose–​response relationship from higher dose rate studies,
such as the LSS (Hsu et  al., 2013). There was a weak but not sta- The Chernobyl Accident
tistically significant relationship between radon exposure and leuke- The Chernobyl accident in 1986 is the worst nuclear accident to date,
mia, and no association between gamma or radon dose with any other and the studies of the exposed children, in particular, are the most
childhood cancers. Previous case-​control studies that utilized a similar important source of information on the risks from internal I-​131 expo-
design were null, or not statistically significant for gamma radiation, sure in the general population. There are ongoing follow-​up studies in
but based on a much smaller sample size (range  =  50 to 860 cases Ukraine, Belarus, Russia, and other Baltic countries of children exposed
vs. 27,447) (Laurier et al., 2001; Raaschou-​Nielsen, 2008). There is to internal radiation, primarily from consumption of contaminated milk
evidence from previous case-​control studies with residential measure- and foods, and of the clean-​up workers. A cohort of Ukrainian children
ments of radon concentration of an increased risk of childhood ALL (n  =  12,514) with I-​131 exposure estimated from measurement and
234

234 PART III:  THE CAUSES OF CANCER


Table 13–​4. Summary of the Characteristics of Recent Epidemiological Studies

Typical
Source Study Exposure Type Exposure Pattern Dose Range Outcomes

Environmental Life Span Study Gamma, neutrons Acute 0–​2 Gy All cancer incidence
Residential radon pooled analysis Radon Chronic 0–​1400 Bq/​m3 Lung cancer incidence
UK Background Gamma Chronic 0–​0.015 Gy Childhood cancer incidence
Chernobyl UKRAM cohort I-​131 Chronic 0–​4 Gy Thyroid cancer incidence
Chernobyl BELAM cohort I-​131 Chronic 0–​10 Gy Thyroid cancer incidence
Swiss Nuclear Power Residents External Chronic na Childhood cancer incidence
France Nuclear Power Residents External Chronic na Childhood cancer incidence
German Nuclear Power Residents External Chronic na Childhood cancer incidence
Techa River Gamma, Cesium Chronic 0–​0.5 Gy Cancer incidence

Medical UK Pediatric CT X-​rays Acute, fractionated 0–​0.1 Gy Cancer incidence


Australian Pediatric CT X-​rays Acute, fractionated na Cancer incidence
US Scoliosis X-​rays Acute, fractionated 0–​1 Gy Breast cancer incidence
BRCA cohort X-​rays Acute, fractionated na Breast cancer incidence
Kuwait Thyroid case-​control X-​rays Acute, fractionated na Thyroid cancer incidence
US Brain Cancer case-​control X-​rays Acute, fractionated na Brain cancer incidence
USRT personal diagnostic exposures X-​rays Acute, fractionated na Thyroid cancer incidence
UKCCS X-​rays Acute, fractionated na Childhood cancer incidence
CCSS X-​rays Acute, fractionated 0–​80 Gy Second cancer incidence
UKCCS X-​rays Acute, fractionated 0–​80 Gy Second cancer incidence
French childhood cancer survivors X-​rays Acute, fractionated 0–​80 Gy Second cancer incidence
Hodgkin’s lymphoma survivors X-​rays Acute, fractionated 0–​80 Gy Second cancer incidence
Second GI cancers study X-​rays Acute, fractionated 0–​80 Gy Second cancer incidence
Retinoblastoma cohort X-​rays Acute, fractionated 0–​80 Gy Second cancer incidence

Occupational Mayak workers Gamma, plutonium Chronic 0–​4 Gy Cancer incidence and mortality
15 Countries Study Gamma Chronic 0–​0.05 Gy Cancer mortality
INWORKS Gamma Chronic 0–​0.05 Gy Cancer mortality
UKNRRW Gamma Chronic 0–​0.05 Gy Cancer incidence
French nuclear workers Gamma Chronic 0–​0.05 Gy Cancer mortality
US nuclear workers Gamma Chronic 0–​0.05 Gy Cancer mortality
Flight crew pooled analysis Cosmic Chronic na Cancer mortality
Scandinavian flight crew Cosmic Chronic na Breast cancer incidence
US flight crew Cosmic Chronic na Breast cancer incidence
Chinese medical workers X-​rays Chronic 0–​0.4 Gy Cancer incidence
USRT X-​rays Chronic 0–​0.1 Gy Cancer incidence

na = not available.

interviews has undergone four rounds of thyroid screening. In the most direct interviews (0.88) than proxy responders (3.98), but confidence
recent analysis, the linear dose–​response relationship for thyroid can- intervals were wide due to the small number of controls with proxy
cer (n = 65, ERR/​Gy = 1.91; 95% CI: 0.43,6.34), was consistent with interviews (Zablotska et al., 2011).
the risk from external acute exposure in the Japanese atomic bomb sur-
vivors (Brenner et al., 2011). Similar results were reported in a cohort Cancer Near Nuclear Power Stations
of children in Belarus (n = 84 thyroid cancers) (Zablotska et al., 2011). Residents living near nuclear power stations remain concerned about
Assessment of dose uncertainty did not significantly alter these risk potential cancer risks, especially childhood leukemia, since the early
estimates (Little et al., 2014, 2015). More details on the relationship reports of potential cancer clusters (Black, 1984). Study design issues
between I-​131 and thyroid cancer are provided in Chapter 44. include small sample size and ecological design. A recent large cohort
Interview-​ based case-​ control studies of adulthood exposures study based on the Swiss Childhood Cancer Registry (n  =  2925)
received by the liquidators reported an increased risk of thyroid can- using distance from residence at birth from a nuclear power plant and
cer (n  =  107, median thyroid dose  =  69 mGy) (Kesminiene et  al., national census data found no evidence of a relationship between dis-
2012) and leukemia (n = 137) including both CLL and non-​CLL (con- tance and childhood leukemia (Spycher et al., 2011). The study could
trols mean RBM dose = 82 mGy) (Zablotska et al., 2013). Although not, however, rule out small risks due to limited statistical power, and
the non-​ CLL leukemia results were consistent with the Japanese distance is a crude and potentially inaccurate proxy for radiation expo-
atomic bomb survivors, the excess risks for CLL and thyroid cancer sure. Two studies with a similar design in France and Germany found
after adulthood exposure were much higher than those observed in some evidence of a relationship between proximity to a power sta-
the LSS (Hsu et al., 2013; Preston et al., 2007). Prevalent case-​control tion and leukemia (Kaatsch et al., 2008; Sermage-​Faure et al., 2012).
studies conducted more than 20 years after the accident could be sub- A  review of all the studies and new UK data concluded that there
ject to recall bias. In the leukemia/​CLL case-​control study, 50% of the remained suggestive evidence of small risks, but confounding could
cases compared to 5% of the controls had died, and dosimetry was not be ruled out, especially given the finding of a similar association
based on proxy responders. The ERR/​Gy was lower in the subset with with distance from sites for proposed nuclear power stations that were
 235

Ionizing Radiation 235


never built (Stather, 2011). A proposed US study was canceled after atomic bomb survivors when restricted to a similar exposure age and
feasibility work suggested that the time and costs involved were exces- follow-​up period. The dose–​response relationship for brain tumors
sive (Nuclear Regulatory Commission, 2015). An earlier US study of (n = 135) and estimated brain dose (mean = 43 mGy) was also statisti-
cancer mortality in 107 counties near nuclear facilities and matched cally significant, but about four times higher than the comparable risk
control counties was null, but with the caution that the risks may have estimate in the LSS. Sensitivity analyses that assessed additional clini-
been too small to detect with the ecological study design (Jablon cal information suggested that there was some evidence of bias due to
et al., 1991). unreported previous brain tumors, and when these were excluded the
ERR/​mGy was still statistically significant but slightly closer in mag-
Techa River Residents nitude to the LSS (Berrington de Gonzalez et al., 2016). The cohort is
The population living near the Techa River received protracted low-​ currently being expanded to include patients who underwent CT scans
dose radiation exposure from internal and external sources following up to 2010. Additional data have been collected to refine the dosimetry
radioactive releases into the river from the Mayak nuclear weap- and conduct analyses of the impact of dose uncertainty (Lee et  al.,
ons plants. In the cohort of residents followed for cancer incidence 2016). The cohort also forms one of the centers of the multi-​center
(n  =  17,433) with a mean organ dose around 50 mGy, there was a EPI-​CT study (Bosch de Basea et al., 2015). With data from nine coun-
statistically significant linear dose–​response relationship for all solid tries across Europe, the pooled study aims to include approximately
cancers and leukemia (excluding CLL) (Krestinina et al., 2007, 2013). one million children, and results are due in the next few years.
There is limited power for site-​specific solid cancer risks (n = 2059), An Australian cohort used Medicare (universal healthcare scheme)
but, of 15 sites evaluated, significant dose–​ response relationships records to identify 680,000 children who had CT scans between 1985
were recently reported for cancer of the esophagus and uterus (Davis and 2005, aged < 20 years, and approximately 10 million children who
et al., 2015). were not exposed (Mathews et  al., 2013). Through record linkage,
3150 cancers were recorded in the exposed children. They reported an
increase in incidence rate ratio for all cancers of 0.16 (95%CI:0.13–​
Medical Exposures 0.19) for each additional CT scan. Site-​specific analyses suggested
increased risks of solid cancers in digestive organs, melanoma, soft tis-
Diagnostic Medical Radiation sue, female genital, urinary tract, brain, and thyroid cancers, and also
The increase in CT scan use and publications reporting unnecessarily leukaemia, myelodysplastic syndromes, and some other lymphoid can-
high dose levels in children in 2001 (Paterson et al., 2001) prompted cers. The results are difficult to compare with the UK cohort because
the initiation of a number of retrospective cohort studies of pediatric of the difference in analytic approach (organ dose versus number of
CT scans and cancer. Collaboration between the investigators ensured CT scans) and different exclusion periods (2–​5 years in the UK versus
that protocols were broadly similar to facilitate future pooling of the 1 year in the Australian study). The lack of specificity by cancer site
data. The UK cohort consists of approximately 180,000 children and and increased risks for cancers not usually considered to be radiation-​
young adults (age < 22 years) who underwent CT scans between 1980 related (e.g., melanoma and lymphomas) raised concerns about poten-
and 2002 in about 100 hospitals (Pearce et al., 2012). The exposure tial bias from preexisting conditions. Dose reconstruction is underway
data were abstracted from electronic databases in radiology depart- using methods similar to the UK CT study, which should facilitate
ments and were linked to national cancer registration and vital status comparisons. A Canadian cohort is also in progress with an identical
databases. Patients with cancer diagnosed before the date of the first design to the UK cohort using electronic medical records from Cancer
CT scan were excluded. Organ-​specific dose estimates were semi-​ Care Ontario linked to cancer registrations (Einstein, 2012). This is the
individualized, as they were based on body region scanned, date of only study currently that will also include adulthood exposures.
scan, and age at scan. With a 2-​year lag period, there was evidence of a Conventional diagnostic X-​rays continue to be a common source
significant dose–​response relationship for leukemia/​MDS (n = 74) and of medical radiation exposure, although doses are typically one-​tenth
red bone marrow dose (mean = 12 mGy) (Figure 13–​2). The ERR/​mGy of the dose of CT scans to the equivalent body region. Direct studies
was statistically compatible with the risk estimate from the Japanese of potential cancer risks face a number of challenges, including low
power and exposure misclassification. Cohorts of special populations
who received unusually high exposure levels (e.g., women with sco-
liosis and tuberculosis) based on medical records have, however, pro-
8
vided important information about the cancer risks from repeated low
doses of radiation. An updated follow-​up of the US scoliosis cohort
7 (n = 3010) reported a borderline significant dose–​response relationship
for breast cancer (ERR/​Gy = 2.86, n = 78, p = 0.058) (Ronckers et al.,
6 2008). The mean breast dose was 120 mGy and the dose–​response
relationship was significantly greater for women who reported a fam-
5 ily history of breast cancer, although this was based on small numbers.
RR (& 95% CI)

Several recent case-​ control studies reported increased risks for


brain and thyroid tumors after dental X-​rays, and breast cancer after
4 chest X-​rays in BRCA mutation carriers (Andrieu et al., 2006; Claus
et al., 2012; Memon et al., 2010). However, recall bias is a concern in
3 case-​control studies that rely on self-​reported exposure information.
In a cohort of US radiologic technologists there was evidence of an
2 increased risk of thyroid cancer after childhood dental X-​rays based
on prospective follow-​up after self-​reporting of X-​ray exposure his-
tory at baseline (Neta et al., 2013). Compared to the general popula-
1
tion, these medical workers should be able to report their history of
diagnostic exposures more reliably. Analyses of other cancer sites and
0 personal diagnostic exposures are ongoing. The UKCCS case-​control
0 10 20 30 40 50 60
study (n = 2690 childhood cancer cases diagnosed 1976–​1996), which
RBM dose (mGy) used record linkage to evaluate diagnostic X-​rays in utero and in early
infancy (< 100 days), reported a non-​statistically significant increased
Figure 13–​2.  Relative risk (and 95% CI) for leukemia/MDS according to risk from in utero X-​rays for all cancers (OR  =  1.14; 95%CI:0.90,
the estimated absorbed red bone marrow (RBM) dose (Gy) from pediatric 1.45) and leukemia (OR = 1.36; 95%CI:0.91, 2.02) (Rajaraman et al.,
CT scans in the UK cohort. Source: Pearce et al. (2012). 2011). The results are compatible with lower doses compared to the
236

236 PART III:  THE CAUSES OF CANCER


period of the earlier studies in the United Kingdom and northeastern Studies of workers at these facilities have provided information on
United States (Doll and Wakeford, 1997). risks of cancer following low-​dose, protracted exposure to external
ionizing radiation. An important feature of these populations is the
Radiotherapy routine dose monitoring, which can be used to reconstruct individ-
With the increasing population of cancer survivors and longer survival, ual long-​term dose histories. These studies generally aim to assess
the risk of a treatment-​related second cancer is an increasing clinical whether the risk of cancer per unit dose at low doses and low-​dose
and public health concern. Approximately half of all cancer patients rates is similar to that estimated from acute doses in the Japanese LSS.
in countries like the United States receive radiotherapy as part of their The major studies published in the past 10 years, which are discussed
initial treatment. Although there are increasing efforts to minimize the in the following paragraph, include those of workers at the Mayak
dose to the normal tissue surrounding the tumor, some very high doses facility and of cleanup workers at the Chernobyl nuclear power plant,
(< 40 Gy) to some proximal organs are unavoidable. In the last decade the 15-​country study by the International Agency for Research on
there has been considerable progress in our understanding of the can- Cancer (IARC), and its successor the International Nuclear Workers
cer risks from partial-​body, high-​dose fractionated exposure, due to Study (INWORKS). Also discussed are major national cohorts of
the publication of a plethora of new dose–​response studies for sec- nuclear workers exposed to external ionizing radiation, such as the UK
ond cancers after radiotherapy (Berrington de Gonzalez et al., 2013; National Registry of Radiation Workers, the Canadian National Dose
NCRP, 2011). In general, these case-​control studies of second solid Registry, and pooled studies of US nuclear workers.
cancers (breast, lung, stomach, pancreas, thyroid, brain, sarcoma, skin, Studies of workers at the plutonium production facility in the
esophagus, bladder, colon and rectum) based on medical records and Russian Federation (former Soviet Union) have been highly informa-
individual dose reconstruction, suggest a linear dose–​response rela- tive on the health effects of protracted exposure to gamma radiation
tionship even for very high, fractioned, organ doses (40+ Gy), which exposure, and provide a rare opportunity to study the risks from plu-
was in contrast to the expectation of a flattening or even downturn tonium exposure. Workers in certain plants at the Mayak Production
in risk at high doses due to cell killing. However, the ERR/​Gy from Association received high plutonium doses (mean lung doses: 0.12 Gy,
these high-​dose fractionated exposures is lower than the risk from the range 0–​1 Gy), due to several spills and accidents that occurred during
acute exposure in the Japanese atomic bomb survivors (Figure 13–​3). periods of heavy production between 1948 and 1958. The key recent
Thyroid cancer is the only clear exception where there is a clear down- studies addressing the effects of plutonium include an updated analysis
turn in risk after about 20 Gy in childhood cancer survivors (Bhatti of lung cancer mortality (Gilbert et al., 2013) and incidence (Labutina
et al., 2010b; Veiga et al., 2012) (Figure 13–​3). et al., 2013), as well as the incidence of liver, bone, and other cancers
Although the ERR/​Gy is lower than reported in the LSS, the very (Hunter et  al., 2013; Labutina et  al., 2013). Strengths of the Mayak
high doses can result in some high absolute risks for common cancers. studies include the large cohort size, high exposures, relatively large
For, example an HL patient treated with 40+ Gy to the chest at age 25 is numbers of exposed female workers, availability of cancer incidence
estimated to have a 30% risk of developing radiation-​related breast can- data, and extensive efforts to characterize dose. Challenges of these
cer by age 55, which is comparable to a BRCA2 mutation carrier (Travis studies include a lack of complete biomonitoring for plutonium expo-
et al., 2005). The ERR/​Gy is generally higher for childhood exposure, sure, resulting in large dose uncertainties, and high rates of smoking
as observed in the LSS, which also contributes to high absolute risks and heavy alcohol consumption, which have led to concerns about
(Berrington de Gonzalez et al., 2013; Grant et al., 2017. As described potential confounding and have raised questions about transfer of risks
earlier, several of these recent case-​control studies of high-​dose radio- to other exposed populations.
therapy provided the first clear evidence that pancreatic cancer (Dores Major attention has focused in the Mayak cohort on cancer in lung,
et  al., 2014; Hauptmann et  al., 2016), rectal cancer (Sakata et  al., liver, and bone, as these are the primary sites associated with plutonium
2012), and sarcomas (Henderson et al., 2012; Rubino et al., 2005) are deposition following inhalation exposure. With 486 deaths from lung
radiation-​related. For more details on second cancers, see Chapter 60. cancer (most among men), the latest study found substantial eleva-
tions in risk with increased plutonium dose: ERR per Gy was 7.4 (95%
CI: 5.0, 11) among men with an attained age of 60 and declined with
Occupational Exposures increasing age (Gilbert et al., 2013). Among women, ERR/​Gy at age
60 was 24 (95% CI: 11, 56). Restricting analyses to workers exposed
Nuclear Workers at lower plutonium doses found similar risks per unit dose as in the full
Hundreds of thousands of workers worldwide have been exposed to cohort, and ruled out anything but very small departures from linearity.
radiation during employment in nuclear weapons and energy facilities. Importantly, the study was able to adjust for tobacco smoking and to

Breast cancer after Childhood cancer (n = 107) Thyroid cancer after Childhood cancer (n = 115)
(Inskip et al., 2009) (Bhatti et al., 2010)
20.0 40.0

35.0

15.0 30.0
RR (& 95% CI)
RR (& 95% CI)

25.0

10.0 20.0

15.0

5.0 10.0

5.0
0.0 0.0
0 10 20 30 40 50 60 0 10 20 30 40 50
Mid-point of dose category (Gy) Mid-point of dose category (Gy)

Figure 13–​3.  Relative risk (and 95% CI) for subsequent cancer according to the estimated absorbed radiation dose (Gy) from fractionated radiotherapy.
Dotted black line indicates fitted dose-response for that study. Dashed grey line indicates the RR for similar age at exposure and attained age based on the
BEIR VII risk models for low-dose exposure (BEIR VII, 2006). The fitted linear dose-response model for Bhatti et al (2010)b is based on the linear term
from the linear-quadratic model.
 237

Ionizing Radiation 237


evaluate the form of interaction between smoking and plutonium dose. the data were restricted to lower dose ranges (< 100 mGy), or when
It found a sub-​multiplicative but super-​additive interaction (discussed excluding or adjusting for exposure to neutrons or internal emitters.
further in the section on effect modification). Analyses of lung cancer Indirect evidence suggested little potential for confounding by smok-
incidence supported the main findings of a steep dose-​response rela- ing in the analysis of all solid cancers, as the risk estimates were
tionship (Hunter et  al., 2013; Labutina et  al., 2013). An analysis by similar when restricted to non-​smoking-​related cancers. The leuke-
lung tumor subtype found a much higher ERR/​Gy for adenocarcinoma mia subtype showing the highest risk was CML, for which the ERR/​
than for squamous cell subtypes, suggesting an important etiologic dif- Gy was 10.45 (90% CI: 4.48, 19.65), and this pattern was consistent
ference for these two subtypes. Bone and connective tissue cancer inci- across the countries (Leuraud et al., 2015). The ERR/​Gy for AML was
dence was also found to be highly elevated (RR = 13.7; 95% CI: 3.0, 1.29 (90% CI: –​0.82, 4.28). Although the mean cohort dose was only
58.5) among a group of unmonitored female workers employed before 25 mGy, the risks per unit dose and subtype patterns were similar to
1950 at a plutonium-​processing facility with heavy exposure potential the recent analysis in the LSS (Hsu et al., 2013). The size, high-​quality
(Cardis et al., 2007). Studies of other cancer types have found a highly dosimetry, and lengthy follow-​up of the INWORKS cohort suggest
significant dose–​response relationship for liver cancer and plutonium, that it will remain an important study through which to understand
but not most other specific cancers (Hunter et al., 2013; Labutina et al., the risk of protracted exposure to low-​dose photons, primarily in adult
2013). Efforts are ongoing to assess the impact of the large uncertain- males. The pooled cohorts will also be informative on site-​specific
ties in the plutonium dose estimates (Stram et al., 2015). cancer risks and effect modification by temporal factors, especially as
The IARC has led a number of pooled international studies of follow-​up is extended for the 78% alive at the end of the recent study.
nuclear workers. In the first decade of the 2000s, a series of stud- Studies of the component cohorts of INWORKS have also yielded
ies was published for solid cancer and leukemia mortality risks in a useful information about risk of grouped and individual cancers. The
pooled study of nuclear workers in 15 countries (Cardis et al., 2005, UK NRRW cohort included 174,541 workers monitored for exter-
2007; Vrijheid et al., 2007, 2008). The study, which included 407,000 nal ionizing radiation (Muirhead et  al., 2009), followed for cancer
workers exposed to an average cumulative dose of 19 mSv, found an incidence and mortality through 2001, with 16% deceased. Major
ERR/​Sv of 0.97 (95% CI: 0.14, 1.97) for all cancers excluding leuke- strengths of the cohort include its large size, well-​ characterized
mia and 1.93 per Sv (95% CI: < 0, 8.47) for leukemia excluding CLL gamma doses, wide representativeness of the facilities included, and
(Cardis et al., 2005). Although no significant heterogeneity in risk was availability of cancer registry linkage. A  limitation of this cohort is
seen in these results by country or large facility (Cardis et al., 2007), a the lack of specificity about which workers may have had substantial
number of subsequent publications questioned the results of the study, internal (e.g., plutonium) dose. The ERRs per Sv observed for leuke-
pointing out that findings contributed by Atomic Energy of Canada mia (excluding CLL) mortality and incidence (respectively) were 1.71
Ltd seemed highly influential on the statistical significance of the solid (90% CI: 0.06, 4.29) and 1.78 (90% CI: 0.17, 4.36). For all cancers
cancer results, and questions arose about the quality of the dosimetry excluding leukemia, the ERR/​Sv was 0.28 (90% CI: 0.02, 0.56) and
data for this cohort (Zablotska et al., 2014; more information is given 0.27 (90% CI:  0.04, 0.51), respectively. Lymphoma showed a bor-
about this later in this chapter). In addition, it was noted that the exclu- derline positive trend with radiation dose (p  =  0.081; 81). Findings
sion of workers with substantial neutron exposure or radionuclide con- for multiple myeloma differed between the mortality and incidence
tamination reduced the overall power of the study, as workers with follow-​up:  the ERR/​Gy was 1.20 (90% CI:  –​0.88, 5.96) for mortal-
neutron dose or internal dose contamination also were more likely to ity (n = 113) and 3.60 (90% CI: 0.77, 8.94) for incidence (n = 149),
have high photon doses (Boice et al., 2006). Despite these criticisms, respectively (Muirhead et al., 2009).
the 15-​country study remains an important contributor to information The US pooled nuclear worker cohort (Schubauer-​Berigan et  al.,
about effects of dose protraction on cancer risk from photon radia- 2015b) includes 119,195 workers monitored for external ionizing radi-
tion exposure, particularly with regard to lymphatic and hematopoietic ation, from four Department of Energy (DOE) nuclear weapons sites
cancers, in which the influence of the Canadian cohort was minimal. (Hanford, Idaho National Laboratory, Oak Ridge National Laboratory,
Strengths of the study include the use of a common protocol and a and Savannah River Site) and the Portsmouth Naval Shipyard. With
focus on evaluation of workers exposed only to external ionizing radi- mortality follow-​up through 2005, 35% of the cohort was deceased,
ation, which minimized the influence of poorly characterized internal and 10,877 cancers (excluding leukemia) and 369 leukemias (exclud-
exposures (IARC, 2012). ing CLL) were observed. Few of these workers received a confirmed
Since the publication of the 15-​country study, it was recognized deposition of internal radiation, and neutron dose was less than 2% of
that continuing mortality follow-​up of its contributing cohorts would the total dose for the cohort. The study found ERR/​Sv values of 1.7
provide an important addition to knowledge on low-​dose cancer risk (95% CI: –​0.22, 4.7) for leukemia and 0.14 (95% CI: –​0.17, 0.48) for
from photon exposure, as only a small percentage of each cohort all cancers excluding leukemia. For the latter group, findings differed
was deceased at the time follow-​up ended for the 15-​country study by cancers classified into those related to smoking (ERR/​Sv: –​0.079;
(NRC, 2006). 95% CI: –​0.43, 0.32) and those unrelated to smoking (ERR/​Sv: 0.70;
INWORKS was formed by a team from the IARC and international 95% CI:  0.058, 1.5). This heterogeneity suggests that negative con-
collaborators involved in the 15-​country study, to study radiation-​ founding may exist with cigarette smoking (where adjustment would
related risks of cancer mortality in the nuclear worker cohorts that shift the risk estimate away from the null), which was also supported
contributed more than 80% of deaths in the 15-​country study: the UK by facility-​specific analyses of lung cancer and chronic obstructive
National Registry of Radiation Workers (NRRW; Muirhead et  al., pulmonary disease. The ERR/​Sv for mesothelioma of 2.5 (95% CI: –​
2009), the French combined nuclear workers study (Metz-​Flamant 1.3, 10) suggests that positive confounding by asbestos exposure may
et  al., 2013), and the US pooled nuclear workers study (Schubauer-​ exist. Other notable findings include significant dose–​response pat-
Berigan et al., 2015b). All three studies have updated mortality follow-​ terns for lymphoma (ERR/​Sv: 1.8; 95% CI: 0.027, 4.4) and multiple
up since the publication of the 15-​country study (Hamra et al., 2015), myeloma (ERR/​Sv: 3.9; 95% CI: 0.60, 9.6).
and in some cases (Schubauer-​Berigan et  al., 2015b) included addi- The French cohorts in INWORKS include 59,021 workers followed
tional cohorts primarily exposed to photon radiation. The Canadian for mortality between 1968 and 2004 at three large nuclear energy
workers were not included, and this avoids the problems with this or weapons cohorts:  Commissariat à l’Energie Atomique (CEA),
cohort described earlier. Electricité de France (EDF), and AREVA Nuclear Cycle (AREVA)
INWORKS includes more than 300,000 workers, with 531 leuke- (Leuraud et al., 2015). Mean cumulative effective dose was 22.5 mSv.
mia (other than chronic lymphocytic) deaths (Leuraud et  al., 2015) With just 11% of the cohort deceased, the ERR/​Sv was 0.34 (90%
and nearly 18,000 deaths from solid cancers (Richardson et al., 2015). CI: –​0.56, 1.4) for all solid cancers, 4.0 (90% CI: < 0, 17) for leuke-
The pooled analysis found a significantly elevated risk of leukemia mia excluding CLL, and was non-​calculable for multiple myeloma and
(ERR/​Gray [Gy]: 2.96; 90% CI: 1.17, 5.21) and solid cancers (ERR/​ lymphoma.
Gy: 0.48; 90% CI: 0.18, 0.79) (Leuraud et  al., 2015; Richardson The Canadian National Dose Registry (CNDR) is a centralized
et al., 2015) (Figure 13–​4). These point estimates changed little when occupational radiation dose registry, containing records for more than
238

238 PART III:  THE CAUSES OF CANCER


(a) Leukemia (b) Solid cancer
1.7
6
1.6
5
1.5

RR (& 90% CI)


RR (& 90% CI) 4 1.4

1.3
3
1.2
2
1.1

1 1.0

0.9
0 0 100 200 300 400 500 600 700
0 50 100 150 200 250 300 350 400 450
Cumulative dose (mGy)
Cumulative RBM dose (mGy)

Figure 13–​4.  Relative risk (and 90% CI) for leukemia (excluding CLL) and solid cancer according to the cumulative radiation dose (Gy) from occupational
radiation exposures in the INWORKS cohort. Source: Leuraud et al. (2015); Richardson et al. (2015).

600,000 workers, including underground miners exposed to radon, in radiation protection such as lead apron usage (Linet et al., 2010;
nuclear power plant workers, and medical personnel (Ashmore et al., Simon et al., 2014).
1998). The cohort or subcohorts within it have been periodically linked
to mortality and cancer registry databases (Sont et  al., 2001). Mean Flight Crew
cumulative doses were highest in nuclear power workers (20 mSv) and Studies of cancer in flight crew have provided equivocal evidence of
lowest in dental and medical workers (0.3 and 4 mSv, respectively). cancer risk following repeated exposure to cosmic radiation. Typical
The study found a significantly elevated ERR/​Sv among male (but doses in these studies are low and have little within-​cohort varia-
not female) registrants, including all leukemias combined, all cancers bility:  for example, a mean absorbed, whole-​body dose of 12 mGy
excluding leukemia, and for many individual cancer sites (Sont et al., (standard deviation 11 mGy) was observed in a recent breast cancer
2001). Since then, studies assessing the impact of dose censoring (i.e., incidence study among flight attendants (Schubauer-​Berigan et  al.,
doses below certain thresholds were unrecorded during the early regis- 2015a). Empirical evidence also suggests that the neutron dose qual-
tration years) and cancer risk in a subgroup of medically exposed reg- ity factor associated with commercial flight conditions is lower than
istrants have been published (Shin et al., 2005; Zielinski et al., 2009). previously thought (e.g., 2–​2.5; Burda et  al., 2013), amplifying the
No extension of follow-​up has occurred past the late 1980s (cancer low-​dose limitations of these studies. Recent studies of flight crew,
incidence) or mid-​1990s (cancer mortality) for the CNDR. Since the including a pooled international study, found no elevation in breast
publication of the 15-​country study, critical attention has focused on cancer mortality (compared to the general population) and no asso-
the completeness of the dosimetry and registry information for one ciation with cosmic radiation (Hammer et al., 2014; Pinkerton et al.,
of the component cohorts (Atomic Energy of Canada Ltd; Ashmore 2012). A large, pooled Scandinavian cancer incidence and case-​control
et  al., 2010). It was noted that risk estimates for this cohort greatly study found a 50% higher risk of breast cancer than in the general pop-
increased in studies based on CNDR data (e.g., Cardis et  al., 2005, ulation, but no association with cosmic radiation or other occupational
2007; Sont et al., 2001; Zablotska et al., 2004) as compared to earlier exposures (Pukkala et al., 2012). Another large study of breast cancer
studies based directly on the facility data (e.g., Cardis et  al., 1995). incidence among US flight attendants found that, while the rate of inci-
Efforts are currently focused on ascertaining that dosimetry and regis- dent breast cancer was 37% higher than among the general US popula-
trants in the CNDR are complete and accurate (Ashmore et al., 2010; tion, the increase was probably due to the much older age at first birth
Zablotska et al., 2014). and lower parity in the cohort compared to US women (Schubauer-​
A new, large-​scale study, the Million Worker Study (MWS) has Berigan et al., 2015a). No association was observed between cosmic
been developed with the aim of evaluating cancer risk from pro- radiation dose and breast cancer incidence, except among high-​parity
tracted radiation exposures of < 100 mGy by combing various groups women and those with younger age at first birth (Pinkerton et al., 2016;
of US workers, including “atomic veterans” (i.e., military personnel Schubauer-​Berigan et al., 2015a). This pattern, which is the opposite
who witnessed atomic test blasts), nuclear weapons workers, nuclear of the expected interaction of radiation with reproductive risk factors
power plant workers, industrial radiographers, and medical workers (e.g., Ronckers et al., 2005), may be associated with circadian disrup-
exposed to radiation (Bouville et al., 2015). While the proposed sam- tion, which was highly correlated with radiation dose in the cohort.
ple size is impressive, at this stage the epidemiological methods for In addition to breast cancer, the occurrence of melanoma has been
obtaining complete and accurate follow-​up for each cohort (including of interest among flight crew. A recent meta-​analysis of 19 studies of
confirming who is alive, and cause of death) have not been described cockpit and cabin crew found that, compared to the general popula-
and may be prohibitively expensive. Inaccurate follow-​up can be an tion, melanoma incidence rates are approximately doubled for flight
important, but often overlooked, source of bias in epidemiological crew, with a similar excess observed in both cockpit and cabin crew
studies; non-​differential outcome misclassification will bias risk esti- (Sanlorenzo et  al., 2015). The authors attribute this excess to occu-
mates towards the null. There are also considerable challenges in the pational sources, including potentially ultraviolet-​ A radiation and
dose reconstruction, including substantial dose from radionuclides cosmic radiation, although no direct adjustment for factors such as
among some contributing DOE cohorts, which will require exten- recreational sun exposure was made.
sive dose reconstruction efforts and possibly involve large uncertain-
ties (e.g., Mound and Rocketdyne; Bouville et  al., 2015). For the Medical Workers
proposed group of 240,000 medical workers, the dosimetric chal- Studies of increased leukemia risks in the earliest radiologists pro-
lenges will include non-​uniform exposures, lack of compliance with vided the first evidence of a link between radiation and cancer (March,
badge monitoring, employment in multiple facilities, and variation 1950). Most of the studies of medical radiation workers have limited
 239

Ionizing Radiation 239


exposure information, particularly for the early workers, when radi- low and protracted doses of radiation have been carried out by the US
ation protection and dosimeters were rare. A  new follow-​up of a National Academies as a series of reports on the “Biological Effects
cohort of 27,000 Chinese medical radiation workers with individual of Ionizing Radiation” (BEIR) and by the United Nations Committee
dose estimates (mean = 0.086 Gy) found a significant dose–​response on the Effects of Atomic Radiation (UNSCEAR, 2010b). The most
relationship for solid cancer incidence that was higher than, but sta- recent BEIR report (BEIR VII Phase 2, 2006)  reviewed the animal
tistically compatible with, risk estimates from other low-​dose stud- and epidemiological data and derived a dose-​and-​dose-​rate adjustment
ies (Sun et al., 2016). Individual dose reconstruction has also recently factor (DDREF) of 1.5 (95% credible interval), which implies that
been completed for the US Radiologic Technologists (USRT) cohort the risk per unit dose is reduced by one-​third if the dose is protracted
of about 110,000 (mostly female) workers (Simon et al., 2014). There or below 100 mGy. However, the recent studies of occupationally
was no evidence of a dose–​response relationship for basal cell skin can- exposed individuals, such as INWORKS, natural background radia-
cer in these workers, even though some received estimated skin doses tion, Techa River residents, and the UK CT (computed tomography)
of > 1 Gy (Lee T et al., 2015), or for brain cancers, even among those study reported dose–​response relationships from these protracted/​frac-
who performed fluoroscopically guided procedures (Kitahara et  al., tionated low-​dose exposures that were compatible with the Japanese
2017). A significant dose–​response relationship was found, however, atomic bomb survivors, implying compatibility with a DDREF of 1. In
for breast cancer for those workers who started work before 1950 and the next few years there will be additional results from nuclear worker
received the highest doses (estimated mean organ dose = 280 mGy) and CT scan cohorts that will provide further information on this ques-
(Preston et al., 2016). Dose–​response analyses for other cancer sites tion, which is central to radiation protection limits.
are ongoing, but power is generally limited for sites other than breast The impact of fractionation and dose level is also critical in the
cancer due to low mean doses and relatively small numbers of cases. study of second cancer risks after radiotherapy to aid clinical decision-​
Some case reports of brain tumors in physicians who perform fluo- making, and also provides insights into biological mechanisms.
roscopically guided procedures have raised concerns about current Mostly the dose–​response studies have found that the risk per unit
exposure levels in these physicians (Roguin et al., 2013). In a subset dose is 5–​10 times lower following these high-​dose (5+ Gy) highly
of the USRT cohort, technologists who reported performing fluoro- fractionated (e.g., 40 fractions) exposures compared to the acute expo-
scopically guided interventional procedures were at an approximately sure of < 2 Gy in the Japanese atomic bomb survivors (Berrington de
two-​fold increased relative risk for brain cancer mortality (HR = 2.55; Gonzalez et al., 2013). The theory is that both cell killing and DNA
95% CI: 1.48, 4.40), as well as modestly elevated risk for incidence repair contribute to the lower cancer risk per unit dose. Surprisingly,
of breast cancer (HR = 1.16; 95% CI: 1.02, 1.32) and melanoma (HR these studies have also shown that there is little evidence that the dose–​
= 1.30; 95% CI: 1.05, 1.61) compared to those who never performed response curve is nonlinear in the direction of a downturn in risk, even
these procedures. No elevated risk was observed for cancers of the thy- at organ doses of ≥ 60 Gy. It was previously assumed that high levels
roid, non-​melanoma skin, prostate, lung, colon-​rectum, or non-​CLL of cell killing would result in a downturn or plateau in risk at very
leukemia in workers who performed these procedures. While exposure high doses (e.g., > 5Gy organ dose; Gray, 1965). Thyroid cancer is
to radiation in the workplace is one possible explanation for these find- the only site currently where there is strong evidence of a downturn
ings, the role of chance or unmeasured confounding by non-​radiation above doses of 20 Gy to the thyroid from radiotherapy for childhood
risk factors cannot be ruled out until these results are replicated in cancer (Veiga et al., 2012). The lack of downturn in other cancer sites
more studies, and as noted above in subsequent analyses there was no has been hypothesized to be related to cellular repopulation between
relationship with dose (Rajaraman et al., 2016; Kitahara et al., 2017). fractions (Sachs and Brenner, 2005).
The first study to examine this question systematically in physicians, The type of ionizing radiation exposure also influences the risk of
did not find evidence of an increased risk of brain tumors (Linet et al., cancer. As described earlier (see section “Units of Exposure”), neu-
2017). Nuclear medicine procedures have also increased dramatically trons may be 20 times more carcinogenic per unit dose than X-​rays.
in the United States, and some radiologic technologists who routinely Most of the estimates of the relative biological effectiveness of dif-
perform these may be exposed above the recommended dose limits. ferent types of radiation exposure come from animal or other labora-
In an analysis of technologists in the USRT cohort who ever (versus tory studies, as few human studies are available where the populations
never) perform various nuclear medicine procedures, there was an are comparable other than with respect to the type of radiation. More
increased risk for squamous cell carcinoma of the skin (HR = 1.29; human studies are needed, however, especially of the RBE of neutrons
95% CI: 1.01, 1.66) with ever performing diagnostic radionuclide pro- and protons due to the increasing use of proton therapy to treat cancer
cedures and of breast cancer (HR = 2.68; 95% CI: 1.10, 6.51) with ever patients. Certain types of proton therapy systems expose the patient to
performing other radionuclide therapy procedures (excluding brachy- a whole-​body scatter dose of neutrons, and the long-​term risks from
therapy and radioactive iodine) (Kitahara et al., 2015). Increasing risks this are uncertain.
were also observed with greater frequency of performing these pro- The impact of age at radiation exposure has been studied exten-
cedures, particularly before 1980. Reconstruction of individual doses sively (BEIR VII Phase 2, 2006). These studies have established that
to a group of nuclear medicine technologists is underway, and further childhood exposure to ionizing radiation results in a higher cancer risk
follow-​up is needed of the potential long-​term risks in these occupa- than adulthood exposure; the one possible exception is for lung can-
tional groups. cer (Preston et al., 2007). Some organs are especially radiosensitive in
childhood, including the red bone marrow, thyroid, breast, and brain
(Braganza et al., 2012; Hsu et al., 2013; Preston et al., 2002a, 2002b;
EXPOSURE AND HOST PARAMETERS Veiga et al., 2016). In contrast, the thyroid and brain seem to have very
THAT INFLUENCE RISK low radiation sensitivity if exposure is in adulthood (Braganza et al.,
2012; Preston et al., 2007; see also Chapter 44 in this volume).
The focus of many of the current radiation epidemiology studies is Long-​term follow-​up of more than 50 years of the Japanese atomic
the understanding of the effect of dose protraction or fractionation. bomb survivors and other studies, including the Canadian fluoros-
This has important practical implications for predicting cancer risk copy cohort (Howe and McLaughlin, 1996), have demonstrated that
from occupational, environmental, and diagnostic medical exposures, the cancer risk from ionizing radiation exposure remains elevated for
which are typically received as low and fractionated (or protracted) decades, probably for the rest of a person’s lifetime; that is, unlike
doses and also contribute most to the collective dose received in the smoking, the risk never returns to baseline (BEIR VII Phase 2, 2006).
general population. Theoretically, the risk per unit dose could be This is consistent with the notion of ionizing radiation being an initia-
lower if the dose is protracted or fractionated because of DNA repair tor, as well as potentially a cancer promoter. The risk does decline,
mechanisms. The results from low-​dose epidemiological studies have however, with time since exposure.
been mixed. However, it is difficult to find exposed populations that In many studies, women have higher cancer risks than men follow-
are identical in all ways other than the manner in which they received ing radiation exposure. For example, the ERR/​Gy in the LSS for all
their radiation exposure. The most authoritative risk assessments for solid cancers (exposure at age 30, attained age 70) is 0.58 for women
240

240 PART III:  THE CAUSES OF CANCER


and 0.35 for men, and differences remain even after removal of sex-​ to direct DNA damage, radiation also causes the formation of reac-
specific cancers (Preston et al., 2007). However, it is uncertain whether tive oxygen species (ROS), which are free radicals involving oxy-
this reflects true biological differences in radiosensitivity, interac- gen, and reactive nitric oxide species (RNOS), which induce stress
tions with other cancer risk factors, or differences in background responses, inflammation, and release of cytokines, growth factors,
cancer rates. and chemokines (Barnett et al., 2009).
There have been a large number of studies of the joint effect of In normal circumstances, when cells detect DNA damage caused
smoking and radiation exposure. The findings are not consistent, by free radicals, they respond by undergoing cell cycle arrest
varying from a complicated joint effect model in the Japanese atomic in order to repair the damage, and the majority of the damage is
bomb survivors to multiplicative in several studies of radiotherapy repaired. When this damage cannot be repaired, the cell under-
and second cancers. Studies of high-​LET radiation, including those goes death via necrosis or apoptosis within a few cell divisions.
of uranium miners exposed to radon progeny and the Mayak pluto- Incorrectly repaired or unrepaired DNA damage, on the other hand,
nium cohort, have found joint effects that appear to be super-​additive can lead to mutation and genomic instability, and cancer can occur
but sub-​multiplicative (Gilbert et al., 2013). Reproductive factors and after many years.
breast cancer risk have also been evaluated in several radiation-​exposed Until recently, research in radiobiology focused mainly on
populations, but there is little consistency with findings regarding the pathways related to the repair of DNA damage. Although the
joint effects (Ronckers et al., 2005). Studies of genetic susceptibility role of repair pathways remains a key area of investigation, there
also have not produced clear-​cut findings, even where there is an obvi- is increasing interest in understanding the mechanisms involved
ous potential mechanism such as DNA repair and BRCA mutation (see in radiation-​induced inflammatory and stress responses to ROS.
discussion of radiosensitivity later in this chapter). Given how well the These mechanisms are particularly important for understanding the
risks from ionizing radiation are known, these difficulties with joint effects of radiation on cells that are not directly irradiated (West and
effects analyses are a reminder about how difficult these questions are Barnett, 2011).
to address via epidemiological studies.

Types of Evidence
MECHANISMS OF CARCINOGENESIS
It is becoming increasingly apparent that the assessment of disease risk
Each of the most common types of ionizing radiation exposure in following radiation exposure is a complex process that needs to extend
humans (fractionated high-​dose exposures such as those experienced beyond traditional assessment by epidemiologic methods to incor-
by patients undergoing cancer radiotherapy; acute low-​ moderate porate biological evaluation of differences in susceptibility between
doses, such as those experienced by many Japanese atomic bomb individuals. Biological changes leading to cancer can be studied at dif-
survivors; chronic low-​dose exposures received by radiation work- ferent levels, and using various endpoints. At the cellular level, radio-
ers; and fractionated low-​dose exposures from diagnostic medical sensitivity measures the degree of response of a cell to radiation, with
examinations) has been associated with increased risk of cancer. The a stronger response indicating higher sensitivity. Typical cellular end-
development of cancer is characterized by distinct biological abilities points include phenomena such as cell killing or measurable chromo-
acquired during the multistep development of human tumors. These somal damage. Radiation response can also be observed at the tissue
include sustained proliferative signaling, evasion of growth suppres- level: tissues can be more or less sensitive to radiation. Finally, differ-
sors, resistance of cell death, induction of angiogenesis, activation ences in susceptibility can be observed at the level of individuals, with
of invasion and metastasis, reprogramming of energy metabolism, some humans being less tolerant of the effects of a fixed amount of
and evasion of immune destruction (Hanahan and Weinberg, 2011). radiation exposure than others (Advisory Group on Ionising Radiation
These hallmarks of cancer are driven by genomic instability (which [AGIR], 2013).
generates genetic diversity leading to the acquisition of hallmark fea- The path from radiation exposure to the development of cancer can
tures) and inflammation (which fosters multiple hallmark functions). be represented as a theoretical continuum of effects that includes exter-
Cellular damage from exposure to ionizing radiation occurs when nal dose, internal dose, early biological effects, and finally, disease
radiation is absorbed by biological tissue and interacts either directly outcome. Potential exposure to other genotoxic agents can also play a
or indirectly with atoms of critical targets. As radiation moves through role. Measures of different biological changes can be used to quantify
the tissue, energy is deposited, causing ionization (ejection of an elec- various points within the continuum between external radiation dose
tron from an atom) along the track, with some clustering at the ends. and final outcome of interest. A biological marker (or biomarker) may
Direct action results when the radiation energy itself causes ionization be suitable as a measure of dose, or as a measure of early or late effect,
of the critical target. This is the dominant process for radiation with or in some scenarios may serve as a measure of both internal dose and
high linear energy transfer (LET), such as neutrons or alpha-​particles effect, whereby some measure (e.g., induction of chromosome aberra-
(Hall and Giaccia, 2006). Indirect action occurs when radiation inter- tions) could tell us not only about the likely dose of ionizing radiation
acts with other atoms or molecules in the cell, such as water, to pro- experienced by an individual, but could also be used to predict risk of
duce highly reactive free radicals that can break chemical bonds and disease outcome(s) of interest (Pernot et al., 2012; see also Chapter 6
damage the critical target, initiating the chain of biological events that in this volume).
eventually leads to cancer. Several factors need to be considered in the selection of appropri-
The carcinogenic effects of ionizing radiation generally result from ate biomarkers for characterization of radiation risk. First, natural
various types of damage to DNA, including damage to nucleotide and meaningful variation of the proposed marker should exist in the
bases, single-​strand breaks (SSBs), double-​strand breaks (DSBs), human population. A test for a reliable biomarker should be sensi-
and DNA crosslinks, often forming clustered/​multiple damage sites tive and specific, and highly reproducible among different labora-
which represent two or more lesions formed within one or two heli- tories. For use in human populations, the ideal biomarker should
cal turns of DNA (BEIR VII Phase 2, 2006). Base damage is repaired be easily obtained with minimal discomfort or risk to the patient.
by mechanisms that involve excision and replacement of individual Given that levels of a biomarker often change over time and can
damaged bases (base-​excision repair) or larger oligonucleotide frag- differ between cells or tissues, details such as the appropriate timing
ments (nucleotide excision repair). The repair of SSBs uses a similar of obtaining the sample from which the biomarker is to be mea-
process to base-​excision repair. The repair of DSBs, on the other sured and the biological source are crucial. Finally, a rapid readout
hand, usually involves several processes. In some instances, DSBs of results is preferable. Although a single test may not possess all of
are rejoined end to end in a process called non-​homologous end these characteristics (e.g., a highly reliable test may not have a rapid
joining. An alternative pathway for DSB repair is the homologous readout), and some characteristics may be more important than oth-
recombination process, in which the broken strand is repaired by ers depending on the intended use of the biomarker, it is useful to
crossing over with an adjacent identical DNA sequence. In addition keep all in mind during biomarker selection.
 241

Ionizing Radiation 241


Cellular Assays to Measure Biological Changes responses for patients with cancers of the colon, upper aerodigestive
in Humans tract, and lung from those of the control population, the sensitivity pro-
The first human cell studies of radiosensitivity were clonogenic assays file of patients with breast cancer was similar to that of the control pop-
performed on fibroblasts cultured from the skin of individuals with ulation, suggesting that mutagen sensitivity response differs between
severe radiation toxicity. In these assays, different levels of radiation tissues (Hsu et al., 1989).
are applied to samples of cells that are then plated in a tissue culture More recent assays for the detection of DNA double-​strand breaks
vessel and allowed to grow, and the resulting colonies are fixed, stained, quantify the levels of nuclear foci of DNA damaged proteins, includ-
and counted. At the conclusion of the experiment, the percentage of ing the gamma-​H2AX and 53BP1 assays, which complement more
cells surviving at each dose is measured, and a cell survival curve is traditional DNA damage/​repair assays, such as pulsed field gel elec-
plotted to graphically represent the dose of ionizing radiation versus trophoretic assessment of DNA double strand breaks and comet assays
survival (Franken et al., 2006). These initial studies indicated extreme (AGIR, 2013). The gamma-​H2AX assay uses immunocytochemistry
radiosensitivity in individuals with severe radiation toxicity (Arlett to visualize foci of phosphorylated histone 2AX (H2AX), which accu-
and Priestley, 1983; Smith et al., 1980; Woods et al., 1988). Further mulates at the site of double-​strand break damage during activation of
studies demonstrated that fibroblast sensitivity significantly differed the DNA damage response signaling pathway (Nikolova et al., 2014).
between cells cultured from individuals with and without known Studies using these newer assays indicate an approximately 1.5-​fold
genetic syndromes associated with radiosensitivity, such as patients reduction in the repair efficiency of strand breaks among AT heterozy-
with ataxia telangectasia (AT) or retinoblastoma (RB) (Deschavanne gotes and RB family members versus phenotypically normal individu-
et  al., 1986). The variation in sensitivity between normal cells and als (Sigurdson and Stram, 2012).
cells with mutations characterized by defects in repair mechanisms Apart from DNA repair assays, other techniques in development
seems to be greater when doses are delivered as chronic low dose-​rate include the ability of fibroblasts to undergo radiation-​induced differ-
exposures versus doses delivered at high dose rates (Kato et al., 2009). entiation, telomere length assays, and combinations of various cellular
Although these observations suggested the possibility of using cellular assays (AGIR, 2013), as well as measuring the expression of markers
radiosensitivity to predict a patient’s probable reaction to radiother- of inflammation (e.g., plasma transforming growth factor [TGF]-​β1) in
apy, studies have not indicated any clear and consistent associations serum or plasma. Given the rapidly evolving nature of this field, work
between cellular radiosensitivity and reaction to radiation exposure in in this research area is likely to undergo considerable evolution in the
patients undergoing radiotherapy for cancers of the breast (Djuzenova next few years.
et al., 2006; Peacock et al., 2000), cervix (West et al., 2001), or head
and neck (Geara et  al., 1993; Rudat et  al., 1999). Furthermore, the
relationship between acute toxicity after radiotherapy and late effects Evidence for Differing Sensitivity to Radiation
(e.g., cancer) remains unclear.
Exposure from Epidemiological Studies
Most cellular assays of cancer susceptibility in human populations
to date have focused on DNA repair capacity. These assays generally As illustrated in Table 13–​3, data from the LSS have clearly indicated
compare DNA damage induced in circulating lymphocytes from can- that different organs and tissues have different sensitivities to the
cer patients to circulating lymphocytes from cancer-​free controls, with effects of radiation exposure. In atomic bomb survivors, for example,
the subsequent quantification of repair in both groups. Ionizing radia- excess relative risks due to radiation are particularly notable for can-
tion (or some other agent, such as the radiomimetic chemical bleomy- cers of the esophagus, colon, lung, breast, ovary, and bladder (Preston
cin) is applied to the cell culture, a specified period of time is allowed et al., 2007).
to pass for repair to occur, and the remaining damage is measured in a The idea that some people are more sensitive to the effects of radia-
variety of ways (e.g., unrepaired SSBs, DSBs, or the incorporation of tion has been accepted for several decades. Individuals with certain rare
a radioisotope). Cellular assays of DNA repair can be broadly grouped hereditary disorders (e.g., ataxia telangiectasia and Nijmegen break-
into the following categories (Berwick and Vineis, 2000): age syndrome) are known to be particularly sensitive to the effects of
radiation (Taalman et al., 1983; Taylor et al., 1975). However, these
1. Tests based on DNA damage to cells induced by chemical or physi-
cancer susceptibility syndromes affect only a small proportion of the
cal agents (e.g., bleomycin or ionizing radiation), such as the muta-
general population. More relevant for the majority of the population is
gen sensitivity assay, the G2-​ radiation assay, the micronucleus
the theory that some part of the genetic contribution defining radiation
assay, and the comet assay.
susceptibility is likely to follow a polygenic model, whereby inheri-
2. Indirect tests of DNA repair, such as assays of unscheduled DNA
tance of several low-​penetrance risk alleles can lead to elevated risk of
synthesis, activity of repair enzymes, or measures of gamma-​
cancer. This theory (the “common-​variant-​common-​disease” model)
H2AX or 53BP1 foci. Levels of enzyme activity or DNA synthesis
is supported by the fact that multiple genetic pathways have been
are measured in radiolabeled cells, usually by scintillation counting
implicated in radiosensitivity, including DNA damage repair, radiation
or radiography. Gamma-​H2AX or 53BP1 foci are assessed using
fibrogenesis, oxidative stress, and endothelial cell damage (Barnett
fluorescence microscopy.
et al., 2009).
3. Tests based on more direct measures of repair kinetics (e.g., plasmid
Most studies of the effects of ionizing radiation in humans have
host cell reactivation assay). Separate sets of fresh or cryopreserved
focused on the DNA repair pathway. A  review of population-​based
lymphocytes are transfected with both damaged and undamaged
studies conducted prior to 2000 found that the combined evidence for
plasmids incorporating the chloramphenicol acetyltransferase (cat)
tests based on DNA damage and direct/​indirect tests of DNA repair
or Luciferase gene as a marker. Repair can then be measured as a
indicated mainly positive associations between DNA repair capac-
rate, such as the amount of radiation or fluorescence at a given point
ity and occurrence of cancer (Berwick and Vineis, 2000; Vineis and
in time.
Perera, 2000). However, these studies faced a number of limitations,
Early assays of DNA repair measured chromatid breaks and gaps fol- including small sample size, the use of the case-​control study design
lowing the administration of bleomycin (Hsu et al., 1989) or gamma-​ (which cannot address the question of temporality between exposure
radiation in the G2 phase of the cell cycle (Sigurdson and Stram, and disease), the use of “convenience controls,” the possibility of con-
2012) to assess repair efficiency following exposure to various muta- founding by factors other than ionizing radiation that could affect DNA
gens in a range of cell types (Berwick and Vineis, 2000; Li et al., 2009; repair, and use of cells different from the target organ. Subsequently,
Wu et al., 2007). Studies using the G2 assay have demonstrated greater a number of population-​based epidemiological studies have examined
sensitivity to radiation in first-​degree relatives of cancer patients com- susceptibility to radiation-​related risk of cancer with respect to genes
pared to non-​cancer controls (Kato et  al., 2009). Tests of genotoxic in the DNA repair and other relevant biological pathways. Results
response to bleomycin in lymphocytes blood cultures indicate a wide from the “candidate-​SNP” approach, which assumes prior knowledge
variance in normal individuals (0.20 to more than 2.00 average chro- of one or more functional single nucleotide polymorphisms (SNPs),
matid breaks per cell). While one study showed distinctly different have not been convincingly replicated to date (Bhatti et  al., 2008,
24

242 PART III:  THE CAUSES OF CANCER


2010a; Bondy et al., 2001; Hu et al., 2002; Liu et al., 2010; Rajaraman be histologically different from the primary cancer, and must have a
et al., 2008; Sigurdson et al., 2007, 2009). latency period of at least 5 years (Cahan et al., 1948). While satisfying
An alternate approach to examine genetic susceptibility is to quan- these criteria is likely to result in an increased probability that the tumor
tify cancer risk in groups of high-​risk individuals. Most (Andrieu is radiation-​related, it does not guarantee that it was radiation-​induced.
et al., 2006; Gronwald et al., 2008; Lecarpentier et al., 2011), but not Early work in search of a radiation signature for sarcoma mainly used
all (John et al., 2013) interview-​based studies of diagnostic chest X-​ conventional cytogenetic analysis to identify large-​scale chromosomal
rays in BRCA1 and BRCA2 mutation carriers have reported a slightly abnormalities. These studies were limited by small sample sizes and
elevated risk of breast cancer in at least one subgroup of X-​ray expo- lack of a comparison group of non-​radiation-​induced tumors. Later
sure (e.g., exposure at younger age). On the other hand, the association studies using polymerase chain reaction followed by direct sequencing
has generally been null for mammograms (Giannakeas et  al., 2014; were able to examine mutations in specific genes such as TP53, but did
Goldfrank et al., 2006; Narod et al., 2006). Breast doses from chest X-​ not address the probability of multi-​gene involvement, and potential
rays are generally very low, and recall bias cannot be ruled out. gene–​gene interactions (Berrington de Gonzalez et al., 2012). A more
Studies have also reported some indication of increased risk follow- recent study using microarray analysis to identify a signature of 135
ing radiotherapy in individuals carrying rare mutants in genes in DNA genes from a learning/​ training set of 12  “radiation-​ induced” and
damage repair pathways. A  case-​only study of contralateral breast 12 “sporadic sarcomas” was able to discriminate “radiation-​induced”
cancer (CBC) reported increased prevalence of pathogenic germline from “sporadic sarcomas” in an independent set of 36 sarcomas with
mutations in DNA repair genes BRCA1, BRCA2, CHEK2, and ATM 96% sensitivity and 62% specificity, but this study remains to be inde-
in women with CBC following radiotherapy for their primary breast pendently replicated (Hadj-​Hamou et al., 2011).
cancer compared with women with CBC who were not irradiated for For thyroid tumors, several lines of evidence indicate that radiation-​
their first breast cancer (Broeks et  al., 2007). A  larger case-​control related tumors may present differently than sporadic tumors. A high
study of women with CBC (cases) compared to matched controls with frequency of RET/​PTC (RET proto-​oncogene/​papillary thyroid carci-
unilateral breast cancer reported evidence of greater CBC risk follow- noma) gene rearrangements has been observed in papillary thyroid car-
ing radiation among women who carried rare ATM missense variants cinomas (PTCs) of children exposed to radioactive fallout in Belarus
(Bernstein et al., 2010) and individuals who carried a rare haplotype after the Chernobyl accident (Fugazzola et al., 1995; Ito et al., 1994;
in RAD50 (Brooks et  al., 2012)  than in unexposed carriers, but no Leeman-​Neill et  al., 2013)  and in thyroid tumors following external
increase in risk with radiation dose in carriers of other rare variants, radiotherapy (Bounacer et al., 1997). The relationship between these
including BRCA1/​2 mutations (Bernstein et al., 2013). gene rearrangements and radiation dose, however, remains unclear: no
The interpretation of these genetic findings (candidate SNP, radia- significant association was observed between RET/​PTC activation
tion exposure in high-​risk genetic populations, or rare mutations in and individual I-​131 doses in post-​Chernobyl cancers occurring in the
patients exposed to radiotherapy) has been complicated by the fact that Russian Federation (Tuttle et al., 2008); higher doses were associated
many of these studies are subject to one or more of the following: lack with higher prevalence of RET/​PTC rearrangements in PTCs of atomic
of replication in an independent population; exposure based on self-​ bomb survivors (Hamatani et  al., 2008); and recent data from post-​
report; lack of a consistent dose–​response association; subgroup find- Chernobyl thyroid cancer in Ukraine indicate a non-​monotonic rela-
ings that could be due to chance; and overlap of study populations. tionship between I-​131 dose and RET/​PTC or PAX8/​PPARγ-​positive
While earlier genetic studies focused on a handful of candidate tumors and dose, with increased risk at low-​to moderate doses and
genes, the development of technologies that can rapidly analyze thou- decreased risk at high doses (Leeman-​Neill et al., 2013).
sands of genetic markers at relatively low cost, along with the mapping A genome-​ wide comparison of somatic copy number alterna-
of linkage disequilibrium between common SNPs across the genome tions (CNAs) between age-​and residence-​ matched exposed and
(International HapMap et  al., 2010)  and the definition of functional non-​exposed PTC patients from Chernobyl observed a copy number
elements critical for regulation and genomic stability (Encode Project gain of chromosome bands 7q11.22-​11.23 in a higher proportion of
Consortium, 2012), have allowed us to comprehensively examine the radiation-​exposed versus unexposed individuals, with specific m-​RNA
approximately 25,000 coding genes and associated functional ele- overexpression of the CLIP2 gene located within this band (Hess
ments. The genome-​wide association study (GWAS) approach has et al., 2011). While a positive dose-​response relationship was observed
successfully identified hundreds of risk loci in germline DNA for vari- between I-​131 dose and binary CLIP2 typing for young PTC cases
ous cancers (Chung and Chanock, 2011). However, the assessment of (age at exposure less than 5 years), this relationship was not observed
gene–​environment interaction for most environmental carcinogens, for those exposed at age ≥5  years (Selmansberger et  al., 2015), and
including radiation, has remained elusive. A GWAS of 100 Hodgkin these results have yet to be confirmed in other populations.
lymphoma survivors treated with radiotherapy identified two loci in As with studies of germline DNA, advances in DNA sequencing
the PRDM1 gene potentially associated with increased risk of sec- technology in the last decade have yielded many insights into somatic
ondary malignancy (Best et  al., 2011), but these loci have yet to be mutations from tumor tissue, largely driven by findings from The
consistently confirmed. Genome-​wide association studies have also Cancer Genome Atlas (TCGA) and the International Cancer Genome
been completed in adult women with contralateral breast cancer in Consortium (ICGC). These projects characterize the genome, as well
the WECARE study (Bernstein et  al., 2004)  and in individuals with as various aspects of the transcriptome and epigenome, to give a
subsequent malignancies in the Childhood Cancer Survivor Study fuller understanding of how genes contribute to tumorigenesis. Over
(Robison et  al., 2009), both studies having detailed radiation doses 30 distinct human tumor types have been analyzed to date through
from radiotherapy. Results from these additional GWAS are expected large-​ scale genome sequencing and integrated multidimensional
to be published shortly. For additional details on genetic susceptibilil- analyses, yielding insights into both individual cancer types and
ity to second cancers following radiotherapy, see Chapter 60. across cancers, particularly with respect to the accurate molecular
classification of tumors (Chin et al., 2011). While most of these dis-
coveries have focused on the genome rather than associated environ-
Radiation Signatures mental factors, a recent paper examining 4,938,362 mutations from
The pursuit of a “molecular signature” that would be able to discrim- 7042 cancers identified strong mutational signatures in tumor tissue
inate radiation-​induced tumors from “sporadic” tumors has obvious marking exposure to tobacco carcinogens and ultraviolet (UV) irra-
and important implications for radiation protection. The search for ion- diation (Alexandrov et al., 2013). The tobacco signal was most evi-
izing radiation signatures started several decades ago, particularly in dent in cancers that are known to be tobacco-​related (lung, head and
the context of sarcomas after radiotherapy and thyroid cancers follow- neck, and liver); and the UV irradiation signal was observed mainly
ing head/​neck radiotherapy and after environmental radiation expo- in malignant melanoma and squamous carcinoma of the head and
sure. For sarcomas, most of the studies of molecular signatures use neck. A recent study identified potential radiation-​associated muta-
some modification of the 1948 Cahan criteria for classifying tumors tional signatures in a small number (n = 17) of radiation-​associ-
as radiation-​induced: the tumor must be in the irradiated field, must ated second malignancies (Behjati, 2016). Similar studies to detect
 243

Ionizing Radiation 243


a radiation signature, whereby tumors caused by ionizing radiation Overall, when all sources of radiation were considered in the UK
exposure could be discriminated from their sporadic counterparts, population, Darby and Parkin estimated that about 2% of cancers could
are currently underway in populations environmentally exposed to be attributable to radiation (Parkin and Darby, 2011). Background
high doses of ionizing radiation. The best target populations for these radiation/​
radon and diagnostic X-​ rays contributed approximately
studies are those with a high proportion of cancers likely to be attrib- equally, followed by radiotherapy.
utable to radiation and well-​characterized doses. Results of these
studies are expected over the next few years.
OPPORTUNITIES FOR PREVENTION

ESTIMATED ATTRIBUTABLE FRACTION There are a number of approaches to reduce unnecessary radiation
exposure to the general population. For example, residential radon
As ionizing radiation is an established carcinogen, it is appropriate exposure can be reduced by a variety of relatively simple and cheap
to estimate the potential fraction of cancers that may be attributable remediation strategies such as improved ventilation, which should
to radiation from various sources, including medical, natural back- also be implemented in schools and workplaces. Diagnostic medical
ground, and nuclear accidents in exposed populations. Some estimates radiation exposure should be limited to clinically justified procedures
come directly from the study populations, such as the LSS of the where the clinical decision-​making depends on the outcome of the test.
Japanese atomic bomb survivors. For low-​dose exposures such as resi- In addition, doses should be kept as low as reasonably possible. There
dential radon or diagnostic radiation exposure, attributable risks can is increased awareness about dose levels from diagnostic procedures,
be estimated, under certain assumptions, using indirect risk projection and new technologies can monitor or automatically control dose, such
methods. Because radiation has been shown to increase cancer risk for as automated exposure control for CT scans. Treatment-​planning sys-
at least 50 years after exposure (Preston et al., 2007), attributable risk tems for radiotherapy aim to maximize tumor control and minimize
estimates should be based on studies with very long-​term follow-​up or dose to the surrounding normal tissue. This should reduce the amount
life-​table methods, with adjustment for competing risks. of normal tissue exposed to very high doses. Many of these systems,
such as IMRT, result in increased whole-​body exposure to low levels
of radiation due to scatter and the number of monitor units involved. It
Environmental Radiation Exposure remains uncertain how this impacts the overall risk–​benefit profile of
newer technologies.
Indirect risk modeling estimates suggest that 5% of leukemia in
The current exceptions are flight crew, who are currently unmoni-
England at any age, and about 15% of childhood leukemias, could
tored, and certain medical workers who perform fluoroscopically
be related to background radiation (Kendall et al., 2011). Residential
guided procedures or who handle radionuclides. The length and com-
radon is estimated to be responsible for about 3% of lung cancers in
plexity of these procedures and the need for close proximity to the
the United Kingdom (Parkin and Darby, 2011). The Chernobyl acci-
beam and patient results in radiation exposure to various exposed parts
dent is estimated to be responsible for 3%–​4% of cancer deaths in the
of the operator’s body, including the hands, lens of the eye, and brain.
most highly exposed groups: cleanup workers, residents, and evacuees
Many of the physicians who perform these procedures have limited
(WHO, 2006). Radioactive fallout in Europe, by comparison, is esti-
training in radiation protection. Heavy protection aprons, gloves, or
mated to be related to only 0.01% of cancers (Cardis et al., 2006).
glasses can make it more difficult to perform the procedures, and there
After 60 years of follow-​up in the LSS of the Japanese atomic bomb
is a need for a careful balance between radiation protection and com-
survivors, it is estimated that about 10% of the solid cancers were
fort and clinical performance. The development of personal protec-
related to the radiation exposure (Preston et al., 2007).
tive technology that does not impede comfort or clinical performance
could help reduce exposures among medical workers. Badge monitors
need to be worn routinely to better monitor the cumulative exposures
Medical Radiation to these physicians.
As described earlier, there are relatively few studies of the cancer risks
from diagnostic radiation exposure due to the low doses per procedure.
However, diagnostic radiation is the largest man-​made source of radia- FUTURE RESEARCH DIRECTIONS
tion, and there are concerns about whether doses are always optimized
and procedures always justified. Indirect modeling approaches esti- As one of the most extensively studied carcinogens, ionizing radiation
mate that in more developed countries between 0.5% (in the UK) and is an ideal candidate for cutting-​edge cancer epidemiology, including
3% (in Japan) of cancers could be attributable to diagnostic medical searching for molecular signatures, risk-​projection modeling, uncer-
radiation (Berrington de Gonzalez and Darby, 2004). With the dra- tainty analyses, and comprehensive assessment of critical exposure
matic increase in CT scans in the United States, the attributable risk periods.
could increase from 1% to 3% if current levels continue (Berrington de Currently, most of the exposures to the general population are low-​
Gonzalez et al., 2009). Not all of these cancers are avoidable because level doses (effective doses < 10 mSv) from diagnostic medical expo-
many of these procedures have important benefits that need to be bal- sures or background radiation. The magnitude of the cancer risks from
anced against the potential risks. these low-​dose exposures, and protracted or fractionated exposures, is
The proportion of second primary cancers that may be related to more uncertain than that from higher, acute doses that have been stud-
the radiotherapy for the first cancer was estimated in the United States ied for many decades in the Japanese atomic bomb survivors. Sample
from the SEER cancer registries as 8% overall for adulthood cancer size, measurement error, and the length of follow-​up needed to evalu-
routinely treated with radiation (Berrington de Gonzalez et al., 2011). ate cancer risks from low doses make it a difficult problem to study
Estimates for the United Kingdom are similar (6% for males and 8% using traditional epidemiological methods. The most promising recent
for females) (Parkin and Darby, 2011). The attributable risk is likely studies have used electronic record linkage with a retrospective study
higher for some childhood and young adulthood cancers with good design, such as the UK natural background radiation case-​control study
survival such as Hodgkin lymphoma, as children are known to be (Kendall et al., 2013), pediatric CT studies (Pearce et al., 2012), and
more radiosensitve, and high relative risks of subsequent cancers have the pooled nuclear workers studies (Schubauer-​Berigan et al., 2015b).
been reported. Estimates for all Hodgkin lymphoma survivors in the This design facilitates large-​ scale populations with individualized
United Kingdom suggest that 16% of second cancers for males could dose estimates. In the next decade there will be new publications in all
be related to radiotherapy, and 19% for females (because of the high of these areas from large-​scale efforts including EPI-​CT, INWORKS,
risk of second breast cancer) (Parkin and Darby, 2011). For testicular the Million Workers Study, the USRT cohort, and the UK natural back-
cancer the estimate was 11%, and for cervical cancer 17%; both occur ground radiation study. The key limitation in many of these studies,
at younger ages, and pelvic radiation exposes a number of organs. however, is the lack of information on confounding factors, such as
24

244 PART III:  THE CAUSES OF CANCER


smoking in the nuclear worker studies and underlying medical condi- study in the US SEER cancer registries. Lancet Oncol, 12(4), 353–​360.
tions in the CT scan studies. Traditional epidemiology, therefore, may PMCID: PMC3086738.
not be able to provide a definitive answer to the question of the risks Berrington de Gonzalez A, and Darby S. 2004. Risk of cancer from diagnostic
from low-​dose or protracted radiation exposure. X-​rays: estimates for the UK and 14 other countries. Lancet, 363(9406),
345–​351. PMID: 15070562.
New molecular techniques could, however, provide new insights. In Berrington de Gonzalez A, Gilbert E, Curtis R, et al. 2013. Second solid can-
particular, the search for “signatures” that can identify radiation expo- cers after radiation therapy:  a systematic review of the epidemiologic
sure as a causal factor for a particular tumor could technically provide a studies of the radiation dose-​response relationship. Int J Radiat Oncol
definitive answer to the low-​dose question if the signature were identi- Biol Phys, 86(2), 224–​233. PMCID: PMC3816386.
fied and then shown to be present in tumors after low-​dose exposure Berrington de Gonzalez A, Kutsenko A, and Rajaraman P. 2012.
with evidence of no other clear cause of cancer. New epidemiologic Sarcoma risk after radiation exposure. Clin Sarcoma Res, 2(1), 18.
studies of ionizing radiation and cancer will also likely continue to fea- PMCID: PMC3507855.
ture a search for common genetic markers that can identify individuals Berrington de Gonzalez A, Mahesh M, Kim KP, et al. 2009. Projected cancer
susceptible to radiation risk effects. These studies still face many chal- risks from computed tomographic scans performed in the United States in
2007. Arch Intern Med, 169(22), 2071–​2077. PMID: 20008689.
lenges, including identifying a population with a high attributable risk, Berrington de Gonzalez A, Salotti JA, McHugh K, et  al. 2016. Relationship
and high-​quality biospesimens, well-​characterized radiation exposure, between paediatric CT scans and subsequent risk of leukaemia and brain
and meaningful replication sets. The thyroid tumors from the Chernobyl tumours: assessment of the impact of underlying conditions. Br J Cancer,
Tissue Bank satisfy many of these requirements, and the first sequenc- 114(4), 388–​394. PMCID: PMC4815765.
ing study is currently in progress. Other populations of interest include Berwick M, and Vineis P. 2000. Markers of DNA repair and susceptibility to
the Japanese atomic bomb survivors and second cancers that occur in or cancer in humans: an epidemiologic review. J Natl Cancer Inst, 92(11),
near the radiation field. The integrated characterization of germline and 874–​897. PMID: 10841823.
somatic alterations as genotyping and analysis methods evolve rapidly Best T, Li D, Skol AD, et al. 2011. Variants at 6q21 implicate PRDM1 in the
in the next decade could be a promising avenue of research. etiology of therapy-​induced second malignancies after Hodgkin’s lym-
phoma. Nat Med, 17(8), 941–​943. PMCID: PMC3229923.
References Bhatti P, Doody MM, Rajaraman P, et al. 2010a. Novel breast cancer risk alleles
and interaction with ionizing radiation among U.S. radiologic technolo-
Advisory Group on Ionising Radiation (AGIR). 2011. Risk of Solid Cancers gists. Radiat Res, 173(2), 214–​224. PMID: 20095854.
Following Radiation Exposure:  Estimates for the UK Population. Bhatti P, Struewing JP, Alexander BH, et  al. 2008. Polymorphisms in DNA
Report of the Independent Advisory Group on Ionising Radiation. repair genes, ionizing radiation exposure and risk of breast can-
London: Health Protection Agency. cer in U.S. Radiologic technologists. Int J Cancer, 122(1), 177–​182.
AGIR. 2013. Human Radiosensitivity. Report of the Independent Advisory PMID: 17764108.
Group on Ionising Radiation. London: Health Protection Agency. Bhatti P, Veiga LH, Ronckers CM et  al. 2010b. Risk of second primary thy-
Alexandrov LB, Nik-​Zainal S, Wedge DC, et  al. 2013. Signatures of muta- roid cancer after radiotherapy for a childhood cancer in a large cohort
tional processes in human cancer. Nature, 500(7463), 415–​ 421. study: an update from the childhood cancer survivor study. Radiat Res,
PMCID: 3776390. 174(6), 741–​752. PMCID: PMC3080023.
Andrieu N, Easton DF, Chang-​Claude J, et al. 2006. Effect of chest X-​rays on Black D. 1984. Investigation of the Possible Increased Incidence of Cancer
the risk of breast cancer among BRCA1/​2 mutation carriers in the inter- in West Cumbria. Report of the Independent Advisory Group.
national BRCA1/​2 carrier cohort study:  a report from the EMBRACE, London: HMSO.
GENEPSO, GEO-​HEBON, and IBCCS Collaborators’ Group. J Clin Boice JD, Jr. 2006. Ionizing radiation. In:  Schottenfeld D, and Fraumeni JF,
Oncol, 24(21), 3361–​3366. PMID: 16801631. Jr. (Eds.), Cancer Epidemiology and Prevention (3rd ed., pp. 259–​293).
Arlett CF, and Priestley A. 1983. Defective recovery from potentially lethal New York: Oxford University Press.
damage in some human fibroblast cell strains. Int J Radiat Biol Relat Stud Boice JD, Jr, Leggett RW, Ellis ED, et al. 2006. A comprehensive dose recon-
Phys Chem Med, 43(2), 157–​167. PMID: 6600729. struction methodology for former rocketdyne/​atomics international radia-
Ashmore JP, Krewski D, Zielinski JM, et al. 1998. First analysis of mortality tion workers. Health Phys, 90(5), 409–​430. PMID: 16607174.
and occupational radiation exposure based on the National Dose Registry Bondy ML, Wang LE, El-​Zein R, et al. 2001. Gamma-​radiation sensitivity and
of Canada. Am J Epidemiol, 148(6), 564–​574. PMID: 9753011. risk of glioma. J Natl Cancer Inst, 93(20), 1553–​1557. PMID: 11604478.
Ashmore JP, Gentner NE, and Osborne RV. 2010. Incomplete data on the Bosch de Basea M, Pearce MS, Kesminiene A, et  al. 2015. EPI-​CT:  design,
Canadian cohort may have affected the results of the study by the challenges and epidemiological methods of an international study on
International Agency for Research on Cancer on the radiogenic cancer cancer risk after paediatric and young adult CT. J Radiol Prot, 35(3),
risk among nuclear industry workers in 15 countries. J Radiol Prot, 30(2), 611–​628. PMID: 26226081.
121–​129. PMID: 20530869. Bounacer A, Wicker R, Caillou B, et  al. 1997. High prevalence of activat-
Barnett GC, West CM, Dunning AM, et al. 2009. Normal tissue reactions to ing ret proto-​oncogene rearrangements, in thyroid tumors from patients
radiotherapy:  towards tailoring treatment dose by genotype. Nat Rev who had received external radiation. Oncogene, 15(11), 1263–​ 1273.
Cancer, 9(2), 134–​142. PMCID: 2670578. PMID: 9315093.
Beck HL, and Bennett BG. 2002. Historical overview of atmospheric nuclear Bouville A, Linet MS, Hatch M, Mabuchi K, and Simon SL. 2014. Guidelines
weapons testing and estimates of fallout in the continental United States. for exposure assessment in health risk studies following a nuclear reactor
Health Phys, 82(5), 591–​608. PMID: 12003011. accident. Environ Health Perspect, 122(1), 1–​5. PMCID: PMC3888574.
BEIR VII Phase 2. 2006. Health Risks from Exposure to Low Levels of Ionizing Bouville A, Simon SL, Miller CW, et al. 2002. Estimates of doses from global
Radiation. Washington, DC:  Board on Radiation Effects Research, fallout. Health Phys, 82(5), 690–​705. PMID: 12003019.
National Research Council. Bouville A, Toohey RE, Boice JD, Jr, et al. 2015. Dose reconstruction for the
Bennett BG. 2002. Worldwide dispersion and deposition of radionu- million worker study: status and guidelines. Health Phys, 108(2), 206–​
clides produced in atmospheric tests. Health Phys, 82(5), 644–​ 655. 220. PMCID: PMC4854640.
PMID: 12003015. Braganza MZ, Kitahara CM, Berrington de Gonzalez A, et al. 2012. Ionizing
Bernstein JL, Langholz B, Haile RW, et  al. 2004. Study design:  evaluat- radiation and the risk of brain and central nervous system tumors: a sys-
ing gene-​environment interactions in the etiology of breast cancer:  the tematic review. Neuro Oncol, 14(11), 1316–​1324. PMCID: PMC3480263.
WECARE study. Breast Cancer Res, 6(3), R199–​R214. PMCID: 400669. Brenner AV, Tronko MD, Hatch M, et al. 2011. I-​131 dose response for incident
Bernstein JL, Haile RW, Stovall M, et al. 2010. Radiation exposure, the ATM thyroid cancers in Ukraine related to the Chornobyl accident. Environ
Gene, and contralateral breast cancer in the women’s environmental can- Health Perspect, 119(7), 933–​939. PMCID: PMC3222994.
cer and radiation epidemiology study. J Natl Cancer Inst, 102(7), 475–​ Broeks A, Braaf LM, Huseinovic A, et al. 2007. Identification of women with
483. PMCID: PMC2902825. an increased risk of developing radiation-​induced breast cancer:  a case
Bernstein JL, Thomas DC, Shore RE et al. 2013. Contralateral breast cancer after only study. Breast Cancer Res, 9(2), R26. PMCID: PMC1868917.
radiotherapy among BRCA1 and BRCA2 mutation carriers: a WECARE Brooks JD, Teraoka SN, Reiner AS, et  al. 2012. Variants in activators and
study report. Eur J Cancer, 49(14), 2979–​2985. PMCID: PMC3755053. downstream targets of ATM, radiation exposure, and contralateral
Berrington de Gonzalez A, Curtis RE, Kry SF, et al. 2011. Proportion of sec- breast cancer risk in the WECARE study. Hum Mutat, 33(1), 158–​164.
ond cancers attributable to radiotherapy treatment in adults:  a cohort PMCID: PMC3240722.
 245

Ionizing Radiation 245


Burda O, Sato T, and Wissmann F. 2013. Quality factor of secondary cosmic radi- exposed to the Chernobyl nuclear accident. Cancer Res, 55(23), 5617–​
ation at flight altitudes. J Radiol Prot, 33(2), 339–​348. PMID: 23480894. 5620. PMID: 7585643.
Cahan WG, Woodard HQ, and et al. 1948. Sarcoma arising in irradiated bone; Furukawa K, Preston DL, Lonn S, et al. 2010. Radiation and smoking effects on
report of 11 cases. Cancer, 1(1), 3–​29. PMID: 18867438. lung cancer incidence among atomic bomb survivors. Radiat Res, 174(1),
Cardis E, Gilbert ES, Carpenter L, et al. 1995. Effects of low doses and low 72–​82. PMCID: PMC3857029.
dose rates of external ionizing radiation: cancer mortality among nuclear Furukawa K, Preston D, Funamoto S, et al. 2013. Long-​term trend of thyroid
industry workers in three countries. Radiat Res, 142(2), 117–​ 132. cancer risk among Japanese atomic-​bomb survivors: 60 years after expo-
PMID: 7724726. sure. Int J Cancer, 132(5), 1222–​1226. PMCID: PMC3910094.
Cardis E, Krewski D, Boniol M, et al. 2006. Estimates of the cancer burden Geara FB, Peters LJ, Ang KK, Wike JL, and Brock WA. 1993. Prospective
in Europe from radioactive fallout from the Chernobyl accident. Int J comparison of in vitro normal cell radiosensitivity and normal tissue reac-
Cancer, 119(6), 1224–​1235. PMID: 16628547. tions in radiotherapy patients. Int J Radiat Oncol Biol Phys, 27(5), 1173–​
Cardis E, Vrijheid M, Blettner M, et al. 2005. Risk of cancer after low doses 1179. PMID: 8262844.
of ionising radiation:  retrospective cohort study in 15 countries. BMJ, Giannakeas V, Lubinski J, Gronwald J, et  al. 2014. Mammography screen-
331(7508), 77. PMCID: PMC558612. ing and the risk of breast cancer in BRCA1 and BRCA2 mutation car-
Cardis E, Vrijheid M, Blettner M, et al. 2007. The 15-​Country Collaborative riers:  a prospective study. Breast Cancer Res Treat, 147(1), 113–​118.
Study of Cancer Risk among Radiation Workers in the Nuclear PMID: 25082516.
Industry: estimates of radiation-​related cancer risks. Radiat Res, 167(4), Gilbert ES, Sokolnikov ME, Preston DL, et al. 2013. Lung cancer risks from
396–​416. PMID: 17388693. plutonium: an updated analysis of data from the Mayak worker cohort.
Chang WP, Chan CC, and Wang JD. 1997. 60Co contamination in recycled Radiat Res, 179(3), 332–​342. PMCID: PMC3661277.
steel resulting in elevated civilian radiation doses: causes and challenges. Goldfrank D, Chuai S, Bernstein JL, et al. 2006. Effect of mammography on
Health Phys, 73(3), 465–​472. PMID: 9287087. breast cancer risk in women with mutations in BRCA1 or BRCA2. Cancer
Chin L, Andersen JN, and Futreal PA. 2011. Cancer genomics:  from dis- Epidemiol Biomarkers Prev, 15(11), 2311–​2313. PMID: 17119064.
covery science to personalized medicine. Nat Med, 17(3), 297–​ 303. Grant EJ, Ozasa K, Preston DL, et al. 2012. Effects of radiation and lifestyle
PMID: 21383744. factors on risks of urothelial carcinoma in the Life Span Study of atomic
Chung CC, and Chanock SJ. 2011. Current status of genome-​wide association bomb survivors. Radiat Res, 178(1), 86–​98. PMID: 22631857.
studies in cancer. Hum Genet, 130(1), 59–​78. PMID: 21678065. Grant EJ, Brenner A, Sugiyama H, et al. In press. Solid cancer incidence among
Clarke RH. 1989. Current radiation risk estimates and implications for the the Life Span Study of atomic bomb survivors: 1958–​2009. Radiat Res,
health consequences of Windscale, TMI and Chernobyl accidents. 187(5), 513–​537. PMID: 28319463.
In:  Crosbie WA, and Gittus JH (Eds.), Medical Response to Effects of Gray LH. 1965. A Symposium Considering Radiation Effects in the Cell and
Ionizing Radiation. New York: Elevier Applied Science. Possible Implications for Cancer Therapy:  A  Collection of Papers,
Claus EB, Calvocoressi L, Bondy ML, et al. 2012. Dental x-​rays and risk of Cellular Radiation Biology (pp. 8–​ 25). Baltimore, MD:  Williams &
meningioma. Cancer, 118(18), 4530–​4537. PMCID: PMC3396782. Wilkins.
Cullings HM, Fujita S, Funamoto S, et al. 2006. Dose estimation for atomic Gronwald J, Pijpe A, Byrski T, et  al. 2008. Early radiation exposures and
bomb survivor studies: its evolution and present status. Radiat Res, 166(1 BRCA1-​associated breast cancer in young women from Poland. Breast
Pt 2), 219–​254. PMID: 16808610. Cancer Res Treat, 112(3), 581–​584. PMID: 18205043.
Cullings H, Grant EJ, Egbert SD, et al. 2016. DS02R1: Improvements to atomic Hadj-​Hamou NS, Ugolin N, Ory C, et al. 2011. A transcriptome signature dis-
bomb survivors’ input data and implementation of Dosimetry System tinguished sporadic from postradiotherapy radiation-​induced sarcomas.
2002 (DS02) and resulting changes in estimated doses. Health Physics, Carcinogenesis, 32(6), 929–​934. PMCID: PMC3314285.
112(1), 56–​97. PMID: 27906788. Hall EJ, and Giaccia AJ. 2006. Radiobiology for the Radiologist.
Darby S, Hill D, Auvinen A, et  al. 2005. Radon in homes and risk of lung Philadelphia: Lippincott Williams & Wilkins.
cancer: collaborative analysis of individual data from 13 European case-​ Hammer GP, Auvinen A, De Stavola BL, et al. 2014. Mortality from cancer and
control studies. BMJ, 330(7485), 223. PMCID: PMC546066. other causes in commercial airline crews: a joint analysis of cohorts from
Davis FG, Yu KL, Preston D, et  al. 2015. Solid cancer incidence in the 10 countries. Occup Environ Med, 71(5), 313–​322. PMID: 24389960.
Techa River incidence cohort:  1956–​2007. Radiat Res, 184(1), 56–​65. Hamra GB, Richardson DB, Cardis E, et  al. 2015. Cohort profile:  The
PMID: 26121228. International Nuclear Workers Study (INWORKS). Int J Epidemiol,
Degteva MO, Vorobiova MI, Tolstykh EI, et  al. 2006. Development of an 45(3), 693–​699. PMCID: PMC4703555.
improved dose reconstruction system for the Techa River population Hanahan D, and Weinberg RA. 2011. Hallmarks of cancer: the next generation.
affected by the operation of the Mayak Production Association. Radiat Cell, 144(5), 646–​674. PMID: 21376230.
Res, 166(1 Pt 2), 255–​270. PMID: 16808612. Hart D, Wall BF, Hillier MC, and Shrimpton PC. 2010. Frequency and
Deschavanne PJ, Debieu D, Fertil B, and Malaise EP. 1986. Re-​evaluation Collective Dose for Medical and Dental X-​Ray Examinations in the UK,
of in vitro radiosensitivity of human fibroblasts of different genetic 2008. Oxfordshire, UK: Health Protection Agency (HPA).
origins. Int J Radiat Biol Relat Stud Phys Chem Med, 50(2), 279–​293. Hauptmann M, Borge Johannesen T, Gilbert ES, et al. 2016. Increased pancre-
PMID: 3488286. atic cancer risk following radiotherapy for testicular cancer. Br J Cancer,
Djuzenova CS, Muhl B, Fehn M, et al. 2006. Radiosensitivity in breast can- 115(7), 901–​908. PMID: 27599043.
cer assessed by the Comet and micronucleus assays. Br J Cancer, 94(8), Henderson TO, Rajaraman P, Stovall M, et  al. 2012. Risk factors associated
1194–​1203. PMCID: PMC2361251. with secondary sarcomas in childhood cancer survivors:  a report from
Doll R, and Wakeford R. 1997. Risk of childhood cancer from fetal irradiation. the childhood cancer survivor study. Int J Radiat Oncol Biol Phys, 84(1),
Br J Radiol, 70, 130–​139. PMID: 9135438. 224–​230. PMCID: PMC3423483.
Dores GM, Curtis RE, van Leeuwen FE, et  al. 2014. Pancreatic cancer risk Hendry JH, Simon SL, Wojcik A, et al. 2009. Human exposure to high natural
after treatment of Hodgkin lymphoma. Ann Oncol, 25(10), 2073–​2079. background radiation: what can it teach us about radiation risks? J Radiol
PMCID: PMC4176454. Prot, 29(2A), A29–​A42. PMCID: PMC4030667.
Drozdovitch V, Minenko V, Khrouch V, et al. 2013. Thyroid dose estimates for Howe GR, and McLaughlin J. 1996. Breast cancer mortality between 1950 and
a cohort of Belarusian children exposed to radiation from the Chernobyl 1987 after exposure to fractionated moderate-​dose-​rate ionizing radiation
accident. Radiat Res, 179(5), 597–​609. PMCID: PMC3682838. in the Canadian fluoroscopy cohort study and a comparison with breast
Einstein AJ. 2012. Beyond the bombs: cancer risks of low-​dose medical radia- cancer mortality in the atomic bomb survivors study. Radiat Res, 145(6),
tion. Lancet, 380(9840), 455–​457. PMCID: PMC3674023. 694–​707. PMID: 8643829.
Encode Project Consortium. 2012. An integrated encyclopedia of DNA ele- Hsu TC, Johnston DA, Cherry LM, et al. 1989. Sensitivity to genotoxic effects
ments in the human genome. Nature, 489(7414), 57–​ 74. PMCID: of bleomycin in humans: possible relationship to environmental carcino-
PMC3439153. genesis. Int J Cancer, 43(3), 403–​409. PMID: 2466800.
Eslinger PW, Napier BA, and Anspaugh LR. 2014. Representative doses to Hsu WL, Preston DL, Soda M, et al. 2013. The incidence of leukemia, lym-
members of the public from atmospheric releases of (131)I at the Mayak phoma and multiple myeloma among atomic bomb survivors:  1950–​
Production Association facilities from 1948 through 1972. J Environ 2001. Radiat Res, 179(3), 361–​382. PMCID: PMC3875218.
Radioact, 135, 44–​53. PMID: 24769389. Hu JJ, Smith TR, Miller MS, Lohman K, and Case LD. 2002. Genetic regula-
Franken NA, Rodermond HM, Stap J, Haveman J, and van Bree C. 2006. tion of ionizing radiation sensitivity and breast cancer risk. Environ Mol
Clonogenic assay of cells in vitro. Nat Protoc, 1(5), 2315–​ 2319. Mutagen, 39(2–​3), 208–​215. PMID: 11921191.
PMID: 17406473. Hunter N, Kuznetsova IS, Labutina EV, and Harrison JD. 2013. Solid cancer
Fugazzola L, Pilotti S, Pinchera A, et al. 1995. Oncogenic rearrangements of incidence other than lung, liver and bone in Mayak workers: 1948–​2004.
the RET proto-​oncogene in papillary thyroid carcinomas from children Br J Cancer, 109(7), 1989–​1996. PMCID: PMC3790189.
246

246 PART III:  THE CAUSES OF CANCER


International Atomic Energy Agency (IAEA). 1998. Dosimetric and Medical Lee C, Kim KP, Bolch WE, Moroz BE, and Folio L. 2015. NCICT: a com-
Aspects of the Radiological Accident in Goiania in 1987. IAEA-​ putational solution to estimate organ doses for pediatric and adult
TECDOC-​1109. Vienna: IAEA. patients undergoing CT scans. J Radiol Prot, 35(4), 891–​909. PMID:
IAEA. 2016. Nuclear Power Reactors in the World. Reference Data Series 2/​ 26609995.
36. Vienna: IAEA. Lee C, Pearce MS, Salotti JA, et al. 2016. Reduction in radiation doses from
Internatinal Agency for Research on Cancer (IARC). 2012. Radiation. IARC paediatric CT scans in Great Britain. Br J Radiol, 89(1060), 20150305.
Monogr Eval Carcinog Risks Hum, 100(Pt D). PMID: 26864156.
International Commission on Radiological Protection (ICRP). 2007. The 2007 Lee T, Sigurdson AJ, Preston DL, et al. 2015. Occupational ionising radiation
Recommendations of the International Commission on Radiological and risk of basal cell carcinoma in US radiologic technologists (1983–​
Protection. ICRP Publication 103. Ann ICRP 37, 2–​4. 2005). Occup Environ Med, 72(12), 862–​869. PMID: 26350677.
International HapMap Consortium Altshuler DM, Gibbs RA, et  al. 2010. Leeman-​Neill RJ, Brenner AV, Little MP, et  al. 2013. RET/​PTC and PAX8/​
Integrating common and rare genetic variation in diverse human popula- PPARgamma chromosomal rearrangements in post-​Chernobyl thyroid
tions. Nature, 467(7311), 52–​58. PMCID: PMC3173859. cancer and their association with iodine-​131 radiation dose and other
Ito T, Seyama T, Iwamoto KS, et  al. 1994. Activated RET oncogene in thy- characteristics. Cancer, 119(10), 1792–​1799. PMCID: PMC3648615.
roid cancers of children from areas contaminated by Chernobyl accident. Leuraud K, Richardson DB, Cardis E, et al. 2015. Ionising radiation and risk
Lancet, 344(8917), 259. PMID: 7913169. of death from leukaemia and lymphoma in radiation-​monitored work-
Jablon S, Hrubec Z, and Boice JD, Jr. 1991. Cancer in populations living near ers (INWORKS): an international cohort study. Lancet Haematol, 2(7),
nuclear facilities: a survey of mortality nationwide and incidence in two e276–​e281. PMCID: PMC4587986.
states. JAMA, 265(11), 1403–​1408. PMID: 1999880. Li C, Wang LE, and Wei Q. 2009. DNA repair phenotype and can-
John EM, McGuire V, Thomas D, et  al. 2013. Diagnostic chest X-​rays and cer susceptibility:  a mini review. Int J Cancer, 124(5), 999–​ 1007.
breast cancer risk before age 50  years for BRCA1 and BRCA2 muta- PMCID: PMC4349428.
tion carriers. Cancer Epidemiol Biomarkers Prev, 22(9), 1547–​1556. Li Y, Guolo A, Hoffman FO, and Carroll RJ. 2007. Shared uncertainty in mea-
PMCID: 4015518. surement error problems, with application to Nevada Test Site fallout
Kaatsch P, Spix C, Schulze-​Rath R, Schmiedel S, and Blettner M. 2008. data. Biometrics, 63(4), 1226–​1236. PMID: 18078484.
Leukaemia in young children living in the vicinity of German nuclear Linet MS, Kim KP, Miller DL, et al. 2010. Historical review of occupational
power plants. Int J Cancer, 122(4), 721–​726. PMID: 18067131. exposures and cancer risks in medical radiation workers. Radiat Res,
Kato TA, Wilson PF, Nagasaw H, et  al. 2009. Variations in radiosensitivity 174(6), 793–​808. PMCID: PMC4098897.
among individuals: a potential impact on risk assessment? Health Phys, Linet MS, Kitahara CM, Ntowe E, et al. 2017. Mortality in 45,634 U.S. phy-
97(5), 470–​480. PMID: 19820456. sicians likely to perform fluoroscopically-​guided interventional proce-
Kendall G, Little MP, and Wakeford R. 2011. Numbers and proportions of leu- dures compared to 64,401 psychiatrists, 1979–​2008. Radiology, 161306.
kemias in young people and adults induced by radiation of natural origin. PMID: 28234559.
Leuk Res, 35(8), 1039–​1043. PMCID: PMC3998761. Little MP, Kukush AG, Masiuk SV, et al. 2014. Impact of uncertainties in expo-
Kendall GM, Little MP, Wakeford R, et al. 2013. A record-​based case-​control sure assessment on estimates of thyroid cancer risk among Ukrainian
study of natural background radiation and the incidence of childhood leu- children and adolescents exposed from the Chernobyl accident. PLoS
kaemia and other cancers in Great Britain during 1980–​2006. Leukemia, One, 9(1), e85723. PMCID: 3906013.
27(1), 3–​9. PMCID: PMC3998763. Little MP, Kwon D, Zablotska LB, et al. 2015. Impact of uncertainties in expo-
Kesminiene A, Evrard AS, Ivanov VK, et al. 2012. Risk of thyroid cancer among sure assessment on thyroid cancer risk among persons in Belarus exposed
chernobyl liquidators. Radiat Res, 178(5), 425–​436. PMID: 22998226. as children or adolescents due to the Chernobyl accident. PLoS One,
Kim KP, Berrington de Gonzalez A, Pearce MS, et  al. 2012. Development 10(10), e0139826. PMCID: PMC4605727.
of a database of organ doses for paediatric and young adult CT scans Liu H, Wakeford R, Riddell A, et al. 2016. A review of job-​exposure matrix
in the United Kingdom. Radiat Prot Dosimetry, 150(4), 415–​ 426. methodology for application to workers exposed to radiation from inter-
PMCID: PMC3400529. nally deposited plutonium or other radioactive materials. J Radiol Prot,
Kitahara CM, Linet MS, Drozdovitch V, et  al. 2015. Cancer and circulatory 36(1), R1–​R22. PMID: 26861451.
disease risks in US radiologic technologists associated with performing Liu Y, Shete S, Wang LE, et al. 2010. Gamma-​radiation sensitivity and poly-
procedures involving radionuclides. Occup Environ Med, 72(11), 770–​ morphisms in RAD51L1 modulate glioma risk. Carcinogenesis, 31(10),
776. PMID: 26220810. 1762–​1769. PMCID: 2981459.
Kopecky KJ, Davis S, Hamilton TE, Saporito MS, and Onstad LE. 2004. Lubin JH, Tomasek L, Edling C, et al. 1997. Estimating lung cancer mortality
Estimation of thyroid radiation doses for the hanford thyroid disease from residential radon using data for low exposures of miners. Radiat
study: results and implications for statistical power of the epidemiologi- Res, 147(2), 126–​134. PMID: 9008203.
cal analyses. Health Phys, 87(1), 15–​32. PMID: 15194919. March HC. 1950. Leukemia in radiologists in a 20 year period. Am J Med Sci,
Krestinina LY, Davis F, Ostroumova E, et al. 2007. Solid cancer incidence and 220(3), 282–​286. PMID: 14771053.
low-​dose-​rate radiation exposures in the Techa River cohort: 1956–​2002. Mathews JD, Forsythe AV, Brady Z, et  al. 2013. Cancer risk in 680,000
Int J Epidemiol, 36(5), 1038–​1046. PMID: 17768163. people exposed to computed tomography scans in childhood or adoles-
Krestinina LY, Davis FG, Schonfeld S, et  al. 2013. Leukaemia incidence in cence:  data linkage study of 11  million Australians. BMJ, 346, f2360.
the Techa River cohort: 1953–​2007. Br J Cancer, 109(11), 2886–​2893. PMCID: PMC3660619.
PMCID: PMC3844904. McKeown SR, Hatfield P, Prestwich RJ, Shaffer RE, and Taylor RE. 2015.
Krewski D, Lubin JH, Zielinski JM, et al. 2006. A combined analysis of North Radiotherapy for benign disease; assessing the risk of radiation-​induced
American case-​control studies of residential radon and lung cancer. J cancer following exposure to intermediate dose radiation. Br J Radiol,
Toxicol Environ Health A, 69(7), 533–​597. PMID: 16608828. 88(1056), 20150405. PMCID: PMC4984935.
Kwon D, Hoffman FO, Moroz BE, and Simon SL. 2016. Bayesian dose-​ Memon A, Godward S, Williams D, Siddique I, and Al-​Saleh K. 2010. Dental
response analysis for epidemiological studies with complex uncertainty x-​rays and the risk of thyroid cancer: a case-​control study. Acta Oncol,
in dose estimation. Stat Med, 35(3), 399–​423. PMID: 26365692. 49(4), 447–​453. PMID: 20397774.
Labutina EV, Kuznetsova IS, Hunter N, Harrison J, and Koshurnikova NA. Mettler FA, Jr., Bhargavan M, Faulkner K, et al. 2009. Radiologic and nuclear
2013. Radiation risk of malignant neoplasms in organs of main deposition medicine studies in the United States and worldwide: frequency, radia-
for plutonium in the cohort of Mayak workers with regard to histological tion dose, and comparison with other radiation sources:  1950–​ 2007.
types. Health Phys, 105(2), 165–​176. PMID: 23799501. Radiology, 253(2), 520–​531. PMID: 19789227.
Land CE, Kwon D, Hoffman FO, et  al. 2015. Accounting for shared and Metz-​Flamant C, Laurent O, Samson E, et al. 2013. Mortality associated with
unshared dosimetric uncertainties in the dose response for ultrasound-​ chronic external radiation exposure in the French combined cohort of
detected thyroid nodules after exposure to radioactive fallout. Radiat Res, nuclear workers. Occup Environ Med, 70(9), 630–​638. PMID: 23716722.
183(2), 159–​173. PMCID: PMC4423551. Molina G. 1990. Lessons learned during the recovery operations in the Ciudad
Laurier D, Valenty M, and Tirmarche M. 2001. Radon exposure and the risk of Juarez accident. In: Proceedings of a Symposium on Recovery Operations
leukemia: a review of epidemiological studies. Health Phys, 81(3), 272–​ in the Event of a Nuclear Accident or Radiological Emergency. STI/​PUB/​
288. PMID: 11513461. 826 (pp. 517–​524). Vienna: IAEA.
Lecarpentier J, Nogues C, Mouret-​Fourme E, et al. 2011. Variation in breast Muirhead CR, O’Hagan JA, Haylock RG, et  al. 2009. Mortality and cancer
cancer risk with mutation position, smoking, alcohol, and chest X-​ray his- incidence following occupational radiation exposure: third analysis of the
tory, in the French National BRCA1/​2 carrier cohort (GENEPSO). Breast National Registry for Radiation Workers. Br J Cancer, 100(1), 206–​212.
Cancer Res Treat, 130(3), 927–​938. PMID: 21761160. PMCID: PMC2634664.
 247

Ionizing Radiation 247


Narod SA, Lubinski J, Ghadirian P, et al. 2006. Screening mammography and Rajaraman P, Simpson J, Neta G, et al. 2011. Early life exposure to diagnostic
risk of breast cancer in BRCA1 and BRCA2 mutation carriers:  a case-​ radiation and ultrasound scans and risk of childhood cancer: case-​control
control study. Lancet Oncol, 7(5), 402–​406. PMID: 16648044. study. BMJ, 342, d472. PMCID: PMC3037470.
National Council on Radiation Protection and Measurements (NCRP). 1989. Richardson DB, Cardis E, Daniels RD, et al. 2015. Risk of cancer from occupa-
Comparative Carcinogenicity of Ionizing Radiation and Chemicals. tional exposure to ionising radiation: retrospective cohort study of work-
Report No. 96. Bethesda, MD: NCRP. ers in France, the United Kingdom, and the United States (INWORKS).
NCRP. 1997. Deposition, Retention and Dosimetry of Inhaled Radioactive BMJ, 351, h5359. PMCID: PMC4612459.
Substances. Report No. 125. Bethesda, MD: NCRP. Robison LL, Armstrong GT, Boice JD, et  al. 2009. The Childhood Cancer
NCRP. 2007. Uncertainties in the Measurement and Dosimetry of External Survivor Study:  a National Cancer Institute–​ supported resource for
Radiation. Report No. 124. Bethesda, MD: NCRP. outcome and intervention research. J Clin Oncol, 27(14), 2308–​2318.
NCRP. 2009. Ionizing Radiation Exposure of the Population of the United PMCID: 2677920.
States. Report No. 160. Bethesda, MD. Roguin A, Goldstein J, Bar O, and Goldstein JA. 2013. Brain and neck tumors
NCRP. 2011. Second Primary Cancers and Cardiovascular Disease After among physicians performing interventional procedures. Am J Cardiol,
Radiation Therapy. Report No. 170. Bethesda, MD. 111(9), 1368–​1372. PMID: 23419190.
National Research Council (NRC). 2006. Review of the Worker and Public Ron E. 2003. Cancer risks from medical radiation. Health Phys, 85(1), 47–​59.
Health Activities Program Administered by the Department of Energy PMID: 12852471.
and the Department of Health and Human Services. Washington, Ronckers CM, Doody MM, Lonstein JE, Stovall M, and Land CE. 2008.
DC: National Academies Press. Multiple diagnostic X-​rays for spine deformities and risk of breast cancer.
NRC. 2012. Analysis of Cancer Risks in Populations Near Nuclear Cancer Epidemiol Biomarkers Prev, 17(3), 605–​613. PMID: 18349278.
Facilities: Phase 1. Washington, DC: National Academy Press. Ronckers CM, Erdmann CA, and Land CE. 2005. Radiation and breast can-
NRC. 2015. Committee on the Analysis of Cancer Risks in Populations near cer:  a review of current evidence. Breast Cancer Res, 7(1), 21–​ 32.
Nuclear Facilities Study: Phase 2 Pilot Planning. Washington, D.C. The PMCID: PMC1064116.
National Academies Press. Rubino C, Shamsaldin A, Le MG, et al. 2005. Radiation dose and risk of soft
Neta G, Rajaraman P, Berrington de Gonzalez A, et  al. 2013. A prospective tissue and bone sarcoma after breast cancer treatment. Breast Cancer Res
study of medical diagnostic radiography and risk of thyroid cancer. Am J Treat, 89(3), 277–​288. PMID: 15754127.
Epidemiol, 177(8), 800–​809. PMCID: PMC3668423. Rudat V, Dietz A, Nollert J et al. 1999. Acute and late toxicity, tumour control
Nikolova T, Dvorak M, Jung F, et al. 2014. The gammaH2AX assay for geno- and intrinsic radiosensitivity of primary fibroblasts in vitro of patients
toxic and nongenotoxic agents:  comparison of H2AX phosphorylation with advanced head and neck cancer after concomitant boost radiochemo-
with cell death response. Toxicol Sci, 140(1), 103–​117. PMID: 24743697. therapy. Radiother Oncol, 53(3), 233–​245. PMID: 10660204.
Parkin DM, and Darby SC. 2011. Cancers in 2010 attributable to ionis- Sachs RK, and Brenner DJ. 2005. Solid tumor risks after high doses of ion-
ing radiation exposure in the UK. Br J Cancer, 105 Suppl 2, S57–​S65. izing radiation. Proc Natl Acad Sci U S A, 102(37), 13040–​ 13045.
PMCID: PMC3252070. PMCID: PMC1199000.
Paterson A, Frush DP, and Donnelly LF. 2001. Helical CT of the body:  are Sakata R, Kleinerman RA, Mabuchi K, et al. 2012. Cancer mortality following
settings adjusted for pediatric patients? AJR Am J Roentgenol, 176(2), radiotherapy for benign gynecologic disorders. Radiat Res, 178(4), 266–​
297–​301. PMID: 11159060. 279. PMCID: PMC3471655.
Peacock J, Ashton A, Bliss J, et al. 2000. Cellular radiosensitivity and compli- Sanlorenzo M, Wehner MR, Linos E, et  al. 2015. The risk of melanoma in
cation risk after curative radiotherapy. Radiother Oncol, 55(2), 173–​178. airline pilots and cabin crew: a meta-​analysis. JAMA Dermatol, 151(1),
PMID: 10799729. 51–​58. PMCID: PMC4482339.
Pearce MS, Salotti JA, Little MP, et  al. 2012. Radiation exposure from Schubauer-​Berigan MK, Anderson JL, Hein MJ, et  al. 2015a. Breast cancer
CT scans in childhood and subsequent risk of leukaemia and brain incidence in a cohort of U.S. flight attendants. Am J Ind Med, 58(3), 252–​
tumours:  a retrospective cohort study. Lancet, 380(9840), 499–​ 505. 266. PMCID: PMC4566958.
PMCID: PMC3418594. Schubauer-​Berigan MK, Daniels RD, Bertke SJ, Tseng CY, and Richardson
Pernot E, Hall J, Baatout S, et  al. 2012. Ionizing radiation biomarkers for DB. 2015b. Cancer mortality through 2005 among a pooled cohort of
potential use in epidemiological studies. Mutat Res, 751(2), 258–​286. U.S. nuclear workers exposed to external ionizing radiation. Radiat Res,
PMID: 22677531. 183(6), 620–​631. PMID: 26010709.
Pinkerton LE, Hein MJ, Anderson JL, et  al. 2016. Breast cancer incidence Sermage-​Faure C, Laurier D, Goujon-​Bellec S, et al. 2012. Childhood leuke-
among female flight attendants:  exposure-​response analyses. Scand J mia around French nuclear power plants: the Geocap study, 2002–​2007.
Work Environ Health, 42(6), 538–​546. PMID: 27551752. Int J Cancer, 131(5), E769–​E780. PMID: 22223329.
Pinkerton LE, Waters MA, Hein MJ, et  al. 2012. Cause-​specific mortality Shin H, Ramsay T, Krewski D, and Zielinski JM. 2005. The effect of censoring
among a cohort of U.S. flight attendants. Am J Ind Med, 55(1), 25–​36. on cancer risk estimates based on the Canadian National Dose Registry of
PMID: 21987391. occupational radiation exposure. J Expo Anal Environ Epidemiol, 15(5),
Preston DL, Kitahara CM, Freedman DM, et al. 2016. Breast cancer risk and 398–​406. PMID: 15592443.
protracted low-​to-​moderate dose occupational radiation exposure in the Shipler DB, Napier BA, Farris WT, and Freshley MD. 1996. Hanford
US Radiologic Technologists Cohort, 1983–​2008. Br J Cancer, 115(9), Environmental Dose Reconstruction Project: an overview. Health Phys,
1105–​1112. PMID: 27623235. 71(4), 532–​544. PMID: 8830754.
Preston DL, Mattsson A, Holmberg E, et al. 2002a. Radiation effects on breast Sigurdson AJ, Bhatti P, Chang SC, et al. 2009. Polymorphisms in estrogen bio-
cancer risk: a pooled analysis of eight cohorts. Radiat Res, 158(2), 220–​ synthesis and metabolism-​related genes, ionizing radiation exposure, and
235. PMID: 12105993. risk of breast cancer among US radiologic technologists. Breast Cancer
Preston DL, Ron E, Yonehara S, et al. 2002b. Tumors of the nervous system and Res Treat, 118(1), 177–​184. PMID: 19214745.
pituitary gland associated with atomic bomb radiation exposure. J Natl Sigurdson AJ, Bhatti P, Doody MM, et al. 2007. Polymorphisms in apoptosis-​
Cancer Inst, 94(20), 1555–​1563. PMID: 12381708. and proliferation-​related genes, ionizing radiation exposure, and risk of
Preston DL, Ron E, Tokuoka S, et al. 2007. Solid cancer incidence in atomic breast cancer among U.S. Radiologic Technologists. Cancer Epidemiol
bomb survivors: 1958–​1998. Radiat Res, 168(1), 1–​64. PMID: 17722996. Biomarkers Prev, 16(10), 2000–​2007. PMID: 17932347.
Pukkala E, Helminen M, Haldorsen T, et  al. 2012. Cancer incidence Sigurdson AJ, and Stram DO. 2012. Genetic predisposition to radiation-​related
among Nordic airline cabin crew. Int J Cancer, 131(12), 2886–​2897. cancer and potential implications for risk assessment. Ann ICRP, 41(3-​4),
PMID: 22447246. 108–​116. PMCID: PMC3916824.
Raaschou-​Nielsen O. 2008. Indoor radon and childhood leukaemia. Radiat Simon SL, Hoffman FO, and Hofer E. 2015. The two-​dimensional Monte
Prot Dosimetry, 132(2), 175–​181. PMID: 19010936. Carlo: a new methodologic paradigm for dose reconstruction for epide-
Rajaraman P, Bhatti P, Doody MM, et  al. 2008. Nucleotide excision repair miological studies. Radiat Res, 183(1), 27–​41. PMCID: PMC4423557.
polymorphisms may modify ionizing radiation-​ related breast cancer Simon SL, Preston DL, Linet MS, et al. 2014. Radiation organ doses received
risk in US radiologic technologists. Int J Cancer, 123(11), 2713–​2716. in a nationwide cohort of U.S. radiologic technologists: methods and find-
PMID: 18767034. ings. Radiat Res, 182(5), 507–​528. PMCID: PMC4406479.
Rajaraman P, Doody MM, Yu CL, et al. 2016. Cancer risks in U.S. radiologic Smith KC, Hahn GM, Hoppe RT, and Earle JD. 1980. Radiosensitivity in vitro
technologists working with fluoroscopically guided interventional proce- of human fibroblasts derived from patients with a severe skin reaction
dures, 1994–​2008. AJR Am J Roentgenol, 206(5), 1101–​1108; quiz 1109. to radiation therapy. Int J Radiat Oncol Biol Phys, 6(11), 1573–​1575.
PMID: 26998721. PMID: 7462061.
248

248 PART III:  THE CAUSES OF CANCER


Sont WN, Zielinski JM, Ashmore JP, et al. 2001. First analysis of cancer inci- UNSCEAR. 2011a. Sources and Effects of Atomic Radiation. UNSCEAR 2008
dence and occupational radiation exposure based on the National Dose Report to the General Assembly with Scientific Annexes, Vol. II, Annex
Registry of Canada. Am J Epidemiol, 153(4), 309–​318. PMID: 11207146. C: Radiation Exposures in Accidents. New York: United Nations.
Spycher BD, Feller M, Zwahlen M, et al. 2011. Childhood cancer and nuclear UNSCEAR. 2011b. Sources and Effects of Atomic Radiation. UNSCEAR
power plants in Switzerland: a census-​based cohort study. Int J Epidemiol, 2008 Report to the General Assembly with Scientific Annexes, Vol. II,
40(5), 1247–​1260. PMCID: PMC3204210. Annex D: Health Effects Due to Radiation from the Chernobyl Accident.
Stather JW. 2011. Childhood leukaemia near nuclear sites:  fourteenth report New York: United Nations.
of the Committee on Medical Aspects of Radiation in the Environment Veiga LH, Holmberg E, Anderson H, et al. 2016. Thyroid cancer after child-
(COMARE). Radiat Prot Dosimetry, 147(3), 351–​354. PMID: 21831861. hood exposure to external radiation:  an updated pooled analysis of 12
Stayner L, Vrijheid M, Cardis E, et al. 2007. A Monte Carlo maximum like- studies. Radiat Res, 185(5), 473–​484. PMCID: PMC4893786.
lihood method for estimating uncertainty arising from shared errors in Veiga LH, Lubin JH, Anderson H, et al. 2012. A pooled analysis of thyroid can-
exposures in epidemiological studies of nuclear workers. Radiat Res, cer incidence following radiotherapy for childhood cancer. Radiat Res,
168(6), 757–​763. PMID: 18088178. 178(4), 365–​376. PMCID: PMC3488851.
Stovall M, Weathers R, Kasper C, et al. 2006. Dose reconstruction for thera- Vineis P, and Perera F. 2000. DNA adducts as markers of exposure to car-
peutic and diagnostic radiation exposures: use in epidemiological studies. cinogens and risk of cancer. Int J Cancer, 88(3), 325–​328. PMID:
Radiat Res, 166(1 Pt 2), 141–​157. PMID: 16808603. 11054658.
Stram DO, Preston DL, Sokolnikov M, et  al. 2015. Shared dosimetry error Vrijheid M, Cardis E, Ashmore P, et al. 2007. Mortality from diseases other
in epidemiological dose-​response analyses. PLoS One, 10(3), e0119418. than cancer following low doses of ionizing radiation:  results from the
PMCID: PMC4370375. 15-​Country Study of nuclear industry workers. Int J Epidemiol, 36(5),
Sun Z, Inskip PD, Wang J, et al. 2016. Solid cancer incidence among Chinese 1126–​1135. PMID: 17666424.
medical diagnostic x-​ray workers, 1950–​1995:  Estimation of radiation-​ Vrijheid M, Cardis E, Ashmore P, et  al. 2008. Ionizing radiation and risk of
related risks. Int J Cancer, 138(12), 2875–​2883. PMCID: PMC4826787. chronic lymphocytic leukemia in the 15-​country study of nuclear industry
Taalman RD, Jaspers NG, Scheres JM, de Wit J, and Hustinx TW. 1983. workers. Radiat Res, 170(5), 661–​665. PMCID: PMC4051434.
Hypersensitivity to ionizing radiation, in vitro, in a new chromosomal West CM, and Barnett GC. 2011. Genetics and genomics of radiother-
breakage disorder, the Nijmegen Breakage Syndrome. Mutat Res, 112(1), apy toxicity:  towards prediction. Genome Med, 3(8), 52. PMCID:
23–​32. PMID: 6828038. PMC3238178.
Taylor AM, Harnden DG, Arlett CF, et al. 1975. Ataxia telangiectasia: a human West CM, Davidson SE, Elyan SA, et al. 2001. Lymphocyte radiosensitivity is
mutation with abnormal radiation sensitivity. Nature, 258(5534), 427–​ a significant prognostic factor for morbidity in carcinoma of the cervix.
429. PMID: 1196376. Int J Radiat Oncol Biol Phys, 51(1), 10–​15. PMID: 11516845.
Terada H, Katata G, Chino M, and Nagai H. 2012. Atmospheric discharge Woods WG, Byrne TD, and Kim TH. 1988. Sensitivity of cultured cells to
and dispersion of radionuclides during the Fukushima Dai-​ichi Nuclear gamma radiation in a patient exhibiting marked in vivo radiation sensitiv-
Power Plant accident. Part II: verification of the source term and analysis ity. Cancer, 62(11), 2341–​2345. PMID: 3179949.
of regional-​scale atmospheric dispersion. J Environ Radioact, 112, 141–​ World Health Organization (WHO). 2006. Health Effects of the Chernobyl
154. PMID: 22721917. Accident and Special Health Care Programmes:  Report of the UN
Thierry-​Chef I, Simon SL, Weinstock RM, Kwon D, and Linet MS. 2012. Chernobyl Forum Health Expert Group. Available from: http://​www.who.
Reconstruction of absorbed doses to fibroglandular tissue of the breast int/​ionizing_​radiation.
of women undergoing mammography (1960 to the present). Radiat Res, Wu X, Gu J, and Spitz MR. 2007. Mutagen sensitivity: a genetic predisposition
177(1), 92–​108. PMCID: PMC3876279. factor for cancer. Cancer Res, 67(8), 3493–​3495. PMID: 17440053.
Till JE, Beck HL, Aanenson JW, et  al. 2014. Military participants at U.S. Zablotska LB, Ashmore JP, and Howe GR. 2004. Analysis of mortality among
Atmospheric nuclear weapons testing: methodology for estimating dose Canadian nuclear power industry workers after chronic low-​dose exposure
and uncertainty. Radiat Res, 181(5), 471–​484. PMCID: PMC4082990. to ionizing radiation. Radiat Res, 161(6), 633–​641. PMID: 15161357.
Travis LB, Hill D, Dores GM, et al. 2005. Cumulative absolute breast cancer Zablotska LB, Bazyka D, Lubin JH, et al. 2013. Radiation and the risk of chronic
risk for young women treated for Hodgkin lymphoma. J Natl Cancer Inst, lymphocytic and other leukemias among chornobyl cleanup workers.
97(19), 1428–​1437. PMID: 16204692. Environ Health Perspect, 121(1), 59–​65. PMCID: PMC3553431.
United Nations Scientific Committee on the Effects of Atomic Radiation Zablotska LB, Lane RS, and Thompson PA. 2014. A reanalysis of cancer
(UNSCEAR). 2006. UNSCEAR 2006 Report to the General Assembly, mortality in Canadian nuclear workers (1956–​1994) based on revised
with Scientific Annexes, Annex A: Epidemiological Studies of Radiation exposure and cohort data. Br J Cancer, 110(1), 214–​223. PMCID:
and Cancer. New York: United Nations. PMC3887280.
UNSCEAR. 2010a. Sources and Effects of Atomic Radiation. UNSCEAR 2010 Zablotska LB, Ron E, Rozhko AV, et al. 2011. Thyroid cancer risk in Belarus
Report to the General Assembly with Scientific Annexes, Vol. I, Annex among children and adolescents exposed to radioiodine after the Chornobyl
A: Medical Radiation Exposures. New York: United Nations. accident. Br J Cancer, 104(1), 181–​187. PMCID: PMC3039791.
UNSCEAR. 2010b. Sources and Effects of Atomic Radiation. UNSCEAR Zielinski JM, Garner MJ, Band PR, et al. 2009. Health outcomes of low-​dose
2010 Report to the General Assembly with Scientific Annexes, Vol. I, ionizing radiation exposure among medical workers: a cohort study of the
Annex B: Exposures of the Public and Workers from Various Sources of Canadian national dose registry of radiation workers. Int J Occup Med
Radiation. New York: United Nations. Environ Health, 22(2), 149–​156. PMID: 19546093.
 249

14 Ultraviolet Radiation

ADÈLE C. GREEN AND DAVID C. WHITEMAN

OVERVIEW biological effect of interest (e.g., DNA mutation, erythema), and then
summing these weighted values across all wavelengths.
Ultraviolet (UV) radiation is the principal cause of over 95% of kera-
tinocyte cancers (basal cell carcinomas and squamous cell carcinomas
of the skin), the most common cancers in white populations world- METHODS OF MEASUREMENT
wide; it also causes the majority (estimated 60%–​90%) of cases of
cutaneous melanoma, the cancer of the skin’s pigment-​ producing At the earth’s surface, solar UV radiation is measured by a wide vari-
cells. In addition, UV radiation is the major cause of many eye dis- ety of detector instruments (the most common of which are radiom-
eases, including ocular cancers and cataract, the most common cause eters), each having different attributes and requirements for calibration
of blindness, and is responsible for the underlying changes in aged and maintenance (Godar, 2005). In addition, portable monitors and
skin on which billions of dollars are spent annually in efforts to repair data-​loggers are increasingly available (Hooke et al., 2014).
the damage. Clinicians first recognized the causal role of sun exposure Broadband radiometers measure UV irradiance over a broad spec-
in keratinocyte cancer development around the turn of the twentieth tral band, integrated over the wavelengths of radiation known to exert
century. The probable role of sunlight in driving high melanoma mor- a particular biological effect (e.g., erythema). The output from broad-
tality in populations of European ancestry living at low latitudes was band radiometers is a single number, calculated from the voltage gen-
identified 50 years later. erated by filtered UV radiation striking a photodiode. These simple
The sun is the universal source of human exposure to UV radiation, instruments are portable and robust, although relatively insensitive
but artificial sources are also encountered in a wide range of settings, to changes in irradiance at specific wavelengths. Networks of radi-
from medical to industrial, and increasingly from commercial tanning ometers were first established during the 1970s and 1980s, often in
outlets. A  century ago, high cumulative UV exposure was the prov- remote locations, and subsequently have provided long-​term informa-
enance of those who worked in outdoor occupations (and in whom tion about levels of biologically effective UV radiation at the earth’s
skin cancers were first described), but during the course of the postwar surface (Frederick et al., 2000; Henderson et al., 2010; Pearson et al.,
decades, social customs changed dramatically such that people with pale 2000). Popular early models (e.g., Robertson-​Berger meters) have
skins sought to spend leisure time outdoors to acquire a suntan, and peo- been largely replaced by newer models that have less technical error
ple living in temperate climates likewise spent their vacations in sunny and are more stable under extremes of temperature, although some
locations and began to patronize tanning salons. By the late twentieth limitations remain (Huber et al., 2002).
century, however, the near epidemic increases in skin cancer incidence Spectroradiometers are more sophisticated instruments that mea-
in white populations, especially in Australia and New Zealand, stimu- sure small changes in UV flux at specific wavelengths across the spec-
lated the initiation of skin cancer awareness and prevention campaigns, trum such as occurs following depletions in atmospheric ozone from
including sun protection and legislation to restrict or ban sunbed use, in time to time (Roy et  al., 1997). Notwithstanding their cost and the
an effort to control this growing public health problem. skill required to operate them, networks of spectroradiometers have
been established worldwide to monitor changes in surface irradiance.
For example, the US National Oceanic and Aerospace Administration
SOLAR RADIATION AND (NOAA) and the Environmental Protection Agency jointly manage the
THE ELECTROMAGNETIC SPECTRUM NEU-​Brew network of spectroradiometers (http://​esrl.noaa.gov/​gmd/​
grad/​neubrew). Since 2006, these instruments have measured daily
Solar UV radiation, a ubiquitous environmental carcinogen, is part of UV radiation and total column ozone at six locations in the western,
the spectrum of electromagnetic radiation emanating from the sun. UV central, and eastern United States (Houston, TX; Table Mountain, CO;
radiation is also generated by artificial sources encountered in a wide Mountain Research Station, CO; Bondville, IL; Raleigh, NC; Ft. Peck,
range of settings. The ultraviolet spectral region spans wavelengths of MT). This latest generation of instruments replaced an earlier network
10 nm–​400 nm, and is divided into several bands: UVA (320–​400 nm); of spectroradiometers operated by the EPA from the 1990s until 2004
UVB (280–​320  nm); UVC (200–​280  nm); far UV (120–​200  nm); (UV-​NET; http://​www.epa.gov/​uvnet/​), of which 14 instruments were
extreme UV (10–​120 nm). Although UV radiation is classified as non-​ located in US National Parks as part of the Park Research and Intensive
ionizing, UV photons are sufficiently energetic to destabilize electron Monitoring of Ecosystems Network (PRIMENet). Similar networks
configurations within molecules such as DNA and to have a biologi- of UV spectroradiometers have been established in many other coun-
cal effect. In terms of solar UV radiation, only UVB and UVA have tries, contributing to a global network coordinated by the World
biological significance, as shorter wavelengths are absorbed by the Meteorological Organization to monitor UV irradiance (World Ozone
atmosphere. and Ultraviolet Radiation Data Centre, administered by Environment
UV radiation is typically absorbed over a surface and is measured Canada, http://​www.woudc.org/​home.php/​). Historical UV data from
as a radiant exposure. The term irradiation represents the dose of radi- global locations can be downloaded from this site for analysis.
ant energy delivered to an area within a given time, and is measured
J/​m2. The rate at which UV energy reaches a surface is termed irradi-
ance (units W/​m2). The total irradiance from any given source of UV DETERMINANTS OF SOLAR IRRADIANCE
radiation is derived by summing the wavelength-​specific irradiances AND SOLAR DOSE
across the spectrum of wavelengths emitted. A “biologically effective
UV irradiance” (UVeff) for a given source is determined by weighting The irradiance at the surface of the earth is much less than in the
the irradiance at each emitted wavelength by its ability to cause the upper atmosphere due to absorption by stratospheric ozone of all

249
250

250 PART III:  THE CAUSES OF CANCER


UVC and a large proportion of UVB before reaching the earth. The erythemal response of human skin when formulated as a film 30–​45
thickness of the ozone mantle varies according to season, latitude, µm thick (Diffey, 1987). These dosimeters have been useful in field
and meteorological conditions and by levels of chlorofluorocarbons studies as they are inexpensive, stable, and easy to apply, all of which
and other gases generated by human activities that can destroy ozone contribute to high rates of compliance in large-​scale studies (Sun et al.,
(Rowland, 2006). International agreements like the Montreal Protocol 2014). However, they do become saturated at relatively modest doses
aimed to reduce the emission of chlorofluorocarbons (Weatherhead of UVR, requiring participants in exposure studies to periodically
and Andersen, 2006), and monitoring shows that increases in terres- change badges.
trial UVB have been modest (Williamson et  al., 2014). However, it More recently, electronic dosimeters have been developed that
is uncertain whether subsequent small improvements in ozone levels comprise miniature battery-​powered UV detectors that can be worn
are due to a decline in the amount of ozone-​depleting substances in on the wrist or other exposed body sites (Allen and McKenzie, 2005;
the Earth’s atmosphere or if they reflect the high natural variability in Heydenreich and Wulf, 2005). These dosimeters have the advantage
ozone levels related to the solar cycle and recent changes in global air of logging UV radiation data at specified intervals, thereby provid-
transport and temperature that make it unlikely that ozone will stabi- ing investigators with information about time-​dependent aspects of
lize at levels observed before 1980 (Weatherhead and Andersen, 2006; sun exposure, such as dose rates and maximum exposure intensity
Williamson et  al., 2014). UV irradiance, especially UVB, increases (Seckmeyer et al., 2012). In addition, electronic dosimeters are able
with decreasing latitude, increasing altitude, in summer, and in the to be reused, do not saturate after short intervals, and their response to
middle hours of the day, and decreases with heavy cloud cover. UVA UVR exposure is linear rather than logarithmic (Allen and McKenzie,
levels are less affected than UVB by atmospheric factors and are more 2005; Seckmeyer et al., 2012).
stable throughout the day. By the end of the twenty-​first century rela- While these various approaches to measuring personal UVB expo-
tive to the present decade (2010–​2020), it is expected that projected sure provide a degree of objectivity, there is attendant measurement
decreases in ozone-​depleting gases and changes in cloud cover will error when compared with gold standard measures of UV radiation as
lead to relatively small decreases in UVB, while reductions in reflec- determined by spectroradiometers. It has been suggested that estimates
tivity of melting Arctic sea ice will lead to decreases of up to 10%. On of UVB exposure from polysulfone badges and electronic dosimeters
the other hand, expected improvement of air quality and in particular may deviate from the gold standard by up to 26% and 15%, respec-
reductions of aerosols over densely populated areas of the Northern tively (Seckmeyer et  al., 2012). In addition to issues of calibration,
Hemisphere are expected to result in increases in UVB of 10%–​20%, the physical location and placement of dosimeters on the body also
with even higher projected increases over China (Bais et al., 2015). cause measurements to vary. Human field studies comparing measure-
ments obtained from dosimeters placed concurrently on various body
sites have shown that maximal readings are obtained from dosimeters
The UV Index located on the head (almost 90% of ambient UVR), with lower read-
The standardization of UV information to the public through the UV ings obtained from dosimeters on the shoulder (55% of ambient) and
Index (UVI) was introduced and endorsed by multiple international wrist (43%) (Thieden et al., 2000). Given that the aim of UV radiation
agencies in the 1990s. UV Index values are available throughout the measurement in most epidemiologic studies is to rank participants by
world from satellite instruments, ground-​based measurements, and relative exposure, the wrist is generally the preferred site of placement
modeled forecasts (Zaratti et al., 2014), and are scaled from 0 to 11+, for reasons of convenience, compliance, and reproducibility (Petersen
with 11+ classified as “extreme.” Increasing levels of sun protection et al., 2014; Sun et al., 2014; Thieden et al., 2006).
are recommended with increasing UVI values to 11+. However, a Numerous field studies have deployed dosimeters and data-​loggers
recent call for modification of the UV Index (Zaratti et al., 2014) was on humans outdoors. Most studies to date have been relatively small in
based on the bias that exists toward European skin types in the cur- scale, and of limited duration, and often have been conducted within
rently depicted UV Index and its failure to properly account for the a single geographic site, so generalizations must be made cautiously.
most extreme values of the UV Index, over 20, experienced by popula- Nevertheless, it appears that most people receive a personal dose of
tions living close to the equator and at high altitudes, such as in the UV radiation less than 5% of daily ambient UV radiation, although
Altiplano region of South America. there is wide inter-​individual variability, and the measured dose is
heavily dependent on environmental, behavioral, and occupational
factors, as well as personal characteristics and the anatomic site of the
MEASURING PERSONAL EXPOSURE dosimeter. As earlier, higher proportions of ambient radiation may be
received on horizontal skin surfaces (such as the vertex of the head,
TO SOLAR RADIATION the top of the shoulders), with lower doses at other sites (Weihs et al.,
2013). Movement, body position, and orientation with respect to the
Humans are exposed to sunlight on a daily basis, yet accurate and
sun all greatly affect the dose received at any given site (Weihs et al.,
reproducible methods for measuring the dose of solar radiation
2013). The level of ambient UV radiation is the strongest influence on
received by an individual have only been developed in the past few
a person’s received dose; however, the influence of ambient UV radia-
decades. In epidemiologic studies, human sun exposure has been mea-
tion on average doses when compared across populations is modified
sured in a variety of ways, ranging from dosimetry to personal recall
by latitude and season. In most non-​equatorial parts of the world, the
of past exposure.
average daily levels of ambient UV radiation are higher in summer
than winter, and the magnitude of seasonal difference increases with
latitude. One might therefore predict that seasonal fluctuations in per-
Dosimetry sonal UV radiation doses would follow a similar temporal course to
Dosimeters are portable units designed to measure personal, incident ambient UVR, with higher doses being received in summer than in
exposure to solar UV radiation. Historically, the two most common winter months. However, dosimetry studies of “free-​living humans”
dosimeters used in human research have been the Bacillus biologi- conducted over a range of latitudes report discordant seasonal trends
cal dosimeter and the polysulfone badge, both of which estimate for different geographic locations. In temperate climates at mid-​to
total doses of sun exposure over relatively short periods (Green and high latitudes, average daily personal UV radiation doses tend to rise
Whiteman, 2006). The biological dosimeter comprises Bacillus sub- in spring and peak in summer in line with ambient UV radiation levels
tilis spores immobilized on a polyester sheet, which are inactivated in (Sun et al., 2014; Thieden et al., 2004b), whereas in subtropical and
a dose-​dependent manner upon exposure to UV radiation (Quintern tropical locations, higher personal UVR doses are observed in winter
et  al., 1992). Dosimeters of this type were used in diverse settings, than in summer (Sun et al., 2014). The accepted explanation is that the
including schools (Munakata et al., 1998), mountains (Moehrle et al., intensity of heat and solar radiation in summer months at low latitudes
2003), and the Arctic (Cockell et al., 2001). Polysulfone is UVR-​sen- discourages fair-​skinned residents from outdoor activities, particularly
sitive polymer that has an action spectrum closely approximating the during the middle part of the day. Conversely, the mild temperatures
 251

Ultraviolet Radiation 251


and less intense levels of ambient UV radiation during winter months The first approach has been to estimate an individual’s average
in tropical settings are more conducive to outdoor work and recreation, exposures to solar UV radiation during defined time periods according
resulting in higher levels of personal sun exposure on average. to places of residence, since ambient solar UV radiation increases with
Few studies have investigated correlations between host character- proximity to the equator (Godar, 2005). Epidemiologists have com-
istics and personal UV radiation; the scant available evidence suggests monly used these ambient exposure measures when conducting eco-
that people with sun-​sensitive phenotypes (such as red or light-​colored logical studies, such as comparing the incidence of melanoma among
hair, a tendency to sunburn, or poor tanning ability) typically receive native residents and migrants with contrasting experiences of past sun
lower average daily doses of UV than those without these character- exposure. Although place of residence is a relatively crude measure of
istics (Cahoon et  al., 2013; Thieden, 2008). Studies from the high-​ personal sun exposure, it is a good proxy for solar dose when compar-
latitude Nordic countries have reported higher average daily personal ing populations (Diffey et  al., 1996; Diffey and Gies, 1998; Godar,
UV radiation doses in women than in men (Thieden et  al., 2004a), 2005) and has the practical advantage of being easy to recall.
whereas studies from mid-​to low-​latitude Australian settings report The other approach has been to ask study participants to recall salient
the opposite (Xiang et  al., 2015). Within any given population, the exposures, such as numbers of sunburns, time spent outdoors in sum-
highest doses of UV radiation are experienced during outdoor activi- mer, or recreational activities associated with high levels of sun exposure
ties. Very high UV doses have been recorded among outdoor workers (English et al., 1998). An episode of sunburn is an integrated measure of
in diverse settings, including alpine environments and the Canadian acute solar dose, and thus the number of sunburns a person experiences
Arctic (Cockell et  al., 2001). As epidemiologic studies often seek gives an indication of accumulated acute exposures to intense solar UV
information on past exposures to the sun, it is interesting to note that radiation (Green et al., 1985; Whiteman and Green, 1994). Recall of past
a Danish study observed high levels of intra-​individual concordance sunburn episodes is reasonable, with most studies suggesting moderate
for personal UVR doses measured at two time points up to 7  years repeatability (kappa scores 0.5–​0.7) (Jennings et al., 2013; Morze et al.,
apart (Thieden et al., 2013), suggesting that individual patterns of sun 2012; van der Mei et al., 2006; Veierod et al., 2008). The number of sun-
exposure tend to be relatively stable over time. burns experienced by a person is due not only to the intensity or dura-
tion of sun exposure, but also to host characteristics including skin type
and pigmentations. Other recalled measures of UV exposure that are not
Biomarkers completely independent of host characteristics include time spent out-
The use of biomarkers seeks to overcome the inherent limitation of doors, vacations in sunny locations, use of tanning salons and outdoor
the dosimeters described earlier, namely that while dosimeters mea- occupations, most of which have moderate to high levels of agreement
sure the amount of ambient UV radiation to which individuals are when subjected to test-​retest reliability studies (Jennings et  al., 2013;
personally exposed, they do not measure the mutagenic effects of sun Morze et al., 2012; van der Mei et al., 2006; Veierod et al., 2008).
exposure of interest to cancer epidemiologists. UV radiation induces
specific types of DNA lesions in the nuclei of skin cells (see later dis- Phenotypic Markers of UV Exposure
cussion), including thymine dimers, and these lesions tend to accu-
mulate in a dose-​dependent manner. These lesions can be detected Because of the challenges in measuring past sun exposure by ques-
readily in DNA harvested from skin biopsies, but such invasive meth- tionnaire-​based methods, and because dosimetry and biomarkers are
ods of sample collection are unsuitable for most epidemiologic stud- limited to recent or current UV exposures, researchers have sought
ies. Moreover, such samples provide a measure of UV-​induced DNA “objective” measures of historical sun exposure based on biological
damage at specific anatomical sites only, which may not correlate with changes that correlate with exposure. Photoaging encompasses a range
a global measure of mutagenesis. Recently, non-​invasive urine tests of visible and microscopic changes to the skin associated with chronic
have been developed to measure the burden of UV-​specific mutations exposure to UV radiation. Silicone-​rubber skin casts capture a record
in human subjects. of the loss of visible surface architecture. These casts have several epi-
During the exquisitely refined process of DNA repair (see later dis- demiologic virtues: they can be obtained rapidly; they are inexpensive,
cussion), the damaged segments of nucleic acid containing thymine safe, and painless to participants; and they can be stored indefinitely.
dimers are excised and are replaced with a new segment of DNA. Moreover, graders can be trained easily to make reliable assessments
The excised fragments of damaged DNA are secreted from the skin of photoaging (kappa values > 0.90) that correlate well with histologi-
cells into the bloodstream, to be excreted by the kidney in the urine cal measures of photoaging (Battistutta et al., 2006).
(Cooke et al., 2013). Although presently a labor-​intensive procedure,
it is possible to quantify the amount of thymine dimers in the urine
ARTIFICIAL SOURCES OF UV EXPOSURE
in a reproducible manner using radio-​labeled high-​performance liq-
uid chromatography (Kotova et al., 2005). Kinetic studies demonstrate
While most people’s only UV exposure comes from sunlight, artificial
that urinary thymine dimer levels reach a maximum 3–​4 days post-​UV
UV sources may contribute to a material proportion of total UV expo-
exposure. In field studies among indoor workers in Sweden, thymine
sure for some. In particular, the prevalence of exposure to artificial
dimers were undetectable in urine samples taken in winter, but fol-
UV radiation for cosmetic tanning has increased markedly in recent
lowing typical working weeks in summer, about 20% of such peo-
decades, with users attaching considerable personal and social impor-
ple exhibited measurable levels of thymine dimers (Liljendahl et al.,
tance to tanned skin (Grange et al., 2015; Lake et al., 2014). A sys-
2013). Levels of urinary thymine dimers are strongly correlated with
tematic review and meta-​analysis in 2013 showed the large extent of
increased UV exposure. These effects have been demonstrated in chil-
ever-​exposure to indoor tanning in Western countries, especially in the
dren and adults in outdoor settings in Sweden (Liljendahl et al., 2012),
young (Wehner et al., 2014), with summary prevalences of 36% for
as well as in Danish skiers, and Danish and Spanish sun-​seekers in a
adults, 55% for university students, and 19% for adolescents.
subtropical beach setting (Petersen et al., 2014). At the present time,
Although the indoor tanning industry generally implies to users
thymine dimers have not been used as measures of UV exposure in
that their use of tanning parlors complying with regulations is safe
epidemiologic studies with skin cancer endpoints.
and might have health benefits (Autier et al., 2011), several reviews
show the high risks of ill effects on eyes and skin, including melanoma
and other skin cancers (Boniol et al., 2012; International Agency for
Personal Recollection Research on Cancer, 2012; Wehner et al., 2012). Surveys have indi-
Because dosimeters and biomarkers measure current or recent solar cated variable compliance by the sunbed industry with guidelines
exposure, they cannot be used in epidemiologic studies that seek to (Chandrasena et al., 2013), and exposure to tanning units can still result
capture past exposure. Accurate recall of past sun exposure is diffi- in high UV doses (Khazova et al., 2015; Nilsen et al., 2011), similar
cult; nevertheless, epidemiologists have attempted to estimate past sun to exposures received while sunbathing in summer at a Mediterranean
exposure in two main ways (Whiteman et al., 2001). resort. Measured UV irradiance from a large number of Norwegian
25

252 PART III:  THE CAUSES OF CANCER


solaria (sunbeds and stand-​up cabinets) in 2008 showed that overall dihydroxyguanine (8-​oxodGuo). Recent experiments have demon-
compliance had increased since 1998–​1999 but total UV irradiance strated that erythemal doses of UVA actually induce more CPDs
had not decreased, mainly because of higher UVA irradiance (Nilsen than 8-​oxodGuo in human skin (Mouret et  al., 2006), although in
et al., 2011). A recent survey in the United Kingdom showed that an relative terms, the amount of CPD damage induced by UVA is about
increase in sunbed emissions had occurred in the last decade, with five-​fold less than an erythemally equivalent dose of UVB (Tewari
UVB irradiance of over 0.3 W m–​2 in more than 85% of all tested et al., 2012). For any given dose of UVA, however, the yield of CPDs
solaria, though exposure per session appeared not to have increased is greater in the cells of the basal epidermis than in more superfi-
(Khazova et al., 2015). cial layers, whereas the potency of UVB diminishes with increas-
Other sources of artificial UV exposure are found in occupational ing depth (Tewari et al., 2012). These findings are important, since
and medical settings. In occupational settings, workers are normally cells in the basal layer include the stem cells and transitional cells
shielded from material UV radiation, though prolonged exposure to that are likely to be the progenitors of keratinocyte cancers (Halliday
intense UV radiation from arc welding is hazardous to both eyes and and Cadet, 2012). Given that around 95% of the daily UV irradiance
skin (International Agency for Research on Cancer, 1992, 2012). In falls in the UVA range, the realization that UVA also causes CPDs in
clinical usage, different UV dose regimens influence the occurrence measurable quantities in the dividing cells of the basal epidermis has
of long-​term carcinogenic side effects. Applications such as PUVA potential ramifications for primary prevention strategies, which have
photochemotherapy that combines UVA with photosensitizing pso- focused mainly on the hazards arising from UVB wavelengths. As
ralens to treat skin diseases such as psoriasis, have been used widely. yet, it is too early to determine how these laboratory findings might
A  systematic literature review of the carcinogenic risks of psoralen affect public health advice; larger scale field studies are required to
UVA therapy versus narrowband UVB therapy, also commonly used address the relative importance of UVA and UVB at a population
for chronic plaque psoriasis, concluded that there is an increased risk level.
of skin cancer following PUVA shown by both US and European The mechanism through which UVA causes CPDs appears to
studies, with a greater risk seen in US studies, likely to be partly involve melanin, the complex pigment produced by melanocytes
explained by higher average UVA doses and lighter phototypes of that shields nuclear material from UV radiation (Abdel-​Malek and
treated patients (Archier et  al., 2012). There were insufficient pro- Cassidy, 2015). Following UVA exposure, various classes of enzymes
spective studies in psoriasis patients treated with UVB to assess the are induced that generate large intracellular quantities of nitric oxide
associated carcinogenic risk. and superoxide. At the same time, UVA sufficiently solubilizes mela-
nin polymers into small fragments that are able to enter the nucleus of
cells. Once exposed to nitric oxide and superoxides, the melanin frag-
BIOLOGIC MECHANISMS FOR UVR CARCINOGENESIS ments undergo electrolysis in an electrolytic reaction, releasing similar
energy to a UV photon, sufficient to generate CPDs when in close
DNA Damage proximity to DNA (Premi et al., 2015). This mechanism is thought to
The principal mechanism of UV carcinogenesis at the molecular level explain the observation that induction of CPDs by UVA in skin cells
is DNA damage. DNA damage may arise directly through highly containing melanin peaks several hours after exposure (with so-​called
energetic UV photons interacting with chemical elements of the dou- dark CPDs), unlike UVB, for which maximal photodamage is almost
ble-​helix, or indirectly through the transfer of energy from photons instantaneous. Of note, it underscores the potential hazards to DNA
through endogenous photosensitizers to DNA. Most DNA damage is integrity from UVA light sources (such as tanning salons) (Abdel-​
repaired with very high fidelity by specific enzymes before the cell Malek and Cassidy, 2015).
replicates. If repair fails, then the damaged nucleotide can be replaced
by an incorrect nucleotide in the DNA of the daughter cell, resulting
DNA Repair
in a permanent change in the sequence known as a mutation. Drawing
a distinction between DNA damage (including lesions, adducts, and Terrestrial organisms have evolved highly efficient mechanisms to
photoproducts) and mutation is critically important in establish- repair DNA damaged by UV radiation. Enzymatic nucleotide exci-
ing the temporal relationship along the pathway to carcinogenesis sion repair (NER) is the only pathway through which UVB-​induced
(Brash, 2015). photoproducts are removed in placental mammals (Vermeulen and
The chemical structure of DNA, particularly the density of pyrim- Fousteri, 2013). NER operates through two mechanisms, called
idine bases (thymine [T]‌and cytosine [C]), renders the molecule global genome NER (GG-​ NER) and transcription-​coupled NER
a potent absorber of UV photons (Pfeifer and Besaratinia, 2012). (TC-​NER), respectively (Marteijn et al., 2014). Other types of DNA
Incident UVB radiation directly damages the integrity of DNA by photodamage induced by UVR are repaired by different mecha-
inducing bulky lesions (cyclobutane pyrimidine dimers [CPDs] and nisms; for example, oxidative lesions are removed by base excision
[6–​4] photoproducts) between adjacent pyrimidine nucleotide bases repair.
that interfere with essential genomic functions such as transcription In GG-​NER, the entire genome is scanned for distortions to the dou-
and replication. If these lesions are not repaired, then characteris- ble-​helix caused by destabilizing DNA lesions. The XPC protein plays
tic mutations occur, most frequently base substitutions of C by T a central role in recognizing DNA damage, and in the specific case
at dipyrimidine sites, and less commonly, CC to TT tandem base of CPDs, recognition is enhanced by UV-​DDB (UVR-​DNA damage
substitutions (Ikehata and Ono, 2011). Such is the specificity of these binding protein). Once detected, a cascade of proteins acts to verify the
mutations for sunlight exposure that they are termed UV signature lesion (XPB and XPD, encoded by ERCC3 and ERCC2 genes, respec-
mutations. Recently there has been renewed appreciation that UV tively), excise the damaged DNA fragment of about 22–​30 nucleotides
damage causes many other nonspecific DNA mutations. Whether (XPF and XPG), and then synthesize the sequence and ligate it to the
or not UV photodamage results in a signature mutation depends on preserved single strand.
the precise sequence of nucleotides at the target site, as well as the The GG-​NER process is relatively slow, however, and it is not
presence of other chemical elements in close proximity at the time uncommon that photoproducts remain unrepaired at the time of
of photon strike (Brash, 2015). It follows that the absence of a UV transcription or cell replication. Stalled transcription can have lethal
signature mutation in a cancer gene of interest does not mean that the consequences for cells, and so the TC-​NER pathway has evolved to
mutation was not caused by sun exposure. remove stalled complexes and to ensure that damaged segments can
While DNA is a major chromophore for UVB radiation, it absorbs be repaired and transcribed in a timely fashion. The precise sequence
photons in the UVA spectrum poorly. Thus UVA induces DNA dam- of events is the subject of intense research scrutiny, but it appears
age indirectly. For many years, it was held that UVA interacted with that the key trigger for TC-​NER is when RNA polymerase II reaches
sensitizers, both endogenous (e.g., bilirubin, porphyrins) and exog- a bulky DNA photolesion and is unable to proceed with RNA syn-
enous (e.g., medications, cosmetics, sunscreens) to produce reactive thesis. This initiates a complex cascade of verification and excision
oxygen species that result in characteristic lesions such as 8-​oxo-​7,8 proteins, which remove the damaged DNA fragment and synthesize
 253

Ultraviolet Radiation 253


a new sequence. The fidelity of the transcription-​ coupled repair This is the tanning response induced by sun exposure (Chen et  al.,
process seems to vary according to conditions, however, with both 2014). From the perspective of cancer epidemiology, it is important to
error-​free and error-​prone repair pathways known to exist (Ikehata recognize that there are two types of melanin, the brown/​black eumela-
and Ono, 2011). Overall, it is estimated that about 90% of CPD dam- nin and the red/​yellow pheomelanin, and the relative abundance of
age is repaired by GG-​NER, with the remainder repaired by TC-​NER each is determined largely by MC1R genotype (Garcia-​Borron et al.,
(Vermeulen and Fousteri, 2013). 2014). MC1R is a highly polymorphic gene with more than 200 vari-
The importance of DNA repair pathways has been shown in people ants recorded. MC1R variants associated with red hair color represent
with xeroderma pigmentosum (XP), who lack enzymes that specifi- a loss-​of-​function mutation that results in the production of pheomela-
cally repair UV-​induced lesions (Kiss and Anstey, 2013). These indi- nin. In addition to its central role in the tanning response, MC1R is
viduals manifest extreme sun sensitivity, premature development of now understood to protect against the adverse effects of UVR through
multiple skin cancers, and early death. Even within the population non-​pigmentary mechanisms (Robinson et al., 2010), including DNA
at large, there is considerable variation in DNA repair efficiency, repair (Swope et al., 2014).
prompting investigations of whether people with less efficient DNA
repair mechanisms are at higher risk of UV-​associated cancers, with
conflicting findings to date (Han et al., 2005; Mocellin et al., 2009; Immunosuppression
Ruczinski et al., 2012). Actively transcribed genes are repaired more UV radiation suppresses immunity in humans and animals; however,
quickly than infrequently expressed genes, and since the pattern of the extent, duration, and type of immunsuppression depend on many
transcriptional activity differs by cell type, so too will the distribution factors (extrinsic and intrinsic) and are subject to complex regulatory
of mutations. control (Halliday et al., 2012; Hart et al., 2011). Both UVA and UVB
induce immune suppression of the skin, probably by different mecha-
nisms (Damian et al., 2011). The molecular mediators of UV-​induced
UV Radiation Mutagenesis local immunosuppression include DNA damage, cis-​urocanic acid,
Mutations occur when DNA repair mechanisms fail to restore the and reactive oxygen species (Schwarz and Schwarz, 2011).
original sequence of nucleotides that existed prior to photodamage. At the cellular level, UV exposure causes Langerhans cells, the den-
There is consensus that certain photolesions confer a higher probabil- dritic antigen-​presenting cells of the skin, to migrate to draining lymph
ity of erroneous repair than others, and hence have higher mutagenic- nodes and activate T and B regulatory cells (Halliday et  al., 2012).
ity (Pfeifer and Besaratinia, 2012). CPD photoproducts account for the Other cell types demonstrated to play a role in UV-​induced immuno-
majority of mutations in mammalian cells, as they are incurred more suppresion of the skin include mast cells (Byrne et al., 2008), natural
commonly than 6–​4 photoproducts following UVR exposure, and are killer cells (Fukunaga et al., 2010), and effector and memory T-​cells
repaired much more slowly. Even for CPDs, however, it appears that (Rana et al., 2008). UV radiation can also suppress immune responses
TT sequences are repaired with higher fidelity than TC sequences and systemically at sites distant from the skin, including lower levels of
that methylated dipyrimidine sites are particular “hot spots” for UVR activity of memory T-​cells in bone marrow and spleen.
mutations. DNA damage arising from oxidative stress is also muta- The evidence that immune surveillance plays a role in protecting
genic, causing G to A mutations. against skin cancer development is indirect, derived mainly from the
It has long been known that mutations in key regulatory genes are high incidence of skin cancers in organ transplant patients treated with
the hallmarks of cancer. Early work focused on the role of tumor sup- immunosuppressive drugs, an effect that is amplified in immunosup-
pressor genes, so named because loss-​of-​function mutations confer pressed patients who experience high sun exposure (Euvrard et  al.,
increased risks of cancer in experimental models. TP53 is one such 2003; Vajdic and van Leeuwen, 2009).
gene, whose protein product (p53) plays a central role in cell cycle
regulation, apoptosis, and DNA repair (Lane, 1992). When this func-
tion is lost through mutation or deletion, cells have a reduced capacity Vitamin D Synthesis
to repair DNA damage, facilitating cell transformation. Early research UVB radiation is necessary for the conversion in the skin of 7-​dehy-
showed that UVR signature mutations in TP53 were detectable in about drocholesterol to cholecalciferol (vitamin D3), the precursor to the
50% of basal cell carcinomas (BCC) and more than 90% of squamous biologically active form of vitamin D.  When measured by dosim-
cell carcinomas (SCC) in humans (Brash et al., 1991; Campbell et al., etry, personal UV exposure contributes modestly (about 8% of the
1993; Rady et al., 1992). variance explained) to serum vitamin D concentrations (Kimlin
Of epidemiological importance has been recent research docu- et  al., 2014)  after controlling for related factors like latitude and
menting the high prevalence of UV signature mutations in non-​can- seasonal factors, use of vitamin D supplements, race, body mass
cerous “normal” human skin. Careful mapping studies have revealed index, and physical activity (Freedman, 2013; Kimlin, 2014; Millen,
that up to 30% of skin cells on the eyelid carry non-​silent driver 2010). Observational studies have suggested that people with low
mutations in known skin cancer genes including NOTCH1, NOTCH2, serum concentrations of vitamin D have increased risks of cancers
TP53, and FAT1 (Martincorena et  al., 2015), suggesting that “pre- at various sites (particularly colon and breast) when compared with
neoplastic clones” are extremely common in sun-​exposed human epi- people who have higher concentrations of vitamin D. At the present
dermis. Understanding how sunlight and genes interact to promote time, however, the observational evidence regarding vitamin D and
tumor development and establishing how the process can be halted cancer falls short of causality (IARC, 2008; Manson and Bassuk,
or reversed once initiated are the focus of intense research (Viros 2015) and is not supported by the findings of clinical trials (Bolland
et al., 2014). et al., 2014).

UV Radiation and Gene Expression CANCERS ASSOCIATED WITH UV EXPOSURE


Exposure to UV radiation invariably leads to DNA damage in kera-
Keratinocyte Cancers
tinocytes. Upon detection of damage, p53 is stabilized to halt kerati-
nocyte replication and allow the damage to be repaired. At the same There is a large body of observational and experimental evidence
time, p53 activates the transcription of pro-​opiomelanocortin, which is that UV radiation is the principal environmental cause of keratino-
then cleaved to alpha-​melanocyte stimulating hormone (α-​MSH) and cyte cancers (basal cell carcinomas and squamous cell carcinomas of
released by keratinocytes. α-​MSH binds to the melanocortin-​1 receptor the skin) (International Agency for Research on Cancer, 1992, 2012;
(MC1R) on the cell membrane of neighboring melanocytes, launching Olsen et al., 2015) (see Chapter 58). Salient points are that keratino-
an intracellular cAMP-​signaling cascade, culminating in increased cyte cancers are more common among those living in areas of high
production of melanin pigments for distribution back to keratinocytes. solar irradiance, and residents of regions with low solar irradiance
254

254 PART III:  THE CAUSES OF CANCER


develop more lesions when they migrate to areas with higher ambient Finally, mouse models support observational human studies showing
UV radiation. Both BCC and SCC of the skin occur almost exclusively that sun exposure in early life has particularly adverse effects (Noonan
on sun-​exposed body sites among fair-​skinned populations and are fre- et al., 2001, 2012; Viros et al., 2014). Numerous epidemiologic studies
quent in patients with constitutional genetic deficiencies preventing have examined the possible association of UV with ocular melanoma,
the repair of UVB-​specific DNA mutations. Field trials have demon- and it appears that occupational sun exposure, especially farming and
strated that regular use of sunscreen reduces the incidence of actinic welding, is associated with higher risks of melanoma of the choroid
keratoses (AK) (Darlington et al., 2003; Thompson et al., 1993) and and ciliary body (Vajdic et al., 2002, 2004).
SCC tumors in humans (Green et al., 1999). Finally recent meta-​analy-
ses have shown that sunbed use significantly increases risks of BCC
and SCC (Wehner et al., 2012). Other Cancers
The era of high-​throughput genomics has revealed the full extent
of UV-​induced mutations for each of the common types of skin can- There has been speculation that exposure to UV radiation may indi-
cer. Sequencing studies have shown that the total mutational loads for rectly affect a person’s risk of developing cancers at sites other
BCC and SCC far exceed those of other common cancers, with up to than skin, but evidence from large prospective studies that have
75 mutations per megabase (Mb) of DNA for BCC and 35 per Mb for tested this hypothesis directly is scarce. The larger of two relevant
SCC (Jayaraman et al., 2014; South et al., 2014). Overwhelmingly, cohort studies, the National Institutes of Health (NIH)-​AARP Diet
the most prevalent mutations in the somatic genome of these tumors and Health Study, linked ambient erythemal UV exposure esti-
are the classic UVR signatures—​C to T substitutions at dipyrimidine mated from satellite Total Ozone Mapping Spectrometer data to
sites (Hodis et al., 2012; South et al., 2014). The key challenge, how- the Census 2000 record of baseline residence for 450,934 white,
ever, has been to separate the small number of critically important non-​Hispanic subjects and examined their cancer outcomes after
mutations that drive tumor development from the thousands of pas- 9  years of follow-​up using cancer registries. After accounting for
senger mutations scattered throughout the genome that are acquired confounding factors, ambient UV exposure was associated with sig-
as the tumor evolves. For SCC, driver mutations appear restricted nificant decreases in non-​Hodgkin’s lymphoma, colon, squamous
to a relatively small subset of genes:  NOTCH1, NOTCH2, TP53, cell lung, pleural, prostate, kidney, and bladder cancers (Lin et al.,
CDKN2A, NRAS, and KRAS (Durinck et al., 2011; South et al., 2014; 2012). On the other hand, a Scandinavian cohort study of 49,261
Wang et al., 2011); these mutations typically carry UVR signatures. women found no evidence of an association between any measure
Driver mutations appear similarly restricted to a small number of of personal cumulative UV exposure at ages 10–​39 and overall can-
genes for BCC (PTCH, TP53) (Jayaraman et  al., 2014). For both cer occurrence after 15 years follow-​up, though sunbathing at ages
types of keratinocyte cancers, other genes have been identified as 10–​29 was related to reduced breast cancer and cancer overall, ≥ 2
candidate drivers at lower frequency, and these await confirmation sunburns/​year was related to reduced lung cancer, and solarium use
in larger studies. was also inversely associated with breast cancer (Yang et al., 2011).
On balance, there is insufficient strong evidence available to draw
firm conclusions.
Melanoma
Cutaneous melanoma is a cancer of the pigment-​producing cells of the OPPORTUNITIES FOR PREVENTION
skin. The causal role of UV exposure is complex and is determined
Reducing Sun Exposure
by phenotypic, genotypic, and behavioral factors (Whiteman et  al.,
2011) (see Chapter 63). The evidence linking UV exposure with cuta- Many health promotion and health education strategies have been
neous melanoma is similar to that for keratinocyte cancer, namely that developed to change knowledge, attitudes, and behaviors to reduce
melanomas develop more commonly among fair-​skinned migrants to susceptible people’s sun exposure. Behavioral interventions aim to
high UV environments than among the population of origin (Whiteman improve personal sun protection through use of clothing, hats, sun-
et al., 2001); and sun-​exposed sites, such as the face and ears, are the screen, and sunglasses, staying in shade, and avoiding exposure during
most commonly affected (per unit area of skin), but intermittently peak UV hours. This section provides a broad overview of approaches
exposed sites, such as the trunk and proximal limbs, are also often to reduce exposure to UV radiation. A more detailed discussion of
affected (Green et al., 1993). Estimates of the population fraction attrib- practical issues regarding the implementation of these approaches is
utable to sun exposure range from 60% to 95% (Olsen et al., 2015). provided in Chapter 62.4.
UV-​ induced mutations are highly prevalent throughout the Preventive interventions can be delivered through a variety of
somatic genome of cutaneous melanomas (at around 14 per Mb), means including education, counseling by healthcare providers,
both as drivers and passengers, as described earlier (Hodis et  al., policy and environmental changes, and community-​wide engage-
2012). Key genes affected in melanoma include BRAF, NRAS, ment through media or social media campaigns (NICE, 2007), or
PTEN, TP53, CDKN2A, and MAP2K1 (Hodis et  al., 2012). Unlike by combining several of these approaches (Saraiya et  al., 2004).
SCC and BCC, however, not all driver mutations for melanoma have A  systematic review of the evidence for behavioral counseling to
the classical UVR signature. The most prevalent somatic mutation prevent skin cancer conducted for the US Preventive Services Tasks
in melanoma, occurring in around 50% of tumors, occurs in BRAF Force (Lin et  al., 2011)  resulted in recommendations to counsel
and involves a T to A transversion at position 1799 (resulting in the children, adolescents, and young adults aged 10–​24  years who
substitution of valine for glutamine at codon 600, hence the abbre- have fair skin to minimize UV exposure to reduce skin cancer risk
viation BRAFV600E). There is conjecture about whether UVR could be (Moyer, 2012), but claimed insufficient evidence to assess the bal-
responsible for such a mutation, since BRAFV600E mutations are also ance of benefits and harms of counseling adults over 24 about skin
commonly found in internal cancers (Davies et al., 2002). The most cancer prevention, despite several successful field trials in adults
favored explanation is that this pattern is consistent with oxidative (Green et  al., 2012). Another systematic review of the evidence
damage in the target cell, for which UVR is one such source (Denat for reducing UV radiation exposure among outdoor workers found
et al., 2014). Converging lines of evidence support this theory, since men more likely to wear hats and protective clothing and women
melanin (particularly pheomelanin) is a potent producer of oxy- more likely to use sunscreen, but found little evidence concerning
gen radicals when exposed to UVR (Napolitano et  al., 2014), and education and prevention policies in the outdoor workplace (Glanz
experimental studies indicate that UVA alone induces melanoma in et al., 2007).
pigmented but not albino mice through oxidative damage (Noonan Multiple-​level community-​wide interventions have been devel-
et al., 2012). It is also possible that pheomelanin induces melanoma oped by cancer agencies in many countries. Australia led the way
independently of sun exposure (Mitra et  al., 2012), although this from the 1980s with mass-​media campaigns to increase the public’s
remains to be established in human studies. knowledge about the link between sun exposure and skin cancer
 25

Ultraviolet Radiation 255


and to reduce high levels of recreational sun exposure. Beginning References
with the “Slip, Slop, Slap” Campaign with a logo that encouraged Abdel-​Malek ZA, and Cassidy P. 2015. Dark CPDs and photocarcinogen-
people to “slip on a shirt, slop on some sunscreen and slap on a esis: the party continues after the lights go out. Pigment Cell Melanoma
hat,” successive campaigns targeted specific groups, for example, Res, 28(4), 373–​374. PMID: 25950490.
adolescents, outdoor workers, and schoolchildren (Iannacone and Allen M, and McKenzie R. 2005. Enhanced UV exposure on a ski-​field com-
Green, 2014). These campaigns have been conducted predomi- pared with exposures at sea level. Photochem Photobiol Sci, 4(5), 429–​
nantly by state Cancer Councils under a “SunSmart” license, rein- 437. PMID: 15875076.
forced by policies to adopt supportive environments such as shade Archier E, Devaux S, Castela E, et al. 2012. Carcinogenic risks of psoralen UV-​
in schools and access to quality sun-​protection products (Iannacone A therapy and narrowband UV-​B therapy in chronic plaque psoriasis: a
systematic literature review. J Eur Acad Dermatol Venereol, 26 Suppl 3,
and Green, 2014). In New Zealand, campaigns have been similar in 22–​31. PMID: 22512677.
their history, with the New Zealand Cancer Society first conduct- Autier P, Dore JF, Breitbart E, et al. 2011. The indoor tanning industry’s double
ing campaigns with an emphasis on children, parents, and teachers, game. Lancet, 377(9774), 1299–​1301. PMID: 21324519.
and later on other sections of the community, informed by quali- Bais AF, McKenzie RL, Bernhard G, et al. 2015. Ozone depletion and climate
tative research (Watts, 2002). A  formal school accreditation pro- change: impacts on UV radiation. Photochem Photobiol Sci, 14(1), 19–​52.
gram for sun protection has had positive results (Reeder, 2012). PMID: 25380284.
Countries in the Northern Hemisphere with lower skin cancer rates Battistutta D, Pandeya N, Strutton GM, et  al. 2006. Skin surface topogra-
have also conducted comprehensive community-​based campaigns, phy grading is a valid measure of skin photoaging. Photodermatol
but with lower levels of intensity and investment of resources Photoimmunol Photomed, 22(1), 39–​45. PMID: 16436180.
Bolland MJ, Grey A, Gamble GD, and Reid IR. 2014. The effect of vitamin
(Hiom, 2010)  and often lacking national coordination (Lazovich D supplementation on skeletal, vascular, or cancer outcomes:  a trial
et al., 2012). For example, the SunWise campaign was launched in sequential meta-​analysis. Lancet Diabetes Endocrinol, 2(4), 307–​320.
2000 by the US Environmental Protection Agency, and its school PMID: 24703049.
program had modest early success (Geller et al., 2003). Economic Boniol M, Autier P, Boyle P, and Gandini S. 2012. Cutaneous melanoma attrib-
analysis indicated that if the SunWise school program continued utable to sunbed use: systematic review and meta-​analysis. Br Med J, 345,
to 2015 it should avert nearly 11,000 skin cancer cases and 960 e4757. PMCID: PMC3404185.
quality-​adjusted life-​years (undiscounted) among its participants Brash D, Rudolph JA, Simon JA, et al. 1991. A role for sunlight in skin can-
(Kyle et al., 2008). For every dollar invested in SunWise, between cer: UV-​induced p53 mutations in squamous cell carcinoma. Proc Natl
approximately $2 and $4 in medical care costs and productivity Acad Sci U S A, 88, 10124–​10128.
Brash DE. 2015. UV signature mutations. Photochem Photobiol, 91(1), 15–​26.
losses could be saved, depending on funding levels (Kyle et  al., PMCID: PMC4294947.
2008). In the United Kingdom, primary prevention efforts com- Byrne SN, Limon-​Flores AY, and Ullrich SE. 2008. Mast cell migration from
menced in the 1990s with the government-​supported national skin the skin to the draining lymph nodes upon ultraviolet irradiation rep-
cancer prevention campaign Sun Know How. Since 2003, Cancer resents a key step in the induction of immune suppression. J Immunol,
Research UK, commissioned by the UK Health Departments, has 180(7), 4648–​4655. PMCID: PMC2391302.
run the SunSmart campaign with varying levels of subsequent Cahoon EK, Wheeler DC, Kimlin MG, et al. 2013. Individual, environmental, and
funding (Hiom, 2010), supported by campaigns conducted by der- meteorological predictors of daily personal ultraviolet radiation exposure
matologists and other charities. measurements in a United States cohort study. PLoS ONE, 8(2), e54983.
Campbell C, Quinn A, Ro Y, Angus B, and Rees J. 1993. p53 mutations are
common and early evens that precede tumor invasion in squamous cell
neoplasia of the skin. J Invest Dermatol, 100, 746–​748.
Reducing Exposure to Artificial UV for Tanning Chandrasena A, Amin K, and Powell B. 2013. Dying for a tan:  a survey to
A review of the trends in legislation to restrict access to indoor tan- assess solarium adherence to world health organization guidelines in
Australia, New Zealand, and the United Kingdom. Eplasty, 13, e62.
ning found that the number of countries with such legislation increased PMCID: PMC3881211.
from two countries in 2003 to 11 countries in 2011 (Pawlak et  al., Chen H, Weng QY, and Fisher DE. 2014. UV signaling pathways within the
2012). Brazil has subsequently banned tanning salons (in 2009) and skin. J Invest Dermatol, 134(8), 2080–​2085. PMCID: PMC4102648.
Australia in 2015. Cockell CS, Scherer K, Horneck G, et al. 2001. Exposure of arctic field sci-
Legislated restrictions of the tanning industry have been fewer in entists to ultraviolet radiation evaluated using personal dosimeters.
the United States, but in 2014, based on the accumulated evidence Photochem Photobiol, 74(4), 570–​578.
of harmful health effects of indoor tanning for cosmetic reasons, Cooke MS, Harry EL, Liljendahl TS, and Segerback D. 2013. DNA nucleotide
the US Food and Drug Administration reclassified tanning lamps excision repair, where do all the cyclobutane pyrimidine dimers go? Cell
from a class I (low risk) medical device to a class II (moderate risk) Cycle, 12(10), 1642. PMCID: PMC3680545.
Damian DL, Matthews YJ, Phan TA, and Halliday GM. 2011. An action spec-
device, with the added requirement for black-​box labeling warn- trum for ultraviolet radiation-​induced immunosuppression in humans.
ing minors about the harms of using tanning lamps (Ernst et  al., Br J Dermatol, 164(3), 657–​659. PMID: 21375518.
2015). Also in 2014, the Surgeon General of the US Department of Darlington S, Williams G, Neale R, Frost C, and Green A. 2003. A randomized
Health and Human Services issued his “Call to Action to Prevent controlled trial to assess sunscreen application and beta carotene supplemen-
Skin Cancer,” which inter alia included the call to “reduce harms tation in the prevention of solar keratoses. Arch Dermatol, 139(4), 451–​455.
from indoor tanning” (US Department of Health & Human Services, Davies H, Bignell GR, Cox C, et  al. 2002. Mutations of the BRAF gene in
2014). It has been noted that some 44 US states now have some kind human cancer. Nature, 417(6892), 949–​954. PMID: 12068308.
of law or regulation related to indoor tanning, though with varying Denat L, Kadekaro AL, Marrot L, Leachman SA, and Abdel-​ Malek ZA.
strength of state laws and compliance enforcement, and some states 2014. Melanocytes as instigators and victims of oxidative stress. J Invest
Dermatol, 134(6), 1512–​1518. PMCID: PMC4418514.
have no laws related to indoor tanning (Pan and Geller, 2015; US Diffey BL. 1987. A comparison of dosimeters used for solar ultraviolet radiom-
Department of Health & Human Services, 2014). etry. Photochem Photobiol, 46(1), 55–​60. PMID: 3615634.
Diffey BL, Gibson CJ, Haylock R, and McKinlay AF. 1996. Outdoor ultra-
violet exposure of children and adolescents. Br J Dermatol, 134(6),
FUTURE RESEARCH 1030–​1034.
Diffey BL, and Gies HP. 1998. The confounding influence of sun exposure in
melanoma. Lancet, 351, 1101–​1102.
Use of new technologies, including social media, is now being Durinck S, Ho C, Wang NJ, et al. 2011. Temporal dissection of tumorigenesis in
researched to reduce deleterious sun exposure. Implementation and primary cancers. Cancer Discov, 1(2), 137–​143. PMCID: PMC3187561.
evaluation programs of behavioral interventions, especially among English DR, Armstrong BK, and Kricker A. 1998. Reproducibility of reported
men, adolescents, children, and their caregivers, are high research measurements of sun exposure in a case-​control study. Cancer Epidemiol
priorities. Biomarkers Prev, 7(10), 857–​863. PMID: 9796629.
256

256 PART III:  THE CAUSES OF CANCER


Ernst A, Grimm A, and Lim HW. 2015. Tanning lamps:  health effects and International Agency for Research on Cancer. 1992. IARC monographs on
reclassification by the Food and Drug Administration. J Am Acad the evaluation of carcinogenic risks to humans:  solar and ultraviolet
Dermatol, 72(1), 175–​180. PMID: 25458016. radiation. IARC Monogr Eval Carcinog Risks Hum, 55, 1–​316. PMID:
Euvrard S, Kanitakis J, and Claudy A. 2003. Skin cancers after organ transplan- 1345607.
tation. N Engl J Med, 348(17), 1681–​1691. PMID: 12711744. International Agency for Research on Cancer. 2012. IARC monographs on the
Frederick JE, Slusser JR, and Bigelow DS. 2000. Annual and interannual evaluation of carcinogenic risks to humans: radiation. IARC Monogr Eval
behavior of solar ultraviolet irradiance revealed by broadband measure- Carcinog Risks Hum, 100D, 1–​68.
ments. Photochem Photobiol, 72(4), 488–​496. PMID: 11045720. Jayaraman SS, Rayhan DJ, Hazany S, and Kolodney MS. 2014. Mutational
Fukunaga A, Khaskhely NM, Ma Y, et  al. 2010. Langerhans cells serve as landscape of basal cell carcinomas by whole-​exome sequencing. J Invest
immunoregulatory cells by activating NKT cells. J Immunol, 185(8), Dermatol, 134(1), 213–​220. PMID: 23774526.
4633–​4640. PMCID: PMC2950871. Jennings L, Karia PS, Jambusaria-​Pahlajani A, Whalen FM, and Schmults CD.
Garcia-​Borron JC, Abdel-​Malek Z, and Jimenez-​Cervantes C. 2014. MC1R, 2013. The Sun Exposure and Behaviour Inventory (SEBI): validation of
the cAMP pathway, and the response to solar UV:  extending the hori- an instrument to assess sun exposure and sun protective practices. J Eur
zon beyond pigmentation. Pigment Cell Melanoma Res, 27(5), 699–​720. Acad Dermatol Venereol, 27(6), 706–​715. PMID: 22487012.
PMCID: PMC4150834. Khazova M, O’Hagan JB, and Robertson S. 2015. Survey of UV emissions
Geller A, Rutsch L, Kenausis K, and Zhang Z. 2003. Evaluation of the SunWise from sunbeds in the UK. Photochem Photobiol, 91(3), 545–​552. PMID:
School Program. J Sch Nurs, 19(2), 93–​99. PMID: 12653621. 25626375.
Glanz K, Buller DB, and Saraiya M. 2007. Reducing ultraviolet radiation expo- Kimlin MG, Lucas RM, Harrison SL, et al. 2014. The contributions of solar ultra-
sure among outdoor workers: state of the evidence and recommendations. violet radiation exposure and other determinants to serum 25-​hydroxyvita-
Environ Health, 6, 22. PMCID: PMC1995198. min D concentrations in Australian adults: the AusD Study. Am J Epidemiol,
Godar DE. 2005. UV doses worldwide. Photochem Photobiol, 81(4), 736–​749. 179(7), 864–​874. PMID: 24573539.
PMID: 15819599. Kiss F, and Anstey AV. 2013. A review of UVB-​mediated photosensitivity dis-
Grange F, Mortier L, Crine A, et al. 2015. Prevalence of sunbed use, and char- orders. Photochem Photobiol Sci, 12(1), 37–​46. PMID: 23023766.
acteristics and knowledge of sunbed users: results from the French popu- Kotova N, Hemminki K, and Segerback D. 2005. Urinary thymidine dimer
lation-​based Edifice Melanoma survey. J Eur Acad Dermatol Venereol, 29 as a marker of total body burden of UV-​ inflicted DNA damage in
Suppl 2, 23–​30. PMID: 25639930. humans. Cancer Epidemiol Biomarkers Prev, 14(12), 2868–​2872. PMID:
Green A, Harwood C, Lear J, et  al. 2012. Skin cancer prevention:  recent 16365002.
evidence from randomized controlled trials. Curr Dermatol Rep, 1(3), Kyle JW, Hammitt JK, Lim HW, et  al. 2008. Economic evaluation of the
123–​130. US Environmental Protection Agency’s SunWise program:  sun protec-
Green A, MacLennan R, Youl P, and Martin N. 1993. Site distribution of cuta- tion education for young children. Pediatrics, 121(5), e1074–​ 1084.
neous melanoma in Queensland. Int J Cancer, 53, 232–​236. PMID: 18450850.
Green A, Siskind V, Bain C, and Alexander J. 1985. Sunburn and malignant Lake JR, Thomson CS, Twelves CJ, and Davies EA. 2014. A qualitative inves-
melanoma. Br J Cancer, 51, 393–​397. tigation of the motivations, experiences and views of female sunbed users
Green A, Williams G, Nèale R, et  al. 1999. Daily sunscreen application and under the age of 18 in England. J Public Health (Oxf), 36(1), 56–​64.
betacarotene supplementation in prevention of basal-​ cell and squa- PMID: 23365261.
mous-​cell carcinomas of the skin: a randomised controlled trial. Lancet, Lane DP. 1992. p53, guardian of the genome. Nature, 359, 486–​487.
354(9180), 723–​729. Lazovich D, Choi K, and Vogel RI. 2012. Time to get serious about skin can-
Green AC, and Whiteman DC. 2006. Solar radiation. In: Schottenfeld Joseph cer prevention. Cancer Epidemiol Biomarkers Prev, 21(11), 1893–​1901.
F. Fraumeni, Jr. (ed.), Cancer Epidemiology and Prevention (3rd ed., PMID: 22962407.
pp. 294–​305). New York: Oxford University Press. Liljendahl TS, Blomqvist A, Andersson EM, Barregard L, and Segerback D.
Halliday GM, and Cadet J. 2012. It’s all about position:  the basal layer of 2013. Urinary levels of thymine dimer as a biomarker of exposure to
human epidermis is particularly susceptible to different types of sun- ultraviolet radiation in humans during outdoor activities in the summer.
light-​
induced DNA damage. J Invest Dermatol, 132(2), 265–​ 267. Mutagenesis, 28(3), 249–​256. PMID: 23339196.
PMID: 22241442. Liljendahl TS, Kotova N, and Segerback D. 2012. Quantification of ultravio-
Halliday GM, Damian DL, Rana S, and Byrne SN. 2012. The suppressive let radiation-​induced DNA damage in the urine of Swedish adults and
effects of ultraviolet radiation on immunity in the skin and internal children following exposure to sunlight. Biomarkers, 17(7), 634–​641.
organs: implications for autoimmunity. J Dermatol Sci, 66(3), 176–​182. PMID: 22889379.
PMID: 22277701. Lin JS, Eder M, and Weinmann S. 2011. Behavioral counseling to prevent skin
Han J, Colditz GA, Liu JS, and Hunter DJ. 2005. Genetic variation in XPD, sun cancer: a systematic review for the U.S. Preventive Services Task Force.
exposure, and risk of skin cancer. Cancer Epidemiol Biomarkers Prev, Ann Intern Med, 154(3), 190–​201.
14(6), 1539–​1544. PMID: 15941969. Lin SW, Wheeler DC, Park Y et al. 2012. Prospective study of ultraviolet radia-
Hart PH, Gorman S, and Finlay-​Jones JJ. 2011. Modulation of the immune tion exposure and risk of cancer in the United States. Int J Cancer, 131(6),
system by UV radiation: more than just the effects of vitamin D? Nat Rev E1015–​1023. PMCID: PMC3402606.
Immunol, 11(9), 584–​596. PMID: 21852793. Manson JE, and Bassuk SS. 2015. Vitamin D research and clinical practice: at a
Henderson S, Javorniczky J, and Gies P. 2010. Spectral measurements of solar crossroads. JAMA, 313(13), 1311–​1312. PMID: 25695911.
UV at several altitudes under Australian conditions. Paper presented at Marteijn JA, Lans H, Vermeulen W, and Hoeijmakers JH. 2014. Understanding
the 2010 UV Workshop, Wellington, New Zealand. https://​www.niwa. nucleotide excision repair and its roles in cancer and ageing. Nat Rev Mol
co.nz/​sites/​niwa.co.nz/​files/​spectral_​measurements_​of_​uv.pdf. Cell Biol, 15(7), 465–​481. PMID: 24954209.
Heydenreich J, and Wulf HC. 2005. Miniature personal electronic UVR dosim- Martincorena I, Roshan A, Gerstung M, et  al. 2015. Tumor evolution:  high
eter with erythema response and time-​stamped readings in a wristwatch. burden and pervasive positive selection of somatic mutations in normal
Photochem Photobiol, 81(5), 1138–​1144. PMID: 15850424. human skin. Science, 348(6237), 880–​886. PMCID: PMC4471149.
Hiom S, Naumann L, Cerny C. 2010. CRUK Expert paper 2: National cam- Millen AE, Wactawski-​ Wende J, Pettinger M, Melamed ML, Tylavsky
paigns (UK and worldwide). Cancer Research UK. FA, Liu S, Robbins J, LaCroix AZ, LeBoff MS, Jackson RD. 2010.
Hodis E, Watson IR, Kryukov GV, et al. 2012. A landscape of driver mutations Predictors of serum 25-​hydroxyvitamin D concentrations among post-
in melanoma. Cell, 150(2), 251–​263. PMCID: PMC3600117. menopausal women:  The Women's Health Initiative Calcium plus
Hooke R, Pearson A, and O’Hagan J. 2014. Autonomous portable solar ultra- Vitamin D clinical trial. The American Journal of Clinical Nutrition.
violet spectroradiometer (APSUS): a new CCD spectrometer system for 91(5):1324–​1235.
localized, real-​time solar ultraviolet (280–​400  nm) radiation measure- Mitra D, Luo X, Morgan A, et al. 2012. An ultraviolet-​radiation-​independent
ment. Photochem Photobiol, 90(4), 903–​910. PMID: 24483759. pathway to melanoma carcinogenesis in the red hair/​fair skin background.
Huber M, Blumthaler M, Schreder J, Bais A, and Topaloglou C. 2002. Effect Nature, 491(7424), 449–​453. PMCID: PMC3521494.
of ambient temperature on Robertson-​berger-​type erythemal dosimeters. Mocellin S, Verdi D, and Nitti D. 2009. DNA repair gene polymorphisms and
Applied Optics, 41(21), 4273–​4277. risk of cutaneous melanoma:  a systematic review and meta-​analysis.
Iannacone MR, and Green AC. 2014. Towards skin cancer prevention and early Carcinogenesis, 30(10), 1735–​1743. PMID: 19706646.
detection:  evolution of skin cancer awareness campaigns in Australia. Moehrle M, Dennenmoser B, and Garbe C. 2003. Continuous long-​term
Melanoma Manage, 1(1), 75–​84. monitoring of UV radiation in professional mountain guides reveals
Ikehata H, and Ono T. 2011. The mechanisms of UV mutagenesis. J Radiat extremely high exposure. Int J Cancer, 103(6), 775–​ 778. PMID:
Res, 52(2), 115–​125. PMID: 21436607. 12516097.
 257

Ultraviolet Radiation 257


Morze CJ, Olsen CM, Perry SL, et al. 2012. Good test-​retest reproducibility South AP, Purdie KJ, Watt SA, et al. 2014. NOTCH1 mutations occur early dur-
for an instrument to capture self-​reported melanoma risk factors. J Clin ing cutaneous squamous cell carcinogenesis. J Invest Dermatol, 134(10),
Epidemiol, 65(12), 1329–​1336. PMID: 22974496. 2630–​2638. PMID: 24662767.
Mouret S, Baudouin C, Charveron M, et  al. 2006. Cyclobutane pyrimidine Sun J, Lucas RM, Harrison SL, et al. 2014. Measuring exposure to solar ultra-
dimers are predominant DNA lesions in whole human skin exposed violet radiation using a dosimetric technique: understanding participant
to UVA radiation. Proc Natl Acad Sci U S A, 103(37), 13765–​13770. compliance issues. Photochem Photobiol, 90(4), 919–​ 924. PMID:
PMCID: PMC1564232. 24571445.
Moyer VA. 2012. Behavioral counseling to prevent skin cancer: U.S. Preventive Swope V, Alexander C, Starner R, et  al. 2014. Significance of the melano-
Services Task Force recommendation statement. Ann Intern Med, 157(1), cortin 1 receptor in the DNA damage response of human melanocytes
59–​65. PMID: 22751761. to ultraviolet radiation. Pigment Cell Melanoma Res, 27(4), 601–​610.
Munakata N, Ono M, and Watanabe S. 1998. Monitoring of solar-​UV exposure PMID: 24730569.
among schoolchildren in five Japanese cities using spore dosimeter and Tewari A, Sarkany RP, and Young AR. 2012. UVA1 induces cyclobutane pyrim-
UV-​coloring labels. Jpn J Cancer Res, 89(3), 235–​245. idine dimers but not 6–​4 photoproducts in human skin in vivo. J Invest
Napolitano A, Panzella L, Monfrecola G, and d’Ischia M. 2014. Pheomelanin-​ Dermatol, 132(2), 394–​400. PMID: 21975824.
induced oxidative stress:  bright and dark chemistry bridging red hair Thieden E. 2008. Sun exposure behaviour among subgroups of the
phenotype and melanoma. Pigment Cell Melanoma Res, 27(5), 721–​733. Danish population. Based on personal electronic UVR dosimetry
PMID: 24814217. and corresponding exposure diaries. Dan Med Bull, 55(1), 47–​68.
National Institute for Health and Clinical Excellence. NICE public health guid- PMID: 18321444.
ance 6: Behaviour change at population, community and individual lev- Thieden E, Agren MS, and Wulf HC. 2000. The wrist is a reliable body site for
els. London; 2007. personal dosimetry of ultraviolet radiation. Photodermatol Photoimmunol
Nilsen LT, Aalerud TN, Hannevik M, and Veierod MB. 2011. UVB and UVA Photomed, 16(2), 57–​61. PMID: 10823313.
irradiances from indoor tanning devices. Photochem Photobiol Sci, 10(7), Thieden E, Heydenreich J, Philipsen PA, and Wulf HC. 2013. People maintain
1129–​1136. PMID: 21445424. their sun exposure behaviour in a 5-​7-​year follow-​up study using personal
Noonan FP, Recio JA, Takayama H, et al. 2001. Neonatal sunburn and mela- electronic UVR dosimeters. Photochem Photobiol Sci, 12(1), 111–​116.
noma in mice. Nature, 413(6853), 271–​272. PMID: 11565020. PMID: 22786662.
Noonan FP, Zaidi MR, Wolnicka-​Glubisz A, et al. 2012. Melanoma induction Thieden E, Philipsen PA, Heydenreich J, and Wulf HC. 2004a. UV radiation
by ultraviolet A but not ultraviolet B radiation requires melanin pigment. exposure related to age, sex, occupation, and sun behavior based on time-​
Nat Commun, 3, 884. PMCID: PMC3621412. stamped personal dosimeter readings. Arch Dermatol, 140(2), 197–​203.
Olsen CM, Wilson LF, Green AC, et al. 2015. Cancers in Australia attribut- PMID: 14967793.
able to exposure to solar ultraviolet radiation and prevented by regu- Thieden E, Philipsen PA, Sandby-​Moller J, Heydenreich J, and Wulf HC.
lar sunscreen use. Aust N Z J Public Health, 39(5), 471–​476. PMCID: 2004b. Proportion of lifetime UV dose received by children, teenagers
PMC4606762. and adults based on time-​stamped personal dosimetry. J Invest Dermatol,
Pan M, and Geller L. 2015. Update on indoor tanning legislation in the United 123(6), 1147–​1150. PMID: 15610527.
States. Clin Dermatol, 33(3), 387–​392. PMID: 25889141. Thieden E, Philipsen PA, and Wulf HC. 2006. Compliance and data reliability
Pawlak MT, Bui M, Amir M, et  al. 2012. Legislation restricting access to in sun exposure studies with diaries and personal, electronic UV dosim-
indoor tanning throughout the world. Arch Dermatol, 148(9), 1006–​1012. eters. Photodermatol Photoimmunol Photomed, 22(2), 93–​99. PMID:
PMID: 22801924. 16606414.
Pearson AJ, Dean SF, Clark IES et al. 2000. NRPB Solar ultraviolet radiation Thompson SC, Jolley D, and Marks R. 1993. Reduction of solar keratoses
measurement network. Radiat Prot Dosim, 91(1–​3), 169–​172. by regular sunscreen use. N Engl J Med, 329(16), 1147–​1151. PMID:
Petersen B, Wulf HC, Triguero-​Mas M, et  al. 2014. Sun and ski holidays 8377777.
improve vitamin D status, but are associated with high levels of DNA US Department of Health & Human Services. 2014. Reports of the Surgeon
damage. J Invest Dermatol, 134(11), 2806–​2813. PMID: 24844860. General. Washington, DC: Office of the Surgeon General.
Pfeifer GP, and Besaratinia A. 2012. UV wavelength-​dependent DNA dam- Vajdic CM, Kricker A, Giblin M, et  al. 2002. Sun exposure predicts risk of
age and human non-​melanoma and melanoma skin cancer. Photochem ocular melanoma in Australia. Int J Cancer, 101(2), 175–​182.
Photobiol Sci, 11(1), 90–​97. PMCID: PMC3289542. Vajdic CM, Kricker A, Giblin M, et  al. 2004. Artificial ultraviolet radiation
Premi S, Wallisch S, Mano CM, et al. 2015. Photochemistry: chemiexcitation and ocular melanoma in Australia. Int J Cancer, 112(5), 896–​900. PMID:
of melanin derivatives induces DNA photoproducts long after UV expo- 15386378.
sure. Science, 347(6224), 842–​847. PMCID: PMC4432913. Vajdic CM, and van Leeuwen MT. 2009. Cancer incidence and risk factors after
Quintern LE, Horneck G, Eschweiler U, and Bucker H. 1992. A biofilm usedas solid organ transplantation. Int J Cancer, 125(8), 1747–​1754.
ultra-​violet dosimeter. Photochem Photobiol, 55, 389–​395. van der Mei IA, Blizzard L, Ponsonby AL, and Dwyer T. 2006. Validity and
Rady P, Scinicariello F, Wagner RF, Jr, and Tyring SK. 1992. p53 mutations in reliability of adult recall of past sun exposure in a case-​control study of
basal cell carcinomas. Cancer Res, 52, 3804–​3806. multiple sclerosis. Cancer Epidemiol Biomarkers Prev, 15(8), 1538–​1544.
Rana S, Byrne SN, MacDonald LJ, Chan CY, and Halliday GM. 2008. PMID: 16896046.
Ultraviolet B suppresses immunity by inhibiting effector and memory Veierod MB, Parr CL, Lund E, and Hjartaker A. 2008. Reproducibility of self-​
T cells. Am J Pathol, 172(4), 993–​1004. PMCID: PMC2276424. reported melanoma risk factors in a large cohort study of Norwegian
Reeder AI, Jopson JA, Gray A. 2012. Sun protection policies and practices in women. Melanoma Res, 18(1), 1–​9. PMID: 18227701.
New Zealand primary schools. N Z Med J, 125(1349), 70–​82. Vermeulen W, and Fousteri M. 2013. Mammalian transcription-​coupled exci-
Robinson S, Dixon S, August S, et al. 2010. Protection against UVR involves sion repair. Cold Spring Harb Perspect Biol, 5(8), a012625. PMID:
MC1R-​mediated non-​pigmentary and pigmentary mechanisms in vivo. 23906714.
J Invest Dermatol, 130(7), 1904–​1913. PMID: 20237490. Viros A, Sanchez-​Laorden B, Pedersen M et  al. 2014. Ultraviolet radiation
Rowland FS. 2006. Stratospheric ozone depletion. Phil Trans R Soc B, accelerates BRAF-​driven melanomagenesis by targeting TP53. Nature,
361(1469), 769–​790. PMID: PMC1609402. 511(7510), 478–​482. PMCID: PMC4112218.
Roy CR, Gies HP, and Toomey S. 1997. Monitoring UV-​B at the earth’s sur- Wang NJ, Sanborn Z, Arnett KL, et  al. 2011. Loss-​of-​function mutations in
face. Austral Meteorol Mag, 46, 203–​210. Notch receptors in cutaneous and lung squamous cell carcinoma. Proc
Ruczinski I, Jorgensen TJ, Shugart YY et al. 2012. A population-​based study Natl Acad Sci U S A, 108(43), 17761–​17766. PMCID: PMC3203814.
of DNA repair gene variants in relation to non-​melanoma skin cancer as a Watts C, Reeder AI, Glasgow H. 2002. A cover-​up story: the Cancer Society
marker of a cancer-​prone phenotype. Carcinogenesis, 33(9), 1692–​1698. Melanoma Prevention programme. In: McKenzie RRA, Watts C (eds.),
PMCID: PMC3514896. UV Radiation and Its Effects: An Update 2002 (pp. 1–​3). Christchurch:
Saraiya M, Glanz K, Briss PA, et al. 2004. Interventions to prevent skin cancer Royal Society of New Zealand.
by reducing exposure to ultraviolet radiation: a systematic review. Am J Weatherhead EC, and Andersen SB. 2006. The search for signs of recovery of
Prev Med, 27(5), 422–​466. the ozone layer. Nature, 441(7089), 39–​45. PMID: 16672963.
Schwarz T, and Schwarz A. 2011. Molecular mechanisms of ultraviolet radia- Wehner MR, Chren MM, Nameth D, et al. 2014. International prevalence of
tion-​induced immunosuppression. Eur J Cell Biol, 90(6–​7), 560–​564. indoor tanning: a systematic review and meta-​analysis. JAMA Dermatol,
PMID: 21035896. 150(4), 390–​400. PMCID: PMC4117411.
Seckmeyer G, Klingebiel M, Riechelmann S, et al. 2012. A critical assessment Wehner MR, Shive ML, Chren MM, et  al. 2012. Indoor tanning and non-​
of two types of personal UV dosimeters. Photochem Photobiol, 88(1), melanoma skin cancer: systematic review and meta-​analysis. BMJ, 345,
215–​222. PMID: 22017203. e5909. PMCID: PMC3462818.
258

258 PART III:  THE CAUSES OF CANCER


Weihs P, Schmalwieser A, Reinisch C, et al. 2013. Measurements of personal Williamson CE, Zepp RG, Lucas RM et al. 2014. Solar ultraviolet radiation in
UV exposure on different parts of the body during various activities. a changing climate. Nature Clim Change, 4(6), 434–​441.
Photochem Photobiol, 89(4), 1004–​1007. PMID: 23587050. Xiang F, Harrison S, Nowak M et al. 2015. Weekend personal ultraviolet radia-
Whiteman D, and Green A. 1994. Melanoma and sunburn. Cancer Causes tion exposure in four cities in Australia: influence of temperature, humid-
Control, 5, 564–​572. ity and ambient ultraviolet radiation. J Photochem Photobiol B, 143,
Whiteman DC, Pavan WJ, and Bastian BC. 2011. The melanomas: a synthesis 74-​81. PMID: 25600266.
of epidemiological, clinical, histopathological, genetic, and biological Yang L, Veierod MB, Lof M et al. 2011. Prospective study of UV exposure and
aspects, supporting distinct subtypes, causal pathways, and cells of origin. cancer incidence among Swedish women. Cancer Epidemiol Biomarkers
Pigment Cell Melanoma Res, 24(5), 879–​897. Prev, 20(7), 1358–​1367. PMID: 21551241.
Whiteman DC, Whiteman CA, and Green AC. 2001. Childhood sun exposure Zaratti F, Piacentini RD, Guillen HA et al. 2014. Proposal for a modification
as a risk factor for melanoma: a systematic review of epidemiologic stud- of the UVI risk scale. Photochem Photobiol Sci, 13(7), 980–​985. PMID:
ies. Cancer Causes Control, 12, 69–​82. 24728468.
 259

15 Electromagnetic Fields

MARIA FEYCHTING AND JOACHIM SCHÜZ

OVERVIEW energy is deposited and can cause heating (cf. microwaves). Heating
of tissue is the established mechanism of interaction with the human
This chapter focuses on the carcinogenic effects of exposure to elec- body at these frequencies, and current exposure guidelines protect
tromagnetic fields, mainly power frequency and radiofrequency fields/​ against excessive heating of tissue, at both localized and whole body
microwaves. It discusses current knowledge on interaction mecha- exposures (ICNIRP, 1998; IEEE, 2005). The energy absorption inside
nisms of relevance for carcinogenicity, describes exposure sources, the body is estimated as the specific absorption rate (SAR), expressed
and reviews the available epidemiological studies. The chapter com- in watts per kilogram (W/​kg). It is, however, not possible to directly
ments on the strengths and weaknesses of the data available, and dis- measure tissue heating inside the body; therefore SAR values are esti-
cusses sources of bias of concern for the interpretation of the findings. mated through measurements in phantoms and theoretical modeling
Based on current evidence, electromagnetic fields are not regarded as (AGNIR, 2012; IARC, 2013). Use of a mobile phone is an example of
carcinogenic to humans, but there are open scientific questions that localized near-​field exposure, and energy is absorbed to a depth of a
merit further research, as will be outlined. few centimeters within the head.

Potential Mechanisms of Carcinogenicity


INTRODUCTION
Despite considerable research efforts, there is currently no known bio-
Electromagnetic fields in the non-​ionizing radiation spectrum are an logical mechanism for a carcinogenic effect of electromagnetic fields
integrated part of modern life, with an ever-​increasing number of at the exposure levels encountered in the general population or in
applications. The properties of electromagnetic fields and their inter- occupational settings. In vitro studies of ELF fields indicate genotoxic
action with the human body are determined by their frequency and effects at exposure levels of 100 µT or higher, which is far above what
wavelength, with the wavelength inversely related to the frequency. is encountered in the human environment for longer than short dura-
Electromagnetic fields generate energy, which is directly proportional tions, while in vivo studies do not provide evidence that ELF fields
to the frequency. In the non-​ionizing part of the electromagnetic spec- cause tumors or enhance tumor growth (SCENIHR, 2015). It is impor-
trum (Figure 15–1), the generated energy is too weak to break chemi- tant to note, however, that an appropriate animal model for childhood
cal bonds, and ions cannot be formed. The low frequency end of the leukemia has not previously been available, and the recent develop-
spectrum includes static fields (0 Hz) and fields related to the pro- ment of such a model (Hauer et al., 2014; Martin-​Lorenzo et al., 2015)
duction, transmission, and use of electricity (usually at 50–​60 Hz). has not yet provided new evidence.
At higher frequencies within the non-​ionizing spectrum are radiofre- For RF fields, studies in cells do not consistently show effects on
quency fields (RF) and microwaves, including applications such as genotoxicity, cell transformation, or cell proliferation, and occasional
radio and television, mobile telephony, radar, and microwave heating. positive findings have not been replicated in independent studies.
Further up the frequency range are infrared light, visible light, and UV Similarly, studies in experimental animals have not shown compelling
radiation. This chapter focuses on the frequencies from above 0 Hz to evidence of a carcinogenic effect, including several long-​term rodent
300 GHz, mainly power frequency and RF fields/​microwaves, as this bioassays, studies in transgenic animals, and studies of co-​carcino-
has been the focus of the research on potential carcinogenic effects of genic effects (AGNIR, 2012; SCENIHR, 2015). Independent studies
electromagnetic fields during the past decades. attempting to replicate a few positive findings for RF field exposure,
Extremely low frequency fields (ELF) have a very long wavelength such as an increased lymphoma incidence in the transgenic Eμ-​Pim1
(6000 km at 50 Hz), and electric and magnetic fields are separated. mouse model (Repacholi et al., 1997), have not been able to confirm
The electric field strength is measured in volts per meter (V/​m), while the original findings. One exception is a recent replication of an earlier
magnetic fields are usually measured as the magnetic flux density in pilot study of a co-​carcinogenic effect of RF fields in mice exposed in
tesla (T), and in the human environment usually in microtesla (µT). At utero to ethylnitrosourea (ENU) (Tillmann et al., 2010). The replica-
these low frequencies the energy is too low to cause heating, and the tion study found an increased occurrence of tumors in mice exposed in
magnetic fields pass through the body. The established mechanism of utero to ENU, and to one of four RF exposure levels (sham, 0.04, 0.4,
interaction between ELF fields and the human body is the induction of or 2 W/​kg), but no evidence of dose–​response pattern (Lerchl et al.,
electric currents. Induced electric currents may cause acute neurologi- 2015). Tumor types with increased occurrence in the replication study
cal effects, and current exposure guidelines are set to protect against were lung, liver, and lymphoma, while the original study found an
these acute effects. The research on the potential carcinogenicity of increase in lung tumors. Discovery of a Helicobacter hepaticus infec-
ELF magnetic fields has focused on exposures that are orders of mag- tion among the mice made results for liver tumors uninterpretable
nitude below the current guidelines (ICNIRP, 2010; IEEE, 2002). (Tillmann et al., 2010). None of the studies found increases in brain
Radiofrequency fields associated with mobile phone technology tumors. Replication of these findings is warranted.
have wavelengths of a few centimeters, depending on the frequency The largest and most comprehensive animal study within this area is
used for communication. They are characterized by their power den- in the process of being published, and was conducted by US National
sity (i.e., the electromagnetic energy per unit area), which is measured Toxicology Program (National Toxicology Program, 2014). Selected
as watts per meter squared (W/​m2). Except for near-​field exposure results have just been made available as a partial report, reporting
(i.e., very close to the source), there is a direct relation between the increased occurrence of two tumor types in male rats (mainly schwan-
power density, the electric field (V/​m), and the magnetic field (A/​m), nomas of the heart and, somewhat weaker, gliomas in the brain), at
and measurement of either the electric or magnetic field is sufficient exposures of 1.5, 3, and 6 W/​kg compared to non-​exposed sham con-
to estimate the other quantities (AGNIR, 2012). At these frequencies, trols. The partial report indicates that no positive findings were seen

259
260

260 PART III:  THE CAUSES OF CANCER

Power lines and Visible


electric appliances light
AM TV
radio FM radio Radar Infrared UV-light Gamma rays
X-rays

10 103 105 107 109 1011 1013 1015 1017 1019 1021
Hz

0 100 104 106 108 1010 1012 1014 1016 1018 1020 1022

Low frequency Radio waves Micro waves

Non-ionizing radiation Cellular Telephones Ionizing radiation


Figure 15–1.  The electromagnetic spectrum. (900–2100 Mhz)

for female rats, or male and female mice, or for any other tumor type in residential exposures exceeding 0.2 µT (Maslanyj et al., 2009), and the
any kind of animal. In addition, survival was significantly lower in the exposure prevalence of averages > 0.5 µT is below 1%.
sham exposed rats, and tumor occurrence was lower than observed his- ELF electric fields are produced wherever there is electric potential,
torically in sham exposed rats. While a full assessment of the implica- and they are directly related to the voltage, irrespective of whether any
tions from this major study has to await the publication of the complete current flows. Hence, high exposures are experienced mainly in occu-
set of results, the already shared findings may renew interest in look- pational settings or in the vicinity of high-​voltage installations, such
ing into mechanistic pathways. Given the inconsistency across animal as overhead power lines or substations. In contrast to magnetic fields,
sexes and species, the high exposure levels and duration compared to which are not perturbed by most materials (and especially not by tis-
common exposures in humans, the main finding in a rare cancer type, sue and therefore penetrating the body), electric fields are shielded
and the lower survival in sham exposed controls, limits the applicabil- by most materials. This is why it is extremely difficult to measure or
ity of the findings to what this means for real life exposures in humans. predict electric field exposures. Distance to the source alone is not a
The available epidemiological evidence on potential cancer risks good surrogate measure, as other buildings or trees may be between
associated with exposure to ELF and RF fields, respectively, is sum- the source and the home and may distort the electric field. Normally,
marized in the two following sections. electric field levels in homes are a few V/​m, but can become as high
as several kV/​m in homes in close vicinity and with an unobstructed
path to high-​voltage power lines and in certain occupational situations.
EXTREMELY LOW FREQUENCY MAGNETIC Most epidemiological studies have investigated magnetic field
AND ELECTRIC FIELDS exposures, and here either residential or occupational exposures. After
approximately 40 years of epidemiological research on cancer effects
First, notably, it must be stressed that there is no “zero” exposure to of ELF electric and magnetic field exposures, most notable evidence is
ELF magnetic fields; due to the electrification of the modern world, an association between residential ELF magnetic fields and a modestly
everyone is exposed to ELF fields to various extents, with typical daily increased risk of leukemia in children (about 1.5-​to 2-​fold), observed
average background levels of less than 0.05 µT but possibly slightly in a large number of studies with some degree of consistency. For other
higher in urban areas. Sources of exposures to ELF magnetic fields exposure and cancer outcome combinations, there are either fewer
above background levels can be roughly subdivided into three types. studies, rather exploratory studies, or the studies are inconsistent or
Exposures exceeding 0.2 µT are related to either the transmission or mostly showing no association. Therefore the remainder of this section
distribution of power (fields from high-​voltage power lines, lower-​ provides a detailed review of the childhood leukemia evidence and
voltage distribution lines, underground cables, transformers, indoor only brief summaries of the other evidence.
wiring) (type 1:  residential exposures), the use of electrical devices
(such as hair dryer, vacuum cleaner) (type 2:  exposure from use of
electrical appliances), or working in electrical occupations (such as
electricians, welders) (type 3:  occupational exposures). Magnetic Exposure Assessment
fields are produced by the flow of the current; thus exposure is deter- Exposure assessment of ELF magnetic fields is challenging. Various
mined by the current and distance to the source. Electric devices may methods have been used to assess residential exposures. Residential
emit magnetic fields of several mT in their immediate vicinity, but exposures are used as surrogates of the individual’s exposure, assum-
only during use; therefore they are sources of high but intermittent ing that total exposure is mostly determined by exposure at home. This
very short-​term exposure. In certain occupations, when working close has shown to be reliable in especially young children given how much
to electric motors, for example, average exposures of 1–​10 µT are pos- time they spend at home, but may lead to some exposure misclassi-
sible, and exposure may span over the whole working day. Residential fication in adults due to exposures and time spent at the workplace
exposures are usually at background level, but if elevated, then they and elsewhere (Forssen et  al., 2002). Personal monitors, carried by
result in rather continuous exposure. Magnetic fields of > 0.2 µT to the individual, which constantly record magnetic field levels and pro-
a few µT may be reached within distances of less than 50–​200 m to vide the most accurate estimate of contemporary exposure, do exist.
nearby high-​voltage power lines (> 200 kV, depending on their power However, studies of residential exposures are almost exclusively case-​
load), distances of less than 20–​50 m to medium-​voltage power lines control studies requiring an accurate estimate of past exposures. This
or other installations (> 10 kV), less than 5–​10 m to low-​voltage power is better achieved by residential measurements, as exposure tends to
lines, their roof connections to buildings, or to transformers (< 1 kV), be more stable over time in cases of elevated field levels, while per-
or in homes with unbalanced currents in indoor wiring or by current sonal monitoring is more influenced by personal behavior. A person’s
produced by ELF fields on closed metallic loops. Even taken together, behavior is likely to change over time, especially when having a cancer
however, these exposure situations are rather uncommon. It is esti- diagnosis, but also by the aging of the subject, so that contemporary
mated that only a few percent (1%–​3%) of residences in Europe and exposure is not a good predictor of past exposures. Exposure assess-
potentially slightly more (1%–​10%) in North America have average ment methods and their features are summarized in Table 15–1.
 261

Electromagnetic Fields 261
Table 15–1.  Main Exposure Assessment Methods Used in Epidemiological Studies of Residential Exposures to Extremely Low-​Frequency Magnetic Fields

Method Description Advantages/​Disadvantages

Personal dosimetry Device carried by study participant over 1 or several Most precise picture of magnetic field exposure of the individual, especially
days, recording the magnetic field level several times when combined with activity diary; rarely used in cancer epidemiology, as
per minute too expensive for large cohorts and limited use in case-​control studies, as
activity patterns change when having cancer and by the aging of subjects
(especially in children) so that contemporary exposure is not a good
predictor of past exposures.
Long-​term stationary Device placed in study participant’s home over 1 or Most precise picture of magnetic field exposure in the home environment;
measurement several days, recording the magnetic field level surveys show good correlation in repeated measurements so that method
several times per minute; either used stationary in appears to be a good surrogate of exposure in retrospective studies; good
the bedroom or at several places in the home for exposure surrogate in children spending most of their time at home; in
calculation of time-​weighted average according adults, time spent at other places or occupational exposures may influence
to how much time the participant spends at each the individual exposure more than the home measurement.
measurement location
Spot measurements As above, but measurements taken only for usually May be as accurate as longer term measurements with stable magnetic
several minutes up to a few hours, at several locations field conditions, but much lesser so when fields vary strongly over time;
within the home but sometimes only outside the especially outdoor measurements have shown rather poor correlation in
residence repeated measurements.
Calculated fields Estimation of magnetic field levels from overhead Accurate retrospective estimation of long-​term magnetic field exposure in
high-​voltage power lines based on distance from line the home environment caused by high-​voltage power lines; neglects other
to home, historic power load of the line, and other line sources of magnetic field exposures in the home environment and when
characteristics not at home; can only be estimated when the respective historic power load
data are available, which is often not the case (used almost exclusively in
the Nordic countries and the UK).
Wire code Estimation of magnetic field levels from overhead power Can be used for retrospective assessment; surveys show some variability on
lines and other electrical installations similar to the how well the method predicts actual magnetic field exposures in the home
preceding method, but rather based on characteristics environment depending on the local conditions, which makes it difficult to
of the source and no data on power load judge how well the method worked in the individual studies; neglects other
sources of magnetic field exposures in the home environment and when not
at home; mostly used in North America.
Distance to power line Measuring or estimating the distance between the home Weakest surrogate of magnetic field exposure; works well in very close
and nearby overhead high-​voltage power lines or other proximity (50 m) to the power lines in countries where power lines are
electrical installations usually run with high power load, but leads to gross misclassification of
exposure in the home environment otherwise; may also be surrogate for
other living conditions and therefore raises issue of confounding.

Exposure assessment methods for occupational settings range from different voltages (Wertheimer and Leeper, 1979). At present, more
categorizations of job titles, to expert ratings of job-​related tasks (Hug than 30 epidemiological studies have investigated this topic. Most of
et al., 2010), to the development of more complex quantitative job-​expo- these were conducted in the late 1980s to early 2000s, and used vari-
sure matrices (JEMs) based on measurement surveys (Bowman et al., able sample sizes and design features. As the studies differ greatly in
2007). While the latter is believed to be the most reliable approach, exposure assessment, exposure indices, and exposure categorizations,
exposure misclassification remains a problem, due to the large exposure as well as analytical strategies, direct comparison is difficult and may
variation within jobs and the usually relatively small number of available even be misleading.
measurements per job category (Gobba et al., 2011; Greenland et al., Therefore, the most informative evidence comes from systematic
2016). Exposure from electrical appliances is usually assessed through reviews or meta-​ analyses. Authoritative hazard identifications or
interviews, with self-​reported type and frequency of use of devices, and health risk assessments were prepared by the International Agency
distance to the source (e.g., for television), and exposure estimates are for Research on Cancer (IARC) program on the evaluation of car-
therefore prone to both random and systematic error. Electric field expo- cinogenic risks to humans, the World Health Organization (WHO)
sures are even more difficult to estimate. Similar approaches to those Environmental Health Criteria, and the evaluation of the European
used with magnetic fields were used in occupational studies, especially Commission’s Scientific Committee on Emerging and Newly Identified
using job categorizations by experts. Measurements of residential expo- Health Risks (SCENIHR). In 2001, the IARC classified ELF magnetic
sures as reported in a few studies (e.g., McBride et al., 1999) must be fields as possibly carcinogenic to humans, based on limited evidence
interpreted with care, as it is questionable how well they predict past from human (epidemiological) studies and inadequate evidence from
exposures. In the example of Canadian children, most time-​weighted studies in experimental animals, including studies published before
average exposures to electric fields were below 20 V/​m (McBride et al., 2001 (IARC, 2002). WHO confirmed the IARC assessment, stating
1999), although in some instances they exceeded 50 V/​m. that evidence from the more recent studies does not alter the classifica-
tion as possible carcinogen (WHO, 2007). SCENIHR also confirmed
that the evidence of possible carcinogenicity remains unchanged, most
Childhood Cancer recently in their update in 2015 (SCENIHR, 2015).
What was considered the strongest evidence in all risk assess-
Childhood Leukemia ments stems from the pooled analyses of the original studies, which
ELF magnetic fields have been studied as a potential risk factor for allowed to some extent the harmonization of the different study
childhood leukemia since the late 1970s, when Wertheimer and Leeper features and was therefore more informative than a comparison of
published a case-​control study from Denver, Colorado, observing a the individual studies. Greenland et al. pooled most of the available
three-​fold increased risk for children living in houses assigned to the studies at the time (Greenland et al., 2000), irrespective of exposure
highest exposure category using the wire code exposure assessment assessment method, which is why they had to combine various expo-
method, a crude categorization of exposure based on proximity of sure indices into a single metric. The combined relative risk estimate
the residence to certain electrical installations, such as power lines of was 1.7 (95% confidence interval [CI] 1.2–​2.3) in children exposed
26

262 PART III:  THE CAUSES OF CANCER


Table 15–2.  Pooled Analyses of Epidemiological Studies on Residential Exposure to Extremely Low Frequency Magnetic Fields and the Risk
of Childhood Leukemia

Ahlbom et al., 2000 Kheifets et al., 2010a Greenland et al., 2000 Schüz et al., 2007

Study base 9 studies (Canada, Denmark, 6 studies (Brazil, Germany, Japan, 12 studies; 8 as in Ahlbom et al. 4 studies (Canada, Germany,
Finland, Germany, New Italy (2), UK); not included in (except UK), 4 additional ones UK, US); all included in
Zealand, Norway, Sweden, Ahlbom et al./​Greenland et al. (2nd from Sweden, other UK, Ahlbom et al. or Kheifets et al.
UK, US) except partly the German study 2nd and 3rd from US) (Germany)
Inclusion Population-​based case-​control As Ahlbom et al., 2000 Inclusive; no restriction but Exposure during the night had
criteria of studies; exposure either delivery of original data to be extractable from the
studies assessed through long-​term exposure measurement
stationary measurements or
calculated fields
Exposure Long-​term stationary As Ahlbom et al., 2000 Measurements (personal, long-​term Long-​term stationary
assessment measurements or calculated or spot), calculated fields measurement
fields
Exposure Average magnetic field level, As Ahlbom et al., 2000 Average magnetic field level, Average exposure during
indices in µT in µT, albeit from different nighttime (10 pm–​6 am), in µT
measurement methods
Numbers of 3247 cases, 10,400 controls 10,818 cases, 12,806 controls 2656 cases, 7084 controls 1842 cases, 3099 controls
subjects
Main result Reference < 0.1 µT Reference ≤ 0.1 µT Reference ≤ 0.1 µT Reference < 0.1 µT
0.1–​< 0.2: OR 1.08, > 0.1–​≤ 0.2: OR 1.07, > 0.1–​≤ 0.2: OR 1.01, 0.1–​< 0.2: OR 1.11,
CI 0.89–​1.31 CI 0.81–​1.41 CI 0.84–​1.21 CI 0.91–​1.36
0.2–​< 0.4: OR 1.11, >0.2–​≤ 0.3: OR 1.16, >0.2–​≤ 0.3: OR 1.06, 0.2–​< 0.4: OR 1.37,
CI 0.84–​1.47 CI 0.69–​1.93 CI 0.78–​1.44 CI 0.99–​1.90
≥ 0.4: OR 2.00, CI 1.27–​3.13 > 0.3: OR 1.44, CI 0.88–​2.36 > 0.3: OR 1.68, CI 1.23–​2.31 ≥ 0.4: OR 1.93, CI 1.11–​3.35
Further results Increase of 15% per 0.2 µT (CI Using ≥ 0.4 µT as highest category Included pooling of 8 studies having Virtually no difference between
4–​27%); small heterogeneity gives OR of 1.46 (CI 0.80–​2.68); wire code data (all from Canada, ORs for nighttime exposure
across studies, although most large overall numbers come from Mexico, or US), with an OR of only and whole-​day exposure;
of the excess cases came from UK study (9665 cases, 9677 1.65 (CI 1.15–​2.35) for highest linear increase in risk with
US study controls), but only few are exposed exposure category, but some increasing exposure
> 0.3 µT (2 cases, 2 controls) heterogeneity across studies

Abbreviations: OR (odds ratio), CI (95% confidence interval), µT (micro-​Tesla).

to average magnetic fields > 0.3 µT compared with those exposed hand, despite the large number of studies, the number of exposed
≤ 0.1 µT. Ahlbom et al. pooled studies that fulfilled certain criteria, children remained small; for instance, combining the pooled data
such as a defined population base for case ascertainment and control sets utilized by Greenland et al. (2000) and Kheifets et al. (2010a),
recruitment, as well as having used longer term measurements or only 125 of 13,474 leukemia cases (0.9%) had exposures > 0.3 µT.
calculated fields for exposure (Ahlbom et al., 2000); the main find- While the consistency of studies and indication of a dose–​response
ing was a relative risk estimate of 2.0 (CI 1.3–​3.1) for exposures trend may favor a causal interpretation, chance and bias cannot be
≥ 0.4 µT compared with exposures < 0.1 µT. While the studies dem- ruled out with reasonable confidence as alternative explanations, so
onstrated a high degree of consistency, a major contribution to the that together with the lack of supportive experimental evidence and
overall excess risk came from a large US study (Linet et al., 1997), plausible mechanistic hypotheses, the overall evidence was classi-
and no association was seen in a large study in the United Kingdom fied as possibly carcinogenic (IARC, 2002; SCENIHR, 2015). With
(UKCCS, 1999). Despite the large number of studies, the numbers respect to confounding, no candidate exposure has been identified
of exposed children were too small to reliably predict the shape of yet (Schüz, 2007); traffic density, use of pesticides in the vicinity
a dose–​response function. Hence, trends are compatible with mon- of power lines, and environmental tobacco smoke are among those
otonic increases, thresholds at 0.3/​0.4 µT, plateauing at higher mag- that were considered. Because of the low prevalence of elevated
netic field levels, as well as no significant increase at all. Kheifets magnetic field exposure, the confounding factor would have to be
and colleagues (2010a) used the approach of Ahlbom et al. (2000) to a strong risk factor itself and, at the same time, be strongly corre-
update the evidence, pooling only the more recent studies, and over- lated with magnetic field strengths from all sources. Hence, while
all confirming the modest association (though somewhat weaker than confounding by an unknown factor is always a possibility, it seems
in the earlier studies). In another pooled analysis of four studies, the to be an unlikely explanation. With regard to exposure assessment
focus was exposure during the night, driven by hypotheses that there methods, there is potential for exposure misclassification. However,
may be less exposure misclassification as the measurement better as it is unlikely that the magnitude of misclassification from most
captures the time when the child is really at home or that nighttime of the exposure assessment methods was associated with disease
exposure may be of more biological relevance. The result was that status, the expected bias would rather be in the direction of dilut-
using nighttime exposure did not yield relative risk estimates differ- ing an association (Schüz, 2007). With regard to selection bias, the
ent from the results when using whole-​day exposure (Schüz et  al., low participation rates in many studies were of concern, especially
2007). Summaries of these four influential pooled analyses are pro- in those studies using measurements. Data suggested that families
vided in Table 15–2 and Figure 15–2. with a lower socioeconomic status were particularly under-​repre-
In summary, based on a large number of studies, a modest asso- sented among controls, resulting in an underestimation of exposure
ciation (1.5-​to 2-​fold) between residential ELF magnetic fields and prevalence in controls (Schüz, 2007). The studies using calculated
childhood leukemia was observed, with—​especially when account- magnetic fields were not affected by selection bias (Feychting
ing for differences in study design features—​a quite high degree et al., 1995; Tynes and Haldorsen, 1997; Verkasalo et al., 1993), as
of consistency across studies carried out in different parts of the no contact with the families was necessary. While they also show
world, at different times, and by different investigators. On the other an association, this is based on much smaller numbers of exposed
 263

Electromagnetic Fields 263
Childhood Leukemia Childhood Leukemia Childhood Brain Tumors
studies before 2000 studies after 2000 All studies
(Ahlbom et al., 2000) (Kheifets et al., 2010a) (Kheifets et al., 2010b)
10 10 10

1 1 1

Figure  15–2. Pooled analyses of epidemiological studies


0.1 0.1 0.1 on residential extremely low frequency magnetic fields and
the risk of childhood leukemia (studies published before and
after 2000) and the risk of brain tumors; pooled relative risk
estimates for three exposure categories compared to the refer-
By exposure categories of < 0.1 µT, 0.1–0.2 µT, 0.2–0.4 µT, and ≥ 0.4 µT ence category of average magnetic fields < 0.1 µT.

children (Ahlbom et  al., 2000; Kheifets et  al., 2010a). Taking all distinguish between electric and magnetic field exposures. Most studies
these considerations into account, selection bias alone is perhaps investigated the risks of leukemia, brain tumors, or breast cancer.
not sufficient to explain the entire association, but a combination of In 2008, Kheifets et al. published a meta-​analysis of studies of occu-
selection bias, confounding, and chance cannot be ruled out as an pational EMF, including 47 studies of brain cancer and 56 of leuke-
alternative explanation for the observed association. mia (Kheifets et al., 2008). For brain cancer, the pooled relative risk
Recently, it was suggested that if ELF magnetic fields would estimate was 1.14 (CI 1.07–​1.22), and for all leukemia it was 1.16
increase the risk of developing leukemia, they may also increase the (CI 1.11–​1.22). There was little variation by diagnostic subtypes, and
risk of a relapse after initial treatment, leading to a lower survival in the associations seen in the more recent studies were slightly weaker
exposed leukemia patients (Foliart et al., 2006). Pooling six studies than in those from the more distant past. Although similarly small
totaling 3073 children with acute lymphoblastic leukemia, however, elevations in risk were seen for both outcomes, the apparent lack of
did not show any association between ELF magnetic field exposure a clear pattern of exposure and risk detracts from a causal interpre-
and overall or event-​free survival from leukemia (Schüz et al., 2012). tation. Studies were of varying quality, especially when it comes to
Some studies also investigated paternal exposure to ELF magnetic exposure assessment; however, restricting the meta-​analysis to only
fields and the risk of childhood leukemia in their offspring, hypothe- studies that were considered of better design quality by the authors
sizing a genetic alteration of the sperm. Meta-​analysis of those studies of the meta-​analyses did not show substantially different results. In
showed a combined relative risk estimate of 1.35 (CI 0.95–​1.91), but a recent multicenter case-​control study in seven countries (InterOcc
at the same time there was some evidence of publication bias (Hug study) involving 1939 cases of glioma, 1822 cases of meningioma,
et al., 2010). Less data are available for maternal exposures, mainly and 5404 controls, in which a JEM based on workplace EMF surveys
because occupational exposure is much less common than in fathers. was applied (Bowman et al., 2007), no association was seen overall
between the ELF magnetic field estimate and brain tumor risk (for
cumulative exposure in µT-​years in the highest exposure category, the
Other Childhood Cancers odds ratios were 0.80 [CI 0.63–​1.00] for glioma and 0.89 [CI 0.70–​
There are much fewer studies on other childhood cancers, includ- 1.12] for meningioma [Turner et  al.,  2014]). In analyses looking at
ing lymphoma, perhaps because those cancers are less frequent in different time windows of exposure, there was an increased risk of
children than leukemia. The only exception is tumors of the central glioma with a dose–​response trend for exposures 1–​4  years before
nervous system (CNS)/​brain. Thirteen studies were combined in a diagnosis, while there was no association in both the 5–​9 years before
literature-​based meta-​analysis, separately analyzing studies based on diagnosis or 10+ years before diagnosis time windows; this may sug-
distance, wire codes, calculated fields, and measurements. For the gest a role of EMF in tumor promotion, but too little data are available
latter, four studies were available and showed a combined relative yet to draw firm conclusions.
risk estimate of 1.14 (CI 0.65–​2.00) for exposures ≥ 0.2 µT compared Residential exposures were also investigated in some studies, but
to < 0.2 µT (Mezei et al., 2008). A recent pooling study having access mainly based on distance to nearby high-​voltage power lines, which
to original data of 10 studies (in Denmark, Finland, Germany, Japan, for adults may introduce substantial exposure misclassification. In a
Norway, Sweden, UK [2]‌, US [2]) showed relative risk estimates of large UK record-​linkage-​based case-​control study using distance to
0.95 (CI 0.65–​1.41), 0.70 (0.40–​1.22), and 1.14 (CI 0.61–​2.13) for power lines as well as calculated fields as exposure metrics, cases of
ELF magnetic field exposures of 0.1–​< 0.2 µT, 0.2–​< 0.4 µT, and leukemia or of brain cancer did not have increased risks compared to
≥ 0.4 µT, compared to < 0.1 µT (Kheifets et al., 2010b; Figure 15–2). cancers not assumed to be related to EMF exposures (used as con-
Taken as a whole, those results were interpreted as providing little trols) (Elliott et al., 2013). With the potential to calculate historic expo-
evidence for an association between residential ELF magnetic field sures from power load of power lines (see Table 15–1), studies from
exposure and childhood brain tumors, suggesting that the observed the Nordic countries are particularly informative. No increased risks
association with an increased risk may be specific to childhood of either brain tumors or leukemias have been observed in Finland
leukemia. (Verkasalo et al., 1996). In Norway, leukemias showed little evidence
of an association (Tynes and Haldorsen, 2003), and for brain tumors
the overall risk was 1.6 (CI 0.9–​2.7), but there was an inverse associa-
Cancer in Adults tion with occupational exposures (Klaeboe et al., 2005). In Sweden,
Studies on EMF exposures and cancer risk in adults included residential risks were slightly elevated for two subtypes of leukemia but not others
and more often occupational studies. Especially the latter often did not and not for brain tumors (Feychting and Ahlbom, 1994).
264

264 PART III:  THE CAUSES OF CANCER


It has also been hypothesized that exposure to ELF magnetic fields consistent associations observed in experimental animals, future epi-
may increase the risk of female breast cancer, assuming that the ELF demiological studies may be better guided by addressing a particular
magnetic fields would affect nocturnal melatonin production (as light mechanistic scenario.
at night does) and that melatonin is protective against breast cancer
(Stevens, 1987), mainly by acting as potent antioxidant (Reiter, 1995).
Several studies addressed the question, looking at occupational expo- RADIOFREQUENCY FIELDS
sures, residential exposures, and use of electric bed heating as expo-
Exposure Sources
sure sources, reviewed in 2006 (Feychting and Forssen, 2006). While
some early smaller studies were inconsistent but showed some positive Currently, the most prevalent sources of radiofrequency (RF) field
associations, the more recent studies reported were mostly negative. In exposure in the general population are related to wireless communica-
a meta-​analysis of almost 25,000 cases and > 60,000 controls from 15 tion, and can be grouped into personal (near-​field) and environmen-
studies published between 2000 and 2009, there was no evidence of an tal (far-​field) exposures. The so-​called bag-​phones and car phones
association with a combined relative risk estimate of 0.99 (CI 0.90–​ that were introduced in the early 1980s had the antenna far from the
1.09) (Chen et al., 2010). Combining results from 23 studies published human body, giving rise to far-​field exposure; handheld mobile phones
from 1990 to 2010, however, showed an overall combined relative risk with the antenna in the handset became available in the late 1980s,
estimate of 1.07 (CI 1.02–​1.13), with the association restricted to pre-​ resulting in near-​field localized exposure to the head. The RF expo-
menopausal breast cancer (Chen et al., 2013). New studies continue to sure decreases rapidly with distance to the exposure source; thus use
appear, such as another well-​conducted occupational study in textile of a wired hands-​free device reduces the RF exposure to the head by
workers in China (Li et al., 2013) showing no increased risk, suggest- approximately 90%. The first handheld mobile phones used the ana-
ing the accumulation of evidence against an association. logue system, which had maximum average radiated powers typi-
Several of the studies were addressing various cancer types as out- cally at 0.6 W (IARC, 2013). In the early 1990s, digital phones were
come, but overall much fewer studies are available for other cancers introduced, which have lower radiated power (maximum average at
in adults than the ones mentioned earlier. For none of the cancers did 0.1–​0.25 W). Development of the technologies for more efficient uti-
any consistent picture emerge (SCENIHR, 2015). Among the most lization of the cellular networks has resulted in even lower exposure
recent cohort studies, no increased risk was seen for leukemia, brain levels during actual use through the introduction of adaptive power
cancer, and breast cancer in Danish electric utility workers (Johansen control (APC), by which mobile phones reduce their output power to
et al., 2007). In the prospective Netherlands cohort study, among pre-​ the level sufficient to maintain a good quality contact with the base
selected cancers (namely lung, breast, and brain cancer), as well as station, and discontinuous transmission (DTX), which means that the
hematological malignancies, no increases were seen for lung, breast, mobile phone transmits only when the user is talking. Through this,
and brain cancer, but among hematological malignancies there was an average RF field levels from mobile phones have decreased consider-
increased risk of acute myeloid leukemia with ever higher exposures ably (AGNIR, 2012). The third generation of mobile phones has the
and an increased risk of follicular lymphoma with both ever and cumu- same maximum average output level as the second generation, but
lative exposures (Koeman et al., 2014). the actual output power in real use is about 10 times lower than that
of second-​generation mobile phones when in use. Another source of
near-​field exposure is cordless phones used within homes, with DECT
Future Research Needs phones being the most successful technology (i.e., wireless handsets
Whether the observed association between residential ELF magnetic connected to the landline phone through a DECT base station in the
fields and childhood leukemia is causal remains an open question that home). As the DECT phone only transmits to a base station that is a
merits further research. Given the consistency of results from epidemi- few meters away, exposure is considerably lower, with average pow-
ological studies, combined with their difficulty of further reducing the ers 10 or more times smaller than those from mobile phones at their
potential of bias and confounding to a level that would address the con- highest power level.
cerns of having created a spurious association, it is unlikely that further Sources of environmental, or far-​field, exposure include, for exam-
case-​control studies of similar design would provide any new insights. ple, mobile phone base stations and radio and television transmitters,
Promising advancements were recently made in experimental set- DECT phone base stations, and wireless local area networks (Wi-​Fi),
tings with the development of a mouse model that better mimics leu- which give rise to exposures that are only a fraction of the exposure
kemia development in children (Hauer et  al., 2014; Martin-​Lorenzo guidelines and less than 1% of the exposure to the head from a hand-
et al., 2015); application of this model is underway. If the results are held mobile phone (AGNIR, 2012). Similarly, devices such as baby
positive, this may provide insights into mechanisms, perhaps suggest- monitors and smart meters used to measure electricity consump-
ing the existence of particularly susceptible children, which could in tion also generate very low RF fields. These technologies have also
turn be addressed by epidemiology. If the results are negative, it adds to changed over time; for example, analog television has been replaced
the evidence that the epidemiological studies may show a non-​causal by digital television broadcasting, which in most cases has resulted in
association and will be a general warning of how cautious the inter- decreased total output power (AGNIR, 2012).
pretation of epidemiological studies investigating the combination of Occupational settings may be associated with higher exposures than
rare exposures and rare diseases must be. If the association was causal, are found in the general population. Workers in occupations involving
due to the rarity of relevant exposure levels, the preventive potential the use of dielectric heaters or induction heaters (e.g., PVC welders)
is limited; only a small proportion of childhood leukemias would be are among the highest exposed group. Other examples are operators of
attributable to ELF magnetic field exposure, recently estimated to be surgical and other medical diathermy devices or magnetic resonance
< 1%–​2% in European countries (Grellier et al., 2014) and possibly imaging (MRI), workers near radar equipment, or those involved in
slightly higher in North America (Greenland and Kheifets, 2006). the maintenance of transmitters such as base stations and radio and
For adults, any future studies should be based on better expo- television towers (AGNIR, 2012). Applications with sometimes inten-
sure measures, alongside testable plausible mechanistic hypotheses. tionally relatively high exposures to the general public are anti-​theft
Further studies linking job titles or other EMF surrogates with can- devices, such as those used in retail stores, but the time that humans
cer outcomes will not provide additional insight. “Better” exposure stay in the close vicinity of those sources is usually very short.
measures need to be validated with respective measurements and
comprehensive analyses of their direction and magnitude of potential
Mobile Phone Use
exposure misclassification. “Testable” hypotheses should be described
by plausible or novel scenarios. Candidates not sufficiently addressed During the last 15 years, studies of RF exposure associated with wire-
are co-​exposures of EMF with known carcinogens (such as chemi- less communication have focused on mobile phone use, which is the
cals or ionizing radiation). Like for childhood leukemia, if there were source that gives the highest localized RF exposure. Consequently,
 265

Electromagnetic Fields 265
the main studied outcomes have been tumors in the head and neck (Benson et al., 2013), questionnaire information will not be affected by
region, as they are located in the areas of the body where most of the recall bias, but non-​differential exposure misclassification may be sub-
energy is absorbed during a mobile phone call (i.e. glioma, meningi- stantial because of the difficulty in reporting accurately about mobile
oma, and acoustic neuroma). Systematic reviews of the scientific evi- phone use among healthy volunteers as well, as described previously.
dence have continuously been conducted by international and national Information obtained from mobile phone operators, if unequivocally
bodies such as the SCENIHR, IARC, and the Independent Advisory linked to a specific person, will reduce non-​differential exposure mis-
Group on Non-​ionizing Radiation (AGNIR) of Public Health England classification considerably. A  Danish cohort study identified mobile
(AGNIR, 2012; IARC, 2013; SCENIHR, 2009, 2015). The WHO is phone users through the subscription registers held by mobile phone
currently working on an update of the Environmental Health Criteria operators during a period when mobile phone use was still uncom-
document on RF fields. All reviews identify considerable method- mon in the general population (Frei et al., 2011; Johansen et al., 2001;
ological limitations and inconsistencies in the available epidemio- McCarthy et al., 2011; Schüz et al., 2006b). With this approach, non-​
logical studies, which prevent firm conclusions. In 2011, the IARC differential exposure misclassification could be limited, but no infor-
classified RF fields as possibly carcinogenic to humans, based on mation was available about the amount of mobile phone use. Thus,
limited evidence in humans from epidemiological case-​control stud- the study would not be able to detect an increased risk in a small sub-
ies of mobile phone use and limited evidence from animal studies group of heavy users. Currently, one cohort study with more elabo-
(IARC, 2013). AGNIR, however, concluded in 2012 that the “accu- rated exposure information from mobile phone operators is underway
mulating evidence on cancer risks … is increasingly in the direction (Schüz et al., 2011a).
of no material effect of exposure” (AGNIR, 2012), also based on the Selection bias may also occur in case-​control studies caused by
epidemiological studies of mobile phone use, combined with infor- non-​participation related to the exposure. Investigation of non-​
mation from incidence trend studies. Based on even more recent data, response in the Interphone study showed that mobile phone users
the SCENIHR review concludes that the evidence for an effect on among both cases and controls were more likely to participate than
glioma has become weaker since the IARC evaluation (SCENIHR, subjects who did not use a mobile phone (Vrijheid et al., 2009b). As
2015). It was noted in all those assessments that conclusions could non-​response was more common among controls than cases, selec-
not be drawn about longer latencies than up to 15  years since first tion bias was introduced, leading to a reduction of the risk estimates
mobile phone use. by approximately 10% for ever regular use of a mobile phone. The
cohort studies have used population-​based cancer registers for case
Methodological Limitations identification, and follow-​up could be made independently of study
Most of the available studies are case-​control studies with retrospec- participants (Benson et  al., 2013; Frei et  al., 2011; Johansen et  al.,
tive assessment of mobile phone use history through structured per- 2001; Schüz et al., 2006b).
sonal interviews or self-​administered questionnaires. This method has
the advantage that detailed information can be collected, such as the Mobile Phone Use Results
preferred ear when using the phone and use of hands-​free devices, There are now many publications on mobile phone use and brain
but it is prone to exposure misclassification, both non-​differential and tumors (e.g., AGNIR, 2012; Ahlbom et  al., 2009; SCENIHR, 2015;
differential, where the former would more likely lead to a dilution of Swerdlow et al., 2011). Several studies are published in multiple pub-
an effect should there be a true association, and the latter would more lications, and have also been included in pooled analyses. Most of the
likely lead to reports of overestimated or even spurious effects. The studies have focused on glioma and acoustic neuroma, and somewhat
early studies of mobile phone use and cancer risk were conducted at fewer on meningioma. In most studies, the main analyses were of
a time period when use of handheld mobile phones was uncommon exposure, defined as time since first mobile phone use. Table 15–3
in the general population, and the technology had only been available includes the original publication of studies with longer induction peri-
for a few years. Thus, these studies could only inform about effects ods, and displays results for glioma and acoustic neuroma. Amount of
after short exposure duration, which are not expected for the type of use in cumulative hours or cumulative number of calls were analyzed
outcomes studied. This review focuses on studies that include induc- in several of the studies.
tion periods of at least 5  years for glioma and meningioma, and at
least 10 years for acoustic neuroma. The changes over time of mobile Time Since First Mobile Phone Use.  For glioma, most of the
phone technologies, the considerably reduced costs associated with epidemiological evidence does not indicate an increased risk associated
buying and using the devices, and the development of new applica- with time since first mobile phone use, regardless of how long a time
tions (e.g., the introduction of smartphones) have all led to a sub- the phone had been used. None of the cohort studies with prospective
stantial increase in mobile phone use over time; at the same time, information on mobile phone use found indications of an increased
as illustrated earlier, the output power of the devices has decreased glioma risk, examining exposure durations of more than 13 years since
substantially with each new technology. This combination is a consid- first mobile phone subscription in the Danish study (Frei et al., 2011)
erable challenge for the retrospective assessment of mobile phone use and more than 10 years in the UK cohort (Benson et al., 2013, 2014).
histories in case-​control studies. Furthermore, studies of brain tumors Exceptions to this pattern are two case-​control studies conducted in
are faced with the additional complication that the tumor itself may Sweden by Hardell and coworkers (Hardell et al., 2006, 2013), which
affect memory function. reported moderately increased risks of malignant brain tumors already
Validation studies conducted as part of the Interphone study, a large after a short period of mobile phone use, and substantially increased
international collaborative study including 16 study centers (Cardis risk estimates after longer exposure durations. The Interphone study,
et  al., 2007), have shown considerable exposure misclassification on the other hand, reported risk estimates for glioma that were consist-
when healthy volunteers were asked to report their amount of mobile ently below unity, which could, at least partly, be explained by selec-
phone use 6 months earlier (Vrijheid et al., 2006). Moreover, it was tion bias caused by non-​participation (Interphone Study Group, 2010;
found that cases tended to overestimate their mobile phone use to Vrijheid et al., 2009b). For meningioma, one study reported a doubling
a greater extent the further back in time they were reporting about of the risk after more than 10 years of use of an analogue mobile phone
(Vrijheid et al., 2009a), which was not observed among controls (the (Hardell et al., 2005), while most other studies reported risk estimates
study covered recall approximately 4 years back in time). A study of close to or below unity (e.g., Interphone Study Group, 2010; Lahkola
self-​reported year when first starting to use a mobile phone also found et al., 2008). A recent French study reported a non-​significantly raised
considerable misclassification (Pettersson et al., 2015), which was also risk of meningioma after 10 years of mobile phone use (Coureau et al.,
found in responses to a seemingly simple question like preferred side 2014).
of the head during mobile phone use (Kiyohara et al., 2015). The few For acoustic neuroma, the pattern is very similar to the glioma find-
cohort studies available have either collected exposure information ings. The Hardell group reported considerably increased risks already
from subscriber registers, or prospectively collected questionnaires. after less than 5 years of mobile phone use (Hardell et al., 2002, 2005),
When collected prospectively, as in the UK Million Women study while other studies found no association between acoustic neuroma
26

Table 15–3.  Epidemiological Studies of Mobile Phone Use and Risk of Malignant Brain Tumors and Acoustic Neuroma That Present Results for at
Least 10 Years Since First Start of Mobile Phone Usea

Location, Time Relative Risk/​SIR


Reference Period, Design Source Outcome Exposure (95% CI)

Schüz et al., 2006 Denmark 420,095 mobile phone Brain and nervous Time since first subscription
1982–​2002 subscribers; the whole system < 1 year 0.90 (0.67–​1.18)
Cohort study Danish population 1–​4 years 1.03 (0.91–​1.17)
5–​9 years 0.96 (0.84–​1.09)
≥ 10 years 0.66 (0.44–​0.95)
Frei et al., 2011 Denmark 358,403 mobile phone Glioma Men
1990–​2007 subscribers; subset Time since first subscription
Cohort study of previous study 1–​4 years 1.20 (0.96–​1.50)
with information on 5–​9 years 1.05 (0.87–​1.26)
socioeconomic status ≥ 10 years 1.04 (0.85–​1.26)
10–​12 years 1.06 (0.85–​1.34)
≥ 13 years 0.98 (0.70–​1.36)

Women
Time since first subscription 0.87 (0.43–​1.75)
1–​4 years 1.02 (0.60–​1.72)
5–​9 years 1.04 (0.56–​1.95)
≥ 10 years
Schüz et al., 2011b Denmark 2,883,665 Danes Acoustic neuroma Mobile phone subscription
1998–​2006 included in the ≥ 11 years
Cohort study CANULI cohort Men
No 1.0
Yes 0.87 (0.52–​1.46)
Women
No No exposed case, 1.6
Yes expected
Benson et al., 2013, UK 791,710 women Glioma Duration of use
2014 1999–​2005—​followed participating in 875 cases < 5 years 0.96 (0.75–​1.23)
through 2011 the UK Million 5–​9 years 0.86 (0.72–​1.02)
Cohort study Women Study, who ≥ 10 years 0.77 (0.62–​0.96)
answered a base-​ Acoustic neuroma Duration of use
line questionnaire 126 cases < 5 years 0.94 (0.53–​1.66)
1999–​2005 5–​9 years 1.46 (0.94–​2.27)
≥ 10 years 1.17 (0.60–​2.27)
Hardell et al., 2002a, Sweden Population-​based Malignant brain Mobile phone use, analog
2002b 1997–​2000 tumors > 1–​6 years 1.09 (0.68–​1.75)
Case-​control study 588 cases > 6 years 1.17 (0.75–​1.81)
Acoustic neuroma Mobile phone use, analog
159 cases > 1–​5 years 3.0 (1.0–​9.3)
> 5–​10 years 3.8 (1.4–​10.2)
> 10 years 3.5 (0.7–​16.8)
Hardell et al., 2005, Sweden Population-​based Malignant brain Mobile phone use, analog
2006 2000–​2003 tumors > 1–​5 years –​
Case-​control study 359 cases > 5–​10 years 1.8 (0.9–​3.5)
> 10 years 3.5 (2.0–​6.4)
Mobile phone use, digital
> 1–​5 years 1.6 (1.1–​2.4)
> 5–​10 years 2.2 (1.4–​3.4)
> 10 years 3.6 (1.7–​7.5)
Acoustic neuroma Mobile phone use, analog
84 cases > 1–​5 years 9.9 (1.4–​69)
> 5–​10 years 5.1 (1.9–​14)
> 10 years 2.6 (0.9–​8.0)
Mobile phone use, digital
> 1–​5 years 1.7 (0.9–​3.5)
> 5–​10 years 2.7 (1.3–​5.7)
> 10 years 0.8 (0.1–​6.7)
 267

Electromagnetic Fields 267
Table 15–3. Continued

Location, Time Relative Risk/​SIR


Reference Period, Design Source Outcome Exposure (95% CI)

Hardell et al., 2013 Sweden Population-​based Malignant brain tumors Mobile phone use, analog
2007–​2009 593 cases > 1–​5 years –​
Case-​control study > 5–​10 years 0.6 (0.1–​3.1)
> 10–​15 years 1.4 (0.7–​3.0)
> 15–​20 years 1.4 (0.7–​2.7)
> 20–​25 years 2.1 (1.1–​4.0)
> 25 years 3.3 (1.6–​6.9)
Mobile phone use, digital
> 1–​5 years 1.8 (1.01–​3.4)
> 5–​10 years 1.6 (0.97–​2.7)
> 10–​15 years 1.3 (0.8–​2.2)
> 15–​20 years 2.1 (1.2–​3.6)
> 20–​25 years –​
> 25 years –​
Interphone Study Group, Interphone Population-​based Glioma Time since first regular use
2010, 2011 13 countries 2708 cases 1–​1.9 years 0.62 (0.46–​0.81)
2000–​2004 2–​4 years 0.84 (0.70–​1.00)
Case-​control study 5–​9 years 0.81 (0.60–​0.97)
≥ 10 years 0.98 (0.76–​1.26)
Acoustic neuroma Time since first regular use
1105 cases 1–​1.9 0.73 (0.49–​1.09)
2–​4 years 0.87 (0.69–​1.10)
5–​9 years 0.90 (0.69–​1.16)
≥ 10 years 0.76 (0.52–​1.11)
Lahkola et al., 2007; Nordic-​UK Interphone Population-​based Glioma Time since first regular use
Schoemaker centers, broader 1496 cases 1.5–​4 years 0.77 (0.65–​0.92)
et al., 2005 age range 5–​9 years 0.75 (0.62–​0.90)
(Partly overlapping with 2000–​2004 ≥ 10 years 0.95 (0.74–​1.23)
the Interphone study) Case-​control study Acoustic neuroma Time since first regular use
676 cases 1.5–​4 years 0.8 (0.7–​1.0)
5–​9 years 0.9 (0.7–​1.2)
≥ 10 years 1.0 (0.7–​1.5)
Han et al., 2012 Pittsburg, US Hospital based Acoustic neuroma Years of mobile phone use
1997–​2007 343 cases < 10 years 0.79 (0.45–​1.37)
Case-​control study ≥ 10 years 1.29 (0.69–​2.43)
Coureau et al., 2014 France Population-​based Glioma Time since first regular use
2004–​2006 190 cases 1–​4 years 1.04 (0.64–​1.69)
Case-​control study 5–​9 years 1.45 (0.91–​2.33)
≥ 10 years 1.45 (0.68–​3.08)
Pettersson et al., 2014 Sweden Population based Acoustic neuroma Time since first regular use
2002–​2007 451 cases 1–​4 years 1.04 (0.72–​1.52)
Case-​control study 5–​9 years 1.40 (0.98–​2.00)
10–​12 years 1.10 (0.68–​1.76)
≥ 13 years 1.12 (0.72–​1.73)

a
Studies with only short-​term mobile phone use are not included in the table (in total 7 studies, published in multiple papers).

risk and time since starting mobile phone use, not even after more than studies from many parts of the world (Barchana et al., 2012; de Vocht
13 years of mobile phone use (Pettersson et al., 2014). et  al., 2011; Deltour et  al., 2012; Dobes et  al., 2011; Kohler et  al.,
Most studies did not take cordless phone use into consideration in 2011; Little et al., 2012); this was particularly so for middle-​aged and
the exposure assessment, which could potentially cause non-​differen- younger persons, which are the more relevant age groups in relation
tial exposure misclassification. Hardell et al. report increased risks of to mobile phone use, while an incidence increase in the elderly, which
brain tumors associated with cordless phone use in the same order of was observed to start even before the introduction of mobile phone
magnitude as for mobile phone use (Hardell et al., 2006, 2013). No technologies, is believed to be due to improved and more readily
associations were observed with cordless phone use in two national available diagnostic imaging techniques (especially MRI). Simulation
Interphone studies (Lonn et al., 2005; Schüz et al., 2006a). In addi- studies have demonstrated that risk increases after short duration of
tion, when Hardell and colleagues included cordless phone users in mobile phone use are incompatible with the observed incidence trends
the unexposed category to mimic the analyses in the Interphone study, (Deltour et  al., 2012; Little et  al., 2012). More details are provided
the results were virtually unchanged (Hardell et al., 2011). Thus, cord- later in this chapter (section on “Incidence Studies”). Second, recall
less phone use does not appear to explain the differences in results bias is indicated by the findings of the strongest effects among per-
between the studies by Hardell and colleagues and other studies. sons reporting to have started mobile phone use at least 25 years prior
The few reported risk increases appear implausible for several rea- to diagnosis when handheld mobile phones had only been available
sons. First, glioma incidence time trends have been stable since the in Sweden 23 years or less at the end of the data collection (Hardell
introduction of mobile phone technologies, as shown in incidence et al., 2013).
268

268 PART III:  THE CAUSES OF CANCER


Also for acoustic neuroma, incidence time trends do not support risk estimates for daily users did not differ from the overall results of
an effect of mobile phone use (Benson et al., 2013; Larjavaara et al., no associations.
2011a), although fewer data are available. Acoustic neuroma is a slow-​ The very high risk increases at quite low amounts of mobile phone
growing tumor, and one would not expect an effect on the tumor occur- use reported in the studies by Hardell et al. are implausible in the
rence after a short latency period. It is likely that most of the acoustic light of the stable incidence time trends. The findings in the French
neuroma tumors diagnosed within 5 years after first mobile phone use case-​control study would also have resulted in increased incidence
were already present when the person started to use a mobile phone trends (Deltour et al., 2012). The results of the Interphone study are
(Thomsen and Tos, 1990). difficult to interpret, considering the lack of a dose–​response pat-
tern. For acoustic neuroma, there were indications of a downward
Amount of  Mobile Phone Use. If high intensity of trend until the 10th percentile. The cases included in the Interphone
mobile phone use is required for effects on tumor development, study were diagnosed in the early 2000s, at which time approxi-
results based on time since first mobile phone use would be less mately 10% of the controls had cumulated at least 1640 hours of
informative, and estimates of amount of use would be needed. mobile phone use. Now, about 15 years later, a considerably larger
Available studies with analyses of amount of mobile phone use proportion of the population in many countries would have accu-
have used different exposure cutoff points, most often based on mulated this amount of use and more, but so far, no corresponding
the exposure distribution among controls. In the Interphone study, increase in the brain tumor incidence has been observed. In addi-
cumulative hours of mobile phone use were categorized with cut- tion, implausibly high cumulative hours of phone use were more fre-
off points approximately at the deciles of hours of use among quent among cases than controls in the Interphone study (Interphone
controls. As shown in Figure 15–3, no trends of increasing risk Study Group, 2010). As mentioned earlier, the results from the vali-
with increasing amount of use were observed for glioma, acous- dation study provide empirical evidence that recall bias is likely to
tic neuroma, or meningioma (Interphone Study Group, 2010; have affected the findings (Vrijheid et al., 2009a), but the question is
2011). In the highest category of cumulative hours (> 1640 h), whether it can explain the small risk increase in heavy users entirely.
a slight risk increase was observed for all three outcomes, 1.40 The cohort study with prospectively collected exposure informa-
for glioma, 1.15 for meningioma, and 1.32 for acoustic neuroma tion, unaffected by recall bias, did not support the hypothesis of an
(although not statistically significant for the latter two). It is note- increased tumor risk with more frequent mobile phone use, but the
worthy that the risk estimates in the 9th decile (735–​1639 h) were highest exposure category was limited to “daily use” (Benson et al.,
among the lowest observed: 0.71 for glioma, 0.76 for meningioma, 2013, 2014).
and 0.48 for acoustic neuroma. With a 5-​year latency period for
acoustic neuroma, the risk estimate in the 10th decile was statisti- Laterality of  Phone Use and Tumor Localization.  RF
cally significant, but the risk estimate in the 9th decile was still the exposure to the head is highly localized during mobile phone use,
lowest of all categories. For glioma and meningioma, the highest and reaches only a few centimeters deep into the brain. Therefore, if
risk increase associated with heavy mobile phone use was observed the exposure is causally related to brain tumor risk, one would expect
within 1–​4 years after starting to use a mobile phone, for acoustic to find an increased risk on the same side of the head as the phone
neuroma among long-​term users. No associations were observed was usually held, and a risk close to unity on the opposite side. Self-​
with cumulative number of calls. reported laterality of mobile phone use is, however, highly uncertain
In the first two studies by the Hardell group (Hardell et al., 1999, (Kiyohara et  al., 2015). Information about laterality of phone use
2002), no consistent associations between cumulative hours of has so far only been collected retrospectively in case-​control studies,
mobile phone use and brain tumor risk were found, but the last and there is a possibility that cases are affected by their knowledge
two studies observed considerable risk increases, also after hav- about the side of the tumor location when reporting the preferred side
ing accumulated relatively few hours; for example, > 80 hours of used during mobile phone calls prior to diagnosis, and they might
analog phone use were associated with a four-​fold risk increase tend to more often report ipsilateral use (Schüz, 2009). Controls will
of malignant brain tumor, and > 64 hours of digital phone use of course report independently of the fictive “tumor” side assigned
with a two-​fold risk increase. Associations were also observed to them by the researchers, as they do not even know which side of
for meningioma and acoustic neuroma. Several other studies their head is the one relevant in the analysis. Results in the case-​
have analyzed higher amounts of use than these, for example two control studies indicate that recall bias may indeed be a problem, as
early studies from the United States (around 500 h as the high- several studies found an increased risk of brain tumors on the same
est cutoff point; Inskip et  al., 2010; Muscat et  al., 2000), and side of the head as the phone was reported to be held, and at the
national Interphone studies (> 500 h; Lahkola et  al., 2007, 2008; same time a reduced risk on the opposite side and/​or among subjects
Lonn et  al., 2005; Schoemaker et  al., 2005). None of these stud- for whom laterality data were missing (Ahlbom et al., 2009; Schüz,
ies found increased risks at such low cumulative levels of use. In 2009; Swerdlow et  al., 2011). Overall, there are strong indications
the most recent study by the Hardell group (Carlberg et al., 2013; that recall bias may have affected results where laterality was taken
Hardell et al., 2013), four categories of cumulative hours of phone into consideration.
use were analyzed, and risk estimates for malignant brain tumors Another way to take the localized exposure into consideration is
were raised in all four categories, but in some categories of bord- to conduct separate analyses of tumors localized in different lobes of
erline statistical significance. In the highest exposure category the brain. The temporal lobe absorbs the highest energy during mobile
(> 2376 h), an almost eight-​fold risk increase was observed for phone calls, but studies have often grouped the different areas of the
analog and a three-​fold risk increase for digital phone use. Also for brain differently, which hampers comparability. Most studies did not
meningioma, considerable risk increases were found in the high- find higher risk estimates for glioma located in the temporal lobe than
est category of analog and digital mobile phone use, respectively, in other locations. One exception is the Interphone combined analysis,
while acoustic neuroma was not analyzed separately in this study. where the glioma risk was slightly higher in the temporal lobe among
A recent French case-​control study also reported raised risk esti- long-​term users and in the highest category of cumulative hours of use,
mates at considerably fewer hours of mobile phone use than the but not overall (Interphone Study Group, 2010). For meningioma, the
Interphone study (Coureau et  al., 2014). The odds ratio for glioma overall analysis found a lower risk estimate for tumors in the tempo-
after 339–​895 hours of phone use was 1.78 (95% CI 0.98–​3.24, and ral lobe than in other locations. The four studies conducted by Hardell
after at least 896 hours it was 2.89 (95% CI 1.41–​5.93). For menin- and colleagues grouped tumor locations differently in all studies, and
gioma, an increased risk was observed only in the highest exposure small numbers probably prevented analyses of temporal lobe tumors
category, with an odds ratio of 2.57 (95% CI 1.02–​6.44). separately in the two first studies. The fourth study reported somewhat
Only one cohort study with prospectively collected information stronger risk estimates for temporal lobe locations combined with over-
on amount of mobile phone use is currently available (Benson et al., lapping lobes (Hardell et al., 2013). The recent French study reported
2013, 2014). The highest exposure category was “daily use,” and the risk estimates in the same order of magnitude for glioma with temporal
 269

Electromagnetic Fields 269
Glioma
hours cases controls RR (95% CI)

–4 141 197 0.70 (0.52, 0.94)

5–12 145 198 0.71 (0.53, 0.94)

13–30 189 179 1.05 (0.79, 1.38)

31–60 144 196 0.74 (0.55, 0.98)

61–114 171 193 0.81 (0.61, 1.08)

115–199 160 194 0.73 (0.54, 0.98)

200–359 158 194 0.76 (0.57, 1.01)

360–734 189 205 0.82 (0.62, 1.08)

735–1639 159 184 0.71 (0.53, 0.96)

1640+ 210 154 1.40 (1.03, 1.89)

.5 .7 .8 1 1.5 2

Acoustic neuroma
hours cases controls RR (95% CI)

–4 58 144 0.77 (0.52, 1.15)

5–12 63 129 0.80 (0.54, 1.18)

13–30 80 136 1.04 (0.71, 1.52)

31–60 66 131 0.95 (0.63, 1.42)

61–114 74 137 0.96 (0.66, 1.41)

115–199 68 128 0.96 (0.65, 1.42)

200–359 50 144 0.60 (0.39, 0.91)

360–734 58 126 0.72 (0.48, 1.09)

735–1639 49 126 0.48 (0.30, 0.78)

1640+ 77 107 1.32 (0.88, 1.97) Figure 15–3.  Results from the Interphone
study for cumulative hours of mobile
phone use, categorized in deciles. The
reference category is non-​regular mobile
.5 .7 .8 1 1.5 2 phone use.

lobe location as for “other locations,” while risk estimates for frontal The other study estimated the total RF dose at the tumor location
lobe tumors were lower (Coureau et al., 2014). Taken together, the evi- or a corresponding location for the controls (Cardis et  al., 2011).
dence does not consistently suggest stronger associations with tumors The total RF dose was estimated based on retrospective self-​reported
in the temporal lobe. information on cumulative hours of mobile phone use, preferred side
Two studies have further developed analyses of tumor localization of the head, frequency band, communication system, and network
beyond lobe of the brain by using information about the exact local- characteristics. Self-​reported cumulative hours of use and tumor loca-
ization of the tumor based on radiological images (Cardis et al., 2011; tion were the only significant predictors of RF dose (43% and 13%
Larjavaara et al., 2011b). The study by Larjavaara and colleagues used of the variation, respectively). Complete information for estimation
a case-​case design based on the assumption that gliomas in mobile of RF dose were available for a subset of the subjects, and results for
phone users on average are located closer to the exposure source (i.e., glioma were almost identical in this subset when based simply on self-​
where the mobile phone handset is held) than tumors in cases who reported cumulative hours of use as when the more elaborated expo-
were not regular mobile phone users. This type of design eliminates sure estimate was used, contrary to what would have been expected if
potential selection bias caused by non-​participation among controls, there were a causal association between RF dose and glioma risk. The
and if the preferred side for phone use is not included, it may also study design did not attempt to reduce recall bias, and the addition of
reduce the effect of recall bias. Analyses were based on 873 glioma more technical details about the mobile communication system did not
cases, and no major difference in distance between the tumor location seem to reduce non-​differential exposure misclassification. The inves-
and a hypothetical mobile phone was found between cases who were tigators also made a case-​case analysis, similar to the analysis in the
regular mobile phone users and those who were non-​users; if anything, Larjavaara study, but based on somewhat fewer cases (N = 556). They
the distance was somewhat shorter for non-​users. found that > 10 years since first mobile phone use were associated with
270

270 PART III:  THE CAUSES OF CANCER


an increased risk of glioma in the brain region with the highest expo- studies would have been detected in incidence time trends. Both stud-
sure, but they also found a reduced risk for those with 5–​9 years since ies concluded that strong risk increases, such as reported in the studies
first mobile phone use, and no consistent dose–​response pattern with by Hardell et al., would have been detected in incidence time trends.
cumulative call time. The Nordic incidence time trend study also had statistical power to
detect smaller risk increases and would most likely also have detected
Children.  Currently there is only one study available on mobile an increased risk after 1640 cumulated hours of mobile phone use
phone use and brain tumor risk in children and adolescents in the age (Deltour et  al., 2012), which was the risk increase observed in the
range 7–​19  years (Aydin et  al., 2011). It used a case-​control design Interphone study. No such incidence increase was observed.
with exposure information collected through interviews with the Several studies reported brain tumor incidence time trends for chil-
parents and children. Information from mobile phone operators was dren and adolescents separately (Aydin et  al., 2011; de Vocht et  al.,
available for a small subgroup of participants who were able to report 2011; Dobes et al., 2011; Inskip et al., 2010; McKean-​Cowdin et al.,
their children’s current and previous mobile phone numbers. Overall, 2013). None of these studies found increases in the brain tumor inci-
a risk estimate close to unity was observed, and risk did not increase dence after the introduction of mobile phones.
with amount of use or by location of the tumor. Having had a mobile A few incidence studies of acoustic neuroma and parotid gland
phone subscription > 2.8 years prior to diagnosis was associated with tumors have also been published (Benson et al., 2013; de Vocht, 2011;
an increased brain tumor risk, but these results were based on only Larjavaara et al., 2011a; Shu et al., 2012; Stangerup et al., 2010). None
about one-​third of the data. Furthermore, this finding was not compat- of them reports changes in incidence trends that could be linked to the
ible with the stable incidence trends observed. Thus, the study does not introduction of handheld mobile phones.
support the hypothesis of an increased risk of brain tumors, but it also
does not provide strong evidence against a risk increase.
Environmental Exposure
Other Tumors.  For other tumor types (e.g., parotid gland tumors, Studies of cancer risk associated with RF exposure from environmen-
ocular melanoma, pituitary tumors, leukemia, lymphoma, and tes- tal sources have usually focused on radio and television transmitters
ticular cancer), the available data are fewer than for brain tumors. or mobile phone base stations. Several studies were conducted as a
Currently, no consistent evidence suggests that mobile phone use result of public concern, and used an ecological design with expo-
is associated with an increased risk of these tumors (AGNIR, 2012; sure simply measured as distance to the nearest transmitter. These
Ahlbom et al., 2009; SCENIHR, 2015). studies are not included in this review, as distance alone is too crude
to measure RF exposure accurately. Two studies have assessed RF
Incidence Studies exposure from radio and television transmitters on an individual level,
Normally, incidence time trend studies would not be regarded as using elaborated modeling with detailed information about important
informative for inferences about etiology because exposure informa- exposure determinants, such as frequency, field strength, and other
tion at the individual level is not available, and there is no information relevant information (Bethke et al., 2008; Ha et al., 2007; Merzenich
about potential confounding factors. In addition, changes in clinical et al., 2008; Schüz et al., 2008), and one study focused on exposure
practices and new diagnostic procedures may affect incidence time from mobile phone base stations, also with elaborated modeling to
trends without any real change in disease occurrence (as it does assess exposure (Elliott et al., 2010). None of the studies found any
indeed for brain tumors, with the detection of smaller tumors follow- indications of increased risk for any types of tumors among children
ing the introduction of computed tomography and later MRI) (Fisher in relation to environmental RF exposure from transmitters. Taken
et al., 2007; Helseth, 1995). Nevertheless, the situation with mobile together, the evidence does not suggest that environmental RF expo-
phone use and brain tumor risk is somewhat different. Brain tumors sure from transmitters affects cancer risk, but few data are currently
are not related to lifestyle factors like smoking or alcohol consump- available.
tion; the only established environmental risk factor is ionizing radia- There are currently no epidemiological studies available on poten-
tion, which explains only a small fraction of the occurrence. During tial effects on cancer risk from exposures emanating from sources
the last decades there has been a rapid increase in the prevalence of such as Wi-​Fi, smart meters, and other low-​level RF-​emitting devices.
mobile phone use in the general population in many countries, from a Although these exposure sources are now ubiquitous in society, the
few percent at the end of the 1980s to near 100% in some age groups level of exposure to individuals from these devices is very low, far
by the middle of the first decade of the 2000s. If RF exposure from below current exposure guidelines (AGNIR, 2012; Tell et al., 2013),
mobile phone use increases the risk of brain tumors, one would expect and considerably lower than the exposure during a mobile phone call,
to see an increasing brain tumor incidence in many countries, in the for example.
age groups that first adopted mobile phone technologies, unless the
latency period is longer than the present risk time, or the risk increase
is restricted to a small subgroup of the population. As mentioned
Occupational Exposure
earlier, brain tumor incidence studies have been published in many
countries, including the Nordic countries, the United Kingdom, the Epidemiological research on occupational exposure to RF fields has
United States, and Australia (Ahlbom and Feychting, 2011; de Vocht not evolved in the same way as for ELF fields. Accurate assessment
et  al., 2011; Deltour et  al., 2009, 2012; Dobes et  al., 2011; Kohler of occupational RF exposure remains a key concern in these studies,
et al., 2011; Little et al., 2012). They have all reported on malignant as they have usually been limited to job title alone, with no actual
brain tumors or gliomas specifically. The time periods included differ knowledge about the exposure levels for the workers in the particu-
between the incidence studies, but the majority cover the incidence lar occupations, to expert assessment, for example by occupational
until 2007 or 2008, and one covers the period until 2009. All stud- hygienists when no objective measurements were available, or by
ies report stable incidence trends in age groups where mobile phone restricting the exposed group to specific areas, such as militaries in the
use has become prevalent, with no indications of increasing incidence navy. Exposure assessment has not been conducted on an individual
after the introduction of mobile phones. In the oldest age groups, level, and information on potential confounding factors is lacking.
however, where mobile phone use has not been prevalent, an increas- Many exposed occupations are also exposed to low frequency fields,
ing brain tumor trend can be observed, which started before the intro- such as radio and television repairmen and radio and telegraph opera-
duction of mobile phones (Deltour et al., 2009; Dobes et al., 2011). tors. Most occupational studies focused on the risk of cancer overall,
Two studies used simulations to predict the shape of the glioma inci- and leukemia and brain tumors (AGNIR, 2012; Ahlbom et al., 2004).
dence trend over time under assumptions of different risk scenarios No consistently increased risks were reported, but findings were often
related to mobile phone use (Deltour et al., 2012; Little et al., 2012). based on small numbers, and exposure misclassification is likely to
The aim was consistency checks, that is, to investigate whether the be considerable. A  few risk increases were observed, generally in
risk of malignant brain tumors reported in some of the case-​control studies with severe methodological shortcomings. The largest studies
 271

Electromagnetic Fields 271
with the most sophisticated exposure assessment provide little support Few data are available on mobile phone use and cancer risk in chil-
for an association (Groves et al., 2002; Morgan et al., 2000). Groves dren, and although the single available case-​control study of brain
et al. reported higher leukemia mortality among groups with the high- tumors in children and adolescents did not indicate an increased risk,
est potential for radar exposure, with a risk estimate of 1.48 (95% and brain tumor incidence time trends in this age group have been sta-
CI 1.01–​2.17), but also a reduced brain cancer mortality in the same ble during the last decades, further studies focused on children are rec-
group, with a risk estimate of 0.65 (95% CI 0.43–​1.01). Morgan and ommended. These must, however, be able to overcome potential bias
colleagues reported risk estimates below unity for both leukemia and from differential recall and selection bias. Prospective cohort studies
nervous system tumors, but results were based on small numbers of would overcome these limitations and could also enable follow-​up into
exposed cases (Morgan et al., 2000). Overall, the data do not suggest adult life. To address cancer risk in children, adolescents, and young
an increased cancer risk associated with occupational RF exposure, adults, however, these cohorts need to be extremely large because of
but the evidence is not sufficient to exclude the possibility of a risk the rarity of the diseases, which is an obstacle for the feasibility of
increase. such studies. Exposure assessment among children poses additional
challenges, as they do not have mobile phone subscriptions in their
own name, and may more often share a mobile phone. In addition,
CONCLUSIONS exposure patterns have changed considerably with the introduction of
smartphones, which are more frequently used for texting and surfing
Overall, the epidemiological evidence does not suggest that exposure than making phone calls.
to RF fields increases cancer risk, taking into consideration the meth- Studies of occupational RF exposure would allow investigation of
odological limitations in the case-​control studies of mobile phone use higher levels of exposure, but to be informative, exposure assessment
and brain tumor risk, lack of associations in the cohort studies, and needs to be improved considerably, to be on an individual level, and to
the stable brain tumor incidence time trends in the age groups that be properly validated. In addition, information on potential confound-
have been frequent mobile phone users for a long time. Some uncer- ing factors must be collected. An obstacle is that exposed occupational
tainty remains regarding the heaviest mobile phone users, although groups are small in numbers, and to achieve sufficient statistical power
the proportion of the population that has reached the highest cumula- international collaboration is necessary.
tive hours of mobile phone use investigated thus far is likely to be Numerous studies of genotoxic or carcinogenic effects of RF fields
substantial by now. Mobile phone use during the more recent years in experimental animals have been conducted, but have not shown
is, however, likely to be associated with considerably lower average consistent effects at exposure levels below guideline levels. Studies of
RF exposure levels, as the third-​generation mobile phone technology RF fields as a co-​carcinogen have been mostly negative, or have had
has become more widespread. Experimental studies, including ani- methodological limitations. The recent finding of increases in tumors
mal studies with long-​term exposure, have not found consistent evi- of the lung and liver, and lymphoma in ENU-​treated mice exposed to
dence of a carcinogenic effect (AGNIR, 2012), and despite the large RF fields, but with no dose response, warrants replication.
amount of research performed, a plausible biological mechanism has
still not been suggested. Thus, the mounting evidence speaks increas-
References
ingly against a carcinogenic effect of exposure to low-​level radiofre-
quency electromagnetic fields. However, neither cohort, case-​control, AGNIR. 2012. Health effects from radiofrequency electromagnetic fields:
nor incidence time trend studies performed to date would be able to report from the Independent Advisory Group on Non-​Ionising Radiation.
In Documents of the Health Protection Agency R, Chemical and
detect a risk increase after an induction period exceeding 20  years,
Environmental Hazards. RCE 20, Health Protection Agency, UK (Ed.).
and limited data are available on potential effects in children. Ahlbom A, Day N, Feychting M, et  al. 2000. A pooled analysis of mag-
netic fields and childhood leukaemia. Br J Cancer, 83(5), 692–​698.
PMCID: PMC2363518.
FUTURE RESEARCH NEEDS Ahlbom A, and Feychting M. 2011. Mobile telephones and brain tumours.
BMJ, 343, d6605.
Although brain tumor incidence time trends have remained stable Ahlbom A, Feychting M, Green A, et  al. 2009. Epidemiologic evidence on
since the introduction of mobile phones, and recall and selection bias mobile phones and tumor risk: a review. Epidemiology, 20(5), 639–​652.
in available case-​control studies are likely explanations for observed Ahlbom A, Green A, Kheifets L, Savitz D, and Swerdlow A. 2004. Epidemiology
risk increases after moderate and short-​term mobile phone use, the of health effects of radiofrequency exposure. Environ Health Perspect,
112(17), 1741–​1754. PMCID: PMC1253668.
uncertainty regarding the most intensive mobile phone use and the
Aydin D, Feychting M, Schüz J, et al. 2011. Mobile phone use and brain tumors
widespread use of mobile phones throughout society and in all age in children and adolescents:  a multicenter case-​control study. J Natl
groups, with an increasing amount of time spent using the technology, Cancer Inst, 103(16), 1264–​1276.
make further research warranted. Barchana M, Margaliot M, and Liphshitz I. 2012. Changes in brain glioma inci-
A continued monitoring of brain tumor incidence time trends in dence and laterality correlates with use of mobile phones: a nationwide pop-
various age groups is recommended, provided that well-​established ulation based study in Israel. Asian Pac J Cancer Prev, 13(11), 5857–​5863.
high-​quality cancer registries are available. Further case-​control Benson VS, Pirie K, Schuz J et  al. 2014. Authors’ response to:  The case of
studies with retrospective self-​reported exposure information are acoustic neuroma: comment on mobile phone use and risk of brain neo-
unlikely to add useful knowledge. Highly recommended are stud- plasms and other cancers. Int J Epidemiol. 43(1):275.
Benson VS, Pirie K, Schüz J, et al. 2013. Mobile phone use and risk of brain
ies that use a cohort design with prospectively collected exposure
neoplasms and other cancers:  prospective study. Int J Epidemiol, 42(3),
information, preferably from independent sources such as mobile 792–​802.
phone operator registers, combined with prospective questionnaire Bethke L, Murray A, Webb E, et  al. 2008. Comprehensive analysis of DNA
information that covers important aspects that may modify expo- repair gene variants and risk of meningioma. J Natl Cancer Inst, 100(4),
sure levels, such as use of hands-​free devices, preferred side of the 270–​276.
head, and DECT phones and other wireless technologies. Access Bowman JD, Touchstone JA, and Yost MG. 2007. A population-​based job expo-
is also needed to population-​based sources for complete follow-​up sure matrix for power-​frequency magnetic fields. J Occup Environ Hyg,
of cancer occurrence, and studies must cover long enough expo- 4(9), 715–​728.
sure durations and be of sufficient size to provide statistically sta- Cardis E, Armstrong BK, Bowman JD, et al. 2011. Risk of brain tumours in rela-
tion to estimated RF dose from mobile phones: results from five Interphone
ble risk estimates. Such an approach would address the sources of
countries. Occup Environ Med, 68(9), 631–​640. PMCID: PMC3158328.
bias identified in currently available epidemiological studies. Focus Cardis E, Richardson L, Deltour I, et al. 2007. The INTERPHONE study: design,
on mobile phone use is warranted, as it is still the main source of epidemiological methods, and description of the study population. Eur
RF exposure in the general population, while exposure levels from J Epidemiol, 22(9), 647–​664.
mobile phone base stations, smart meters, and other far-​field sources Carlberg M, Soderqvist F, Hansson Mild K, and Hardell L. 2013. Meningioma
are considerably lower. patients diagnosed 2007–​2009 and the association with use of mobile
27

272 PART III:  THE CAUSES OF CANCER


and cordless phones:  a case-​control study. Environ Health, 12(1), 60. meningioma in patients diagnosed 2000–​2003. Neuroepidemiology, 25(3),
PMCID: PMC3733594. 120–​128.
Chen C, Ma X, Zhong M, and Yu Z. 2010. Extremely low-​frequency electro- Hardell L, Carlberg M, and Hansson Mild K. 2011. Re-​analysis of risk for
magnetic fields exposure and female breast cancer risk: a meta-​analysis glioma in relation to mobile telephone use: comparison with the results
based on 24,338 cases and 60,628 controls. Breast Cancer Res Treat, of the Interphone international case-​control study. Int J Epidemiol, 40(4),
123(2), 569–​576. 1126–​1128.
Chen Q, Lang L, Wu W, et al. 2013. A meta-​analysis on the relationship between Hardell L, Carlberg M, and Mild KH. 2006. Case-​control study of the asso-
exposure to ELF-​EMFs and the risk of female breast cancer. PLoS One, ciation between the use of cellular and cordless telephones and malig-
8(7), e69272. PMCID: PMC3712018. nant brain tumors diagnosed during 2000–​2003. Environ Res, 100(2),
Coureau G, Bouvier G, Lebailly P, et  al. 2014. Mobile phone use and brain 232–​241.
tumours in the CERENAT case-​control study. Occup Environ Med, 71(7), Hardell L, Carlberg M, Soderqvist F, and Mild KH. 2013. Case-​control study
514–​522. of the association between malignant brain tumours diagnosed between
de Vocht F. 2011. Cell phones and parotid cancer trends in England. 2007 and 2009 and mobile and cordless phone use. Int J Oncol, 43(6),
Epidemiology, 22(4), 608–​609. 1833–​1845. PMCID: PMC3834325.
de Vocht F, Burstyn I, and Cherrie JW. 2011. Time trends (1998–​2007) in Hardell L, Hallquist A, Mild KH, et al. 2002. Cellular and cordless telephones
brain cancer incidence rates in relation to mobile phone use in England. and the risk for brain tumours. Eur J Cancer Prev, 11(4), 377–​386.
Bioelectromagnetics, 32(5), 334–​339. Hardell L, Nasman A, Pahlson A, Hallquist A, and Hansson Mild K. 1999. Use
Deltour I, Auvinen A, Feychting M, et al. 2012. Mobile phone use and inci- of cellular telephones and the risk for brain tumours: a case-​control study.
dence of glioma in the Nordic countries 1979–​2008: consistency check. Int J Oncol, 15(1), 113–​116.
Epidemiology, 23(2), 301–​307. Hauer J, Borkhardt A, Sanchez-​Garcia I, and Cobaleda C. 2014. Genetically
Deltour I, Johansen C, Auvinen A, et al. 2009. Time trends in brain tumor inci- engineered mouse models of human B-​cell precursor leukemias. Cell
dence rates in Denmark, Finland, Norway, and Sweden, 1974–​2003. J Cycle, 13(18), 2836–​2846.
Natl Cancer Inst, 101(24), 1721–​1724. Helseth A. 1995. The incidence of primary central nervous system neoplasms
Dobes M, Khurana VG, Shadbolt B, et al. 2011. Increasing incidence of glio- before and after computerized tomography availability. J Neurosurg,
blastoma multiforme and meningioma, and decreasing incidence of 83(6), 999–​1003.
Schwannoma (2000–​2008):  findings of a multicenter Australian study. Hug K, Grize L, Seidler A, Kaatsch P, and Schüz J. 2010. Parental occupational
Surg Neurol Int, 2, 176. PMCID: PMC3263004. exposure to extremely low frequency magnetic fields and childhood can-
Elliott P, Shaddick G, Douglass M, et al. 2013. Adult cancers near high-​voltage cer: a German case-​control study. Am J Epidemiol, 171(1), 27–​35.
overhead power lines. Epidemiology, 24(2), 184–​190. IARC. 2002. Non-​ionizing radiation. Part  1, static and extremely low-​fre-
Elliott P, Toledano MB, Bennett J, et al. 2010. Mobile phone base stations and quency (ELF) electric and magnetic fields. Vol. 80. Lyon: International
early childhood cancers: case-​control study. BMJ, 340, c3077. Agency for Research on Cancer.
Feychting M, and Ahlbom A. 1994. Magnetic fields, leukemia, and central ner- IARC. 2013. Non-​ionizing radiation. Part 2, Radiofrequency electromagnetic
vous system tumors in Swedish adults residing near high-​voltage power fields. IARC monographs on the evaluation of carcinogenic risks to
lines. Epidemiology, 5(5), 501–​509. humans. Vol. 102. Lyon: International Agency for Research on Cancer.
Feychting M, and Forssen U. 2006. Electromagnetic fields and female breast ICNIRP. 1998. Guidelines for limiting exposure to time-​ varying electric,
cancer. Cancer Causes Control, 17(4), 553–​558. magnetic, and electromagnetic fields (up to 300 GHz). International
Feychting M, Schulgen G, Olsen JH, and Ahlbom A. 1995. Magnetic fields and Commission on Non-​Ionizing Radiation Protection. Health Phys, 74(4),
childhood cancer: a pooled analysis of two Scandinavian studies. Eur J 494–​522.
Cancer, 31A(12), 2035–​2039. ICNIRP. 2010. Guidelines for limiting exposure to time-​varying electric and
Fisher JL, Schwartzbaum JA, Wrensch M, and Wiemels JL. 2007. Epidemiology magnetic fields (1 Hz to 100 kHz). Health Phys, 99(6), 818–​836.
of brain tumors. Neurol Clin, 25(4), 867–​890, vii. IEEE. 2002. Std C95.6–​2002, IEEE standard for safety levels with respect to
Foliart DE, Pollock BH, Mezei G, et  al. 2006. Magnetic field exposure and human exposure to electromagnetic fields, 0–​3 kHz. https://​standards.
long-​term survival among children with leukaemia. Br J Cancer, 94(1), ieee.org/​findstds/​standard/​C95.6-​2002.html. Accessed June 26, 2016.
161–​164. PMCID: PMC2361064. IEEE. 2005. Std C95.1–​2005, IEEE standard for safety levels with respect
Forssen UM, Ahlbom A, and Feychting M. 2002. Relative contribution of to human exposure to radio frequency electromagnetic fields, 3 kHz–​
residential and occupational magnetic field exposure over twenty-​ 300 GHz. https://​standards.ieee.org/​findstds/​standard/​C95.1-​2005.html.
four hours among people living close to and far from a power line. Accessed June 26, 2016.
Bioelectromagnetics, 23(3), 239–​244. Inskip PD, Hoover RN, and Devesa SS. 2010. Brain cancer incidence trends
Frei P, Poulsen AH, Johansen C, et al. 2011. Use of mobile phones and risk in relation to cellular telephone use in the United States. Neuro Oncol,
of brain tumours:  update of Danish cohort study. BMJ, 343, d6387. 12(11), 1147–​1151. PMCID: PMC3098028.
PMCID: PMC3197791. Interphone Study Group. 2010. Brain tumour risk in relation to mobile tele-
Gobba F, Bravo G, Rossi P, Contessa GM, and Scaringi M. 2011. Occupational phone use: results of the INTERPHONE international case-​control study.
and environmental exposure to extremely low frequency-​ magnetic Int J Epidemiol, 39(3), 675–​694.
fields: a personal monitoring study in a large group of workers in Italy. J Interphone Study Group. 2011. Acoustic neuroma risk in relation to mobile
Expo Sci Environ Epidemiol, 21(6), 634–​645. telephone use:  results of the INTERPHONE international case-​control
Greenland S, Fischer HJ, and Kheifets L. 2016. Methods to explore uncertainty study. Cancer Epidemiol, 35(5), 453–​464.
and bias introduced by job exposure matrices. Risk Anal, 36(1):74–​82. Johansen C, Boice J, Jr, McLaughlin J, and Olsen J. 2001. Cellular telephones
Greenland S, and Kheifets L. 2006. Leukemia attributable to residential mag- and cancer-​-​a nationwide cohort study in Denmark. J Natl Cancer Inst,
netic fields:  results from analyses allowing for study biases. Risk Anal, 93(3), 203–​207.
26(2), 471–​482. Johansen C, Raaschou Nielsen O, Olsen JH, and Schüz J. 2007. Risk for
Greenland S, Sheppard AR, Kaune WT, Poole C, and Kelsh MA. 2000. A leukaemia and brain and breast cancer among Danish utility work-
pooled analysis of magnetic fields, wire codes, and childhood leuke- ers:  a second follow-​ up. Occup Environ Med, 64(11), 782–​ 784.
mia. Childhood Leukemia-​EMF Study Group. Epidemiology, 11(6), PMCID: PMC2078420.
624–​634. Kheifets L, Ahlbom A, Crespi CM, et al. 2010a. Pooled analysis of recent stud-
Grellier J, Ravazzani P, and Cardis E. 2014. Potential health impacts of resi- ies on magnetic fields and childhood leukaemia. Br J Cancer, 103(7),
dential exposures to extremely low frequency magnetic fields in Europe. 1128–​1135. PMCID: PMC2965855.
Environ Int, 62, 55–​63. Kheifets L, Ahlbom A, Crespi CM, et al. 2010b. A pooled analysis of extremely
Groves FD, Page WF, Gridley G et  al. 2002. Cancer in Korean war navy low-​frequency magnetic fields and childhood brain tumors. Am J
technicians:  mortality survey after 40  years. Am J Epidemiol, 155(9), Epidemiol, 172(7), 752–​761. PMCID: PMC2984256.
810–​818. Kheifets L, Monroe J, Vergara X, Mezei G, and Afifi AA. 2008. Occupational
Ha M, Im H, Lee M, et  al. 2007. Radio-​frequency radiation exposure from electromagnetic fields and leukemia and brain cancer: an update to two
AM radio transmitters and childhood leukemia and brain cancer. Am J meta-​analyses. J Occup Environ Med, 50(6), 677–​688.
Epidemiol, 166(3), 270–​279. Kiyohara K, Wake K, Watanabe S, et al. 2015. Recall accuracy of mobile phone
Hardell L, Carlberg M, and Hansson Mild K. 2005. Case-​control study on calls among Japanese young people. J Expo Sci Environ Epidemiol.
cellular and cordless telephones and the risk for acoustic neuroma or Mar 18. doi: 10.1038/​jes.2015.13. [Epub ahead of print]
 273

Electromagnetic Fields 273
Klaeboe L, Blaasaas KG, Haldorsen T, and Tynes T. 2005. Residential and SCENIHR. 2009. Scientific Committee on Emerging and Newly Identified
occupational exposure to 50-​Hz magnetic fields and brain tumours in Health Risks:  Health Effects of Exposure to EMF. http://​ec.europa.eu/​
Norway: a population-​based study. Int J Cancer, 115(1), 137–​141. health/​archive/​ph_​risk/​committees/​04_​scenihr/​docs/​scenihr_​o_​022.pdf.
Koeman T, van den Brandt PA, Slottje P, et al. 2014. Occupational extremely Accessed October 19, 2012.
low-​frequency magnetic field exposure and selected cancer outcomes in a SCENIHR. 2015. Scientific Committee on Emerging and Newly Identified
prospective Dutch cohort. Cancer Causes Control, 25(2), 203–​214. Health Risks:  Potential Health Effects of Exposure to Electromagnetic
Kohler BA, Ward E, McCarthy BJ, et  al. 2011. Annual report to the nation Fields (EMF). http://​ec.europa.eu/​health/​scientific_​committees/​emerg-
on the status of cancer, 1975–​ 2007, featuring tumors of the brain ing/​docs/​scenihr_​o_​041.pdf. Accessed August 15, 2015.
and other nervous system. J Natl Cancer Inst, 103(9), 714–​ 736. Schoemaker MJ, Swerdlow AJ, Ahlbom A, et al. 2005. Mobile phone use and risk
PMCID: PMC3086878. of acoustic neuroma: results of the Interphone case-​control study in five North
Lahkola A, Auvinen A, Raitanen J, et al. 2007. Mobile phone use and risk of European countries. Br J Cancer, 93(7), 842–​848. PMCID: PMC2361634.
glioma in 5 North European countries. Int J Cancer, 120(8), 1769–​1775. Schüz J. 2007. Implications from epidemiologic studies on magnetic fields and
Lahkola A, Salminen T, Raitanen J, et al. 2008. Meningioma and mobile phone the risk of childhood leukemia on protection guidelines. Health Phys,
use: a collaborative case-​control study in five North European countries. 92(6), 642–​648.
Int J Epidemiol, 37(6), 1304–​1313. Schüz J. 2009. Lost in laterality: interpreting “preferred side of the head during
Larjavaara S, Feychting M, Sankila R et al. 2011a. Incidence trends of vestibu- mobile phone use and risk of brain tumour” associations. Scand J Public
lar schwannomas in Denmark, Finland, Norway and Sweden in 1987–​ Health, 37(6), 664–​667.
2007. Br J Cancer, 105(7), 1069–​1075. PMCID: PMC3185956. Schüz J, Böhler E, Berg G, et al. 2006a. Cellular phones, cordless phones, and
Larjavaara S, Schüz J, Swerdlow A, et al. 2011b. Location of gliomas in rela- the risks of glioma and meningioma (Interphone Study Group, Germany).
tion to mobile telephone use: a case-​case and case-​specular analysis. Am Am J Epidemiol, 163(6), 512–​520.
J Epidemiol, 174(1), 2–​11. Schüz J, Elliott P, Auvinen A, et al. 2011a. An international prospective cohort
Lerchl A, Klose M, Grote K, et  al. 2015. Tumor promotion by exposure to study of mobile phone users and health (Cosmos): design considerations
radiofrequency electromagnetic fields below exposure limits for humans. and enrolment. Cancer Epidemiol, 35(1), 37–​43.
Biochem Biophys Res Comm, 459(4), 585–​590. Schüz J, Grell K, Kinsey S, et al. 2012. Extremely low-​frequency magnetic fields
Li W, Ray RM, Thomas DB, et al. 2013. Occupational exposure to magnetic and survival from childhood acute lymphoblastic leukemia:  an interna-
fields and breast cancer among women textile workers in Shanghai, tional follow-​up study. Blood Cancer J, 2, e98. PMCID: PMC3542478.
China. Am J Epidemiol, 178(7), 1038–​1045. PMCID: PMC3783098. Schüz J, Jacobsen R, Olsen JH, et al. 2006b. Cellular telephone use and cancer
Linet MS, Hatch EE, Kleinerman RA, et al. 1997. Residential exposure to mag- risk: update of a nationwide Danish cohort. J Natl Cancer Inst, 98(23),
netic fields and acute lymphoblastic leukemia in children. N Engl J Med, 1707–​1713.
337(1), 1–​7. Schüz J, Philipp J, Merzenich H, Schmiedel S, and Bruggemeyer H. 2008.
Little MP, Rajaraman P, Curtis RE, et al. 2012. Mobile phone use and glioma Re: Radio-​frequency radiation exposure from AM radio transmitters and
risk: comparison of epidemiological study results with incidence trends in childhood leukemia and brain cancer. Am J Epidemiol, 167(7), 883–​884.
the United States. BMJ, 344, e1147. PMCID: PMC3297541. Schüz J, Steding-​Jessen M, Hansen S, et al. 2011b. Long-​term mobile phone
Lonn S, Ahlbom A, Hall P, and Feychting M. 2005. Long-​term mobile phone use and the risk of vestibular schwannoma: a Danish nationwide cohort
use and brain tumor risk. Am J Epidemiol, 161(6), 526–​535. study. Am J Epidemiol, 174(4), 416-​422.
Martin-​Lorenzo A, Hauer J, Vicente-​Duenas C, et al. 2015. Infection exposure Schüz J, Svendsen AL, Linet MS, et al. 2007. Nighttime exposure to electro-
is a causal factor in B-​cell precursor acute lymphoblastic leukemia as a magnetic fields and childhood leukemia: an extended pooled analysis. Am
result of Pax5-​inherited susceptibility. Cancer Discov, 5(12), 1328–​1343. J Epidemiol, 166(3), 263–​269.
Maslanyj M, Simpson J, Roman E, and Schüz J. 2009. Power frequency mag- Shu X, Ahlbom A, and Feychting M. 2012. Incidence trends of malig-
netic fields and risk of childhood leukaemia: misclassification of expo- nant parotid gland tumors in Swedish and nordic adults 1970 to 2009.
sure from the use of the ‘distance from power line’ exposure surrogate. Epidemiology, 23(5), 766–​767.
Bioelectromagnetics, 30(3), 183–​188. Stangerup SE, Tos M, Thomsen J, and Caye-​Thomasen P. 2010. True incidence of
McBride ML, Gallagher RP, Theriault G, et al. 1999. Power-​frequency elec- vestibular schwannoma? Neurosurgery, 67(5), 1335–​1340; discussion 1340.
tric and magnetic fields and risk of childhood leukemia in Canada. Am J Stevens RG. 1987. Electric power use and breast cancer: a hypothesis. Am J
Epidemiol, 149(9), 831–​842. Epidemiol, 125(4), 556–​561.
McKean-​Cowdin R, Razavi P, Barrington-​Trimis J, et  al. 2013. Trends in Swerdlow AJ, Feychting M, Green AC, Leeka Kheifets LK, and Savitz DA. 2011.
childhood brain tumor incidence, 1973–​2009. J Neurooncol, 115(2), Mobile phones, brain tumors, and the interphone study: where are we now?
153–​160. Environ Health Perspect, 119(11), 1534–​1538. PMCID: PMC3226506.
Merzenich H, Schmiedel S, Bennack S, et al. 2008. Childhood leukemia in Tell RA, Kavet R, and Mezei G. 2013. Characterization of radiofrequency
relation to radio frequency electromagnetic fields in the vicinity of TV field emissions from smart meters. J Expo Sci Environ Epidemiol, 23(5),
and radio broadcast transmitters. Am J Epidemiol, 168(10), 1169–​1178. 549–​553.
Mezei G, Gadallah M, and Kheifets L. 2008. Residential magnetic field expo- Thomsen J, and Tos M. 1990. Acoustic neuroma: clinical aspects, audiovestibu-
sure and childhood brain cancer: a meta-​analysis. Epidemiology, 19(3), lar assessment, diagnostic delay, and growth rate. Am J Otol, 11(1), 12–​19.
424–​430. Tillmann T, Ernst H, Streckert J, et  al. 2010. Indication of cocarcinogenic
Morgan RW, Kelsh MA, Zhao K, et  al. 2000. Radiofrequency exposure and potential of chronic UMTS-​modulated radiofrequency exposure in an
mortality from cancer of the brain and lymphatic/​hematopoietic systems. ethylnitrosourea mouse model. Int J Radiat Biol, 86(7), 529–​541.
Epidemiology, 11(2), 118–​127. Turner MC, Benke G, Bowman JD, et  al. 2014. Occupational exposure to
Muscat JE, Malkin MG, Thompson S, et al. 2000. Handheld cellular telephone extremely low-​frequency magnetic fields and brain tumor risks in the
use and risk of brain cancer. JAMA, 284(23), 3001–​3007. INTEROCC study. Cancer Epidemiol Biomarkers Prev, 23(9), 1863–​
National Toxicology Program. 2014. Cell Phone Radiofrequency Radiation 1872. PMCID: PMC4154968.
Studies. http://​www.niehs.nih.gov/​health/​assets/​docs_​a_​e/​cell_​phone_​ Tynes T, and Haldorsen T. 1997. Electromagnetic fields and cancer in chil-
radiofrequency_​radiation_​studies_​508.pdf. Accessed December dren residing near Norwegian high-​voltage power lines. Am J Epidemiol,
19, 2015. 145(3), 219–​226.
Pettersson D, Bottai M, Mathiesen T, Prochazka M, and Feychting M. 2015. Tynes T, and Haldorsen T. 2003. Residential and occupational exposure to 50
Validation of self-​reported start year of mobile phone use in a Swedish Hz magnetic fields and hematological cancers in Norway. Cancer Causes
case-​control study on radiofrequency fields and acoustic neuroma risk. Control, 14(8), 715–​720.
J Expo Sci Environ Epidemiol, 25(1), 72–​79. UKCCS. 1999. Exposure to power-​frequency magnetic fields and the risk of child-
Pettersson D, Mathiesen T, Prochazka M, et al. 2014. Long-​term mobile phone hood cancer. UK Childhood Cancer Study Investigators. Lancet, 354(9194),
use and acoustic neuroma risk. Epidemiology, 25(2), 233–​241. 1925–​1931.
Reiter RJ. 1995. Reported biological consequences related to the suppression Verkasalo PK, Pukkala E, Hongisto MY, et  al. 1993. Risk of cancer in
of melatonin by electric and magnetic field exposure. Integr Physiol Finnish children living close to power lines. BMJ, 307(6909), 895–​899.
Behav Sci, 30(4), 314–​330. PMCID: PMC1679080.
Repacholi MH, Basten A, Gebski V, et  al. 1997. Lymphomas in E mu-​Pim1 Verkasalo PK, Pukkala E, Kaprio J, Heikkila KV, and Koskenvuo M. 1996. Magnetic
transgenic mice exposed to pulsed 900 MHZ electromagnetic fields. fields of high voltage power lines and risk of cancer in Finnish adults: nation-
Radiat Res, 147(5), 631–​640. wide cohort study. BMJ, 313(7064), 1047–​1051. PMCID: PMC2352388.
274

274 PART III:  THE CAUSES OF CANCER


Vrijheid M, Armstrong BK, Bedard D, et al. 2009a. Recall bias in the assess- Wertheimer N, and Leeper E. 1979. Electrical wiring configurations and child-
ment of exposure to mobile phones. J Expo Sci Environ Epidemiol, 19(4), hood cancer. Am J Epidemiol, 109(3), 273–​284.
369–​381. WHO. 2007. World Health Organization. Environmental Health Criteria (EHC)
Vrijheid M, Cardis E, Armstrong BK, et al. 2006. Validation of short term recall Document on ELF Fields, Doc. No. 238, downloadable from the WHO
of mobile phone use for the Interphone study. Occup Environ Med, 63(4), EMF Project website, www.who.int/​emf.
237–​243. PMCID: PMC2078087.
Vrijheid M, Richardson L, Armstrong BK, et al. 2009b. Quantifying the impact
of selection bias caused by nonparticipation in a case-​control study of
mobile phone use. Ann Epidemiol, 19(1), 33–​41.
 275

16 Occupational Cancer

KYLE STEENLAND, SHELIA HOAR ZAHM, AND A. BLAIR

OVERVIEW of studies of cancer in the workplace. Exposures in the workplace are


often considerably higher than those in the non-​occupational setting.
Occupational cancer was one of the earliest areas of cancer research, Occupational exposures can often be measured more precisely than
as astute clinicians observed clusters of cancer in workers exposed to non-​occupational exposures because of the availability of production
high levels of workplace toxins. These early observations were fol- records, personal and area measurements, and operating procedures,
lowed by some of the earliest epidemiologic cohort studies, beginning which provide information that can be used to characterize exposures.
after World War II, which followed the leads from the early clusters. Occupational studies are needed to protect workers from workplace
Many of today’s recognized carcinogens were discovered via occu- hazards. Many workers have little flexibility regarding how tasks are
pational studies, including classic carcinogens such as asbestos and performed, equipment and ingredients used, and the location of work,
arsenic, as well as more recently established carcinogens, such as and thus have relatively little individual control over their personal
silica and diesel fumes. The earlier discoveries were characterized by exposures. Occupational exposures are, therefore, largely involun-
high exposures and high risks; later discoveries of agents with lower tary. Although the excesses of cancer from occupational exposures
exposure and lower risks have required a multiplicity of large studies may occur in worker groups of relatively small size, the high level of
to establish causality, and to provide sufficient data for risk assessment exposure can result in large relative risks. Because of this, over three
to be performed by regulatory agencies. It is likely that this pattern decades ago Doll and Peto (1981) noted that “detection of occupational
will continue, with cancer risks from more difficult to characterize hazards should therefore have a higher priority in any program of can-
exposures like shift work, currently considered a probable but not defi- cer prevention than their proportional importance might suggest.” This
nite carcinogen, requiring a large body of evidence before acceptance. conclusion is still true today. It should be noted that many occupational
Most occupational exposures recognized in the past were of a chemi- exposures also occur in non-​occupational settings and have adverse
cal or particle nature. A more expansive view of occupational expo- impacts beyond the workplace (e.g., arsenic, diesel fumes, polychlo-
sures today should consider a broader range of infectious, medical, rinated biphenyls [PCBs], dioxins, polycyclic aromatic hydrocarbons,
behavioral, and physical factors that might impact cancer, such as shift and pesticides).
work, sedentary behavior, or workplace stress. Despite the many established links between occupational expo-
Occupational carcinogen discovery has always been controversial sures and cancer, much remains unknown. A starting place for a list of
because such findings have economic as well as public health impli- needed studies could be those occupational exposures already classified
cations. Consequently, decision-​making and action may involve more as possible or probable human carcinogens by the IARC. These often
political activity than findings in other epidemiologic areas. The exam- have limited or no epidemiologic information, but are already suspect,
ple of diesel exhaust, recently declared a definite carcinogen by the and clarity is needed. The selection priority for the agents on this list
International Agency for Research on Cancer (IARC) after 25 years of would depend upon factors such as number of workers exposed, typical
controversy, is a good example (see later discussion in this chapter). levels of exposure, likely magnitude of relative risk, opportunities for
In this chapter we summarize the past history of occupational can- high-​quality investigations, and the possibility of exposures outside the
cer epidemiology, and indicate which occupational exposures are pres- workplace. In addition, there are many occupations or industries where
ently considered to be definite or probable carcinogens. We describe cancer excesses have been noted, but specific agents that might be
the basic study designs of occupational cancer research, which are involved have not been clearly identified. Despite the obvious need for
similar to the designs of non-​occupational studies, but which have additional investigations (Blair et al., 2011; Siemiatycki, 2014; Straif,
their own peculiarities, particularly in regard to exposure assessment. 2012; Ward et al., 2010), the number of publications on cancer in the
We discuss the type of evidence that has stimulated occupational can- workplace is decreasing and is now only about one-​third the number of
cer studies and how data generated by occupational cancer studies are just a decade ago (Raj et al., 2014). Explanations for this decline are not
used in risk assessment for workplace regulations, and the calculation clear, but may include a change in the political/​social climate that no
of attributable fractions to quantify the burden of occupational cancer. longer supports this type of health research, a decrease in heavy indus-
Finally, we discuss some current controversies and propose likely try in high-​income countries, a weakening of labor unions which have
future directions for occupational epidemiology. These include a focus encouraged research, cycles of focus and popularity in public health
on “exposures” like shift work and sedentary work habits, which are research, a belief that control of hazardous occupational exposures has
not traditional toxins. In addition, it will be important to document the been accomplished because of previous successes, and the failure in
carcinogenic effects of established occupational carcinogens in less recent times to find occupational exposures with such dramatic impacts
developed countries where they have not been studied, as a means to on cancer as previously identified agents such as asbestos, benzidine,
affect policy and ensure safe workplaces. vinyl chloride, benzene, and chromium (Blair et al., 2011; Raj et al.,
2014; Siemiatycki, 2014).
Most recognized occupational carcinogens have been identified
INTRODUCTION through studies of men in high-​income countries (Hohenadel et al.,
2015; Zahm et al., 1994, 1999). The inclusion of women in studies
Studies of workplace exposures have provided a wealth of information of the workplace has increased since the early 1990s, but studies
regarding the causes of cancer from early (Ramazzini, 1713) to recent of men still predominate and typically have more detailed analyses
times (Siemiatycki, 2014). Approximately 40% of the factors classified on men than on women, possibly due to small numbers or a lower
as sufficient, probable, or possible human carcinogens by the IARC priority placed on investigating risks among women (Hohenadel
were initially investigated as occupational exposures (Siemiatycki et al., 2015). Furthermore, in the past few decades, low-​and mid-
et al., 2004). There are many practical and disease prevention benefits dle-​income countries have assumed a larger and larger fraction of

275
276

276 PART III:  THE CAUSES OF CANCER


the world’s production in many industries, but there does not yet mortality in the areas where the plants were located, compared with
appear to have been a comparable increase in investigations of occu- other areas in England.
pational exposures in most low-​income countries (Raj et al., 2014). Asbestos had been known to cause asbestosis since before World
Studies are particularly needed in low-​and middle-​income countries War II. There were case reports of lung cancer among asbestos work-
because exposures may be less well controlled (Siemiatycki, 2014). ers, and some published literature began to draw links between the
two before the war (Greenberg, 1999). However, the first solid epide-
miologic evidence came from the landmark cohort mortality study of
HISTORY asbestos workers by Doll in 1955, which showed the lung cancer risk
among men employed for 20 or more years to be 10 times that of the
The era of modern occupational epidemiology, with systematic assem- general population (Doll, 1955). The first strong epidemiologic evi-
bling and follow-​up of occupational cohorts, did not begin until after dence for a link between asbestos and mesothelioma followed in 1960
World War II. Most early identification of the occupational origin of among asbestos miners in South Africa (Wagner et al., 1960). Wagner
some cancers was done by astute clinicians seeing cancer in clear et al. (1960) described a case series of 33 cases living near or working
excess (i.e., clusters) among workers who were exposed at the time. in crocidolite asbestos mines (i.e., a cluster). The fact that the tumor
Perhaps the earliest identification of occupational cancer was particu- was so rare and did not occur elsewhere in South Africa provided com-
larly prescient. Bernardo Ramazzini, the father of occupational medi- pelling evidence of a link. The evidence of the relationship is now so
cine, noted in 1713 that nuns, who can be considered members of a strong that mesothelioma is considered a sentinel tumor or marker for
specific occupation, were more likely to have breast cancer than other asbestos exposure (Friedman, 2011).
women. He speculated that this might be due to a lack of sexual activ- Following these classic early studies, subsequent cohort studies
ity (Olson, 2002). Today we know that Ramazzini was right about the found excess cancer associated with a large number of occupational
phenomenon but (partly) wrong about the cause, which we now attri- exposures, including dioxin, radon in mines, ethylene oxide, silica,
bute to nulliparity and increased exposure of the breast to estrogen. vinyl chloride, diesel fumes, cadmium, benzene, beryllium, PCBs,
Ramazzini also described diseases among miners, painters, weavers, wood dust, acid mists, and others all of which are classified as IARC
and printers. Group 1 (definite) human carcinogens, as detailed in the next section
Another famous clinician who identified occupational cancer was of this chapter. Ionizing radiation, beyond the radon that caused lung
Percival Pott, who noted in 1775 that chimney sweepers in London had cancer among miners (Darby et  al., 1995; IARC, 2012d), has also
an excess of scrotal cancer, which he attributed to soot accumulation in been identified as an occupational carcinogen. Long-​term follow-​up
the scrotum (Dobson, 1972; Pott, 1775; Waldron, 1983). Subsequently of nuclear workers from three countries, exposed primarily to γ-​radia-
there were other reports of scrotal cancer among other workers exposed tion, showed the risk of developing leukemia, particularly chronic
to soot, also known as coal tar. Coal tar can be considered the first rec- myeloid leukemia, to be linked to the protracted low doses of radiation
ognized occupational carcinogen, for which workmen’s compensation (Leuraud et  al., 2015). Nuclear weapons workers exposed to pluto-
was given in England as of 1907 (Waldron, 1983). Another clinician nium had increased risk of lung cancer, liver cancer, and bone sarcoma
who identified an early occupational carcinogen was Ludwig Rehn, (Gilbert, et al., 2000, 2004; IARC, 2012d; Koshurnikova et al., 2000).
who reported a cluster of bladder cancers among aniline dye workers Workers involved in cleanup after the Chernobyl nuclear accident
in 1895 (Dietrich and Dietrich, 2001). In the 1920s, mineral oil was were exposed to X-​and γ-​radiation and have excess risk of leukemia,
also recognized as a cause of scrotal cancer, and other skin cancers, including chronic lymphocytic leukemia (Kesminiene et  al., 2008;
due to a cluster of cases among cotton spinners (Waldron, 1983). Zablotska et al., 2013).
Kuroda and Kawahata identified a cluster of lung cancer among Several of the carcinogens mentioned in the preceding paragraph
workers exposed to coal gas in steel mills in Japan in 1935 (Takahashi are also present in the general environment at lower levels (e.g.,
and Ishii, 2013). In one of the earliest examples of calculation of stan- dioxin, diesel fumes, PCBs, indoor radon). In addition, two occupa-
dardized mortality ratios (SMRs) using death certificates and census tions, painting and foundry work, have been identified by the IARC as
data by occupation, Kennaway and Kennaway (1936) showed that coal increasing cancer risk (Group 1), without being able to specify which
gas workers in England had excess cancer of the lung (coal gas was specific carcinogens are responsible.
later replaced by coke in steel mills, which also caused lung cancer) While most occupational carcinogens have been discovered via
(Lloyd, 1971). Other notable clusters included bone and blood cancer retrospective cohort studies, large population-​ based case-​ control
in radium dial painters, identified by Harrison Martland in New Jersey studies have often confirmed these findings, using self-​reported job
in 1931 (Loutit, 1970), and lung cancer in nickel refinery workers in histories and assessment of potential occupational exposures by indus-
Wales, identified by Bridge in 1933 (Bridge, 1933). There were also trial hygienists. Perhaps the most important model for this approach
several German case reports of lung cancer among chromate workers has been the numerous studies in Montreal by the group led by Jack
in the 1930s (Hayes et al., 1988). Siemiatycki (Siemiatycki et al., 1991, 1997). Siemiatycki’s team elic-
After World War II, occupational epidemiologists, primarily in ited detailed job histories through a semi-​structured questionnaire
England, systematically began using retrospective cohort studies with supplementary questionnaires for selected occupations, followed
based on industry records to study cancer mortality. Defining the by comprehensive review by chemists and industrial hygienists, who
study population as all workers in a particular job or industry and translated each job into a list of potential exposures using a checklist
following them over time, instead of studying case series or clusters of 294 agents.
among current workers only, allowed the inclusion of cancers occur-
ring after leaving work, and allowed statistical comparisons of cancer
rates in exposed workers versus low-​or non-​exposed referents. Most ACCEPTED HUMAN OCCUPATIONAL CARCINOGENS
of the previously observed clusters were confirmed in such studies,
including bladder cancer in dye workers (Case et al., 1954), arsenic Several groups regularly convene expert panels to evaluate the car-
and lung/​skin cancer (Hill and Faning, 1948), chromates and lung cinogenicity of agents, with the most long-​standing being the IARC,
cancer (Machle and Gregorius, 1948), nickel refining and lung can- an agency of the World Health Organization. The IARC Monograph
cer (Doll, 1958), and coal gas and lung cancer (Doll, 1952). Radium Program assembles interdisciplinary working groups to evaluate epi-
decays into radon gas, and after World War II investigators linked demiologic, experimental, exposure, and mechanistic data to classify
radon in uranium miners to lung cancer (Wagoner et al., 1965). All agents, mixtures, or exposure circumstances as carcinogenic (Group 1),
of these studies were retrospective cohort mortality studies, with two probably carcinogenic (Group 2A), possibly carcinogenic (Group 2B),
exceptions. In the case of arsenic and nickel, investigators did not have not classifiable (Group 3), or probably not carcinogenic (Group 4) to
access to plant records, and instead conducted studies of proportional humans. Although there are some critics of the IARC process who
 27

Table 16–1.  Agents, Occupations, and Industries Classified by IARC as Human Carcinogens (Group 1) That Are Primarily Occupational Exposures

Agent Cancer Main Industry or Use

Acid mists containing sulfuric acid, strong Larynx Chemical


inorganic
4-​Aminobiphenyl Bladder Rubber
Arsenic and arsenic compounds Lung, skin, bladder Glass, metals, pesticides
Asbestos (all forms) Larynx, lung, pleura, ovary Insulation, construction, renovation
Benzene Leukemia Chemical production, solvent, fuel
Benzidine Bladder Dye/​pigment manufacture
Benzo(a)pyrene N/​A* Coal liquefaction and gasification, coke
production, coke ovens, roofing, paving,
aluminum production
Beryllium and beryllium compounds Lung Aerospace industry, metals
Bis(chloromethyl) ether; chloromethyl Lung Chemical intermediate/​byproduct
methyl ether
1,3-​Butadiene Leukemia, lymphoma Plastic, rubber
Cadmium and cadmium compounds Lung Dye/​pigment manufacture, batteries
Chromium (VI) compounds Nasal cavity, lung Metal plating, dye/​pigment manufacture
Coal tar pitch Skin, lung Construction, electrodes
1,2-​Dichloropropane Biliary tract Chemical, paint stripping, printing
Diesel engine exhaust Lung Transport, mining
Erionite Pleura Hydrocarbon cracking, agriculture
Ethylene oxide N/​A* Chemical, sterilant
Fission products, including strontium-​90 Leukemia, lymphoma Nuclear workers
Formaldehyde Nasopharynx, leukemia Plastics, textiles
Gallium arsenide N/​A* Semiconductors
Ionizing radiation (all types, including Thyroid, leukemia, salivary gland, lung, Radiology, nuclear industry, underground mining
radon-​222 Progeny) bone, esophagus, stomach, colon, rectum,
skin, breast, kidney, bladder, brain
Leather dust Nasal cavity Leather industry
4,4′-​Methylenebis (2-​chloroaniline) (MOCA) N/​A* Rubber
Mineral oils, untreated and mildly treated Skin Lubricant
β-​Naphthylamine Bladder Pigment
Nickel compounds Nasal cavity, lung Metal alloy
3,4,5,3′,4′-​Pentachlorobiphenyl (PCB-​126) N/​A* Electrical components
2,3,4,7,8-​Pentachlorodibenzofuran N/​A* Incineration, smelting, refining
Polychlorinated biphenyls Skin Electrical components
Polychlorinated biphenyls (“dioxin-​like”) N/​A* Electrical components
Shale oils Skin Lubricant, fuel
Silica dust, crystalline, in the form of Lung Construction, mining
quartz or cristobalite
Solar radiation Skin Outdoor workers
Soot Skin, lung Chimney sweeps, masons, firefighters
Sulfur mustard Lung War gas
2,3,7,8-​Tetrachlorodibenzo-​p-​dioxin N/​A* Chemical
Tobacco, secondhand Lung Restaurant and bar workers, offices
Ortho-​Toluidine Bladder Pigment
Trichloroethylene Kidney Solvent, dry cleaning
Ultraviolet radiation Skin, eye Outdoor workers
Vinyl chloride Liver Plastic
Wood dust Nasal cavity Wood, furniture

Occupation or Industry Cancer Main Industry or Use

Acheson process Lung Graphite and silicon carbide production


Aluminum production Lung, bladder Dye manufacture
Auramine, production of Bladder Dye manufacture
Coal gasification Skin, lung, bladder Power plants, chemical industry
Coal-​tar distillation Skin Coke oven plants
Coke production Skin, lung, kidney Coke oven plants
Hematite mining, underground Lung Mining
Iron and steel founding Lung Iron and steel founding
Isopropyl alcohol manufacture Nasal cavity Chemical
using strong acids
Magenta production Lung, bladder Painting
Painter (occupation as) Lung, bladder Painting
Rubber manufacturing industry Leukemia, lymphoma, bladder, lung, stomach Rubber

Source: Benbrahim et al., 2014; Boyle and Levin, 2008; Grosse et al., 2014; Guyton et al., 2015; IARC, 2013, 2014a, 2014b, 2015c, 2015d, 2015e; Stewart and Wild, 2014.
** N/​A—​Not applicable (agent classified in Group 1 on the basis of mechanistic evidence).
278

278 PART III:  THE CAUSES OF CANCER


claim the reviews have not sufficiently taken into account potential carbide), iron and steel founding, isopropyl alcohol production by the
methodological weaknesses in epidemiologic data in general and may strong-​acid process, magenta production, working as a painter, frying
be biased by working group members with vested interests, there is (emissions from high temperatures), working as a hairdresser or bar-
very broad involvement of the scientific community in the IARC ber, spraying and applying non-​arsenical insecticides, petroleum refin-
Monograph Program, overwhelming support for the scientific rigor ing, and shift work that involves circadian disruption. The exposures
of the process, and ongoing international agreement with its decisions associated with these processes and occupations could change over
(Pearce et al., 2015) time or by geographic location, and change the cancer risk previously
The IARC does not systematically indicate which carcinogens are associated with them. For example, if the cancer risk associated with
occupational, and thus lists of occupational carcinogens may vary occupation as a hairdresser were due to exposure to hair dyes, changes
depending on the criteria used. Table 16–1 presents the agents classi- in the formulations of hair dyes might cause the risk to change over
fied by IARC as Group 1 carcinogens that we consider to be mostly calendar time or geographic region.
occupational exposures. Note that occupational exposures can Table 16–3 presents the most common types of occupational cancer;
involve a broad range of infectious, medical, behavioral, and physi- these are lung cancer, bladder cancer, mesothelioma, and leukemia,
cal factors, in addition to the long-​recognized toxic chemicals. For although occupational exposures have been linked to many cancer
example, chemotherapeutic medications are an occupational expo- sites (IARC, 2015b).
sure for some healthcare workers. Medical radiation workers have In 2008–​2009, the IARC held six working group meetings during
excess leukemia and cancers of the skin and breast related to their which expert panels reviewed the evidence for substances and factors
occupational exposure (Linet et  al., 2010; Yoshinaga et  al., 2004). that the IARC had classified as known (Group  1) carcinogens in the
Tobacco, in the form of secondhand smoke, is an occupational expo- past. This re-​review reaffirmed the earlier Group 1 classifications, and
sure for bar and restaurant workers, resulting from the failure to added new cancer sites for some agents. For example, formaldehyde
ban smoking in these workplaces in some localities. Restrictions on had originally been classified in 2006 as a human carcinogen based
smoking in public indoor areas have recently decreased this expo- on excess risk of nasopharyngeal cancer, but the recent review in 2009
sure in some areas and countries, but not all (Schmidt, 2007). Shift added leukemia, particularly myeloid leukemia, as having sufficient
work adversely affects diurnal rhythm (IARC, 2010a). Physical inac- evidence of carcinogenicity due to formaldehyde (IARC, 2012f). To
tivity and sedentary work are the norm in most modern workplaces date, the accumulation of additional evidence has never caused the
(Brownson et  al., 2005). Sunlight is an occupational exposure for IARC to downgrade an agent that was previously classified as a known
persons who work outdoors (Carey et al., 2014). carcinogen, although some agents classified as possible carcinogens
Agents classified by the IARC as Group 1 carcinogens must have (Group 2B, possibly carcinogenic to humans) have been downgraded
sufficient evidence for carcinogenicity in humans, or exceptionally to Group 3 (inadequate evidence), such as amitrole, atrazine, and eth-
limited evidence in humans but sufficient evidence of carcinogenic- ylene thiourea.
ity in animals, along with strong evidence in exposed humans that
the agent acts through a mechanism relevant to human carcinogenic-
ity (IARC, 2015a). Ethylene oxide, which is used as an intermedi- EVIDENCE STIMULATING EPIDEMIOLOGIC STUDIES
ate in the production of several industrial chemicals, as a fumigant, OF OCCUPATIONAL CANCER
and as a sterilant for medical equipment and supplies, was classified
as a carcinogen based on limited evidence from humans, sufficient There are several types of evidence that by themselves have suggested
evidence from animals, and compelling genotoxicity data from stud- the presence of occupational cancer hazards and have stimulated epide-
ies of exposed workers (IARC, 2012f). Similarly, mechanistic data miologic studies of occupational exposures or groups. These informa-
on the presence of DNA adducts, sister chromatid exchanges, and tive approaches include investigations of cancer clusters, searches for
micronuclei among workers exposed to 4,4'-​Methylenebis (2-​chlo- explanations of unusual geographic patterns, whole animal cancer bio-
roaniline) (MOCA) resulted in its classification as a known human assays, Ames testing, and animal and human biomarker and mechanistic
carcinogen (IARC, 2010b), despite limited human evidence. 2,3,7,8-​ studies.
Tetrachlorodibenzo-​p-​dioxin, which is a byproduct of the synthesis A cancer cluster is the apparent excess of cancer cases among a
of chlorophenols or chlorophenoxy acid herbicides, waste incinera- relatively small group of people. As noted earlier, occupational can-
tion, metal production, and wood combustion, was also classified as cer clusters have led to the identification of carcinogens and in-​depth
a Group 1 carcinogen based on limited human data, sufficient animal epidemiologic studies dating back centuries. In the modern era after
data, and evidence that TCDD acts through a mechanism involving World War II, a plant physician noted three cases of angiosarcoma
the Ah receptor, which is involved in normal cell homeostasis and is of the liver among workers exposed to vinyl chloride monomer in a
present in both humans and animals. Other occupational agents clas- polymerization operation at a chemical plant (Creech and Johnson,
sified as carcinogens based on limited human evidence, but sufficient 1974). The extremely rare nature of angiosarcoma, and the fact
animal data and mechanistic data, include 2,3,4,7,8-​pentachlorod- that three cases came from the same workplace, contributed to the
ibenzofuran, “dioxin-​like” polychlorinated biphenyls, dyes metabo- ability of the physician to recognize the excess cancer incidence.
lized to benzidine, and benzo(a)pyrene. Epidemiologic investigations confirmed the excess of angiosarcoma
Table 16–2 presents the occupational agents classified as probable of the liver and discovered that vinyl chloride also caused hepatocel-
carcinogens (Group 2A), generally based on limited evidence of car- lular carcinoma, a more common cancer that would be less likely to
cinogenicity in humans and sufficient evidence of carcinogenicity in be noted as excessive among an occupational group without a formal
experimental animals. In some cases, agents are classified Group 2A study (IARC, 2012f).
based on lower levels of evidence of carcinogenicity in humans, but A similar story of discovery occurred in 2013 with the observation
with strong evidence that the carcinogenesis is mediated by a mecha- of 11 male patients with intrahepatic or extrahepatic biliary tract cancer
nism that also operates in humans or that the agent clearly belongs, (cholangiocarcioma) among workers exposed to 1,2-​dichloropropane
based on mechanistic considerations, to a class of agents for which among 62 male workers at a printing company in Japan (Kumagai et al.,
one or more members have been classified as Group 1 or Group 2A. 2013). The rarity of cholangiocarcioma, the very high relative risk, the
Agents on this list have considerable potential for human carcinoge- young ages of the patients, the absence of non-​occupational risk fac-
nicity, so those with widespread human exposure have a high prior- tors, and the intensity of the exposure, along with sufficient evidence of
ity for research to resolve the uncertainty in their classification (Ward carcinogenicity among experimental animals, led the IARC to classify
et al., 2010). 1,2-​dichloropropane as a known human carcinogen (Benbrahim-​Tallaa
These lists (Tables 16–1 and 16–2) include some exposure scenar- et al., 2014; Kubo et al., 2014; Kumagai et al., 2013).
ios that have been linked to cancer, but for which the specific agents Geographic patterns of cancer occurrence have provided clues to
responsible have not been identified. These include exposures asso- occupational carcinogens. Publication of the first US cancer mortality
ciated with the Acheson process (synthesis of graphite and silicon atlas in 1975 revealed a string of counties along the southeast Atlantic
 279

Occupational Cancer 279
Table 16–2.  Agents, Occupations, Industries, and Occupational Circumstances Classified by IARC as Probable Human Carcinogens (Group 2A) That
Are Primarily Occupational Exposures

Agent Cancer Main Industry or Use

Acrylamide –​ Plastics
Bitumens (combustion products during roofing) Lung Roofing
Captafol –​ Pesticide
α-​Chlorinated toluenes –​ Pigment, chemical
4-​Chloro-​o-​toluidene Bladder Pigment, textile
Cobalt metal with tungsten carbide Lung Hard metal production
Creosotes Skin Wood
Diazinon Lymphoma, lung Agriculture, pest control
Dibenz[a,j]acridine –​ Mastic asphalt worker
Dichloromethane (methylene chloride) Biliary tract, lymphoma Plastics, solvent, paint stripping
Diethyl sulfate –​ Chemical
Dimethycarbamoyl carbide –​ Chemical
1.2-​Dimethylhydrazine –​ Research
Dimethysulfate –​ Chemical
Epichlorhydrin –​ Plastic
Ethylene dibromide –​ Fumigant
Glycidol –​ Pharmaceutical industry
Glyphosate Lymphoma Agriculture, forestry
Indium phosphide –​ Semiconductors
Lead compounds, inorganic Lung, stomach Metals, pigments
Malathion Lymphoma, prostate Agriculture, pest control
Methyl methanesulfonate –​ Chemical
6-​Nitrochrysene –​ Transportation, underground mining
1-​Nitropyrene –​ Transportation, underground mining
2-​Nitrotoluene –​ Dye production
Non-​arsenical insecticides Leukemia Agriculture
Polybrominated biphenyls –​ Flame retardants, plastics
1,3-​Propane sultone –​ Chemical, batteries
Silicon carbide whiskers Lung Plastics
Styrene-​7,8-​oxide –​ Plastic
Tetrachloroethylene Liver, lymphoma Solvent, dry cleaning
Tetrafluoroethylene –​ Chemical
1,2,3-​Trichloropropane –​ Solvent
Tris(2,3-​dibromopropyl)phosphate –​ Plastic, textile
Vinyl bromide –​ Plastic, textile
Vinyl fluoride –​ Chemical

Occupation, Industry, or Occupational Circumstance Cancer Main Industry or Use

Art glass, glass containers, and pressed ware, manufacture Lung, stomach Glass manufacture
Carbon electrode manufacture Lung Carbon electrode manufacture
Food frying at high temperature Lung Restaurant
Hairdresser or barber Bladder, lung Cosmetology
Petroleum refining Leukemia, skin Petroleum refining
Shift work that involves circadian disruption Breast Health care, manufacturing, custodial work

Source: Benbrahim et al., 2014; Boyle and Levin, 2008; Grosse et al., 2014; Guyton et al., 2015; IARC, 2013, 2014a, 2014b, 2015c, 2015d, 2015e; Stewart and Wild, 2014.

coast with markedly elevated rates of lung cancer (Mason et al., 1975). and the overwhelming majority of established human carcinogens have
A series of case-​control studies conducted in the areas found that the also been shown to cause cancer in animals (Huff, 1999, 2008). There
excess lung cancer was due to asbestos exposures associated with are many chemicals that were found to cause cancer in animals before
short-​term shipyard work that took place largely during World War epidemiologic studies demonstrated effects in humans. Some notable
II (Blot et al., 1978, 1979b, 1980; Tagnon et al., 1980). Many of the carcinogens first identified in animal studies and later found to cause
shipyards had been closed for years. cancer among exposed workers are dioxin (IARC, 1997, 2012f), 1,3-​
In recent time, a common motivation for an epidemiologic study of butadiene (Melnick and Huff, 1992), and formaldehyde (IARC, 2006).
a suspect occupational carcinogen is demonstration of cancer among Concordance overall between animal and human carcinogens, as
experimental animals. Animal bioassays have characteristics that must well as concordance of the specific affected sites, has improved with
be considered when extrapolating the results to humans, such as ani- better bioassay methods, such as improved histopathology with more
mal/​human species metabolic differences, typically greater doses to detailed examination of more organs per test animal, use of vari-
animals than are experienced by humans, different routes of exposure, ous routes of administration, and different forms of the compounds
fewer concomitant exposures that could affect cancer risk, and some (Maronpot et  al., 2004). For example, arsenic was recognized as a
target organs that may not exist in humans (e.g., the rodent Zymbal human carcinogen in 1987, while animal bioassays were negative or
glands) or that rarely develop cancer among humans (e.g., the pituitary provided limited evidence until many years later, when organic metab-
gland). However, despite these limitations, the results of long-​term can- olites of arsenic were studied and found to be positive (Huff et  al.,
cer bioassays are highly predictive for identifying human carcinogens, 2000; IARC, 2012c).
280

280 PART III:  THE CAUSES OF CANCER


Table 16–3.  IARC Human Carcinogens (Group 1) That Are Primarily Table 16–3. Continued
Occupational Exposures by Cancer Site
Cancer Site Occupational Exposure
Cancer Site Occupational Exposure
Pleura Asbestos
Bladder Aluminum production Erionite
4-​Aminobiphenyl Painter (occupation as)
Arsenic and inorganic arsenic
compounds Skin Arsenic and inorganic arsenic
Auramine production compounds
Benzidine Coal-​tar distillation
Magenta production Coal-​tar pitch
2-​Naphthylamine Mineral oils, untreated or mildly treated
Painter (occupational exposure as) Polychlorinated biphenyls
Rubber production industry Shale oils
Ortho-​Toluidine Solar radiation
X-​ and γ-​radiation Soot
X-​ and γ-​radiation
Bone Plutonium
Radium-​224 and its decay products Stomach Rubber production industry
Radium-​226 and its decay products
Radium-​228 and its decay products Source: IARC, 2015b, 2015f.
X-​ and γ-​radiation
Breast X-​ and γ-​radiation Measures of exposure and biomarkers of effects in humans can also be
Eye Welding critical in assessing and understanding carcinogenesis. They have also
stimulated occupational epidemiologic investigations. Early reports of
Gallbladder and biliary tract 1,2-​Dichloropropane
chromosomal damage from ethylene oxide (Ehrenberg and Hallstrom,
Kidney Trichloroethylene 1967), a sterilant gas, were instrumental in leading to two epidemiologic
X-​ and γ-​radiation studies, which documented excess leukemia (Hogstedt et  al., 1979a;
Larynx Acid mists, strong inorganic Hogstedt et al., 1979b). Animal studies subsequently showed that inhaled
Asbestos ethylene oxide caused leukemia in rodents (Snellings et al., 1984).
Leukemia/​Lymphoma Benzene
1,3-​Butadiene
Fission products, including
DESIGN OF HUMAN STUDIES
strontium-​90
Formaldehyde The role of occupational exposures in the development of cancer has
Rubber production industry been investigated using various epidemiologic designs, including cohort,
X-​ and γ-​radiation case-​control, cross-​sectional, and ecological studies (Checkoway et al.,
Liver Plutonium 2007; Steenland and Moe, 2016). The type of study design has a major
impact on the type and source of information available on occupational
Lung Acheson process exposures as well as potential bias, and thus on the quality of the data
Aluminum production generated by the study and the conclusions that can be drawn.
Arsenic and inorganic arsenic
compounds
Asbestos
Beryllium and beryllium compounds
Cohort Studies
Bis(chloromethyl)ether; chloromethyl In a cohort study, typically exposed and non-​exposed groups are iden-
methyl ether (technical grade) tified and followed over time to ascertain disease outcomes (although
Cadmium and cadmium compounds sometimes highly exposed workers are compared to lower exposed
Chromium (VI) compounds
Coal gasification
workers in an internal analysis). The occurrence of disease among the
Coal-​tar pitch two groups is compared. For many occupational studies, the “non-​
Coke production exposed” group has been the general population of the country or some
Diesel engine exhausts other political entity in which the exposed group is located. Such a com-
Hematite mining parison group is typically selected for practical reasons (i.e., information
Iron and steel founding on the incidence or mortality experience is already available, and no spe-
Nickel compounds cific unexposed population can be assembled without considerable effort
Painter (occupational exposure as) and cost). Although use of a non-​worker referent population has practi-
Plutonium cal advantages, it has a methodological limitation known as the healthy
Radon-​222 and its decay products worker effect. This occurs when the exposed population (i.e., workers)
Silica dust, crystalline
Soot
is made up of relatively healthy individuals, while the referent group
Sulfur mustard includes non-​working people who are not employed because of health
Tobacco smoke, secondhand problems, so the two groups are not comparable (Fox and Collier 1976).
X-​ and γ-​radiation The healthy worker effect can take two forms; one involves the selection
of healthy workers into the workforce, and a second involves healthy
Nasal cavity and paranasal sinus Isopropyl alcohol manufacture using
strong acids workers staying in the workforce while others who develop illnesses
Leather dust leave the workforce early (the latter is called the healthy worker survivor
Nickel compounds effect and is important when considering cumulative exposures). The
Radium-​226 and its decay products healthy worker effect can obscure an increase in disease among work-
Radium-​228 and its decay products ers due to an occupational exposure, which may be offset by the gener-
Wood dust ally healthy nature of the working population. The healthy worker effect
Nasopharynx Formaldehyde is a particular problem for diseases that are disabling and which may
Wood dust occur relatively early in life, such as cardiovascular disease, but it is not
entirely absent for cancer. However, the healthy worker effect wears off
Ovary Asbestos
 281

Occupational Cancer 281
with further follow-​up, when more of the cohort is no longer working, observations on deaths only. Full information on the occupational
which is when most cancers occur. When a referent group of workers is group from which the deaths arose is not available. Without enumera-
available, its use not only avoids the healthy worker effect, but also has tion of the population at risk, it is not possible to calculate incidence or
the advantage that it is likely to be similar to the exposed population mortality rates and compare them to an unexposed population. Instead,
in other often unmeasured ways (e.g., sharing lifestyle factors such as the proportion of deaths due to a specific cause out of the total number
smoking habits). Thus it can help limit unmeasured confounding. of deaths among the workers is compared to the proportion in a com-
A cohort study can assess multiple outcomes (e.g., all types of can- parison population, often the general population. The statistic used to
cer), but unless the cohort is extremely large, the number of subjects express the results, the proportional mortality ratio, is the origin of
with rare diseases will be small. The cohort design is particularly the common name for this design, the PMR study. This approach is
useful for the study of occupational exposures that would otherwise most common when a company, union, government agency, or insur-
be uncommon in most samples of the general population. An increas- ance company may have death certificates available for study, but no
ing challenge, particularly in the United States, is gaining access to records on the identity or work histories for all persons who worked
workplaces and to the historical occupational and exposure infor- in the job of interest, regardless of their current employment or vital
mation necessary to conduct cohort studies. In the United States, status. Examples include the NIOSH National Occupational Mortality
until recently most occupational cohort studies ascertained mortality Surveillance (NOMS) system, which has occupation and industry-​
only, not cancer incidence, which means that some associations with coded death data from 30 US states from over a 20-​year time period
cancers that have good survival (e.g., bladder, prostate, and breast (Robinson et  al., 2015), a study of deaths among unionized plumb-
cancer) may have been missed. With better coverage of the United ers, pipefitters, and allied trades (Lehman et al., 2008), and a study of
States by cancer registries, more incidence-​ based occupational deaths among cement workers recorded by a Greek insurance com-
cohorts are appearing, such as the study of US firefighters (Daniels pany (Rachiotis et al., 2012).
et al., 2014) and the Agricultural Health Study (Blair et al., 2015b). Proportional mortality studies are generally inexpensive and quick
Countries with national cancer registries, such as Scandinavian coun- to conduct; they have been used as a surveillance tool for routine detec-
tries, have long been able to study cancer incidence among workers, tion of any occupational excess and as an inexpensive initial approach
such as the studies of dry cleaners in Denmark, Norway, Sweden, to provide some information on a new issue. However, they may be
and Finland (Lynge et al., 2006). A retrospective cohort study identi- biased if the death certificates on hand are not representative of all
fies the exposed and unexposed groups (either some general popu- deaths that occurred in the occupational group. Often such studies are
lation or a specific unexposed worker population) at some point in based on deaths among active workers or retirees from the company or
the past and follows them to the present, while a prospective cohort union under study, but miss deaths among persons who left employ-
study identifies the two groups in the present time and follows them ment with that company or union before death or before becoming
into the future. In general, most occupational studies have been retro- eligible for retirement. Also, because the proportion of deaths for all
spective in nature, which must rely upon historically collected expo- causes must total 100%, a higher proportion of deaths from one cause
sure information, but provide results more quickly than prospective must be offset by a lower proportion of deaths from another cause, or
studies, which must wait for cancers to develop in the population. vice versa, which can distort results. For example, if aerial pesticide
There are some notable exceptions to the almost exclusive use of applicators have a large real excess of deaths due to accidents, the
retrospective cohort studies to assess occupational carcinogens (Blair mathematical requirement that the PMR for all causes combined equal
et al., 2015a), including the prospective cohort study of cancer among 1.00 may cause some one or other causes of death to appear deficient,
Swedish construction workers that began in the mid-​1960s (Bergdahl even if their absolute mortality rates are excessive compared to an
and Järvholm, 2003; Järvholm and Englund, 2014), the US Agricultural unexposed population.
Health Study (Alavanja et al., 1996), the French Agricultural and Cancer
(AGRICAN) study (Levêque-​Morlias et al., 2015), studies of Chernobyl
cleanup workers (Kesminiene et al., 2002; Romanenko et al., 2008), and Case-​Control Studies
the recently launched World Trade Center Rescue and Recovery Workers
Study (Solan et  al., 2013)  and the GuLF Study (Sandler et  al., 2014). In a case-​control study, persons with the disease of interest (“cases”)
Some prospective cohort studies that were initiated primarily to investi- and persons without the disease (“controls”) are identified and past
gate non-​occupational factors have been utilized to evaluate cancer and exposures are ascertained. The relative occurrence of exposures among
occupational cancers as well, such as the Nurses’ Health Study, Shanghai cases and controls is compared. Case-​control studies are of three
Women’s Study, EPIC study, and Sister Study (Blair et al., 2015a). types: population-​based, clinic or hospital-​based, and nested within a
A limitation of most retrospective cohort studies is the lack of cohort. A population-​based case-​control study is the optimal approach
information on important potential confounders, such as occupational for the study of rare cancers, which would be infrequent in a cohort
exposures from other jobs besides the one under study and lifestyle study, and allows for ascertainment of important lifestyle factors, such
factors such as smoking and alcohol consumption and diet. However, as smoking, that are not typically recorded in work records, upon
for smoking, which is usually the potential confounder of most con- which most occupational cohort studies rely. Case-​control studies can
cern, a considerable amount of research based on studies with smoking assess multiple exposures (e.g., all past occupations). Generally, these
data, and based on hypothetical simulations, has been published which studies include subjects with a very wide range of jobs, each held by
shows that smoking rarely confounds disease risks in occupational a small number of persons (except for a few common occupations),
studies, and changes of effect measures (e.g., rate ratios) greater than which can limit the power to evaluate rare exposures and hence limit
20% are extremely unlikely (Axelson and Steenland, 1988; Blair et al., the usefulness of this approach for studies of occupational exposures.
2007). It is still preferable to collect smoking data, to control for pos- Case-​control studies can sometimes restrict the study area to enhance
sible confounding, but also to assess if smoking modifies the effect of the proportion of the population exposed, for example, studies of pesti-
an occupational exposure, as it does for asbestos (IARC, 2012e) and cides have been located in rural states, or portions of states, to increase
diesel exhaust (Silverman et al., 2012). For example, smokers who are the proportion of farmers who use pesticides (e.g., Miligi et al., 2003;
also exposed to asbestos have a risk of developing lung cancer that Zahm et al., 1990).
is greater than the individual risks from asbestos and smoking added Most case-​control studies conduct interviews with study subjects,
together, but less than the risks multiplied together (IARC, 2012e). and rely upon self-​reported occupational histories. Workers are usu-
ally able to report job title, industry, and dates of employment with
high validity, but often not the actual exposures sustained on the job
(Teschke et  al., 2002). Workers in some occupations do better than
Proportional Mortality Studies others. Farmers, for example, can provide quite accurate informa-
A variation on the cohort design in which a defined population is tion about pesticides used and conditions of handling and application
followed over time is a proportional mortality study, which includes (Blair et al., 2002). However, for many case-​control studies, specific
28

282 PART III:  THE CAUSES OF CANCER


exposures must be inferred from the job history through assessment by department, and time period, and then use that information to esti-
experts, use of a job-​exposure matrix, or, rarely, biological measure- mate exposures experienced by individuals in the study (Friesen et al.,
ments (Teschke et al., 2002) (see the section “Exposure Assessment” 2015). Data sources for such estimates include any existing personal
later in this chapter). and area industrial hygiene sampling data (noting if the samples were
Nested case-​control studies of occupational carcinogens are case-​ taken for routine monitoring or in response to a known or suspected
control studies conducted within a cohort, and cases are compared to high-​exposure excursion), descriptions of the industrial process and
a set of non-​cases in the cohort. More extensive information on the changes over time, production levels, exposure control devices and
occupational exposure or important potential confounders or effect efforts, and interviews with longtime workers with knowledge of his-
modifiers can be more easily obtained for the cases and a set of con- torical conditions. Experts then use these data to develop qualitative or
trols than for the full cohort (Rothman et al., 2008). quantitative estimates of exposure by job, department, and time. The
exposure data may incorporate level and probability of the exposure,
as well as the expert’s confidence in the assessment. Reliability and
Cross-​Sectional Studies validity assessments are sometimes performed to provide an indication
of the reliability and accuracy of the exposure assessment, such as in
In cross-​sectional studies, the exposure and cancer come from approx- studies of diesel exposure (Stewart et al., 2010) and of ethylene oxide
imately the same point in time. The major limitation of this approach (Hornung et al., 1994).
is that the temporal sequence of the exposure and the disease may be Population-​ based studies (either case-​ control or cohort studies)
difficult, if not impossible, to determine. For cancer, which has a long typically include a wide variety of occupations and industries, with a
latency, the assumption that current exposure status reflects exposures distribution representative of the general population. With the excep-
when the disease process was initiated may be particularly problem- tion of a few common jobs, the number of subjects holding any spe-
atic. Despite this limitation, a series of informative studies of the cor- cific job can be small. In population-​based case-​control studies using
relation between the concentration of various industries with cancer interviews, self-​reported occupational histories have good reliability
mortality in US counties yielded clues about occupational carcino- for job, industry, and dates of employment, but poorer reliability for
gens, and were followed up with case-​control and cohort studies (Blot reporting specific exposures (Friesen et al., 2015). Checklists of expo-
et al., 1979a). sures may help improve reporting of the job history itself, as well as
Cross-​sectional studies are often used in the study of biomarkers exposures of interest, but the data are prone to false positives and false
related to occupational exposures. For example, cadmium in the urine negatives. Reporting is somewhat better for exposures that are easily
of smelter workers has been measured at the same time as urinary pro- sensed and recognizable, or if the person was involved with the deci-
teins beta-​2-​microglobulin and retinol binding protein showing kid- sion-​making about the exposure, such as a farmer who buys, applies,
ney damage (Thun, 1992). Benzene levels in air and urine of exposed and tracks pesticide use. For many exposures, however, self-​reported
workers were correlated with measures of hematotoxicity, demonstrat- exposure histories can have serious limitations and may be affected by
ing its impact at air levels below the US occupational standard of 1 case recall bias in case-​control studies. Even more problematic than
part per million, and among genetically susceptible subpopulations self-​reports are proxy reports. Spouses and children provide less accu-
(Lan et al., 2004). A cross-​sectional study that showed formaldehyde-​ rate job and exposure histories than the subjects themselves (Brown
associated hematotoxicity among exposed workers was cited as a key et al., 1991). Coworkers have provided job and exposure information
justification for the IARC’s classification of formaldehyde as a human for deceased subjects in some studies (e.g., Chernobyl cleanup work-
leukemogen (IARC, 2012f; Zhang et al., 2010). ers [Romanenko et  al.,  2008]). Generic JEMs (vs. industry-​specific
Cross-​sectional studies of biomarkers can shed light on potential JEMs described earlier) can be used to assign exposure status based
mechanisms, identify early events in the natural history, improve expo- on the occupational histories, although one must consider possible dif-
sure assessment, and, in the case of molecular biomarkers, identify ferences across geographic areas before applying a JEM developed in
individuals susceptible to disease (Mayeux, 2004; Schulte, 1993). one country to a study conducted in another (Friesen et al., 2015; Hoar
However, a cross-​sectional study may be affected by reverse causation et al., 1980).
or disease bias, that is, the biomarker may be the effect of the disease In some studies, detailed sets of questions are triggered if the sub-
rather than a cause (Checkoway et al., 2007). jects have held jobs for which additional data are needed to assess
the potential for exposures of interest. These “occupational modules”
gather information on materials and equipment used, sensory descrip-
Exposure Assessment tions (e.g., smell), dermal exposure, work practices, engineering con-
Although studies relating occupation and cancer are informative in trols, and personal protective equipment used to improve the accuracy
suggesting leads to occupational hazards, eventually investigations of the assessment of exposure potential and amount at the individual
must focus on specific exposures to provide solid information for haz- subject level (Siemiatycki et al., 1997; Stewart et al., 1998)
ard and risk assessment. Quantitative occupational exposure assess- Expert reviews and exposure assignments are time-​ consuming,
ment is important in evaluating potential carcinogens and critical in expensive, not transparent, and may not always be reproducible, so
risk assessment. The data sources and approaches to develop such more systematic approaches are being developed (Fritschi et al., 2009;
quantitative assessments differ in industry-​based and population-​based Russ et al., 2014, 2016). Current efforts that aim to overcome these
epidemiologic studies (Friesen et al., 2015). limitations include the development of algorithm-​and measurement-​
Industry-​ based studies (generally cohort studies) are typically based decision rules, applying machine learning methods to classifica-
focused on one or a few agents in one or a few types of occupational tion tree models, computerized decision rules (e.g., OccIDEAS: http://​
settings. It is important to remember, however, that although the focus www.occideas.org/​#!whousing/​c1ykh) (Fritschi et  al., 2009), and
of the research may be on “one or a few” exposures, there are very few computer-​assisted occupational coding software (e.g., SOCcer: http://​
workplaces where is there is such a small number of exposures. The soccer.nci.nih.gov) (Russ et al., 2014, 2016).
subjects’ job histories are usually obtained from preexisting company In many settings, it may be important to consider multiple expo-
or union records. With the notable exception of radiation workers, per- sures simultaneously to assess interaction among exposures, to evalu-
sonal exposure measurements for a large proportion of the subjects ate possible confounding, and to clarify the credibility of associations.
in retrospective cohort studies are not usually available. There may De Roos et al. (2003) performed an integrative assessment of multiple
be some measurements for a few past workers in some settings and pesticides as risk factor for non-​Hodgkin’s lymphoma. Consideration
area measurements at various work locations, but generally exposure of mixtures is important to evaluate if there are enhanced effects from
measurements for the time period critical for cancer initiation are lim- a mixture, which is greater than that expected from single exposures;
ited. To overcome this limitation, researchers collect information from this has been reported in a few studies for pesticides (De Roos et al.,
a variety of sources to reconstruct historical exposures by task, job, 2003; Hohenadel et  al., 2011; Lerro et  al., 2015). In contrast, there
 283

Occupational Cancer 283
might be diminished effects from combinations of exposures, such as environmental hazards. Quantitative risk assessment is typically con-
the observation that the risk of adenocarcinoma of the lung associated ducted by government regulatory agencies. Quantitative risk assess-
with diesel exposure was ameliorated in presence of endotoxin expo- ment may be based on either animal data or human data. The former
sure (Tual et al., 2015). has an advantage in that exposures in observational human studies
Attention must also be given to consideration of gender differences are not assigned as in animal experiments, and thus there can be con-
that may affect exposure assessment (Friesen et al., 2012; Locke et al., siderable uncertainty in exposure estimates. However, animal studies
2014). Men and women with the same job title do not always have the require extrapolation from animals to humans and hence involve much
same duties and exposures. There are also gender differences in self-​ more uncertainty. For this reason, human (epidemiological) data are
reports of exposures, work practices, personal hygiene, and the effec- generally preferred, but are not always available. When human data are
tiveness of protective equipment and engineering controls that affect available, occupational quantitative risk assessment, aimed at setting
the accuracy of exposure assessment (Zahm and Blair, 2003). permissible exposure levels, is based on exposure–​response data from
It would be ideal if it were possible to use a biomarker of exposure, one or more occupational studies. These results provide information on
which would characterize the total internal dose rather than external the increased rate of disease per unit of exposure (exposure–​response
exposure. However, biomarkers of exposure are often short-​lived in data) for an exposed population that must typically be converted to the
the body. To fully characterize exposure over the years would require excess risk of disease over a lifetime for specific exposures to iden-
multiple biologic collections, or there would need to be (1) limited tify a permissible exposure level. The US Occupational Safety and
intra-​individual variation of the biomarker over time, (2) an accurate Health Administration (OSHA) usually seeks to limit excess risk to 1
laboratory assay, and (3) long enough persistence in the body so that in 1000 deaths (or serious disease) for exposed populations. For exam-
the measure could reflect an exposure that occurred during the time ple, if the background lifetime risk for lung cancer in the non-​exposed
period when carcinogenesis was initiated. Furthermore, for case-​con- general population is 5% (0.05), then OSHA will seek the level of
trol studies, the biomarker would need to be not affected by disease exposure that will not increase lifetime risk for an exposed population
status or treatment (Baris et  al., 2000; Steenland and Moe, 2016). beyond 5.1%. Other countries have adopted similar approaches for
Nonetheless, biomarkers can often help in the assessing the relation- carcinogens that are genotoxic (Nielsen and Ovrebø, 2008).
ship between historical exposures and dose. There are two major issues of concern for quantitative risk assess-
ment based on epidemiological exposure–​response models. The first
is the shape of the exposure–​response curve. When data are sparse,
Genetic Susceptibility and sometimes even when they are not, it may be difficult to choose
Because occupational studies are likely to have better assessment of among competing models used for setting permissible limits, and dif-
work-​related exposures than studies of many other types of exposures, ferent models can have very different consequences. Typical questions
they offer excellent opportunity to explore potential interactions with involving model selection might be whether the exposure–​response
genetic susceptibility. For example, using the candidate gene approach, relationship shows a linear increase in disease rate per unit of expo-
it was established that the slow N-​acetylation phenotype, caused by var- sure, whether there is a threshold below which there is no risk followed
iation in the NAT2 gene, confers increased risk for bladder cancer among by an increase, or conversely, whether there is a cut-​point above which
workers exposed to aromatic amines (Gu and Wu, 2011). Technological disease risk begins to flatten out or even decrease. In general, one
advances have enabled researchers to look at extremely large numbers expects that greater exposure to a carcinogen would lead to a greater
of genetic variants in an agnostic fashion in genome-​wide association occurrence of cancer. However, risk for some established carcinogens
studies (GWAS), accelerating the search for gene-​environment inter- plateaus at higher doses; examples include ethylene oxide, dioxin,
actions. A small genome-​wide study of gene–​environment interaction beryllium, diesel fumes, and nickel (Stayner et  al., 2003; Steenland
for asbestos exposure in lung cancer susceptibility has been conducted et al., 2011). Plateaus in disease rate at higher exposure levels in occu-
with some promising results, particularly when a pathway approach was pational studies may occur due to (1) bias introduced by the healthy
used, but more research with a larger number of subjects is needed (Wei worker survivor effect; (2) depletion of the number of susceptible peo-
et al., 2012). Some recent examples of possible gene-​exposure interac- ple in the population at high exposure levels; (3) a natural limit on the
tions include a large GWAS study of bladder cancer that found evidence relative risk for diseases with a high background rate; (4) inaccurate
of interaction for rs798766 (TMEM129-​TACC3-​FGFR3) with expo- measurements or misclassification of exposure; (5) influence of other
sure to straight metalworking fluids, with the interaction more apparent risk factors that vary by the level of the main exposure (confounding);
among patients with tumors positive for FGFR3 expression (Figueroa and (6) saturation of key enzyme systems or other processes involved
et  al., 2015). Gene–​environment interactions between prostate cancer with the development of disease (Stayner et al., 2003). As an example
susceptibility loci identified via GWAS and occupational exposure to of a biological process that affects the shape of the dose–​response
pesticides have been identified among applicators in the Agricultural curve, high doses of radiation result in cell death, while lower doses
Health Study (Koutros et  al., 2010, 2013). In addition to genomics, damage cells and result in malignant transformation, so risk for thy-
the occupational setting can be a fertile ground for the application of roid cancer, but not other sites, plateaus and may even decrease with
epigenomics, transcriptomics, proteomics, and metabolomics and other increasing dose (Berrington de González et al., 2013).
“-​omics” technologies (Vlaanderen et al., 2010). A second question typical of quantitative risk assessment is the
nature of the exposure–​response relationship in the low-​dose region,
In summary, occupational cancer epidemiology is a powerful tool for where there may be few data. This question often arises when occupa-
(1) protecting workers’ health through identifying and reducing expo- tional epidemiological studies (high exposure) are used for risk assess-
sure to carcinogens; (2) protecting the public by identifying and reduc- ment for general environmental exposures (lower exposure), such as
ing exposure to occupational carcinogens that otherwise would enter diesel fumes, dioxin, or asbestos. Traditionally, when data are sparse in
the non-​workplace environment or consumer products; and (3) eluci- the low-​dose region, a linear extrapolation of risk down to zero expo-
dating mechanisms of carcinogenesis. sure is used by regulatory agencies, starting from a point of departure
that is a function of the lowest level of risk for which observed data
are considered reliable. This procedure may also provide the best fit
QUANTITATIVE RISK ASSESSMENT to data in which the exposure–​response relationship plateaus at higher
exposures (Steenland et al., 2011).
Evaluation of the carcinogenicity of occupational exposures consists By way of example of quantitative risk assessment, consider a
of not only identification of carcinogens (qualitative risk assessment), recent occupational cancer risk assessment for lung cancer and die-
such as that done by the IARC Monograph Program, but also of quan- sel fumes (Vermeulen et  al., 2014). These authors took three occu-
titative risk assessment, which is the characterization of the proba- pational studies of workers exposed to diesel fumes (two truck driver
bility of human cancer resulting from specific levels of exposures to studies, one miner study), which were the only epidemiologic studies
284

284 PART III:  THE CAUSES OF CANCER


with quantitative exposure–​response data using the same exposure behavior (7%), geophysical factors (3%), alcohol (3%), and medicines
metric (cumulative exposure to elemental carbon to characterize die- (1%). In addition, the risk from occupational exposures is borne largely
sel exhaust), therefore permitting a joint analysis. They combined the by blue-​collar workers who represented only about 25% of the US
three studies to derive a common exposure–​response curve, which population in 2000 (those in the construction, extraction, maintenance,
indicated that for each 1µg/​m3-​year of elemental carbon exposure, the transportation, and material moving industries; see https://​www.cen-
log RR (relative risk) for lung cancer would increase by 0.00098; this sus.gov/​prod/​2003pubs/​c2kbr-​25.pdf); thus the AF among blue-​collar
translates to an RR of about 2 for 700 µg/​m3-​years, which in turn to workers must be considerably larger than 4%.
an average exposure of about 35 µg/​m3 elemental carbon for someone Doll and Peto’s original estimate has stood up relatively well to the
exposed for 20 years (the miners had an average exposure of about test of time (Blot and Tarone, 2015). Since then, there have been a num-
this level). Next, the authors translated this finding into an estimate ber of other estimates in the United States and other countries, as more
of excess lifetime risk for death from lung cancer (above background information accumulated on the relative risks of cancer from specific
lifetime risk for an individual not occupationally exposed to diesel agents, and as the prevalence of occupational exposure has changed.
fumes, which is about 5%), at different exposure levels over a hypo- Steenland et al. (2003) estimated that 2.3%–​4.8% of US cancer deaths
thetical 45-​year working lifetime. US OSHA uses the 45-​year work- were attributable to occupational factors (0.8%–​1.0% among females,
ing lifetime as a default assumption for setting permissible levels for 3.3%–​7.3% among males), which accounted for 13,000 to 26,000 can-
workers. Exposure for 45 years at 1 µg/​m3 (45 ug/​m3-​years) would be cer deaths annually. The highest AFs were for lung cancer, estimated
expected to lead to an excess lifetime risk of 1.7/​1000 (i.e., increasing to be between 6% and 17% for men, and to be about 2% for women.
a background risk of 0.0500 to 0.0517). Given that OSHA seeks to To give a more recent example of an AF for a specific agent and a spe-
keep excess lifetime risks below 1/​1000, this would argue for a per- cific cancer, we can use the example of diesel fumes and lung cancer
missible exposure level for workers of slightly less than 1 µg/​m3. Risk discussed earlier (Vermeulen et al., 2014). These authors estimated that
assessors must then weigh the optimal standard from a health stand- at current exposure levels to diesel fumes, the occupational exposure is
point versus economic feasibility. Currently, with the exception of responsible for about 1% of lung cancer deaths in the United States and
miners, exposed workers have diesel exhaust exposures in the range of England, and another 5% of lung cancer is due to environmental expo-
3–​13 µg/​m3, while members of the public in urban areas are exposed sures to diesel fumes.
to about 0.8 µg/​m3 (Vermeulen et al., 2014). AFs from other countries and worldwide are not dissimilar. For
example, Rushton et al. (2012) estimated that 5.3% (8.2% men, 2.3%
women) of cancer deaths in England in 2005 were due to occupational
ATTRIBUTABLE FRACTION exposures. The principal cancer sites considered were mesothelioma,
sinonasal, lung, nasopharynx, breast, non-​ melanoma skin cancer,
To estimate the impact of occupational cancer, we can calculate the bladder, esophagus, soft tissue sarcoma, and stomach. The principal
attributable fraction (AF) of cases or deaths for a specific cancer carcinogens were asbestos, mineral oils, solar radiation, silica, diesel
(or all cancers) due to occupation; this is sometimes also called the engine exhaust, coal tars and pitches, dioxins, environmental tobacco
attributable risk. The AF is calculated by AF = [(RR – ​1)p]/​[(RR – ​1) •  smoke, radon, tetrachloroethylene, and arsenic, as well as occupa-
p + 1], where RR is the relative risk of disease for the exposed ver- tional circumstances such as shift work and occupation as a painter
sus non-​exposed, and p is the proportion of the population exposed. or welder.
The proportion of the population exposed includes those exposed cur- Recent estimates on a global scale were made by the Global Burden
rently and those exposed in the past. An overall RR for exposed versus of Disease (GBD) 2013 group (GBD 2013 Risk Factors Collaborators,
non-​exposed and an overall proportion of the population exposed are 2015), which estimated that IARC Group  1 (definite) occupational
commonly used. However, sometimes the population exposed is bro- carcinogens accounted for about 3.7% of worldwide cancer deaths in
ken down into different levels and/​or duration of exposure, and RRs 2013 (304,000 deaths). More detailed information available from the
specific by level/​duration of exposure are used to calculate separate GBD website (https://​vizhub.healthdata.org/​gbd-​compare/​), estimates
AFs by exposure level or duration. There is considerable judgment that the principal cancers caused by occupational exposures were
involved in these calculations; for example, if one wants to calculate lung cancer (269,000 deaths), mesothelioma (25,000 deaths), laryn-
the AF for all cancers due to occupation, one must choose which can- geal cancer (4600 deaths), and leukemia (3200 deaths). Similarly, the
cer/​occupations associations are to be included. RRs for some spe- GBD 2103 group estimated that there were 5.8 million occupational
cific cancers and occupations/​exposures may be based on one large cancer DALYs out of a total of 197 million all-​cause cancer DALYs,
and very well-​conducted study, but more often use a meta-​analysis so cancer DALYs attributable to occupation were 2.9% (GBD 2013;
of existing studies to determine an average RR across studies. Often, Risk Factors Collaborators, 2015). The principal occupational cancers
authors restrict themselves to occupational carcinogens considered contributing to DALYS were lung cancer (5.0  million), mesotheli-
definite or probable according to IARC (Group 1 or 2A). A common oma (514,000), leukemia (108,000), and laryngeal cancer (102,000)
difficulty is that estimates of the proportion of the population exposed (https://​vizhub.healthdata.org/​gbd-​compare/​).
are not routinely available. The case of mesothelioma is particularly interesting, because it is
AFs can be calculated for both incident cancers and for fatal can- (1)  only caused by asbestos, largely an occupational exposure, and
cers. Further calculations can be done to estimate disability-​adjusted (2) a highly fatal cancer that has a very long latency (e.g., typically
life years lost (DALYs). DALYs attempt to quantify years lost due to 40  years instead of approximately 20  years for most solid tumors).
premature death, or years lived with lower quality of life following a Thus while the burden of many occupational cancers is gradually
disease. They are a weighted estimate of the number of years lived with decreasing in developed countries, where exposures have been low-
disability, where the weighting is based on the severity of the disability. ered and heavy industry has decreased (which is not the case in less
There have been a number of estimates of the AF for occupational developed countries), the burden from mesothelioma in the developed
cancer in the United States and other countries. One of the first and per- countries may have not yet peaked. Figure 16–1 shows the increas-
haps the best known was published by Doll and Peto in 1981, in which ing mortality rate of mesothelioma in England over time among older
they estimated that 4% (1.2% for females, 7% for males) of US cancer men. The number of male mesothelioma deaths per year in England
deaths were due to occupational cancer, primarily from lung cancer and is expected to peak at around 2500/​year in the year 2020 (Health and
with the principal occupational exposure being asbestos (Doll and Peto, Safety Executive, 2014).
1981). Doll and Peto noted that this estimate could be off by a factor of It should be remembered that these estimates of the burden of can-
two in either direction. It is important to put the estimate of AF of 4% cer associated with occupational exposures do not account for second-
for occupational exposures in perspective with other cancer risk factors. ary exposures. In some circumstances, occupational exposures have
Doll and Peto (1981) estimated that tobacco had an AF of 30% and diet been observed to increase the risk of cancer among the spouses and
of 35%, but all others were generally in the range estimated for occu- children of workers. Asbestos fibers carried home on workers’ cloth-
pational exposures, for example, infections (10%), reproductive/​sexual ing, skin, and hair pose a risk for family members (Ferrante et al.,
 285

Occupational Cancer 285
1000 all situations, and especially where individuals have little control
over exposures that may cause the disease. Studies linking work-
place exposures to cancer may lead government regulators to control
exposure. Because regulation has economic consequences, studies
of cancer and occupational exposures receive intense scrutiny. They
are often reviewed and dissected beyond what may occur for other
cancer risk factors. Because of these pressures, intense debates and
disagreements regarding the quality of individual studies and the
weight of evidence are standard for each occupational exposure–​can-
100 cer relationship when associations are new and the amount of scien-
tific information is less robust than it will eventually be. Eventually,
however, sufficient scientific information may accumulate to allow
general agreement among scientists, stakeholders, and the public
regarding an issue. All widely accepted occupational carcinogens
Rate per 1,000,000 person-years

(e.g., arsenic, asbestos, benzene, and silica) went through this per-
iod—​sometimes a lengthy period—​of debate and controversy. Even
after general acceptance that a substance is a carcinogenic hazard to
humans by the public and scientific community, there may be disa-
greement regarding the exposure–​response relationship and other risk
10 assessment issues. Examples of established occupational carcinogens
where disagreements continue include arsenic (where risk from expo-
sure at low levels is debated [Tsuji et al., 2014]), for chrysotile asbes-
tos (although classified as a human carcinogen by the IARC [2012c],
there is continuing debate about whether the cancer risk associated
with chrysotile asbestos is actually due to concomitant amphibole
asbestos exposure [Bernstein et al., 2013; Kanarek, 2011; van der Bij
et  al., 2013; Yarborough,  2006]), and benzene (where there is disa-
greement on the range of lymphohematopoietic cancers affected and
1 the risks at lower levels of exposure [Hayes et  al., 1997; Schnatter
et al., 2012]). The debate regarding the adequacy of the data to con-
Age group clude that a substance is a carcinogenic hazard is still ongoing for sub-
85+ stances of more recent concern, for example, diesel exhaust (Gamble
75–84 et al., 2012; Sun et al., 2014), formaldehyde (Checkoway et al., 2012;
65–74 Gentry et al., 2013; McLaughlin and Tarone, 2014), trichloroethylene
55–64 (Alexander et al., 2007), and shift work (Brudnowska and Peplonska,
45–54 2011; Erren et al., 2010).
35–44 The example of diesel exhaust, now an established lung carcino-
0.1 gen, is instructive regarding the controversy and special interests often
1970 1980 1990 2000 2010 associated with studies of occupational cancer. The National Cancer
Year of death
Institute (NCI) and the National Institute for Occupational Safety and
Health (NIOSH) initiated a study of diesel-​exposed miners in 1992,
which was not published until 2012. The study faced numerous legal
Figure 16–1.  Age-​adjusted mortality rate for mesothelioma among males challenges from the mining industry, through its lobbying group The
in the United Kingdom, 1969–​2013. Methane Awareness Research Group (MARG), and was subject to
Congressional oversight throughout protocol development, data col-
lection, analysis, peer review, and journal publication. These pres-
2007; IARC, 1987, 2012c). In fact, secondary exposure to asbestos is sures delayed the study for years (Monforton, 2006; Morris, 2012;
thought to be the major risk factor for mesothelioma among women. Kean, 2012).
Family members on farms where pesticides are applied may be
exposed via contact with contaminated clothing of the applicator as
well as environmental exposure to airborne pesticides drifting over FUTURE DIRECTIONS
residential areas, residential dust, and ingestion (Deziel et al., 2015).
Spouses and children of farmers had a doubling of the urinary levels There are many occupational exposures that require further evalu-
of a metabolite of the pesticide carbaryl following application by the ation. An obvious starting point for selection of a few high-​priority
farmer on the family farm, documenting secondary exposure (Shealy candidates could be the substances in the probable (2A) and possible
et al., 1997). In the Agricultural Health Study, pesticide levels in the (2B) categories of IARC classification that affect large populations.
homes of farmers have exceeded those in non-​farm homes (Curwin For these, there is already some information suggesting an associa-
et al., 2005). Children of the Agricultural Health Study participants tion, and this information can provide direction for new investiga-
have excess lymphoma (Flower et al., 2004). Other studies of paren- tions. There are an even larger number of occupational exposures
tal exposure to pesticides have suggested that the children’s second- that have been linked to cancer in animal bioassays for which epi-
ary exposures play a role in the risk of leukemia and brain cancer, demiologic data are largely nonexistent. Even for those exposures
and less consistently with Wilms tumor, Ewing sarcoma, soft-​tissue that are already classified as human carcinogens, additional studies
sarcoma, and Hodgkin’s lymphoma (Bailey et  al., 2014; Daniels could provide important information. The recent IARC re-​evalua-
et al., 1997; Rodvall et al., 2003; Zahm and Ward, 1998). tions in Volumes 100 A through F of factors in Group 1 (sufficient
evidence for human carcinogenicity) show that even studies of
known carcinogens can be beneficial in that they reveal links to spe-
CONTROVERSY cific cancers not previously documented, as well as new mechanis-
tic and toxicological understanding, and data on exposure–​response
Occupational studies often generate considerable controversy. relationships. It appears, however, that, despite numerous leads and
Cancer is a devastating disease with special emotional overtones in the clear need for study of occupational exposures, research efforts
286

286 PART III:  THE CAUSES OF CANCER


on occupational cancer have been diminishing in recent years (Raj Pesticides
et al., 2014).
Future studies should take advantage of the new technologies that Pesticides provide many important benefits in food production and
allow characterization of mechanistic pathways, gene–​exposure inter- protection, public health, and aesthetics, but some may present risk
actions, and epigenetic influences. Also, new techniques in exposure of cancer and other diseases (Blair et  al., 2015b). Experimental
assessment are available that reduce exposure misclassification and investigations have been conducted for most pesticides, but epide-
result in more accurate estimates of exposure–​response relationships. miologic data are much more limited. The IARC, for example, has
Effective use of these new opportunities will require less reliance on only evaluated a few specific pesticides, but has recently described
the historical cohort design, which has been the design of choice in plans for a series of monographs on pesticides currently in use, and
occupational investigations in the past, and more use of prospective has conducted two. The initial IARC effort concerned commonly
designs (Blair et al., 2015a) that allow the incorporation of biologic used organophosphate insecticides and a very common herbicide
and mechanistic components. The changing dynamics and geographi- (glyphosate, commercial name Round-​Up). The monograph clas-
cal location of various industries also indicate that future investiga- sified tetrachlorvinphos and parathion as possibly carcinogenic to
tions should expand beyond studies that are composed of white men humans (2B) and malathion, diazinon, and glyphosate as probably
in high-​income countries, which have predominated in the past (Raj carcinogenic to humans (2A) (Guyton et  al., 2015). In a second
et al., 2014). evaluation, the IARC classified DDT as a probable carcinogen (2A),
Although it is difficult to precisely predict specific exposures or lindane (an organophosphate largely banned) as a definite carcinogen
workplace conditions that are most important for study in the future, (1), and 2-​4-​5-​T (a widely used herbicide) as a possible carcinogen
following are a few that deserve consideration, based on some evi- (2B) (Loomis et al., 2015). DDT use remains common in Africa and
dence of hazard, number of people exposed, and possible magnitude India for malaria control, although it was banned as an insecticide in
of population burden. most countries in the late 1970s and early 1980s. Many pesticides
currently in use, however, have not been adequately evaluated in epi-
demiologic studies. In addition to inadequate studies on many pes-
ticides currently in use, new pesticides are periodically developed
Sedentary Behavior and introduced to overcome pest resistance that develops over time.
Lack of physical activity contributes to a number of diseases, includ- Consequently, there is a growing need to initiate new epidemiologic
ing cancer (US Department of Health and Human Services, 1996). studies to evaluate the cancer hazard that may emanate from estab-
Amount of time spent in sedentary behaviors appears to be associ- lished and new pesticides.
ated with an additional disease burden beyond that from low levels of
physical activity (Matthews et  al., 2012). In the past, most physical
activity was associated with work, but currently, many occupations Diesel Exhaust
make a negligible contribution to overall activity, and this leads to Diesel exhaust has recently been classified by the IARC as a human
health threats (Straker et al., 2009). The high level of physical activity carcinogen (IARC, 2014a). Epidemiologic studies that made major
in earlier times is dramatically demonstrated in an Old Order Amish contributions to this decision (Attfield et  al., 2012; Garshick et  al.,
community whose lifestyle is similar to that common 150 years ago 2008; Silverman et al., 2012; Steenland et al., 1990) evaluated expo-
(Bassett et al., 2004). Their physical activity may play a role in their sures largely from an older engine technology. These older engines
low overall cancer rate (Westman et al., 2010). In general, over time will eventually be replaced by newer, cleaner-​ burning engines
there has been a drastic decrease in the proportion of jobs that require a designed to reduce exhaust exposures. There is a need for new epide-
moderate intensity of physical activity, with a corresponding increase miologic investigations to evaluate the potential hazard from exhausts
in jobs with largely sitting and sedentary behavior (Church et  al., from these new engines.
2011). Consequently, many individuals must rely almost entirely on
leisure time activities to achieve appropriate levels of physical activ-
ity, especially in developed countries. However, there are certainly Nanoparticles
many occupations in developed countries that still require consider- Naturally occurring nanoparticles are found in engine exhausts and
able physical activity. burning carbon materials, but most of the future concern is in relation
to engineered nanoparticles. Human exposure to engineered nanopar-
ticles can occur during creation in the laboratory, large-​scale manu-
Shift Work
facture, use, and environmental contamination (Kumar and Dhawan,
Shift work has been classified as probably carcinogenic in humans (2A) 2013; Rim et al., 2013). NIOSH recently conducted an industry-​wide
based on limited evidence in humans, sufficient evidence in experimen- study and exposure assessment (Dahm et al., 2015; Schubauer-​Berigan
tal animals, and mechanistic actions that may be relevant in humans et al., 2011) and found that to date there are only about 1000 workers
(IARC, 2010a). In humans the strongest association was with breast in the United States judged as exposed. However, many nanoparticles
cancer, but links with other sites (prostate, endometrium, and colon) are composed of known or suspect carcinogenic agents, and the num-
were observed in some studies. Night work suppresses melatonin, ber of exposed individuals is expected to grow rapidly, particularly in
which inhibits tumors, although other mechanisms have also been sug- the chemical industry, photochemical electricity generation, and imag-
gested (Fritschi et al., 2011). The overall evidence regarding shift work ing and molecular diagnosis (Stark et al., 2015). The IARC evaluated
and ill health is still in flux. A  recent study provided some evidence nanoparticles in 2014; most were judged not classifiable in humans
for a relationship with lung cancer (Gu et al., 2015). This suggests that (Group 3), but one type was found to have sufficient animal evidence
additional studies are needed for breast cancer, which is most strongly of carcinogenicity, and was classified Group 2B (possible carcinogen)
associated with shift work at the present time, but perhaps studies are (Grosse et al., 2014).
also needed to consider links with other cancers. Studies should also
evaluate different forms of shift work, characterize exposure more
accurately, evaluate timing issues regarding exposure to light at night, ACKNOWLEDGMENT
explore possible mechanistic actions, and assess possible interaction
with other lifestyle and occupational factors. It should be noted that The authors would like to thank David Check for graphics contributions.
this is a common exposure. For example, Purdue et  al. (2015) have
estimated that if shift work causes breast cancer, an estimated 6% of References
US incident breast cancers in 2010 would have been attributable to Alavanja MC, Sandler DP, McMaster SB, et al. 1996. The Agricultural Health
shift work. Study. Environ Health Perspect, 104(4), 362–​369. PMCID: PMC1469343.
 287

Occupational Cancer 287
Alexander DD, Kelsh MA, Mink PJ, et  al. 2007. A meta-​analysis of occu- Checkoway H, Boffetta P, Mundt DJ, and Mundt KA. 2012. Critical review
pational trichloroethylene exposure and liver cancer. Int Arch Occup and synthesis of the epidemiologic evidence on formaldehyde exposure
Environ Health, 81(2), 127–​143. PMID: 17492303. and risk of leukemia and other lymphohematopoietic malignancies.
Axelson O, and Steenland K. 1988. Indirect methods of assessing the effects Cancer Causes Control, 23(1), 1747–​1766. PMCID: PMC3465649.
of tobacco use in occupational studies. Am J Ind Med, 13(1), 105–​118. Checkoway H, Pearce N, and Kriebel D. 2007. Selecting appropriate study
PMID: 3344750. designs to address specific research questions in occupational epidemiol-
Bailey HD, Fritschi L, Infante-​Rivard C, et al. 2014. Parental occupational pes- ogy. Occup Environ Med, 64(9), 633–​638. PMCID: PMC2092571.
ticide exposure and the risk of childhood leukemia in the offspring: find- Church TS, Thomas DM, Tudor-​Locke C, et al. 2011. Trends over 5 decades in
ings from the childhood leukemia international consortium. Int J Cancer, the U.S. occupation-​related physical activity and their associations with
135(9), 2157–​2172. PMID: 24700406. obesity. PLoS One, 6(5), e19657. PMCID: PMC3102055.
Baris D, Kwak LW, Rothman N, et al. 2000. Blood levels of organochlorines Creech JL, Jr, and Johnson MN. 1974. Angiosarcoma of liver in the manufacture
before and after chemotherapy among non-​Hodgkin’s lymphoma patients. of polyvinyl chloride. J Occup Med, 16(3), 150–​151. PMID: 4856325.
Cancer Epidemiol Biomarkers Prev, 9(2), 193–​197. PMID: 10698481. Curwin BD, Hein MJ, Sanderson WT, et  al. 2005. Pesticide contamination
Bassett DR, Schneider PL, and Huntington GE. 2004. Physical activity in inside farm and nonfarm homes. J Occup Environ Hyg, 2(7), 357–​367.
an Old Order Amish community. Med Sci Sports Exer, 36(1), 79–​85. PMID: 16020099.
PMID: 14707772. Dahm M, Schubauer-​Berigan M, Evans D, Birch E, Fernback J, and Deddens J.
Benbrahim-​Tallaa L, Lauby-​Secretan B, Loomis D, et al. 2014. Carcinogenicity 2015. Carbon nanotube and nanofiber exposure assessments: an analysis
of perfluorooctanoic acid, tetrafluoroethylene, dichloromethane, 1,2-​ of 14 site visits. Ann Occup Hyg, 59(6), 705–​723. PMCID: PMC4507369.
dichloropropane, and 1,3-​propane sultone. Lancet Oncol, 15(9):  924–​ Daniels JL, Olshan AF, and Savitz DA. 1997. Pesticides and childhood cancers.
925. PMID: 25225686. Environ Health Perspect, 105(10), 1068–​1077. PMCID: PMC1470375.
Bergdahl IA, and Järvholm B. 2003. Cancer morbidity in Swedish asphalt Daniels RD, Kubale TL, Yiin JH, et al. 2014. Mortality and cancer incidence
workers. Am J Ind Med, 43(1), 104–​108. PMID: 12494427. in a pooled cohort of US firefighters from San Francisco, Chicago
Bernstein D, Dunnigan J, Hesterberg T, et al. 2013. Health risk of chrysotile and Philadelphia (1950–​2009). Occup Environ Med, 71(6), 388–​397.
revisited. Crit Rev Toxicol, 43(2), 154–​183. PMCID: PMC3581056. PMCID: PMC4499779.
Berrington de González A, Gilbert E, Curtis R, et al. 2013. Second solid can- Darby SC, Whitley E, Howe GR, et al. 1995. Radon and cancers other than lung
cers after radiation therapy:  a systematic review of the epidemiologic cancer in underground miners:  a collaborative analysis of 11 studies. J
studies of the radiation dose-​response relationship. Int J Radiat Oncol Natl Cancer Inst, 87(5), 378–​384. PMID: 7853419.
Biol Phys, 86(2), 224–​233. PMCID: PMC3816386. De Roos AJ, Zahm SH, Cantor KP, et al. 2003. Integrative assessment of mul-
Blair A, Hines CJ, Thomas K, et  al. 2015. Investing in prospective cohort tiple pesticides as risk factors for non-​Hodgkin’s lymphoma among men.
studies on occupational exposures. Am J Ind Med, 58(2), 113–​ 122. Occup Environ Med, 60:E11. PMCID: PMC1740618.
PMCID: PMC4516175. Deziel NC, Friesen MC, Hoppin JA, Hines CJ, Thomas K, and Beane Freeman
Blair A, Marrett L, and Beane Freeman L. 2011. Occupational cancer in devel- LE. 2015. A review of nonoccupational pathways for pesticide exposure
oped countries. Environ Health, 10(Suppl 1):S9. PMCID: PMC3073201 in women living in agricultural areas. Environ Health Perspect, 123(6),
Blair A, Ritz B, Wesseling C, et al. 2015. Pesticides and human health. Occup 515–​524. PMCID: PMC4455586.
Environ Med, 72:81–​82. PMID: 25540410. Dietrich H, and Dietrich B. 2001. Ludwig Rehn (1849–​1930): pioneering find-
Blair A, Stewart P, Lubin JH, et  al. 2007. Methodological issues regard- ings on the aetiology of bladder tumours. World J Urol, 19(2), 151–​153.
ing confounding and exposure misclassification in epidemiological PMID: 11374319.
studies of occupational exposures. Am J Ind Med, 50(3), 199–​ 207. Dobson J. 1972. Percivall Pott. Ann R Coll Surg Engl, 50(1), 54–​ 65.
PMID: 17096363 PMCID: PMC2388056.
Blair A, Tarone R, Sandler D, et al. 2002. Reliability of reporting on lifestyle Doll R. 1952. The causes of death among gas-​workers with special reference to
and agricultural factors by a sample of participants in the Agricultural cancer of the lung. Br J Ind Med, 9(3), 180–​185. PMCID: PMC1037402.
Health Study from Iowa. Epidemiology, 13(1), 94–​99. PMID: 11805592. Doll R. 1955. Mortality from lung cancer in asbestos workers. Br J Ind Med,
Blot WJ, Fraumeni JF Jr, Mason TJ, et al. 1979a. Developing clues to environ- 12(2), 81–​86. PMCID: PMC1037613.
mental cancer: a stepwise approach with the use of cancer mortality data. Doll R. 1958. Cancer of the lung and nose in nickel workers. Br J Ind Med,
Environ Health Perspect, 32(Oct.), 53–​58. PMCID: PMC1637941. 15(4), 217–​223. PMCID: PMC1037926.
Blot WJ, Harrington JM, Toledo A, et  al. 1978. Lung cancer after employ- Doll R, and Peto R. 1981. The causes of cancer: quantitative estimates of avoid-
ment in shipyards during World War II. N Engl J Med, 299(12), 620–​624. able risks of cancer in the United States today. J Natl Cancer Inst, 66(6),
PMID: 683235. 1191–​1308. PMID: 7017215.
Blot WJ, Morris LE, Stroube R, et  al. 1980. Lung and laryngeal cancers in Ehrenberg L, and Hallstrom T. 1967. Haematologic studies on persons occu-
relation to shipyard employment in coastal Virginia. J Natl Cancer Inst, pationally exposed to ethylene oxide. In:  Radiosterilization of Medical
65(3), 571–​575. PMID: 6931936 Products (pp. 327–​334). Vienna:  International Atomic Energy Agency
Blot WJ, Stone BJ, Fraumeni JF Jr et al. 1979b. Cancer mortality in U.S. coun- SM 92/​96.
ties with shipyard industries during World War II. Environ Res, 18(2), Erren TC, Falaturi P, Morfeld P, Knauth P, Reiter R, and Piekarski C. 2010.
281–​290. PMID: 510250 Shiftwork and cancer: the evidence and the challenge. Dtsch Arztebl Int,
Blot WJ, and Tarone RE. 2015. Doll and Peto’s quantitative estimates of cancer 107(38), 657–​662. PMCID: PMC2954516.
risks:  holding generally true for 35  years. J Natl Cancer Inst, 107(4). Ferrante D, Bertolotti M, Todesco A, Mirabelli D, Terracini B, and Magnani
PMID: 25739419 C. 2007. Cancer mortality and incidence of mesothelioma in a cohort of
Boyle P, and Levin B (Eds.). 2008. World Cancer Report 2008. Lyon, wives of asbestos workers in Casale Monferrato, Italy. Environ Health
France: International Agency for Research on Cancer Perspect, 115(10), 1401–​1405. PMID: 17938727.
Bridge JC. 1933. Annual Report of the Chief Inspector of Factories and Workshops Figueroa J, Koutros S, Colt JS, et al. 2015. Modification of occupational expo-
for the Year 1932 (pp. 103–​104). London: Her Majesty’s Stationery Office. sures on bladder cancer risk by common genetic polymorphisms. J Natl
Brown LM, Dosemeci M, Blair A, and Burmeister L. 1991. Comparability of Cancer Inst, 107(11), Epub Sep 14 (Print Nov 2015). PMID: 26374428.
data obtained from farmers and surrogate respondents on use of agricul- Flower KB, Hoppin JA, Lynch CF, et al. 2004. Cancer risk and parental pes-
tural pesticides. Am J Epidemiol, 134(4), 348–​355. PMID: 1877595. ticide application in children of Agricultural Health Study participants.
Brownson RC, Boehmer TK, and Luke DA. 2005. Declining rates of physical Environ Health Perspect, 112(5), 631–​635. PMCID: PMC1241933
activity in the United States: what are the contributors? Ann Rev Public Fox AJ, and Collier PF. 1976. Low mortality rates in industrial cohort studies
Health, 26, 421–​443. PMID: 15760296. due to selection for work and survival in the industry. Br J Prev Soc Med,
Brudnowska J, and Peplonska B. 2011. Night shiftwork and cancer risk: a lit- 30 (4), 225–​230. PMCID: PMC478970.
erature review. Med Pr, 62(3), 323–​338. PMID: 21870422. Friedman GK. 2011. Malignant diseases attributable to asbestos exposure.
Carey RN, Glass DC, Peters S, et  al. 2014. Occupational exposure to solar In:  Dodson FR, and Hammar SP (Eds.), Asbestos:  Risk Assessment,
radiation in Australia: who is exposed and what protection do they use? Epidemiology, and Health Effects (2nd ed., p.  575). Boca Raton,
Aust N Z J Public Health, 38(1), 54–​59. PMID: 24494947. FL: CRC Press.
Case R, Hosker M, McDonald DB, and Pearson JT. 1954. Tumours of the uri- Friesen M, Lavoue J, Teschke K, and Van Tongeren M. 2015. Occupational expo-
nary bladder in workmen engaged in the manufacture and use of certain sure assessment in industry-​and population-​based epidemiologic stud-
dyestuff intermediates in the British chemical industry. I. The role of ani- ies. In:  Nieuwenhuijsen MJ (Ed.), Environmental Exposure Assessment
line, benzidine, alpha-​naphthylamine, and beta-​naphthylamine. Br J Ind for Epidemiology Studies (2nd ed., pp. 139–​ 162). Oxford:  Oxford
Med, 11(2), 75–​104. PMCID: PMC1037533. University Press.
28

288 PART III:  THE CAUSES OF CANCER


Friesen MC, Zahm SH, Ward MH, and Silverman DT. 2012. Occupation and Huff J, Chan P, and Nyska A. 2000. Is the human carcinogen arsenic carcino-
cancer. In:  Goldman MB, Troisi R, and Rexrode K (Eds), Women and genic to laboratory animals? Toxicol Sci, 55(1), 17–​23. PMID: 10788555.
Health (2nd ed., pp. 629–​645). San Diego, CA: Academic Press/​Elsevier. Huff J, Jacobon MF, and Davis DL. 2008. The limits of two-​year bioassay
Fritschi L, Friesen MC, Glass D, Benke G, Girschik J, and Sadkowsky T. 2009. exposure regimens for identifying chemical carcinogens. Environ Health
OccIDEAS: retrospective occupational exposure assessment in commu- Perspect, 116(11), 1439–​1442. PMCID: PMC2592260.
nity-​based studies made easier. J Environ Public Health, 2009, Article ID IARC. 1987. Overall evaluations of carcinogenicity:  an updating of IARC
957023. doi: 10.1155/​2009/​957023. PMCID: PMC2778192. monographs volumes 1 to 42. IARC Monogr Eval Carcinog Risks Hum,
Fritschi L, Glass DC, Heyworth JS et  al. 2011. Hypotheses for mecha- 7, 1–​440. PMID: 3482203.
nisms linking shiftwork and cancer. Med Hypotheses, 77(3), 430–​436. IARC. 1997. Polychlorinated dibenzo-​ para-​
dioxins and polychlorinated
PMID: 21723672. dibenzofurans. IARC Monogr Eval Carcinog Risks Hum, 69, 1–​631.
Gamble JF, Nicolich MJ, and Boffetta P. 2012. Lung cancer and diesel PMID: 9379504.
exhaust: an updated critical review of the occupational epidemiology lit- IARC. 2006. Formaldehyde, 2-​butoxyethanol and 1-​tert-​butoxypropan-​2-​ol.
erature. Crit Rev Toxicol, 42(7), 549–​598. PMCID: PMC3441149. IARC Monogr Eval Carcinog Risks Hum, 88, 1–​478. PMID: 17366697.
Garshick E, Laden F, Hart JE, et al. 2008. Lung cancer and vehicle exhaust in IARC. 2010a. Painting, firefighting, and shiftwork. IARC Monogr Eval
trucking industry workers. Environ Health Perspect, 116(10), 1327–​1332. Carcinog Risks Hum, 98, 9–​764. PMID: 21381544.
PMCID: PMC2569090. IARC. 2010b. Some aromatic amines, organic dyes, and related exposures.
Gentry PR, Rodricks JV, Tumbull D, et al. 2013. Formaldehyde exposure and IARC Monogr Eval Carcinog Risks Hum, 99, 1–​658. PMID: 21528837.
leukemia: Critical review and reevaluation of the results from a study that IARC. 2012a. Pharmaceuticals. Volume 100 A.  A  review of human car-
is the focus for evidence of biological plausibility. Crit Rev Toxicol, 43(8), cinogens. IARC Monogr Eval Carcinog Risks Hum, 100(Pt A), 1–​401.
661–​670. PMID: 23902349. PMID: 23189749.
Gilbert ES, Koshurnikova NA, Sokolnikov M, et  al. 2000. Liver cancers in IARC. 2012b. Biological agents. Volume 100 B.  A  review of human car-
Mayak workers. Radiat Res, 154(3), 246–​522. PMID: 10956429. cinogens. IARC Monogr Eval Carcinog Risks Hum, 100(Pt B), 1–​441.
Gilbert ES, Koshurnikova NA, Sokolnikov ME, et  al. 2004. Lung cancer in PMID: 23189750.
Mayak workers. Radiat Res, 162(5), 505–​516. PMID: 15624305. IARC. 2012c. Arsenic, metals, fibres, and dusts. Volume 100 C. A review of
GBD 2013 Risk Factors Collaborators. 2015. Global, regional and national human carcinogens. IARC Monogr Eval Carcinog Risks Hum, 100(Pt C),
comparative risk assessment of 79 behavioral, environmental/​occupa- 11–​465. PMID: 23189751.
tional and metabolic risks or clusters of risks in 188 countries 1990–​ IARC. 2012d. Radiation. Volume 100 D. A review of human carcinogens. IARC
2013:  a systematic analysis for the GBD 2013. Lancet, 386 (10010), Monogr Eval Carcinog Risks Hum, 100(Pt D), 7–​303. PMID: 23189752.
2287–​2323. PMID: 26364544. IARC. 2012e. Personal habits and indoor combustions. IARC Monogr Eval
Greenberg M. 1999. A study of lung cancer mortality in asbestos workers: Doll, Carcinog Risks Hum, 100(Pt E), 1–​538. PMID: 23193840.
1955. Am J Ind Med, 36(3), 331–​347. PMID: 10469998. IARC. 2012f. Chemical agents and related occupations. IARC Monogr Eval
Grosse Y, Loomis D, Guyton KZ, et al. 2014. Carcinogenicity of fluoro-​edenite, Carcinog Risks Hum, 100(Pt F), 9–​562. PMID: 23189753.
silicon carbide fibres and whiskers, and carbon nanotubes. Lancet Oncol, IARC. 2013. Diesel and gasoline engine exhausts and some nitroarenes. IARC
15(13), 1427–​1428. PMID: 25499275. Monogr Eval Carcinog Risks Hum, 105, 9–​699, PMID: 26442290.
Gu F, Han J, Laden F et  al. 2015. Total and cause-​ specific mortality of IARC. 2014a. Diesel and gasoline engine exhausts and some nitroarenes. IARC
U.S.  nurses working rotating night shifts. Am J Prev Med, 48(3), 241–​ Monogr Eval Carcinog Risks Hum, 105, 9–​699. PMID: 26442290.
252. PMCID: PMC4339532. IARC. 2014b. Trichloroethylene, tetrachloroethylene, and some other chlo-
Gu J, and Wu X. 2011. Genetic susceptibility to bladder cancer risk and out- rinated agents. IARC Monogr Eval Carcinog Risks Hum, 106, 1–​512.
come. Per Med, 8(3), 365–​374. PMID: PMCID: PMC3172962. PMID: 26214861.
Guyton KZ, Loomis D, Grosse Y, et  al. 2015. Carcinogenicity of tetrachlor- IARC. 2015a. Preamble. Available from:  http://​monographs.iarc.fr/​ENG/​
vinphos, parathion, malathion, diazinon, and glyphosate. Lancet Oncol, Preamble/​currentb6evalrationale0706.php. Accessed April 21, 2015.
16(5), 490–​491. PMID: 25801782. IARC. 2015b. Organ Site Poster, Updated March 2015. Available from: http://​
Hayes RB. 1988. Review of occupational epidemiology of chromium monographs.iarc.fr/​ENG/​Publications/​OrganSitePoster.pdf. Accessed
chemicals and respiratory cancer. Sci Total Environ, 71(3), 331–​339. July 6, 2015.
PMID: 3043655. IARC. 2015c. Polychlorinated biphenyls and polybrominated biphenyls. IARC
Hayes RB, Dosemeci M, Wacholder S, et  al. 1997. Benzene and the dose-​ Monogr Eval Carcinog Risks Hum, 107, 1–​513. Available from:  http://​
related incidence of hematologic neoplasms in China. J Natl Cancer Inst, monographs.iarc.fr/​ENG/​Monographs/​vol107/​mono107.pdf. Accessed
89(14), 1065–​1071. PMID: 9230889. March 28, 2016.
Health and Safety Executive England. 2014. Mesothelioma in Great Britain IARC. 2015d. Some drugs and herbal products. IARC Monogr Eval Carcinog
2014. Available from:  www.hse.gov.uk/​statistics/​causdis/​mesothelioma. Risks Hum, 108, 1–​438. Available from: http://​monographs.iarc.fr/​ENG/​
Accessed September 2, 2015. Monographs/​vol108/​mono108.pdf. Accessed March 28, 2016.
Hill AB, and Faning EL. 1948. Studies in the incidence of cancer in a factory IARC. 2015e. Outdoor air pollution. IARC Monogr Eval Carcinog Risks Hum,
handling inorganic compounds of arsenic; mortality experience in the fac- 109, 1–​454. Available from: http://​monographs.iarc.fr/​ENG/​Monographs/​
tory. Br J Ind Med, 5(1), 1–​6. PMCID: PMC1035980. vol109/​mono109.pdf. Accessed March 28, 2016.
Hoar SK, Morrison AS, Cole P, and Silverman DT. 1980. An occupation and IARC. 2015f. List of classifications by cancer sites with sufficient or limited evi-
exposure linkage system for the study of occupational carcinogenesis. dence in humans, Volumes 1 to 113. Available from: http://​monographs.
J Occup Med, 22(11), 722–​726. PMID: 7441390. iarc.fr/​ENG/​Classification/​Table4.pdf. Accessed September 2, 2015.
Hogstedt C, Malmqvist N, and Wadman B. 1979b. Leukemia in workers Järvholm B, and Englund A. 2014. The impact of asbestos exposure in Swedish
exposed to ethylene oxide. JAMA, 241(11), 1132–​1133. PMID: 105157. construction workers. Am J Ind Med, 57(1), 49–​55. PMID: 24108505.
Hogstedt C, Rohlén O, Berndtsson BS, Axelson O, and Ehrenberg L. 1979b. Kanarek MS. 2011. Mesothelioma from chrysotile asbestos:  update. Ann
A cohort study of mortality and cancer incidence in ethylene oxide Epidemiol, 21(9), 688–​697. PMID: 21820631.
production workers. Br J Ind Med, 36(4), 276–​ 280. PMCID: Kean S. 2012. Daylight at last for study of diesel lung cancer risks. Available
PMC1008604. from:  http://​news.sciencemag.org/​health/​2012/​03/​daylight-​last-​study-​
Hohenadel K, Raj P, Demers PA, Zahm SH, and Blair A. 2015. The inclu- diesel-​lung-​cancer-​risks. Accessed August 29, 2015.
sion of women in studies of occupational cancer:  a review of the epi- Kennaway NM, and Kennaway EL. 1936. A study of the incidence of cancer of
demiologic literature from 1991–​2009. Am J Ind Med, 58(3), 276–​281. the lung and larynx. J Hyg (Lond), 36(2), 236–​267. PMCID: PMC2170982.
PMID: 25678456. Kesminiene A, Cardis E, Tenet V, et  al. 2002. Studies of cancer risk among
Hohenadel K, Harris SA, McLaughlin JR, et  al. 2011. Exposure to mul- Chernobyl liquidators:  materials and methods. J Radiol Prot, 22(3A),
tiple pesticides and risk of non-​Hodgkin lymphoma in men from six A137–​A141. PMID: 12400962.
Canadian provinces. Int J Environ Res Public Health, 8(6), 2320–​2330. Kesminiene A, Evrard AS, Ivanov VK, et  al. 2008. Risk of hematological
PMCID: PMC3138027. malignancies among Chernobyl liquidators. Radiat Res, 170(6), 721–​735.
Hornung RW, Greife AL, Stayner LT, et al. 1994. Statistical model for predic- PMCID: PMC2904977.
tion of retrospective exposure to ethylene oxide in an occupational mor- Koshurnikova NA, Gilbert ES, Sokolnikov M et  al. 2000. Bone cancers in
tality study. Am J Ind Med, 25(6), 825–​836. PMID: 8067360. Mayak workers. Radiat Res, 154(3), 237–​245. PMID: 10956428.
Huff J. 1999. Long-​term chemical carcinogenesis bioassays predict human can- Koutros S, Beane Freeman LE, Berndt SI, et al. 2010. Pesticide use modifies
cer hazards: issues, controversies, and uncertainties. Ann N Y Acad Sci, the association between genetic variants on chromosome 8q24 and pros-
895, 56–​79. PMID: 10676409. tate cancer. Cancer Res, 70(22), 9224–​9233. PMCID: PMC2982856.
 289

Occupational Cancer 289
Koutros S, Berndt SI, Hughes Barry K, et al. 2013. Genetic susceptibility loci, Morris J. 2012. Diesel report’s publication delayed as industry demands to
pesticide exposure and prostate cancer risk. PLoS One, 8(4), e58195. see documents first, Feb 6, 2012. Available from:  http://​www.washing-
PMCID: PMC3617165. tonpost.com/​national/​health-​science/​diesel-​reports-​publication-​delayed-​
Kubo S, Nakanuma Y, Takemura S, et  al. 2014. Case series of 17 patients as-​industry-​demands-​to-​see-​documents-​first/​2012/​02/​01/​gIQA5wrFtQ_​
with cholangiocarcinoma among young adult workers of a print- story.html. Accessed August 29, 2015.
ing company in Japan. J Hepatobiliary Pancreat Sci, 21(7), 479–​488. Olson JS. 2002. Bathsheba’s Breast: Women, Cancer and History (pp. 32–​33).
PMID: 24420816. Baltimore, MD: The Johns Hopkins University Press.
Kumagai S, Kurumatani N, Arimoto A, and Ichihara G. 2013. Pearce N, Blair A, Vineis P, et al. 2015. IARC monographs: 40 years of evalu-
Cholangiocarcinoma among offset colour proof-​printing workers exposed ating carcinogenic hazards to humans. Environ Health Perspect, 123(6),
to 1,2-​dichloropropane and/​or dichloromethane. Occup Environ Med, 507–​514. PMCID: PMC4455595.
70(7), 508–​510. PMID: 23493378. Pott P. 1775. Chirurgical Observations Relative to the Cataract, the Polypus of
Kumar A, and Dhawan A. 2013. Genotoxic and carcinogenic potential of the Nose, the Cancer of the Scrotum, the Different Kinds of Ruptures and
engineered nanoparticles:  an update. Arch Toxicol, 87(11), 1883–​1990. the Mortification of the Toes and Feet. London: TJ Carnegy.
PMID: 24068037. Purdue MP, Hutchings SJ, Rushton L, and Silverman DT. 2015. The proportion
Lan Q, Zhang L, Li G, et al. 2004. Hematotoxicity in workers exposed to low lev- of cancer attributable to occupational exposures. Ann Epidemiol, 25(3),
els of benzene. Science, 306(5702), 1774–​1776. PMCID: PMC1256034. 188–​192. PMID: 25487971.
Lehman EJ, Hein MJ, and Estill CF. 2008. Proportionate mortality study of the Rachiotis G, Drivas S, Kostikas K, Makaropoulos V, and Hadjichristodoulou
United Association of Journeyman and Apprentices of the Plumbing and C. 2012. Respiratory tract mortality in cement workers: a proportionate
Pipe Fitting Industry. Am J Ind Med, 51(12), 950–​963. PMID: 18942099. mortality study. BMC Pulm Med, 12, 30. doi: 10.1186/​1471-​2466-​12-​30.
Lerro CC, Koutros S, Andreotti G, et al. 2015. Use of acetochlor and cancer PMCID: PMC3425084.
incidence in the Agricultural Health Study. Int J Cancer, 137(5), 1167–​ Raj P, Hohenadel K, Demers P, Zahm SH, and Blair A. 2014. Recent trends
1175. PMCID: PMC4492894. in published occupational cancer epidemiology research: results from a
Leuraud K, Richardson DB, Cardis E, et al. 2015. Ionising radiation and risk comprehensive review of the literature. Am J Ind Med, 57(3), 259–​264.
of death from leukaemia and lymphoma in radiation-​monitored work- PMID: 24488816.
ers (INWORKS): an international cohort study. Lancet Haematol, 2(7), Ramazzini B. 1713. Diseases of Workers. Translated from the Latin text: De
e276–​e281. PMID: 26436129. Morbis Artificum of 1713 by WC Wright. New  York Academy of
Levêque-​Morlais N, Tual S, Clin B, Adjemian A, Baldi I, and Lebailly P. 2015. Medicine, History of Medicine Series No. 23, 1964. New York: Hafner.
The AGRIculture and CANcer (AGRICAN) cohort study:  enrollment Rim KT, Song SW, and Kim HY. 2013. Oxidative DNA damage from nanopar-
and causes of death for the 2005–​2009 period. Int Arch Occup Environ ticle exposure and its application to workers’ health: a literature review.
Health, 88(1), 61–​73. PMID: 24599726. Saf Health Work, 4(4), 177–​186. PMCID: PMC3889076.
Linet MS, Kim KP, Miller DL, Kleinerman RA, Simon SL, and Berrington de Robinson CF, Walker JT, Sweeney MH, et al. 2015. Overview of the National
González A. 2010. Historical review of occupational exposures and can- Occupational Mortality Surveillance (NOMS) system:  leukemia and
cer risks in medical radiation workers. Radiation Res, 174(6), 793–​808. acute myocardial infarction risk by industry and occupation in 30 US
PMCID: PMC4098897. states 1985–​1999, 2003–​2004, and 2007. Am J Ind Med, 58(2), 123–​137.
Lloyd JW. 1971. Long-​term mortality study of steelworkers. V. Respiratory can- PMCID: PMC4573401.
cer in coke plant workers. J Occup Med, 13(2), 53–​68. PMID: 5546197. Rodvall Y, Dich J, and Wiklund K. 2003. Cancer risk in offspring of male pes-
Locke SJ, Colt JS, Stewart PA, et  al. 2014. Identifying gender differ- ticide applicators in agriculture in Sweden. Occup Environ Med, 60(10),
ences in reported occupational information from three US popula- 798–​801. PMCID: PMC1740388.
tion-​based case-​control studies. Occup Environ Med, 71(12), 855–​864. Romanenko A, Bebeshko V, Hatch M et  al. 2008. The Ukrainian-​American
PMCID: PMC4177972. study of leukemia and related disorders among Chornobyl cleanup
Loomis D, Guyton K, Grosse Y, et al. 2015. Carcinogenicity of lindane, DDT, workers from Ukraine: I. Study methods. Radiat Res, 170(6), 691–​697.
and 2,4-​ dichlorophenoxyacetic acid. Lancet Oncol, 16(8), 891–​ 892. PMCID: PMC2856482.
PMID: 26111929. Romanenko AY, Finch SC, Hatch M et  al. 2008. The Ukrainian-​American
Loutit JF. 1970. Malignancy from radium. Br J Cancer, 24(2), 195–​ 207. study of leukemia and related disorders among Chornobyl cleanup work-
PMCID: PMC2008571. ers from Ukraine:  III. Radiation risks. Radiat Res, 170(6), 711–​720.
Lynge E, Andersen A, Rylander L, et al. 2006. Cancer in persons working in PMCID: PMC2856603.
dry cleaning in the Nordic countries. Environ Health Perspect, 114(2), Rothman K, Greenland S, and Lash T. 2008. Modern Epidemiology (pp. 122–​
213–​219. PMCID: PMC1367834. 124). New York: Lippincott, Williams and Wilkins.
Machle W, and Gregorius F. 1948. Cancer of the respiratory system in the Rushton L, Hutchings SJ, Fortunato L, et  al. 2012. Occupational cancer
United States chromate producing industry. Public Health Rep, 63(35), burden in Great Britain. Br J Cancer, 107(Suppl 1), S3–​S7. PMCID:
1114–​1127. PMCID: PMC1995382. PMC3384015.
Maronpot RR, Flake G, and Huff J. 2004. Relevance of animal carcinogen- Russ DE, Ho KY, Colt JS, et al. 2016. Computer-​based coding of free-​text job
esis findings to human cancer predictions and prevention. Toxicol Pathol, descriptions to efficiently identify occupations in epidemiologic studies.
32(Suppl 1), 40–​48. PMID: 15209402. Occup Environ Med, 73(6), 417–​424. PMCID: PMC4871757.
Mason TJ, McKay FW, Hoover R, Blot WJ, and Fraumeni JF, Jr. 1975. Atlas Russ DE, Ho KY, Johnson CA, and Friesen MC. 2014. Computer-​based coding
of cancer mortality for U.S.  counties:  1950–​1969. [DHEW Publ No. of occupation codes for epidemiological analyses. Proc IEEE Int Symp
(NIH)75–​780]. Washington, DC: US Government Printing Office. Comput Based Med Syst, 2014, 347–​350. PMCID: PMC4161468.
Matthews CE, George SM, Moore SC, et al. 2012. Amount of time spent in sed- Sandler DP, Kwok RK, Engel LS, et al. 2014. GuLF STUDY: Gulf Long-​Term
entary behaviors and cause-​specific mortality in US adults. Am J Clinical Follow-​ Up Study. Study Protocol, Version 24.0, December 6, 2014.
Nutr, 95(2), 437–​445. PMCID: PMC3260070. Available from: http://​www.niehs.nih.gov/​research/​atniehs/​labs/​epi/​stud-
Mayeux R. 2004. Biomarkers:  potential uses and limitations. NeuroRx, 1(2), ies/​gulfstudy/​publications/​gulf_​study_​protocol_​1262014.pdf. Accessed
182–​188. PMCID: PMC534923. September 2, 2015.
McLaughlin JK, and Tarone RE. 2014. Mortality from solid tumors in Schmidt CW. 2007. A change in the air:  smoking bans gain momen-
the updated formaldehyde cohort. Am J Ind Med, 57(4), 486–​ 487. tum worldwide. Environ Health Perspect, 115(8), A412–​A415.
PMID: 24501017. PMCID: PMC1940108.
Melnick RL, and Huff J. 1992. 1,3-​Butadiene: toxicity and carcinogenicity in Schnatter AR, Glass DC, Tang G, Irons RD, and Rushton L. 2012.
laboratory animals and in humans. Rev Environ Contam Toxicol, 124, Myelodydplastic syndrome and benzene exposure among petroleum
111–​144. PMID: 1732994. workers:  an international pooled analysis. J Natl Cancer Inst, 104(22),
Nielsen GD, and Ovrebø S. 2008. Background, approaches and recent trends 1724–​1737. PMCID: PMC3502195.
for setting health-​based occupational exposure limits: a minireview. Regul Schubauer-​Berigan M, Dahm M, and Yencken M. 2011. Engineered carbona-
Toxicol Pharmacol, 51(3), 253–​69. PMID: 18502550. ceous nanomaterials manufacturers in the United States: workforce size,
Miligi L, Costantini AS, Bolejack V, et al. 2003. Non-​Hodgkin’s lymphoma, characteristics, and feasibility of epidemiologic studies. J Occup Environ
leukemia, and exposures in agriculture: results from the Italian multicenter Med, 53(6 Suppl), S62–​S67. PMID: 21654420.
case-​control study. Am J Ind Med, 44(6), 627–​636. PMID: 14635239. Schulte PA. 1993. A conceptual and historical framework for molecu-
Monforton C. 2006. Weight of the evidence or wait for the evidence? Protecting lar epidemiology. In:  Schulte PA, and Perera FP (Eds.), Molecular
underground miners from diesel particulate matter. Am J Public Health, Epidemiology:  Principles and Practices (pp. 3–​ 44). San Diego,
96(2), 271–​276. PMCID: PMC1470492. CA: Academic Press.
290

290 PART III:  THE CAUSES OF CANCER


Shealy DB, Barr JR, Ashley DL, Patterson DG Jr, Camann DE, and Bond AE. Thun M. 1992. Kidney dysfunction in cadmium workers. In:  Steenland K
1997. Correlation of environmental carbaryl measurements with serum (Ed.), Case Studies in Occupational Epidemiology. Oxford:  Oxford
and urinary 1-​naphthol measurements in a farmer applicator and his fam- University Press.
ily. Environ Health Perspect, 105(5), 510–​513. PMCID: PMC1469864. Tsuji JS, Alexander DD, Perez V, and Mink PJ. 2014. Arsenic and bladder
Siemiatycki J. 1991. Risk Factors for Cancer in the Workplace. CRC Press, cancer: quantitative assessment of studies in human populations to detect
Boca Raton, FL: CRC Press. risks at low doses. Toxicology, 317, 17–​30. PMID: 24462659.
Siemiatycki J. 2014. Historical overview of occupational cancer research. Tual S, Silverman DT, Koutros S, et  al. 2015. Use of dieselized farm
In:  Anttila S, and Boffetta P (Eds.), Occupational Cancer (pp. 1–​20). equipment and incident lung cancer:  findings from the Agricultural
London: Springer-​Verlag. Health Study cohort. Environ Health Perspect, 124 (5), 611-​ 618.
Siemiatycki J, Fritschi L, Nadon L, and Gérin M. 1997. Reliability of an expert PMCID: PMC4858397.
rating procedure for retrospective assessment of occupational exposures US Department of Health and Human Services. 1996. Physical Activity and
in community-​based case-​control studies. Am J Ind Med, 31(3), 280–​286. Health: A Report of the Surgeon General. Atlanta, GA: US Department
PMID: 9055950. of Health and Human Services, Centers for Disease Control and
Siemiatycki J, Richardson L, Straif K, et  al. 2004. Listing occupa- Prevention, National Center for Chronic Disease Prevention and Health
tional carcinogens. Environ Health Perspect, 112(15), 1447–​ 1459. Promotion.
PMCID: PMC1247606. van der Bij S, Koffijberg H, Lenters V, et  al. 2013. Lung cancer risk at low
Silverman D, Samanic C, Lubin JH, et  al. 2012. The diesel exhaust in min- cumulative asbestos exposure: meta-​regression of the exposure-​response
ers study: a nested case-​control study of lung cancer and diesel exhaust. relationship. Cancer Causes Control, 24(1), 1–​12. PMID: 23187858.
J Natl Cancer Inst, 104(11), 855–​868. PMCID: PMC3369553. Vermeulen R, Silverman DT, Garshick E, Vlaanderen J, Portengen L, and
Snellings WM, Weil CS, and Maronpot RR. 1984. A two-​year inhalation study Steenland K. 2014. Exposure-​response estimates for diesel engine exhaust
of the carcinogenic potential of ethylene oxide in Fischer 344 rats. Toxicol and lung cancer mortality based on data from three occupational cohorts.
Appl Pharmacol, 75(1), 105–​117. PMID: 6464016. Environ Health Perspect, 122(2), 172–​177. PMCID: PMC3915263.
Solan S, Wallenstein S, Shapiro M, et  al. 2013. Cancer incidence in world Vlaanderen J, Moore LE, Smith MT, et  al. 2010. Application of OMICS
trade center rescue and recovery workers, 2001–​2008. Environ Health technologies in occupational and environmental health research; cur-
Perspect, 121(6), 699–​704. PMCID: PMC3672914. rent status and projections. Occup Environ Med, 67(2), 136–​ 143.
Stark WJ, Stoessel PR, Wohlleben W, and Hafner A. 2015. Industrial applications PMCID: PMC2910417.
of nanoparticles. Chem Soc Rev, 44(16), 5793–​5805. PMID:25669838. Wagner J, Sleggs C, and Marchand P. 1960. Diffuse pleural mesothelioma and
Stayner L, Steenland K, Dosemeci M, and Hertz-​Picciotto I. 2003. Attenuation of asbestos exposure in the North Western Cape Province. Br J Ind Med,
exposure-​response curves in occupational cohort studies at high exposure 17(Oct), 260–​271. PMCID: PMC1038078.
levels. Scand J Work Environ Health, 29(4), 317–​324. PMID: 12934726. Wagoner J, Archer V, Lundin F, Holday D, and Lloyd J. 1965. Radiation as
Steenland K, Burnett C, Lalich N, Ward E, and Hurrell J. 2003. Dying for the cause of lung cancer among uranium miners. N Engl J Med, 273(4),
work: the magnitude of US mortality from selected causes of death asso- 181–​188. PMID: 14306333.
ciated with occupation. Am J Ind Med, 43(5), 461–​482. PMID: 12704620. Waldron HA. 1983. A brief history of scrotal cancer. Br J Ind Med, 40(4), 390–​
Steenland K, and Moe C. 2016. Environmental and occupational epidemiology. 401. PMCID: PMC1009212.
In: Frumkin H (Ed.), Environmental Health: From Global to Local (2nd Ward EM, Schulte PA, Straif K et  al. 2010. Research recommendations for
ed., pp. 83–​109). San Francisco: John Wiley & Sons. selected IARC-​ classified agents. Environ Health Perspect, 118(10),
Steenland K, Seals R, Klein M, Jinot J, and Kahn HD. 2011. Risk estimation 1355–​1362. PMCID: PMC2957912.
with epidemiologic data when response attenuates at high-​exposure lev- Wei S, Wang LE, McHugh MK et al. 2012. Genome-​wide gene-​environment
els. Environ Health Perspect, 119(6), 831–​837. PMID: 21220221. interaction analysis for asbestos exposure in lung cancer susceptibility.
Steenland NK, Silverman DT, and Hornung RW. 1990. Case-​control study Carcinogenesis, 33(8), 1531–​1537. PMCID: PMC3499061.
of lung cancer and truck driving in the Teamsters Union. Am J Public Westman JA, Ferketich AK, Kauffman RM, et  al. 2010. Low cancer inci-
Health, 80(6), 670–​674. PMCID: PMC3114819. dence rates in Ohio Amish. Cancer Causes Control, 21(1), 69–​ 75.
Stewart B, and Wild C (Eds.). 2014. World Cancer Report 2014. Lyon, PMCID: PMC4308689.
France: International Agency for Research on Cancer. Yarborough CM. 2006. Chrysotile as a cause of mesothelioma:  an assess-
Stewart PA, Stewart WF, Siemiatycki J, Heineman EF, and Dosemeci M. 1998. ment based on epidemiology. Crit Rev Toxicol, 36(2), 165–​ 187.
Questionnaires for collecting detailed occupational information for com- PMID: 16736942.
munity-​based case control studies. Am Ind Hyg Assoc J, 59(1), 39–​44. Yoshinaga S, Mabuchi K, Sigurdson AJ, Doody MM, and Ron E. 2004. Cancer
PMID: 9438334. risks among radiologists and radiologic technologists:  review of epide-
Stewart PA, Coble JB, Vermeulen R, et al. 2010. The diesel exhaust in miners miologic studies. Radiology, 233(2), 313–​321. PMID: 15375227.
study: I. Overview of the exposure assessment process. Ann Occup Hyg, Zablotska LB, Bazyka D, Lubin JH et al. 2013. Radiation and the risk of chronic
54(7), 728–​746. PMCID: PMC2953554. lymphocytic and other leukemias among Chornobyl cleanup workers.
Straif K. 2012. Estimating the burden of occupational cancer as a strate- Environ Health Perspect, 121(1), 59–​65. PMCID: PMC3553431.
gic step to prevention. Brit J Cancer, 107(Suppl 1), S1–​S2. PMCID: Zahm SH, and Blair A. 2003. Occupational cancer among women:  where
PMC3384019. have we been and where are we going? Am J Ind Med, 44(6), 565–​575.
Straker L, and Mathiassen SE. 2009. Increased physical work loads in modern PMID: 14635233.
work: a necessity for better health and performance? Ergonomics, 52(10), Zahm SH, Pottern LM, Lewis DR, Ward MH, and White DW. 1994. Inclusion
1215–​1225. PMID: 19787501. of women and minorities in occupational-​cancer epidemiologic studies.
Sun Y, Bochmann F, Nold A, and Mattenklott M. 2014. Diesel exhaust exposure J Occup Med, 36(8), 842–​847. PMID: 7807263.
and risk of lung cancer:  a review of the epidemiologic evidence. Int J Zahm SH, Ward MH, and Silverman DT. 1999. Occupational cancer.
Environ Res Public Health, 11(2), 1312–​1340. PMCID: PMC3945540. In: Goldman MB, and Hatch MC (Eds.), Women and Health (pp. 493–​502).
Tagnon I, Blot WJ, Stroube RB, et al. 1980. Mesothelioma associated with the San Diego, CA: Academic Press.
shipbuilding industry in coastal Virginia. Cancer Res, 40(11), 3875–​3879. Zahm SH, and Ward MH. 1998. Pesticides and childhood cancer. Environ
PMID: 7471040. Health Perspect, 106(Suppl 3), 893–​908. PMCID: PMC1533072.
Takahashi K, and Ishii Y. 2013. Historical developments of administrative mea- Zahm SH, Weisenburger DD, Babbitt PA et  al. 1990. A case-​control study
sures for occupational diseases in Japan, a report commissioned by the of non-​ Hodgkin’s lymphoma and the herbicide 2,4-​ dichlorophenoxy-
ILO, 10 July 2013. Available from:  http://​www.ilo.org/​wcmsp5/​groups/​ acetic acid (2,4-​D) in eastern Nebraska. Epidemiology, 1(5), 349–​56.
public/​ - ​ - ​ - ​ e d_​ p rotect/ ​ - ​ - ​ - ​ p rotrav/ ​ - ​ - ​ - ​ s afework/ ​ d ocuments/ ​ p ublication/​ PMID: 2078610.
wcms_​234221.pdf. Accessed September 2, 2015. Zhang L, Tang X, Rothman N, et al. 2010. Occupational exposure to formal-
Teschke K, Olshan AF, Daniels JL, et al. 2002. Occupational exposure assess- dehyde, hematotoxicity, and leukemia-​specific chromosome changes in
ment in case-​ control studies:  opportunities for improvement. Occup cultured myeloid progenitor cells. Cancer Epidemiol Biomarkers Prev,
Environ Med, 59(9), 575–​593. PMCID: PMC1740358. 19(1), 80–​88. PMCID: PMC2974570.
 291

17 Air Pollution

JONATHAN M. SAMET AND AARON J. COHEN

OVERVIEW air pollution increases lung cancer risk. By 2013, the totality of the
evidence on outdoor air pollution and cancer had become sufficiently
A wide variety of man-​made and naturally occurring air pollutants cohesive to support a conclusion by the International Agency for
are known to cause cancer. Diverse exposures such as tobacco smoke, Research on Cancer (IARC) that outdoor air pollution is a Group  1
radionuclides (radon), chemicals (benzene, mustard gas, and volatile carcinogen (IARC, 2015).
organic compounds), fibers (asbestos), and metals and metalloids During recent decades, a number of airborne carcinogens, viewed
(chromium, nickel, and arsenic) have long been classified as carcino- as potential threats to public health, have been particularly contro-
genic to humans. Historically, the evidence base for these classifica- versial. The energy crisis of the early 1970s led to increased man-
tions predominantly concerned high levels of exposure in occupational ufacture of diesel-​powered vehicles; recognition that diesel-​ soot
settings. Over the last 30–​40 years, scientific attention has focused on particles were mutagenic raised concern that the increasing numbers
quantifying the adverse health effects of indoor and outdoor air pollut- of diesel-​powered vehicles would increase lung cancer risk for the
ants at exposure levels several orders of magnitude lower than those population. Three indoor carcinogens received widespread attention
studied initially. These include secondhand smoke, household expo- from the scientific community and the public during the 1980s and
sure to radon, residential and environmental exposure to asbestos, soot 1990s: tobacco smoke inhaled by non-​smokers, radon, and asbestos
from diesel powered engines, ambient exposures to small particles fibers. Policies and programs for reducing the risks to the popula-
(PM2.5), and indoor air pollution from the combustion of biomass and tion of each of these carcinogens became extremely controversial.
coal. This chapter provides an overview of recent epidemiologic stud- Critics questioned whether the risks were exaggerated and whether
ies of air pollutants and cancer. It is necessarily selective, because the the high costs of control, particularly for radon and asbestos, were
evidence on air pollution and lung cancer in particular is now exten- justified. The scientific evidence on secondhand smoke was repeat-
sive. Issues fundamentally important to policy development concern edly challenged by the tobacco industry as it sought to maintain con-
dose–​response relationships and risk assessment. Of great concern is troversy, even as review panels concluded that secondhand smoke
that exposures in economically developing countries rival and often caused lung cancer (IARC, 1986; National Research Council and
dramatically exceed those found in high-​income Western countries. Committee on Passive Smoking, 1986). Now, definitive and incon-
trovertible conclusions on the carcinogenicity of secondhand smoke
have been made by both the IARC (2004) and the Surgeon General
INTRODUCTION (2006). In the United States, there has also been extensive litigation
around indoor asbestos in the past. For these and other inhaled car-
In the early decades of the twentieth century, lung cancer was a rare cinogens, substantial research has been motivated by concern for the
disease. By mid-​century, however, it was evident that an epidemic of public’s health, and by the need to develop the scientific foundation
lung cancer was occurring among males in the United States and a of evidence for control strategies to address carcinogens for which
number of European countries, and a parallel epidemic followed at exposure is widespread.
mid-​century among women. By then, there were already several estab- This chapter provides an overview of the evidence that outdoor
lished and suspect causes of lung cancer. The evidence was stron- and indoor air pollution are causally related to lung and other types of
gest for radon, linked to lung cancer in underground miners based cancer. The topic of secondhand smoke and lung cancer is addressed
on epidemiological and clinical observations in mines in Eastern in Chapter 11 of this volume as well. The evidence on air pollution
Europe and the measurement of high levels of radon in these mines. and lung cancer is now extensive and this review is necessarily selec-
Epidemiological research was implemented to search for causal fac- tive, emphasizing the most recent findings, primarily from the epi-
tors, targeting specific groups of workers at high risk as well as the demiologic literature. A 1990 monograph provides a more complete
general population. By the 1920s and 1930s, clinicians were already review of the earlier literature on outdoor air pollution (Tomatis,
beginning to make the link between smoking and lung cancer, and the 1990). The IARC has published a review monograph that compre-
ubiquitous and serious air pollution in cities was also considered as a hensively covers combustion products, including indoor and outdoor
potential cause, a hypothesis supported by the presence of an urban–​ air pollution, and cancer, as well as completing a series of volumes
rural gradient in risk (Hoffman, 1929, 1931; Macklin and Macklin, on the carcinogenicity of these agents (IARC, 2010a, 2010b, 2013b,
1940; Overholt and Rumel, 1940; Stocks and Campbell, 1955). These 2013c, 2015). This chapter focuses on outdoor, primarily urban, air
two non-​exclusive hypotheses were given equal weight by Doll and pollution and indoor air contaminants in relation to lung cancer in
Hill (Doll and Hill, 1952)  as a rationale for their case-​control study high-​income countries. Mounting evidence indicates that people in
of lung cancer in London. After smoking was quickly identified as developing countries have exposures to indoor and outdoor environ-
a powerful cause of lung cancer, concern persisted that air pollution ments that rival and often dramatically exceed those found in high-​
may cause lung and other cancers, and research was ongoing on this income Western countries (Gordon et al., 2014). Indoor air pollution
hypothesis. Additionally, by the 1950s, other respiratory carcinogens from coal combustion and cooking fumes are thought to account for
had been identified, including radon and asbestos. the high lung cancer risk among women in China and Hong Kong,
The hypothesis that air pollution causes lung cancer had a strong despite the low prevalence of smoking (Bruce et al., 2015). Rising
basis in the known release of carcinogens into outdoor air from pollution of outdoor air in the mega-​cities of the developing world
industrial sources, power plants, and motor vehicles, and in the rec- also poses a risk for lung cancer. These topics are addressed else-
ognition that indoor air is contaminated by respiratory carcinogens. where (Brauer et al., 2012, 2016; Katsouyanni, 2013; Krzyzanowski
Historically, epidemiological studies have shown for decades that et al., 2014).

291
29

292 PART III:  THE CAUSES OF CANCER

EXPOSURES TO INHALED CARCINOGENS Table 17–​1. Summary Concentrations of Air Toxics “Pollutants


of Interest” in the United States for 2010

Factors That Affect Exposure Compound Median 25th Percentile 75th Percentile
Exposures to inhaled carcinogens take place in a variety of settings, PAHsa
including the home, the workplace, other public and commercial Acenaphthene 2.04 0.983 4.30
locations, and outdoors. The concept of total personal exposure pro- Benzo(a)pyrene 0.02 0 0.1
vides a useful framework for conceptualizing exposures to inhaled Fluorene 2.91 1.75 5.39
carcinogens and evaluating the contributions of outdoor and indoor Naphthalene 66.4 36.6 117
air pollution (National Research Council et al., 2012). Total personal
exposure represents the integrated exposure to an agent accumulated Metalsb
in multiple microenvironments (i.e., environments having a relatively Arsenic (PM10) 0.415 0.240 0.700
homogeneous concentration of the agent of interest during a speci- Beryllium (PM10) 0.002 0.0003 0.004
fied time period). For an inhaled carcinogen such as benzo[a]‌pyrene, Cadmium (PM10) 0.084 0.050 0.176
relevant microenvironments over the day might include outdoor air Lead (PM10) 2.32 1.45 3.74
contaminated by vehicle exhaust and industrial emissions, workplace Manganese (PM10) 4.03 2.15 7.97
exposures, and air contaminated by tobacco smoke in a bar. For each Nickel (PM10) 0.845 0.594 1.22
microenvironment, the exposure received depends on the concentra- Hexavalent chromium 0.018 0 0.032
tion of the carcinogen and the time spent in the microenvironment. Carbonylsc
Total personal exposure is the sum of the products of concentration Acetaldehyde 0.893 0.597 1.29
with time spent in each microenvironment. Formaldehyde 1.66 1.09 2.47
Also relevant is the total volume of inhaled air, which for adults VOCsd
is about 10,000 liters per day. Because of the large volume inhaled, Acrylonitrile 0 0 0
meaningful doses of carcinogens may reach the lung at relatively low Benzene 0.245 0.173 0.373
concentrations. The actual lung dose of the carcinogen will further 1,3-​Butadiene 0.026 0.012 0.048
depend on the exposed person’s ventilation rate and pattern, physical Carbon tetrachloride 0.037 0.013 0.272
characteristics of the agent, and other factors that affect the location Chloroform 0.020 0.014 0.031
and amount of deposition in the lung (National Research Council and p-​Dichlorobenzene 0.007 0 0.019
Panel on Dosimetric Assumptions Affecting the Application of Radon 1,2-​Dichloroethane 0 0 0
Risk Estimates, 1991). Inhaled carcinogens may be absorbed into the Ethylbenzene 0.053 0.03 0.1
systemic circulation. Doses to other organs (e.g., the urinary bladder) Tetrachloroethylene 0.016 0.008 0.03
depend on uptake, metabolism, distribution, and excretion. Trichlororethylene 0 0 0
The relevant microenvironments vary depending on the carcinogen Vinyl chloride 0 0 0
of concern. Because most time is typically spent at home, exposures to
contaminants in household air dominate for certain pollutants such as
PAHs: polycyclic aromatic hydrocarbons; PM10: particulate matter with particles of
radon. Based on time-​activity patterns and policies regarding smoking, aerodynamic diameter, 10μm; SD: standard deviation; VOCs: volatile organic compounds.
the workplace can be a significant source of exposure to secondhand a
PAHs: unit is ng/​m3.
smoke as well as industrial carcinogens. In high-​income countries, b
Metals: unit is ng/​m3.
adults generally spend little time outdoors, but pollutants in ambient
c
Carbonyls: unit is ppbv.
d
VOCs: unit is ppbv.
air can diffuse into homes. The microenvironments in which exposures Source: IARC, 2015
occur influence the choice of control strategies. Some environments
are shared and public; initiatives to control exposure in these settings
require action at the societal level, often involving regulations. Other Incinerator emissions are also mutagenic; their activity reflects PAH
environments are private, and control lies with individuals. Whereas content and nitroarenes (DeMarini et al., 1998). The IARC has sum-
regulations are often essential to control toxic exposures in public set- marized the findings of hundreds of studies of the mutagenicity of par-
tings, education is usually the mainstay of initiatives to reduce expo- ticles collected in outdoor air (IARC, 2015). Investigators have used
sures to carcinogens in the home. biomarkers of exposure to ambient air pollutants to better characterize
patterns of exposure. These studies have been primarily focused on
polycyclic aromatic hydrocarbons and markers of oxidative stress. The
OUTDOOR AIR POLLUTION results have been mixed. In a 1992 study in Poland (Perera et al., 1992),
Perrera and colleagues measured PAH-​DNA adducts in lymphocytes
as markers of molecular and genetic damage. Residents of a highly
Exposures to Carcinogens in Outdoor Air
industrialized and polluted area had higher mean levels of PAH-​DNA
Ambient air, particularly in densely populated urban environments, adducts than residents of a comparison area. Autrup and colleagues
contains a variety of known human carcinogens, including organic (Autrup et  al., 1999)  compared measurements of bulky carcinogen-​
compounds such as benzo[a]‌ pyrene and benzene, inorganic com- DNA adducts and 2-​amino-​apidic semialdehyde, a marker of oxidative
pounds such as arsenic and chromium, and radionuclides (Table 17–​1) stress, in non-​smoking Danish bus drivers and postal workers. The lev-
(IARC, 2015). These substances are present as components of com- els of adducts, but not the biomarker of oxidative stress, were higher in
plex mixtures, which may include carbon-​based particles to which the the bus drivers in central Copenhagen. In a study in Germany, markers
organic compounds are adsorbed, oxidants such as ozone, and aerosols of exposure to benzo[a]‌pyrene tended to be higher in city dwellers,
of sulfuric acid. The combustion of fossil fuels for power generation or compared with suburban dwellers. In the AULIS project in Greece,
transportation is the main source of organic and inorganic compounds, the mean levels of bulky DNA adducts were higher in residents of
oxidants, and acids; combustion byproducts contribute heavily to par- the the rural town of Halkida than in Athens (Georgiadis et al., 2001;
ticulate air pollution in most urban settings. Radionuclides also result Kyrtopoulos et  al., 2001), although the levels were also higher in
from fuel combustion, as well as from mining operations. those exposed to secondhand smoke. The findings in relation to resi-
The plausibility that outdoor air pollutants cause cancer is sup- dence location were unanticipated, given that prior studies tended to
ported by studies that have addressed its genotoxicity using in vitro show higher levels of markers of genotoxicity in urban workers and
assay systems and biomarkers. Urban air samples are mutagenic in residents (Kyrtopoulos et  al., 2001). By contrast, in a study of stu-
the Salmonella assay, and much of the activity is attributable to poly- dents in Copenhagen, measured personal exposure to small particles
cyclic aromatic hydrocarbons (PAHs) (DeMarini, 2013; IARC, 2015). was associated with 7-​hydro-​8-​oxo-​2′-​deoxyguanosine, a marker of
 293

Air Pollution 293


DNA damage in lymphocytes; other associations between exposure risk assessment (see later in this chapter). Armstrong and colleagues
and biomarker levels were not found (IARC, 2015). The differences in have reviewed the literature on cancer risk in relation to occupational
findings may reflect the differing levels of exposure and the markers and environmental exposure to PAHs (Armstrong et al., 2004). They
considered. concluded that PAHs are associated with increased lung cancer risk in
Unfortunately, there are few databases that record long-​term occupational and urban environments. Workers exposed to mixtures of
measurements of carcinogens and other products from fossil fuel polycyclic compounds in coke-​ovens and coal gasification plants (Doll
combustion that could be used in epidemiologic studies. Available et al., 1972; Redmond, 1983) have increased lung cancer risk (IARC,
data indicate that there have been improvements in some indi- 2012b). The levels of POM encountered in urban air are substantially
ces of air quality in the United States and some other developed less than in these settings.
countries in recent decades (US Environmental Protection Agency
[EPA], 2016). Particles
Particulate matter with an aerodynamic diameter < 2.5 μm, PM2.5, has The IARC’s Outdoor Air Pollution (Volume 109) classified particulate
been the airborne pollutant of greatest current interest with respect to matter in outdoor air pollution as a Group 1 carcinogen (IARC, 2015).
lung cancer because particles of size 2.5 μm or less can be inhaled and Like POM, particulate air pollution is not a single entity, but rather
deposited in the lung. These generally originate from combustion and a chemically and physically diverse group of pollutants. Particles
may transport carcinogens, such as polycylic aromatic hydrocarbons, derive from sea spray and crystal dust, as well as from combustion of
on their surfaces. Comprehensive monitoring of PM2.5 has only been in diesel fuel. Carbonaceous particles produced by burning fossil fuels
place since about 2000; previously, the US Environmental Protection are in the respirable range (generally < 1.0 μm in aerodynamic diam-
Agency (EPA) monitored particles with diameters up to PM10; before eter) and can transport carcinogens, such as PAHs, adsorbed to their
1988, it monitored only total suspended particulates (TSP), some of surfaces. Carcinogenicity appears to be modified by the rate of expo-
which are too large to be inhaled into the lung. Progressive declines sure. Experiments in animals demonstrate that even relatively pure
in these indicators of combustion product pollution have been docu- carbon particles can induce lung cancer in rats when administered
mented, following emissions reductions for these pollutants and their at high concentrations, suggesting that particles per se might, under
precursors (US EPA, 2016). some conditions, be carcinogenic (Mauderly, 1997). The relevance
The data discussed in the preceding refer to ambient air pollu- of these findings for humans is controversial, since the tumors in
tion over relatively large geographic areas. However, the exposure of rats were observed at doses that far exceeded the exposures to die-
human populations to carcinogens in ambient air may result from local sel exhaust and overwhelmed lung clearance mechanisms (Winer and
sources, such as small businesses (e.g., automotive body or chrome Busby Jr, 1995).
plating shops), municipal facilities (e.g., waste incinerators), or areas Gaseous pollutants, such as sulphur dioxide (SO2) and oxides of
with high vehicular traffic (Health Effects Institute, 2010). Studies in nitrogen (NOx), are produced by the combustion of fossil fuels and
Harlem show that diesel particle exposures to pedestrians can be sub- then are converted into fine particles in the atmosphere. Epidemiologic
stantial (Kinney et al., 2000) and that high school students are exposed studies have not found consistent evidence of lung cancer risk from
to a variety of toxic air pollutants throughout the day (Kinney et al., occupational exposure to SO2. The IARC has classified strong sulphuric
2002). Levy and colleagues collected spot air samples along sidewalks acid aerosol as a known human carcinogen based on increased lung
in a section of Boston with heavy diesel bus traffic, and detected “hot and laryngeal cancer in heavily exposed occupational groups (IARC,
spots” for exposure to benzo-​(a)-​pyrene, a marker for combustion 2012b).
emissions (IARC, 2013b; Levy et al., 2000).
Diesel
Diesel exhaust is a ubiquitous component of air pollution worldwide.
The IARC recently summarized current estimates of the proportionate
Combustion Products contribution of diesel exhaust to ambient PM2.5. These ranged from
As noted earlier, the combustion of fossil fuels for transportation and approximately 2% in remote rural locations to more than 30% in cities
power generation generates many known or suspected carcinogens. (IARC, 2013b). The IARC has classified diesel exhaust as a human
The following section discusses some of the more significant pollut- carcinogen, based on occupational studies and supporting animal and
ants in terms of exposure prevalence and/​or lung carcinogenicity. The mechanistic data (IARC, 2013b).
IARC’s Outdoor Air Pollution (Volume 109) provides more detailed
information on these pollutants (IARC, 2015). Other
The combustion of fossil fuels contributes known or suspected indi-
Polycyclic Organic Matter vidual carcinogenic chemicals to urban ambient air. In addition to such
Polycyclic organic matter (POM), comprises a large and varied class known human lung carcinogens as arsenic, chromium, and nickel, sev-
of chemical compounds that include polycyclic aromatic hydrocar- eral others are of note, and are discussed briefly here.
bons (PAH) and nitro-​PAHs. The IARC classifies both categories as
carcinogenic and mutagenic (IARC, 2010b, 2013a). These POM have Radionuclides.  Alpha-​emitting radionuclides can also be mea-
common chemical features (one or more benzene rings and a boiling sured in outdoor air. These include isotopes of lead, radium, thorium,
point > 100°C), and can be found in both the solid and gas phases and uranium (Natusch, 1978) that are naturally present in fossil fuels
contaminants of ambient air, depending on their exact chemical struc- and are released by combustion. These contribute a relatively small
ture. Those with > 5 benzene rings tend to be associated with the par- proportion of the radiation dose received by the general population
ticle phase. In addition to the compounds released directly into the (National Council on Radiation Protection and Measurements, 2009).
environment by combustion, others (such as the byproducts of diesel
combustion) are created secondarily by chemical and photochemical 1,3-​butadiene.  1,3-​butadiene is a volatile organic compound used
reactions in the environment (Greenberg, 1987; Natusch, 1978; Winer since the 1930s to produce synthetic rubber. Industrial emissions contrib-
and Busby Jr, 1995; Zielinska et al., 2010). ute to its presence in some urban areas. However, the major source in the
Although the combustion of fossil fuels is a ubiquitous source of United States is automotive exhaust. Emissions are substantially greater
POM in the urban air, it is not the only source of human exposure to for vehicles with more than two axles (Sapkota and Buckley, 2003).
POM, and for some individuals it may not be the main source. Other Levels of 1,3-​butadiene in ambient air generally range from 1 to 10 ppb,
exposure comes from inhaling tobacco or wood smoke, and from diet about 1000-​fold less than occupational exposure levels (IARC, 1992).
(e.g., from the consumption of grilled meat). 1,3-​butadiene has been classified by the IARC as a human car-
Benzo-​(a)-​pyrene is a constituent of POM that has been extensively cinogen (Group 1) based largely on animal experiments, which found
studied and is known to be carcinogenic. It is frequently used as a sur- increased tumor occurrence at multiple sites, including the lung (IARC,
rogate or marker for combustion sources in epidemiologic studies and 2012b). Epidemiologic studies of occupationally exposed populations
294

294 PART III:  THE CAUSES OF CANCER


(rubber workers and butadiene monomer production workers) have EPIDEMIOLOGIC EVIDENCE ON OUTDOOR
consistently observed increases in hematopoietic cancers, but not can- AIR POLLUTION AND CANCER
cers of the respiratory system (IARC, 2012b).

Aldehydes.  Various aldehydes are classified as hazardous air pol- Research Approaches
lutants by the US EPA. Formaldehyde and acetaldehyde are present in Epidemiologists have used three approaches to examine the role of
urban ambient air largely because of the combustion of gasoline and indoor and outdoor air pollution, in the etiology of lung and other can-
diesel fuel (Health Effects Institute, 2010). Exposures to formaldehyde cers: cohort, case-​control, and ecologic designs. Historically, some of
and acetaldehyde in outdoor air tend to be highly correlated. Outdoor the earliest studies of outdoor air pollution were based on ecologi-
concentrations of formaldehyde generally range from 1 to 20 μg/​m3. cal approaches, comparing lung cancer mortality rates in urban and
Levels up to 100 μg/​m3 have been observed in heavy traffic or episodes rural areas (Stocks and Campbell, 1955). Ecological studies have also
of air pollution (IARC, 2012b). Combustion of alternative fuels, such been used in exploratory assessments of the association of indoor
as methanol, and oxygenated fuels containing the additive MBTE also radon with lung cancer. However, the recent and most critical evi-
yields aldehydes (Health Effects Institute, 1993). Formaldehyde is dence comes largely from cohort and case-​control studies, which use
present in indoor as well as outdoor air. more refined approaches for exposure assessment than earlier studies
Formaldehyde has been classified as a human carcinogen by the (IARC, 2015).
IARC and the National Toxicology Program (IARC, 2006, 2012b; Cohort studies of outdoor air pollution have usually defined expo-
National Toxicology Program, 2014), based on evidence from animal sure based on residence location. Thus, the information on age, gen-
experiments and occupationally exposed workers with an excess of der, potential confounders such as cigarette smoking, and exposure
nasal and nasopharyngeal cancer and leukemia. to air pollution were classified at the individual level, whereas the
information on the covariates was ecological. Recent studies have
refined exposure estimates based on statistical models of the location
Point Sources of residence (Krewski et  al., 2009; Raaschou-​Nielsen et  al., 2013).
Industrial point sources also contribute to air pollution. Electrical The case-​control approach provides investigators with an efficient way
power plants fueled by fossil fuels (e.g., coal, oil, and natural gas) emit to estimate the relative risk of lung cancer in relation to air pollution
known and suspected carcinogens (Natusch, 1978). These include met- exposure without requiring access to an existing cohort or the collec-
als and metalloids (chromium, nickel, and arsenic), radionuclides such tion of information on an entire population. In case-​control studies,
as radon and uranium, and POM such as benzo(a)pyrene. Non-​ferrous cases of lung cancer that occur in the population are identified and
metal smelters emit inorganic arsenic and other metals, and sulphur classified according to their exposure to outdoor or indoor air pollu-
dioxide (Pershagen et  al., 1977). Municipal solid waste incinerators tion; a sample of the study population—​the controls—​is selected and
emit heavy metals (e.g., lead and cadmium), PAHs, organic compounds similarly classified according to their exposures. While most recent
(such as dioxins), and acidic gases (World Health Organization, 1988). data come from cohort studies, nested case-​control studies are useful
Unfortunately, these sources of air pollution are often located in or for assessing biomarkers.
near poor, working-​class communities. Reports describing cancer rates In contrast to the cohort and case-​control designs, ecologic, or
around point sources have addressed chemical disasters (e.g., dioxin in aggregate-​level, studies do not collect information on individual sub-
Seveso, Italy) (Bertazzi et al., 1993), radiation from the nuclear reactor jects, but instead compare the incidence or mortality rates of lung or
accident at Three Mile Island, Pennsylvania (Hatch et al., 1990, 1991), other cancers in relation to the levels of air pollution. This approach
and other sources (Pershagen, 1990). Such studies have been useful uses routinely collected data on both lung cancer rates and regional
for examining specific issues but are not essential for addressing the measurements of air pollution. In these studies, surrogate measures
broader issue of air pollution and cancer. Methods have been devel- of exposure have been used based on geologic features, housing char-
oped for studying point sources (Elliott, 2006). acteristics, or measurements of indoor concentrations. Relative risks
can be estimated from ecologic data, but the interpretation of these
estimates is more complicated than for estimates derived from cohort
Fibers and case-​control studies. In most cases, data are not available to take
Asbestos fibers contaminate ambient air in rural and urban environ- into account inter-​individual and between-​region differences in other
ments, and apparently have been present in the ambient environment lung cancer risk factors. Ecological studies of air pollution are no lon-
for at least 10,000 years. The literature on levels of asbestos in outdoor ger informative.
air was reviewed as part of a comprehensive report on the health effects All three designs potentially suffer from a common problem when
of asbestos in public buildings (Health Effects Institute et al., 1991). applied to the study of air pollution and lung cancer: the difficulty of
The median levels of asbestos fibers in rural environments, where characterizing accurately the subjects’ exposure to air pollution and
there are no known natural sources of asbestos, were on the order of mixtures of diverse carcinogens inhaled in indoor and outdoor envi-
0.01–​0.001 ng/​m3; few individual measurements exceeded 1  ng/​m3. ronments. As described earlier, an individual’s exposure to carcino-
In urban environments, where there is both a greater prevalence of gens in outdoor and indoor environments may be complex and may
asbestos-​containing materials and a higher frequency of release of occur in multiple microenvironments; therefore it may be difficult to
fibers from building materials and vehicular brake linings, the median estimate exposure for the purpose of epidemiologic analysis. Some
levels ranged from 0.02 to 10 ng/​m3 and a much larger proportion of of the exposure assessment approaches used are listed in Table 17–​2
individual measurements exceeded 1 ng/​m3 (Health Effects Institute (Di et al., 2016; van Donkelaar et al., 2015); these range from cat-
et al., 1991). egorical indicators, such as self-​report and residence location, to
Epidemiologic studies of occupational groups, such as asbestos contemporary geographic information systems and statistical models
miners and asbestos textile production workers, who were exposed based on monitoring and satellite data, and land use characteristics.
to asbestos at concentrations several orders of magnitude greater The early lung cancer studies often classified exposure to outdoor
than those cited in the preceding, have consistently observed air pollution based on urban or rural residence, a crude indicator of
increased rates of lung cancer. Based on a large body of experi- current or lifetime exposure. In some subsequent studies, approaches
mental and epidemiologic evidence, asbestos is considered to be a based on residence location were refined by more fully capturing
known human carcinogen (IARC, 2012a). Lung cancer occurrence the residential history and incorporating duration of residence into
was not increased in relation to exposure at levels observed in the exposure indexes.
ambient urban environment in a study of chrysotile-​asbestos min- Exposure estimates have been refined by using available monitoring
ing regions (Camus et al., 1998). The risks of different fiber types data in combination with air pollution models that incorporate land-​
remain contentious. use information (e.g., roadways and major point sources and, most
 295

Air Pollution 295


Table 17–​2. Methods of Assessing Exposure efforts of the tobacco industry to discredit the extensive evidence that
secondhand smoke causes lung cancer (Proctor, 2012).
Source of Information Type of Information All exposure estimates in studies of air pollutants are subject to some
misclassification. Usually this misclassification is non-​differential, in
Source strength Emission rate (mass per time), traffic that its effects are similar in subjects with and without lung cancer.
density
This form of misclassification attenuates the association between an
Geographical information Distance of place of residence from the
source
air pollutant and cancer (Jurek et  al., 2005; Szpiro et  al., 2011)  and
Dispersion models Spatiotemporal concentration distributions
can obscure a monotonic relationship with exposure (Birkett, 1992;
from modeling of emission rates, Dosemeci et al., 1990; Jurek et al., 2005). Studies may differ in the
meteorology, air chemistry, geography degree of misclassification, however. This can create the misimpres-
Outdoor–​indoor penetration Modeling from outdoor concentration, sion that their results are in conflict (Lubin et  al., 1995a, 1995b;
building, and ventilation characteristics Rothman and Greenland, 1998). Differential misclassification can
Stationary monitoring Concentration over time and by place spuriously elevate as well as attenuate estimates of association. This
Questionnaires and interviews Source strength, distance from the source, form of bias is of less concern than confounding, however. Most stud-
time activity ies of air pollution attempt to collect data on other lung cancer risk
Personal monitoring Continuous or cumulated concentrations factors, such as cigarette smoking or occupational exposures. Errors
over time in the measurement of potential confounders can introduce bias, even
Human samples Concentrations of biomarkers of exposure if air pollution exposures are estimated with relatively little error; the
in biospecimens result may be either over-​or underestimation of the air pollution effect
Toxicological models Concentration and dose of pollutants (Greenland, 1980).
in target organs modeling from
concentration, breathing rate, metabolism
Monitoring data, land use regression,
Spatial models satellite data
Epidemiologic Studies of Ambient Air Pollution
and Lung Cancer
Source: van Donkelaar et al., 2015; Di et al., 2016. The first studies on lung cancer and air pollution compared lung can-
cer death rates in urban and rural areas. These studies coincided with
the early studies of smoking and lung cancer; consequently, some (but
recently, satellite data) to assign concentrations to the residences where not all) attempted to control for smoking. Most studies found over-
study participants live. For example, the European Study of Cohorts all excesses of lung cancer mortality of about 30%–​40% in the urban
for Air Pollution Effects (ESCAPE) pooled data from 17 cohorts areas. The attribution of these excesses to differences in air quality was
across Europe. Measurements were made in each study site, and then strengthened by evidence of urban/​rural differences in ambient levels
a land-​use regression model was developed to estimate concentrations of carcinogens, such as benzo-​(a)-​pyrene, and by the persistence of
of particulate matter and nitrogen oxides at the home addresses of par- the urban excess after adjustment for cigarette smoking status in some
ticipants (Raaschou-​Nielsen et al., 2013). These approaches have been studies.
continually refined and are now complemented by the use of satellite Later studies have used increasingly sophisticated methods to esti-
data to estimate concentrations of airborne particulate matter and other mate exposure and control for potential confounders. Detailed descrip-
pollutants (Di et al., 2016; van Donkelaar et al., 2015). tions of these studies are provided in the IARC’s Outdoor Air Pollution
There has also been extensive attention given to the development of (Volume 109; IARC, 2015). Groundbreaking findings came from the
approaches to estimating exposures to indoor carcinogens, particularly Harvard Six Cities Study and the American Cancer Society’s (ACS)
secondhand smoke and radon, and, to a lesser extent, asbestos. While Cancer Prevention (CPS) II Study, with initial findings published in
the association of secondhand smoke with lung cancer is not specifi- 1993 and 1995, respectively. Both found increased risk for lung cancer
cally covered in this chapter, the conceptual approaches to assessing in association with higher levels of exposure to PM2.5, a finding repli-
exposure to secondhand smoke are covered because of their relevance cated in many subsequent studies. A meta-​analysis of 18 studies, car-
to the topic of air pollution and cancer. For each of these agents, the ried out in support of the IARC’s Volume 109, found that lung cancer
validity of exposure assessment approaches and the degree of misclas- risk increased by nearly 10% (meta-​relative risk 1.09; 95% CI: 1.04,
sification and resulting bias in risk estimates have figured prominently 1.14) with each 10 μg/​m3 increase of PM2.5 (Figure  17–1) (Hamra
in the interpretation of the epidemiological findings. et al., 2014). There was no strong evidence for variation by geographic
The micro-​environmental model has become the model for expo- region or smoking status.
sure assessment. For radon, individual-​and population-​level expo- In most of the cohort studies, the exposures were assigned based
sures are largely driven by exposures at home, as mentioned earlier. on air pollution data at a single point in time or averaged over
These reflect both the time spent at home and the home concentra- extended periods. Consequently, most studies have not examined
tions. Radon concentrations can be measured in homes, using passive the relationship between air pollution and lung cancer with respect
and relatively inexpensive but accurate measuring devices that record to the timing of exposure. Analyses of extended follow-​ups of both
the average concentration across intervals ranging from days to a the Six Cities and ACS cohorts suggest that more recent exposures
year. A technique to measure longer-​term exposures has been devel- may have the strongest effects on all-​ cause mortality (Krewski
oped that uses levels of surface radioactivity in glass (Steck et  al., et al., 2000).
2002). Issues related to misclassification based on these approaches The current evidence suggests that lung cancer attributable to air
are considered in the section of this chapter on indoor radon. pollution may occur among both smokers and non-​smokers (Hamra
For secondhand smoke, exposure measurement has involved quali- et al., 2014); therefore, residual confounding and effect modification
tative assessments based on questionnaires and quantitative assess- by air pollution must be considered, especially among the smokers.
ments based on biomarkers or air measurements (Apelberg et al., 2013; Most studies report air pollution relative risks adjusted for cigarette
Avila-​Tang et al., 2013a, 2013b). Study participants can describe the smoking, but the adjustment may not have controlled completely for
smoking status of their spouse, the number of smokers at home, and potential confounding. Most cohort studies have information on ciga-
the extent of exposure at work or in other settings in the recent past. rette smoking only at the beginning of follow-​up. The possibility that
Cotinine and other biomarkers of tobacco smoke provide objective changes in tobacco use were correlated with air pollution exposure
evidence of exposure within the past 3–​7 days. Self-​reported informa- cannot be excluded, since patterns of smoking cessation have varied
tion on exposures in childhood and at various periods throughout life geographically in the United States. On the other hand, the associa-
cannot be validated directly, but can provide qualitative information tion of lung cancer with air pollution was largely unaffected in the
about past exposures. The issue of misclassification was central to the Six Cities Study (Dockery et al., 1993), when longitudinal information
296

296 PART III:  THE CAUSES OF CANCER

Study by region RR (95% CI) Weight


North America
McDonell et al. 2000 1.39 (0.79, 2.46) 0.66
Krewski et al. 2009 1.09 (1.05, 1.13) 21.19
Hart et al. 2011 1.18 (0.95, 1.48) 3.77
Lipsett et al. 2011 0.95 (0.70,1.28) 2.22
Lepeule et al. 2012 1.37 (1.07, 1.75) 3.20
Hystad et al. 2013 1.29 (0.95, 1.76) 2.14
Jerrett et al. 2013a 1.12 (0.91, 1.37) —
Puett et al. 2014 1.06 (0.90, 1.24) 6.48
Subtotal (/ 2 = 0.0%, p = 0.490) 1.11 (1.05, 1.16) 39.67
Europe
Beelen et al. 2008 0.81 (0.63, 1.04) 3.11
Carey et al. 2013 1.11 (0.86, 1.43) 3.07
Cesaroni et al. 2013 1.05 (1.01, 1.10) 20.21
Raaschou-Nielsen et al. 2013 1.39 (0.91, 2.13) 1.17
Subtotal (/ 2 = 50.0%, p = 0.112) 1.03 (0.89, 1.20) 27.56
Other
Cao et al. 2011 1.03 (1.00, 1.07) 21.25
Katanoda et al. 2011 1.24 (1.12, 1.37) 11.52
Subtotal (/2 = 91.0%, p = 0.001) 1.13 (0.94, 1.34) 32.77

Overall (/2 = 53.0%, p = 0.010) 1.09 (1.04, 1.14) 100.00

0.5 1 2 3

Figure 17–​1.  Estimates of lung cancer relative risk associated with relative risks. Source: Hamra et al. (2014).

on cigarette smoking was used in a reanalysis. Several case-​control low. Doll and Peto based their estimate on past and current estimates
studies have adjusted for smoking using time-​varying information and of benzo-​(a)-​pyrene in urban air and extrapolation from occupational
still found increased risk for air pollution exposure (e.g., Nyberg et al., studies of PAH-​exposed workers (Doll and Peto, 1981). They esti-
2000). With one exception (Turner et al., 2011), the studies that show mated that less than 1% of future lung cancer in the United States
increased lung cancer risks among self-​reported never-​smokers have would be due to air pollution from the burning of fossil fuels. They did
been small and the effect estimates imprecise. However, Turner and col- note, however, that perhaps 10% of the lung cancers then occurring
leagues (2011) estimated that long-​term residential exposure to PM2.5 in large cities might have been due to air pollution. In 1990, the US
increased lung cancer mortality by 15% to 27% per 10μg/​m3 increase EPA (US EPA, 1990) estimated that 0.2% of all cancer, and probably
in exposure in 188,699 lifelong never-​smokers in the American Cancer less than 1% of lung cancer, could be attributed to air pollution. This
Society’s CPS II cohort (Turner et al., 2011). A subsequent analysis estimate was obtained by applying the unit risks for over 20 known or
of this cohort by Turner et  al. (2014) found that the effects of joint suspected human carcinogens found in outdoor air to estimates of the
exposure to PM2.5 and tobacco smoking were greater than additive, ambient concentrations and numbers of persons potentially exposed.
corroborating the results of several earlier studies (IARC, 2013c). The Global Burden of Disease project provides the most com-
Limitations of approaches to exposure estimation also contribute to prehensive worldwide estimates of the global burden of lung can-
uncertainty in risk estimates. The ACS and Six Cities studies estimated cer due to ambient and household air pollution (Cohen et  al., 2016;
the exposure of each participant based solely on long-​term average con- Forouzanfar et al., 2015; Institute for Health Metrics and Evaluation,
centrations in the metropolitan area of residence. While this approach 2016). Exposure to ambient PM2.5 was estimated to have contributed
may accurately reflect exposure to pollutants that are distributed homog- to 387,000 (UI: 350, 000–​420,000) lung cancer deaths (23.6% of the
enously over large areas for several decades, it does not capture finer total), and a resultant loss of 8.3 million (uncertainty interval [UI]: 7.5–​
spatial and temporal variation; more recent studies in Europe and North 9.1 million) disability-​adjusted life years (DALY) worldwide in 2013.
America are now incorporating spatial statistical methods to estimate The estimated global lung cancer mortality rate attributable to ambient
individual long-​term exposure histories, linking residential histories, PM2.5 increased from 4.64 to 5.41 per 100,000 from 1990 to 2013.
measurements of traffic density on nearby streets, and long-​term records Fifty-​two percent (52%) of global lung cancer mortality due to PM2.5
of specific air pollutants. With this approach, the variations in exposures in 2013 was estimated to have occurred in China.
can be estimated in relation to time and space, perhaps with less mis- When all causes of death were considered, ambient PM2.5 was the
classification (Hoek et al., 2002; Nyberg et al., 2000; Reynolds et al., seventh ranking global mortality risk factor in 2013. Exposure to
2001). Hoek and colleagues observed larger relative risk estimates for ambient PM2.5 contributed to an estimated 2.93  million (UI:  2.78–​
cardiopulmonary mortality among subjects who lived near major roads 3.07 million) premature deaths and a loss of 69.7 million (UI: 65.5–​
than were observed in studies that used larger-​scale urban and regional 75.5 million) DALY in 2013, or 5.3% of total global deaths and 2.9%
measurements (Hoek et  al., 2002). The highest relative risk estimates of global DALY. Sixty percent of this burden occurs in in low-​and
were with exposure 20 or more years before diagnosis (Nyberg et al., middle-​income countries in East and South Asia. The absolute number
2000). By providing exposure estimates at the individual level, these of deaths estimated as attributable to ambient PM2.5 increased by 31%
studies also reduce the possibility of aggregate-​level (ecologic) bias. from 1990 to 2013. Exposure to ozone was estimated to cause an addi-
tional 217,000 (UI: 161,000–​272,000) premature deaths and a loss of
5.1 million (UI: 3.6–​6.6 million) DALY in 2013. Worldwide, ambient
Risk Attribution PM2.5 was the second leading cause of lung cancer mortality in 2013
Estimates of the population attributable risk of lung cancer due to out- after tobacco smoking.
door air pollution have been based on markedly different methods and Household air pollution (HAP) from burning of solid fuels was the
vary by an order of magnitude. The early estimates of burden were eighth ranking global mortality risk factor in 2013 (Institute for Health
 297

Air Pollution 297


Metrics and Evaluation, 2013). Exposure to HAP contributed to an (2002) and the California EPA (1998) based their risk estimates on the
estimated 2.89 million (UI: 2.46–​3.30 million) premature deaths and occupational epidemiology studies available at the time. Both agen-
81.1 million (UI: 70.0–​92.8 million) DALY in 2013, largely in low-​ cies have concluded that the evidence from occupational studies is
and middle-​income countries in East and South Asia. The absolute consistent with a causal relationship with lung cancer. The California
number of deaths attributable to HAP increased by 1.3% from 1990 EPA calculated a unit risk value (1998), which ranged from 1 to 24
to 2013. excess deaths in 10,000 people per 1 μg/​m3 diesel PM lifetime expo-
HAP was the fifth ranking cause of lung cancer in 2013, contributing sure. The US EPA estimated a possible range of lung cancer risk from
to 127,701 (UI: 57,000–​202,900) lung cancer deaths, and 2.9 million environmental exposure to diesel exhaust (0.1 to 10 excess deaths
(UI: 1.3–​4.6 million) DALY worldwide in 2013, with the preponder- in 10,000 people per 1 μg/​m3 diesel PM lifetime exposure). The US
ance again in low-​and middle-​income countries in East and South EPA, however, acknowledged recent improvements in diesel technol-
Asia (Institute for Health Metrics and Evaluation, 2013). Between ogy, and uncertainty as to the relevance of these risk estimates to pres-
1990 and 2013 the estimated global lung cancer mortality rate (per ent engines, a concern that was recently underscored by the result of a
100 persons) attributable to HAP declined by 6.1%. major toxicologic study of the carcinogenicity of emissions from the
latest diesel engines. The ACES study of the Health Effects Institute
reported “dramatic improvements in emissions and the absence of any
Diesel Exhaust and Cancer of the Lung and Bladder significant health effects (especially cancer)” and concluded that “the
The epidemiological evidence regarding the carcinogenicity of diesel overall toxicity of exhaust from modern diesel engines is significantly
exhaust has been reviewed extensively and repeatedly, partly because decreased compared to emissions from traditional-​technology diesel
of the large numbers of people exposed and the implications for engines (p. 5)” (HEI, 2015; US EPA, 2002).
regulation. Most of the evidence comes from studies of occupational
exposure rather than general populations, for whom exposure is dif- Childhood Cancer and Exposure to Traffic-​Related
ficult to estimate accurately (HEI Diesel Epidemiology Panel, 2015; Air Pollution
IARC, 2013b).
Epidemiologic evidence indicates that workers exposed to diesel Motor vehicle exhaust contains a number of known carcinogens,
exhaust for prolonged periods in a variety of occupational settings are including benzene, a known leukemogen (Chapter 38). In 1989 Savitz
at increased risk of lung cancer. Workers in jobs that entailed prolonged and Feingold, using data from an earlier case-​control study of resi-
exposure to diesel exhaust in the trucking and railroad industries and in dential exposure to electric and magnetic fields (Savitz and Feingold,
urban transport were shown to have approximately 20%–​50% higher 1989), reported that vehicular traffic density at the residence of can-
risk of lung cancer mortality in meta-​analyses of studies dating from cer cases was associated with an increased risk of leukemia and brain
the 1950s through the mid-​1990s. This increased risk was independent cancer among children in Denver. As interest in the health effects of
of tobacco smoking (Cohen and Higgins, 1995; Larkin et  al., 2000; air pollution on children has increased, and spatial statistical methods,
Lipsett and Campleman, 1999). such as the geographic information system (GIS), have become more
Occupational exposure to diesel exhaust is also linked to increased widely available, this relationship has been studied by others with
risk of bladder cancer, based on less consistent evidence. The IARC’s inconsistent results (IARC, 2015). The IARC’s Outdoor Air Pollution
Monograph 105 (“Diesel and gasoline engine exhausts and some (Volume 109)  reviewed studies of all childhood cancers combined,
nitroarenes”) noted that case-​control studies generally reported an leukemia and lymphoma, and brain tumors. The evidence was con-
increased risk in occupationally exposed workers but cohort studies sidered to be subject to potential publication bias, but indicative of
did not (Cohen and Higgins, 1995; IARC, 2013b). possible weak association.
Until recently, few studies had linked lung cancer risk to quantitative
estimates of long-​term exposure to diesel exhaust; the resulting uncer-
tainty limited both causal attribution and quantitative risk assessment INDOOR AIR POLLUTION
(Health Effects Institute, 1999). New studies of underground miners and
trucking industry workers have linked long-​term exposure to respirable Carcinogens in indoor air have large potential implications for risk
elemental carbon (EC), a component of diesel particles, to lung cancer because people spend substantial amounts of time indoors. Indoor
in these settings. Underground miners exposed to extremely high levels air pollution may stem from incoming outdoor air or may originate
of diesel particles experienced 2-​to 3-​fold increases in lung cancer mor- indoors from tobacco smoking, building materials, soil gas, household
tality in analyses that controlled for tobacco smoking; trucking industry products, and combustion from heating and cooking (Spengler, 1991).
workers experienced a 40% increase (Garshick et al., 2012; Silverman In more developed countries, two of the most important indoor pollut-
et al., 2012). A recent reanalysis of these studies by the Health Effects ants that influence lung cancer risk in never-​smokers are passive smok-
Institute corroborated the reported results and concluded that the new ing (US Department of Health and Human Services, 1986, 2006) and
studies provided a basis for quantitative risk assessment of lung cancer radon (National Research Council and Committee on Health Risks of
due to diesel exhaust exposure (HEI Diesel Epidemiology Panel, 2015). Exposure to Radon, 1999). Asbestos exposure may pose a risk to build-
IARC’s Monograph 105 provides the most recent and comprehen- ing occupants, but the resulting risk is estimated to be minimal, par-
sive summary of the numerous cohort and case-​control studies of die- ticularly if asbestos-​containing materials are appropriately managed
sel exhaust and cancer. This included the newer and more definitive in place (Health Effects Institute et al., 1991). Other carcinogens are
studies, and largely corroborated the conclusions of the earlier meta-​ present in indoor air, but have received less attention in high-​income
analyses (IARC, 2013b). countries.
Diesel exhaust is now considered by IARC to be “carcinogenic to One major concern in economically developing countries is indoor
humans” (Group 1), based on epidemiologic and toxicologic evidence air pollution from the use of unprocessed solid fuels, notably coal,
(IARC, 2013b). With the finding that diesel exhaust is carcinogenic, for cooking and space heating (Martin et al., 2013). The high risk of
emphasis has shifted to quantitative risk assessment (HEI Diesel lung cancer among non-​smoking Chinese women has been mentioned
Epidemiology Panel, 2015). To this end, Vermeulen et al. pooled the earlier.
results of three cohort studies (two of truckers and one of miners)
(Vermeulen et al., 2014). They estimated that lifetime environmental
Asbestos
exposure to 0.8 μg/​m3 EC would cause 21 excess lung cancer deaths
per 10,000 persons. They further estimated that “[b]‌ased on broad Asbestos, a well-​established occupational carcinogen, includes several
assumptions regarding past occupational and environmental expo- forms of fibrous, naturally occurring silicate minerals that have been
sures . . . approximately 6% of annual [US and UK] lung cancer deaths widely used in products found in homes and public and commercial
may be due to DEE exposure (p. 175).” This estimate is broadly con- buildings (Health Effects Institute et al., 1991). The epidemiologic evi-
sistent with earlier estimates by US regulatory agencies. Both the EPA dence on asbestos and lung cancer dates to the 1950s, although clinical
298

298 PART III:  THE CAUSES OF CANCER


case series had previously led to the hypothesis that asbestos causes and Committee on the Biological Effects of Ionizing Radiation,
lung cancer (Lynch and Smith, 1939; Wedler, 1944). In a retrospec- 1988; Proctor, 2012). While lesser risks have been observed for more
tive cohort study published in 1955, Doll observed that asbestos textile recent worker cohorts, the newer epidemiological studies show clear
workers at a factory in the United Kingdom had a 10-​fold elevation evidence of a continued increase in cancer risk (Hunter et al., 2013;
in lung cancer risk and that the risk was most heavily concentrated Tirmarche et al., 2012). Cigarette smoking and radon decay products
during the time frame before regulations were implemented to limit synergistically influence lung cancer risk in a manner that is supra-​
asbestos dust in factories (Doll, 1955). A 7-​fold excess of lung cancer additive but sub-​multiplicative (Lubin et al., 1995a; National Research
was subsequently observed among insulation workers in the United Council and Committee on Health Risks of Exposure to Radon, 1999).
States (Selikoff et al., 1964, 1979). The peak incidence occurred 30–​ Radon is of broad societal interest because it is a ubiquitous indoor
35 years after the initial exposure to asbestos (Selikoff et al., 1980). air pollutant, entering buildings in soil gas. On average, indoor expo-
Subsequently, numerous studies of specific worker groups and of the sures to radon for the general population are much less than those
general population have repeatedly documented the carcinogenicity of received by occupational groups such as uranium miners. For exam-
asbestos (IARC, 2012a). The risk of lung cancer has been noted to ple, even the lowest historical radon concentration in a uranium mine
increase with increased exposure to asbestos and to be associated with is still roughly an order of magnitude higher than in the average home
the principal commercial forms of asbestos. Asbestos and cigarette (Lubin et  al., 1995a). However, measurements of indoor radon con-
smoking are both independent causes of lung cancer; in combination, centrations in homes show a log-​normal distribution with all homes
they act synergistically to increase risk in a manner compatible with a having some radon detectable at an average around 1 picocurie per
multiplicative effect (Hammond et al., 1979; IARC, 2004). liter (37 becquerels per m3), but with a highly skewed tail of the dis-
With the recognition in the 1970s and 1980s that asbestos-​containing tribution that corresponds to exposures in uranium mines. As a basis
materials were prevalent in indoor environments and a potential source for developing control strategies, risk assessments have estimated the
of airborne fibers, concern was raised as to risks to the general popula- magnitude of the problem and the extent to which exposures must be
tion for both mesothelioma and lung cancer. In the United States, pro- reduced to protect public health. Because of the prevalence of low and
grams were initiated to either remove or manage asbestos-​containing average exposures in the population, these are of greatest concern.
materials to limit releases of fibers. Because the concentrations were After the problem of indoor radon was first widely recognized in the
generally very low, the risks could not be directly addressed using early 1980s, case-​control studies were carried out to estimate the risks
epidemiological approaches. A  risk assessment was carried out by directly. These studies complemented the information from cohort
the Health Effects Institute that used all available measurements of studies of underground miners, which generally had much higher
indoor asbestos fiber concentrations and a concentration–​risk relation- levels of exposure. Most studies incorporated measurements of radon
ship from studies of workers (Health Effects Institute et  al., 1991). concentrations in the homes of lung cancer cases and controls. The
The concentration information considered by the Health Effects interpretation of the findings was limited by exposure misclassifica-
Institute showed that exposures in indoor environments were gener- tion, a particularly difficult problem in estimating lifetime exposure
ally quite low and were not elevated in comparison with levels in urban (Lubin et al., 1995a). Measurement of implanted polonium-​210 in the
outdoor air. surface of glass (e.g., window glass or glass covering a picture) has
The US EPA estimates that an individual who continuously breathed been proposed as an indicator of long-​term concentration and has been
air containing asbestos at an average of 0.000004 fibers/​cm3 over his or applied in several epidemiological studies (Brownson and Alavanja,
her entire lifetime would theoretically have no more than a 1 in 1 mil- 2000; Field et  al., 2002; Steck et  al., 2002). Nonetheless, the risk
lion increased risk of developing cancer from this exposure. Similarly, estimates from individual case-​control studies have been relatively
the EPA estimates that breathing air containing 0.00004 fibers/​cm3 imprecise. A  meta-​analysis of the findings through the mid-​1990s
would result in not greater than a 1 in 100,000 increased chance of did show evidence for a significant and positive exposure–​response
developing cancer, and air containing 0.0004 fibers/​cm3 would result relationship that was consistent with the extrapolated risks in the
in not greater than a 1 in 10,000 increased chance of developing cancer miner studies (National Research Council and Committee on Health
(US EPA, 1999). This risk was considered sufficiently low as to not Risks of Exposure to Radon, 1999). Subsequent pooled analyses of
warrant removal of asbestos from buildings. The EPA now promotes the European and North American case-​control studies provide risk
in-​place management of asbestos-​containing materials. estimates that are also consistent with the risk models based on the
underground miner studies (Darby et al., 2005; Krewski et al., 2005).
For the purpose of risk assessment and policy formulation, risk
Radon models have been developed based on the findings in the underground
Radon is an inert gas produced naturally from radium in the decay miners (US EPA, 2003). There are inherent uncertainties in extending
series of uranium. It decays into a series of relatively short-​lived such models to the general population, particularly the extrapolation of
particle progeny; two of the short-​lived decay products emit alpha findings from highly exposed uranium miners to the much lower expo-
particles that can, by virtue of their high energy and mass, damage sures indoors. The studies of miners lacked information on women and
DNA in respiratory epithelial cells. Laboratory experiments involving children and had little control over other differences between mines
exposure of cells to single alpha particles show that even one “hit” and homes, such as exposure to diesel exhaust. Effect modification by
from an alpha particle causes permanent change in a cell. Even though smoking added further uncertainty; the evidence from the miners indi-
cancer is a multistage phenomenon, this finding implies that radon cated synergy between smoking and radon, but the combined effect
exposure may result in increased risk at any level (National Research was sub-​multiplicative (National Research Council and Committee on
Council and Committee on Health Risks of Exposure to Radon, 1999). Health Risks of Exposure to Radon, 1999).
Research on radon is now quite consistent regarding carcinogenicity. From the policy perspective, the most critical uncertainty is whether
The various lines of evidence (dosimetric, in vitro, and epidemiologi- radon causes lung cancer at all exposures—​that is, is there a thresh-
cal) consistently support a linear no-​threshold risk relationship. old exposure below which radon does not cause cancer? Biological
Epidemiologic studies of underground miners of uranium and evidence supports the assumption that a single hit to a cell by an
other ores have long established that exposure to radon and its decay alpha particle causes permanent cellular change, an assumption lead-
products causes lung cancer (Lubin et al., 1995a; National Research ing to a non-​threshold dose–​response relationship (Hei et  al., 1994,
Council and Committee on Health Risks of Exposure to Radon, 1999; 1997; National Research Council and Committee on Health Risks
National Research Council and Committee on the Biological Effects of Exposure to Radon, 1999). Additionally, the case-​control study
of Ionizing Radiation, 1988). In fact, radon exposure was probably findings to date provide evidence for increased risk down to levels
the first occupational respiratory carcinogen to be identified, based of exposure not substantially greater than that associated with typical
on the extremely high rates of lung cancer documented in the under- indoor concentrations. When combined in a meta-​analysis, studies of
ground metal miners of Schneeberg and Joachimsthal and the high the general population show a significant association between indoor
levels of radon measured in the mines (National Research Council radon and lung cancer in the general population that is quantitatively
 29

Air Pollution 299


compatible with risk models derived from the underground miners. In 1992 the US EPA (1992a) published its risk assessment of SHS
This coherence lends support to using extrapolation of the miner data as a carcinogen. The agency’s evaluation drew on the toxicologic evi-
with a linear, non-​threshold model to estimate the risk of indoor radon. dence on SHS and the extensive literature on active smoking. A meta-​
Consequently, the risk model developed by the National Research analysis of the 31 studies published to that time was central in the
Council’s Biological Effects of Ionizing Radiation (BEIR) VI decision to classify SHS as a class A carcinogen—​namely, a known
Committee (National Research Council and Committee on Health human carcinogen. Overall, the analysis found a significantly increased
Risks of Exposure to Radon, 1999) and subsequently applied by the risk of lung cancer in never-​smoking women married to smoking men;
US EPA incorporates a linear model without a threshold. The assump- for the studies conducted in the United States, the estimated relative
tions made by the Environmental Protection Agency (US EPA, 1992b) risk was 1.19 (90% CI: 1.04, 1.35).
and the Biological Effects of Ionizing Radiation (BEIR) IV and VI Subsequent conclusions reaffirmed those of the earlier reports.
Committees of the National Research Council (National Research A 2004 report by the IARC (2004) reiterated the conclusion with the
Council and Committee on Health Risks of Exposure to Radon, 1999; following statement: “evidence is sufficient to conclude that involun-
National Research Council and Committee on the Biological Effects tary smoking is a cause of lung cancer in never-​smokers. The magni-
of Ionizing Radiation, 1988)  led to estimates that approximately tudes of the observed risks are reasonably consistent with predictions
21,000 lung cancer deaths per year in the United States are attribut- based on studies of active smoking in many populations.” The 2006
able to radon (with an uncertainty range of 8000 to 45,000), an esti- report of the US Surgeon General (p.  15), which focused on SHS,
mate that makes indoor radon the second-​leading cause of lung cancer concluded: “1. The evidence is sufficient to infer a causal relationship
(US EPA, 2003). The story of research on radiation and lung cancer is between secondhand smoke exposure and lung cancer among lifetime
relatively complete and illustrates how epidemiological evidence can nonsmokers. This conclusion extends to all secondhand smoke expo-
inform policy decisions. sure, regardless of location and 2. The pooled evidence indicates a 20 to
30 percent increase in the risk of lung cancer from secondhand smoke
exposure associated with living with a smoker” (US Department of
Secondhand Smoke Health and Human Services, 2006). The burden of lung cancer attrib-
In 1981 reports were published from Japan (Hirayama, 1981)  and utable to passive smoking has been estimated as 31,620 deaths glob-
from Greece (Trichopoulos et  al., 1981)  that indicated increased ally (Global Burden of Disease Study 2013 Collaborators, 2015).
lung cancer risk in non-​smoking women married to cigarette smok-
ers. This once controversial association has subsequently been
examined in numerous investigations in the United States and other EXPOSURE IN LOW-​AND MIDDLE-​INCOME
countries. A causal relationship between secondhand smoke (SHS) COUNTRIES
and lung cancer is biologically plausibile, given the many carcino-
gens in sidestream smoke and the lack of a documented threshold Current knowledge about ambient air pollution and lung cancer is
dose for respiratory carcinogens in active smokers (IARC, 2004; US based largely on the experience of populations of Western industri-
Department of Health and Human Services, 2006, 2010). Moreover, alized nations. The levels of exposures in low-​and middle-​income
genotoxic activity has been demonstrated for many components of countries (LMICs) in Asia, Africa, and the Middle-​East rival and often
SHS (Claxton et  al., 1989; Hillerdal, 1996; IARC, 2004; Lofroth, exceed those commonly observed in high-​income countries.
1989; Weiss, 1989). Experimental exposure of non-​ smokers to Some LMICs face a double air pollution challenge: the first from
SHS has validated the excretion of 4-​ (methylnitrosamino)-​
1-​(3-​ increasing levels of ambient air pollution due to urbanization and
pyridyl)-​1 butanol (NNAL), a tobacco-​specific carcinogen, in urine industrialization, and the second from a continuing reliance on solid
as a valid biomarker of exposure to tobacco smoke (Anderson et al., fuels for domestic energy needs. The latter is most common among
2001; Hecht et al., 1993). Non-​smokers exposed to SHS also have the rural poor, who still lack access to clean energy (Balakrishnan
increased concentrations of adducts of tobacco-​related carcinogens et al., 2014). Although the proportion of the global population that
(Crawford et al., 1994; Maclure et al., 1989). uses solid fuels such as coal or biomass for household cooking or
Through the 1980s and 1990s, the tobacco industry sought to heating has declined over the past three decades, the populations
maintain controversy and uncertainty about the carcinogenicity of have grown and the number of people who rely on solid fuels has
SHS (Proctor, 2012). In fact, the US courts found the tobacco indus- remained largely unchanged at 2.8 billion. These exposures occur
try guilty of fraud and deception about the risks of SHS exposure mostly in sub-​Saharan Africa and South and East Asia. Forty-​one
(Kessler, 2006). percent (41%) of global households and 60% percent of households
There are long-​standing authoritative conclusions that SHS causes in sub-​Saharan Africa and South Asia were estimated to have burned
lung cancer. By 1986, the evidence had mounted, and three consensus solid fuels in 2010 (Bonjour et  al., 2013). Indoor burning of solid
reports published in the same year concluded that SHS was a cause fuels has also been associated with increased rates of cancer of
of lung cancer. The IARC (1986) concluded that “passive smoking the upper airways. Exposure to household air pollution is highest
gives rise to some risk of cancer.” In its monograph on tobacco smok- in women due to their role in food preparation (Forouzanfar et al.,
ing, the agency supported this conclusion on the basis of the char- 2015). Exposures to levels of fine particles (PM2.5) and other health-​
acteristics of sidestream and mainstream smoke, the absorption of damaging air pollutants, such as carbon monoxide, are many-​fold
tobacco smoke materials during involuntary smoking, and the nature higher in households where solid fuels are burned in unventilated liv-
of dose–​response relationships for carcinogenesis. In the same year, ing areas. The emissions from the combustion of solid fuels include
the National Research Council (1986) and the US Surgeon General known human carcinogens such as polycyclic aromatic hydrocar-
(1986) also concluded that involuntary smoking increases the inci- bons, formaldehyde, and benzene (IARC, 2010a). Household com-
dence of lung cancer in non-​smokers. In reaching this conclusion, the bustion of solid fuels also contributes to ambient air pollution in
National Research Council (1986) cited the biological plausibility of settings where solid fuels are widely used, and was estimated to be
the association between exposure to SHS and lung cancer and the sup- responsible for 12% of global population-​weighted PM2.5, 37% and
porting epidemiological evidence. Based on a pooled analysis of the 26% in sub-​Saharan Africa and South Asia, respectively, in 2010
epidemiological data adjusted for bias, the report concluded that the (Chafe et al., 2014).
best estimate for the excess risk of lung cancer in non-​smokers mar- Indoor burning of coal causes lung cancer in humans and is associ-
ried to smokers was 25%. The 1986 report of the Surgeon General ated with a 2-​fold increase in lung cancer risk (Hosgood et al., 2011;
(1986) characterized involuntary smoking as a cause of lung cancer IARC, 2010a). The risk from indoor combustion of low-​grade “smoky”
in non-​smokers. This conclusion was based on the extensive informa- coal approximates the risks from active smoking. Indoor burning of
tion already available about the carcinogenicity of active smoking, the biomass, such as wood and dung, is also associated with increased
qualitative chemical similarities between SHS and mainstream smoke, lung cancer (Bruce et  al., 2015; Hosgood et  al., 2010). In addition,
and the epidemiological data on involuntary smoking. cooking-​oil emissions from high-​temperature frying may also cause
30

300 PART III:  THE CAUSES OF CANCER


lung cancer (IARC, 2010a). In some parts of China, household coal References
burning has resulted in markedly elevated lung cancer rates among Anderson KE, Carmella SG, Ye M, et  al. 2001. Metabolites of a tobacco-​
non-​smoking women, and the venting of emissions and introduction of specific lung carcinogen in nonsmoking women exposed to environmen-
cleaner burning stoves have been demonstrated to reduce lung cancer tal tobacco smoke. J Natl Cancer Inst, 93(5), 378–​381. PMID: 11238699.
rates in exposed women (Hosgood et al., 2011; Lan et al., 2002). The Apelberg BJ, Hepp LM, Avila-​Tang E, et  al. 2013. Environmental monitor-
use of cleaner fuels, such as compressed natural gas, is associated with ing of secondhand smoke exposure. Tob Control, 22(3), 147–​ 155.
reduced rates of lung cancer and other adverse health outcomes rela- PMCID: PMC3639351.
tive to the use of coal and biomass. Armstrong B, Hutchinson E, Unwin J, and Fletcher T. 2004. Lung can-
There has been little research on ambient air pollution and lung cer risk after exposure to polycyclic aromatic hydrocarbons:  a review
and meta-​analysis. Environ Health Perspect, 112(9), 970–​ 978.
cancer among urban residents of the burgeoning cities in LMICs, PMCID: PMC1247189.
although mounting levels of urban air pollution from local station- Autrup H, Daneshvar B, Dragsted LO, et al. 1999. Biomarkers for exposure to
ary and, increasingly, mobile sources are recognized as an important ambient air pollution: comparison of carcinogen-​DNA adduct levels with
environmental problem (World Health Organization [WHO], 2002). other exposure markers and markers for oxidative stress. Environ Health
In the cities of the poorest developing countries, WHO’s Global Perspect, 107(3), 233–​238.
Environmental Monitoring System observed average ambient con- Avila-​Tang E, Al-​Delaimy WK, Ashley DL et  al. 2013a. Assessing second-
centrations of total suspended particles of 300 μg/​m3; the exposure hand smoke using biological markers. Tob Control, 22(3), 164–​ 171.
in places where coal is used for fuel, such as poor communities in PMCID: PMC3639350.
South Africa, may exceed 1 mg/​m3 (WHO, 1990). Estimated levels Avila-​Tang E, Elf JL, Cummings KM, et  al. 2013b. Assessing secondhand
smoke exposure with reported measures. Tob Control, 22(3), 156–​163.
of PM10 in cities in developing countries also exceed those com- PMCID: PMC3639349.
monly encountered in Europe and North America (Pandey et  al., Balakrishnan K, Cohen A, and Smith KR. 2014. Addressing the burden of
2006). One might predict that the high levels of ambient air pol- disease attributable to air pollution in India: the need to integrate across
lution found in cities in the developing world would be associated household and ambient air pollution exposures. Environ Health Perspect,
with greater excess lung cancer occurrence than has been observed 122(1), A6–​A7. PMCID: PMC3888581.
in Western industrialized settings. Although there are currently few Bertazzi A, Pesatori AC, Consonni D, et al. 1993. Cancer incidence in a popu-
relevant investigations, a case-​control study in Shenyang, China, lation accidentally exposed to 2,3,7,8-​ tetrachlorodibenzo-​ para-​
dioxin.
observed a 200% increase in lung cancer risk after adjustment for Epidemiology, 4(5), 398–​406. PMID: 8399687.
age, education, and smoking, among residents in “smoky” areas of Birkett NJ. 1992. Effect of nondifferential misclassification on estimates of
odds ratios with multiple levels of exposure. Am J Epidemiol, 136(3),
the city and a 50% increase among those in “somewhat or slightly 356–​362. PMID: 1415154.
smoky” areas (Xu et al., 1989) Bonjour S, Adair-​Rohani H, Wolf J, et al. 2013. Solid fuel use for household
As a greater proportion of the world’s population moves from rural cooking: country and regional estimates for 1980-​2010. Environ Health
communities to the rapidly expanding and highly polluted cities of Perspect, 121(7), 784–​790. PMCID: PMC3701999.
Asia and the Southern Hemisphere, there is a clear need for research Brauer M, Amann M, Burnett RT, et al. 2012. Exposure assessment for estima-
on outdoor air pollution in the developing world. Epidemiologic tion of the global burden of disease attributable to outdoor air pollution.
studies in these settings will present even greater challenges than Environ Sci Technol, 46(2), 652–​660. PMCID: PMC4043337.
those in the industrialized West. In addition to the generic problem Brauer M, Freedman G, Frostad J, et al. 2016. Ambient air pollution exposure
of estimating long-​term exposure to air pollution discussed earlier, estimation for the Global Burden of Disease 2013. Environ Sci Technol,
50(1), 79–​88. PMID: 26595236.
the ambient air pollution mixture in urban centers in the developing Brownson RC, and Alavanja MC. 2000. Previous lung disease and lung cancer
countries is changing, due in part to the increase in vehicular traffic. risk among women (United States). Cancer Causes Control, 11(9), 853–​
Careful planning will be required to characterize these changes over 858. PMID: 11075875.
time, including the choice and development of methods to measure Bruce N, Dherani M, Liu R, et al. 2015. Does household use of biomass fuel
indicator pollutants for different sources. In addition, the current dra- cause lung cancer? A systematic review and evaluation of the evidence for
matic increases in cigarette smoking in the developing world (Peto the GBD 2010 study. Thorax, 70(5), 433–​441. PMID: 25758120.
and Lopez, 2003), and the thoroughly predictable consequences, California Environmental Protection Agency. 1998. Health Risk Assessment
will complicate the interpretation of studies of air pollution and lung for Diesel Exhaust: Proposed Identification of Diesel Exhaust as a Toxic
cancer. Air Contaminant. Sacramento:  Office of Environmental Health Hazard
Assessment, California Environmental Protection Agency.
Camus M, Siemiatycki J, and Meek B. 1998. Nonoccupational exposure to
chrysotile asbestos and the risk of lung cancer. N Engl J Med, 338(22),
FUTURE DIRECTIONS 1565–​1571. PMID: 9603793.
Chafe ZA, Brauer M, Klimont Z, et al. 2014. Household cooking with solid
There is now abundant evidence that specific air pollutants increase fuels contributes to ambient PM2.5 air pollution and the burden of disease.
lung cancer risk in smokers and non-​smokers. Carcinogens can be Environ Health Perspect, 122(12), 1314–​1320. PMCID: PMC4256045.
measured in indoor and outdoor environments; toxicologic data indi- Claxton LD, Morin RS, Hughes TJ, and Lewtas J. 1989. A genotoxic assess-
cate the potential for human carcinogenicity. Epidemiologic research ment of environmental tobacco smoke using bacterial bioassays. Mutat
shows evidence of effects of indoor and outdoor air pollution on lung Res, 222(2), 81–​99. PMID: 2645519.
cancer risk, albeit weak for some agents. Various indoor air pollutants Cohen AJ, Brauer M, and Burnett R. 2016. The Global Burden of Disease
attributable to ambient air pollution: estimates of current burden and 23-​
are confirmed causes of cancer, as is outdoor air pollution generally. year trends from the GBD 2013 Study.
Research is still needed on air pollution and lung cancer to guide Cohen AJ, and Higgins MWP. 1995. Health Effects of Diesel
policies for protection of public health. Critical remaining questions Exhaust: Epidemiology Diesel Exhaust: A Critical Analysis of Emissions,
relate to the magnitude of the risk at the lower pollution levels now Exposure, and Health Effects. A Special Report of the Institute’s Working
present in many high-​income countries and to the specific pollutants Group (pp. 251–​292). Cambridge, MA: Health Effects Institute.
leading to increased lung cancer risk. Direct epidemiologic obser- Crawford FG, Mayer J, Santella RM, et al. 1994. Biomarkers of environmental
vation of exposed populations can provide the best information for tobacco smoke in preschool children and their mothers. J Natl Cancer
evaluating the magnitude of outdoor air pollution–​related excess lung Inst, 86(18), 1398–​1402. PMID: 8072033.
cancer. In general, large-​scale epidemiologic studies of air pollution Darby S, Hill D, Auvinen A, et  al. 2005. Radon in homes and risk of lung
cancer: collaborative analysis of individual data from 13 European case-​
and lung cancer are needed if we are to obtain sufficiently informative control studies. BMJ, 330(7485), 223. PMCID: PMC546066.
and precise data on risks. Large numbers of cases will be necessary to DeMarini DM. 2013. Genotoxicity biomarkers associated with exposure to
measure the effects of air pollution among women and ethnic minori- traffic and near-​road atmospheres:  a review. Mutagenesis, 28(5), 485–​
ties and to measure the joint effects of air pollution and other factors, 505. PMID: 23945473.
such as occupation and smoking. Evidence is particularly needed for DeMarini DM, Shelton ML, Abu-​Shakra A, Szakmary A, and Levine JG. 1998.
low-​and middle-​income countries. Spectra of spontaneous frameshift mutations at the hisD3052 allele of
 301

Air Pollution 301


Salmonella typhimurium in four DNA repair backgrounds. Genetics, Review and Synthesis of Current Knowledge (p. 227). Cambridge,
149(1), 17–​36. PMCID: PMC1460133. MA: Health Effects Institute.
Di Q, Kloog I, Koutrakis P, et  al. 2016. Assessing PM exposures with high Hecht SS, Carmella SG, Murphy SE, et al. 1993. A tobacco-​specific lung car-
spatio-​temporal resolution across the continental United States. Environ cinogen in the urine of men exposed to cigarette smoke. N Engl J Med,
Sci Technol, 50(9), 4712–​4721. PMID: 27023334. 329(21), 1543–​1546.
Dockery DW, Pope CA, 3rd, Xu X, et al. 1993. An association between air pol- HEI Diesel Epidemiology Panel. 2015. Diesel emissions and lung cancer: an
lution and mortality in six U.S. cities. N Engl J Med, 329(24), 1753–​1759. evaluation of recent epidemiological evidence for quantitative risk assess-
PMID: 8179653. ment. Res Rep Health Eff Inst, Spec No. 19(19), 1–​149. PMID: 26846065.
Doll R. 1955. Mortality from lung cancer in asbestos workers. Br J Ind Med, Hei TK, Piao CQ, Willey JC, Thomas S, and Hall EJ. 1994. Malignant trans-
12(2), 81–​86. PMCID: PMC1037613. formation of human bronchial epithelial cells by radon-​simulated alpha-​
Doll R, and Hill AB. 1952. A study of the aetiology of carcinoma of the lung. particles. Carcinogenesis, 15(3), 431–​437. PMCID: PMC20515.
Br Med J, 2(4797), 1271–​1286. PMCID: PMC2022425. Hei TK, Wu LJ, Liu SX et al. 1997. Mutagenic effects of a single and an exact
Doll R, and Peto R. 1981. The causes of cancer: quantitative estimates of avoid- number of alpha particles in mammalian cells. Proc Natl Acad Sci U S A,
able risks of cancer in the United States today. J Natl Cancer Inst, 66(6), 94(8), 3765–​3770. PMCID: PMC20515.
1191–​1308. PMID: 7017215. Hillerdal G. 1996. Long-​term survival of patients with lung cancer from a
Doll R, Vessey MP, Beasley RW, et  al. 1972. Mortality of gasworkers defined geographical area before and after radiological screening. Lung
-​final report of a prospective study. Br J Ind Med, 29(4), 394–​406. Cancer, 15(1), 21–​30. PMID: 8865120.
PMCID: PMC1009455. Hirayama T. 1981. Non-​smoking wives of heavy smokers have a higher risk
Dosemeci M, Wacholder S, and Lubin JH. 1990. Does nondifferential misclas- of lung cancer: a study from Japan. Br Med J (Clin Res Ed), 282(6259),
sification of exposure always bias a true effect toward the null value? Am 183–​185. PMCID: PMC1503989.
J Epidemiol, 132(4), 746–​748. PMID: 2403115. Hoek G, Brunekreef B, Goldbohm S, Fischer P, and van den Brandt PA. 2002.
Elliott P. 2006. Point sources of air pollution: investigation of possible health Association between mortality and indicators of traffic-​related air pollu-
effects using small area methods. In: Ayres J, Maynard R, and Richards tion in the Netherlands: a cohort study. Lancet, 360(9341), 1203–​1209.
R (Eds.), Air Pollution and Health (Vol. 3, pp. 49–​67). London: Imperial PMCID: PMC12401246.
College Press. Hoffman FL. 1929. Cancer of the lungs. Am Rev Tuberc, 19, 392–​406.
Field RW, Smith BJ, Steck DJ, and Lynch CF. 2002. Residential radon Hoffman FL. 1931. Cancer and smoking habits. Annals Surg, 93(1), 50–​67.
exposure and lung cancer:  variation in risk estimates using alternative PMCID: PMC1398760.
exposure scenarios. J Expo Anal Environ Epidemiol, 12(3), 197–​203. Hosgood HD, 3rd, Boffetta P, Greenland S, et  al. 2010. In-​home coal and
PMID: 12032816. wood use and lung cancer risk:  a pooled analysis of the International
Forouzanfar MH, Alexander L, Anderson HR, et al. 2015. Global, regional, and Lung Cancer Consortium. Environ Health Perspect, 118(12), 1743–​1747.
national comparative risk assessment of 79 behavioural, environmental PMCID: PMC3002194.
and occupational, and metabolic risks or clusters of risks in 188 countries, Hosgood HD, 3rd, Wei H, Sapkota A, et al. 2011. Household coal use and lung
1990–​2013: a systematic analysis for the Global Burden of Disease Study cancer: systematic review and meta-​analysis of case-​control studies, with
2013. Lancet, 386(10010), 2287–​2323. PMCID: PMC4685753. an emphasis on geographic variation. Int J Epidemiol, 40(3), 719–​728.
Garshick E, Laden F, Hart JE, et al. 2012. Lung cancer and elemental carbon PMCID: PMC3147068.
exposure in trucking industry workers. Environ Health Perspect, 120(9), Hunter N, Muirhead CR, Tomasek L, et  al. 2013. Joint analysis of three
1301–​1306. PMCID: PMC3440130. European nested case-​ control studies of lung cancer among radon
Georgiadis P, Topinka J, Stoikidou M, et al. 2001. Biomarkers of genotoxicity exposed miners:  exposure restricted to below 300 WLM. Health Phys,
of air pollution (the AULIS project): bulky DNA adducts in subjects with 104(3), 282–​292. PMID: 23361424.
moderate to low exposures to airborne polycyclic aromatic hydrocarbons International Agency for Research on Cancer (IARC). 1986. Tobacco smok-
and their relationship to environmental tobacco smoke and other param- ing. IARC Monogr Eval Carcinog Risk Chem Hum, 38, 35–​ 394.
eters. Carcinogenesis, 22(9), 1447–​1457. PMCID: PMC11532867. PMID: 3460963.
Global Burden of Disease Study 2013 Collaborators. 2015. Global, regional, IARC. 1992. IARC Monographs on the Evaluation of Carcinogenic Risks
and national incidence, prevalence, and years lived with disability for 301 to Humans:  Occupational Exposures to Mists and Vapours from
acute and chronic diseases and injuries in 188 countries, 1990–​2013: a Strong Inorganic Acids; And Other Industrial Chemicals. Lyon,
systematic analysis for the Global Burden of Disease Study 2013. Lancet, France: International Agency for Research on Cancer.
386(9995), 743–​800. PMCID: PMC4561509. IARC. 2004. Tobacco smoke and involuntary smoking. IARC Monogr Eval
Gordon SB, Bruce NG, Grigg J, et al. 2014. Respiratory risks from household Carcinog Risks Hum, 83, 1–​1438. PMCID: PMC4781536.
air pollution in low and middle income countries. Lancet Respir Med, IARC. 2006. Formaldehyde, 2-​ butoxyethanol and 1-​ tert-​
butoxypropan-​
2(10), 823–​860. PMCID: PMC5068561. 2-​ol. IARC Monogr Eval Carcinog Risks Hum, 88, 1–​ 478.
Greenberg A. 1987. Analyses of polycyclic aromatic hydrocarbons. In:  Lioy PMCID: PMC4781641.
PJ, and Daisey JM (Eds.), Toxic Air Pollution: A Comprehensive Study of IARC. 2010a. Household use of solid fuels and high-​temperature frying. IARC
Non-​Criteria Pollutants (pp. 93–​118). Boca Raton, FL: Lewis Publishers. Monogr Eval Carcinog Risks Hum, 95, 1–​430. PMCID: PMC5046077.
Greenland S. 1980. The effect of misclassification in the presence of covariates. IARC. 2010b. Some non-​heterocyclic polycyclic aromatic hydrocarbons and
Am J Epidemiol, 112(4), 564–​569. PMID: 7424903. some related exposures. IARC Monogr Eval Carcinog Risks Hum, 92,
Hammond EC, Selikoff IJ, and Seidman H. 1979. Asbestos exposure, cigarette 1–​853. PMCID: PMC4781319.
smoking and death rates. Ann N Y Acad Sci, 330, 473–​490. PMID: 294198. IARC. 2012a. Arsenic, metals, fibres, and dusts. IARC Monogr Eval Carcinog
Hamra GB, Guha N, Cohen A et al. 2014. Outdoor particulate matter exposure Risks Hum, 100(Pt C), 11–​465. PMCID: PMC4781271.
and lung cancer: a systematic review and meta-​analysis. Environ Health IARC. 2012b. Chemical agents and related occupations. IARC Monogr Eval
Perspect, 122(9), 906–​911. PMCID: PMC4154221. Carcinog Risks Hum, 100(Pt F), 9–​562. PMCID: PMC4781612.
Hatch MC, Beyea J, Nieves JW, and Susser M. 1990. Cancer near the Three IARC. 2013a. Bitumens and bitumen emmissions, and some N-​and S-​
Mile Island nuclear plant: radiation emissions. Am J Epidemiol, 132(3), heterocyclic polycyclic aromatic hydrocarbons. IARC Monogr Eval
397–​412; discussion 413–​397. PMID: 2389745. Carcinog Risks Hum, 103, 9–​303. PMCID: PMC4781167.
Hatch MC, Wallenstein S, Beyea J, Nieves JW, and Susser M. 1991. Cancer IARC. 2013b. Diesel and gasoline engine exhausts and some nitroarenes. IARC
rates after the Three Mile Island nuclear accident and proximity of Monogr Eval Carcinog Risks Hum, 105, 9–​699. PMCID: 4781216.
residence to the plant. Am J Public Health, 81(6), 719–​724. PMCID: IARC. 2013c. IARC Scientific Publication No. 161. Air Pollution and Cancer.
PMC1405170. Press W Ed. Geneva: World Health Organization.
Health Effects Institute. 1993. Research Priorities for Mobile Air Toxics [HEI IARC. 2015. Outdoor Air Pollution. Vol. 109. Lyon, France:  International
Communication Number 2]. Cambridge, MA: Health Effects Institute. Agency for Research on Cancer.
Health Effects Institute. 1999. Diesel Emissions and Lung Cancer: Epidemiology Institute for Health Metrics and Evaluation. 2013. GBD Compare. Available from:
and Quantitative Risk Assessment. Cambridge, MA:  Health Effects https://​vizhub.healthdata.org/​gbd-​compare/​. Accessed August 22, 2016.
Institute. Institute for Health Metrics and Evaluation. 2016. Global Burden of Disease
Health Effects Institute. 2010. Special Report 17:  Traffic-​ Related Air (GBD). Available from: http://​www.healthdata.org/​gbd. Accessed August
Pollutions: A Critical Review of the Literature on Emissions, Exposure, 22, 2016.
and Health Effects. Boston: Health Effects Institute. Jurek AM, Greenland S, Maldonado G, and Church TR. 2005. Proper interpre-
Health Effects Institute, Asbestos Research Committee, and Literature Review tation of non-​differential misclassification effects: expectations vs obser-
Panel. 1991. Asbestos in Public and Commercial Buildings: A Literature vations. Int J Epidemiol, 34(3), 680–​687. PMID: 15802377.
302

302 PART III:  THE CAUSES OF CANCER


Katsouyanni K. 2013. Geographical distribution of air pollutants. In: Straif K, National Research Council, and Committee on Passive Smoking. 1986.
Cohen A, and Samet J (Eds.), IARC Scientific Publication No. 161, Air Environmental Tobacco Smoke:  Measuring Exposures and Assessing
Pollution and Cancer (p. 15). Lyon, France: WHO Press. Health Effects. Washington, DC: National Academy Press.
Kessler G. 2006. Final Opinion Civil Action No. 99–​2496 (GK). Available National Research Council, and Committee on the Biological Effects of
from:  http://​www.justice.gov/​sites/​default/​files/​civil/​legacy/​2014/​09/​11/​ Ionizing Radiation. 1988. Health Risks of Radon and Other Internally
amended opinion_​0.pdf Accessed on June 8, 2015. Deposited Alpha-​Emitter: BEIR IV. Washington, D.C.
Kinney PL, Aggarwal M, Northridge ME, Janssen NA, and Shepard P. 2000. National Research Council, and Panel on Dosimetric Assumptions Affecting the
Airborne concentrations of PM(2.5) and diesel exhaust particles on Application of Radon Risk Estimates. 1991. Comparative Dosimetry of
Harlem sidewalks:  a community-​ based pilot study. Environ Health Radon in Mines and Homes: Companion to BEIR IV Report. Washington,
Perspect, 108(3), 213–​218. PMCID: PMC1637978. DC: National Academy Press.
Kinney PL, Chillrud SN, Ramstrom S, Ross J, and Spengler JD. 2002. National Toxicology Program. 2014. Formaldehyde CAS No. 50-​99-​0 13th
Exposures to multiple air toxics in New  York City. Environ Health Report on Carcinogens (13th ed., p.  11). Triangle Park, NC:  US
Perspect, 110 Suppl 4, 539–​546. PMCID: PMC1241202. Department of Health and Human Services, Public Health Service.
Krewski D, Burnett RT, Goldberg MS, et al. 2000. Reanalysis of the Harvard Natusch DF. 1978. Potentially carcinogenic species emitted to the atmosphere
Six Cities Study and the American Cancer Society Study of Particulate Air by fossil-​fueled power plants. Environ Health Perspect, 22, 79–​ 90.
Pollution and Mortality. Investigators’ Reports Parts I and II. Cambridge, PMCID: PMC1637149.
MA: Health Effects Institute. Nyberg F, Gustavsson P, Jarup L, et  al. 2000. Urban air pollution and lung
Krewski D, Jerrett M, Burnett RT, et  al. 2009. Extended follow-​up and spa- cancer in Stockholm. Epidemiology, 11(5), 487–​495. PMID: 10955399.
tial analysis of the American Cancer Society study linking particulate air Overholt RH, and Rumel WR. 1940. Clincial studies of primary carcinoma of
pollution and mortality. Res Rep Health Eff Inst 140, 5–​114; discussion the lung. An analysis of seventy-​five cases, twenty-​one of which were
115–​136. PMID: 19627030. treated by pneumonectomy or lobectomy. JAMA, 114 (9), 735–​742.
Krewski D, Lubin JH, Zielinski JM, et al. 2005. Residential radon and risk of Pandey KD, Wheeler D, Ostro B, et  al. 2006. Ambient Particulate Matter
lung cancer: a combined analysis of 7 North American case-​control stud- Concentrations in Residential and Pollution Hotspot Areas of World
ies. Epidemiology, 16(2), 137–​145. PMID: 15703527. Cities: New Estimates Based on the Global Model of Ambient Particulates
Krzyzanowski M, Apte JS, Bonjour SP, et al. 2014. Air pollution in the mega-​ (GMAPS). Washington, DC:  The World Bank Development Economics
cities. Curr Environ Health Rep, 1, 185–​191. Research Group and the Environment Department Working Paper, The
Kyrtopoulos SA, Georgiadis P, Autrup H, et al. 2001. Biomarkers of genotoxic- World Bank.
ity of urban air pollution. Overview and descriptive data from a molecular Perera FP, Hemminki K, Gryzbowska E, et  al. 1992. Molecular and genetic
epidemiology study on populations exposed to moderate-​to-​low levels of damage in humans from environmental pollution in Poland. Nature,
polycyclic aromatic hydrocarbons: the AULIS project. Mutat Res, 496(1–​ 360(6401), 256–​258. PMID: 1436106.
2), 207–​228. PMCID: PMC11551497. Pershagen G. 1990. Air pollution and cancer. In:  Vainio H, Sorsa M, and
Lan Q, Chapman RS, Schreinemachers DM, Tian L, and He X. 2002. Household McMichael AJ (Eds.), Complex Mixtures and Cancer Risk (pp. 240–​251).
stove improvement and risk of lung cancer in Xuanwei, China. J Natl Lyon, France: International Agency for Research on Cancer.
Cancer Inst, 94(11), 826–​835. PMID: 12048270. Pershagen G, Elinder CG, and Bolander AM. 1977. Mortality in a region sur-
Larkin EK, Smith TJ, Stayner L, et al. 2000. Diesel exhaust exposure and lung rounding an arsenic emitting plant. Environ Health Perspect, 19, 133–​
cancer:  adjustment for the effect of smoking in a retrospective cohort 137. PMCID: PMC1637390.
study. Am J Ind Med, 38(4), 399–​409. PMID: 10982980. Peto R, and Lopez AD. 2003. Smoking Data:  Clinical Trial Service Unit.
Levy JI, Houseman EA, Ryan L, Richardson D, and Spengler JD. 2000. Particle Available from: http://​www.ctsu.ox.ac.uk/​. Accessed January 10, 2016.
concentrations in urban microenvironments. Environ Health Perspect, Proctor R. 2012. Golden Holocaust: Origins of the Cigarette Catastrophe and
108(11), 1051–​1057. PMCID: PMC11102296. the Case for Abolition. Berkeley: University of California Press.
Lipsett M, and Campleman S. 1999. Occupational exposure to diesel exhaust Raaschou-​Nielsen O, Andersen ZJ, Beelen R, et  al. 2013. Air pollution and
and lung cancer: a meta-​analysis. Am J Public Health, 89(7), 1009–​1017. lung cancer incidence in 17 European cohorts: prospective analyses from
PMCID: PMC1508841. the European Study of Cohorts for Air Pollution Effects (ESCAPE).
Lofroth G. 1989. Environmental tobacco smoke:  overview of chemical Lancet Oncol, 14(9), 813–​822. PMID: 23849838.
composition and genotoxic components. Mutat Res, 222(2), 73–​ 80. Redmond CK. 1983. Cancer mortality among coke oven workers. Environ
PMID: 2645518. Health Perspect, 52, 67–​73. PMCID: PMC1569361.
Lubin JH, Boice JD, Jr, Edling C, et al. 1995a. Lung cancer in radon-​exposed Reynolds P, Elkin E, Scalf R, Von Behren J, and Neutra RR. 2001. A case-​
miners and estimation of risk from indoor exposure. J Natl Cancer Inst, control pilot study of traffic exposures and early childhood leukemia
87(11), 817–​827. PMID: 7791231. using a geographic information system. Bioelectromagnetics, Suppl 5,
Lubin JH, Boice JD, Jr, and Samet JM. 1995b. Errors in exposure assessment, S58–​S68. PMID: 11170118.
statistical power and the interpretation of residential radon studies. Radiat Rothman KJ, and Greenland S. 1998. Modern Epidemiology.
Res, 144(3), 329–​341. PMID: 7494877. Philadelphia: Lippincott-​Raven.
Lynch KM, and Smith WA. 1939. Pulmonary asbestosis. V. A report of bron- Sapkota A, and Buckley TJ. 2003. The mobile source effect on curbside 1,3-​
chial carcinoma and epithelial metaplasia. Am J Cancer, 36, 567–​573. butadiene, benzene, and particle-​bound polycyclic aromatic hydrocar-
Macklin MT, and Macklin CC. 1940. Does chronic irritation cause primary bons assessed at a tollbooth. J Air Waste Manag Assoc, 53(6), 740–​748.
carcinoma of the human lung? Arch Pathol, 30, 924–​955. PMID: 12828334.
Maclure M, Katz RB, Bryant MS, Skipper PL, and Tannenbaum SR. 1989. Savitz DA, and Feingold L. 1989. Association of childhood cancer with res-
Elevated blood levels of carcinogens in passive smokers. Am J Public idential traffic density. Scand J Work Environ Health, 15(5), 360–​363.
Health, 79(10), 1381–​1384. PMCID: PMC1350179. PMID: 2477895.
Martin WJ, 2nd, Glass RI, Araj H, et al. 2013. Household air pollution in low-​ Selikoff IJ, Churg J, and Hammond EC. 1964. Asbestos exposure and neopla-
and middle-​income countries: health risks and research priorities. PLoS sia. JAMA, 188, 22–​26. PMID: 14107207.
Med, 10(6), e1001455. PMCID: PMC3672215. Selikoff IJ, Hammond EC, and Seidman H. 1979. Mortality experience of
Mauderly JL. 1997. Relevance of particle-​induced rat lung tumors for assess- insulation workers in the United States and Canada, 1943–​1976. Ann N Y
ing lung carcinogenic hazard and human lung cancer risk. Environ Health Acad Sci, 330, 91–​116. PMID: 294225.
Perspect, 105 Suppl 5, 1337–​1346. PMCID: PMC1470153. Selikoff IJ, Hammond EC, and Seidman H. 1980. Latency of asbestos disease
National Council on Radiation Protection and Measurements. 2009. Ionizing among insulation workers in the United States and Canada. Cancer,
Radiation Exposure of the Population of the United States. Report No. 46(12), 2736–​2740. PMID: 7448712.
160. 1st ed. Washington, DC: National Council on Radiation. Silverman DT, Samanic CM, Lubin JH, et  al. 2012. The Diesel Exhaust in
National Research Council, and Committee on Health Risks of Exposure to Miners study:  a nested case-​control study of lung cancer and diesel
Radon. 1999. Health Effects of Exposure to Radon: BEIR VI. Washington, exhaust. J Natl Cancer Inst, 104(11), 855–​868. PMCID: PMC3369553.
DC: National Academy Press. Spengler JD. 1991. Sources and concentrations of indoor air pollution. In: Samet
National Research Council, Committee on Human and Environmental JM, and Spengler JD (Eds.), Indoor Air Pollution: A Health Perspective
Exposure Science in the 21st Century, Board on Environmental Studies (pp. 33–​67). Baltimore, MD: Johns Hopkins University Press.
and Toxicology, and Division on Earth and Life Studies. 2012. Exposure Steck DJ, Alavanja MC, Field RW, et al. 2002. 210Po implanted in glass sur-
Science in the 21st Century:  A  Vision and a Strategy. Washington, faces by long term exposure to indoor radon. Health Phys, 83(2), 261–​
DC: National Academies Press. 271. PMCID: PMC12132714.
 30

Air Pollution 303


Stocks P, and Campbell JM. 1955. Lung cancer death rates among non-​smokers US EPA. 1999. Integrated Risk Information System (IRIS) on Asbestos.
and pipe and cigarette smokers:  an evaluation in relation to air pollu- Washington, DC: National Center for Environmental Assessment, Office
tion by benzpyrene and other substances. Br Med J, 2(4945), 923–​929. of Research and Development.
PMCID: PMC1981086. US EPA. 2003. EPA Assessment of Risks from Radon in Homes: Air and Radiation
Szpiro AA, Paciorek CJ, and Sheppard L. 2011. Does more accurate exposure (6608J) (p. 98). Washington, DC: Office of Radiation and Indoor Air.
prediction necessarily improve health effect estimates? Epidemiology, US EPA. 2002. Health Assessment Document for Diesel Engine Exhaust (Final
22(5), 680–​685. PMCID: PMC3195520. 2002). Washington, DC:  US Environmental Protection Agency, Office of
Tirmarche M, Harrison J, Laurier D, et al. 2012. Risk of lung cancer from radon Research and Development, National Center for Environmental Assessment.
exposure: contribution of recently published studies of uranium miners. US EPA. 1990. Cancer Risk from Outdoor Exposure to Air Toxics (Vol. 1, Final
Ann ICRP, 41(3–​4), 368–​377. PMID: 23089036. Report, p.  162). Research Triangle Park, NC:  US Government Printing
Tomatis L. 1990. Air Pollution and Human Cancer. Berlin, Germany: Office.
Springer-​Verlag. US EPA. 2016. Our Nation’s Air: Status and Trends Through 2015. Available
Trichopoulos D, Kalandidi A, Sparros L, and MacMahon B. 1981. Lung cancer from: https://​gispub.epa.gov/​air/​trendsreport/​2016/​. Accessed September
and passive smoking. Int J Cancer, 27(1), 1–​4. PMID: 7251227. 9, 2016.
Turner MC, Cohen A, Jerrett M, et  al. 2014. Interactions between cigarette van Donkelaar A, Martin RV, Brauer M, and Boys BL. 2015. Use of satellite
smoking and fine particulate matter in the Risk of Lung Cancer Mortality observations for long-​term exposure assessment of global concentrations
in Cancer Prevention Study II. Am J Epidemiol, 180(12), 1145–​1149. of fine particulate matter. Environ Health Perspect, 123(2), 135–​143.
PMID: 25395026. PMCID: PMC4314252.
Turner MC, Krewski D, Pope CA, 3rd, et  al. 2011. Long-​term ambient fine Vermeulen R, Silverman DT, Garshick E, et al. 2014. Exposure-​response esti-
particulate matter air pollution and lung cancer in a large cohort of mates for diesel engine exhaust and lung cancer mortality based on data
never-​smokers. Am J Respir Crit Care Med, 184(12), 1374–​ 1381. from three occupational cohorts. Environ Health Perspect, 122(2), 172–​
PMID: 21980033. 177. PMCID: PMC3915263.
US Department of Health and Human Services. 1986. The Health Consequences Wedler HW. 1944. Asbestosis and pulmonary carcinoma (Asbestose und lun-
of Involuntary Smoking: A Report of the Surgeon General. Washington, genkrebs). Bull Hygiene, 19(5), 362–​362.
DC:  US Department of Health and Human Services, Public Health Weiss SJ. 1989. Tissue destruction by neutrophils. N Engl J Med, 320(6), 365–​
Service, Office on Smoking and Health. 376. PMID: 2536474.
US Department of Health and Human Services. 2006. The Health Consequences Winer AM, and Busby WF Jr. 1995. Atmospheric Transport and Transformation
of Involuntary Exposure to Tobacco Smoke:  A  Report of the Surgeon of Diesel Emissions (pp. 83–​105). Cambridge, MA: The Health Effects
General. Atlanta, GA:  US Department of Health and Human Services, Institute.
Centers for Disease Control and Prevention, Coordinating Center for World Health Organization (WHO). 1988. Emissions of Heavy Metal and
Health Promotion, National Center for Chronic Disease Prevention and PAH Compounds from Municipal Solid Waste Incinerators:  Control
Health Promotion, Office on Smoking and Health. Technology and Health Effects:  Report on a WHO Meeting, Florence.
US Department of Health and Human Services. 2010. How Tobacco Smoke Copenhagen: WHO.
Cuases Disease:  The Biology and Behavioral Basis for Smoking-​ WHO. 1990. Global Estimates for Health Situation Assessment and Projections.
Atrributable Disease: A Report of the Surgeon General. Atlanta, GA: US Geneva: WHO.
Department of Health and Human Services, Centers for Disease Control WHO. 2002. The World Health Report:  Reducing Risks, Promoting Healthy
and Prevention, National Center for Chronic Disease Prevention and Life. Geneva: World Health Organization.
Health Promotion, Office on Smoking and Health. Xu ZY, Blot WJ, Xiao HP, et al. 1989. Smoking, air pollution, and the high rates
US Environmental Protection Agency (EPA). 1992a. Respiratory Health Effects of lung cancer in Shenyang, China. J Natl Cancer Inst, 81(23), 1800–​
of Passive Smoking:  Lung Cancer and Other Disorders. Washington, 1806. PMID: 2555531.
DC: US Government Printing Office. Zielinska B, Samy S, McDonald JD, and Seagrave J. 2010. Atmospheric
US EPA. 1992b. Technical Support Document for the 1992 Citizen’s Guide to Transformation of Diesel Emissions. HEI Research Report 147 (p. 86).
Radon. Washington, DC: US Government Printing Office. Boston, MA: Health Effects Institute.
304
 305

18 Water Contaminants

KENNETH P. CANTOR, CRAIG M. STEINMAUS, MARY H. WARD,


AND LAURA E. BEANE FREEMAN

OVERVIEW severe symptoms and even death may follow shortly after ingestion of
even a small inoculum of bacteria. The short incubation period virtu-
Humans have long recognized the hazards of microbial contamination ally eliminates the opportunity for those exposed to cholera to move
of drinking water. Only since the 1960s, however, have epidemiologic or change their water supply before the onset of disease. Snow’s map
studies systematically examined whether naturally occurring and/​or depicting the cross-​sectional clustering of cases along the distribution
man-​made pollutants in drinking water affect cancer risk. Ironically, pipes of the contaminated water system strongly implicated drink-
some of the measures taken to reduce microbial hazards have increased ing water as the vector for disease transmission, and argued against
exposure to other contaminants. This chapter will begin by discuss- ‘miasma’ as a plausible alternative explanation.
ing three waterborne exposures that affect large numbers of people In contrast to the study of infectious agents, epidemiologic studies
and have been studied most extensively: inorganic arsenic, disinfec- of cancer typically confront exposures for which the induction period
tion byproducts, and nitrate. Of these, only arsenic and its compounds may last 20, 50, or more years, depending on the stage(s) of carcino-
are currently designated as carcinogenic to humans. We then discuss genesis that are affected. Retrospective studies (case control or his-
the evidence concerning two emerging issues: the carcinogenicity of torical cohort) and ecological analyses are usually more feasible than
toxins from cyanobacteria, an ancient and ubiquitous family of prokar- prospective studies for estimating risk, although these studies require
yotic organisms formerly known as blue-​green algae, now affected by that researchers reconstruct historical exposures that occurred many
climate change, and the methods of studying cancer in local communi- decades in the past. Mobility creates logistical problems in identifying
ties where the water supply has been contaminated by industrial chem- and tracking the exposed population (Meliker et  al., 2007; Nuckols
icals. Methodologic challenges complicate studies of these issues. The et al., 2011), and in characterizing exposure at different periods of life.
long induction period following exposure, especially when the expo- Studies often rely on a single, contemporaneous questionnaire seek-
sure affects early stages of carcinogenesis, requires that researchers ing information about historical levels of water consumption that are
reconstruct historical levels that existed many decades in the past. The assumed to accurately represent lifetime patterns. Exposed populations
exposed populations must be sufficiently large, stable, and well defined may not be sufficiently large and/​or well defined that an epidemiologic
that epidemiologic studies can detect associations with specific cancer study can detect associated levels of risk in site-​specific cancers.
sites. The presence of multiple exposures complicates efforts to eval- Despite these obstacles, there has been substantial progress in study-
uate individual exposures in relation to a ­priori hypotheses. Despite ing the potential carcinogenicity of waterborne pollutants and in devel-
these obstacles, there have been important advances in studying the oping opportunities to integrate genetic and metabolomic analyses into
potential carcinogenicity of waterborne pollutants, as well as new this area of research (Antonelli et al., 2014; Cantor et al., 2010). The
opportunities to integrate genetic and metabolomic approaches into present chapter will discuss developments in the field since the third
future studies. edition of this text (Cantor et al., 2006).

INTRODUCTION INORGANIC ARSENIC

Water is essential to life. However, drinking water may be contami- Exposure


nated by biological, chemical, and other pollutants that lead to ill
health and death. Access to clean water has been a prerequisite for Human exposure to inorganic arsenic occurs primarily through water
successful human settlements since ancient times. The engineering ingestion, although exposure can also occur via air, food, and tobacco
feats of the Romans more than 2000 years ago (Gagarin, 2010) and the smoking (IARC, 2012a). The most common source of high exposure is
Incas prior to the Spanish Conquest (Wright et al., 1997) attest to the from drinking water contaminated with arsenic from naturally occur-
central importance of uncontaminated water in human cultures. ring geologic sources. Industrial exposure occurs through the inhala-
People have long taken steps to reduce microbial contamination of tion of arsenic-​containing particulates, especially during the mining
drinking water. Efforts to assess the relationship between waterborne and smelting of copper, tin, and lead; some agricultural workers con-
contaminants and cancer are relatively recent, however. Not until the tinue to be exposed to arsenical pesticides. Medications containing
1960s did epidemiologic studies begin to evaluate systematically arsenic were a staple of the pharmacopeia until the 1940s; patients
whether naturally occurring and/​or man-​made chemicals and other were treated for a wide range of ailments, particularly dermatologic
substances in drinking water affect cancer risk (International Agency conditions (Cantor et al., 2006; Cuzick et al., 1992). With the excep-
for Research on Cancer [IARC], 2012a). tion of arsenic trioxide, used to treat promyelocytic leukemia (Coombs
Epidemiologic studies of water pollution and cancer confront et  al., 2015), medicinal arsenic is no longer a significant source of
special challenges that are not generally problematic when studying exposure.
microbial contaminants in drinking water. These can be illustrated In natural waters, inorganic arsenic occurs primarily as pentavalent
by contrasting the seminal study of waterborne cholera in mid-​nine- arsenate [As(V)] or trivalent arsenite [As(III)] (Cantor et  al., 2006).
teenth-​century London by Dr. John Snow (1855) with an evaluation Arsenic can also occur in organic forms such as arsenobetaine or other
of cancer risk in relation to chemical contamination of water. In the arsenosugars. These occur more commonly in food (e.g., seafood) and
case of cholera, the incubation period is measured in hours, or at most are considerably less toxic than inorganic arsenic. Tens of millions of
a few days. The consequences of infection are rapid and unambiguous; people worldwide are exposed to naturally occurring inorganic arsenic

305
306

306 PART III:  THE CAUSES OF CANCER


in drinking water, including an estimated 50  million in Bangladesh, shows the temporal trend of arsenic concentration in drinking water in
30 million in India (mostly in West Bengal), 15 million in China, and relation to the rate ratio (RR) estimates for lung and bladder cancer
millions more in Europe and South and Central America (Ravenscroft, mortality. The RR estimates for both cancer sites started to increase
2007). Exposures in Bangladesh and West Bengal are primarily the about 10 years after the large initial increase in arsenic exposure and
result of widespread introduction of tube wells, beginning in the 1970s, continued to rise until the late 1980s. Subsequent analyses have shown
to reduce exposure to microbial pathogens in surface water. By 1993, that cancer mortality rates in this area remained high up to 2000, the
widespread arsenic contamination of tube wells was confirmed, lead- last year assessed, approximately 40  years after the high exposures
ing to enormous public health concerns (Smith et al., 2000). A 1998–​ began and approximately 30 years after the maximum exposure was
1999 survey of 3534 tube wells throughout Bangladesh found that ended (Smith et al., 2012).
25%–​27% of wells had arsenic concentrations > 50 µg/​l, five times
the current World Health Organization recommended maximum of Biologic Metrics of Arsenic Exposure
10 µg/​l, and 9% had concentrations > 200 µg/​l. It was estimated that Humans excrete arsenic primarily through the urine, and urinary levels
57 million people in Bangladesh were exposed to water with arsenic of inorganic arsenic and its metabolites are commonly used to estimate
concentrations > 10 µg/​l, and 35 million were exposed to concentra- recent internal absorption (IARC, 2012a; NRC, 1999). The biological
tions > 50 µg/​l (British Geological Survey, Government of the People’s half-​life of arsenic in urine following ingestion is approximately 39–​
Republic of Bangladesh, 2001). Mitigation efforts have reduced the 59 hours (Buchet et al., 1981); thus, urinary concentrations are most
proportion of wells with arsenic concentrations > 50 µg/​l from 25% valuable for assessing recent exposures or exposures among people
to 13% (Flanagan et al., 2012). Despite this, an estimated 22 million whose intake levels have not changed much over time. Arsenic can
people in Bangladesh still rely on the highly contaminated wells. also be measured in blood, hair, and nails (Orloff et  al., 2009). The
In most other areas with naturally contaminated water, arsenic half-​life of arsenic in blood is fairly short, so blood levels are only
water concentrations are generally lower than those in Bangladesh. useful for detecting recent exposures. Arsenic levels in the hair and
According to the Environmental Protection Agency (EPA) in the nails have a longer half-​life, so these can be used to assess exposures
United States, an estimated 12% of all public water systems (5252 over several months or more. Hair and nails can be contaminated by
of 43,443) have arsenic concentrations between 5–​10 µg/​l (US EPA, external exposures to arsenic, so specialized cleaning procedures are
2000a). Several million people in the United States are thought to be employed when using hair and nails to assess internally absorbed arse-
exposed to higher levels from drinking water drawn from unregulated nic (Orloff et al., 2009).
private wells. Arsenic concentrations > 50 µg/​l have been reported in
many US states, but levels > 200 µg/​l are rare (Chappells et al., 2014). Statistical Modeling of Exposure
A case-​control study of skin, bladder, and kidney cancer conducted Historical exposure records are rarely available, especially in regions
in parts of Hungary, Romania, and Slovakia reported that 25% of the where private wells are common. Several studies have used statisti-
population was exposed to an average concentration of arsenic in cal modeling to estimate arsenic exposures from sources that lack
drinking water of > 10 µg/​l, and 8% were exposed to > 50 µg/​l. Levels direct measurements. For example, in a bladder cancer case-​control
above 200 µg/​l were reported in only a few wells (Hough et al., 2010). study in New England, investigators measured arsenic concentra-
Another source of arsenic exposure is food, which, on average, tions in water samples from 2611 current and 448 past residences of
accounts for about 8–​10 μg of ingested arsenic per day (Dabeka et al., study subjects (Nuckols et al., 2011). In this region, almost half the
1993; Tao and Bolger, 1999). Arsenic can be taken up by plants grown population obtains drinking water from private wells. The available
in soil with elevated arsenic levels. Some arsenic in soil can result from measurements covered residences that accounted for only 27% of
previous use of arsenic-​based pesticides, or irrigation with arsenic-​ the total lifetime person-​years of participants in the study. In order
contaminated water (Rahman and Hasegawa, 2011). Exposure to arse- to estimate exposures during the remaining residential history of
nic in rice has received considerable attention. Rice actively extracts the subjects, the researchers developed statistical regression mod-
arsenic from soil, and detectable levels have been seen in many rice els that combined empirical measurements from the sampled wells
products. In 2012, the US Food and Drug Administration (FDA) found with geological data for two major strata and geochemical data for
arsenic in almost all of the nearly 200 brands of rice, rice cakes, rice public water supplies drawn from different sources. Model results
baby foods, and rice cereals it tested (US FDA, 2012). A 2011 study of were applied to estimate the arsenic level at each historic residence
229 pregnant women in the United States found that women who ate rice where direct data were not available. In a validation survey done
had significantly higher urinary arsenic levels than those who did not. as part of this investigation, measures of sensitivity, specificity, and
Eating 0.56 cups of cooked rice per day was associated with an arsenic overall agreement varied. For example, using a dichotomous cutoff
intake equivalent to that from drinking one liter per day of water with of 5 µg/​l, overall agreement between measured and predicted level
an arsenic concentration of 10 µg/​l (the current standard for drinking of drinking water arsenic from bedrock wells (from one of the mod-
water specified by the United States and WHO; Gilbert-​Diamond et al., els) varied between 57% and 70%, depending on the buffer radius
2011). Rice also absorbs arsenic from cooking water as well as during that was used for the estimate. Sensitivity varied from 58% to 78%
growth. Thus, intake from rice is likely higher in areas where arsenic-​ and specificity from 52% to 73%. While these results are adequate
contaminated water is used for cooking (Rahman and Hasegawa, 2011). for some purposes, the level of exposure misclassification in these
estimates is high enough to compromise findings in epidemiologic
analyses, highlighting the difficulties that can occur using modeling
Exposure Measurement in Epidemiologic Studies approaches to estimate past exposure. Other modeling or statistical
approaches have been used, including geological averages, kriging,
Residential Exposure and using the arsenic concentration in nearest neighboring wells.
Epidemiologic studies of arsenic and cancer often use residential his- Reported correlation coefficients between the arsenic concentrations
tories in combination with historical records of water concentration to predicted by these methods and concentrations in known wells in
estimate exposure in regions with highly contaminated drinking water. validation surveys have ranged from fair to good (i.e., correlation
For example, Marshall et al. investigated cancer mortality following a coefficients between 0.4 and 0.8) (Dauphine et  al., 2013; James
13-​year period of very high arsenic exposure in a desert area (Region et al., 2014; Meliker et al., 2008).
II) in northern Chile (Marshall et al., 2007). The period of high expo-
sure began in 1958 when two rivers with arsenic water concentrations
near 860 µg/​l were piped to the city for drinking, and ended in 1971 Cancer Outcomes
when an arsenic treatment plant was installed. The epidemiologists
measured cancer mortality rates for the years before, during, and after Experimental Studies in Animals
this high exposure period, and compared these with death rates in a Until recently, arsenic has been one of the few human carcinogens
socio-​demographically similar region in Chile (Region V). Figure 18–​1 without an animal model. More recent experiments using arsenical
 307

Water Contaminants 307


Lung Cancer Mortality Lung Cancer Mortality
Men aged 30 and over Women aged 30 and over
4 600 4 600

3 450 3 450

Arsenic µg/L
Arsenic µg/L

Rate Ratio
Rate Ratio

2 300 2 300

1 150 1 150

0 0 0 0
1950 1960 1970 1980 1990 2000 1950 1960 1970 1980 1990 2000
Years Years

Bladder Cancer Mortality Bladder Cancer Mortality


Men aged 30 and over Women aged 30 and over
12 600 12 600

10 10
450 450
8 8

Arsenic µg/L
Arsenic µg/L
Rate Ratio

Rate Ratio
6 300 6 300

4 4
150 150
2 2

0 0 0 0
1950 1960 1970 1980 1990 2000 1950 1960 1970 1980 1990 2000
Years Years

Figure 18–​1.  Lung and bladder cancer mortality rate ratios comparing Region II with Region V, Chile, for men and women aged 30 and above, separately,
as estimated by Poisson regression with smoothing. The circles are the mortality rate ratios plotted at the midpoint of each successive 3-​year period. The
shading represents the 95% confidence intervals of these rate ratios. Histograms (gray lines) of the population-​weighted average arsenic water concen-
trations for Region II, from 1950 to 1994 in 5-​year increments, are also presented (vertical axes at right). The dramatic rise and decline in arsenic water
concentrations were in the years 1958 and 1970, respectively. They were incorrectly drawn in the original published graphs shown here.

methylation metabolites and early-​life exposures have shown increases surface water. Ecological studies in the 1960s through 1990s reported
in cancer in rodents (IARC, 2012a; Tokar et al., 2010a). elevated mortality from cancers of the skin, bladder, lung, liver, and
There are important interspecies differences in the metabolism kidney (Cantor et al., 2006). More recently, cohort and case-​control
of arsenic between laboratory animal species and humans. These studies that collected individual data on arsenic drinking water lev-
may account for differences in the toxicity and dose–​response rela- els and potential confounders have confirmed these associations. For
tionships in different species (Vahter, 1999b). For example, mice example, in a prospective cohort study of 8086 residents of north-
are very effective at metabolizing arsenic. In rats, methylated inor- western Taiwan, relative risks of urothelial cancer associated with
ganic arsenic accumulates in red blood cells; storage inside red residential arsenic water concentrations of < 10, 10–​49.9, 50–​99.9,
blood cells may limit toxicity to other organs. This does not occur 100–​299.9, and ≥ 300 µg/​l at the time of study recruitment were 1.00
in humans. Consequently, long-​term feeding studies of laboratory (reference), 1.85 (95% confidence interval [CI]: 0.45, 7.61), 2.19 (CI:
animals may not reliably predict the carcinogenic effects in humans. 0.43, 11.1), 5.50 (CI: 1.39, 21.8), and 10.8 (CI: 2.90, 40.3), respec-
The designation of inorganic arsenic as a human carcinogen by the tively (Chen et al., 2010b). Relative risks for lung cancer were 1.00
IARC is based on data from humans rather than animals (IARC, (reference), 1.10 (CI: 0.74, 1.63), 0.99 (CI: 0.59, 1.68), 1.54 (CI:
2012a), and refers primarily to cancers of the skin, bladder, and 0.97, 2.46), and 2.25 (CI: 1.43, 3.55) (Chen et al., 2010a). In northern
lung. Associations have been observed with cancers of the kidney, Chile, arsenic concentrations in drinking water have been measured
liver, and prostate, but these are not currently regarded as definitive longitudinally in almost all sources for decades, allowing researchers
(IARC, 2012a). to estimate lifetime exposures (Ferreccio et al., 2000). A 2007–​2010
Among the first human evidence linking arsenic to cancer in case-​control study in Chile reported odds ratios for lifetime average
humans were case reports describing skin and internal cancers fol- arsenic water concentrations of < 26, 26–​79, 80–​197, and > 197 µg/​l
lowing ingestion of medicinal arsenic, arsenic in drinking water, or of 1.00 (reference), 0.92 (95% CI: 0.52, 1.61), 2.62 (CI: 1.53, 4.50),
wine contaminated with arsenical pesticides. The earliest quantitative and 6.00 (CI: 3.38, 10.64), respectively, for bladder cancer, and 1.00
epidemiologic studies of ingested arsenic and cancer were ecologi- (reference), 0.98 (CI: 0.62, 1.53), 1.70 (CI: 1.05, 2.75), and 3.18 (CI:
cal in design, conducted primarily in areas with chronic arsenicism 1.90, 5.30) for lung cancer (Steinmaus et al., 2013). Elevated odds
and highly contaminated drinking water, such as Taiwan, Argentina, ratios were also reported for kidney cancers, helping to confirm pre-
and Chile. The arsenic exposure in Taiwan began in the 1940s, long vious ecologic mortality observations for kidney cancer (Chen et al.,
before the tragic exposures in Bangladesh, but also resulted from a 1988; Ferreccio et al., 2013a). Extensive summaries of other epide-
shift from surface water to wells to avoid microbial contaminants in miologic studies on arsenic and cancer in humans and experimental
308

308 PART III:  THE CAUSES OF CANCER


animal studies are provided elsewhere (Agency for Toxic Substances Metabolism
and Disease Registry [ATSDR], 2007; IARC, 2012a; NRC, 1999, In humans, ingested inorganic arsenic is metabolized through a com-
2014). plex series of reduction and methylation steps (Figure 18–​2). Ingested
Interestingly, not all of the effects of arsenic on cancer are detrimen- inorganic pentavalent arsenate [As(V)] is rapidly reduced to arsenic
tal. As mentioned previously, arsenic trioxide is an effective treatment trioxide [As(III)], which then accepts a methyl group from S-​adenosyl
for promyelocytic leukemia (Coombs et  al., 2015), once considered methionine (SAM) to produce the methylated pentavalent species
among the most fatal forms of leukemia. Also provocative is that a monomethylarsonic acid, MMA(V). MMA(V) is then reduced to tri-
study from northern Chile found a 49% decrease in breast cancer mor- valent monomethylarsonous acid, MMA(III). An additional methyl
tality among women over age 30 years during the 13-​year period of group can then be added to yield dimethylarsinic acid, DMA(V),
high arsenic water concentrations (near 860 µg/​l) in the area (Smith which can be reduced to small amounts of DMA(III) (NRC, 2014). As
et al., 2014). In fact, during the latter part of the high-​exposure period mentioned, urine is the primary route of arsenic excretion in humans.
(1965–​1970), breast cancer mortality among exposed women under A potentially useful marker of an individual’s ability to metabolize
age 60 years was 70% lower than that among unexposed women. inorganic arsenic is the percentage that different metabolites contrib-
Regional differences in diagnosis, treatment, and confounding vari- ute to the total amount in urine. Although there is substantial inter-​
ables were found to be unlikely causes for the lower risk. Although this individual variation, on average, As(III) and As(V) together account
finding has not yet been replicated elsewhere, its biologic plausibility for 10%–​30% of the total, MMA(III) and MMA(V) contribute 10%–​
is supported by in vivo evidence of increased apoptosis of breast can- 20%, and DMA(III) and DMA(V) combined account for the remain-
cer cells at exposure levels similar to those in the Chile study area (Sun ing 60%–​70% (Hopenhayn-​Rich et al., 1993; Vahter, 1999a).
et al., 2011; Xia et al., 2012). The methylation of arsenic was previously thought to contribute pri-
marily to detoxification, since the pentavalent metabolites, MMA(V)
and DMA(V), are acutely less toxic in vitro and more rapidly excreted
Factors That May Affect Risk than arsenic in its inorganic form (Vahter and Concha, 2001). However,
more recent in vitro studies have shown that MMA(III) may be more
Several factors have been reported to influence susceptibility to arsenic-​ toxic, at least acutely, than inorganic arsenic (Styblo et al., 2000). This
related cancers. These include demographic traits (age, gender), meta- suggests that MMA(III) could be the principal toxicant and that people
bolic capability, genetic susceptibility, nutrition, and smoking. who are less able to metabolize MMA(III) to DMA(V) may be more
susceptible to arsenic toxicity. MMA(III) is difficult to measure in
Demographic Factors epidemiologic studies because it is rapidly oxidized to its pentavalent
Age at exposure appears to modify susceptibility to certain arsenic-​ form in urine (Kalman et al., 2014).
related cancers. Exposure in utero or during childhood is more strongly Epidemiologic studies have found that people with high urinary
associated with lung and bladder cancer than the same level of expo- proportions of total MMA or low proportions of total DMA have 2–​5
sure in adulthood. In the Chile case-​control study (noted earlier), odds times higher risk of arsenic-​related cancers and other diseases than
ratios of 5.24 (95% CI:  3.05, 9.00) for lung cancer and 8.11 (95% individuals who do not (Smith and Steinmaus, 2009). For example, in
CI: 4.31, 15.25) for bladder cancer were seen among adults exposed to the highly exposed southwest coast of Taiwan, odds ratios for urothe-
high arsenic water concentrations in utero or as children. Odds ratios lial carcinoma were calculated for different strata of lifetime exposure
were lower among those whose exposure began later (Steinmaus et al., to arsenic, further stratified according to high or low proportions of
2014). Ecologic studies in Chile also found evidence of increases in urinary inorganic arsenic excreted as MMA (%MMA). Subjects in
kidney, laryngeal, and liver cancer mortality among adults who had the lower category of both cumulative exposure (< 20 mg/​l-​years) and
been highly exposed in early life (Smith et al., 2012). These findings %MMA (< 11.40%) served as the reference group. The odds ratios for
are supported by studies in mice showing that prenatal arsenic expo- urothelial cancer in subjects with cumulative exposure ≥ 20 mg/​l-​year
sure increases the occurrence of lung, liver, and other tumors later in were 1.5 (95% CI: 0.4, 5.9) in those with %MMA < 11.40%, versus
life, whereas the same level of arsenic exposures in adulthood does not 3.7 (95% CI: 1.2, 11.6) in those with higher %MMA values (Huang
affect risk (Tokar et al., 2010a). The mechanisms by which in utero et al., 2008). Similar patterns have been seen in Chile for lung cancer,
or long-​term/​early-​life exposure affect risk are unknown, although where odds ratios for arsenic water concentrations >200 ug/​l were 3.16
the in utero period is a time of major epigenetic reorganization, and (95% CI: 1.59, 6.32) in subjects with low %MMA values (< 12.5%)
arsenic has been linked to epigenetic changes in laboratory animals but 6.81 (95% CI: 3.24, 14.31) in subjects with higher %MMA values
and humans (Kile et al., 2014). Epigenetic changes could affect gene (Melak et al., 2014).
expression and regulation later in life. Other proposed mechanisms
relate to the effects of arsenic on the developing immune system or Genetic Susceptibility
other rapidly developing organs, to exposure before the develop- Polymorphisms in genes that code for the enzymes involved in
ment of detoxification enzymes or to the higher dose relative to body arsenic methylation have been linked to poor arsenic metabo-
mass index (BMI) in fetuses and children compared to adults (Miller lism. The most consistent evidence for this involves the arsenic
et al., 2001). (3+) methyltransferase (AS3MT) genes (Figure 18–​2), for which

As (V) As(III) MMA (V) MMA (III) DMA (V)


GSH GSSG SAM SAH SAM SAH
OH OH CH3
OH OH
I I I
I I
O = Asv – OH AsIII – CH3 O = AsV – CH3
OH – AsIII – OH O = AsV – CH3
I I I
O– AS3MT OH AS3MT OH

Figure 18–​2.  Arsenic metabolism: in humans, ingested inorganic arsenic is metabolized through a series of reduction and methylation steps. In the past,
this was thought to be primarily a detoxification process since DMA(V) is more readily excreted and is less toxic than inorganic arsenic (As(V) or As(III)).
However, more recent evidence suggests that the intermediary species, MMA(III), may be more toxic than As(V) or As(III).
Abbreviations: As(V) = arsenate; GSH = glutathione; GSSG = glutathione disulfide; As(III) = arsenite; SAM = S-​adenosyl methionine; SAH = S-​adenosyl homo-
cysteine; AS3MT = arsenic-​3-​methyltransferase; MMA(V) = monomethylarsonic acid; MMA(III) = monomethylarsonous acid; DMA(V) = dimethylarsinic acid.
 309

Water Contaminants 309


polymorphisms have been statistically significantly associated with Much of the controversy about the effects of arsenic at low expo-
urinary methylation patterns (Antonelli et al., 2014). For example, sure levels reflects the difficulty of measuring long-​term exposures.
in an arsenic-​exposed region in Mexico, subjects with the C allele Because of the long latency of arsenic-​associated cancer, retrospec-
of Met287Thr in the AS3MT gene had higher mean urinary %MMA tive studies must collect or estimate exposure data from the distant
levels (15 vs. 10%, p < 0.05) and a higher odds of having arsenic-​ past (which are not commonly available), and prospective studies must
related premalignant skin lesions (odds ratio [OR]  =  4.28; 95% follow subjects over a period of decades (which can be prohibitively
CI:  1.0, 18.5) (Valenzuela et  al., 2009). Polymorphisms in other expensive). Assessing past or long-​term exposure patterns can be dif-
genes have also been linked to arsenic metabolism, including sev- ficult if subjects change water sources throughout their lives, or use
eral glutathione-​s-​transferase genes. different water sources with different arsenic concentrations through-
out the day (i.e., at home, work, and school). Single measurements of
Nutrition arsenic in urine or hair will likely only represent a small portion of
The availability of certain micronutrients may also affect a person’s lifetime exposure for many people. In addition, among people with
ability to methylate arsenic. For example, low intakes of methionine, relatively low drinking water arsenic concentrations (e.g., < 10 µg/​
protein, and choline have been shown to impair arsenic methylation l), food-​borne arsenic can become a more important exposure source
in experimental animals (Vahter and Marafante, 1987). Low body (Meliker et al., 2006). Because food consumption patterns and arsenic
mass or weight has been associated with increased risks of arsenic-​ concentrations in foods may change over time, assessing past or long-​
related skin lesions in India and Bangladesh (Ahsan et al., 2006; Guha term exposure to arsenic from food can also be challenging. Inaccurate
Mazumder et  al., 1998). Folate affects one carbon metabolism, and or missing data can increase misclassification of exposure, can lead to
therefore may modify the transfer of methyl groups from S-​adenosyl errors in estimating study subjects’ true arsenic exposure, and there-
methionine that occur in arsenic methylation (Hall and Gamble, 2012; fore can limit the ability of a study to identify true associations (Cantor
Kile and Ronnenberg, 2008). In arsenic-​exposed areas of Bangladesh, and Lubin, 2007).
folate supplementation was associated with decreased blood arsenic Other issues relate to statistical power and the limits at which rela-
levels and an increased percentage of inorganic arsenic excreted as tively small increases in risk can be detected in epidemiologic stud-
DMA (Gamble et al., 2006; Peters et al., 2015). Folate deficiency has ies. In other words, if arsenic concentrations that are currently allowed
been linked to arsenic-​caused skin lesions (Melkonian et  al., 2012); in drinking water were truly associated with increased cancer risks,
most of these studies have been cross-​sectional, and clear prospec- these increases would expected to be fairly small (e.g., relative risks of
tive associations between folate intake and cancer have not been seen < 1.15 at the current US and World Health Organization limits of
to date. 10 µg/​l) (Gibb et al., 2011). Such small increases in risk, while impor-
Selenium increases the non-​enzymatic methylation of inorganic tant from a public health standpoint, are difficult to detect in epide-
arsenic, and has been associated with reduced arsenic toxicity and miologic studies without very large sample sizes, detailed exposure
increased arsenic excretion in animal and laboratory tests (Zwolak and assessment, and rigorous control of confounding. Even small increases
Zaporowska, 2012). Both selenium and arsenic affect DNA methyla- in risk can be important if the exposure and endpoints are common.
tion in vivo and in vitro, suggesting that in cytosine DNA methyltrans- Lung and bladder cancer are among the most common cancer types
ferase, selenium, and arsenic compete for methyl donation from SAM. worldwide. Given the potential importance of studies of low exposure,
Higher levels of blood selenium have been associated with lower such studies must be carefully designed and executed to provide mean-
percentages of MMA in blood or urine (Basu et  al., 2011; Pilsner ingful estimates of risk (Cantor and Lubin, 2007).
et  al., 2011), and with lower risks of premalignant arsenic-​related
skin lesions (Chen et al., 2007), but little information is available on
whether selenium modifies cancer risks related to arsenic. Mechanism(s) of Carcinogenicity
Currently, the primary mechanisms by which arsenic exerts its car-
Other Factors cinogenic effects in humans are unknown. Numerous pathways
Synergistic associations have been seen between arsenic exposure in have been proposed. Evidence suggests that arsenic does not inter-
either air or water, and tobacco smoking (Chen et  al., 2004; Hertz-​ act directly with DNA (Nesnow et al., 2002), but may be genotoxic
Picciotto et al., 1992). For example, in Chile, arsenic water concentra- through other mechanisms. One possibility is that the metabolism of
tions > 200 µg/​l were associated with lung cancer odds ratios of 8.0 arsenic generates reactive oxygen species and/​or decreases antioxidant
(95% CI:  1.7, 52.3) among never-​smokers, but 32.0 (95% CI:  7.22, defenses, leading to DNA damage and chromosomal changes (Kitchin
198.0) in ever-​smokers (Ferreccio et al., 2000). Some evidence of syn- and Conolly, 2010). Chromosomal alterations from arsenic exposure
ergy between arsenic in water and certain occupational exposures has are evidenced by the induction of micronuclei in human lymphocytes
been reported in relation to premalignant or malignant skin lesions or and in exfoliated bladder cells (Cantor et al., 2006). Arsenic may also
other cancers for pesticides, silica, asbestos, and wood dust (Ferreccio alter enzymes involved in DNA repair (Andrew et al., 2009; Hsu et al.,
et al., 2013b; Melkonian et al., 2011). Recently, evidence of synergy 2008) or DNA methylation. Both hypo-​and hypermethylation of DNA
between arsenic and excess BMI were reported in a cancer case-​ have been associated with arsenic exposure in exposed populations
control study in Chile (Steinmaus et al., 2015). (Bailey and Fry, 2014; Ren et al., 2011). The role of these changes in
human cancer remains unclear. Cytotoxicity following sulfhydral pro-
tein binding of reactive metabolite species and subsequent regenerative
Cancer Risks at Lower Exposures cell proliferation or hyperplasia might also lead to the development of
The most convincing epidemiologic evidence of the carcinogenicity cancer (Cohen et al., 2013). Modulations in the immune system have
of ingested arsenic comes from studies involving high exposure lev- been linked to cancer risks (Finn, 2012), and arsenic has been associ-
els, that is, drinking water arsenic concentrations > 100 µg/​l. Findings ated with a variety of inflammatory and infectious diseases, as well
at exposure levels below this are mixed (Gibb et  al., 2011)  (Table as to biomarkers of altered immune response that may be related to
18–​1). This issue is important because millions of people worldwide immune suppression and altered immune surveillance of cancerous
are exposed to these lower levels, but the associated cancer risks are cells (Dangleben et al., 2013). Increasing evidence regarding the role
unknown. Linear extrapolation from higher dose studies suggests that of stem cells in cancer development, as well as the links between early-​
the cancer risks at (lifetime) exposures near the US and World Health life arsenic exposure and adult disease, has led to theories regarding
Organization limits for arsenic in water (10 µg/​l) may be on the order the role of stem cells in arsenic carcinogenicity (Tokar et al., 2011).
of one extra cancer case for every 300 people (NRC, 2001). Some have In a rat kidney stem cell or partially differentiated progenitor cell
suggested that the dose–​response relationship between ingested arse- line, low-​level chronic arsenic exposure induced phenotypic changes
nic and cancer is not linear and that relatively low arsenic exposure lev- of cancer, including increases in colony formation (indicative of the
els may be associated with little or no excess risk (Cohen et al., 2013). loss of contact inhibition), increased proliferation, increases in matrix
310

Table 18–​1. Cancer Studies in Populations with Arsenic Water Concentrations < 100 µg/​L

Study Area/​Outcome Results Description

Bates et al., Utah Cumulative arsenic ≥ 53 mg 1. Case-​control study


1995 Bladder cancer OR = 1.41 (90% CI: 0.7, 2.9) 2. No past arsenic water concentration measurements
3. No data on private wells
4. OR > 1.0 but not statistically significant in all subjects.
Corresponding OR in smokers = 3.32 (90% CI: 1.1, 10.3)
Lewis et al., Utah Cumulative arsenic > 5000 μg/​L-​yr 1. Retrospective cohort mortality study
1999 Multiple cancers Lung cancer: SMR = 0.44 (p < 0.05) 2. Rare population: rural Mormon population 1900+.
in males and 0.22 (p > 0.05) 3. Lifestyle factors (smoking, diet, other) not assessed
in females 4. Unusual results: low SMR for lung cancer potentially an
indication of major confounding
Karagas et al., New Hampshire Toenail arsenic > 97th percentile 1. Case-​control study
2001 Skin cancer OR = 2.07 (0.92, 4.66) 2. Toenails measure only a few months of exposure: past exposure
unknown
3. OR > 1.0 but not statistically significant
4. No clear dose–​response relationship
Steinmaus California/​Nevada Arsenic intake > 80 μg/​day 1. Case-​control study
et al., 2003 Bladder cancer OR = 1.78 (0.89, 3.56) for exposures > 2. Low power to assess past exposures
40 years before cancer diagnosis 3. < 9% of subjects lived in highly exposed areas > 40 years before
diagnosis
4. OR > 1.0 but not statistically significant
Karagas et al., New Hampshire Toenail arsenic > 97th percentile 1. Case-​control study
2004 Bladder cancer OR = 2.17 (0.92, 5.11) among smokers 2. Toenails measure only a few months of exposure: past exposure
unknown
3. OR > 1.0 but not statistically significant
4. OR not increased in never smokers
5. No clear dose–​response relationship
Lamm et al., United States County median arsenic concentration 1. Ecologic mortality study
2004 Bladder cancer ≥ 3 μg/​L 2. County median used despite wide variability within counties
SMR = 0.94 (0.90, 0.98) 3. Only a fraction of all wells within exposed counties were
measured
4. No data on past exposure
5. No data on smoking, diet, or other potential confounders
Heck et al., New Hampshire and Toenail arsenic ≥ 0.1137 μg/​g 1. Case-​control study
2009 Vermont OR = 0.89 (0.46, 1.75) All types 2. ORs much lower without multiple adjustments; the confounder
Lung cancer OR = 2.75 (1.00, 7.57) Squamous/​ causing this is unknown
small cell 3. Toenails measure only a few months of exposure: past exposures
unknown
4. No clear dose–​response relationship
Gilbert-​ New Hampshire Urine arsenic ≥ 5.31 μg/​L 1. Cross-​sectional: urine samples measured at the time of cancer
Diamond Skin cancer OR = 1.43 (0.91, 2.27) diagnosis
et al., 2013 2. Urine measures only a few weeks of exposure
3. No direct data on past exposure
4. OR > 1.0 but not statistically significant
5. OR not elevated in long-​term residents
Meliker et al., Michigan Exposed counties 1. Ecologic mortality study
2007 Multiple cancers Lung cancer: SMR = 1.02 (99% 2. Ecologic exposure data based on county
CI: 0.98, 1.06) in males and 1.02 3. Wide variability in exposure within counties
(99% CI: 0.96, 1.07) in females 4. Past exposure not assessed
Bladder cancer: SMR = 0.94 (99% 5. No data on smoking, diet, or other potential confounders
CI: 0.82, 1.08) in males and 0.98
(99% CI: 0.80, 1.19) in females
Meliker et al., Michigan Time weighted lifetime average water 1. Case-​control study
2010 Bladder cancer arsenic concentration > 10 μg/​L 2. Exposure in past private wells based on statistical modeling
OR = 1.10 (0.65, 1.86)
Garcia-​ Arizona, Oklahoma, and 80th vs. 20th percentile urinary arsenic 1. Prospective cohort mortality study
Esquinas North/​South Dakota HR = 3.30 (1.28, 8.48) Prostate cancer 2. Unusual findings: highest hazard ratios are for outcomes not
et al., 2013 Multiple cancers HR = 2.46 (1.09, 5.58) Pancreas cancer commonly linked to arsenic (prostate and pancreas cancer)
(Strong HR = 1.56 (1.02, 2.39) Lung cancer 3. Urine measures only a few weeks of exposure
Heart Bladder cancer: insufficient number
Study) of cases
Dauphine California/​Nevada Arsenic water concentration ≥ 85 μg/​L 1. Case-​control study
et al., 2013 Lung cancer OR = 1.39 (0.55, 3.53) for exposures > 2. Low power to assess past exposures
40 years before cancer diagnosis 3. < 15% of subjects lived in highly exposed areas > 40 years
before diagnosis
4. OR > 1.0 but not statistically significant
Baastrup Denmark Cumulative exposure 5 mg 1. Retrospective cohort study
et al., 2008 Multiple cancers IRR = 1.00 (0.98, 1.03) Lung cancer 2. Very low exposures: median exposures of 0.7–​2.1 ug/​L and 95th
IRR = 1.01 (0.98, 1.04) Bladder cancer percentiles of 2.0–​2.5 ug/​L
IRR = 0.94 (0.81, 1.09) Kidney cancer 3. Exposures from food not assessed
IRR = 0.99 (0.97, 1.01) Skin cancer
 31

Water Contaminants 311


Table 18–​1.  Continued

Study Area/​Outcome Results Description

Chen et al., Northeastern Taiwan For arsenic water concentration < 100 1. Cohort study
2010a Lung cancer µg/​L: 2. Arsenic water concentrations measured at the residence at the
RR = 1.10 (0.74, 1.63) for 10–​49.9 µg/L​ time of study recruitment
RR = 0.99 (0.59, 1.68) for 50–​99.9
ug/​L
Chen et al., Northeastern Taiwan For arsenic water concentration < 100 1. Cohort study
2010b Urothelial cancer µg/​L: 2. Arsenic water concentrations measured at the residence at the
RR = 1.85 (0.45, 7.61) for 10–​49.9 µg/L​ time of study recruitment
RR = 2.19 (0.43, 11.1) for 50–​99.9
ug/​L
Chen et al., Northeastern and For arsenic water concentration 10–​99 1. Cohort study
2004 southwest Taiwan µg/​L: 2. Includes some subjects in Chen et al., 2010
Lung cancer RR = 1.09 (0.63, 1.91) 3. Less than lifetime exposure in many subjects
Han et al., Idaho, US County average arsenic concentrations 1. Ecologic study
2009 Multiple cancers No statistically significant 2. Used county average arsenic well concentrations
correlations with lung, bladder, or 3. Mostly low exposures
kidney cancer incidence in adjusted 4. Did not account for latency
analyses
Buchet and Belgium Arsenic water concentrations 20–​50 1. Ecologic mortality study
Lison, 1998 Multiple cancers µg/​L 2. Limited exposure data
Males 3. Exposures from water and air, subjects living near zinc smelters
SMR = 1.05 (0.94, 1.18) Lung cancer
SMR = 0.92 (0.63, 1.35)
Bladder cancer
SMR = 1.13 (0.67, 1.91) Kidney cancer
Females
SMR = 1.24 (0.83, 1.87) Lung cancer
SMR = 1.20 (0.63, 2.31)
Bladder cancer
SMR = 0.91 (0.45, 1.81) Kidney cancer
Ferreccio Chile Arsenic water concentrations < 89 µg/​L 1. Case-​control study
et al., 2000 Lung cancer OR = 0.3 (0.1, 1.2) for 10–​29 µg/​L 2. Exposure based on average concentrations for the years
OR = 1.8 (0.5, 6.9) for 30–​59 µg/​L 1958–​1970
OR = 4.1 (1.8, 9.6) for 60–​89 µg/​L
Kurttio et al., Finland Arsenic water concentrations ≥ 0.5 µg/​L 1. Retrospective cohort study
1999 Bladder and kidney RR = 1.51 (0.67, 3.38) Bladder cancer 2. Very low exposures
cancer RR = 1.07 (0.46, 2.52) Kidney cancer 3. Elevated risk ratios seen in smokers: RR = 10.3 (1.16, 92.6) for
For exposures > 10 years before cancer bladder cancer
diagnosis
Steinmaus Chile As conc (µg/​L) OR   (90% CI) 1. Case-​control study
et al., 2014 Lung cancer   6.5 1.00 (ref) 2. Median arsenic water concentrations
23.0 1.43 (0.82, 2.52) 3. Some data obtained in interviews with next-​of-​kin
58.5 2.01 (1.14, 3.52)

As: arsenic; CI: confidence interval; HR: hazard ratio; IRR: incidence rate ratio; OR: odds ratio; RR: relative risk; SMR: standardized mortality ratio.
95% confidence intervals in parentheses unless otherwise noted.

metalloproteinases, and dysregulated signaling pathways (Tokar et al., 2010; Sancha et al., 2000; Su et al., 2011; US EPA, 2012). Other treat-
2013). Other studies have shown that arsenic may affect stem cells in ments include reverse osmosis, activated alumina, microfiltration, and
other cell types, including skin and prostate (Sun et al., 2012; Tokar ion exchange (US EPA, 2000b). Smaller reverse osmosis filters are
et al., 2010b). Although a number of different mechanisms have been commonly used to reduce arsenic concentrations in private domestic
proposed, the mechanism or combination of mechanisms most likely wells, where most of the other approaches are prohibitively expensive
responsible for the human health effects of arsenic is unknown. (Slotnick et al., 2006). The effectiveness of these filters may vary con-
siderably due to inconsistent maintenance and other factors (George
et al., 2006; Thomson et al., 2000; Walker et al., 2008).
Opportunities for Prevention A variety of approaches have been used to reduce arsenic exposures
The most effective and practical way to reduce cancer risks from arse- in Bangladesh. A mass survey of over 5 million wells measured arse-
nic is to reduce exposure. Several countries have promulgated regula- nic concentrations and used colored paint to indicate the level of con-
tory limits for arsenic concentrations in drinking water. In the United tamination. Wells with arsenic concentrations < 50 µg/​l were painted
States, these apply only to public water sources, not household wells. green; wells over this level (approximately 1.4 million) were painted
Large municipal water suppliers have reduced arsenic concentrations red (Johnston and Sarker, 2007). Contamination in this area primarily
through various methods, including the development of new water affected tube wells that accessed shallower aquifers (i.e., < 150 m).
sources (e.g., tapping new aquifers, desalinization of ocean water), Mitigation efforts have closed highly contaminated wells, expanded
mixing water from wells with higher arsenic concentrations with water the use of surface water, and created deep tube wells, shallow dug
with lower arsenic concentrations, and coagulation with iron salts and wells, and sand pond filters. Educational programs were implemented
filtration (Albuquerque Bernalillo County Water Utility Authority, in highly exposed areas in Araihazar, Bangladesh. Participation in
312

312 PART III:  THE CAUSES OF CANCER


a school-​ based educational and intervention program resulted in 2010; Lourencetti et  al., 2012), blood (Aggazzotti et  al., 1990), and
a five-​fold increase in switching to a lower arsenic well, and sig- urine (Caro and Gallego, 2007, 2008).
nificant declines in urinary arsenic levels among participants (Khan
et al., 2015).
Chelation therapy with British anti-​lewisite (BAL; dimercaprol) or Types of Cancer
its analogs unithiol (DMPS) and succimer (DMSA) is used to treat very Soon after THMs were identified, ecologic studies conducted in the
high-​dose acute poisonings (e.g., occupational accidents, homicide, or United States, Norway, and Finland showed elevated cancer rates
suicide attempts). Chelation has not been shown to be appropriate for associated with chlorination byproducts or surrogate measures (Cantor
prevention or treatment of arsenic exposures from food or drinking et al., 2006). The exposures in these studies were broadly defined, with
water (Kosnett, 2013). Biomonitoring and early detection have been some studies comparing the use of chlorinated to unchlorinated water,
proposed in areas where arsenic exposures have already been reduced and others comparing surface water (which tends to have higher lev-
(Adonis et al., 2014). To date, the effectiveness of such programs has els of DBPs) to groundwater. Levels of THM and mutagenicity were
not been established. estimated. The most commonly identified cancer sites with elevated
rates were bladder, colon, and rectum. Subsequently, a number of
death-​certificate-​based case-​control studies were conducted, focusing
DISINFECTION BYPRODUCTS on these and other sites. Information on cause of death and current
residence was obtained from death certificates (Cantor et  al., 2006).
Although these studies had methodologic limitations, including a
Exposure
lack of historical exposure information and information on potential
The disinfection of drinking water is one of the triumphs of pub- confounders, they generally supported an association between chlori-
lic health, responsible for significant reductions in mortality and nated byproducts and risk of cancers of the bladder, colon, and rectum
morbidity due to infectious disease (Cutler and Miller, 2005). An (Cantor et al., 2006).
unintended consequence of this approach, however, has been the Following the ecological studies of cancer mortality, a series of
formation of disinfection byproducts (DBPs) that may affect can- case-​ control studies of incident cancers collected more informa-
cer risk. DBPs are formed by the reaction of chlorine or other dis- tion on longer-​term exposures, and variables that might confound or
infectants with organic matter; they include a complex mixture of interact with DBPs. The site with the strongest evidence of a causal
compounds that depends on both the treatment regimen and charac- relationship was bladder, although risks associated with DBP were
teristics of the source water. The formation of specific compounds generally modest, with ORs around 1.5 associated with estimated
is influenced by the level and type of organic matter present, the total THM levels of 50 μg/​L (Costet et al., 2011). Three cohort and
existence of bromide and/​or iodide, and the pH and temperature of eight case-​control studies evaluated this association, in addition to
the water (Richardson et al., 2007). Trihalomethanes (THMs) com- several reviews and meta-​analyses (Costet et  al., 2011; Villanueva
prise one category of DBP. They were discovered in 1974 (Rook, et al., 2003). The cohort studies and all but two of the case-​control
1974)  and are composed of chloroform (typically the dominant studies were reviewed in the previous edition of this text and will not
THM), dibromochloromethane, bromodichloromethane, and bromo- be discussed here (Cantor et  al., 2006). Many of the earlier studies
form. More than 700 DBPs have now been identified. Major classes focused on surrogates of exposure to DBPs, such as use of a chlori-
that are regulated in the United States include the THMs, haloacetic nated water source for drinking water, or duration of use of surface
acids (HAAs), and oxyhalides (bromate and chlorite). There are also versus groundwater. A meta-​analysis published in 2003 reported an
multiple unregulated classes, such as the halonitromethanes, iodo-​ odds ratio of 1.2 (95% CI: 1.1, 1.4) associated with ever consuming
acids and other halo-​ acids, chlorinated hydroxy furanones (e.g., chlorinated water, and 1.4 (95% CI: 1.2, 1.7) for long-​term exposure
MX), haloamides and haloacetonitriles, nitrosamines, and aldehydes (Villanueva et al., 2003). Although results were generally consistent
(Richardson et  al., 2007). Many of these compounds have been among studies, variation existed particularly with respect to apparent
shown to be mutagenic and/​or genotoxic. As indicated earlier, only sex differences. Some studies showed stronger effects among men.
three classes of compounds are regulated in the United States; it is However, none of the individual studies had adequate power to detect
assumed that reduced levels of these compounds will reduce overall associations among women.
exposure to DBPs (US EPA, 2003). The regulatory maximum con- Since the previous edition of this text, two case-​control studies of
taminant level (MCL) in the United States is 80 ppb total for total bladder cancer have been conducted, one from six regions in Spain
THM, 60 ppb for the HAAs, 10 μg/​l for bromate, and 1000 μg/​l for (Villanueva et al., 2007) and another in three northern New England
chlorite (US EPA, 2006). Concern about bladder cancer was a key states in the United States (Beane Freeman et al., 2017). Both expanded
consideration when the rules and cost–​benefit analysis were updated upon previous exposure assessment efforts by considering routes of
in 2006 (US EPA, 2005). exposure other than ingestion, such as showering, bathing, and use
In 1991, the IARC determined that there was inadequate evidence of swimming pools (Costet et  al., 2011; Villanueva et  al., 2004). In
to classify the use of chlorinated drinking water as carcinogenic to Spain, THM levels were estimated by using historical information
humans (IARC, 1991). Since then, IARC has evaluated individual from individual municipalities (Villanueva et al., 2006). Compared to
DBPs, none of which is listed as a Class I carcinogen (IARC, 2004, participants in the lowest quartile of long-​term average THM concen-
2013, 2014). Chloral and chloral hydrate were designated probable tration in water (< 8 µg/​l), those in the highest quartile (> 49 μg/​l) had
human carcinogens (Group 2A), whereas dichloroacetic acid, trichlo- a two-​fold increase of bladder cancer risk (OR = 2.10; 95% CI: 1.09,
roacetic acid, dibromoacetic acid, bromochloroacetic acid, and MX 4.02). In a metric that incorporated both the THM concentration and
were listed as possible human carcinogens (Group 2B) (IARC, 2004, the amount of water ingested, there was a non-​significant increase in
2013, 2014). bladder cancer risk among those with the highest intake of THM (> 35
People can be exposed to DBPs through ingestion, dermal absorp- μg/​d) compared to those with no exposure (OR = 1.35; CI: 0.92, 1.99)
tion, and inhalation. While ingestion has received the most attention (Villanueva et  al., 2007). Information about the duration of shower-
in epidemiologic studies, recent work has evaluated exposure through ing and bathing was combined with the average THM concentration
inhalation or dermal absorption of low molecular weight, volatile, to evaluate inhalation and dermal exposure. Those with the highest
non-​polar constituents (Richardson et  al., 2007). Studies have dem- exposure levels had significantly elevated risk (OR = 1.83; CI: 1.17,
onstrated increased THM levels in blood following exposure, with 2.87). Bladder cancer risk was also elevated among those who reported
higher THM blood levels associated with showering than with inges- ever use of swimming pools, another potentially large source of DBP
tion (Backer et al., 2000). Additionally, studies of swimmers in chlo- exposure (OR = 1.57; CI: 1.18, 2.09), although risk did not increase
rinated pools show increased THM concentrations in exhaled air with increasing total lifetime hours of use. The results for ingestion
(Aggazzotti et  al., 1993; Caro and Gallego, 2007; Kogevinas et  al., and concentration were stronger among men than women, whereas the
 31

Water Contaminants 313


associations with showering/​bathing and swimming pool exposures Table 18–​3), nor with individual THMs or HAAs in models adjusted
were similar in both sexes. for water nitrate level (Jones et  al., 2016). More recently, a small
In the population-​based case control study in New England (Beane study conducted within the National Health and Nutrition Survey
Freeman et al., 2017), historical information from public utilities and demonstrated an association between blood levels of brominated,
residential histories were used to estimate total THM exposures, as but not chlorinated, THMs and cancer mortality overall (Min and
well as levels of chlorinated and brominated compounds. At the 95th Min, 2016). Other sites that have been evaluated include brain,
percentile of average daily intake (> 103.9 µg/​day), bladder cancer esophagus, pancreas, and childhood leukemia (Cantor et al., 2006).
risk was significantly associated with total THM (OR = 1.53; 95% CI: Each of these sites has at least one epidemiologic study that shows
1.01, 2.32) and brominated THMs (OR = 1.98; 95% CI: 1.19, 3.29). some evidence of association with increasing exposure to DBPs.
For cumulative intake (mg) of chlorinated and brominated THMs from As indicated, for both colon and bladder cancer there is some sug-
age 10 through diagnosis or interview, bladder cancer risk was elevated gestion that risk may differ by sex, with many studies showing associa-
among those with > 95th percentile of total THM (OR = 1.45; 95% CI: tions with THMs only in men (Costet et al., 2011; King et al., 2000).
0.95, 2.2), and chlorinated and brominated THMs (OR = 1.77; CI: However, it should be noted that most studies are underpowered to
1.05, 2.99 and OR = 1.78; CI: 1.05, 3.00, respectively). There was detect associations in women. The two largest studies of bladder can-
no evidence of bladder cancer risk with swimming pool use (OR = cer, with the most women, did not demonstrate sex differences (Beane
0.94; CI: 0.55, 1.59 for > 12,174 lifetime hours compared to those who Freeman et al., 2017; Cantor et al., 1987).
never used swimming pools).
Although both studies showed increased risk of bladder cancer
associated with increased THM levels, the risk estimates in the New Mechanisms of Carcinogenesis
England study were lower than in Spain. It should be noted that the
estimated exposure to THMs was also much lower in New England Given the number of compounds identified as DBPs, it is not surpris-
(Beane Freeman et  al., 2017; Salas et  al., 2013). Differences in the ing that several mechanisms for carcinogenicity have been proposed.
association with swimming pool use may be due to chance or to dif- Some DBPs are genotoxic or mutagenic, either alone or as part of a
ferences in swimming pool disinfection that are not well understood. mixture; chloroform is considered to be mutagenic only at very high
The DBPs in swimming pools are affected by the same characteristics levels of exposure (Richardson et al., 2007). Water samples from five
as drinking water. The specific DBPs formed in swimming pools vary European countries demonstrated cytotoxicity in mammalian cells that
by the disinfection processes used (Lee et  al., 2010), the number of increased with the number of DBP compounds identified in each sam-
swimmers in the pool, and the presence of urine, other body fluids, and ple (Jeong et al., 2012). Although there were differences in genotoxic-
personal care products (Chowdhury et al., 2014). These issues should ity among the samples, it did not appear that the genomic DNA damage
be studied further. in these samples correlated with the chemical analyses. Therefore, it
Much less epidemiologic research has been conducted on the may be that the observed associations were due to unidentified DBPs,
association between DBP exposure and cancers other than bladder. or perhaps to combinations of DBPs. There has been interest in the
Colon and rectum cancer are the sites with the most evidence for effect of genetic susceptibility in modifying risk. In an analysis from
an association. A case-​control study in Spain and Italy reported no the Spanish bladder cancer study, Cantor and colleagues (Cantor et al.,
association between total THMs and colorectal cancer, although 2010) demonstrated significant interaction between genetic variation
there was a suggestive increase in risk among men with the highest in key metabolizing pathways and DBP levels on bladder cancer risk.
quartile of brominated THM exposure (OR = 1.43; 95% CI: 0.83, Exposed individuals with high-​risk variants of GSTT1, GSTZ1, and
2.46, p trend = 0.04) (Villanueva et al., 2017). There was no associa- CYP2E1 were at increased risk compared with those without the high-​
tion with showering and bathing in water with higher levels of total risk forms. Other studies have not reported attempts to replicate these
THMs, chloroform, or brominated compounds, nor were risks ele- findings.
vated in women. There was a suggestion of an interaction between It has been suggested that epigenetic mechanisms may also be
brominated THMs and four genetic polymorphisms in the CYP2E1 important, particularly with regard to long-​term, lower-​level expo-
gene. An interaction between this gene and total THMs was observed sures. In controls from the bladder cancer case-​control study from
in the Spanish study of bladder cancer. Results from this study were Spain, the levels of DNA methylation at some transposons (LINE-​1 5-​
similar for both colon and rectal cancers. Other studies, however, methylcytosine levels) in granulocytes were associated with increased
have suggested that the risks may differ for colon and rectum can- lifetime exposure to THM (Salas et al., 2014). There was also some
cer. For example, a case-​control study of colon and rectum cancer evidence that levels of methylation modified the association between
in Ontario, Canada, reported increased risk of colon cancer in men, THM levels and bladder cancer risk (p-​interaction = 0.03). In another
with increasing cumulative exposure to THMs, duration of exposure epigenome-​wide study, genes that have been associated with cancer
to chlorinated surface water, and duration of exposure to THM lev- at several sites, including colorectal and bladder, were found to have
els > 50 µg/​l. No associations were observed with rectum cancer or methylation levels that differed by THM level (Salas et al., 2015).
in women (King et al., 2000). A case-​control study in Iowa reported
increased risk of rectal cancer (Hildesheim et al., 1998). Recently,
two studies have suggested an increased risk of rectal cancer at higher NITRATE
levels of bromoform exposure. A case control study of rectal cancer
in New York showed a statistically significantly increased risk at the
highest levels of bromoform exposure (1.69–​15.43 µg/​day), but not Exposure
with other THMs (Bove et al., 2007). An ecologic study in Australia Since the mid-​1920s, human activities have doubled the natural rate
evaluated colon and rectal cancers in relation to total THMs and at which nitrogen is deposited onto land. Most important has been the
individual species of THMs (Rahman et  al., 2014). The authors production and application of nitrogen fertilizers, the combustion of
reported a statistically significantly increased incidence rate ratio fossil fuels, and replacement of natural vegetation with nitrogen-​fixing
for colon cancer with an increasing interquartile range of bromo- crops such as soybeans (Davidson EA, 2012; Vitousek et al., 1997). In
form (IQR = 2 µg/​l) in men, but not women. No other individual spe- 1909 the Haber-​Bosch process was developed to produce ammonia by
cies were associated with risk, nor was total THM level. There were reacting nitrogen gas with hydrogen. This allowed a dramatic increase
no significant associations with rectal cancer. Although the study in the production of synthetic fertilizers and expansion of global
was ecological, a strength was that it evaluated all THMs, separately agriculture. Fertilizer production has increased exponentially since
and in combination. A recent analysis of kidney cancer in a cohort 1960; half of all synthetic fertilizers have been produced since 1985
of postmenopausal women in Iowa found no association between (Howarth, 2008). As a result, nitrogen inputs to the land have steadily
kidney cancer and long-​term average total THM (HRQ4vsQ1 = 0.70; increased. Approximately half of all applied nitrogen drains from agri-
95% CI: 0.41=, 1.20), HAA5 (HRQ4vsQ1 = 0.65; 95% CI; 0.39, 1.08; cultural fields to contaminate surface and groundwater (Davidson EA,
314

314 PART III:  THE CAUSES OF CANCER


2012). Nitrate levels in groundwater under agricultural land can be epidemiologic evidence for drinking water nitrate was considered
several to 100 times higher than levels under natural vegetation (Nolan inadequate. In human studies, it is important to assess effect modi-
BT, 2000). Approximately 15% of the US population relies on pri- fication by exposure to modifiers of endogenous nitrosation. To date,
vately owned household wells for drinking water (Hutson, 2004). As there are few studies of any cancer site that have assessed long-​term
mentioned, these are not regulated by the EPA. The US Geological exposure from drinking water and potential interactions with dietary
Survey assessed available private well measurements in US aquifers factors that affect NOC formation.
sampled in 1991–​2004 and found that 8% exceeded the 10 mg/​l maxi-
mum contaminant level (MCL) for nitrate-​nitrogen (NO3-​N) (Focazio
et al., 2006). Other private well surveys show that a significant propor- Epidemiologic Studies
tion of wells, particularly those less than 30 meters in depth in agricul-
tural areas, exceed the MCL for nitrate (Centers for Disease Control Most early epidemiologic studies were ecologic studies of mortality
and Prevention, 1998; Johnson and Kross, 1990). Almost all public from stomach cancer that used exposure estimates based on measure-
water supplies have nitrate levels below the MCL. However, in the past ments of nitrate in drinking water concurrent with death from cancer.
few decades, nitrate levels have risen to levels approaching the MCL in Results were mixed, with some studies showing positive associations,
some public supplies located in agricultural areas (Ward et al., 2010). many showing no association, and a few showing inverse associations.
The results of ecologic studies through 1995 were reviewed in Cantor
(1997). Since that review, an ecologic study of cancer incidence in
Mechanisms of Carcinogenesis Slovakia (Gulis et al., 2002) evaluated 20-​year average nitrate levels in
The MCL for nitrate in drinking water was established at 10 mg/​l NO3-​ public water supplies (highest quartile: 6–​10 mg/​l NO3-​N) and found
N to protect against infant methemoglobinemia, not cancer or other a positive correlation with stomach cancer incidence among women
health outcomes (US EPA, 2016). Nevertheless, nitrate is a precur- but not men. Other ecologic studies of cancer incidence were largely
sor compound of N-​nitroso compounds (NOC), many of which are null: endpoints in these studies included the brain (Barrett et al., 1998;
potent animal carcinogens. NOC cause cancer in every animal spe- van Leeuwen et al., 1999), bladder and colon (Gulis et al., 2002; van
cies tested and at multiple organ sites (IARC, 2010; Lijinsky, 1986). Leeuwen et al., 1999), esophagus and stomach (Barrett et al., 1998),
Approximately 5% of nitrate ingested in drinking water or in the diet kidney (Gulis et al., 2002), ovary (van Leeuwen et al., 1999), and non-​
is reduced to nitrite by bacteria. This occurs mostly because of the Hodgkin lymphoma (NHL) (Cocco et al., 2003; Law et al., 1999; van
oral microbiome, but continues in the stomach, small intestine, colon, Leeuwen et  al., 1999). Among the limitations of ecologic studies is
and bladder when nitrate-​reducing bacteria are present (IARC, 2010). their inability to assess individual-​level exposure or dietary factors
Nitrite can then react with amines and amides to form nitrosamines that might interact with nitrate metabolism and affect the endogenous
and nitrosamides, the two main types of NOC. formation of NOC. They also do not identify potentially susceptible
Nitrate is found in many foods, with the highest levels occurring in subgroups or other interactions.
certain green leafy and root vegetables (NRC, 1981). Average daily Since the previous version of this chapter was published, 8 case
intakes are estimated to be in the range of 30–​130 mg/​day as nitrate control and 2 cohort studies have evaluated historical nitrate levels in
(IARC, 2010). Endogenous NOC formation is blocked by ascorbic public water supplies in relation to several cancers. The levels were
acid, alpha-​tocopherol, and other polyphenols. Because these inhibi- largely below 10 mg/​l NO3-​N. Most of these studies evaluated poten-
tors are present at high levels in most vegetables, dietary nitrate intake tial confounders and factors affecting nitrosation. Details of 9 case-​
from vegetables may not result in substantial NOC formation (Mirvish, control and 2 cohort studies published through 2005 were presented in
1995). Drinking water contributes the majority of nitrate intake when the previous version of this chapter (Cantor et al., 2006). Table 18–​2
levels are near the MCL (Chilvers et al., 1984; IARC, 2010). Sources shows the study designs and results of more recent studies, includ-
of exposure to preformed NOC include processed meats and fish, beer, ing the results of periodic follow-​ups of a cohort study of postmeno-
certain occupations, cosmetics, and some drugs. It is estimated that pausal women in Iowa (Inoue-​Choi et  al., 2012, 2015; Jones et  al.,
45%–​75% of human exposure to NOC comes from in vivo formation, 2016, 2017; Ward et al., 2010). In the first analysis of drinking water
however (Tricker, 1997). An IARC Working Group reviewed the epi- nitrate in the Iowa cohort with follow-​up through 1998, Weyer and
demiologic studies of ingested nitrate and nitrite through mid-​2006. colleagues reported that ovarian and bladder cancer were significantly
The evidence for dietary nitrite ingestion and cancer is considered associated with the long-​term average residential nitrate levels at the
limited (IARC, 2010), based on epidemiologic studies of stomach time of enrollment (Weyer et  al., 2001). They also observed signifi-
and esophageal cancer. However, another recent IARC review con- cant inverse associations for uterine and rectal cancer, but no associa-
cluded that processed meats, which usually contain added nitrite and/​ tions with cancers of the breast, colon, rectum, pancreas, kidney, lung,
or nitrate, cause cancer in humans, based mostly on the evidence for melanoma, NHL, or leukemia. Analyses of public water supply nitrate
colorectal cancer (Bouvard et al., 2015). concentrations and cancers of the thyroid, breast, ovary, bladder, and
Endogenous NOC formation due to ingestion of water with elevated kidney were published after additional follow-​up of the cohort. The
nitrate has been demonstrated in human volunteers (Mirvish et  al., exposure assessment was improved by (a) the computation of aver-
1992; Møller et al., 1989; Vermeer et al., 1998), generally under con- age nitrate levels and years of exposure at or above 5 mg/​l NO3-​N,
ditions of low antioxidant intake (e.g., low vitamin C). NOC also have based on time in residence (vs. using one public water supply average
been measured in human feces after ingestion of nitrate via drinking nitrate estimate used by Weyer and colleagues); and (b) by estimation
water (Rowland et al., 1991). In each of these four studies, increased of THM levels and dietary nitrite intake. Thyroid cancer was evaluated
nitrosation was observed at nitrate concentrations that exceeded the for the first time after follow-​up of the cohort through 2004. A total of
MCL. Some of the NOCs that were formed were known carcinogens. 40 cases was identified (Ward et al., 2010). Among women with > 10
In the hundreds of animal studies conducted since the 1970s, NOCs years of public water supply whose levels exceeded 5 mg/​l NO3-​N for
cause tumors of the upper and lower gastrointestinal tract, urinary 5 years or more, thyroid cancer risk was 2.6 times higher than that of
tract, lung, thyroid, breast, and ovary. Transplacental exposure induces women whose supplies never exceeded 5 mg/​l. Higher dietary nitrate
brain tumors in offspring (Rice, 1989). Long-​term exposure to lower intake was also associated with increased thyroid cancer risk. With fol-
NOC concentrations has a stronger carcinogenic effect than short-​term low-​up through 2010, the risk of ovarian cancer remained significantly
higher exposures (Lijinsky, 1986). increased among women in the highest quartile of average nitrate in
The IARC Working Group classified ingested nitrate or nitrite as public supplies (Inoue-​Choi et al., 2015). Ovarian cancer risk among
probably carcinogenic to humans (2A), when ingested under condi- private well users was also elevated compared to the lowest nitrate
tions that result in endogenous nitrosation (IARC, 2010). This desig- quartile for public supplies. Associations were stronger when vitamin
nation was based on human mechanistic studies, sufficient evidence in C intake was below median levels with a significant interaction for
animals for the carcinogenicity of nitrite in combination with amines users of private wells. Higher intake of processed meat sources of
or amides, and limited epidemiologic evidence for dietary nitrite. The dietary nitrite was also associated with increased risk. Overall, breast
 315
Table 18–​2. Case-​Control and Cohort Studies of Drinking Water Nitrate Levels and Cancer (2004–​2017)

First Author
(Year) Study Design, Years
Country Regional Description Exposure Description Cancer Sites Included Summary of Findingsa,b Comments

Mueller (2004) Population-​based case-​control Dipstick measurements of nitrate Childhood (< 15 years) > 11 mg/​L NO3-​N vs. ND OR = 1.0 Strongest associations for
6 countries Incidence, 1976–​1994 and nitrite in the pregnancy water malignant brain (CI: 0.4, 2.2); exclude bottled water astroglial tumors with dipstick
Los Angeles County, San supply among women who had not tumors users OR = 1.5 (CI: 0.6, 3.8) nitrate and nitrite measurements
Francisco Bay Area, Seattle–​ moved (185 cases, 341 controls); > 1.5 mg/​L NO2-​N vs. ND OR = 2.1 at pregnancy residence
Puget Sound region of the population excluding bottled water (CI: 0.6, 7.4); exclude bottled water
United States; Paris, France; users (131 cases, 241 controls) users OR = 5.2 (CI: 1.2, 23.3)
Milan, Italy; Valencia, Spain; Well as water source for entire
Winnipeg, Canada pregnancy including periconceptual
period (vs. public supply) associated
with increased risk in Canada and
Seattle but not other centers
Ward (2006) Population-​based case-​control Nitrate levels in public water Non-​Hodgkin Private wells: > 5.0 mg/​L NO3-​N vs. No effect modification by vitamin
United States Incidence, 1998–​2000 supplies among those with nitrate lymphoma ND OR = 0.8 (CI: 0.2, 2.5) C, smoking
Iowa estimates for > 70% of person-​ Public supply average: > 2.9 mg/​
years > 1960 (181 cases, 142 L NO3-​N vs. < 0.63 OR = 1.2
controls); nitrate measurements (CI: 0.6, 2.2)
for private well users at time of Years > 5mg/​L NO3-​N: 10+ vs. 0
interviews (1998–​2000; 54 cases, OR = 1.4 (CI: 0.7, 2.9)
44 controls).
Zeegers (2006) Cohort 1986 nitrate level in 364 pumping Bladder Nitrate intake from water (highest No effect modification by vitamin
Netherlands Incidence, 1986–​1995 stations, exposure data available quintile > 1.7 mg/​day NO3-​N C, E, smoking
204 municipal registries across the for 871 cases, 4359 members of [median in quintile = 2.4 mg/​day]
Netherlands the subcohort vs. lowest RR = 1.11 (CI: 0.87,
1.41; p-​trend = 0.14)
Ward (2007) Population-​based case control Nitrate levels in public water Kidney (renal cell Public supply average: > 2.8 mg/​ Joint effects of water nitrate
United States Incidence, 1986–​1989 supplies among those with nitrate carcinoma) L NO3-​N vs. < 0.62 OR = 0.89 and vitamin C showed
Iowa estimates for > 70% of person-​ (CI: 0.57, 1.39); Years > 5mg/​ similar pattern to red meat
years > 1960 (201 cases, 1244 L NO3-​N 11+ vs. 0 OR = 1.03 (p-​interaction = 0.13)
controls) (CI: 0.66, 1.60)
Joint effect for 11+ years > 5 mg/​
L NO3-​N and red meat (> 1.2
servings/​day) OR = 1.91 (CI: 1.04,
3.51); p-​interaction = 0.01
McElroy (2008) Population-​based case-​control, Limited to women in rural areas with Colorectum Private wells > 10.0 mg/​L NO3-​N vs. Interactions with dietary intakes
United States women no public water system (475 cases, < 0.5: Colorectal cancer OR = 1.52 not evaluated; no interaction
Incidence, 1990–​1992 and 1447 controls); nitrate levels at (CI: 0.95, 2.44); proximal colon with smoking
1999–​2001 residence (presumed to be private cancer: OR = 2.91 (CI: 1.52, 5.56);
Wisconsin wells) estimated by kriging using no association for distal colon
data from a 1994 representative cancer or rectal cancer
sample of 289 private wells
Ward (2008) Population-​based case control Controls from prior study of Stomach and esophagus Average nitrate quartiles (> 4.32 vs. > 2.7mg/​L NO3-​N & > 30g/​
United States Incidence, 1988–​1993 lymphohematopoetic cases and (adenocarcinomas < 2.45 mg/​L NO3-​N): stomach day processed meat vs.
Nebraska controls interviewed in 1992–​ only) OR = 1.2 (CI: 0.5, 2.7); esophagus low intakes both: stomach
1994; proxy interviews for 80%, OR = 1.3 (CI: 0.6, 3.1); OR = 2.0 (CI: 0.8, 4.8);
76%, 61% of stomach, esophagus, Years > 10 mg/​L NO3-​N (9+ vs. p-​interaction = 0.21; no
controls, respectively. 0): stomach OR = 1.1 (CI: 0.5, 2.3); interaction for esophagus; no
Nitrate levels (1965–​1985) in public esophagus OR = 1.2 (CI: 0.6, 2.7) interaction with vitamin C, red
water supplies for > 70% of Private well users (> 4.5mg/​L NO3-​ meat for either cancer
person-​years (79 distal stomach, N vs. < 0.5) stomach OR = 5.1
84 esophagus, 321 controls); (CI: 0.5, 52; 4 cases, 13 controls);
private wells sampled at interview esophagus OR = 0.5 (CI: 0.1, 2.9; 8
(15 stomach, 22 esophagus, 44 cases; 13 controls)
controls)
(continued)
316
Table 18–​2  Continued

First Author
(Year) Study Design, Years
Country Regional Description Exposure Description Cancer Sites Included Summary of Findingsa,b Comments

Ward (2010) Cohort of women ages 55–​69 Nitrate levels in public water Thyroid Average nitrate quartiles (> 2.46 vs Dietary nitrate intake quartiles
United States Incidence, 1986–​2004 supplies (1955–​1988) and private < 0.36 mg/​L NO3-​N) HR = 2.18 positively associated with risk
Iowa well use among women > 10 years (CI: 0.83, 5.76; p-​trend = 0.02) (p-​trend = 0.046)
at residence (21,977 women; 40 Years > 5 mg/​L (> 5 years vs.
thyroid cases); no measurements 0) HR = 2.59 (CI: 1.09, 6.19;
for private wells p-​trend = 0.04);
private well (vs. < 0.36 mg/​L NO3-​N)
HR = 1.13 (CI: 0.83, 3.66)
Inoue-​Choi (2012) Cohort of women ages 55–​69 Nitrate levels in public water Breast Average nitrate quintiles (> 3.8 vs. Interaction with vitamin C and
United States Incidence, 1986–​2008 supplies (1955–​1988) and private < 0.32 mg/​L NO3-​N) HR = 1.14 smoking not evaluated
Iowa well use among women > 10 years (CI: 0.95, 1.36; p-​trend = 0.11);
at residence (20,147 women; 1751 private well (vs. < 0.32 mg/​L
breast cases); no measurements for NO3-​N) HR = 1.14 (CI: 0.97, 1.34)
private wells Subgroup with folate > 400 µg/​
d: (> 3.8 vs. < 0.32 mg/​L NO3-​
N) HR = 1.40 (CI: 1.05, 1.87;
p-​trend = 0.04); private well (vs.
< 0.32 mg/​L NO3-​N) HR = 1.38
(CI: 1.05, 1.82)
No association among those with low
folate < 400 µg/​d
Inoue-​Choi (2015) Cohort of women ages 55–​69 Nitrate levels in public water Ovary Highest vs. lowest quartile average (> Associations with average water
United States Incidence, 1986–​2010 supplies (1955–​1988) and private 2.98 mg/​L vs. < 0.47 mg/​L NO3-​ nitrate and private well use
Iowa well use among women > 10 years N) HR = 2.03 (CI: 1.22, 3.38; p-​ were stronger among women
at residence with nitrate and trend = 0.003), adjusted for TTHM; with < median vitamin C intake
trihalomethane estimates (17,216 years > 5 mg/​L (> 4 years vs. (average nitrate p-​trend = 0.005;
women; 190 ovarian cases); no 0) HR = 1.52 (CI: 1.00, 2.31; p-​ p-​interaction = 0.33; private
measurements for private wells trend = 0.05), adjusted for TTHM well p-​interaction = 0.01)
Adjusted for total trihalomethanes Private well users (vs. < 0.47 mg/​L No interaction with red meat
(TTHM) (1955–​1988), measured NO3-​N) HR = 1.53 (CI: 0.93, 2.54) intake
TTHM levels in 1980s, prior years
estimated by expert)
Espejo-​Herrera (2015) Hospital-​based case-​control Nitrate levels in public supplies Bladder Average level (age 18-​interview) No interaction with vitamin C,
Spain Incidence, 1998–​2001 (1979–​2010) and bottled water > 2.26 vs. 1.13 mg/​L NO3-​N E, red meat, processed meat,
Asturias, Alicante, Barcelona, (measurements of brands with OR = 1.04 (CI: 0.60, 1.81) average THM level
Valles-​Bages, Tenerife highest consumption based on a Years > 2.15 mg/​L NO3-​N (75th
provinces Spanish survey); analyses limited percentile): > 20 vs. 0 years
to those with > 70% of residential OR = 1.41 (CI: 0.89, 2.24)
history with nitrate estimate (531
cases, 556 controls)
Jones (2016) Cohort of women ages 55–​69 Nitrate levels in public water Bladder Highest vs. lowest quartile average Significant interaction with
United States Incidence, 1986–​2010 supplies (1955–​1988) and private (> 2.98 vs. < 0.47 mg/​L NO3-​N) smoking: Current smokers
Iowa well use among women > 10 years HR = 1.47 (CI: 0.91, 2.38; p-​ with > 2.98 mg/​L NO3-​N vs.
at residence with nitrate and trend = 0.11), adjusted for average non-​smokers < 0.47 mg/​L
trihalomethane estimates (20,945 TTHM levels NO3-​N HR = 3.67 (CI: 1.43,
women; 170 bladder cases); no Years > 5 mg/​L (> 4 years vs. 9.38); p-​interaction = 0.03;
measurements for private wells 0) HR = 1.61 (CI: 1.05, 2.47; p-​ no significant interaction with
Adjusted for total trihalomethanes trend = 0.03), adjusted for average vitamin C, TTHM levels
(TTHM) (1955–​1988), measured TTHM levels
TTHM levels in 1980s, prior years Private well users (vs. < 0.47 mg/​L
estimated by expert) NO3-​N) HR = 1.53 (CI: 0.93, 2.54)
 317
Espejo-​Herrera (2016) Pooled case-​control studies Nitrate levels in public supplies Colorectal cancer Water nitrate intake based on average Stronger associations for men and
Spain, Italy Incidence, 2008–​2013 (2004–​2010) for 349 water nitrate levels (estimated 30 to among those with high red meat
Spain (9 provinces) and supply zones, bottled water 2 years prior to interview) and intake; no significant interaction
population-​based controls; Italy (measurements of brands with water intake (L/​day) with red meat, vitamin C, E,
(two provinces) and hospital-​ highest consumption based on > 2.3 vs. < 1.1 mg /​day NO3-​N fiber
based controls surveys in Spain and Italy), OR = 1.49 (CI: 1.24, 1.78), adjusted
and private wells and springs for diet/​other colorectal cancer risk
(measurements in 2013 in 21 factors; colon OR = 1.52 (1.24,
municipalities of León, Spain, 1.86), rectum OR = 1.62 (1.23,
the area with highest non-​public 2.14)
water use).
Analyses include those with nitrate
estimates for > 70% of period
30 years before interview (1869
cases, 3530 controls)
Espejo-​Herrera (2016) Hospital-​based case-​control Nitrate levels in public supplies Breast Water nitrate intake based on average Water nitrate intake estimated
Spain Incidence, 2008–​2013 (2004–​2010), bottled water nitrate levels (age 18 to 2 years from age 18 to 30 and from
Spain (8 provinces) (measurements of brands with prior to interview) and water 15 to 2 years before interview
highest consumption based on a intake (L/​day). Post-​menopausal showed similar results.
Spanish survey), and private wells women: > 2.0 vs. 0.5 mg/​day Stronger associations among
and springs (measurements in NO3-​N OR = 1.32 (0.93, 1.86); postmenopausal women with
2013 in 21 municipalities of León, Premenopausal women: > 1.4 vs. high red or processed meat
Spain, the area with highest non-​ 0.4 mg/​day NO3-​N OR = 1.14 intake; no significant interaction
public water use). (0.67, 1.94) with red meat, vitamin C, E,
Analyses include women with > 70% smoking
of period from age 18 to 2 years
before interview (1245 cases, 1520
controls)
Jones (2017) Cohort of women ages 55–​69 Nitrate levels in public water Kidney 95th percentile vs. lowest quartile of No significant interaction with
United States Incidence, 1986–​2010 supplies (1955–​1988) and private average nitrate level (> 5.00 vs. smoking, vitamins C or E
Iowa well use among women > 10 years < 0.47 mg/​L NO3-​N) HR = 2.23
at residence with nitrate and (CI: 1.19, 4.17; p-​trend = 0.35),
trihalomethane estimates (20,945 adjusted for TTHM
women; 163 kidney cases); no Years >5 mg/​L (> 4 years vs.
measurements for private wells 0) HR = 1.54 (CI; 0.97, 2.44; p-​
Adjusted for total trihalomethanes trend = 0.09), adjusted for THM
(TTHM) (1955–​1988), measured Private well users (vs. < 0.47 mg/​L
levels in 1980s, prior year levels NO3-​N) HR = 0.96 (CI: 0.59, 1.58)
estimated by expert)

ND: not detected
a
Nitrate or nitrite levels presented in the publications as mg/​L of the ion were converted to mg/​L as NO3-​N or NO2-​N
b
Odds ratios (OR) for case-​control studies, incidence rate ratios (RR), and hazard ratios (HR) for cohort studies, and 95% confidence intervals (CI).
318

318 PART III:  THE CAUSES OF CANCER


cancer risk was not associated with water nitrate levels with follow-​up no significant interactions with red meat, vitamins C and E, and fiber
through 2008 (Inoue-​Choi et  al., 2012). Among women with folate intakes. Postmenopausal and premenopausal breast cancers were not
intake ≥ 400 μg/​day, risk was significantly increased for those in the associated with water nitrate ingestion in a hospital-​based case-​control
highest average nitrate quintile (HR = 1.40; 95% CI: 1.05, 1.87) and study in Spain (Espejo-​Herrera et al., 2016b).
among private well users (HR = 1.38; 95% CI: 1.05, 1.82), compared Animal studies demonstrate that in utero exposure to nitrosamides
to those with the lowest average nitrate quintile. There was no asso- can cause brain tumors in the exposed offspring. Water intake during
ciation with nitrate exposure among women with lower folate intake. pregnancy was estimated in a multicenter case-​control study of child-
Analyses of effect modification by other dietary factors were not pre- hood brain tumors in five countries based on the maternal residen-
sented. With follow-​up through 2010, there were 130 bladder cancer tial water source (Mueller et al., 2004). Nitrate/​nitrite levels in water
cases among women who had used publicly supplied water > 10 years. supplies were measured using a dipstick method for a subset of the
Risk remained elevated among women with the highest average nitrate women; however, most of these measurements occurred many years
levels and was significantly increased among women whose drinking after the pregnancy. Drinking water from private wells versus public
water concentration exceeded 5 mg/​l NO3-​N for at least 4 years (Jones water supplies was not consistently associated with risk of childhood
et al., 2016). Risk estimates were not changed by adjustment for THM brain tumors. However, higher nitrite levels (> 1.5 mg/​l nitrite-​N) in
levels. Smoking, but not vitamin C intake, modified the association the drinking water were associated with significantly increased risk of
with nitrate in water; increased risk was apparent only in current smok- childhood brain tumors, especially astroglial tumors.
ers. Dietary nitrite intake was not associated with risk. With follow-​
up through 2010, there were 125 kidney cancer cases among women
in the public water supply analysis; risk was significantly increased Biomarkers of Genetic Damage
among those in the 95th percentile of average nitrate (> 5.0 mg/​L
NO3-​N) compared with the lowest quartile (HR = 2.2; 95% CI: 1.2, Two cross-​sectional studies of the genotoxic effects of nitrate in drink-
4.2). There was no positive trend with the average nitrate level and no ing water included individuals drinking well water with nitrate con-
increased risk for women using private wells, compared to those with centrations ranging from 11 to 65 mg/​l as NO3-​N. The first study found
low average nitrate in their public water supply (Jones et al., 2016). no increase in the frequency of peripheral lymphocyte sister chromatid
Higher risk was evident in the 95th percentile of dietary nitrite intake exchanges with increasing levels of nitrate (Kleinjans et  al., 1991).
from processed meats but with no significant trend. A  subsequent study employed the hypoxanthine-​guanine phosphori-
In contrast to the positive findings for bladder cancer among the bosyltransferase (HPRT) variant frequency test in peripheral lympho-
cohort of Iowa women, a cohort study of men and women aged 55–​69 cytes (van Maanen et  al., 1996). An increased prevalence of HPRT
in the Netherlands with lower nitrate levels in public supplies found variants in subjects drinking medium and high levels of nitrate was
no association between water nitrate ingestion (median in top quin- observed. An inverse correlation between the labeling index in lym-
tile = 2.4 mg/​day NO3-​N) and bladder cancer risk (Zeegers et al., 2006). phocytes and nitrate exposure was suggestive of an exposure-​related
Dietary intake of vitamins C and E and history of cigarette smoking immunosuppressive effect. Future studies examining the genotoxic
did not modify this observation. A hospital-​based case-​control study potential of nitrate in drinking water can make valuable contributions
of bladder cancer in multiple areas of Spain (Espejo-​Herrera et  al., to understanding the adverse effects of nitrate exposure and warrant
2015) assessed lifetime water sources and usual intake of tap water. further exploration.
Nitrate levels in public water supplies were low, with almost all aver-
age levels below 2 mg/​l NO3-​N. Risk of bladder cancer was not associ-
ated with the nitrate level in drinking water or with estimated nitrate CYANOBACTERIAL TOXINS
ingestion, and there was no evidence of interaction with factors affect-
ing endogenous nitrosation. Cyanobacteria (formerly known as blue-​green algae) are an ancient and
Several case-​control studies conducted in the Midwestern United ubiquitous family of prokaryotic organisms, with fossil remains dat-
States obtained lifetime histories of drinking water sources and esti- ing back ~3.5 billion years. Photosynthesis by these bacteria is likely
mated exposure for users of public water supplies. In contrast to responsible for earth’s oxygen-​enriched atmosphere, and subsequent
findings of an increased risk of NHL associated with higher average evolution of higher organisms (Olson, 2006; Paerl et al., 2011; Schopf,
nitrate levels in Nebraska public water supplies in an earlier study 2000). Cyanobacteria from more than 40 genera produce a wide vari-
(Ward et al., 1996), there was no association with similar concentra- ety of toxins (cyanotoxins). Among these are hepato-​, nephro-​, neuro-​,
tions in public water sources in a case-​control study of NHL in Iowa and dermatotoxins belonging to several chemical classes (Chorus and
(Ward et  al., 2006). A  study of renal cell carcinoma in Iowa (Ward Bartram, 1999). Adverse human health effects range from minor skin
et  al., 2007)  found no association with the level of nitrate in public irritation to death from severe liver damage and possibly amyotrophic
water supplies, including the number of years that the supply exceeded lateral sclerosis (Cox and Sacks, 2002).
5 or 10 mg/​l NO3-​N. Higher nitrate levels in public water supplies Evidence for carcinogenicity is strongest for microcystins (MCs),
increased risk among subgroups that reported above the median intake nodularin, and cylindrospermopsin (Zegura et al., 2011). Microcystins,
of red meat intake or below the median intake of vitamin C, however. usually the most common toxins found in fresh water, are cyclic
These interactions were statistically significant. A small case-​control peptides containing seven amino acids, with two variable positions.
study of adenocarcinoma of the stomach and esophagus among men There are at least 80 analogs. Microcystin-​LR, the most thoroughly
and women in Nebraska (Ward et  al., 1997)  estimated nitrate levels studied, contains lysine (L) and arginine (R) in the variable positions
among long-​term users of public water supplies and found no asso- (Butler, 2009).
ciation between average nitrate levels and risk. A case-​control study Epidemiologic evidence implicating cyanotoxins (particularly
among rural women in Wisconsin estimated nitrate levels in private microcystin-​LR) as human carcinogens comes largely from southeast-
wells using spatial interpolation of nitrate concentrations from a 1994 ern coastal China. Here, elevated rates of hepatocellular carcinoma
water quality survey and found a significant increased risk of proximal (HCC) have long been recognized. The main causes are known to be
colon cancer among women estimated to have nitrate levels > 10 mg/​l endemic infection with hepatitis B (and more recently C) viruses, in
NO3-​N compared to levels < 0.5 mg/​l. Risks of distal colon cancer and combination with consumption of grain contaminated by aflatoxin.
rectal cancer were not associated with the nitrate level (McElroy et al., An additional cofactor, however, may be consumption of surface
2008). Water nitrate ingestion from public supplies, bottled water, water contaminated by cyanotoxins. Many studies in this region have
and private wells and springs over the adult lifetime was estimated in reported much higher risk of HCC among populations that regularly
analyses that pooled case-​control studies of colorectal cancer in Spain used surface waters (largely pond, ditch, or river water) as their pri-
and Italy (Espejo-​Herrera et  al., 2016a). Risk of colorectal cancer mary water source, compared to those who consumed water from
was significantly increased among those with > 2.3 mg/​day NO3-​N shallow or deep wells (Shen et al., 1985; Su, 1979; Yu, 1989; Zhang,
(vs. < 1.1 mg/​day); risks were stronger among men, but there were 1993). For example, in the period 1972–​1981 in Qidong County, Shen
 319

Water Contaminants 319


et al. (1985) observed annual incidence rates of HCC of 141.4, 72.3, Working Group classified microcystin-​LR as “possibly carcinogenic
43.5, 22.3, and 11.7 per 100,000 for water consumers of house ditch, to humans,” microcystis extracts as “not classifiable as to their car-
field ditch, river, shallow wells, and deep wells, respectively. Among cinogenicity to humans,” and nodularin also as “not classifiable”
99 HCC cases and 99 matched controls in Guangxi Autonomous (IARC, 2010).
Region, Zhang et al. (1993) found an odds ratio of 3.7 (95% CI: 1.3, A limited number of studies have evaluated the association between
11.0) for consuming pond/​ditch water, compared with well water. In cyanobacteria and liver cancer in other populations. In central Serbia,
a review, Yu (1989) reported several studies of incidence and mortal- more than 80% of drinking water reservoirs have experienced cya-
ity from HCC conducted in Qidong, Nanhue, and Haimen counties in nobacterial blooms over the past 80 years, many with measureable
1973–​1983. These showed a consistent pattern of higher incidence and levels of microcystin-​LR (Svircev et al., 2013). The mean incidence
mortality rates among those who drank water from ponds and ditches of HCC was elevated in three Serbian districts where cyanobacterial
than among those who only used wells. For example, the standardized blooms occurred annually for the past 25 years. The incidence rate
incidence ratios (SIR) in Qidong were 2.6 and 1.4 per 100,000 for of HCC in these districts averaged 27/​100,000 in the period 1999–​
pond and ditch water users, and 0.34 for shallow well water consum- 2008, nearly four times the rate (7.2/​100,000) in another region of the
ers. Variability in hepatitis virus infection or aflatoxin (Yu, 1989) in the country that uses groundwater. The geographic distribution of other
affected populations did not explain the risk differences. In a public risk factors (hepatitis B virus, hepatitis C virus, and liver cirrhosis,
health intervention (1973–​1978), the population was encouraged to as measured by mortality), did not explain the localized increases in
shift drinking water source from surface to wells, resulting in substan- risk. An ecologic study in Florida, which evaluated HCC incidence
tially decreased HCC risks (Su, 1979). among people consuming publically-​supplied surface waters with a
Prior to 1990, researchers suspected that chemical contaminants in history of contamination by microcystins, relative to those who used
surface waters in the endemic region probably accounted for the asso- uncontaminated groundwater sources, was equivocal (Fleming et al.,
ciation between liver cancer and drinking surface water in the endemic 2002). Microcystin-​LR and nodularin are potent inhibitors of protein
areas. Pesticides and other agricultural chemicals were considered phosphatases 1 and 2A; they belong to the okadaic acid class of tumor
strong candidates. Empirical evidence for their involvement was lack- promotors (Fujiki and Suganuma, 1994, 2011). Microcystin-​LR upreg-
ing, however. ulates production of tumor necrosis factor α gene (TNF-​α), which has
Extracts from cyanobacteria have long been known to be highly tumor-​promoting activity, as well as affecting early-​response genes
toxic to humans (Schwimmer and Schwimmer, 1955). In particular, such as c-​jun, jun B, jun D, c-​fos, fos B, and fra-​1 (Sueoka et al., 1997).
gastroenteritis, hepatotoxicity, and occasionally death from these The World Health Organization in 1998 first recommended a pro-
toxins have been observed (Chorus and Bartram, 1999; Griffiths visional limit for microcystin-​LR in drinking water of 1 µg/​l (WHO,
and Saker, 2003; Jochimsen et  al., 1998). Experimental demonstra- 2011). Many countries have accepted this level and have issued recom-
tion of skin tumor–​promoting activity in mice that ingest extracts of mendations for its implementation (Burch, 2008). A Health Advisory
Microcystis aeruginosa (common in freshwater) was reported in 1989 issued by the US EPA calls for a limit of 0.3 µg/​l total microcystin
(Falconer and Buckley, 1989), followed by experimental results show- for infants and preschool children and 1.6 µg/​l for older children and
ing that microcystin-​LR specifically promotes liver tumors in rodents adults (US EPA, 2015). Few countries have issued legally binding lim-
(Fujiki and Suganuma, 2011; Lian et al., 2006; Nishiwaki-​Matsushima its (Burch, 2008).
et al., 1992). Cyanobacteria proliferate rapidly in fresh and marine waters under
Field results from the endemic HCC region of China have demon- suitable environmental conditions. So-​ called “water blooms” are
strated the presence of microcystin in surface water. A series of 989 exacerbated by phosphorus and nitrogen runoff from agricultural and
water samples, tested with an enzyme-​linked immunosorbent assay municipal sources, and by warming of lakes and other drinking water
(ELISA) (detection limit of 50 pg/​ml) revealed that 17% of pond/​ditch sources driven by the changing global climate (Elliott, 2012; Heisler
water samples (average = 101 pg/​ml), 32% of river water samples (160 et al., 2008; Newcombe et al., 2012; O’Reilly et al., 2015; Paerl and
pg/​ml), and 4% of shallow well water samples (68 pg/​ml) were posi- Paul, 2012). The public health implications of these combined factors
tive for microcystin. No microcystin was detected in deep well water deserves further study.
(Ueno et al., 1996). Environmental surveys and epidemiologic studies
of HCC incidence or mortality from this era further implicated micro-
cystin as a risk factor for HCC (Chen and Kensler, 2014; Harada et al.,
COMMUNITIES EXPOSED TO INDUSTRIAL CHEMICALS
1996; Ling, 2000; Yu et al., 2001; Yu and Chen, 1994). Adding to this
evidence is statistical modeling of HCC case-​control data that showed IN DRINKING WATER
a multiplicative interaction between consumption of water from ponds
or ditches and infection with hepatitis B virus and aflatoxin ingestion Here, we describe three settings in which elevated cancer risks have
(Zhao et al., 1994), consistent with the promotional effects of MC-​LR been observed in communities exposed to water contaminated by
seen experimentally. Most of the risk from consumption of microcys- industrial chemicals. Such studies are intrinsically opportunistic. In
tin results from its joint effect with hepatitis B infection. Compared one instance (Toms River, NJ), elevated rates of childhood cancers
to groundwater users negative for hepatitis B antibody (HBab) (OR were observed. The exposure involved poorly defined mixtures and
[ref] = 1.00), the OR estimate among subjects who were negative for did not allow the attribution of risk to specific chemical(s). In the other
HBAb but consumed pond/​ditch water was 1.74; among subjects posi- two scenarios, the predominant chemical exposures were known and
tive for HBAb who consumed groundwater, the OR was 6.57, and observations of elevated risk of specific types of cancer were consis-
among subjects who were both HBab positive and consumed ditch tent with occupational exposures and/​or experimental findings.
water, the OR was 11.42.
The finding that high-​risk populations consumed water contami-
Camp Lejeune
nated with microcystin, and that this interacted with hepatitis virus
infection to cause HCC, suggests that microcystin is an important risk At Camp Lejeune, a US Marine Corps Base Camp in coastal North
factor for HCC. Individual-​level data on dose–​response relationships Carolina, two of eight drinking water systems that served most of
are lacking, however. Collecting such information would be difficult, the base’s population (including family members of Marines) were
given the poor predictability and temporal variability of microcystin contaminated by solvents from the 1950s through 1985, as reported
blooms and the long induction period for liver cancer. Further research by the US ATSDR (2016). One supply was contaminated by an off-​
on whether microcystin interacts with other HCC risk factors would base dry cleaner, primarily with perchloroethylene (PCE) (tetrachlo-
be valuable. roethylene), reaching a maximum measured level of 215 ug/​l; other
Studies in China have also found an association between likely contaminants included trichloroethylene (TCE), trans-​1,2-​dichloro-
exposure to microcystin in drinking water and elevated risk of colorec- ethylene, and VC, a degradation product of PCE. Another affected
tal cancer (Chen et  al., 2003; Zhou et  al., 2002). In 2006, an IARC water supply was contaminated by sources located on the base
320

320 PART III:  THE CAUSES OF CANCER


involving industrial spills, waste disposal sites, and leaking under- water from private wells and public water supply wells in six nearby
ground storage tanks. The primary contaminant was TCE (maximum districts. Environmental fate and transport modeling revealed that
detected level of 1400 ug/​l); levels of PCE were lower (maximum extensive contamination of groundwater occurred via deposition of
of 100 ug/​l), and benzene was detectable. A third water supply that airborne PFOA from plant stacks and subsequent transport to the aqui-
served 2100 family housing units was not contaminated. The US fer (Shin et al., 2011b). There were also direct releases into the Ohio
ATSDR conducted a detailed assessment of exposure and estimated River. A  class-​action suit brought by local residents alleging health
historical average monthly levels of TCE, PCE, VC, and benzene damages was resolved by a pretrial settlement agreement calling for
exposure in water provided to work areas and housing units on the DuPont to fund epidemiologic studies in the surrounding communities
base (Maslia et al., 2007, 2013). and among plant workers (Frisbee et al., 2009). This was implemented
Two retrospective cohort studies evaluated the risk of total and by the “C8 Science Panel,” composed of three senior chronic disease
site-​specific cancer mortality. One cohort was comprised of Marines epidemiologists who designed and oversaw the studies, reviewed data,
and Navy personnel who began service in 1975–​1985 (N = 154,932); and reported to the court whether selected adverse health outcomes
another included civilian personnel employed at the base during 1973–​ were “more probably than not” caused by exposure to PFOA. This
1985 (N = 4,647). Follow-​up of both cohorts began in 1979 and ended criterion is a legal rather than a scientific concept. Any positive results
in 2008 (Bove et al., 2014a, 2014b) The primary comparison was with reported would require DuPont to support continuing medical moni-
uniformed personnel (N = 154,969) and civilian employees (N = 4690), toring for the relevant condition(s).
respectively, at Camp Pendleton, a Marine base of similar size and Blood serum measurements of PFOA concentrations by the C8
demographic composition located in California, with no known his- Health Project in 2005–​2006 revealed an overall median concen-
tory of comparable drinking water contamination. Among Marines and tration of 28.2 ng/​ml in this population, compared with 4 ng/​ml in
Navy personnel, mortality due to all cancers combined was elevated the United States overall, as measured in the National Health and
(hazard ratio [HR] = 1.10; 95% CI: 1.00, 1.20). Hazard ratio estimates Nutrition Examination Survey (Steenland et al., 2009). A number
were also increased for cancer sites of “primary interest” (based on of studies were conducted to evaluate the risk of 55 health out-
occupational studies or experimental evidence), although these results comes potentially associated with PFOA. These studies included
were not statistically significant. These included kidney cancer (HR = individual exposure assessment in a community (N  =  43,449)
1.35; CI: 0.84, 2.16), liver cancer (HR = 1.42; CI: 0.92, 2.20), esopha- (Shin et  al., 2011a, 2013), a survey (including personal medical
geal cancer (HR = 1.43; CI: 0.85, 2.38), cervical cancer (HR = 1.33; CI: information and background) of PFOA concentrations in blood
0.24, 7.32), Hodgkin lymphoma (HR = 1.47; CI: 0.71, 3.06), and multi- (serum) and drinking water in the six major contaminated water
ple myeloma (HR = 1.68; CI: 0.76, 3.72). There was an increasing trend districts (Winquist et  al., 2013), a geographic analysis of cancer
of risk of kidney cancer mortality with total contaminant level, and of incidence in the region (Vieira et  al., 2013), a cohort mortality
Hodgkin lymphoma with TCE and with benzene. Increases in risk (not study of exposed workers (Steenland and Woskie, 2012), and a
statistically significant) were also found among the civilian employees study of incident cancers among adults (Barry et  al., 2013). The
for mortality from all cancer combined (HR = 1.12; CI: 0.92, 1.36), cancer incidence study included 32,254 adult community resi-
kidney cancer (HR = 1.92; CI: 0.58, 6.34) and multiple myeloma (HR dents or plant workers, most of whom had participated in a 2005–​
= 1.84; CI: 0.45, 7.58). A study of incident cancer is underway (2017). 2006 baseline survey with serum PFOA measurements. Yearly
A separate case-​control study measured mortality from 13 hematopoi- serum concentrations were estimated for each person from 1952
etic cancers among children born in the interval 1968–​1985 to moth- to 2011 (Shin et  al., 2011a). Subsequent interviews were held in
ers with household exposure to drinking water at Camp Lejeune. Odds 2008–​2011 to assess health problems (later confirmed by medical
ratio estimates of 1.6 (CI: 0.5, 4.8) and 1.6 (CI: 0.5, 4.7) were observed records and/​or cancer registry information). Estimated cumulative
for first trimester exposure to any PCE or any VC, respectively (Ruckart serum PFOA concentrations were positively associated with kid-
et al., 2013). Another small case-​control study, of 71 male breast cancer ney and testicular cancer. The HR estimates for kidney cancer with
cases identified from the Department of Veterans Affairs cancer regis- increasing exposure quartiles were 1.0, 1.23, 1.48, and 1.58 (linear
try, found an adjusted OR of 1.14 (CI: 0.65, 1.97) for ever having been trend test p  =  0.18); those for testicular cancer were 1.0, 1.04,
stationed at Camp Lejeune (Ruckart et al., 2015). 1.91, and 3.17 (trend p = 0.04).
As mentioned, the 95% CI associated with most cancer sites in Based on findings from this study and the literature, the C8 Science
the Camp Lejeune studies did not exclude the null hypothesis. When Panel concluded that “there is a probable link between PFOA and both
viewed in isolation, these investigations do not provide evidence of testicular and kidney cancer” (C8 Science Panel, 2012). The Panel also
causal associations for the contaminants involved. However, the concluded, “there is no probable link between PFOA and either thy-
observed risks are consistent with positive epidemiologic evidence roid cancer or melanoma, for which limited but insufficient evidence
from more highly exposed occupational populations, where the expo- was found.” A working group of the IARC classified PFOA as “pos-
sure levels are better documented, and there are mechanistic and/​or sibly carcinogenic to humans,” based primarily on these epidemiologic
experimental data that support a causal relationship. Based on the findings (IARC, 2016). The epidemiologic evaluation of the public
overall evidence, primarily from occupational settings, an IARC work- health impact of widespread exposure to PFOA played a central role
ing group classified TCE, the contaminant found at highest levels at in responding to the health concerns of the community and in estab-
Camp Lejeune, as “carcinogenic to humans” (IARC, 2014). The US lishing possible human carcinogenicity, consistent with laboratory
EPA has come to a similar conclusion (Chiu et  al., 2013), as has a observations that it causes cancer of the testicles, liver, and pancreas in
draft report from the US National Toxicology Program (National rodents (Kennedy, 2015).
Toxicology Program, 2015). Vinyl chloride and benzene have been
classified as human carcinogens (IARC, 2012b), and PCE was evalu-
ated by an IARC working group that designated it a “probable” human Toms River, New Jersey
carcinogen (IARC, 2014).
Responding to residents of Toms River, New Jersey, who reported an
unusual number of childhood cancers, the New Jersey Department
Community Exposed to Perfluorooctanoic Acid of Health and Senior Services (NJDHSS), in collaboration with the
(PFOA) and Related Compounds U.S. Agency for Toxic Substances and Disease Registry (ATSDR)
and community organizations, undertook several studies (Maslia
Perfluorooctanoic acid (PFOA or C8) has been used historically in et  al., 2005; NJ Department of Health, 2016). A 1997 analysis of
the production of Teflon, Gore-​Tex, non-​
stick cookware surfaces, cancer incidence in Dover Township, the location of Toms River,
and other products. Manufacturing using PFOA began at the DuPont found a significant elevation in the incidence of all childhood can-
Washington Works on the Ohio-​West Virginia border in 1951. Nearby cers combined (90 observed, 67 expected) for the years 1979–​1995,
populations were exposed, primarily through contaminated drinking based on the New Jersey State cancer registry (Berry, 1997). None
 321

Water Contaminants 321


of the other 32 townships in Ocean County had a significantly particles, metals and metalloids other than arsenic, and water hardness,
elevated rate. The risk of pediatric cancers was especially elevated the epidemiologic evidence at this time is limited and equivocal.
in the Toms River census tracts. Overall, 24 cases were observed,
with 14.1 expected. In females under age 5 years, 10 cases were
observed, with 1.6 expected. The NJDHSS subsequently conducted CLIMATE CHANGE
two case-​control studies of leukemia and neurological cancers, one
based on an extensive interview of parents and another based on The capacity of the global atmosphere to hold water increases by about
birth certificates. These used 4 and 10 controls, respectively, per 7% per 1oC warming, leading to increasing water vapor in the air and
case, matched on age and gender. more intense precipitation events (Trenberth, 2011). By the year 2015,
An important element of these studies involved detailed model- average annual global land surface temperature had increased by
ing by ATSDR of the water distribution system in order to generate 0.90oC (1.62oF) above the average for the twentieth century (National
month-​by-​month estimates of potential individual residential expo- Oceanic & Atmospheric Administration [NOAA], 2015). Climate
sures during the time period 1962–​1996 for each of several public well change is associated with major perturbations in the hydrologic cycle,
water sources. Two sources were contaminated by chemicals from and therefore in the quantity and quality of water available for human
separate Superfund sites (Maslia et al., 2001). A former Ciba Chemical consumption. With respect to its possible effects on carcinogenic water
Corporation (later, Ciba-​Geigy) plant opened in 1952 and produced contaminants, research is currently focused in two areas: (1) increases
organic dyes and intermediate products, epoxy resins, and specialty in cyanobacterial growth patterns, and (2) elevated levels of disinfec-
chemicals. Until 1966, the plant discharged treated wastewater directly tion byproducts. The first is occurring in response to elevated tempera-
into the Toms River. After 1966, the wastewater was pumped via a 10-​ tures of freshwater bodies and the release of plant nutrients, especially
mile pipeline into the Atlantic Ocean. The pipeline experienced four nitrate and phosphate, from agricultural regions (Elliott, 2012; Heisler
major breaks over time, causing local chemical contamination. Tens et al., 2008; Newcombe et al., 2012; O’Reilly et al., 2015; Paerl and
of thousands of drums with solid and liquid chemical wastes were dis- Paul, 2012). Increasing levels of disinfection byproducts may result
posed on site, resulting in extensive soil and groundwater contami- from the mobilization of precursor humic and fulvic acids from soils
nation. A down-​gradient well field of the local water supply (Holly during more intense rainfall events (Delpla et al., 2016). The design of
Street) was contaminated with dyes, nitrobenzene, and other com- most existing water treatment plants predates concerns about climate
pounds during the mid-​1960s. The other Superfund site is the Reich change. Increasing attention is now directed on enhancing these facili-
Farm, where, for 4 months in 1971, over 4500 drums of toxic waste ties to cope with changes in water quantity and quality due to both high
from a Union Carbide plant were dumped illegally. These leaked into rainfall and, conversely, water shortages (Levine et al., 2016).
the sandy subsoil, forming a plume that contaminated another public
supply well field (Parkway) (Fagin, 2013). A modeling study of public
drinking water reconstructed the monthly well-​field origins of water EFFECTS OF EXPOSURE MISCLASSIFICATION
provided to each residence during the relevant time period (Maslia IN EPIDEMIOLOGIC STUDIES
et al., 2000, 2001). However, the residential exposures were estimated
after extensive remediation, when most chemical contaminants were The degree to which risk estimates from epidemiologic studies of
no longer detectable, obviating the possibility of linking specific cancer reflect “true” risks depends on the accuracy of the exposure
chemicals or mixtures with modeled exposures from individual well estimates during the relevant exposure period, commonly measured in
fields. This shortcoming in exposure assessment, and the relatively decades prior to appearance of a tumor. Relatively small errors in esti-
small numbers of cases, limited the utility of the case control studies. mating historical exposure can have profound effects on the observed
Air pollution from the Ciba-​Geigy plant was also modeled and used in risk. When a true risk is present and the misclassification of expo-
case-​control data analyses. sure in case-​control studies is non-​differential (similar among cases
Parents of 40 children diagnosed with leukemia or nervous system and non-​cases), the risk estimate is typically biased toward the null
cancer while residing in Dover Township in 1979–​1996 participated (Cantor and Lubin, 2007). For example, given a “true” relative risk of
in the interview study, as did 159 matched controls. Birth mothers and 2.0 (an actual 100% risk increase), the observed relative risk would be
fathers were interviewed by telephone, providing information on resi- decreased to 1.5 (an observed 50% increase) if the correlation coef-
dence location, volume of water consumed, parents’ occupations, and ficient between the actual exposure and the estimated exposure from
several other factors. The focus of most statistical analyses was the the study were 0.6 (Vineis, 2004). This level of exposure misclassi-
relative level of consumption of water originating in the contaminated fication is common in studies of environmental factors and leads to
Holly Street or Parkway well fields. No consistent associations were special concern in low-​exposure settings, where expected excess risks
observed with postnatal exposures (Fagliano et  al., 2003). Prenatal are relatively small and the error in exposure estimates can lead to
exposure to water from the Parkway well field in the period 1982–​ erroneously missing important risks. Careful consideration of this is
1996 was associated with leukemia among female children of all ages warranted when evaluating the findings of epidemiologic studies of
(OR = 3.4; CI:1.1, 10.4). Findings among male children were essen- lower levels of arsenic in drinking water (< 100 ug/​l) and for all studies
tially null. The study, albeit small, was well designed and was sup- of disinfection byproducts and of nitrate in drinking water. A corol-
ported by an exposure assessment that was extensive and detailed, but lary is that when an association between cancer risk and a waterborne
that lacked important information on specific chemical contaminants. exposure is consistently observed among different populations and
Cancer risk was not measured in adults in Toms River who consumed varying circumstances (e.g., bladder cancer and DBP), quantitative
contaminated water. The complex story of the origin and findings of estimates of risk almost certainly underestimate the true risk. When
these studies conducted by NJDHSS and ATSDR and the historical, planning future studies, careful assessment of statistical power issues,
social, legal, and political context of water contamination in Toms in the face of unavoidable errors in historical exposure assessment,
River are described by Fagin (2013). must be conducted.

OTHER WATERBORNE EXPOSURES FUTURE DIRECTIONS

Classes of waterborne carcinogens not described here are addressed


elsewhere in the current or previous edition of this text (Cantor et al., Inorganic Arsenic
2006). Schistosomiasis is an established infectious cause of bladder Inorganic arsenic is an established human carcinogen, with most evi-
cancer (Chapters 24 and 52); radionuclides such as radon and radium dence regarding ingested exposure from populations with prolonged
226 and 228 are naturally occurring contaminants of drinking water consumption of drinking water at concentrations well over 100 µg/​l. In
(Chapter 13). For some other waterborne factors, such as asbestiform the observed range, the dose–​response curve appears to be linear. An
32

322 PART III:  THE CAUSES OF CANCER


unanswered question concerns the level of risk and shape of the dose–​ References
response curve when exposures are below 100 µg/​l. Addressing this Adonis MI, Diaz J, Miranda VR, et al. 2014. Biomarkers for screening of lung
issue is fraught with many methodologic challenges. The most feasible cancer and pre-​neoplastic lesions in a high risk Chilean population. Biol
approach would be to evaluate risk in populations with exposure in the Res, 47(1), 62. PMCID: PMC4335787.
50–​100 µg/​l range, if such groups could be identified. Other research Aggazzotti G, Fantuzzi G, Tartoni PL, and Predieri G. 1990. Plasma chloro-
questions concern the mechanism(s) of action and the role of genetic form concentrations in swimmers using indoor swimming pools. Arch
polymorphisms in arsenic metabolism and cancer risk. Better under- Environ Health, 45(3), 175–​179. PMID: 2386423.
standing of the mechanism(s) will likely shed light on the effects of Aggazzotti G, Fantuzzi G, Righi E, et  al. 1993. Chloroform in alveolar air
low exposure levels. Elevated MMA in the urine of exposed individu- of individuals attending indoor swimming pools. Arch Environ Health,
48(4), 250–​254. PMID: 8357275.
als places them at elevated cancer risk and risk of other adverse health Ahsan H, Chen Y, Parvez F et al. 2006. Arsenic exposure from drinking water
outcomes. The reasons for this observation are not well understood. and risk of premalignant skin lesions in Bangladesh:  baseline results
from the Health Effects of Arsenic Longitudinal Study. Am J Epidemiol,
163(12), 1138–​1148. PMID: 16624965.
Disinfection Byproducts Albuquerque Bernalillo County Water Utility Authority. 2010. Water Resources
Education. Arsenic Removal Facility. Available from:  http://​www.abc-
Considerable evidence implicates exposure to disinfection byproducts wua.org/​education/​31_​Arsenic.html. Accessed June 12, 2015.
in the risk of bladder cancer, although the specific agents associated Andrew AS, Mason RA, Kelsey KT, et al. 2009. DNA repair genotype inter-
with this risk have yet to be determined. Currently, the data for other acts with arsenic exposure to increase bladder cancer risk. Toxicol Lett,
cancer sites are limited and sometimes inconsistent. Well-​designed 187(1), 10–​14. PMCID: PMC2680739.
studies of colon and rectal cancers, renal cell carcinoma, and brain Antonelli R, Shao K, Thomas DJ, Sams R, 2nd, and Cowden J. 2014. AS3MT,
cancer would be informative. It may not be possible to determine GSTO, and PNP polymorphisms:  impact on arsenic methylation and
which of the compounds in the complex and varying mixtures that implications for disease susceptibility. Environ Res, 132, 156–​ 167.
constitute DBPs are most strongly linked to risk. However, it is fea- PMID: 24792412.
sible to refine exposure assessment in future studies to specific types Aschebrook-​Kilfoy B, Heltshe SL, Nuckols JR, et  al. 2012. Modeled nitrate
levels in well water supplies and prevalence of abnormal thyroid condi-
or classes of DBPs in order to guide mitigation efforts. To that end, tions among the Old Order Amish in Pennsylvania. Environ Health, 11, 6.
epidemiologic studies should include lifetime residential histories that PMCID: PMC3305600.
can then be linked to public water supply databases in order to esti- Agency for Toxic Substances and Disease Registry (ATSDR). 2007.
mate long-​term exposures. Of particular importance is the question Toxicological Profile for Arsenic. Available from:  http://​www.atsdr.cdc.
of whether risk differs for brominated and chlorinated compounds. gov/​toxprofiles/​tp.asp?id=22&tid=3. Accessed February 11, 2016.
Future work should also investigate the degree of correlation that other ATSDR. 2016. Camp Lejeune Drinking Water Public Health Assessment,
chemical classes have with THMs or HAAs. Direct measurement and U.S. Marine Corps Base Camp Lejeune, North Carolina. Atlanta,
estimation of other classes would be valuable where possible. Another GA: (ATSDR) Agency for Toxic Substances and Disease Registry.
major knowledge gap is the extent to which inhalation and dermal Baastrup R, Sorensen M, Balstrom T, et al. 2008. Arsenic in drinking-​water and
risk for cancer in Denmark. Environ Health Perspect, 116(2), 231–​237.
routes of exposure are important. Exposure studies suggest that these PMID: 18288323.
routes could result in internal doses of the volatile DBPs much higher Backer LC, Ashley DL, Bonin MA, et al. 2000. Household exposures to drink-
than ingestion. The two epidemiologic studies of bladder cancer that ing water disinfection by-​products: whole blood trihalomethane levels. J
evaluated these routes of exposure are inconsistent. Further evaluation Expo Anal Environ Epidemiol, 10(4), 321–​326. PMID: 10981726.
is warranted. Bailey K and Fry RC. 2014. Long-​term health consequences of prenatal arsenic
exposure: links to the genome and the epigenome. Rev Environ Health,
29(1–​2), 9–​12. PMCID: PMC4049250.
Nitrate Barrett JH, Parslow RC, McKinney PA, Law GR, and Forman D. 1998. Nitrate
in drinking water and the incidence of gastric, esophageal, and brain
Although the number of analytic epidemiologic studies with histori- cancer in Yorkshire, England. Cancer Causes Control, 9(2), 153–​159.
cal exposure data has increased since the early 1990s, there remain PMID: 9578292.
few studies of any single cancer site, making interpretation difficult. Barry V, Winquist A, and Steenland K. 2013. Perfluorooctanoic acid
The recent analytic studies have generally included historical data of (PFOA) exposures and incident cancers among adults living near a
nitrate levels from public water supplies and have evaluated potential chemical plant. Environ Health Perspect, 121(11–​12), 1313–​1318.
confounders and factors affecting nitrosation. However, users of pri- PMCID: PMC3855514.
vate wells, which often have higher nitrate levels than public sources Basu A, Mitra S, Chung J, et  al. 2011. Creatinine, diet, micronutrients, and
arsenic methylation in West Bengal, India. Environ Health Perspect,
of drinking water, are usually excluded due to a lack of long-​term mea- 119(9), 1308–​1313. PMCID: PMC3230402.
surements of nitrate. Recent efforts to model nitrate levels in private Bates M, Smith A, and Cantor K. 1995. Case-​control study of bladder can-
wells for epidemiologic studies using geographic information systems cer and arsenic in drinking water. Am J Epidemiol, 141(6), 523–​530.
(Aschebrook-​Kilfoy et al., 2012; Wheeler et al., 2015) will be impor- PMID: 7900719.
tant for understanding the relationship with nitrate ingestion over a Beane Freeman L, Cantor KP, Karagas M, et al. 2017. Exposure to disinfec-
broader range of intake. With the increase in use of nitrogen-​based fer- tion by-​products and bladder cancer in New England. Environ Health
tilizers, nitrate levels in water supplies have been increasing in many Perspect. 125(6):067010. PMID: 28636529.
areas worldwide; therefore, additional studies, preferably of popula- Berry M. 1997. Childhood Cancer Incidence Health Consultation: A Review
tions with well-​characterized and higher exposures, are warranted. and Analysis of Cancer Registry Data, 1979–​1995 for Dover Township,
Ocean County, New Jersey. Trenton, New Jersey: New Jersey Department
of Health and Senior Services.
Bouvard V, Loomis D, Guyton KZ, et al. 2015. Carcinogenicity of consump-
Cyanotoxins tion of red and processed meat. Lancet Oncol, 16(16), 1599–​ 1600.
Microcystin is a likely cofactor for elevated rates of HCC in south- PMID: 26514947.
eastern coastal China. The expanding worldwide contamination of Bove FJ, Ruckart PZ, Maslia M, and Larson TC. 2014a. Evaluation of mortal-
freshwater used for drinking with nitrate and phosphorous, combined ity among marines and navy personnel exposed to contaminated drinking
water at USMC base Camp Lejeune: a retrospective cohort study. Environ
with increased water temperatures, is leading to more blooms of cya- Health, 13(1), 10. PMCID: PMC3943370.
nobacteria and consequent contamination with microcystin and other Bove FJ, Ruckart PZ, Maslia M, and Larson TC. 2014b. Mortality study of
cyanotoxins. Further studies in China and other regions of the world civilian employees exposed to contaminated drinking water at USMC
are warranted and should include other possibly carcinogenic cyano- Base Camp Lejeune: a retrospective cohort study. Environ Health, 13, 68.
toxins such as nodularin and cylindrospermopsin. PMCID: PMC4237831.
 32

Water Contaminants 323


Bove GE, Jr, Rogerson PA, and Vena JE. 2007. Case control study of the geo- Chiu WA, Jinot J, Scott CS, et al. 2013. Human health effects of trichloroethyl-
graphic variability of exposure to disinfectant byproducts and risk for rec- ene: key findings and scientific issues. Environ Health Perspect, 121(3),
tal cancer. Int J Health Geogr, 6, 18. PMCID: PMC1890278. 303–​311. PMCID: PMC3621199.
British Geological Survey. Government of the People’s Republic of Bangladesh Chorus I and Bartram J (Eds.). 1999. Toxic Cyanobacteria in Water: A Guide
MoLG, Rural Development and Co-​operatives, Department of Public to Their Public Health Consequences, Monitoring and Management.
Health Engineering. 2001. Arsenic Contamination of Groundwater in London: WHO.
Bangladesh. BGS Technical Report WC/​00/​19, Volume 1, Kinniburg D, Chowdhury S, Alhooshani K, and Karanfil T. 2014. Disinfection byproducts in
and Smedley P (Eds.). swimming pool: occurrences, implications and future needs. Water Res,
Buchet JP and Lison D. 1998. Mortality by cancer in groups of the Belgian 53, 68–​109. PMID: 24509344.
population with a moderately increased intake of arsenic. Int Arch Occup Cocco P, Broccia G, Aru G, et al. 2003. Nitrate in community water supplies
Environ Health, 71(2), 125–​130. PMID: 9553788. and incidence of non-​Hodgkin’s lymphoma in Sardinia, Italy. J Epidemiol
Buchet JP, Lauwerys R, and Roels H. 1981. Comparison of the urinary excre- Community Health, 57(7), 510–​511. PMCID: PMC1732520.
tion of arsenic metabolites after a single oral dose of sodium arsenite, Cohen SM, Arnold LL, Beck BD, Lewis AS, and Eldan M. 2013. Evaluation
monomethylarsonate, or dimethylarsinate in man. Int Arch Occup Environ of the carcinogenicity of inorganic arsenic. Crit Rev Toxicol, 43(9), 711–​
Health, 48(1), 71–​79. PMID: 6894292. 752. PMID: 24040994.
Burch MD. 2008. Effective doses, guidelines and regulations. Adv Exp Med Coombs CC, Tavakkoli M, and Tallman MS. 2015. Acute promyelocytic leuke-
Biol, 619, 831–​853. PMID: 18461792. mia: where did we start, where are we now, and the future. Blood Cancer
Butler N, Carlisle JC, Linville R, and Washburn B. 2009. Microcystins: A Brief J, 5, e304. PMCID: PMC4450325.
Overview of Their Toxicity and Effects, with Special Reference to Fish, Costet N, Villanueva CM, Jaakkola JJ, et  al. 2011. Water disinfection by-​
Wildlife, and Livestock. Sacramento, CA, USA: California EPA. products and bladder cancer:  is there a European specificity? A  pooled
C8 Science Panel. 2012. Probable Link Evalution of Cancer. Available from: and meta-​analysis of European case-​control studies. Occup Environ Med,
http:// ​ w ww.c8sciencepanel.org/ ​ p dfs/ ​ P robable_ ​ L ink_ ​ C 8_ ​ C ancer_​ 68(5), 379–​385. PMID: 21389011.
16April2012_​v2.pdf. Cox PA and Sacks OW. 2002. Cycad neurotoxins, consumption of flying foxes,
Cantor KP Hoover R, Hartge P, et  al. 1987. Bladder cancer, drinking water and ALS-​PDC disease in Guam. Neurology, 58(6), 956–​959. PMID:
source, and tap water consumption: a case-​control study. J Natl Cancer 11914415.
Inst, 79(6), 1269–​1279. PMID: 3480378. Cutler D and Miller G. 2005. The role of public health improvements in health
Cantor KP. 1997. Drinking water and cancer. Cancer Causes Control, 8(3), advances: the twentieth-​century United States. Demography, 42(1), 1–​22.
292–​308. PMID: 9498894. PMID: 15782893.
Cantor KP, Ward MH, Moore LE, and Lubin JH. 2006. Water contaminants. Cuzick J, Sasieni P, and Evans S. 1992. Ingested arsenic, keratoses, and bladder
In:  Schottenfeld D, and Fraumeni JF, Jr. (Eds.), Cancer Epidemiology cancer. Am J Epidemiol, 136(4), 417–​421. PMID: 1415161.
and Prevention (3rd ed., pp. 382–​404). New  York:  Oxford University Dabeka RW, McKenzie AD, Lacroix GM, et al. 1993. Survey of arsenic in total
Press. diet food composites and estimation of the dietary intake of arsenic by
Cantor KP, and Lubin JH. 2007. Arsenic, internal cancers, and issues in infer- Canadian adults and children. J AOAC Int, 76(1), 14–​25. PMID: 8448438.
ence from studies of low-​level exposures in human populations. Toxicol Dangleben NL, Skibola CF, and Smith MT. 2013. Arsenic immunotoxicity: a
Appl Pharmacol, 222(3), 252–​257. PMCID: PMC2692340. review. Environ Health, 12(1), 73. PMCID: PMC3848751.
Cantor KP, Villanueva CM, Silverman DT, et  al. 2010. Polymorphisms in Dauphine DC, Smith AH, Yuan Y, et al. 2013. Case-​control study of arsenic in
GSTT1, GSTZ1, and CYP2E1, disinfection by-​ products, and risk of drinking water and lung cancer in California and Nevada. Int J Environ
bladder cancer in Spain. Environ Health Perspect, 118(11), 1545–​1550. Res Public Health, 10(8), 3310–​3324. PMCID: PMC3774440.
PMCID: PMC2974691. Davidson EA, David MB, Galloway JN, et al. 2012. Excess nitrogen in the U.S.
Caro J and Gallego M. 2007. Assessment of exposure of workers and swimmers environment: trends, risks, solutions. Issues Ecol, 15, 1–​16.
to trihalomethanes in an indoor swimming pool. Environ Sci Technol, Delpla I, Scheili A, Guilherme S, Cool G, and Rodriguez MJ. 2016. Variations
41(13), 4793–​4798. PMID: 17695931. of disinfection by-​product levels in small drinking water utilities accord-
Caro J and Gallego M. 2008. Alveolar air and urine analyses as biomarkers of ing to climate change scenarios: a first assessment. J Water Clim Change,
exposure to trihalomethanes in an indoor swimming pool. Environ Sci 7(1), 1–​15.
Technol, 42(13), 5002–​5007. PMID: 18678040. Elliott JA. 2012. Is the future blue-​green? A review of the current model predic-
Centers for Disease Control and Prevention (CDC). 1998. A Survey of the tions of how climate change could affect pelagic freshwater cyanobacte-
Quality of Water Drawn from Domestic Wells in Nine Midwest States (p. ria. Water Res, 46(5), 1364–​1371. PMID: 22244968.
25). Atlanta, GA: CDC. Espejo-​ Herrera N, Cantor KP, Malats N, et  al. 2015. Nitrate in drinking
Chappells H, Parker L, Fernandez CV, et al. 2014. Arsenic in private drinking water and bladder cancer risk in Spain. Environ Res, 137, 299–​307.
water wells:  an assessment of jurisdictional regulations and guidelines PMID: 25601732.
for risk remediation in North America. J Water Health, 12(3), 372–​392. Espejo-​Herrera N, Gracia-​Lavedan E, Boldo E, et al. 2016. Colorectal cancer
PMID: 25252340. risk and nitrate exposure through drinking water and diet. Int J Cancer,
Chen CJ, Kuo TL, and Wu MM. 1988. Arsenic and cancer [letter]. Lancet, 1, 139(2), 334–​346. PMID: 26954527.
414–​415. Espejo-​ Herrera N, Gracia-​ Lavedan E, Pollan M, et  al. 2016b. Ingested
Chen CL, Chiou HY, Hsu LI, et  al. 2010a. Ingested arsenic, characteris- nitrate and breast cancer in the Spanish Multicase-​Control Study on
tics of well water consumption and risk of different histological types Cancer (MCC-​Spain). Environ Health Perspect, 124(7), 1042–​ 1049.
of lung cancer in northeastern Taiwan. Environ Res, 110(5), 455–​462. PMCID: PMC4937871.
PMID: 19735913. Fagin D. 2013. Toms River:  A  Story of Science and Salvation.
Chen CL, Chiou HY, Hsu LI, et  al. 2010b. Arsenic in drinking water and New York: Bantam Books.
risk of urinary tract cancer:  a follow-​ up study from northeastern Fagliano JA, Berry M, Kohler BA, Klotz JB, and Imtiaz R. 2003. Case-​Control
Taiwan. Cancer Epidemiol Biomarkers Prev, 19(1), 101–​110. PMID: Study of Childhood Cancers in Dover Township, Ocean County, New
20056628. Jersey, Vol I–​Vol V. Trenton, New Jersey, USA: New Jersey Department
Chen CL, Hsu LI, Chiou HY, et al. 2004. Ingested arsenic, cigarette smoking, of Health and Senior Services in cooperation with Agency for Toxic
and lung cancer risk:  a follow-​up study in arseniasis-​endemic areas in Substances and Disease Registry.
Taiwan. JAMA, 292(24), 2984–​2990. PMID: 15613666. Falconer IR and Buckley TH. 1989. Tumor promotion by microcystis sp a
Chen JG and Kensler TW. 2014. Changing rates for liver and lung cancers in blue-​green-​alga occurring in water-​supplies. Med J Australia, 150(6),
Qidong, China. Chem Res Toxicol, 27(1), 3–​6. PMCID: PMC3946948. 351–​351.
Chen K, Cai J, Liu XY, et  al. 2003. Nested case-​control study on the risk Ferreccio C, Gonzalez C, Milosavjlevic V, et al. 2000. Lung cancer and arsenic
factors of colorectal cancer. World J Gastroenterol, 9(1), 99–​ 103. concentrations in drinking water in Chile. Epidemiology, 11(6), 673–​679.
PMCID: PMC4728259. PMID: 11055628.
Chen Y, Hall M, Graziano JH, et  al. 2007. A prospective study of blood Ferreccio C, Smith AH, Duran V, et al. 2013a. Case-​control study of arsenic in
selenium levels and the risk of arsenic-​ related premalignant skin drinking water and kidney cancer in uniquely exposed Northern Chile.
lesions. Cancer Epidemiol Biomarkers Prev, 16(2), 207–​213. PMCID: Am J Epidemiol, 178(5), 813–​818. PMCID: PMC3817454.
PMC1924921. Ferreccio C, Yuan Y, Calle J, et al. 2013b. Arsenic, tobacco smoke, and occu-
Chilvers C, Inskip H, Caygill C, et al. 1984. A survey of dietary nitrate in well-​ pation:  associations of multiple agents with lung and bladder cancer.
water users. Int J Epidemiol, 13(3), 324–​331. PMID: 6490303. Epidemiology, 24(6), 898–​905. PMID: 24036609.
324

324 PART III:  THE CAUSES OF CANCER


Finn OJ. 2012. Immuno-​oncology:  understanding the function and dysfunc- Hough RL, Fletcher T, Leonardi GS, et al. 2010. Lifetime exposure to arsenic
tion of the immune system in cancer. Ann Oncol, 23 Suppl 8, viii6–​9. in residential drinking water in Central Europe. Int Arch Occup Environ
PMCID: PMC4085883. Health, 83(5), 471–​481. PMID: 20401490.
Flanagan SV, Johnston RB, and Zheng Y. 2012. Arsenic in tube well water Howarth RW. 2008. Coastal nitrogen pollution: a review of sources and trends
in Bangladesh:  health and economic impacts and implications for globally and regionally. Harmful Algae, 8(1), 14–​20.
arsenic mitigation. Bull World Health Organ, 90(11), 839–​ 846. Hsu LI, Chiu AW, Huan SK, et  al. 2008. SNPs of GSTM1, T1, P1, epoxide
PMCID: PMC3506399. hydrolase and DNA repair enzyme XRCC1 and risk of urinary transi-
Fleming LE, Rivero C, Burns J, et al. 2002. Blue green algal (cyanobacterial) tional cell carcinoma in southwestern Taiwan. Toxicol Appl Pharmacol,
toxins, surface drinking water, and liver cancer in Florida. Harmful Algae, 228(2), 144–​155. PMID: 18199464.
1(2), 157–​168. Huang YK, Huang YL, Hsueh YM, et  al. 2008. Arsenic exposure, uri-
Focazio MJ, Tipton D, Shapiro SD, and Geiger LH. 2006. The chemical quality nary arsenic speciation, and the incidence of urothelial carcinoma:  a
of self-​supplied domestic well water in the United States. Ground Water twelve-​year follow-​up study. Cancer Causes Control, 19(8), 829–​839.
Monit Remediat, 26(3), 92–​104. PMID: 18351295.
Frisbee SJ, Brooks AP, Jr., Maher A, et al. 2009. The C8 health project: design, Hutson SS, Barber NL, Kenney JF, Linsey KS, Lumia DS, and Maupin MA.
methods, and participants. Environ Health Perspect, 117(12), 1873–​1882. 2004. Estimated Use of Water in the United States in 2000. Washington,
PMCID: PMC2799461. DC: US Geological Survey.
Fujiki H and Suganuma M. 1994. Tumor necrosis factor-​alpha, a new tumor International Agency for Research on Cancer (IARC). 1991. Chlorinated drinking-​
promoter, engendered by biochemical studies of okadaic acid. J Biochem, water; chlorination by-​products; some other halogenated compounds; cobalt
115(1), 1–​5. PMID: 8188614. and cobalt compounds. IARC Monogr Eval Carcinog Risks Hum, 52.
Fujiki H and Suganuma M. 2011. Tumor promoters: microcystin-​LR, nodularin IARC. 2004. Some drinking water disinfectants and contaminants, including
and TNF-​alpha and human cancer development. Anticancer Agents Med arsenic. IARC Monogr Eval Carcinog Risks Hum, 84.
Chem, 11(1), 4–​18. PMID: 21269254. IARC. 2010. Ingested nitrate and nitrite, and cyanobacterial peptide toxins.
Gagarin MF (Ed.). 2010. The Oxford Encyclopedia of Ancient Greece and IARC Monogr Eval Carcinog Risks Hum, 94, 329–​412.
Rome, Volume 1. Oxford: Oxford University Press. IARC. 2012a. A review of human carcinogens:  arsenic, metals, fibres, and
Gamble MV, Liu X, Ahsan H, et  al. 2006. Folate and arsenic metabolism: a dusts. IARC Monogr Eval Carcinog Risks Hum, 100(Pt C).
double-​ blind, placebo-​controlled folic acid-​ supplementation trial in IARC. 2012b. Chemical agents and related occupations. IARC Monogr Eval
Bangladesh. Am J Clin Nutr, 84(5), 1093–​1101. PMCID: PMC2046214. Carcinog Risks Hum, 100(Pt F), 9–​562.
Garcia-​Esquinas E, Pollan M, Umans JG, et  al. 2013. Arsenic exposure and IARC. 2013. Some chemicals in industrial and consumer products, some food
cancer mortality in a US-​based prospective cohort: the Strong Heart contaminants and flavourings, and water chlorination by-​products. IARC
Study. Cancer Epidemiol Biomarkers Prev, 22(11), 1944–​1953. PMID: Monogr Eval Carcinog Risks Hum, 101.
23800676. IARC. 2014. Trichloroethylene, tetrachloroethylene, and some other chlori-
George CM, Smith AH, Kalman DA, and Steinmaus CM. 2006. Reverse osmo- nated agents. IARC Monogr Eval Carcinog Risks Hum, 106.
sis filter use and high arsenic levels in private well water. Arch Environ IARC. 2016. Perfluorooctanoic acid, tetrafluoroethylene, dichloromethane, 1,2-​
Occup Health, 61(4), 171–​175. PMCID: PMC3849398. dichloropropane, and 1,3-​propane sultone. IARC Monogr Eval Carcinog
Gibb H, Haver C, Gaylor D, et  al. 2011. Utility of recent studies to assess Risks Hum, 110.
the National Research Council 2001 estimates of cancer risk from Inoue-​Choi M, Jones RR, Anderson KE, et al. 2015. Nitrate and nitrite inges-
ingested arsenic. Environ Health Perspect, 119(3), 284–​290. PMCID: tion and risk of ovarian cancer among postmenopausal women in Iowa.
PMC3059988. Int J Cancer, 137(1), 173–​182. PMCID: PMC4405451.
Gilbert-​Diamond D, Cottingham KL, Gruber JF, et al. 2011. Rice consumption Inoue-​Choi M, Ward MH, Cerhan JR, et  al. 2012. Interaction of nitrate and
contributes to arsenic exposure in US women. Proc Natl Acad Sci U S A, folate on the risk of breast cancer among postmenopausal women. Nutr
108(51), 20656–​20660. PMCID: PMC3251121. Cancer, 64(5), 685–​694. PMCID: PMC3403733.
Gilbert-​Diamond D, Li Z, Perry AE, Spencer SK, Gandolfi AJ, and Karagas James KA, Meliker JR, Buttenfield BE, et al. 2014. Predicting arsenic concen-
MR. 2013. A population-​based case-​control study of urinary arsenic spe- trations in groundwater of San Luis Valley, Colorado:  implications for
cies and squamous cell carcinoma in New Hampshire, USA. Environ individual-​level lifetime exposure assessment. Environ Geochem Health,
Health Perspect, 121(10), 1154–​1160. PMID: 23872349. 36(4), 773–​782. PMID: 24429726.
Griffiths DJ and Saker ML. 2003. The Palm Island mystery disease 20 years Jeong CH, Wagner ED, Siebert VR, et  al. 2012. Occurrence and toxicity of
on: a review of research on the cyanotoxin cylindrospermopsin. Environ disinfection byproducts in European drinking waters in relation with
Toxicol, 18(2), 78–​93. PMID: 12635096. the HIWATE epidemiology study. Environ Sci Technol, 46(21), 12120–​
Guha Mazumder DN, Haque R, Ghosh N, et al. 1998. Arsenic levels in drink- 12128. PMCID: PMC4790091.
ing water and the prevalence of skin lesions in West Bengal, India. Int J Jochimsen EM, Carmichael WW, An JS, et al. 1998. Liver failure and death
Epidemiol, 27(5), 871–​877. PMID: 9839746. after exposure to microcystins at a hemodialysis center in Brazil. N Engl
Gulis G, Czompolyova M, and Cerhan JR. 2002. An ecologic study of nitrate J Med, 338(13), 873–​878. PMID: 9516222.
in municipal drinking water and cancer incidence in Trnava District, Johnson CJ and Kross BC. 1990. Continuing importance of nitrate contami-
Slovakia. Environ Res, 88(3), 182–​187. PMID: 12051796. nation of groundwater and wells in rural areas. Am J Ind Med, 18(4),
Hall MN and Gamble MV. 2012. Nutritional manipulation of one-​ carbon 449–​456. PMID: 2248249.
metabolism: effects on arsenic methylation and toxicity. J Toxicol, 2012, Johnston RB and Sarker MH. 2007. Arsenic mitigation in Bangladesh: national
595307. PMCID: PMC3317163. screening data and case studies in three upazilas. J Environ Sci Health A
Han YY, Weissfeld JL, Davis DL, and Talbott EO. 2009. Arsenic levels in Tox Hazard Subst Environ Eng, 42(12), 1889–​1896. PMID: 17952790.
ground water and cancer incidence in Idaho: an ecologic study. Int Arch Jones RR, Weyer PJ, Dellavalle CT, et al. 2017. Ingested nitrate, disinfection
Occup Environ Health, 82(7), 843–​849. PMID: 18941772. by-​products, and kidney cancer risk in older women. Epidemiology,
Harada K, Oshikata M, Uchida H, et al. 1996. Detection and identification of [epub ahead of print]. PMID: 28252454.
microcystins in the drinking water of Haimen City, China. Nat Toxins, Jones RR, Weyer PJ, Dellavalle CT, et al. 2016. Nitrate from drinking water and
4(6), 277–​283. PMID: 9029553. diet and bladder cancer among postmenopausal women in Iowa. Environ
Heck JE, Andrew AS, Onega T, et al. 2009. Lung cancer in a U.S. population Health Perspect, 124(11), 1751–​1758. PMID: 27258851.
with low to moderate arsenic exposure. Environ Health Perspect, 117(11), Kalman DA, Dills RL, Steinmaus C, et al. 2014. Occurrence of trivalent mono-
1718–​1723. PMID: 20049123. methyl arsenic and other urinary arsenic species in a highly exposed
Heisler J, Glibert PM, Burkholder JM, et al. 2008. Eutrophication and harmful juvenile population in Bangladesh. J Expo Sci Environ Epidemiol, 24(2),
algal blooms: a scientific consensus. Harmful Algae, 8(1), 3–​13. 113–​120. PMID: 23549402.
Hertz-​Picciotto I, Smith AH, Holtzman D, Lispett M, and Alexeef G. 1992. Karagas MR, Stukel TA, Morris JS, et al. 2001. Skin cancer risk in relation
Synergism between occupational arsenic exposure and smoking in the to toenail arsenic concentrations in a US population-​based case-​control
induction of lung cancer. Epidemiology, 3(1), 23–​31. study. Am J Epidemiol, 153(6), 559–​565. PMID: 11257063.
Hildesheim ME, Cantor KP, Lynch CF, et  al. 1998. Drinking water source Karagas MR, Tosteson TD, Morris JS, et al. 2004. Incidence of transitional
and chlorination byproducts. II. Risk of colon and rectal cancers. cell carcinoma of the bladder and arsenic exposure in New Hampshire.
Epidemiology, 9(1), 29–​35. PMID: 9430265. Cancer Causes & Control, 15, 265–​272. PMID: 15286466.
Hopenhayn-​Rich C, Smith AH, and Goeden HM. 1993. Human studies do not Kennedy GL and Symons JM, 2015. Carcinogenicity of perfluoroalkyl com-
support the methylation threshold hypothesis for the toxicity of inorganic pounds. In: DeWitt J (Ed.), Toxicological Effects of Perfluoroalkyl and
arsenic. Environ Res, 60(2), 161–​177. Polyfluoroalkyl Substances (pp. 265–​304). London: Springer-​Verlag.
 325

Water Contaminants 325


Khan K, Ahmed E, Factor-​Litvak P, et  al. 2015. Evaluation of an elemen- and Distribution of Drinking Water Within the Service Areas of the
tary school-​based educational intervention for reducing arsenic expo- Hadnot Point and Holcomb Boulevard Water Treatment Plants and
sure in Bangladesh. Environ Health Perspect, 123(12), 1331–​ 1336. Vicinities, U.S. Marine Corps Base Camp Lejeune, North Carolina,
PMCID: PMC4671245. Chapter A: Summary and Findings. Available from: http://​www.atsdr.cdc.
Kile ML and Ronnenberg AG. 2008. Can folate intake reduce arsenic toxicity? gov/​sites/​lejeune/​hadnotpoint.html.
Nutr Rev, 66(6), 349–​353. PMCID: PMC4013823. McElroy JA, Trentham-​Dietz A, Gangnon RE, et  al. 2008. Nitrogen-​nitrate
Kile ML, Houseman EA, Baccarelli AA, et al. 2014. Effect of prenatal arse- exposure from drinking water and colorectal cancer risk for rural women
nic exposure on DNA methylation and leukocyte subpopulations in cord in Wisconsin, USA. J Water Health, 6(3), 399–​409. PMID: 19108561.
blood. Epigenetics, 9(5), 774–​782. PMCID: PMC4063836. Melak D, Ferreccio C, Kalman D, et al. 2014. Arsenic methylation and lung
King WD, Marrett LD, and Woolcott CG. 2000. Case-​control study of colon and bladder cancer in a case-​control study in northern Chile. Toxicol Appl
and rectal cancers and chlorination by-​products in treated water. Cancer Pharmacol, 274(2), 225–​231. PMCID: PMC4344188.
Epidemiol Biomarkers Prev, 9(8), 813–​818. PMID: 10952098. Meliker JR, Avruskin GA, Slotnick MJ, et al. 2008. Validity of spatial models
Kitchin KT and Conolly R. 2010. Arsenic-​induced carcinogenesis-​-​oxidative of arsenic concentrations in private well water. Environ Res, 106(1), 42–​
stress as a possible mode of action and future research needs for more 50. PMCID: PMC2271042.
biologically based risk assessment. Chem Res Toxicol, 23(2), 327–​335. Meliker JR, Franzblau A, Slotnick MJ, and Nriagu JO. 2006. Major contribu-
PMID: 20035570. tors to inorganic arsenic intake in southeastern Michigan. Int J Hyg
Kleinjans JC, Albering HJ, Marx A, et al. 1991. Nitrate contamination of drink- Environ Health, 209(5), 399–​411. PMID: 16731038.
ing water:  evaluation of genotoxic risk in human populations. Environ Meliker JR, Slotnick MJ, Avruskin GA, et al. 2007. Individual lifetime expo-
Health Perspect, 94, 189–​193. PMCID: PMC1567968. sure to inorganic arsenic using a space-​time information system. Int Arch
Kogevinas M, Villanueva CM, Font-​Ribera L, et al. 2010. Genotoxic effects in Occup Environ Health, 80(3), 184–​197. PMID: 16897097.
swimmers exposed to disinfection by-​products in indoor swimming pools. Meliker JR, Slotnick MJ, AvRuskin GA, et al. 2010. Lifetime exposure to
Environ Health Perspect, 118(11), 1531–​1537. PMCID: PMC2974689. arsenic in drinking water and bladder cancer: a population-​based case-​
Kosnett MJ. 2013. The role of chelation in the treatment of arsenic and mercury control study in Michigan, USA. Cancer Causes Control, 21(5), 745–​
poisoning. J Med Toxicol, 9(4), 347–​354. PMCID: PMC3846971. 757. PMID: 20084543.
Kurttio P, Pukkala E, Kahelin H, Auvinen A, and Pekkanen J. 1999. Arsenic Melkonian S, Argos M, Pierce BL, et al. 2011. A prospective study of the syn-
concentrations in well water and risk of bladder and kidney cancer in ergistic effects of arsenic exposure and smoking, sun exposure, fertilizer
Finland. Environ Health Perspect, 107(9), 705–​710. PMID: 10464069. use, and pesticide use on risk of premalignant skin lesions in Bangladeshi
Lamm S, Engel A, Kruse M, Feinleib M, Byrd D, Lai S, and Wilson R. 2004. men. Am J Epidemiol, 173(2), 183–​191. PMCID: PMC3011951.
Arsenic in drinking water and bladder cancer mortality in the US:  an Melkonian S, Argos M, Chen Y, et  al. 2012. Intakes of several nutrients are
analysis based on 133 U.S. counties and thirty years of observation. J Occ associated with incidence of arsenic-​ related keratotic skin lesions in
Environ Med, 48, 298–​306. PMID: 15091293. Bangladesh. J Nutr, 142(12), 2128–​2134. PMCID: PMC3497963.
Law G, Parslow R, McKinney P, and Cartwright R. 1999. Non-​Hodgkin’s Miller MD, Marty AM, Arcus A, et al. 2001. Differences between children and
lymphoma and nitrate in drinking water:  a study in Yorkshire, United adults: implications in risk assessment at California EPA. Int J Toxicol,
Kingdom. J Epidemiol Community Health, 53(6), 383–​384. PMCID: 21, 403–​418.
PMC1756892. Min JY and Min KB. 2016. Blood trihalomethane levels and the risk of
Lee J, Jun MJ, Lee MH, et al. 2010. Production of various disinfection byprod- total cancer mortality in US adults. Environ Pollut, 212, 90–​96. PMID:
ucts in indoor swimming pool waters treated with different disinfection 26840521.
methods. Int J Hyg Environ Health, 213(6), 465–​474. PMID: 20961810. Mirvish SS. 1995. Role of N-​nitroso compounds (NOC) and N-​nitrosation in
Levine A, Yang Y, and Goodrich J. 2016. Enhancing climate adaptation capac- etiology of gastric, esophageal, nasopharyngeal and bladder cancer and
ity for drinking water treatement facilities. J Water Clim Change, 7, contribution to cancer of known exposures to NOC. Cancer Lett, 93(1),
485–​497. 17–​48. PMID: 7600541.
Lewis DR, Southwick JW, Ouellet-​Hellstrom R, Rench J, and Calderon RL. Mirvish SS, Grandjean AC, Møller H, et al. 1992. N-​nitrosoproline excretion
1999. Drinking Water Arsenic in Utah: A Cohort Mortality Study. Environ by rural Nebraskans drinking water of varied nitrate content. Cancer
Health Perspect, 107(5), 359–​365. PMID: 10210691. Epidemiol Biomarkers Prev, 1(6), 455–​461. PMID: 1302557.
Lian M, Liu Y, Yu SZ, et al. 2006. Hepatitis B virus x gene and cyanobacte- Møller H, Landt J, Pedersen E, et al. 1989. Endogenous nitrosation in relation
rial toxins promote aflatoxin B-​1-​induced hepatotumorigenesis in mice. to nitrate exposure from drinking water and diet in a Danish rural popula-
World J Gastroenterol, 12(19), 3065–​3072. tion. Cancer Res, 49(11), 3117–​3121. PMID: 2720669.
Lijinsky W. 1986. The significance of N-​nitroso compounds as environmental Mueller BA, Searles Nielsen S, Preston-​Martin S, et al. 2004. Household water
carcinogens. Environ Carcinog Rev, 4(1), 1–​45. source and the risk of childhood brain tumours: results of the SEARCH
Ling B. 2000. Health impairments arising from drinking water polluted with International Brain Tumor Study. Int J Epidemiol, 33(6), 1209–​1216.
domestic sewage and excreta in China. Schriftenr Ver Wasser Boden PMID: 15567873.
Lufthyg, 105, 43–​46. PMID: 10842792. National Oceanic & Atmospheric Administration (NOAA). 2015. Global
Lourencetti C, Grimalt JO, Marco E, et al. 2012. Trihalomethanes in chlorine Highlights:  Calendar Year 2015. Available from:  http://​www.ncdc.noaa.
and bromine disinfected swimming pools:  air-​water distributions and gov/​sotc/​summary-​info/​global/​201512. Accessed September 23, 2016.
human exposure. Environ Int, 45, 59–​67. PMID: 22572118. National Toxicology Program. 2015. Report on Carcinogens: Monograph on
Marshall G, Ferreccio C, Yuan Y, et al. 2007. Fifty-​year study of lung and blad- Trichloroethylene. Research Triangle Park, NC: US Department of Health
der cancer mortality in Chile related to arsenic in drinking water. J Natl and Human Services.
Cancer Inst, 99(12), 920–​928. PMID: 17565158. Nesnow S, Roop BC, Lambert G, et al. 2002. DNA damage induced by methyl-
Maslia ML, Reyes JJ, Gillig RE, et al. 2005. Public health partnerships address- ated trivalent arsenicals is mediated by reactive oxygen species. Chem
ing childhood cancer investigations:  case study of Toms River, Dover Res Toxicol, 15(12), 1627–​1634. PMID: 12482246.
Township, New Jersey, USA. Int J Hyg Environ Health, 208(1–​2), 45–​54. Newcombe G, Chorus I, Falconer I, and Lin TF. 2012. Cyanobacteria: impacts
PMID: 15881978. of climate change on occurrence, toxicity and water quality management.
Maslia ML, Sautner JB, Aral MM, et al. 2000. Using water-​distribution system Water Res, 46(5), 1347–​1348. PMID: 22265251.
modeling to assist epidemiologic investigations. J Water Res Pl-​ASCE, New Jersey Department of Health. 2016. Toms River Township Childhood
126(4), 180–​198. Cancer Investigation. Available from:  http://​www.nj.gov/​health/​eohs/​
Maslia ML, Sautner JB, Faye RE, et al. 2007. Analysis of Groundwater Flow, dovertwp.shtml -​ Case-​control%20studies. Accessed April 18, 2016.
Contaminant Fate and Transport, and Distribution of Drinking Water at Nishiwaki-​Matsushima R, Ohta T, Nishiwaki S, et al. 1992. Liver tumor pro-
Tarawa Terrace and Vicinity, U.S. Marine Corps Base Camp Lejeune, motion by the cyanobacterial cyclic peptide toxin microcystin-​LR. J
North Carolina: Historical Reconstruction and Present-​Day Conditions—​ Cancer Res Clin Oncol, 118(6), 420–​424. PMID: 1618889.
Executive Summary. Available from:  http://​www.atsdr.cdc.gov/​sites/​ Nolan BT and Stoner JD, 2000. Nutrients in groundwaters of the conterminous
lejeune/​tarawaterrace.html. United States, 1992–​1995. Environ Sci Technol, 34, 1156–​1165.
Maslia ML, Sautner JB, and Mustafa MA. 2001. Summary of Findings: Historical National Research Council (NRC). 1981. The Health Effects of Nitrate, Nitrite,
Reconstruction of the Water-​ Distribution System Serving the Dover and N-​Nitroso Compounds. Washington DC: Committee on Nitrite and
Township Area, New Jersey. Atlanta, GA: Agency for Toxic Substances Alternative Curing Agents in Foods, National Research Council.
and Disease Registry (ATSDR). NRC. 1999. Arsenic in Drinking Water (pp. 229–​ 250). Washington,
Maslia ML, Suarez-​Soto RJ, Sautner JB, et al. 2013. Analyses and Historical DC:  Subcommittee on Arsenic in Drinking Water, National Research
Reconstruction of Groundwater Flow, Contaminant Fate and Transport, Council.
326

326 PART III:  THE CAUSES OF CANCER


NRC. 2001. Arsenic in Drinking Water 2001 Update. Washington, Perspectives on Drinking Water Risk Assessment and Management.
DC: Subcommittee to Update the 1999 Arsenic in Drinking Water Report, Proceedings of the Santiago (Chile) Symposium, September 1998. IAHS
National Research Council. Pub. no. 260.
NRC. 2014. Critical Aspects of EPA’s IRIS Assessment of Inorganic Schopf JW. 2000. Solution to Darwin’s dilemma:  discovery of the missing
Arsenic:  Interim Report. National Research Council. Available Precambrian record of life. Proc Natl Acad Sci U S A, 97(13), 6947–​6953.
from: http://​www.nap.edu/​catalog.php?record_​id=18594. Accessed June PMCID: PMC34368.
2, 2014. Schwimmer M and Schwimmer D. 1955. The Role of Algae and Plankton in
Nuckols JR, Freeman LE, Lubin JH, et al. 2011. Estimating water supply arse- Medicine. New York: Grune & Stratton.
nic levels in the New England Bladder Cancer Study. Environ Health Shen ZC, Li WG, and Chen JG. 1985. An epidemiological study of drinking
Perspect, 119(9), 1279–​1285. PMCID: PMC3230387. water and PLC in Qidong (in Chinese). Jiangsu Med J, 11, 28–​30.
O’Reilly CM, Sharma S, Gray DK, et  al. 2015. Rapid and highly variable Shin HM, Vieira V, Ryan PB, et  al. 2011a. Retrospective exposure estima-
warming of lake surface waters around the globe. Geophysical Research tion for perfluorooctanoic acid (PFOA) for participants in the C8 Health
Letters, 42(24), 10,773–​10,781. Project. Epidemiology, 22(1), S83–​S83.
Olson JM. 2006. Photosynthesis in the Archean era. Photosynth Res, 88(2), Shin HM, Vieira VM, Ryan PB, et  al. 2011b. Environmental fate and trans-
109–​117. PMID: 16453059. port modeling for perfluorooctanoic acid emitted from the Washington
Orloff K, Mistry K, and Metcalf S. 2009. Biomonitoring for environmental Works Facility in West Virginia. Environ Sci Technol, 45(4), 1435–​1442.
exposures to arsenic. J Toxicol Environ Health B Crit Rev, 12(7), 509–​ PMID: 21226527.
524. PMID: 20183531. Shin HM, Vieira VM, Ryan PB, Steenland K, and Bartell SM. 2013.
Paerl HW, Hall NS, and Calandrino ES. 2011. Controlling harmful cyanobacte- Retrospective exposure estimation and predicted versus observed serum
rial blooms in a world experiencing anthropogenic and climatic-​induced perfluorooctanoic acid concentrations for participants in the C8 Health
change. Sci Total Environ, 409(10), 1739–​1745. PMID: 21345482. Project. Environ Health Perspect, 199(4), 1760.
Paerl HW and Paul VJ. 2012. Climate change: links to global expansion of Slotnick MJ, Meliker JR, and Nriagu JO. 2006. Effects of time and point-​of-​use
harmful cyanobacteria. Water Res, 46(5), 1349–​1363. PMID: 21893330. devices on arsenic levels in Southeastern Michigan drinking water, USA.
Peters BA, Hall MN, Liu X, et al. 2015. Folic acid and creatine as therapeutic Sci Total Environ, 369(1–​3), 42–​50. PMID: 16750243.
approaches to lower blood arsenic: a randomized controlled trial. Environ Smith AH, Lingas EO, and Rahman M. 2000. Contamination of drinking-​water
Health Perspect, 123(12), 1294–​1301. PMCID: PMC4671237. by arsenic in Bangladesh: a public health emergency. Bull World Health
Pilsner JR, Hall MN, Liu X, et  al. 2011. Associations of plasma selenium Organ, 78(9), 1093–​1103. PMCID: PMC2560840.
with arsenic and genomic methylation of leukocyte DNA in Bangladesh. Smith AH, Marshall G, Liaw J, et al. 2012. Mortality in young adults follow-
Environ Health Perspect, 119(1), 113–​118. PMCID: PMC3018489. ing in utero and childhood exposure to arsenic in drinking water. Environ
Rahman MA and Hasegawa H. 2011. High levels of inorganic arsenic in rice in Health Perspect, 120(11), 1527–​1531. PMCID: PMC3556614.
areas where arsenic-​contaminated water is used for irrigation and cook- Smith AH, Marshall G, Yuan Y, et al. 2014. Rapid reduction in breast cancer
ing. Sci Total Environ, 409(22), 4645–​4655. PMID: 21899878. mortality with inorganic arsenic in drinking water. EBioMedicine, 1(1),
Rahman MB, Cowie C, Driscoll T, et al. 2014. Colon and rectal cancer inci- 58–​63. PMCID: PMC4286879.
dence and water trihalomethane concentrations in New South Wales, Smith AH and Steinmaus CM. 2009. Health effects of arsenic and chromium
Australia. BMC Cancer, 14, 445. PMCID: PMC4088985. in drinking water: recent human findings. Annu Rev Public Health, 30,
Ravenscroft P. 2007. Predicting the Global Distribution of Natural Arsenic 107–​122. PMCID: PMC2762382.
Contamination of Groundwater. Symposium on Arsenic: The Geography Snow J. 1855. On the Mode of Communication of Cholera, 2nd edition.
of a Global Problem. Royal Geographical Society, London. Available London: Churchill.
from:  http://​www.geog.cam.ac.uk/​research/​projects/​arsenic/​symposium/​ Steenland K, Jin C, MacNeil J, et al. 2009. Predictors of PFOA levels in a com-
S1.2_​P_​Ravenscroft.pdf. Accessed March 16, 2012. munity surrounding a chemical plant. Environ Health Perspect, 117(7),
Ren X, McHale CM, Skibola CF, et  al. 2011. An emerging role for epigen- 1083–​1088. PMCID: PMC2717134.
etic dysregulation in arsenic toxicity and carcinogenesis. Environ Health Steenland K and Woskie S. 2012. Cohort mortality study of workers exposed
Perspect, 119(1), 11–​19. PMCID: PMC3018488. to perfluorooctanoic acid. Am J Epidemiol, 176(10), 909–​917. PMID:
Rice JM, Rehm S, Donovan PJ, and Perantoni AO. 1989. Comparative trans- 23079607.
placental carcinogenesis by directly acting and metabolism-​dependent Steinmaus C, Castriota F, Ferreccio C, et al. 2015. Obesity and excess weight
alkylating agents in rodents and nonhuman primates. IARC Sci Publ, in early adulthood and high risks of arsenic-​related cancer in later life.
96, 17–​34. Environ Res, 142, 594–​601. PMCID: PMC4664040.
Richardson SD, Plewa MJ, Wagner ED, Schoeny R, and Demarini DM. 2007. Steinmaus C, Ferreccio C, Acevedo J, et  al. 2014. Increased lung and blad-
Occurrence, genotoxicity, and carcinogenicity of regulated and emerg- der cancer incidence in adults after in utero and early-​ life arsenic
ing disinfection by-​products in drinking water: a review and roadmap for exposure. Cancer Epidemiol Biomarkers Prev, 23(8), 1529–​ 1538.
research. Mutat Res, 636(1–​3), 178–​242. PMID: 17980649. PMCID: PMC4344186.
Rook JJ. 1974. Formation of haloforms during chlorination of natural waters. Steinmaus CM, Ferreccio C, Romo JA, et al. 2013. Drinking water arsenic in
Water Treat Exam, 23, 234–​243. northern chile: high cancer risks 40 years after exposure cessation. Cancer
Rowland IR, Granli T, Bockman OC, Key PE, and Massey RC. 1991. Epidemiol Biomarkers Prev, 22(4), 623–​630. PMCID: PMC3848421.
Endogenous N-​nitrosation in man assessed by measurement of apparent Styblo M, Del Razo LM, Vega L, et al. 2000. Comparative toxicity of trivalent
total N-​nitroso compounds in faeces. Carcinogenesis, 12(8), 1395–​1401. and pentavalent inorganic and methylated arsenicals in rat and human
PMID: 1860160. cells. Arch Toxicol, 74(6), 289–​299. PMID: 11005674.
Ruckart PZ, Bove FJ, and Maslia M. 2013. Evaluation of exposure to contami- Su CC, Lu JL, Tsai KY, and Lian Ie B. 2011. Reduction in arsenic intake from
nated drinking water and specific birth defects and childhood cancers at water has different impacts on lung cancer and bladder cancer in an arse-
Marine Corps Base Camp Lejeune, North Carolina: a case-​control study. niasis endemic area in Taiwan. Cancer Causes Control, 22(1), 101–​108.
Environ Health, 12, 104. PMCID: PMC3880212. PMID: 21052815.
Ruckart PZ, Bove FJ, Shanley E, 3rd, and Maslia M. 2015. Evaluation of con- Su D. 1979. Drinking water and liver cell cancer: an epidemiologic approach to
taminated drinking water and male breast cancer at Marine Corps Base the etiology of this disease in China. Chin Med J, 92, 748–​756.
Camp Lejeune, North Carolina: a case control study. Environ Health, 14, Sueoka E, Sueoka N, Okabe S, et al. 1997. Expression of the tumor necrosis
74. PMCID: PMC4571057. factor alpha gene and early response genes by nodularin, a liver tumor
Salas LA, Bustamante M, Gonzalez JR, et al. 2015. DNA methylation levels and promoter, in primary cultured rat hepatocytes. J Cancer Res Clin Oncol,
long-​term trihalomethane exposure in drinking water: an epigenome-​wide 123(8), 413–​419. PMID: 9292703.
association study. Epigenetics, 10(7), 650–​661. PMCID: PMC4622514. Sun RC, Board PG, and Blackburn AC. 2011. Targeting metabolism with arse-
Salas LA, Cantor KP, Tardon A, et al. 2013. Biological and statistical approaches nic trioxide and dichloroacetate in breast cancer cells. Mol Cancer, 10,
for modeling exposure to specific trihalomethanes and bladder cancer 142. PMCID: PMC3240126.
risk. Am J Epidemiol, 178(4), 652–​660. PMCID: PMC3736753. Sun Y, Tokar EJ, and Waalkes MP. 2012. Overabundance of putative cancer
Salas LA, Villanueva CM, Tajuddin SM, et  al. 2014. LINE-​1 methyla- stem cells in human skin keratinocyte cells malignantly transformed by
tion in granulocyte DNA and trihalomethane exposure is associated arsenic. Toxicol Sci, 125(1), 20–​29. PMCID: PMC3243751.
with bladder cancer risk. Epigenetics, 9(11), 1532–​1539. PMCID: Svircev Z, Drobac D, Tokodi N, et  al. 2013. Epidemiology of primary liver
PMC4622716. cancer in Serbia and possible connection with cyanobacterial blooms. J
Sancha A, O’Ryan R, and Perez O. 2000. The removal of arsenic from drink- Environ Sci Health C Environ Carcinog Ecotoxicol Rev, 31(3), 181–​200.
ing water and associated costs:  the Chilean case. Interdisciplinary PMID: 24024518.
 327

Water Contaminants 327


Tao SS and Bolger PM. 1999. Dietary arsenic intakes in the United States: FDA van Maanen JM, Welle IJ, Hageman G, et al. 1996. Nitrate contamination of
Total Diet Study, September 1991–​December 1996. Food Addit Contam, drinking water: relationship with HPRT variant frequency in lymphocyte
16(11), 465–​472. PMID: 10755138. DNA and urinary excretion of N-​nitrosamines. Environ Health Perspect,
Thomson B, Fox K, and O’Grady M. 2000. Arsenic removal by point-​of-​use 104(5), 522–​528. PMCID: PMC1469364.
treatment systems at San Ysidro, NM. Paper presented at the American Vermeer IT, Pachen DM, Dallinga JW, Kleinjans JC, and van Maanen JM.
Water Works Annual Conference, Denver, CO. 1998. Volatile N-​nitrosamine formation after intake of nitrate at the ADI
Tokar EJ, Benbrahim-​Tallaa L, Ward JM, et al. 2010a. Cancer in experimen- level in combination with an amine-​rich diet. Environ Health Perspect,
tal animals exposed to arsenic and arsenic compounds. Crit Rev Toxicol, 106(8), 459–​463. PMCID: PMC1533225.
40(10), 912–​927. PMCID: PMC3076186. Vieira V, Hoffman K, and Fletcher T. 2013. Assessing the spatial distribution
Tokar EJ, Diwan BA, and Waalkes MP. 2010b. Arsenic exposure transforms of perfluorooctanoic acid exposure via public drinking water pipes using
human epithelial stem/​progenitor cells into a cancer stem-​like pheno- geographic information systems. Environ Health Toxicol, 28, e2013009.
type. Environ Health Perspect, 118(1), 108–​115. PMCID: PMC2831952. PMCID: PMC3761072.
Tokar EJ, Person RJ, Sun Y, Perantoni AO, and Waalkes MP. 2013. Chronic Villanueva CM, Cantor KP, Cordier S, et  al. 2004. Disinfection byproducts
exposure of renal stem cells to inorganic arsenic induces a cancer pheno- and bladder cancer:  a pooled analysis. Epidemiology, 15(3), 357–​367.
type. Chem Res Toxicol, 26(1), 96–​105. PMCID: PMC3921970. PMID: 15097021.
Tokar EJ, Qu W, and Waalkes MP. 2011. Arsenic, stem cells, and the devel- Villanueva CM, Cantor KP, Grimalt JO, et  al. 2006. Assessment of lifetime
opmental basis of adult cancer. Toxicol Sci, 120 Suppl 1, S192–​S203. exposure to trihalomethanes through different routes. Occup Environ
PMCID: PMC3043086. Med, 63(4), 273–​277. PMCID: PMC2078091.
Trenberth KE. 2011. Changes in precipitation with climate change. Climate Villanueva CM, Cantor KP, Grimalt JO, et  al. 2007. Bladder cancer and
Res, 47(1-​2), 123–​138. exposure to water disinfection by-​products through ingestion, bathing,
Tricker AR. 1997. N-​nitroso compounds and man: sources of exposure, endog- showering, and swimming in pools. Am J Epidemiol, 165(2), 148–​156.
enous formation and occurrence in body fluids. Eur J Cancer Prev, 6(3), PMID: 17079692.
226–​268. PMID: 9306073. Villanueva CM, Fernandez F, Malats N, Grimalt JO, and Kogevinas M. 2003.
US Environmental Protection Agency (EPA). 2000a. Arsenic Occurrence in Meta-​analysis of studies on individual consumption of chlorinated drink-
Public Drinking Water Supplies. Washington, DC: US Environmental ing water and bladder cancer. J Epidemiol Community Health, 57(3),
Protection Agency. EPA-​815-​R-​00-​23. 166–​173. PMCID: PMC1732410.
US EPA. 2000b. Technologies and Costs for Removal of Arsenic from Drinking Villanueva CM, Gracia-​Lavedan E, Bosetti C, et  al. 2017. Colorectal cancer
Water. Washington, DC:  Office of Water, US Environmental Protection and long-​term exposure to trihalomethanes in drinking water:  a multi-
Agency. Report No. 815R-​00-​028. center case-​control study in Spain and Italy. Environ Health Perspect,
US EPA. 2003. National Primary Drinking Water Regulations:  Stage 2 125(1), 56–​65. PMID: 27383820.
Disinfectants and Disinfection Byproducts Rule; National Primary Vineis P. 2004. A self-​fulfilling prophecy: are we underestimating the role of the
and Secondary Drinking Water Regulations:  Approval of Analytical environment in gene-​environment interaction research? Int J Epidemiol,
Methods for Chemical Contaminants (Vol. 168). Washington, DC:  US 33(5), 945–​946. PMID: 15319401.
Environmental Protection Agency. Vitousek PM, Aber JD, Howarth RW, et al. 1997. Human alteration of the global
US EPA. 2005. Economic Analysis for the Final Stage 2 Disinfectant and nitrogen cycle: sources and consequences. Ecol Appl, 7(3), 737–​750.
Disinfectant Byproducts Rule. Washington, DC:  US Environmental Walker M, Seiler RL, and Meinert M. 2008. Effectiveness of household
Protection Agency. reverse-​osmosis systems in a Western U.S. region with high arsenic in
US EPA. 2006. National Primary Drinking Water Regulations:  Stage 2 groundwater. Sci Total Environ, 389(2–​3), 245–​252. PMID: 17919687.
Disinfectants and Disinfection Byproducts Rule, The Federal Register Ward MH, Cerhan JR, Colt JS, and Hartge P. 2006. Risk of non-​Hodgkin lym-
(Vol. 71, pp. 388). Washington, DC: US Environmental Protection Agency. phoma and nitrate and nitrite from drinking water and diet. Epidemiology,
US EPA. 2012. Fallon, NV: Pooling Resources to Construct Arsenic Treatment 17(4), 375–​382. PMID: 16699473.
Facility. Available from:  http://​water.epa.gov/​lawsregs/​rulesregs/​sdwa/​ Ward MH, Heineman EF, Markin RS, and Weisenburger DD. 2008.
arsenic/​nv_​fallon.cfm. Accessed May 3, 2015. Adenocarcinoma of the stomach and esophagus and drinking water and
US EPA. 2015. Drinking Water Health Advisory for the Cyanobacterial dietary sources of nitrate and nitrite. Int J Occup Environ Health, 14(3),
Microcystin Toxins. Washington, DC:  US Environmental Protection 193–​197. PMID: 18686719.
Agency. EPA-​820R15100. Ward MH, Kilfoy BA, Weyer PJ, et  al. 2010. Nitrate intake and the risk of
US EPA. 2016. Regulated Drinking Water Contaminants:  Inorganic. Available thyroid cancer and thyroid disease. Epidemiology, 21(3), 389–​ 395.
from: https://​www.epa.gov/​ground-​water-​and-​drinking-​water/​table-​regulated-​ PMCID: PMC2879161.
drinking-​water-​contaminants -​ Inorganic. Accessed September 23, 2016. Ward MH, Mark SD, Cantor KP, et  al. 1996. Drinking water nitrate and
US Food and Drug Administration (FDA). 2012. Arsenic in Rice: Full Analytical the risk of non-​ Hodgkin’s lymphoma. Epidemiology, 7(5), 465–​ 471.
Results from Rice/​Rice Product Sampling, September 2012. Available PMID: 8862975.
from:  http://​www.fda.gov/​Food/​FoodborneIllnessContaminants/​Metals/​ Ward MH, Rusiecki JA, Lynch CF, and Cantor KP. 2007. Nitrate in public water
ucm319916.htm. Accessed November 29, 2012. supplies and the risk of renal cell carcinoma. Cancer Causes Control,
Ueno Y, Nagata S, Tsutsumi T, et al. 1996. Detection of microcystins, a blue-​ 18(10), 1141–​1151. PMID: 17717631.
green algal hepatotoxin, in drinking water sampled in Haimen and Fusui, Ward MH, Sinha R, Heineman EF, et al. 1997. Risk of adenocarcinoma of the
endemic areas of primary liver cancer in China, by highly sensitive immu- stomach and esophagus with meat cooking method and doneness prefer-
noassay. Carcinogenesis, 17(6), 1317–​1321. PMID: 8681449. ence. Int J Cancer, 71(1), 14–​19. PMID: 9096659.
Vahter M and Marafante E. 1987. Effects of low dietary intake of methionine, Weyer PJ, Cerhan JR, Kross BC, et al. 2001. Municipal drinking water nitrate
choline or proteins on the biotransformation of arsenite in the rabbit. level and cancer risk in older women: the Iowa Women’s Health Study.
Toxicol Lett, 37(1), 41–​46. PMID: 3590229. Epidemiology, 12(3), 327–​338. PMID: 11338313.
Vahter M. 1999a. Variation in human metabolism of arsenic. In: Chappell WR, Wheeler DC, Nolan BT, Flory AR, DellaValle CT, and Ward MH. 2015.
Abernathy CO, and Calderon RL (Eds.), Arsenic: Exposure and Health Modeling groundwater nitrate concentrations in private wells in Iowa. Sci
Effects (pp. 267–​279). Amsterdam: Elsevier. Total Environ, 536, 481–​488. PMID: 26232757.
Vahter M. 1999b. Methylation of inorganic arsenic in different mamma- Winquist A, Lally C, Shin HM, and Steenland K. 2013. Design, methods, and
lian species and population groups. Sci Prog, 82 (Pt 1)(Pt 1), 69–​88. population for a study of PFOA health effects among highly exposed mid-​
PMID: 10445007. Ohio Valley community residents and workers. Environ Health Perspect,
Vahter M and Concha G. 2001. Role of metabolism in arsenic toxicity. 121(8), 893–​899. PMCID: PMC3734501.
Pharmacol Toxicol, 89(1), 1–​5. PMID: 11484904. World Health Organization (WHO). 2011. WHO Guidelines for Drinking-​
Valenzuela OL, Drobna Z, Hernandez-​Castellanos E, et al. 2009. Association of Water Quality, 4th edition. Available from:  http://​www.who.int/​water_​
AS3MT polymorphisms and the risk of premalignant arsenic skin lesions. sanitation_​health/​publications/​2011/​dwq_​guidelines/​en.
Toxicol Appl Pharmacol, 239(2), 200–​207. PMCID: PMC2910909. Wright K, Kelly J, and Zegarra, AV. 1997. Macchu Picchu: ancient hydraulic
van Leeuwen JA, Waltner-Toews D, Abernathy T, Smit B, Shoukri M, 1999. engineering. J Hydraul Eng, 123, 838–​843.
Associations between stomach cancer incidence and drinking water con- Xia J, Li Y, Yang Q, et  al. 2012. Arsenic trioxide inhibits cell growth and
tamination with atrazine and nitrate in Ontario (Canada) agroecosystems, induces apoptosis through inactivation of notch signaling pathway in
1987–​1991. Int J Epidemiol 28, 836–​840. breast cancer. Int J Mol Sci, 13(8), 9627–​9641. PMCID: PMC3431819.
328

328 PART III:  THE CAUSES OF CANCER


Yu S, Zhao N, and Zi X. 2001. The relationship between cyanotoxin (microcys- Zhang MD. 1993. [A population-​based case control study of primary liver
tin, MC) in pond-​ditch water and primary liver cancer in China. Zhonghua cancer in Fusui]. Zhonghua Liu Xing Bing Xue Za Zhi, 14(1), 14–​18.
Zhongliu Zazhi, 23(2), 96–​99. PMID: 11783030. PMID: 8504447.
Yu SZ. 1989. Drinking water and primary liver cancer. In: Tang ZY, Wu MC, Zhao N, Yu S, and Sun WM. 1994. [Interaction among the relative risk factors
and Xia SS (Eds.), Primary Liver Cancer (pp. 30–​37). Beijing:  China of primary liver cancer in a case-​control study]. Zhonghua Liu Xing Bing
Academic Publishers. Xue Za Zhi, 15(2), 90–​93. PMID: 7923340.
Yu SZ and Chen G. 1994. Blue-​green algae toxins and liver cancer. Chinese J Zhou L, Yu H, and Chen K. 2002. Relationship between microcystin in drink-
Cancer Res, 6(1), 9–​17. ing water and colorectal cancer. Biomed Environ Sci, 15(2), 166–​171.
Zeegers MP, Selen RF, Kleinjans JC, Goldbohm RA, and van den Brandt PMID: 12244757.
PA. 2006. Nitrate intake does not influence bladder cancer risk:  the Zwolak I and Zaporowska H. 2012. Selenium interactions and toxicity: a
Netherlands cohort study. Environ Health Perspect, 114(10), 1527–​1531. review. Selenium interactions and toxicity. Cell Biol Toxicol, 28(1), 31–​
PMCID: PMC1626405. 46. PMID: 21913064.
Zegura B, Straser A, and Filipic M. 2011. Genotoxicity and potential carcino-
genicity of cyanobacterial toxins: a review. Mutat Res, 727(1–​2), 16–​41.
PMID: 21277993.
 329

19 Diet and Nutrition

MARJORIE L. MCCULLOUGH AND WALTER C. WILLETT

OVERVIEW consumption (Chapter 12). Associations with diet are also covered in


cancer-​specific chapters, where applicable. The goal of this chapter is
The formal study of diet, nutrition, and cancer is still relatively young, to provide a “wide-​angle” perspective on these interrelated factors that
with most epidemiological studies and randomized controlled trials affect cancer.
(RCTs) having occurred in the past 20–​30 years. Despite the method-
ological challenges of studying diet and cancer in free-​living popula-
tions, there is scientific consensus that overweight and obesity increase Other Authoritative Reviews
the risk of certain cancers, as well as growing evidence that dietary The World Cancer Research Fund (WCRF) and its American affiliate,
patterns rich in vegetables, fruits, and whole grains and low in red the American Institute for Cancer Research (AICR), have a valuable
and processed meat are associated with lower risk of colorectal cancer series of comprehensive systematic literature reviews of diet, physical
and total cancer mortality. Although it is more difficult to isolate the activity, obesity, and alcohol and cancer. These have broader cover-
specific components of diet that affect risk, several key factors appear age of nutritional issues than the reviews by the International Agency
to play a role. Dietary composition appears to operate by affecting for Research on Cancer (IARC) and are updated more frequently. The
energy intake and also independent of energy intake. Despite extensive most recent complete WCRF/​AICR report, published in 2007 (WCRF/​
research, evidence does not support an important impact of the mac- AICR, 2007), synthesized the evidence for 20 cancers and based its
ronutrient composition of diet on cancer risk. Individual nutrients and conclusions largely on meta-​analyses. The WCRF/​AICR “Continuous
phytochemicals, including folate, vitamin D, calcium, and carotenoids Update Project (CUP)” follows a staggered schedule for updating the
(such as lycopene), appear to be associated with lower risk of certain evidence for each cancer site and more recently for cancer survivors.
cancers and are under active investigation. However, thus far, RCTs do An expert panel reviews and judges the evidence according to crite-
not support chemoprevention with high-​dose antioxidant supplements, ria for inferring causality, classifying relationships as “convincing,”
and in some cases these may cause harm. The assessment of lifelong “probable,” “limited-​suggestive,” “limited-​no conclusion,” and “sub-
diet and timing of exposures continues to be challenging. Future direc- stantial effect on risk unlikely.” Throughout this chapter, we will refer
tions in diet and cancer research include the examination of early life to the WCRF/​AICR reports, in addition to studies of nutritional bio-
exposures in relation to carcinogenicity, the role of the microbiome markers and dietary patterns (not covered in the reports), and to other
in cancer etiology, and risk factors for cancer subtypes defined by the high quality meta-​analyses and individual studies as appropriate.
molecular characteristics of tumors.

Estimates of Attributable Fraction


INTRODUCTION
Estimates of the proportion of cancers that can be attributed to
The relationship between nutritional factors and human cancer has dietary factors varies depending on the definition of “diet,” popula-
been debated for decades, but it is only in the past 20–​30 years that tion analyzed (e.g., national, global, individual studies), methodol-
large epidemiologic studies and RCTs have begun to examine the ogy employed, and period of study. Diet is a complex entity that can
issue systematically. Dietary factors are believed to contribute to the be defined narrowly to include only the composition and volume of
large geographic and temporal variations in cancer rates observed food consumed, or more broadly to encompass related factors such as
among human populations and to changes in rates among migrants energy balance, body weight, distribution of body fat, and metabolic
who move to countries with higher or lower rates than the country of byproducts of digestion. In 1981, Doll and Peto estimated that 35%
origin. A series of ecological reports in the 1970s showed high corre- of cancer deaths in the United States in the late 1970s could be attrib-
lations between specific dietary factors and cancer rates (e.g., fat and uted to diet, with a range of acceptable estimates of 10%–​70% (Doll
breast cancer, meat and colon cancer) across countries, and stimulated and Peto, 1981). This figure was calculated by summing the estimated
hypotheses on diet and cancer (Armstrong and Doll, 1975). Initially, fraction of deaths from specific cancer sites that might be avoided by
case-​control studies supported a role of these and other dietary factors practical dietary means (for example, stomach 90%, female breast
in cancer etiology (WCRF/​AICR, 1997). Over time, studies of diet 50%, bladder 20%, other types 10%) (Doll and Peto, 1981). The Doll
and cancer became progressively larger, and prospective investiga- and Peto analysis did not provide separate estimates of the attributable
tions, described herein, refuted some but not all of these associations fractions for body weight and physical inactivity, although the text
(WCRF/​AICR, 2007). Meta-​analyses and pooling of individual-​level stated that “[o]‌vernutrition should perhaps come first rather than last
data have also improved the precision and reproducibility of risk esti- on a list of aspects of diet which may affect the incidence of cancer,
mates. Current and future prospective studies using repeated measures even though the relevant mechanisms remain obscure.”
of diet, biological markers, and tumor molecular phenotype will con- In 2015, the WCRF/​AICR proposed new estimates, based on sys-
tinue to advance our understanding of the link between diet and cancer tematic reviews of the updated literature, and regional data on the
etiology. prevalence of known risk factors for various types of cancer. The latest
This chapter focuses on research regarding dietary composition in WCRF/​AICR update estimated that 20 to 22% of all incident cancers
relation to the most common forms of cancer, the most thoroughly in the United States and United Kingdom were due to the combination
studied exposures, and issues of current interest. Because of the of diet, physical inactivity, and overweight/​obesity; this figure rose to
broad nature of the topic, we will only briefly mention related topics 29% when the analysis was restricted to the 13 most common cancers
such as energy balance, obesity and body composition (Chapter 20), (WCRF International, 2015). The estimated fractions of all cancers
physical inactivity and sedentary behavior (Chapter 21), and alcohol preventable by diet in China and Brazil were about 15%, where obesity

329
30

330 PART III:  The Causes of Cancer


rates are lower (estimates rose to 19 and 22% for the 13 most common Inherent Challenges in Measuring Dietary Intake
cancers, respectively). The estimates for the United States were lower
than those of Doll and Peto (1981), probably because WCRF/​AICR Diet is a complex exposure composed of hundreds of nutrients and
considered only specific dietary factors for which the evidence for cau- non-​nutrient components, some known, some yet to be fully char-
sality was judged to be “convincing” or “probable.” acterized. Dietary behaviors tend to cluster, complicating efforts to
Other estimates have attempted to separate the effects of dietary isolate the effects of individual foods. Nutrients and foods are con-
composition from related factors such as overweight/​obesity, physi- sumed in combinations that may enhance or block their absorption,
cal inactivity, and alcohol consumption. Colditz and Wei attributed metabolism, or physiologic effects. For example, tomatoes consumed
5.0% of incident cancers in the United States specifically to dietary as part of a highly processed diet with large amounts of refined car-
composition, largely because of high consumption of red and pro- bohydrate may have a different effect on cancer risk than tomatoes
cessed meat and low consumption of folate (Colditz and Wei, 2012). consumed as part of a Mediterranean diet alongside fish, vegetables,
Parkin proposed similar attributable fraction estimates for the United nuts, and olive oil. Food processing (e.g., milling, fermentation), stor-
Kingdom, although consideration was also given to low consumption age (e.g., salting, drying, freezing, bottling) and preparation methods
of fruit, vegetables, and fiber and high intake of salt as well as meat (e.g., steaming, grilling) can affect nutrient concentration in a food,
(Parkin, 2011). In 2003, the IARC estimated attributable fractions for and can add carcinogens or unknown substances. The timing of a
low fruit and vegetable intake alone at 5%–​12% (IARC, 2003). Given nutritional exposure may modify its effects on cancer risk, although
that many dietary hypotheses remain unresolved and that the long-​ this is seldom recognized in advance. Vitamin supplements are used
term impact of early life exposures is largely unexplored, estimates by over half the US population (Dickinson et  al., 2014), and some
of the percentage of cancers attributable to dietary composition alone vitamin formulations provide concentrated, high doses that may have
may continue to change beyond the current estimates of 5%–​12%. different physiologic effects than when derived from food. In addi-
tion, individuals eat varied diets formed by individual preference,
cultural models, intolerance, and influenced by lifestyle and avail-
Measures of Dietary Intake ability. The complexity of diet and the challenge in quantifying intake
in free-​living populations are aptly illustrated in a photojournalistic
Measurement of diet in free-​living populations is a comprehensive series on the typical diet in the week of a family in various parts of
field of study with specific methodological considerations, and read- the world (Menzel, 2005).
ers are referred elsewhere for a detailed description (Willett WC,
2013). Most epidemiologic studies of cancer risk in humans use
Food Frequency Questionnaires (FFQs) to estimate diet, because Dietary Patterns
these are designed to assess usual intake efficiently for time periods
of up to 1  year, thereby avoiding the day-​to-​day fluctuations that One method of capturing multiple correlated aspects of diet is to char-
affect 24-​hour recalls. FFQs consist of a food list and questions on acterize intake in terms of dietary patterns instead of single foods or
frequency of consumption, and some include portion sizes (Willett nutrients. This is illustrated by contrasts between the “Mediterranean
WC, 2013). FFQs can be simple or complex, short or long, and are diet” and the “Western diet,” as discussed in Chapter  20, and is
population specific, reflecting commonly consumed foods and con- expanded on later in this chapter.
stituents of interest in a study population. Their use has been shown
to be sufficiently valid for assessing intake of foods and nutrients,
and identifying important diet–​disease relationships in large popula- Study Designs
tion studies (Block et al., 1990; Hu et al., 1999; Rimm et al., 1992; The study designs most commonly used to examine the relationship of
Willett et al., 1985). FFQs compare well with biochemical indicators diet and cancer risk in humans are summarized in Figure 19–​1 (Harris
of intake and more detailed questionnaire assessments (Willett WC, et  al., 2009). Each design contributes valuable information, but all
2013). Weighed food records or 24-​hour dietary recalls reflect only have strengths and weaknesses. Ecological studies and migrant studies
very recent intake. However, if collected repeatedly over a year, they play an important role in generating hypotheses about diet and can-
could approximate usual intake, and have the advantage of measur- cer, including interest in the 1970s in the fat–​breast cancer and meat–
ing exactly what participants eat and drink (assuming an adequately colon cancer hypotheses (Armstrong and Doll, 1975). Major changes
detailed nutrient database). Whereas serial 24-​hour recalls were pro- in disease rates within a population provide key evidence that expo-
hibitively expensive in the past due to interviewer and training costs, sures acquired during life have an important effect on cancer risk in
these methods may prove feasible with technological advancements populations. Through the 1990s, most information on diet and cancer
(Kirkpatrick et  al., 2014), including applications to smartphones was obtained from case-​control studies. Subsequently, concern about
and tablets. Whether large populations will provide data on a suffi- recall bias (the diagnosis influencing self-​reported diet) and selection
cient number of days to provide meaningful assessment of usual diet bias (overly health-​conscious controls being more likely to participate)
remains to be demonstrated. Combining these methods with an FFQ has limited their use in studies of diet and cancer. Indeed, many of
may lead to improved precision in dietary exposure assessment. the findings from early case-​control studies have not been confirmed
in subsequent research. Public health guidelines for diet and nutrition
tend to place the most weight on RCTs and prospective cohort studies,
Biomarkers which have complementary strengths and weaknesses, and for which
Biochemical indicators of diet can be useful in some situations, but for the exposure precedes cancer diagnosis. Although RCTs can reduce
many dietary factors of interest, such as total fat, fiber, and sucrose, or eliminate confounding, they are expensive and are not always fea-
no useful indicators exist. Recently, studies of the food metabolome sible or ethical. For example, an RCT of processed meat in relation
have identified potential biomarkers of foods such as red meat (3-​ to incident colorectal cancer is both infeasible and unethical, analo-
methylhistidine) (Cross et al., 2011) and whole grains (alkylresorcinols) gous to prohibitions against RCTs of smoking or excess weight gain
(Kyro et al., 2014a, 2014b; Scalbert et al., 2014). To be useful in epide- and cancer risk. Systematic literature reviews and meta-​analyses can
miological studies, the within-​person variation of biomarkers will need to also contribute importantly to understanding diet–​cancer relationships,
be relatively low (Van Dam and Hunter, 2013). Other biomarkers of inter- provided that the inclusion or exclusion of individual studies is ade-
est include DNA specimens that permit the testing of nutritional hypothe- quately explained and justified and the definitions of dietary and other
ses using Mendelian randomization. This approach examines variation in exposures are appropriately harmonized across studies. Publication
genes of known function to study the causal effect of a related exposure bias is a serious concern in meta-​analyses of published studies. Large
on cancer risk in non-​experimental studies (Gray and Wheatley, 1991). pooled analyses of individual-​level data from consortia of prospective
Studies of gene–​diet interactions also hold promise, but these require studies have advantages over meta-​analyses, because publication bias
very large numbers, and few clear examples exist at present. is minimized, original data are harmonized, and a uniform approach
 31

Diet and Nutrition 331


Strength of Evidence adenocarcinoma (IARC; Lauby-​Secretan, Scoccianti, Loomis, et al.,
2016). The strongest empirical support for mechanisms explaining
these associations involves the endocrine and metabolic effects of
Randomized trials Prospective cohorts obesity. Hormonal effects primarily involve alterations of peptide
of disease outcomes of disease outcomes (e.g., IGF, insulin) and steroid (estradiol, testosterone) hormones and
their binding proteins (Calle and Kaaks, 2004). For certain cancers,
such as esophageal adenocarcinoma and gall bladder tumors, chronic
Randomized trials of local inflammation from esophageal reflux or recurrent gallstones
physiologic measures plays an important role. Adipose tissue also generates multiple pro-​
inflammatory mediators.
The effects of dietary composition on long-​term weight change
and adiposity are of great scientific interest and public health impor-
Retrospective case-control
studies of disease outcomes tance. In 2007, WCRF/​AICR characterized the evidence as “probable”
that energy-​dense foods, sugary drinks, and fast foods increase risk
(WCRF/​AICR, 2007). A meta-​analysis of 53 randomized clinical trials
found that weight loss was slightly greater with low-​carbohydrate than
Ecologic studies Prevalence studies Animal studies
with low-​fat diets (Tobias et al., 2015). Many aspects of diet consistent
with a Mediterranean dietary pattern appear to contribute to long-​term
weight maintenance (Mozaffarian et al., 2011). In a 6-​year trial of diet
Case series/Case reports
and weight loss with only 2 years of active intervention, those assigned
to a Mediterranean diet had greater sustained weight loss compared
with those on low-​fat or high-​fat diets (Shai et al., 2008).
Figure  19–​1.  Hierarchy of quality and strength of evidence of different
study designs for evaluating causation of chronic disease. Studies of physi-
ological endpoints may not fully account for all effects of an intervention;
thus evidence is strongest when derived from well-​conducted studies of MACRO-​AND MICRONUTRIENTS
disease endpoints. Well-​controlled prospective observational studies and
well-​performed randomized clinical trials have different, and highly com- The relationship of diet to cancer risk can be evaluated in terms of
plementary, strengths and limitations. The major strength of randomized either individual dietary constituents or broad dietary patterns. This
trials is to minimize residual confounding; limitations include potential section of the chapter discusses recent epidemiologic research on
lower generalizability to external populations, the inability to evaluate the macro-​and micronutrients and their relationship (or lack of relation-
effects of many risk factors of interest (e.g., smoking, biomarker levels) ship) to cancer etiology, biology, and prevention.
due to ethical or practical limitations, inadequacy of duration and/​or dose
tested, challenges in testing multiple doses, noncompliance, unblinding,
differential loss to follow-​up, and treatment crossover. The major limita- Dietary Fat
tion of prospective observational studies is potential residual confound- In the 1980s, dietary fat was postulated to be the most important aspect
ing, whereas major strengths generally include the converse of each of of diet with respect to cancer risk. This hypothesis was largely fueled
the major limitations of randomized trials. Thus, evidence is most robust by the striking international correlations between per capita fat intake
when studies of both designs provide concordant results. Source:  Harris and incidence or death rates from breast, colon, and prostate cancer
WS, et al. (2009). (Armstrong and Doll, 1975; Prentice et al., 1988). For breast cancer,
the positive associations seen in some case-​control studies (Howe
et al., 1990; WCRF/​AICR, 1997) have not generally been replicated
can be used to define exposure, cut-​points, covariates, and statistical in prospective studies (Key et  al., 2011; Smith-​Warner et  al., 2001;
modeling (Smith-​Warner et al., 2006). WCRF/​AICR, 2010), although a recent large European study found
Recognizing these methodologic considerations, the remainder of a weak positive association of saturated fatty acids with ER+PR+
this chapter discusses elements of diet that have received particular and HER2-​tumor subtypes (Sieri et  al., 2014). The lack of agree-
attention with regard to cancer risk. ment between the case-​control and cohort studies reinforces concerns
about recall and selection biases in case-​control studies of diet. The
Women’s Health Initiative (WHI) study, designed and implemented
ENERGY BALANCE at the end of the previous century (The Women’s Health Initiative
Study Group, 1998), included a randomized clinical trial to test the
There is strong evidence that energy balance has important effects effect of a low-​fat diet intervention (by instruction) on breast cancer
on the incidence of some cancers, as described in detail in Chapter incidence. The initial goal was to reduce fat intake among women in
20. Experimental studies in animals during the first half of the twen- the intervention arm to 20% of calories, and compare this to women
tieth century indicated that energy restriction profoundly reduced the consuming approximately 37% of calories from fat. The actual con-
development of mammary tumors (Tannenbaum, 1942; Tannenbaum trast was smaller, however, because women in the intervention arm
and Silverstone, 1953). This finding has consistently been replicated reported consuming 27% of calories from fat and the control arm
in a wide variety of mammary and other tumor models (Birt et al., averaged 33%. No differences were observed in plasma lipid fractions
1992; Nair et al., 1995; Ross and Bras, 1971; Weindruch and Walford, known to be influenced by dietary fat between the intervention and
1982). For example, 30% restriction in energy intake reduces mam- control groups, indicating that little or no difference in fat intake was
mary tumors by as much as 90% (Boissonneault et  al., 1986). In achieved between the two groups (Willett, 2010). At trial completion,
humans, energy balance is best assessed by determining body size there was a non-​significantly lower risk of breast cancer in the inter-
and composition, or changes in weight or body composition, rather vention arm (Prentice RL et al., 2006). In an extended follow-​up after
than by comparing energy intake to expenditure, since both param- active intervention, there was no difference in rates of breast or other
eters are measured with poor precision in free-​living humans (Willett cancers between the two randomized groups (Thomson et al., 2014b).
WC, 2013). The IARC found sufficient evidence that excess body This inability to test the original hypothesis illustrates at least two
fat (starting at ≥25 kg/​m2) is associated with increased risk of can- serious limitations in using randomized trials to study dietary factors
cers of the gastric cardia, colon and rectum, liver, gall bladder, pan- and cancer etiology. The first is the difficulty of achieving and sustain-
creas, breast (post-​menopause), corpus uteri, ovary, kidney (renal ing the desired contrast in the exposure of interest. The second is that
cell), thyroid and multiple myeloma, meningioma, and esophageal even a small change in the exposure (in this case dietary fat intake)
32

332 PART III:  The Causes of Cancer


can secondarily change other factors (in this case, weight loss of 1–​2 Protein
kg among women on the lower fat diet) (Prentice et al., 2006). Had
there been a statistically significant reduction in the incidence rate of Epidemiologic studies have not found a clear association between an
breast or other cancers, the intervention could not have distinguished overall high intake of protein in adulthood and risk of cancer. Most
weight loss from reductions in fat intake. In a second large random- studies find no evidence of a harmful association with some foods that
ized trial of dietary fat reduction, conducted among women with high are major sources of protein, such as fish, poultry, and plant foods.
mammographic density, a non-​significant increase in risk of breast Red and processed meat and dairy products are other major protein
cancer was seen among those randomized to a low-​fat diet (Martin sources, discussed later in the chapter.
et al., 2011).
The debate over dietary fat intake and breast cancer incidence con-
tinues with respect to the timing of exposure, type of fat, and disease MICRONUTRIENTS
subtypes. Limited data suggest that animal fat intake during young
adulthood may influence risk of premenopausal breast cancer (Farvid Vitamin D
et  al., 2014b). High intake of polyunsaturated fatty acids increases
mammary cancer incidence in rodents, but has not been associated with Vitamin D is best known for its role in maintaining calcium homeosta-
increased risk of breast cancer in humans (Smith-​Warner et al., 2001). sis and bone health. It is found naturally in a limited number of foods
The findings regarding total dietary fat intake and prostate cancer (e.g., fatty fish and eggs), and is added to milk and other products in
have been inconsistent. Although earlier studies suggested a positive the United States and many other countries. It is also available from
relationship with animal fat (WCRF/​AICR, 1997), this association dietary supplements at a wide range of doses. The most important
has weakened over time (WCRF/​AICR, 2014b). A  large interna- source of vitamin D, however, is cutaneous production due to UVB
tional collaboration including over 5000 prostate cancer cases and > radiation. The recommended dietary allowance for vitamin D for
6600 controls found no strong evidence that circulating specific fatty adults is 600 IU/​day, with a tolerable upper limit of 4000 IU (Institute
acids were important predictors of prostate cancer risk (Crowe et al., of Medicine, 2011). A cup of milk contains about 100 IU of vitamin
2014). An unexpected modest positive association was observed D. In comparison, approximately 10,000–​20,000 IU is produced by a
with two omega-​3 fatty acids, eicosapentaenoic and docosapentae- fair-​skinned adult in a bathing suit who develops a light pink sunburn
noic acid. The significance of this finding is unclear, since it conflicts from UVB exposure (Holick, 2004). Higher levels of sun exposure
with meta-​analyses of studies of fish consumption, the major source convert the skin precursors of vitamin D to inert metabolites, so that
of these fatty acids (Szymanski et  al., 2010), which is inversely vitamin D toxicity from sun exposure is highly unlikely. The potential
associated with prostate cancer incidence (Lovegrove et  al., 2015; benefits of sun exposure for vitamin D production need to be balanced
Szymanski et al., 2010). A limitation of this study is that it included against the harmful effects of UV radiation for skin cancer, however
all incident cases of prostate cancer, instead of restricting the end- (see Chapter 62.4). Seasonal variation occurs in the circulating storage
point to advanced or fatal cases. Since many cases are diagnosed by form of vitamin D [25(OH)D], with a peak during summer months and
prostate specific antigen (PSA) screening, dietary factors associated a nadir in mid-​winter. This variation is accentuated in populations who
with screening tend to be positively associated with overall incidence live further from the equator (McCullough et al., 2010).
of prostate cancer. In 1941, Frank Apperly observed lower cancer mortality rates in
North American regions that have higher levels of solar radiation
(Apperly, 1941), an observation later hypothesized to reflect variations
in vitamin D exposure (Garland and Garland, 1980). In vitro and in
Carbohydrates vivo animal studies provide strong biological support for a role for
Carbohydrates comprise a diverse group of macronutrients that may vitamin D in cancer prevention. The active form of the vitamin, 1,25
differ in their effects on chronic disease risk. For example, carbohy- dihydroxyvitamin D (1,25(OH)2D), also called calcitriol, is the natural
drates with a high glycemic index (Wolever and Jenkins, 1986) cause ligand for the vitamin D receptor. This receptor is a nuclear transcrip-
larger spikes in postprandial blood glucose and insulin concentra- tion factor that modulates expression of over 200 genes, including
tions than those with a lower glycemic index (Miller, 1994). High some that regulate cell growth, limit inflammation, and reduce levels
glycemic carbohydrates also cause higher fasting insulin levels in of VEGF (Feldman et al., 2014).
insulin-​resistant states (Pereira et  al., 2002). Fasting plasma insu- The main storage form of vitamin D [25(OH)D] has a half-​life of
lin concentrations are inversely correlated with insulin-​like growth 3–​4 weeks. Serum concentrations of 25(OH)D reflect input from diet,
factor-​binding protein 1 (IGFBP-​1) and thus increase bioactive IGF-​ supplements, and skin synthesis and are considered the preferred bio-
1 concentrations (Giovannucci, 2001). The glycemic index (GI) of a marker of exposure. Prospective cohort studies with pre-​diagnostic
carbohydrate reflects its potential to raise blood sugar, whereas the measures of 25(OH)D provide the strongest support for an inverse
glycemic load (GL) considers both the GI and the amount of carbo- relationship between vitamin D and cancer risk in humans. The most
hydrate consumed. Neither parameter was associated with the risk of consistent evidence is for colorectal cancer, where the incidence rate is
colorectal or pancreatic cancer in a meta-​analysis (Mulholland et al., approximately one-​third lower in those with high versus low circulat-
2009). In contrast, the risk of endometrial cancer is consistently asso- ing levels of 25(OH)D (Lee et al., 2011a; Ma et al., 2011). The evi-
ciated with increased GL. An increase of 50 units of GL is associated dence for other cancers is mixed (Helzlsouer, 2010; Kim and Je, 2014;
with a 15% higher risk (WCRF/​AICR, 2013). Endometrial cancer is Stolzenberg-​Solomon et al., 2009; Wolpin et al., 2012). For prostate
also the cancer site most strongly associated with obesity and insulin cancer the evidence is highly inconsistent (Ahn et al., 2008; Xu et al.,
resistance. It has not yet been determined whether subsets of people 2014), although some studies suggest a “U-​shaped” association, with
are at higher cancer risk from dietary GL. higher risks at either end of the 25(OH)D distribution (Kristal et al.,
Grain consumption in the United States has increased by 41% 2014). Vitamin D metabolism in the prostate may vary by stage and
over the past several decades (Wells and Buzby, 2008), concurrent grade of the tumor (Tannour-​Louet et  al., 2014), complicating the
with national dietary guidelines to reduce total dietary fat intake. interpretation of the role of vitamin D in prostate cancer. Case-​control
Approximately 15% of total energy intake in the United States is studies of breast cancer suggest lower risk with higher 25(OH)D con-
from added sugars, with soda and energy drinks contributing 36% of centrations measured after diagnosis (Yin et al., 2010), but the findings
added sugars (Welsh et  al., 2011). Consumption of sugar-​sweetened are generally weak or null in prospective studies (Kim and Je, 2014),
beverages correlates with increasing energy intake (Vartanian et  al., suggesting possible disease-​related effects on vitamin D status.
2007), and contributes to weight gain and increasing prevalence of Determining optimal 25(OH)D levels for disease prevention is chal-
overweight and obesity (WCRF/​AICR, 2007). Therefore, excess sugar lenging because circulating concentrations vary considerably across
consumption, especially as beverages, likely increases risk of cancer populations, and assays differ in their quantification of 25(OH)D
indirectly through weight gain. (Binkley et al., 2014). A large, ongoing, international consortium of 21
 3

Diet and Nutrition 333


prospective cohorts (Circulating Biomarkers of Breast and Colorectal or by increasing the misincorporation of uracil in DNA (Blount and
Cancer Consortium) is addressing this issue by calibrating serum Ames, 1994). Considerable evidence from both case-​ control and
25(OH)D levels to a single laboratory and using the same method cohort studies supports a lower risk of colon cancer with higher folate
for standardizing serum 25(OH)D concentrations to account for sea- intake (Kennedy et al., 2011), and some evidence suggests that higher
sonal variation in analyses of prediagnostic vitamin D and breast and folate intake may attenuate the risk of colorectal cancer associated
colorectal cancer (Ziegler, et al., 2015). Preliminary results show sta- with alcohol (Nan et al., 2013). An association between a functional
tistically significant inverse associations between circulating 25(OH)D polymorphism in the folic acid metabolizing gene, methylene tetrahy-
(up to 100 nmol/​L) and colorectal (unpublished), but not breast cancer drofolate reductase (MTHFR), and incidence of colorectal cancer adds
risk (Visvanathan et al., 2015). support for a causal relationship (Zhao et al., 2013). Results of studies
Results of RCTs of vitamin D supplementation and cancer out- of circulating folate and risk of colorectal cancer have been incon-
comes have not definitively confirmed or negated a protective effect sistent, but blood levels of folate are influenced by genetic determi-
of vitamin D and cancer, due to questions about dose and crossover. nants of metabolism as well as intake. For example, in an analysis that
The large Women’s Health Initiative found no reduction in the risk included folate intake and MTHFR genotype, higher blood levels due
of colorectal (Wactawski-​Wende et  al., 2006)  or breast (Chlebowski to diet were associated with lower risk of colorectal cancer, but higher
et  al., 2008)  cancer among women randomized to 400 IU vitamin levels due to genotype were associated with higher risk, potentially
D plus 1000 mg calcium compared to the placebo group. However, due to blockage of a metabolic pathway that reduces risk (Lee et al.,
400 IU is now considered a low dose of vitamin D, and most women 2012). This illustrates some of the complexity of using biomarkers of
(including those on placebo) were taking vitamin D by the end of the dietary intake. In a randomized controlled trial among patients with
study. Secondary analyses of osteoporosis trials reported some lower- a recent history of colorectal adenomas, folic acid supplementation
ing of cancer risk, but the number of cases was generally too small to increased risk of more advanced adenomas (Cole et  al., 2007), rais-
be conclusive (Avenell et al., 2012; Lappe et al., 2007). No association ing concerns that very high intakes of this vitamin may actually be
with recurrent polyps was observed in a recent trial of 1000 IU vitamin carcinogenic. These conflicting observations have been postulated to
D and 1200 IU calcium (Baron et al., 2015). Ongoing trials of vita- reflect different stages of carcinogenesis, whereby high folate intake
min D supplementation and chronic disease risk in the United States might inhibit the initiation of neoplasia but promote the growth and/​
(Manson et al., 2012) and elsewhere (Manson and Bassuk, 2015) are or progression of existing lesions. Reassuringly, recent studies of
testing higher doses. folate intake continue to find an inverse association with colorectal
cancer risk, even among older individuals with very high intakes of
folate (Gibson et al., 2011; Stevens et al., 2011). In a detailed analysis
Calcium of the timing of folate intake in relation to colorectal adenoma and
cancer, greater folate intake was inversely associated with the risk of
Higher calcium intake has been shown to reduce the incidence of colorectal cancer when exposure occurred before the development
bowel tumors in animals (Lamprecht and Lipkin, 2001), possibly by of adenoma (Lee et al., 2011b). Lower risk of colorectal cancer was
precipitating bile acids. Calcium supplementation reduced the recur- observed only after a 12-​year delay. Also reassuringly, mortality due to
rence of adenomatous polyps by about 20% in one randomized trial colorectal cancer continued to decline steadily after implementation of
(Baron et al., 1999), but not in a recent trial of similar design (Baron folic acid fortification in 1998 in the United States (http://​seer.cancer.
et al., 2015). In observational studies, a pooled analysis of cohort stud- gov/​statfacts/​html/​colorect.html). The role of folate in numerous other
ies found a statistically significant 20% lower risk of colorectal cancer cancers has been studied, but the results are inconsistent and provide
in those with the highest versus lowest total calcium intake (Cho et al., no clear evidence of an association.
2004). Higher calcium intake has been associated with lower breast
cancer risk in some prospective studies (Cui and Rohan, 2006), but not
others (Larsson et al., 2009). The aforementioned randomized trial of Fiber
1000 mg calcium and 400 IU vitamin D found no reduction in either Interest in the relation between fiber intake and colon cancer was
colorectal (Wactawski-​Wende et al., 2006) or postmenopausal breast stimulated by Burkitt’s observation that colon cancer was rare in areas
cancer incidence (Chlebowski et  al., 2008)  over 7  years of the trial. of Africa where fiber consumption and stool bulk were high (Burkitt,
It is possible that the generally high calcium intake in both trial arms 1971). Fiber has been hypothesized to dilute potential carcinogens and
limited the ability to detect a modest reduction in risk. accelerate transit through the colon. The anti-​carcinogenic mechanisms
Higher calcium intake (> 2000 mg/​day) from diet and nutritional of a diet high in fiber and low in fat were explored in a recent study
supplements was associated with increased risk of total, lethal, of the microbiome. In a 2-​week food exchange experiment, O’Keefe
and high-​grade prostate cancer in 24-​years of follow-​up of men in et al. (2015) gave African American men a high-​fiber, low-​fat African-​
the Health Professionals Follow-​ up Study (Wilson et  al., 2015). style diet and rural Africans a high-​fat, low-​fiber Western-​style diet. In
Phosphorous intake was correlated with calcium and was also associ- comparison with their usual diets, the interventions resulted in recipro-
ated with increased risk. In addition, a meta-​analysis of 32 prospective cal alterations in mucosal biomarkers of cancer risk and in aspects of
studies found that dietary calcium, and its major source, dairy product the microbiota and metabolome related to cancer risk.
intake, was significantly associated with higher total prostate cancer In observational studies, dietary fiber was inversely associated
risk, but supplemental calcium intake was the only form significantly with risk of colorectal cancer in early case-control studies (Howe
associated with increased risk of fatal prostate cancer (Aune et  al., et  al., 1992; Trock et  al., 1990), but not consistently in subse-
2015). Measured levels of total and ionized serum calcium were asso- quent prospective studies. In a prospective US cohort of almost
ciated with higher risk of fatal prostate cancer, but only during the a half million retired men and women, no association was found
early years of follow-​up, suggesting that serum calcium might be a for dietary fiber intake and colorectal cancer risk (Schatzkin et al.,
marker of extant prostate cancer (Schwartz and Skinner, 2012). 2007). Similarly, in a large pooled analysis of over 700,000 men
Thus, while evidence supports a modest benefit of higher calcium and women from multiple prospective cohort studies, no associa-
intake in colorectal cancer prevention, the practical implications are tion was seen except a possible small increase in risk in those with
presently unclear because high calcium intake or dairy product con- very low fiber intake (Park et al., 2005). In contrast, a 2012 analysis
sumption has been associated with higher risk of prostate cancer from the large European Prospective Investigation into Cancer and
(Kushi et al., 2012). Nutrition (EPIC) cohort found statistically significantly lower colo-
rectal cancer risk with higher intake of dietary fiber, including fiber
from fruit, vegetables, and cereals (Murphy et al., 2012). The most
Folate recent WCRF/​AICR update of the epidemiologic evidence for colon
Folate deficiency has long been known to cause tumors in animals, cancer ranks “foods containing dietary fibre” (see http://​www.wcrf.
possibly by influencing gene expression through DNA methylation org/​sites/​default/​files/​Colorectal-​Cancer-​2011-​Report.pdf) to be
34

334 PART III:  The Causes of Cancer


convincingly related to lower risk (WCRF/​AICR, 2011). However, no benefit of high β-​carotene intake for lung cancer, and current can-
the inconsistency among large prospective studies remains unex- cer prevention guidelines (Kushi et al., 2012) and the US Preventive
plained, and is one of the few examples of important heterogeneity Services Task Force (Moyer and Force, 2014) specifically recommend
in findings among such studies. RCTs of supplemental wheat bran against taking β-​carotene supplements.
fiber (Alberts et  al., 2000) and isphaghula husk (psyllium fiber) Neither preformed vitamin A nor carotenoid intake has been con-
(Bonithon-​Kopp et al., 2000) failed to reduce the risk of recurrent sistently associated with risk of colon cancer, although these relation-
adenomatous polyps. A trial that combined instructions for both ships have been examined in several studies (WCRF/​AICR, 2011).
high fiber intake and a reduction in total fat also failed to reduce Similarly, β-​carotene—​as well as vitamins C and E—​did not affect
recurrent adenomatous polyps (Schatzkin et al., 2000). the recurrence of adenomatous colon polyps (Greenberg et al., 1994).
Higher intake of fiber has also been hypothesized to lower breast Recent pooled analyses of prospectively collected blood suggest that
cancer risk by interfering with enterohepatic metabolism of estro- several carotenoids may reduce breast cancer risk (Bakker et al., 2016;
gens (Gorbach and Goldin, 1987). Although most individual pro- Eliassen et  al., 2015). Both β-​carotene and α-​carotene serum levels
spective cohort studies have not found a statistically significant were inversely associated with ER–​breast tumors. These findings are
association of breast cancer risk with greater dietary fiber intake, consistent with a pooled analysis in which vegetable consumption
two meta-​analyses of 10 (Dong et  al., 2011)  and 15 (Aune et  al., was associated inversely with ER–​but not ER+ breast cancer (Jung
2012a) prospective cohort studies found a statistically significant et al., 2013).
5%–​10% lower risk of breast cancer associated with each 10 g of The question of whether lycopene, or tomato products, its major
increase in dietary fiber intake. In the large EPIC cohort, a weak food source, protect against prostate cancer risk has been studied
inverse relation was seen with fiber from vegetable sources, but not extensively in epidemiologic and clinical studies. Lycopene is the
with fiber from grains or fruits; this inverse association was pri- most abundant carotenoid measured in prostate tissue (Arab et  al.,
marily with estrogen receptor–​negative (ER–​) breast cancer (Ferrari 2001). Although the totality of the evidence for prostate cancer has not
et  al., 2013). Earlier life exposure to fiber may also impact breast been convincing (WCRF/​AICR, 2014b), a recent large pooled analy-
cancer risk. In a recent study, higher fiber intake during high school sis reported statistically significantly lower risk of aggressive prostate
was associated with a 16% lower risk of breast cancer overall, and cancer with higher prediagnostic circulating lycopene levels (Key
a 24% lower risk of premenopausal breast cancer (Farvid et  al., et al., 2015), adding important support to the possibility of benefit.
2016). Whether fiber or other components of high-​fiber foods, such
as carotenoids, are responsible for this beneficial association is
unclear. Vitamins C and E
Extensive damage to DNA, protein, and lipids can occur from oxidant
byproducts of smoking and normal metabolism. Although DNA repair
Carotenoids mechanisms and antioxidant defenses exist, they are imperfect (Ames
et  al., 1995). Antioxidants may reduce the risk of cancer by neutral-
Carotenoids, ubiquitous in fruits and vegetables, are fat-​soluble pig-
izing free radicals and reactive oxygen species that can damage DNA.
ments that absorb light energy for plant photosynthesis and protect
In humans, vitamin E is the major lipid-​soluble, membrane-​localized
chlorophyll from damage. Over 700 naturally occurring carotenoids
antioxidant, and vitamin C is the major water-​soluble antioxidant.
have been identified, and six (α-​carotene, β-​carotene, β-​cryptoxanthin,
Vitamin C can interfere with formation of nitrosamines—​carcinogens
lycopene, and lutein+zeaxanthin) comprise greater than 95% of total
formed endogenously from nitrogenous precursors in diet and tobacco
carotenoids in human blood (Maiani et al., 2009). These compounds
smoke—​in the stomach. However, chemoprevention trials of high-​risk
are hypothesized to reduce cancer risk by lowering oxidative stress
populations have not provided strong support for a benefit from anti-
and inflammation. Pro-​vitamin A carotenoids (α-​carotene, β-​carotene,
oxidants against cancer. Chemoprevention trials of stomach cancer in
and β-​cryptoxanthin) are precursors to vitamin A, which may influence
high-​risk populations have shown regression of gastric dysplasia from
carcinogenesis by regulating cell differentiation (Maiani et al., 2009).
several antioxidant nutrients (Correa et  al., 2000), but not a specific
Preclinical and biomarker-​ based studies in the 1980s (Menkes
benefit from vitamin C supplements (Blot et  al., 1993). A  secondary
et al., 1986; Stahelin et al., 1984) provided evidence for a protective
analysis of the aforementioned ATBC trial found a 34% lower inci-
relationship between carotenoid intake and risk of lung cancer after
dence of prostate cancer in heavy smokers who received supplemental
controlling for cigarette smoking. Inconsistencies were noted, how-
α-​tocopherol (50 mg/​day), despite no effect on lung cancer (The Alpha-​
ever, in that these studies did not show a relationship with preformed
Tocopherol Beta-​Carotene Cancer Prevention Study Group, 1994). The
vitamin A (Willett and Colditz, 2013), and a pooled analysis of seven
Selenium and Vitamin E Cancer Prevention Trial (SELECT), initiated
prospective cohort studies provided little support for a protective
partly because of the ATBC trial results for vitamin E, found no ben-
role of β-​carotene intake and lung cancer (Mannisto, Smith-​Warner,
efit from selenium for prostate cancer, and an unexpected borderline
Spiegelman, et  al., 2004). Counter to the hypothesis that β-​carotene
adverse effect among men randomized to vitamin E alone (400 IU/​
might protect against lung cancer, two large trials in smokers and/​or
day vitamin E of all rac α-​tocopheryl acetate) or in combination with
asbestos workers, the Alpa-​Tocopherol, Beta-​Carotene (ATBC) (The
selenium (200 µg/​d of L-​selenomethionine). This adverse effect was
Alpha-​ Tocopherol Beta-​ Carotene Cancer Prevention Study Group,
sustained and became statistically significant during post-​trial follow-​
1994) and the CARET (Omenn et al., 1996) trials reported statistically
up (Klein et al., 2011). The SELECT trial tested a higher dose of vita-
significant increases in lung cancer occurrence among those random-
min E than ATBC. However, an RCT using a similar vitamin E dose in
ized to 20–​30 mg β-​carotene per day. This is several times the amount
mostly non-​smokers found no effect of the vitamin on prostate cancer
typically present in the diet. However, in mostly non-​smokers, the
(Gaziano et al., 2009). Overall, epidemiologic studies and RCTs have
Physicians’ Health Study (Hennekens et al., 1996) and the Women’s
not supported a role of vitamin C or vitamin E in cancer prevention
Health Study (Lee et al., 1999) observed no effect of β-​carotene sup-
(Fortmann et al., 2013; WCRF/​AICR, 2007). Despite the lack of dem-
plements on lung or overall cancer incidence. Many explanations have
onstrated benefit, any aspect of diet that acted by blocking the initial
been offered for the lack of benefit, or even harm, in the randomized
steps in carcinogenesis would need to be present at that time, presum-
trials, including interference with the metabolism of other beneficial
ably two or three decades earlier in the case of smoking and lung can-
carotenoids by the relatively high dose of β-​carotene in the supple-
cer. This is far longer than the duration of any of the intervention trials.
ments, or by induction of carcinogen-​activating enzymes in the highly
oxidized lungs of smokers (Liu et al., 2003; Wang and Russell, 1999).
The lack of benefit from β-​carotene supplements does not exclude
Selenium
the possibility that other nutrients or biologically active constituents
in fruits and vegetables might prove to be protective in the future. Selenium defends against oxidative stress through selenoproteins,
However, epidemiologic evidence and randomized trials both indicate including selenium-​dependent glutathione peroxidases. The amount
 35

Diet and Nutrition 335


of selenium in food may vary up to 10-​fold, depending on the sele- however, global cancer-​
prevention guidelines recommend limiting
nium content of soil where the plant was grown or where the ani- sodium intake (WCRF/​AICR, 2007).
mal was raised. This variability makes food composition databases
for selenium unreliable (Panel on Dietary Antioxidants and Related
Compounds, 2000). Therefore, most epidemiologic evidence on sele- Plant Polyphenols
nium and cancer risk is from randomized trials and biomarkers of Polyphenols are secondary metabolites widely distributed in plant
selenium exposure. For example, several prospective studies have foods. There are four main classes:  flavonoids, lignans, phenolic
reported inverse associations between selenium levels and prostate acids and stilbenes. Polyphenols are of considerable interest in anti-
cancer incidence and progression using measures of selenium in toe- cancer research because of their antioxidant, anti-​inflammatory, anti-​
nails (Vogt et al., 2003; Yoshizawa et al., 1998), as a marker of sele- estrogenic, and anticarcinogenic properties (Adolphe et  al., 2010).
nium intake over the past year, or in the plasma or serum (Brooks Advances in measurement and reporting of the polyphenol content of
et  al., 2001; Li et  al., 2004; Nomura et  al., 2000). Selenium was foods (Harnly et al., 2006; Rothwell et al., 2013) facilitate population
strongly associated with reduced prostate cancer risk as a second- studies of these compounds. Herein, we focus on evidence for lignans
ary endpoint in one small trial of selenium supplementation and skin and flavonoids.
cancer (Clark et al., 1996). However, the SELECT trial (Klein et al.,
2001), the aforementioned large randomized placebo controlled trial
with selenium (200 µg) and vitamin E (400 IU) was ended 4  years Lignans
early, with the study showing no protective effect of selenium on total Lignans, found primarily in flaxseed and other seeds, berries, tea, and
prostate cancer incidence (Lippman et  al., 2009). This interpreta- wine, comprise one of three main groups of plant compounds classi-
tion of this trial was limited because it included only one fatal case fied as phytoestrogens, the other two being isoflavonoids and coumes-
of prostate cancer. Recent research suggests that polymorphisms in tans (Webb and McCullough, 2005). Studies in humans, animals, and
genes that code for selenoproteins should be considered when exam- cell culture provide support for a chemopreventive action of lignans,
ining the relationship between selenium biomarker concentrations potentially through anti-​ estrogenic, anti-​ angiogenic, proapoptotic,
and prostate cancer risk (Xie et al., 2016). and antioxidant mechanisms. The two primary lignans, matairesinol
(MAT) and secoisolariciresinol (SEC), are converted by intestinal
microflora to the biologically relevant mammalian lignans, enterodiol
Vitamin and Mineral Combinations (EDL) and enterolactone (ENL). These latter two compounds, mea-
sured in blood and urine, reflect only very recent intake. Some studies
Few trials have tested combinations of multivitamins and/​or minerals support an inverse association of these biomarkers with risk of post-
in relation to cancer incidence. In a nutritionally depleted population menopausal breast (Velentzis et al., 2009) and bladder (Zamora-​Ros
in Linxian, China, a trial of over 25,000 men and women found that et al., 2014) cancer. While evidence is inconsistent for dietary lignan
a cocktail of 50 μg selenium, 30 mg vitamin E, and 15 mg β-​carotene intake and prostate cancer risk (Saarinen et al., 2010), a recent meta-​
reduced total mortality, cancer-​specific mortality, and gastric cancer, analysis supports an inverse association with circulating enterolactone
compared to placebo (Blot et  al., 1993). Ten-​year post-​trial follow-​ (He et al., 2015). The short half-​life of these circulating compounds
up showed sustained benefits (Qiao et al., 2009). In contrast, as noted and the large between-​individual variability in metabolism present
earlier, most vitamin and/​or mineral supplement trials in nutritionally challenges to research in this area.
replete populations have not shown benefit (Fortmann et  al., 2013),
although benefits among subgroups with less than optimal diet cannot Flavonoids
be excluded. Recently, two RCTs conducted in populations presumed Flavonoids are bioactive, polyphenolic non-​nutrient constituents in
to be nutritionally replete found reduced incidence of all cancers com- plants. Common classes of flavonoids include anthocyanidins, flavo-
bined in individuals randomized to relatively low-​dose, multi-​nutrient nols, flavanones, flavones, flavanols, proanthocyanidins, and isofla-
supplements. In the Physicians’ Health Study (Gaziano et al., 2012), vones, the latter being the most thoroughly studied. Isoflavones, found
randomization of adult men to a Centrum Silver multivitamin, which primarily in soy foods, have weak estrogenic activity and are therefore
contained 26 nutrients at the US RDA level (recommended level from of interest in relation to hormone-​dependent cancers. Because isofla-
diet for most US adults), was associated with a significant, 8% lower vones were shown to increase estrogenic markers in MCF-​7 cells, an
incidence of all cancers combined. A  similar study in US women is ER+ breast cancer cell line, and to produce estrogenic effects in rodent
ongoing. The French SUVIMAX study of men and women (Hercberg reproductive tissues (Messina and Wood, 2008), there has been con-
et  al., 2004)  reported a statistically significant 31% lower risk of all cern among medical professionals and the general public that eating
cancers combined in men, but not women, who were randomized to soy food could fuel breast cancer growth. However, soy is metabolized
an antioxidant mix (120 mg of ascorbic acid, 30 mg of vitamin E, 6 differently in humans than it is in mice and rats, so findings in rodents
mg of β-​carotene, 100 μg of selenium, and 20 mg of zinc) versus pla- may not apply to people (Setchell et al., 2011). A meta-​analysis of soy
cebo. Women in that trial had higher baseline plasma antioxidant status. feeding studies in humans does not support effects of soy consumption
Based on the preponderance of data on RCTs of high-​dose, individual on circulating estrogen (Fritz et al., 2013).
supplements, several organizations recommend against taking antioxi- Another meta-​analysis of 14 prospective studies showed that in
dant or other supplements for cancer prevention (Kushi et  al., 2012; Asian women, those who ate the most (compared to the least) soy iso-
WCRF/​ AICR, 2007), or prevention of chronic diseases (Fortmann flavones had a 24% lower risk of incident breast cancer, while there
et al., 2013). However, a possible modest benefit of lower dose supple- was no association in Western countries such as the United States
ments in cancer prevention deserves further study, especially in groups (Dong and Qin, 2011). However, consumption levels in US women
with suboptimal diets. would typically correspond to levels in the reference group from stud-
ies in Asian countries. A systematic review concluded that soy intake
equivalent to 2–​3 servings daily (25–​50 mg/​d, similar to the traditional
Sodium Japanese diet) may reduce breast cancer risk (Fritz et al., 2013). It is
Foods preserved in salt or other sources of sodium are thought to con- also important to consider differences in lifelong soy exposure in Asian
tribute to a higher risk of gastric cancer in Asia and South America, and Western countries when comparing associations between soy and
where salting fish and/​or brining vegetables is common practice. High breast cancer across these populations. In Chinese women, consistently
concentrations of salt irritate the gastric epithelium, especially in the higher soy consumption during adolescence and adulthood was asso-
context of Helicobacter pylori (H. pylori) infection (see Chapter 31). ciated with 60% lower risk of pre-​menopausal breast cancer, but not
In the United States and other Western countries, dietary salt or with postmenopausal breast cancer (Lee SA et al., 2009). Isoflavone
sodium has not been associated with increased risk of stomach or other exposure in utero may likewise be important (Nagata, 2010). For
cancers. Because salt preservation is an issue in certain populations, prostate cancer, dietary intake and serum biomarkers of daidzein and
36

336 PART III:  The Causes of Cancer


genistein (major isoflavone types), but not total isoflavones, were asso- the human estrogen receptor, it is classified as an endocrine disruptor
ciated with a significantly lower risk of total prostate cancer in a recent (Geens et al., 2012). Although BPA is not thought to be a direct car-
meta-​analysis (He et al., 2015). Inter-​individual variability in the abil- cinogen, studies in rodents show that it sensitizes mammary tissue to
ity of gut microbiota to convert daidzein to equol may explain some of effects of chemical carcinogens (Rochester, 2013). BPA exposure may
the inconsistencies in the literature on soy and cancer (Lampe, 2009; have significant implications for human health and fertility, especially
Vastag, 2007). Of note, few studies have comprehensively evaluated exposure at developmental ages, and in sensitive populations. In 2012,
whether soy supplements or isolated soy proteins (found in energy the FDA banned its use in baby bottles and sippy cups (FDA, 2012a,
bars and other processed foods) influence cancer risk. 2012b).
Flavonoid classes other than isoflavones are more commonly con- Pesticides and genetic modification of crops to resist pests or
sumed in Western populations. Despite considerable interest in these improve crop yield are frequently the focus of public concern. Studies
other flavonoids with respect to human cancer, the evidence remains of the effects of herbicides and pesticides in humans are complicated,
limited, in part because flavonoid databases have only recently permit- however, because the amounts in foods are highly variable and not
ted comprehensive analyses of these compounds in relation to cancer known to the consumer. For many of these compounds, no practical
risk (Peterson et al., 2015). Also, flavonoid content varies by plant spe- biomarker of exposure has been identified. The IARC (Guyton et al.,
cies and can be affected by factors such as processing method. Food 2015) and the state of California (State of California, 2015) have con-
sources of these compounds include green tea (flavanols), black tea cluded that glyphosate (Roundup), an extensively used herbicide, is a
(theaflavins and thearubigins), vegetables (flavonols), berries (antho- class II carcinogen. This is troublesome because Roundup use is cur-
cyanidins), celery and garlic (flavones), citrus fruits (flavanones), and rently widespread for commercial and non-​commercial purposes.
cocoa (proanthocyanidins), among others. Emerging studies from Cadmium, used in nickel-​cadmium batteries and for stabilizing
European and US cohorts support a cancer protective role of flava- plastics, is a heavy metal that enters the food supply from water con-
nols in relation to hepatocellular cancer risk (Zamora-​Ros et al., 2013), taminated by landfills. It bioacumulates in the liver and kidney, and has
flavonols and urothelial cell carcinoma (Zamora-​Ros et  al., 2014), a half-​life of 7–​16 years (Kjellstrom and Nordberg, 1978). Since 1993,
anthocyanidins, flavonols, flavones, flavanols, and total flavonoids the IARC has classified cadmium as a Group 1 carcinogen, citing suf-
and gastric cancer (Zamora-​Ros et  al., 2012), and flavonols and fla- ficient evidence in humans for the carcinogenicity of cadmium and
vanones and ovarian cancer (Cassidy et  al., 2014). However, not all cadmium compounds (IARC, 2012a). Cadmium is thought to influ-
studies are consistent (Wang et al., 2009), and some even suggest harm ence cancer risk through increasing oxidative stress and reducing DNA
(Romagnolo and Selmin, 2012). damage repair. It is also considered an endocrine disrupter since it has
the potential to mimic estrogens and androgens in the body. Studies
generally find no association of dietary estimates of cadmium with
breast cancer risk (Wu et al., 2015), potentially because of difficulty
Food Contaminants estimating exposure. A  meta-​analysis of mostly case-​control studies
A comprehensive review of potentially carcinogenic food and bever- reported a significant increased risk of breast cancer with higher uri-
age contaminants is beyond the scope of this chapter. Herein we focus nary cadmium concentrations (Larsson et al., 2015).
on some key contaminants that are established or suspected cancer
risk factors.
Aflatoxins are mycotoxins produced primarily by the common fun- SPECIFIC FOODS AND BEVERAGES
gus Aspergillus flavus and the closely related species A. parasiticus,
and found primarily in corn, peanuts, cottonseed, and tree nuts, in Fruits and Vegetables
tropical or semitropical areas (IARC, 2012b). They are classified as
Group 1 carcinogens by the IARC (IARC, 2012b), and are an estab- Fruits and vegetables contain numerous constituents thought to influ-
lished cause of liver cancer. Presence of the hepatitis B virus sensitizes ence carcinogenesis, including vitamins, phytochemicals, and dietary
liver cells to the mutagenic effects of aflatoxin, and recent studies doc- fiber (IARC, 2003). Based largely on case-​control studies, the 1997
ument decreases in liver cancer incidence in high-​risk areas in China WCRF/​AICR report designated the evidence “convincing” that fruits
coinciding with lower aflatoxin biomarkers in urine and blood (Chen and vegetables lower the risk of several cancers (WCRF/​AICR, 1997).
et  al., 2013; Sun et  al., 2013). Reductions in aflatoxin exposure are Ten years later, the 2007 WCRF/​AICR report concluded that the evi-
currently the main reason for the decrease in liver cancer at older ages dence for fruits, vegetables, and cancer risk was “probable,” primar-
in these high-​risk areas. Neonatal vaccination against hepatitis B will ily for gastrointestinal cancers (WCRF/​AICR, 2007). This shift was
become more important as vaccinated cohorts age (Sun et al., 2013). chiefly due to weaker findings from prospective cohort studies, which
An industrial chemical, acrylamide, has been classified as “prob- are not subject to the recall and selection biases of case-​control studies.
ably carcinogenic” in humans (Group 2A) by IARC since 1994, based Nevertheless, fruits and vegetables may influence cancer risk in
primarily on genotoxicity experiments in animals (IARC, 1994). more nuanced ways. A  higher vegetable intake was associated with
Discovery in the first decade of the 2000s that acrylamide could be a significantly lower risk of ER–​breast cancer in two large prospec-
formed in (mostly) carbohydrate-​containing foods during high-​heat tive analyses (Emaus et al., 2016; Jung et al., 2013), including a pool-
cooking stimulated a surge of research into the association of esti- ing project of 20 cohort studies (Jung et al., 2013) (Figure 19–​2). In
mated acrylamide intake and cancer risk. Dietary acrylamide assess- another pooled analysis of 13 prospective cohort studies, intake of
ment using an FFQ was shown to correlate moderately (r  =  0.34) fruits and vegetables was inversely associated with risk of renal cell
with hemoglobin adducts of acrylamide and its metabolite, glycidam- cancer (Lee JE et al., 2009), although the WCRF CUP for kidney can-
ide (Wilson et  al., 2009). Two large meta-​analyses (Pelucchi et  al., cer considered the totality of the evidence to be limited (WCRF/​AICR,
2011; Virk-​Baker et al., 2014) found no association between dietary 2015a). In a meta-​analysis of published studies, intake of fruits and
acrylamide and risk of several cancers. Potential for confounding by vegetables was associated with a nonlinear lower risk of colorectal
tobacco (an important source of acrylamide) and difficulty in assess- cancer, with higher risk reported among those with the lowest intakes,
ing exposure in a rapidly changing marketplace remain challenges in and a flattened association at higher intake levels (Aune et al., 2011).
studying the health effects of this compound. Fruits and vegetables are consistently inversely associated with can-
Bisphenyl A  (BPA) is an industrial chemical used in materials cers of the esophagus and other aerodigestive tract sites, although
intended to come into contact with food (e.g., reusable plastic bot- this is based largely on case-​control studies (WCRF/​AICR, 2007). As
tles, feeding-​bottles, microwave ovenware, storage containers, etc.), described later in this chapter, overall diet patterns higher in fruits and
whereas the epoxy resins are used for internal coating of food and vegetables are associated with lower risk of certain cancers, cancer
beverage cans (European Food Safety Authority, 2006). First pro- mortality, cardiovascular diseases (CVD), and death from all causes.
duced in 1891, its estrogenic potential was hypothesized in the It is worth noting that fruits and vegetables are highly heteroge-
1930s (Dodds and Lawson, 1936). Because of its ability to activate neous in composition, and some types of fruits and vegetables may
 37

Diet and Nutrition 337


1.2
formation of nitrosamines in the gut is catalyzed by heme iron, result-
1 P-trend, 0.06
ing in oxidative DNA damage (Joosen et  al., 2009). Polycyclic aro-
matic hydrocarbons (PAHs) and heterocyclic aromatic amines (HCA
0.8 P-trend, < 0.001 or HAA) are carcinogens formed during high-​heat cooking of meat
Relative Risk

(Sinha et al., 1998a, 1998b). In a study from the NIH-​AARP Cohort


0.6 including 300,948 US men and women (Cross et al., 2010) in which
dietary intakes were linked with a meat carcinogen database, a posi-
ER-positive tive association was observed for heme iron, nitrate from processed
0.4
ER-negative meats, and heterocyclic amine intake, specifically 2-​amino-​3,8-​dime-
0.2 thylimidazo[4,5-​f]quinoxaline (MeIQx) and 2-​amino-​3,4,8-​trime-
thylimidazo[4,5-​f]quinoxaline (DiMeIQx). Not all studies of specific
meat carcinogens are consistent (Ollberding et  al., 2012). The 2015
0
Q1 Q2 Q3 Q4 Q5
US Dietary Guidelines Advisory Committee report recommended that
individuals reduce their intake of red and processed meat (regardless
Quintile of Vegetable Intake of fat content) because of its associations with cancer and other dis-
eases and the environmental impact on agriculture and greenhouse
Figure  19–​2. Association of vegetable intake and RR of breast cancer gasses (US Dietary Guidelines Advisory Committee, 2015). However,
by hormone receptor status. The study included 993,466 women from 20 this recommendation did not survive opposition from industry lobby-
prospective cohort studies, with 19,869 estrogen receptor positive (ER+) ists and Congress (US Department of Health and Human Services and
and 4821 ER− breast cancers diagnosed during 11–​20 years of follow-​up. US Department of Agriculture, 2015).
Source: Jung S, et al. (2013).

Grains
have potential deleterious effects. Potatoes and some fruit juices, for Whole-​grain foods, made from the entire grain seed, are much higher
example, have a high glycemic load and increase insulin secretion. In in fiber and many vitamins, minerals, and other phytochemicals than
the United States, potatoes and orange juice are the most commonly processed (refined) flour products. While research on whole grains
consumed vegetables and fruit, respectively, and broccoli and legumes in relation to cancer risk has generally been inconsistent, some stud-
are among the least frequently consumed vegetables (Produce for ies support an inverse association with gastrointestinal cancers. In
Better Health Foundation, 2015). Therefore, careful attention should the NIH-​AARP Diet and Health Study of 291,988 men and 197,623
be paid to the types of foods that contribute to risk estimates. women, whole-​grain foods were associated with a significant 20%
lower risk of colorectal cancer (Schatzkin et al., 2007), and a statisti-
cally borderline inverse association with rare small intestinal tumors
Red and Processed Meat (Schatzkin et al., 2008). The evidence for other cancers, including pan-
The IARC Monographs Programme recently reviewed the evidence on creatic (Chan et al., 2007) and breast (Egeberg et al., 2009), is incon-
red and processed meat in relation to cancer, and classified processed clusive. Whole-​grain foods are strongly protective against CVD (Cho
meat (e.g., hot dogs, bacon, sausage, deli meats, etc.) as a Group 1 car- et al., 2013) and diabetes (de Munter et al., 2007). As described later
cinogen, based on sufficient evidence in humans for colorectal cancer in this chapter, dietary patterns rich in whole grains, nuts, vegetables,
(Bouvard et  al., 2015). Consumption of unprocessed red meat (e.g., fruits, and low in red and processed meat are associated with lower risk
beef, pork, lamb) was classified as probably carcinogenic to humans of certain cancers.
(Group 2A), based on limited evidence in humans for colorectal can-
cer, and strong mechanistic evidence supporting a carcinogenic effect
(Bouvard et al., 2015). These conclusions are consistent with those of
Dairy Products
the WCRF/​AICR Continuous Update Report (WCRF/​AICR, 2011), In the United States, dairy products are the major source of dietary
which considered the association between red and processed meats calcium and vitamin D.  Consistent with the evidence for calcium
and colorectal cancer to be “convincing.” A meta-​analysis of prospec- and vitamin D, epidemiologic evidence supports an inverse associa-
tive cohort studies reported 17%–​18% higher risk of colorectal cancer tion between dairy products and colorectal cancer (Cho et al., 2004).
for each 100 g of red or 50 g of processed meat consumed per day A  recent meta-​ analysis further suggests that milk and total dairy
(Chan et al., 2011). The IARC noted that consumption of processed products (but not cheese) lower the risk of colon but not rectal cancer
meat is also positively associated with stomach cancer; consumption (Aune et al., 2012b).
of red meat is positively associated with pancreatic and prostate cancer High consumption of milk during childhood and adolescence is
(Bouvard et  al., 2015). Because the relationships for colorectal can- associated with increased height (Berkey et al., 2009), which in turn is
cer appear linear up to 140 g/​day (Chan et  al., 2011), the evidence associated with multiple forms of cancer (WCRF/​AICR, 2007). Milk
does not allow for a determination of a safe level of consumption. The consumption also increases blood levels of IGF-​1 (Cadogan et  al.,
American Cancer Society’s cancer prevention guidelines recommend 1997; Rich-​Edwards et  al., 2007), which have been associated with
reducing consumption in general, rather than indicating a specific higher risks of breast and prostate cancer (Key et al., 2010; Neuhouser
amount (Kushi et al., 2012). et al., 2013; Rowlands et al., 2009). Prospective studies of breast can-
The timing when meat is consumed may be important. For example, cer risk in relation to dairy intake have been inconsistent and mostly
a large pooled analysis of eight cohort studies observed no association null (Missmer et al., 2002), although some studies suggest that breast
between breast cancer and consumption of red or total meat in adult- cancer incidence may be lower with higher intakes of low-​fat dairy
hood (Missmer et al., 2002). However, the consumption of red meat products during mid-​and later life (Cui and Rohan, 2006; Shin et al.,
during high school was associated with higher risk of breast cancer in 2002). In contrast, high intake of dairy products has been associated
young women (Farvid et al., 2014a). It was considered plausible that with an increased risk of other hormonally related cancers, includ-
breast tissue would be most susceptible to carcinogens during adoles- ing prostate (Aune et al., 2015), ovary (Genkinger et al., 2006), and
cence. Furthermore, modeling substitution of red meat with poultry, endometrium, especially among women not using hormone therapy
fish, or legumes and nuts was associated with 14%–​24% lower risks of (Ganmaa et al., 2012). Evidence on milk and dairy consumption dur-
overall or postmenopausal breast cancer. ing childhood and cancer risk in adulthood remains limited; in one
There are several mechanisms by which the consumption of red report, adolescent milk consumption was not significantly associated
and processed meats could influence colorectal carcinogenesis. The with future risk of breast cancer (Linos et al., 2010).
38

338 PART III:  The Causes of Cancer

Soy relative risk for drinking 4 or more cups per day compared with less
than 2 cups per day was 0.25 (95% CI: 0.11, 0.62); this was weakened
Soy foods (such as tofu, tempeh, edamame, miso, many veggie after control for biomarkers of hepatocellular injury and inflamma-
burgers, and other products made with soy flour) contain isoflavones tion, suggesting that these mechanisms may underlie the association
which, as described earlier, have weak estrogenic activity and can act (Aleksandrova et al., 2015). Whether coffee is related to a lower risk
as anti-​estrogens by competitive inhibition for the estrogen receptor of cancers of the kidney (Lee et al., 2007), breast (Bhoo-​Pathy et al.,
(Nagata, 2010). Because of this, soy foods have been hypothesized 2015), colon and rectum (Li et  al., 2013), or prostate (particularly
to reduce the risks of breast and other hormonally related cancers. lethal or advanced forms) (Wilson et al., 2011) is the subject of con-
In a recent meta-​analysis including 14 epidemiologic studies, Asian tinued research.
women who ate the most (compared to the least) soy isoflavones The 2016 IARC Expert Review Committee also reviewed the evi-
had a 24% lower risk of developing breast cancer, while there dence for maté (a traditional South American caffeine-​rich infused
was no association in Western countries such as the United States drink), and beverage temperature. The IARC concluded that drink-
(Dong and Qin, 2011). The benefit of soy has been observed primar- ing very hot beverages at above 65 degrees Centigrade (149 degrees
ily with consumption during childhood or early adult life (Lee SA Fahrenheit), including maté, was “probably carcinogenic to humans”
et al., 2009). (Group 2A) (Loomis et al., 2016). As most of the evidence in humans
is from case-​control studies, it will be important to confirm these find-
ings in additional prospective studies.
Coffee
In addition to caffeine, coffee contains multiple biologically active com- Tea
pounds such as chlorogenic acid, kahweol, and N-​methylpyridinium,
which in animals and in vitro induce apoptosis, inhibit inflammation, Tea is rich in polyphenols and other compounds with antioxidant and
angiogenesis, and metastasis, and regulate genes involved in DNA anti-​cancer properties. Green tea, more commonly consumed in Asian
repair and detoxification processes (Bohn et al., 2014). countries, is particularly rich in catechins, a type of flavonoid. In con-
In 1991, an expert Working Group organized by the International trast, black tea, more commonly consumed in Western countries, is
Agency for Research on Cancer (IARC) Monographs Program clas- higher in theaflavins and thearubigens (Yuan et al., 2011). Case-​control
sified coffee as a 2B carcinogen, concluding that “[c]‌offee is possibly studies of tea consumption and cancer risk generally find stronger
carcinogenic to the human urinary bladder” (IARC Working Group on inverse associations than prospective studies. A  meta-​analysis of 57
the Evaluation of Carcinogenic Risks to Humans, 1991); however, a prospective studies (including 20 from the US and 22 from Asia) that
2016 review by IARC concluded that coffee drinking was considered included over 8 million individuals and almost 50,000 incident cancer
unclassifiable as to its carcinogenicity to humans (Group 3) (Loomis cases found tea consumption to be inversely associated with oral can-
et al., 2016). Most of the studies considered in the 1991 IARC review cer (RR 0.72; 95% CI: 0.54, 0.95, high vs. low tea consumption), but
were case-​control studies, which are susceptible to recall and selec- with none of the other cancers examined (gastric, colon, rectum, lung,
tion biases. A 2012 meta-​analysis of coffee consumption and bladder liver, pancreas, breast, prostate, ovarian, bladder, or glioma). Thus, the
cancer risk continued to show positive associations for case-​control evidence to date suggests that the relationship between tea and can-
studies, but estimates from five prospective cohort studies were null cer is likely to be minimal, but additional large studies of oral cancer
(Zhou et al., 2012). Further, coffee consumption is generally associ- would be valuable to confirm this finding.
ated with smoking, which is strongly related to bladder cancer and
difficult to control for.
The potential for residual confounding due to smoking is a concern Alcohol
for the relationship of coffee consumption with other cancers as well. As described in Chapter  12, alcohol is considered a Group  1 car-
In the AARP cohort, there was a two-​fold increased risk of mortality cinogen by the IARC (IARC, 2010), and an established cause of
due to cancer in age-​adjusted models for both men and women; after cancers of the upper gastrointestinal tract, liver, colorectum, and
controlling for smoking and other risk factors, risk estimates were breast (female). In contrast, there is evidence for lack of carcino-
substantially attenuated and were no longer statistically significant in genicity for kidney cancer and non-​Hodgkin lymphoma. For pan-
any category of consumption, although the trend remained statistically creatic cancer, risk begins to increase at greater than 3 servings/​day
significant in men only (Freedman et  al., 2012). Significant interac- (Gapstur et al., 2011), whereas the increase for breast cancer begins
tions by smoking status were observed (p < 0.01), with weak inverse at as few as 3 servings/​week (Chen et al., 2011; Smith-​Warner et al.,
trends in never smokers and either a positive or no trend in current and 1998). On the other hand, alcohol is associated with lower CVD
former smokers. risk and mortality (Thun et  al., 1997), and is an integral part of
The WCRF/​ AICR CUP considers the protective association certain healthful dietary patterns, such as the Mediterranean diet
between coffee consumption and endometrial cancer to be “probable” (Trichopoulou et  al., 2003). Dietary recommendations for cancer
(WCRF/​AICR, 2013) although a recent meta-​analysis concluded that prevention advise limiting alcohol consumption among those who
there is no association (Yang TO et al., 2015). Of 12 cancers examined drink to no more than one drink/​day for women and two drinks/​day
in relation to coffee consumption in the Prostate, Lung, Colorectal, and for men (WCRF/​AICR, 2007).
Ovarian (PLCO) screening cohort, a statistically significant inverse
association was observed only for endometrial cancer; the association
was non-​statistically significantly inverse when limiting the analysis to DIETARY PATTERNS
never smokers (Hashibe et al., 2015).
Despite methodologic limitations, coffee consumption has been Characterizing “diet” as an overall diet pattern provides a more com-
strongly and consistently related to lower risk of hepatocellular cancer plete picture of dietary intake than studies of single foods or nutri-
(HCC) (Bravi et al., 2013), and the 2014 WCRF/​AICR Liver Cancer ents. Dietary patterns also potentially reflect additive and synergistic
CUP considers it “probable” that coffee consumption protects against effects of the components of diet on disease risk. This approach may
liver cancer (WCRF/​AICR, 2015b). In a recent pooling project of nine be particularly useful in the study of cancer etiology, where associa-
US cohorts including over 1 million persons, higher coffee consump- tions with certain dietary risk factors may be small and difficult to
tion was associated with lower risk of HCC (relative risk for > 3 cups/​ detect in isolation.
day vs. non-​drinker = 0.73; 95% CI: 0.53, 0.99, P, trend cups/​day = The two most common approaches to characterize dietary patterns
< 0.0001) (Petrick et al., 2015). The relative risk was stronger for caf- in observational studies involve either statistically driven empirical
feinated coffee than for decaffeinated coffee, although the confidence methods, such as factor analysis, or creation of a priori dietary indexes
interval for the latter was wide. In the European EPIC cohort, the or scores (Hu, 2002), as described later in this chapter.
 39

Diet and Nutrition 339


Vegetarians Hypertension (DASH) diet, the Healthy Eating Index (reflecting US
Dietary Guidance), and an Alternate Healthy Eating Index (reflecting
Levels of vegetarianism range from complete exclusion of all animal the Harvard Healthy Eating Plate & Food Guide Pyramid) were each
products (including fish and dairy; “vegan”) to lacto-​ovo vegetarian associated with an approximate 20% statistically significant reduction
(includes eggs and dairy), to pescovegetarian (includes fish). In the in cancer mortality, comparing the highest to lowest quintile (Liese
Adventist Health Study II, compared to non-​ vegetarians, vegetar- et al., 2015). These diet scores share several commonalities, including
ians of any type—​defined according to protein sources reported on a being rich in a variety of plant foods and generally higher in healthy
FFQ—​had a statistically significant 28% lower risk of colorectal can- protein sources, and lower in sugar and saturated and trans fat, but vary
cer (Orlich et al., 2015). Likewise, in a recent British study (Key et al., in the specific fatty acids, protein sources, and alcohol recommenda-
2014), vegans, vegetarians, and pescovegetarians had a significantly tions. Of individual cancers, diet scores have tended to be most con-
lower risk of several cancers, compared to meat eaters. sistently inversely associated with gastrointestinal cancers. Despite the
many advantages of studying diet patterns, such exposures are more
complex to interpret, as they may mask subcomponents that are mini-
Mediterranean Diet mally or maximally predictive of disease outcome.
Probably the most studied dietary pattern has been the Mediterranean
diet, characterized by high consumption of olive oil (or unsaturated
fats in some scoring systems), fruits, vegetables, nuts, legumes, and Cancer Prevention Guidelines
unprocessed cereals, low consumption of meat and meat products, low Dietary guidelines focused on cancer prevention emphasize not only
consumption of dairy (with the exception of long preservable cheeses), healthy diet, but also behaviors that affect body weight, physical
and moderate intake of alcohol, usually wine consumed with meals activity, and alcohol consumption (Kushi et al., 2012; WCRF/​AICR,
(Trichopoulou et  al., 2014). A  meta-​analysis of 11 cohort studies 2007). These recommendations encourage diets that are mostly plant-​
found that higher (vs. lower) scores reflecting a Mediterranean-​like based, high in non-​starchy vegetables, whole fruit, and whole (vs.
diet were associated with a lower risk of all cancers combined (relative refined) grains, and low in red and processed meat. Scores reflecting
risk = 0.87; 95% CI: 0.81, 0.93), and specific cancer sites (colorectum, the diet, physical activity, body weight, and alcohol recommendations
breast, prostate, stomach, liver, pancreas, head and neck, and respira- of cancer prevention guidelines have been developed and examined
tory cancers) (Schwingshackl and Hoffmann, 2015). This conclusion in relation to cancer incidence (Kabat et al., 2015; Romaguera et al.,
was further supported by a recent analysis of three cohort studies in 2012; Thomson et al., 2014a) and mortality (McCullough et al., 2011;
which adherence to a Mediterranean diet, calculated using identical Vergnaud et  al., 2013). Lifelong non-​ smokers whose dietary pat-
scoring methods, was associated with lower all-​cancer mortality (Liese terns, weight, and physical activity more closely correspond with the
et  al., 2015). The Spanish PREDIMED randomized trial random- American Cancer Society (ACS) guidelines have a 24%–​30% lower
ized participants to three diet pattern interventions: a Mediterranean death rate from cancer, and a 42% lower all-​cause mortality rate,
diet (via instruction) with supplementary nuts, a Mediterranean diet compared to those whose patterns are least consistent (McCullough
with supplementary extra virgin olive oil, or a control (low-​fat) diet et al., 2011). Another large study of the AARP cohort reported that,
(Estruch et al., 2013). Both Mediterranean diet groups had unequivo- in analyses controlling for smoking, the incidence rates for 14 out of
cal health benefits for CVD; although based on a small number of 25 cancer sites were significantly lower in study participants whose
cases, the group randomized to extra virgin olive oil had a substantially behaviors were consistent with the ACS guidelines than in those who
lower risk of breast cancer compared to the control group (RR = 0.32; were not (Kabat et al., 2015). In the Women’s Health Initiative cohort,
95% CI: 0.13, 0.79) (Toledo et al., 2015). significant inverse associations with cancer incidence and death rates
were reported among white, black, and Hispanic postmenopausal
women, with stronger associations appearing in the last two racial/​eth-
Other Diet Patterns nic groups (Thomson et al., 2014a). In models with mutual adjustment,
the individual score components, including diet, generally remained
Empirically derived patterns, which use statistical approaches to
independently associated with lower cancer risk. Results using the
extract unique correlated eating behaviors within a population, typi-
global WCRF/​AICR score, with different weighting for lifestyle fac-
cally identify two or three such patterns, each explaining up to 5%–​
tors, found qualitatively similar inverse associations with cancer risk
10% of the variation in diet. As expected, patterns vary across study
and mortality (Romaguera et al., 2012; Vergnaud et al., 2013). Overall,
populations, but two have been commonly identified across interna-
these studies underscore the significant potential for cancer prevention
tional borders:  the first, a so-​called “Western” pattern, is higher in
through combined health behaviors including diet, physical activity,
red and processed meat, potatoes, sugar, and lower in vegetables and
and healthy body weight.
whole fruit; the second, a “Prudent” pattern, is higher in fruits, veg-
etables, poultry, fish, and nuts, and low in high-​fat dairy, meat, and
sugar. The “Western” pattern is positively associated with colon can-
cer incidence (Magalhaes et al., 2011) and with cancer recurrence and Effects of Diet on Cancer Survival
mortality among colorectal cancer patients (Meyerhardt et al., 2007). Approximately 14.5 million children and adult cancer survivors were
Although the Prudent pattern is associated with lower colorectal can- alive on January 1, 2014 (American Cancer Society, 2014). Compared
cer risk, it appears that this is not as strong as avoiding a Western-​type to the evidence base on diet and cancer prevention, the study of diet
diet (Fung et al., 2003). in relation to cancer recurrence, cancer-​specific survival, and overall
A priori indices, such as those used to study the Mediterranean diet, survival is in its infancy. Therefore guidelines on nutrition and physi-
quantify the degree of dietary concordance with hypothesized healthy cal activity for cancer survivors also recommend following cancer
diet patterns using a set of criteria, with low scores generally reflect- prevention guidelines (Rock et al., 2012). Timing is a central issue in
ing poor concordance, and high scores reflecting high concordance to studies of diet and cancer survival. For a cancer patient, the relevant
multiple dietary components. Healthy diet scores have been based on time is after the diagnosis, and many of the existing studies assessed
dietary guidelines, hypothesized general or disease-​specific diets, cul- diet before diagnosis. Optimally an epidemiologic study should have
turally defined healthful models of eating, and specific mechanisms dietary assessments both before and after diagnosis because these tend
mediated by diet, such as its antioxidant or anti-​inflammatory poten- to be correlated, and we would want to know their independent con-
tial. Recently, associations of four hypothesized healthful diet patterns tribution to survival. Only a few randomized trials of dietary inter-
were examined in relation to cause-​specific mortality using a uniform ventions and cancer survival have been conducted, as described in the
approach to diet score calculations and analysis across three prospec- following paragraph.
tive cohort studies (Liese et al., 2015). Scores reflecting dietary con- Breast cancer survivors comprise over 3.1 million survivors in the
cordance with the Mediterranean diet, the Dietary Approaches to Stop United States, larger than any other group (American Cancer Society,
340

340 PART III:  The Causes of Cancer


2014). A report based on a systematic and quantitative literature review who might benefit most from increased or decreased exposure to
found no convincing or probable dietary factors associated with recur- specific dietary factors. The same concept underlies “personalized
rence, cause-​specific, or total mortality for breast cancer survivors medicine” or “personalized prevention.” The potential for practical
(WCRF/​AICR, 2014a), although limited support exists for soy foods, applications of this concept has weakened, especially for prevention,
high dietary fiber, and low saturated fat. A limitation is that many stud- with the realization that the effects of common genetic variants on
ies contributing to this report only examined diet prior to diagnosis. In disease risk are considerably smaller than anticipated 15  years ago.
Western diets where soy consumption is less common, lignans are the Apart from this application, the identification of gene–​environment
predominant phytoestrogen. A  recent population-​based case-​control interactions can be of value in etiologic studies if Mendelian random-
study showed that pre-​diagnostic dietary lignan intake was associ- ization supports a particular mechanism for a diet and disease relation-
ated with statistically significantly lower risk of breast cancer mortal- ship that cannot be tested in a randomized clinical trial (Willett W,
ity (McCann et al., 2010). The Women’s Intervention Nutrition Study 2013). Furthermore, the identification of a subset of individuals who
(WINS) trial, a low-​fat dietary intervention trial, found a 24% mar- are uniquely responsive to the effects of a particular dietary factor can
ginally significant lower risk of recurrence (Chlebowski et al., 2006); increase the statistical power of observational studies of this relation-
however, because women on the low-​fat arm also lost weight, it is ship. The study of gene–​diet interactions in relation to cancer risk has
possible that the effect was due to weight loss and not because of a evolved from studies of single nucleotide polymorphisms (SNPs) in
reduction in dietary fat per se (Gapstur and Khan, 2007). The Women’s candidate genes to genome-​wide association studies (GWAS) using
Healthy Eating and Living (WHEL) study found no difference in a more agnostic approach. A  recent systematic literature review of
breast cancer outcomes between women counseled on a high fruit, gene–​diet interactions and colorectal cancer risk in candidate gene
vegetable, fiber, low-​fat diet versus those counseled on eating “five studies found suggestive evidence for interactions between meat, cru-
a day” (fruits and vegetables) (Pierce et al., 2007). As breast cancer ciferous vegetables, dietary fiber, calcium, vitamins, and alcohol and
patients typically have a relatively good prognosis, they are subject to SNPs in the ABCB1, NFKB1, GSTM1, GSTT1, CCND1, VDR, MGTM,
the same diseases as the general population and actually tend to be at IL10, and PPARG genes (Andersen et  al., 2013). More recently, a
higher risk of other chronic diseases, including CVD, relative to non-​ comprehensive GWAS approach examined diet–​ gene interactions
cancer survivors (Rock et al., 2012). Thus, understanding the potential with colorectal cancer risk in over 9000 cases and 9000 controls from
role of diet on both cancer and non-​cancer outcomes in this patient 10 studies (Figueiredo et al., 2014). The study uncovered strong evi-
group has strong clinical and population health relevance. As would dence for a gene–​diet interaction and colorectal cancer risk between
be expected, a growing literature in breast cancer survivors from pro- a genetic variant (rs4143094) on chromosome 10p14 near the gene
spective cohort studies suggests that women who consume healthy diet GATA3 and processed meat consumption (p = 8.7E-​09), although the
patterns (assessed via diet scores) rich in a variety of plant foods and functional significance of this interaction is still unknown (Figueiredo
whole grains and low in red and processed meat after diagnosis have et al., 2014).
lower risk of non-​breast-​cancer causes of death (George et al., 2014; The impact of epigenetic modifications can vary from full gene
Inoue-​Choi et al., 2013; Izano et al., 2013; Kim et al., 2011); however, silencing to complete expression, and the effect may be permanent
none of the diet scores examined was associated with breast cancer–​ or transient (Teegarden et al., 2012). Multiple bioactive food compo-
specific mortality. nents, including folate, selenium, polyphenols, isothiocyanates, and
For colorectal cancer survivors, post-​diagnostic dietary intake from others, have been shown to affect epigenetic processes in vitro; how-
two cohorts suggests a deleterious effect of a Western dietary pat- ever, more evidence is needed from animal and human studies (Ong
tern, high red and processed meat intake, and a high glycemic load et al., 2011). At present, no gene–​environment interactions involving
diet on colorectal cancer recurrence or survival (Van Blarigan and nutritional factors and cancer have been identified with sufficient cer-
Meyerhardt, 2015). In another type of analysis, post-​diagnostic data tainty and impact to have clinical application.
were used to predict plasma vitamin D status. In turn, higher predicted
vitamin D was associated with lower colorectal cancer-specific and
overall mortality (Ng et al., 2009). SUMMARY
Like breast cancer, clinical recurrence and death often occur many
years after a prostate cancer diagnosis, thus providing a wide time Methodologic Considerations
window for potential dietary and lifestyle influences on the outcome
(Chan et  al., 2014). Although the data on post-​diagnostic diet and Existing methodologic approaches have identified important rela-
survival are limited, a higher prostate-​specific mortality with higher tionships between nutrition and cancer, especially regarding dietary
intake of saturated fat and a lower total mortality with higher intake patterns, anthropometric measures, and alcohol consumption.
of vegetable fats were observed in a recent report from the Physician’s Diet composition can be reasonably well characterized by food
Health Study (Van Blarigan et  al., 2015). Similar findings were frequency questionnaires, repeated measures of short-​term intake
observed in another population of men with prostate cancer (Richman (e.g., multiple food records and recalls), and biological markers
et al., 2013). Adherence to a Western dietary pattern has been associ- of dietary intake. The methods of assessing diet composition con-
ated with higher prostate-​specific and total mortality, and adherence tinue to improve as nutrient databases are expanded, technologies
to a Prudent dietary pattern was associated with lower total mortality are adapted and/​or developed to measure dietary intake more pre-
(Yang M et al., 2015). Numerous short-​term interventions have been cisely, new biomarkers are discovered, and metabolites that medi-
conducted with changes in PSA or other surrogate variables as out- ate between food intake and biological effects on cancer risk are
comes; possible benefits have been seen with several combinations of identified. Further improvements in these methodologies will likely
nutritional supplements, but clinical outcomes are lacking (Hackshaw-​ increase the precision of estimates of absolute or relative risk. New
McGeagh et al., 2015). biomarkers are needed to assess aspects of diet that cannot be evalu-
The available evidence supports the same recommendations for can- ated from self-​report; markers of food processing and contaminants
cer survival as for cancer prevention (Rock et al., 2012). Fortunately, would be particularly useful. It is inherently difficult to separate
these recommendations also apply to the prevention of CVD, and thus the effects of highly correlated nutrients and foods in observational
provide broad benefits for the prevention of chronic disease. studies; distinguishing among them may be facilitated by Mendelian
randomization studies and short-​ term randomized studies with
intermediate endpoints. These issues become unimportant in studies
GENE–​DIET INTERACTIONS of overall dietary patterns.
No single type of study is likely to resolve most hypothesized diet
Nutrigenomics seeks to identify genetic polymorphisms and epigen- and cancer relationships. Large randomized trials of diet and cancer
etic alterations that modify individual response to nutrients (Teegarden incidence may be definitive if “positive,” but are not a gold standard, as
et al., 2012; Zeisel, 2007). The underlying goal is to identify individuals they may be unable to detect important effects due to poor adherence
 341
Table 19–1.  Relationship of Dietary Factors with Risk of Selected Individual Cancer Sitesa

Dietary Factor Colorectum Breast Prostate Lung Stomach Esophagus Pancreas Liver Ovary Endometrium All Cancers

Macronutrients/​energy balance
Obesity ↑↑ ↑↑˅ ↑˅ ↑↑˅ ↑↑˅ ↑↑ ↑↑ ↑ ↑↑ ↑↑

Abdominal fatness ↑↑ ↑
Carbohydrates/​sugars ↑§

Glycemic Load ↑

Height ↑↑ ↑↑ ↑ ↑ ↑↑ ↑ ↑

Nutrients (from food unless


otherwise specified)
Vitamin D [25(OH)D] ↓ φ Non-​linear Conflicting

Calcium ↓ ↑ §

Folate ↓§

Fiber ↓↓

Carotenoids ↓˅ Lycopene↓§ ↓

ß-​carotenesupplements φ ↑↑˅

Vitamin E supplements φ §

Selenium supplements φ

Antioxidant (combin​ation) ϕ φ
supplements
Salt preservation ↑↑

Contaminants ↑↑arsenic in ↑↑Aflatoxin


drinking water
Foods

Fruits ↓** ↓ ↓^

Vegetables ↓˅ ↓**
↓^

Red meat ↑↑ ↑§
↑† ↑†

Processed meat ↑↑ ↑↑ †
↑^

Other protein sources, fish, ↓§



poultry, nuts
Whole grains ↓

Dairy or milk ↓ ↑˅§

Soy ↓˅

Coffee φ ↓↓§ ↓

(continued)
342
Table 19–1. Continued

Dietary Factor Colorectum Breast Prostate Lung Stomach Esophagus Pancreas Liver Ovary Endometrium All Cancers

Tea φ

Alcohol ↑↑˅ ↑↑ ↑↑ ↑↑ ↑§ ↑↑ ↑↑

Diet patterns*
Empirical diet patterns

“Western” diet ↑↑

“Prudent” diet ↓ ↓˅

Dietary indices

Mediterranean diet ↓ ↓§ ↓˅ ↓ ↓

“DASH” diet ↓ ↓

USDA Healthy Eating Index ↓ ↓ ↓ ↓ ↓ ↓

Harvard “Alternate” Healthy ↓ ↓ ↓


Eating Index
Dietary Inflammatory Index ↑

Cancer lifestyle guidelines‡

ACS guidelines ↓ ↓ ↓˅ ↓ ↓ ↓˅ ↓˅ ↓ ↓↓
WCRF/AICR Guidelines ↓ ↓ ↓ ↓ ↓ ↓ ↓ ↓↓

a
Two arrows denotes convincing or consistent evidence; One arrow denotes probable association; Grey arrow denotes only one or limited studies; ϕ mark=unlikely to be an association; ˅ denotes lower risk in subgroups, e.g. obesity and
postmenopausal (not premenopausal) breast cancer, aggressive prostate, gastric cardia and esophageal adenocarcinoma; vegetables, carotenoids and ER-​breast cancer; beta-​carotene supplements and lung cancer in smokers; processed meat and
gastric cardia cancer; fruit, vegetables, processed meat and squamous cell esophageal cancer; dairy and aggressive prostate cancer; soy and breast cancer primarily in Asian countries; alcohol and colorectal cancer risk convincing in men, probable in
women. For diet patterns, prudent diet related to lower risk of ER-​breast tumors, Mediterranean diet associated with esophageal squamous cell cancer; ACS guidelines related to lower liver, lung and pancreatic cancer in men only. § denotes author
conclusions (note: breast cancer and red meat includes early life exposure); † also noted in IARC 2015 conclusions *Evidence on diet patterns is based on a limited number of studies. ‡ In addition to food-based recommendations, the ACS and
WCRF/​AICR scores also include body mass index, physical activity and alcohol consumption. Sources: WCRF/​AICR 2007 report or CUP reports if available; **This association may be confounded by smoking.
 34

Diet and Nutrition 343


(especially with complex dietary patterns), limited duration, and diffi- responsive to the dietary factor being investigated could increase our
culty blinding the intervention. For most hypotheses, the best evidence ability to detect diet and cancer relationships and to provide targeted
will probably come from replicated cohort studies in combination with interventions. Cohort studies with prospectively collected archived
short-​term randomized trials with intermediate biomarkers. stool, blood, and urine samples will be positioned to evaluate proposed
biomarkers from “omics” studies in relation to cancer risk.
Molecular characterization is increasingly used in oncology and
Estimates of Attributable Fraction cancer epidemiology to identify tumor subtypes that differ in their
The population attributable fraction for diet and all cancers, based clinical characteristics and etiology. As with the identification of gene–​
on more precise estimates for relationships that are established with environment interactions, identifying biologically relevant exposures
reasonable certainty, is about 20% overall, which is weaker than and pathways related to specific cancer subtypes can increase the abil-
previously estimated, and much of this is related to overweight and ity to detect relationships and to develop support for causality. This
inactivity. This also does not include the contribution of dietary com- work will benefit from collaborative efforts to provide the large num-
position to adiposity, which has become better established in recent bers of study participants and cases required.
years. The proportion contributed by dietary composition independent
of adiposity is unclear because some dietary factors have yet to be References
identified or established with sufficient certainty, and associations are Adolphe JL, Whiting SJ, Juurlink BH, Thorpe LU, and Alcorn J. 2010. Health
likely underestimated because of measurement error or misspecifica- effects with consumption of the flax lignan secoisolariciresinol digluco-
tion of temporal relationships. Estimates based on reasonably docu- side. Br J Nutr, 103(7), 929–​938. PMID: 20003621.
mented relationships suggest an etiologic contribution of 5%–​12%, Ahn J, Peters U, Albanes D, et al. 2008. Serum vitamin D concentration and
but this could be appreciably higher. prostate cancer risk:  a nested case-​control study. J Natl Cancer Inst,
100(11), 796–​804.
Alberts DS, Martinez ME, Roe DJ, et al. 2000. Lack of effect of a high-​fiber
Specific Diet and Cancer Relationships cereal supplement on the recurrence of colorectal adenomas. N Engl J
Med, 342, 1156–​1162.
Progress in diet and cancer prevention over the past 30+ years has Aleksandrova K, Bamia C, Drogan D, et  al. 2015. The association of cof-
been slow compared to accrual of knowledge relating diet to CVD and fee intake with liver cancer risk is mediated by biomarkers of inflam-
diabetes, but several findings come into focus: mation and hepatocellular injury:  data from the European Prospective
Investigation into Cancer and Nutrition. Am J Clin Nutr, 102(6), 1498–​
1. Individual nutritional supplements in high doses are unlikely to 1508. PMCID: PMC4658462.
prevent cancer and may even be harmful, but evidence suggests a American Cancer Society. 2013. Breast cancer facts & fi­ gures  2013–​2014.
modest benefit of RDA-​level multi-​nutrient supplements that may Atlanta: American Cancer Society.
American Cancer Society. 2014. Cancer treatment and survivorship facts &
be most important in subsets with suboptimal diets.
­figures 2014–​2015. Atlanta: American Cancer Society.
2. Certain constituents (nutrients, phytochemicals) may significantly Ames BN, Gold LS, and Willett WC. 1995. The causes and prevention of can-
reduce the risk of various cancers. Low folate intake likely contrib- cer. Proc Natl Acad Sci USA, 92, 5258–​5265.
utes to colon and possibly other cancers, and considerable evidence Andersen V, Holst R, and Vogel U. 2013. Systematic review: diet-​gene interac-
supports a role of plant foods, lycopene, vitamin D, and constitu- tions and the risk of colorectal cancer. Aliment Pharmacol Ther, 37(4),
ents of coffee in human cancer. 383–​391. PMCID: PMC3565452.
3. Processed and red meat consumption is likely to increase risk of Apperly. 1941. The relation of solar radiation to cancer mortality in North
colorectal and possibly other cancers, and dairy foods are likely America. Cancer Res, 1, 191–​195.
to influence risk of colorectal and prostate cancers in opposite Arab L, Steck-​Scott S, and Bowen P. 2001. Participation of lycopene and beta-​
carotene in carcinogenesis: defenders, aggressors, or passive bystanders?
directions.
Epidemiologic Rev, 23(2), 211–​230.
4. Alcohol consumption increases the risk of breast, colorectal, esoph- Armstrong B, and Doll R. 1975. Environmental factors and cancer incidence
ageal, and other cancers but is unlikely to increase risk of kidney and mortality in different countries, with special reference to dietary prac-
cancer or non-​Hodgkin lymphoma. tices. Int J Cancer, 15, 617–​631.
5. Several culturally or a priori defined diet patterns, including the Aune D, Chan DS, Greenwood DC, et al. 2012a. Dietary fiber and breast cancer
Mediterranean diet, are associated with lower cancer risk and mor- risk: a systematic review and meta-​analysis of prospective studies. Ann
tality in prospective epidemiological studies. These diet patterns Oncol, 23(6), 1394–​1402. PMID: 22234738.
are higher in plant foods (vegetables, fruits, cereals, vegetable pro- Aune D, Lau R, Chan DS et al. 2011. Nonlinear reduction in risk for colorectal
tein) and fish, and lower in processed meat and red meat and gener- cancer by fruit and vegetable intake based on meta-​analysis of prospec-
tive studies. Gastroenterology, 141(1), 106–​118. PMID: 21600207.
ally moderate in dairy and alcohol (Table 19–​1).
Aune D, Lau R, Chan DS, et al. 2012b. Dairy products and colorectal cancer
risk: a systematic review and meta-​analysis of cohort studies. Ann Oncol,
FUTURE DIRECTIONS 23(1), 37–​45. PMID: 21617020.
Aune D, Navarro Rosenblatt DA, Chan DS, et al. 2015. Dairy products, cal-
The precision of dietary estimates can be improved by combining sev- cium, and prostate cancer risk: a systematic review and meta-​analysis of
eral different approaches to exposure measurement, including the use cohort studies. Am J Clin Nutr, 101(1), 87–​117. PMID: 25527754.
of biomarkers, food frequency questionnaires, and multiple 24-​hour Avenell A, MacLennan GS, Jenkinson DJ, et al. 2012. Long-​term follow-​up for
mortality and cancer in a randomized placebo-​controlled trial of vitamin
recalls or food records. Pooled analyses of dietary intakes or biomark-
D(3) and/​or calcium (RECORD trial). J Clin Endocrinol Metab, 97(2),
ers in relation to cancer outcomes across multiple prospective stud- 614–​622. PMID: 22112804.
ies can minimize variation due to chance and biases from selective Bakker MF, Peeters PH, Klaasen VM, et al. 2016. Plasma carotenoids, vitamin
recall and publication. This is optimal if measurements of absolute C, tocopherols, and retinol and the risk of breast cancer in the European
intake and biomarker concentration are standardized and/​or calibrated Prospective Investigation into Cancer and Nutrition cohort. Am J Clin
across studies. Probably the greatest limitation of currently available Nutr, 103(2), 454–​464. PMID: 26791185.
evidence is that it derives mainly from diets assessed in mid-​or later Baron JA, Beach M, Mandel JS, et al. Calcium supplements for the prevention
life with follow-​up of less than 15 years. Dietary assessments earlier in of colorectal adenomas. N Engl J Med 1999;340:101–​7.
life, including in utero and during puberty, combined with long follow-​ Baron JA, Barry EL, Mott LA et al. 2015. A trial of calcium and vitamin D for
the prevention of colorectal adenomas. N Engl J Med, 373(16), 1519–​
up using repeated dietary assessments, may uncover exposures that
1530. PMCID: PMC4643064.
are relevant. Berkey CS, Colditz GA, Rockett HR, Frazier AL, and Willett WC. 2009. Dairy
Technological advancements in biomarker analysis permit com- consumption and female height growth: prospective cohort study. Cancer
prehensive examination of an individual’s metabolome, microbiome, Epidemiol Biomarkers Prev, 18(6), 1881–​1887. PMCID: PMC3740511.
genome, and epigenome. Use of these methods to assess internal Bhoo-​Pathy N, Peeters PH, Uiterwaal CS, et al. 2015. Coffee and tea consump-
exposures and identify subsets of individuals who are particularly tion and risk of pre-​and postmenopausal breast cancer in the European
34

344 PART III:  The Causes of Cancer


Prospective Investigation into Cancer and Nutrition (EPIC) cohort study. Clark LC, Combs GF Jr, Turnbull BW, et al. 1996. Effects of selenium supple-
Breast Cancer Res, 17, 15. PMCID: PMC4349221. mentation for cancer prevention in patients with carcinoma of the skin: a
Binkley N and Sempos CT for the Vitamin D Standardization Program. 2014. randomized controlled trial. Nutritional Prevention of Cancer Study
Standardizing vitamin D assays:  the way forward. J Bone Miner Res, Group. JAMA, 276(24), 1957–​1963.
29(8), 1709–​1714. PMID: 24737265. Colditz GA, and Wei EK. 2012. Preventability of cancer:  the relative con-
Birt DF, Kris ES, Choe M, and Pelling JC. 1992. Dietary energy and fat effects tributions of biologic and social and physical environmental deter-
on tumor promotion. Cancer Res, 52(7 Suppl), 2035s–​2039s. PMID: minants of cancer mortality. Annu Rev Public Health, 33, 137–​ 156.
1544138. PMCID: PMC3631776.
Block G, Hartman AM, and Naughton D. 1990. A reduced dietary question- Cole BF, Baron JA, Sandler RS, et al. 2007. Folic acid for the prevention of
naire: development and validation. Epidemiol, 1, 58–​64. colorectal adenomas: a randomized clinical trial. JAMA, 297(21), 2351–​
Blot WJ, Li JY, Taylor PR, et al. 1993. Nutrition intervention trials in Linxian, 2359. PMID: 17551129.
China: supplemenation with specific vitamin/​mineral combinations, can- Correa P, Fontham ETH, Bravo JC, et al. 2000. Chemoprevention of gastric dys-
cer incidence, and disease-​specific mortality in the general population. J plasia: randomized trial of antioxidant supplements and anti-​Helicobacter
Natl Cancer Inst, 85(18), 1483–​1492. pylori therapy. J Natl Cancer Inst, 92(23), 1881–​1888.
Blount BC, and Ames BN. 1994. Analysis of uracil in DNA by gas Cross AJ, Ferrucci LM, Risch A, et al. 2010. A large prospective study of meat
chromatography-​mass spectrometry. Analytical Biochemistry 219, consumption and colorectal cancer risk:  an investigation of potential
195–​200. mechanisms underlying this association. Cancer Res, 70(6), 2406–​2414.
Bohn SK, Blomhoff R, and Paur I. 2014. Coffee and cancer risk, epidemio- PMCID: PMC2840051.
logical evidence, and molecular mechanisms. Mol Nutr Food Res, 58(5), Cross AJ, Major JM, and Sinha R. 2011. Urinary biomarkers of meat consump-
915–​930. PMID: 24668519. tion. Cancer Epidemiol Biomarkers Prev, 20, 1107–​1111.
Boissonneault GA, Elson CE, and Pariza MW. 1986. Net energy effects of Crowe FL, Appleby PN, Travis RC, et  al. 2014. Circulating fatty acids and
dietary fat on chemically induced mammary carcinogenesis in F344 rats. prostate cancer risk: individual participant meta-​analysis of prospective
J Natl Cancer Inst, 76(2), 335–​338. PMID: 3080637. studies. J Natl Cancer Inst, 106(9). PMCID: PMC4188122.
Bonithon-​Kopp C, Kronborg O, Giocosa A, Rath U, and Faivre J. 2000. Calcium Cui Y, and Rohan TE. 2006. Vitamin D, calcium, and breast cancer risk:  a
and fibre supplementation in the prevention of colorectal adenoma recur- review. Cancer Epidemiol Biomarkers Prev, 15(8), 1427–​1447.
rence: a randomised intervention trial. Lancet, 356, 1300–​1306. de Munter JS, Hu FB, Spiegelman D, Franz M, and van Dam RM. 2007.
Bouvard V, Loomis D, Guyton KZ, et  al. 2015. Carcinogenicity of con- Whole grain, bran, and germ intake and risk of type 2 diabetes:  a pro-
sumption of red and processed meat. Lancet, 16, 1599–​1600 PMID: spective cohort study and systematic review. PLoS Med, 4(8), e261.
26514947 PMCID: PMC1952203.
Bravi F, Bosetti C, Tavani A, Gallus S, and La Vecchia C. 2013. Coffee Dickinson A, Blatman J, El-​Dash N, and Franco JC. 2014. Consumer usage and
reduces risk for hepatocellular carcinoma: an updated meta-​analysis. Clin reasons for using dietary supplements: report of a series of surveys. J Am
Gastroenterol Hepatol, 11(11), 1413–​1421 e1411. PMID: 23660416. Coll Nutr, 33(2), 176–​182. PMID: 24724775.
Brooks JD, Metter EJ, Chan DW, et  al. 2001. Plasma selenium level before Dodds EC, and Lawson W. 1936. Synthetic œstrogenic agents without the
diagnosis and the risk of prostate cancer development. J Urol, 166(6), phenanthrene nucleus. Nature, 137, 996.
2034–​2038. PMID: 11696701. Doll R, and Peto R. 1981. The causes of cancer: quantitative estimates of avoid-
Burkitt DP. 1971. Epidemiology of cancer of the colon and rectum. Cancer, able risks of cancer in the United States today. J Natl Cancer Inst, 66,
28, 3–​13. 1191–​1308.
Cadogan J, Eastell R, Jones N, and Barker ME. 1997. Milk intake and bone Dong JY, He K, Wang P, and Qin LQ. 2011. Dietary fiber intake and risk of
mineral acquisition in adolescent girls: randomised, controlled interven- breast cancer: a meta-​analysis of prospective cohort studies. Am J Clin
tion trial. BMJ, 315(7118), 1255–​1260. PMCID: PMC2127785. Nutr, 94(3), 900–​905. PMID: 21775566.
Calle EE, and Kaaks R. 2004. Overweight, obesity and cancer: epidemiological Dong JY, and Qin LQ. 2011. Soy isoflavones consumption and risk of breast
evidence and proposed mechanisms. Nature Rev, 4, 579–​591. cancer incidence or recurrence:  a meta-​analysis of prospective studies.
Cassidy A, Huang T, Rice MS, Rimm EB, and Tworoger SS. 2014. Intake of Breast Cancer Res Treat, 125(2), 315–​323. PMID: 21113655.
dietary flavonoids and risk of epithelial ovarian cancer. Am J Clin Nutr, Egeberg R, Olsen A, Loft S, et al. 2009. Intake of whole grain products and risk
100(5), 1344–​1351. PMCID: PMC4196485. of breast cancer by hormone receptor status and histology among post-
Chan DS, Lau R, Aune D et al. 2011. Red and processed meat and colorectal menopausal women. Int J Cancer, 124(3), 745–​750. PMID: 19004010.
cancer incidence: meta-​analysis of prospective studies. PLoS One, 6(6), Eliassen AH, Liao X, Rosner B, et  al. 2015. Plasma carotenoids and risk of
e20456. PMCID: PMC3108955. breast cancer over 20 y of follow-​up. Am J Clin Nutr, 101(6), 1197–​1205.
Chan JM, Van Blarigan EL, and Kenfield SA. 2014. What should we tell pros- PMCID: PMC4441811.
tate cancer patients about (secondary) prevention? Curr Opin Urol, 24(3), Emaus MJ, Peeters PH, Bakker MF, et al. 2016. Vegetable and fruit consump-
318–​323. PMCID: PMC4084902. tion and the risk of hormone receptor-​defined breast cancer in the EPIC
Chan JM, Wang F, and Holly EA. 2007. Whole grains and risk of pancre- cohort. Am J Clin Nutr, 103(1), 168–​177. PMID: 26607934.
atic cancer in a large population-​based case-​control study in the San Estruch R, Ros E, Salas-​Salvado J, et al. 2013. Primary prevention of cardio-
Francisco Bay Area, California. Am J Epidemiol, 166(10), 1174–​1185. vascular disease with a Mediterranean diet. N Engl J Med, 368(14), 1279–​
PMID: 17881383. 1290. PMID: 23432189.
Chen JG, Egner PA, Ng D, et al. 2013. Reduced aflatoxin exposure presages European Food Safety Authority. 2006. Opinion of the Scientific Panel on
decline in liver cancer mortality in an endemic region of China. Cancer Food Additives, Flavourings, Processing Aids and Materials in Contact
Prev Res (Phila), 6(10), 1038–​1045. PMCID: PMC3800239. with Food on a request from the Commission related to 2,2-​ BIS(4-​
Chen WY, Rosner B, Hankinson SE, Colditz GA, and Willett WC. HYDROXYPHENYL)PROPANE (Bisphenol A). EFSA, 428, 1–​75.
2011. Moderate alcohol consumption during adult life, drink- Farvid MS, Cho E, Chen WY, Eliassen AH, and Willett WC. 2014a. Dietary
ing patterns, and breast cancer risk. JAMA, 306(17), 1884–​ 1890. protein sources in early adulthood and breast cancer incidence: prospec-
PMCID: PMC3292347. tive cohort study. BMJ, 348, g3437. PMCID: PMC4051890.
Chlebowski RT, Blackburn GL, Thomson CA, et al. 2006. Dietary fat reduction Farvid MS, Cho E, Chen WY, Eliassen AH, and Willett WC. 2014b.
and breast cancer outcome:  interim efficacy results from the Women’s Premenopausal dietary fat in relation to pre-​and post-​ menopausal
Intervention Nutrition Study. J Natl Cancer Inst, 98(24), 1767–​1776. breast cancer. Breast Cancer Res Treat, 145(1), 255–​ 265.
PMID: 17179478. PMCID: PMC4126228.
Chlebowski RT, Johnson KC, Kooperberg C, et  al. 2008. Calcium plus vita- Farvid MS, Eliassen AH, Cho E, et  al. 2016. Dietary fiber intake in young
min D supplementation and the risk of breast cancer. J Natl Cancer Inst, adults and breast cancer risk. Pediatrics, 137(3), 1–​11. PMID: 26908709.
100(22), 1581–​1591. FDA. 2012a. Food additive petitions: American Chemistry Council. Available
Cho E, Smith-​Warner SA, Spiegelman D et al. 2004. Dairy foods, calcium, and at:  https://​plastics.americanchemistry.com/​Product-​Groups-​and-​Stats/​
colorectal cancer: a pooled analysis of 10 cohort studies. J Natl Cancer PolycarbonateBPA-​Global-​Group/​FDA-​Petition-​Letter-​ACC.pdf
Inst, 96, 1015–​1022. FDA. 2012b. Indirect food additives: polymers. Federal Register (Vol. 77, pp.
Cho SS, Qi L, Fahey GC, Jr., and Klurfeld DM. 2013. Consumption of cereal 41899–​41902): FDA, Washington, D.C.
fiber, mixtures of whole grains and bran, and whole grains and risk reduc- Feldman D, Krishnan AV, Swami S, Giovannucci E, and Feldman BJ. 2014.
tion in type 2 diabetes, obesity, and cardiovascular disease. Am J Clin The role of vitamin D in reducing cancer risk and progression. Nat Rev
Nutr, 98(2), 594–​619. PMID: 23803885. Cancer, 14(5), 342–​357. PMID: 24705652.
 345

Diet and Nutrition 345


Ferrari P, Rinaldi S, Jenab M, et  al. 2013. Dietary fiber intake and risk of Council on Epidemiology and Prevention. Circulation, 119(6), 902–​907.
hormonal receptor-​defined breast cancer in the European Prospective PMID: 19171857.
Investigation into Cancer and Nutrition study. Am J Clin Nutr, 97(2), Hashibe M, Galeone C, Buys SS, et al. 2015. Coffee, tea, caffeine intake, and
344–​353. PMID: 23269820. the risk of cancer in the PLCO cohort. Br J Cancer, 113(5), 809–​816.
Figueiredo JC, Hsu L, Hutter CM, et al. 2014. Genome-​wide diet-​gene interac- PMCID: PMC4559834.
tion analyses for risk of colorectal cancer. PLoS Genet, 10(4), e1004228. He J, Wang S, Zhou M, et al. 2015. Phytoestrogens and risk of prostate can-
PMCID: PMC3990510. cer:  a meta-​analysis of observational studies. World J Surg Oncol, 13,
Fortmann SP, Burda BU, Senger CA, Lin JS, and Whitlock EP. 2013. Vitamin 231. PMCID: PMC4521376.
and mineral supplements in the primary prevention of cardiovascular Helzlsouer K. 2010. Overview of the Cohort Consortium Vitamin D Pooling
disease and cancer: An updated systematic evidence review for the U.S. Project of Rarer Cancers. Am J Epidemiol, 172(1), 4–​ 9. PMID:
Preventive Services Task Force. Ann Intern Med, 159(12), 824–​ 834. 20562193
PMID: 24217421. Hennekens CH, Buring JE, Manson JE, et  al. 1996. Lack of effect of long-​
Freedman ND, Park Y, Abnet CC, Hollenbeck AR, and Sinha R. 2012. term supplementation with beta carotene on the incidence of malignant
Association of coffee drinking with total and cause-​specific mortality. N neoplasms and cardiovascular disease. N Engl J Med, 334, 1145–​1149.
Engl J Med, 366(20), 1891–​1904. PMCID: PMC3439152. Hercberg S, Galan P, Preziosi P, et al. 2004. The SU.VI.MAX study. A random-
Fritz H, Seely D, Flower G, et  al. 2013. Soy, red clover, and isoflavones ized, placebo-​controlled trial of the health effects of antioxidant vitamins
and breast cancer:  a systematic review. PLoS One, 8(11), e81968. and minerals. Arch Intern Med, 164, 2335–​2342. PMID: 15557412
PMCID: PMC3842968. Holick MF. 2004. Sunlight and vitamin D for bone health and prevention of
Fung T, Hu FB, Fuchs C, et  al. 2003. Major dietary patterns and the risk of autoimmune diseases, cancers, and cardiovascular disease. Am J Clin
colorectal cancer in women. Arch Intern Med, 163, 309–​314. Nutr, 80(Suppl), 1678S–​1688S. PMID: 15585788
Ganmaa D, Cui X, Feskanich D, Hankinson SE, and Willett WC. 2012. Milk, Howe GR, Benito E, Castelleto R, et  al. 1992. Dietary intake of fiber and
dairy intake and risk of endometrial cancer:  a 26-​year follow-​up. Int J decreased risk of cancers of the colon and rectum:  evidence from the
Cancer, 130(11), 2664–​2671. PMCID: PMC3359127. combined analysis of 13 case-​control studies. J Natl Cancer Inst, 84(24),
Gapstur SM, Jacobs EJ, Deka A, et al. 2011. Association of alcohol intake with 1887–​1896.
pancreatic cancer mortality in never smokers. Arch Intern Med, 171(5), Howe GR, Hirohata T, Hislop TG, et al. 1990. Dietary factors and risk of breast
444–​451. PMID: 21403041. cancer: combined analysis of 12 case-​control studies. J Natl Cancer Inst,
Gapstur SM, and Khan S. 2007. Fat, fruits, vegetables, and breast cancer survi- 82(7), 561–​569. PMID: 2156081.
vorship. JAMA, 298(335–​336). PMID: 17635896 Hu FB. 2002. Dietary pattern analysis: a new direction in nutritional epidemiol-
Garland CF, and Garland FC. 1980. Do sunlight and vitamin D reduce the like- ogy. Curr Opin Lipidol, 13(1), 3–​9.
lihood of colon cancer? Int J Epidemiol, 9(3), 227–​231. Hu FB, Rimm E, Smith-​Warner SA, et al. 1999. Reproducibility and validity of
Gaziano JM, Glynn RJ, Ghristen WG, et  al. 2009. Vitamins E and C in dietary patterns assessed with a food-​frequency questionnaire. Am J Clin
the prevention of prostate and total cancer in men. The Physician’s Nutr, 69(2), 243–​249. PMID: 9989687.
Health Study II Randomized Controlled Trial. JAMA, 301, 52–​ 62. IARC. 1994. Acrylamide. In:  International Agency for Research on Cancer
PMCID: PMC2774210 (Ed.),: Some industrial chemicals. IARC Monographs on the Evaluation
Gaziano JM, Sesso HD, Christen WG, et al. 2012. Multivitamins in the preven- of Carcinogenic Risks to Humans (Vol. 60). Lyon, France: IARC Press.
tion of cancer in men: the Physicians’ Health Study II randomized con- IARC. 2003. Fruit and vegetables. IARC Monographs on the Evaluation of
trolled trial. JAMA, 308(18), 1871–​1880. PMCID: PMC3517179. Carcinogenic Risks to Humans (Vol. 8). Lyon: IARC Press.
Geens T, Aerts D, Berthot C, et al. 2012. A review of dietary and non-​dietary IARC. 2010. Alcohol consumption and ethyl carbamate. IARC Monographs
exposure to bisphenol-​ A. Food Chem Toxicol, 50(10), 3725–​ 3740. on the Evaluation of Carcinogenic Risks to Humans (Vol. 96). Lyon,
PMID: 22889897. France: IARC Press.
Genkinger JM, Hunter DJ, Spiegelman D, et al. 2006. Dairy products and ovar- IARC. 2012a. Arsenic, metals, fibres, and dust. IARC Monographs on the
ian cancer:  a pooled analysis of 12 cohort studies. Cancer Epidemiol Evaluation of Carcinogenic Risks to Humans (Vol. 100C). Lyon,
Biomarkers Prev, 15(2), 364–​372. PMID: 16492930. France: IARC Press.
George SM, Ballard-​Barbash R, Manson JE, et  al. 2014. Comparing indices IARC. 2012b. Chemical agents and related occupations. IARC Monographs
of diet quality with chronic disease mortality risk in postmenopausal on the Evaluation of Carcinogenic Risks to Humans (Vol. 100F). Lyon,
women in the Women’s Health Initiative Observational Study: evidence France: IARC Press.
to inform national dietary guidance. Am J Epidemiol, 180(6), 616–​625. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans.
PMCID: PMC4157698. 1991. Coffee, tea, mate, methylxanthines and methylglyoxal. IARC
Gibson TM, Weinstein SJ, Pfeiffer RM, et  al. 2011. Pre-​and postfortifica- Monographs on the Evaluation of Carcinogenic Risks to Humans (Vol.
tion intake of folate and risk of colorectal cancer in a large prospective 51, pp. 1–​513). Lyon, France: IARC Press.
cohort study in the United States. Am J Clin Nutr, 94(4), 1053–​1062. Inoue-​Choi M, Robien K, and Lazovich D. 2013. Adherence to the WCRF/​
PMCID: PMC3173023. AICR guidelines for cancer prevention is associated with lower mortal-
Giovannucci E. 2001. Insulin, insulin-​like growth factors and colon cancer: a ity among older female cancer survivors. Cancer Epidemiol Biomarkers
review of the evidence. J Nutr, 131, 3109S–​3120S. Prev, 22(5), 792–​802. PMCID: PMC3650116.
Gorbach SL, and Goldin BR. 1987. Diet and the excretion and enterohepatic Institute of Medicine. 2011. Dietary reference intakes for calcium and vitamin
cycling of estrogens. Prev Med, 16(4), 525–​531. PMID: 3628202. D. Washington, DC: Institute of Medicine (IOM).
Gray R, and Wheatley K. 1991. How to avoid bias when comparing bone Izano MA, Fung TT, Chiuve SS, Hu FB, and Holmes MD. 2013. Are diet qual-
marrow transplantation with chemotherapy. Bone Marrow Transplant, ity scores after breast cancer diagnosis associated with improved breast
7(Suppl 3), 9–​12. cancer survival? Nutr Cancer, 65(6), 820–​826. PMCID: PMC3776311.
Greenberg ER, Baron JA, Tosteson TD, et  al. 1994. A clinical trial of anti- Joosen AM, Kuhnle GG, Aspinall SM, et  al. 2009. Effect of processed and
oxidant vitamins to prevent colorectal adenoma. Polyp Prevention Study red meat on endogenous nitrosation and DNA damage. Carcinogenesis,
Group. N Engl J Med, 331(3), 141–​147. PMID: 8008027. 30(8), 1402–​1407. PMCID: PMC2718076.
Guyton KZ, Loomis D, Grosse Y, et  al. 2015. Carcinogenicity of tetrachlor- Jung S, Spiegelman D, Baglietto L, et al. 2013. Fruit and vegetable intake and
vinphos, parathion, malathion, diazinon, and glyphosate. Lancet Oncol, risk of breast cancer by hormone receptor status. J Natl Cancer Inst,
16(5), 490–​491. PMID: 25801782. 105(3), 219–​236. PMCID: PMC3593764.
Hackshaw-​McGeagh LE, Perry RE, Leach VA, et al. 2015. A systematic review Kabat GC, Matthews CE, Kamensky V, Hollenbeck AR, and Rohan TE. 2015.
of dietary, nutritional, and physical activity interventions for the preven- Adherence to cancer prevention guidelines and cancer incidence, cancer
tion of prostate cancer progression and mortality. Cancer Causes Control, mortality, and total mortality: a prospective cohort study. Am J Clin Nutr,
26(11), 1521–​1550. PMCID: PMC4596907. 101(3), 558–​569. PMCID: PMC4340061.
Harnly JM, Doherty RF, Beecher GR et  al. 2006. Flavonoid content of Kennedy DA, Stern SJ, Moretti M, et  al. 2011. Folate intake and the risk
U.S. fruits, vegetables, and nuts. J Agric Food Chem, 54(26), 9966–​9977. of colorectal cancer:  a systematic review and meta-​ analysis. Cancer
PMID: 17177529. Epidemiol, 35(1), 2–​10. PMID: 21177150.
Harris WS, Mozaffarian D, Rimm E, et al. 2009. Omega-​6 fatty acids and risk Key TJ, Appleby PN, Cairns BJ, et al. 2011. Dietary fat and breast cancer: com-
for cardiovascular disease:  a science advisory from the American Heart parison of results from food diaries and food-​frequency questionnaires in
Association Nutrition Subcommittee of the Council on Nutrition, Physical the UK Dietary Cohort Consortium. Am J Clin Nutr, 94(4), 1043–​1052.
Activity, and Metabolism; Council on Cardiovascular Nursing; and PMID: 21865329.
346

346 PART III:  The Causes of Cancer


Key TJ, Appleby PN, Crowe FL, et  al. 2014. Cancer in British vegetari- Lee JE, Wei EK, Fuchs CS, et al. 2012. Plasma folate, methylenetetrahydro-
ans: updated analyses of 4998 incident cancers in a cohort of 32,491 meat folate reductase (MTHFR), and colorectal cancer risk in three large
eaters, 8612 fish eaters, 18,298 vegetarians, and 2246 vegans. Am J Clin nested case-​control studies. Cancer Causes Control, 23(4), 537–​545.
Nutr, 100 (Suppl 1), 378S–​385S. PMCID: PMC4144109. PMCID: PMC3721151.
Key TJ, Appleby PN, Reeves GK, Roddam AW, and Endogenous Hormones Lee JE, Willett WC, Fuchs CS, et al. 2011b. Folate intake and risk of colorectal
Breast Cancer Collaborative Group. 2010. Insulin-​like growth factor 1 cancer and adenoma: modification by time. Am J Clin Nutr, 93(4), 817–​
(IGF1), IGF binding protein 3 (IGFBP3), and breast cancer risk: pooled 825. PMCID: PMC3057549.
individual data analysis of 17 prospective studies. Lancet Oncol, 11(6), Lee SA, Shu XO, Li H, et al. 2009. Adolescent and adult soy food intake and
530–​542. PMCID: PMC3113287. breast cancer risk: results from the Shanghai Women’s Health Study. Am
Key TJ, Appleby PN, Travis RC, et al. 2015. Carotenoids, retinol, tocopherols, J Clin Nutr, 89(6), 1920–​1926. PMCID: PMC2683002.
and prostate cancer risk: pooled analysis of 15 studies. Am J Clin Nutr, Li G, Ma D, Zhang Y, Zheng W, and Wang P. 2013. Coffee consumption and
102(5), 1142–​1157. PMCID: PMC4625592. risk of colorectal cancer: a meta-​analysis of observational studies. Public
Kim EH, Willett WC, Fung T, Rosner B, and Holmes MD. 2011. Diet quality Health Nutr, 16(2), 346–​357. PMID: 22694939.
indices and postmenopausal breast cancer survival. Nutr Cancer, 63(3), Li H, Stampfer MJ, Giovannucci EL, et al. 2004. A prospective study of plasma
381–​388. PMCID: PMC3113520. selenium levels and prostate cancer risk. J Natl Cancer Inst, 96(9), 696–​
Kim Y, and Je Y. 2014. Vitamin D intake, blood 25(OH)D levels, and breast can- 703. PMID: 15126606.
cer risk or mortality: a meta-​analysis. Br J Cancer, 110(11), 2772–​2784. Liese AD, Krebs-​Smith SM, Subar AF, et  al. 2015. The Dietary Patterns
PMCID: PMC4037823. Methods Project:  synthesis of findings across cohorts and relevance to
Kirkpatrick SI, Subar AF, Douglass D, et al. 2014. Performance of the auto- dietary guidance. J Nutr, 145(3), 393–​402. PMCID: PMC4336525.
mated self-​ administered 24-​ hour recall relative to a measure of true Linos E, Willett WC, Cho E, and Frazier L. 2010. Adolescent diet in relation
intakes and to an interviewer-​administered 24-​h recall. Am J Clin Nutr, to breast cancer risk among premenopausal women. Cancer Epidemiol
100(1), 233–​240. PMCID: PMC4144101. Biomarkers Prev, 19(3), 689–​696. PMCID: PMC2866323.
Kjellstrom T, and Nordberg GF. 1978. A kinetic model of cadmium metabolism Lippman SM, Klein EA, Goodman PJ, et al. 2009. Effect of selenium and vita-
in the human being. Environ Res, 16(1–​3), 248–​269. PMID: 679914. min E on risk of prostate cancer and other cancers:  the Selenium and
Klein EA, Thompson IM, Jr., Tangen CM, et al. 2011. Vitamin E and the risk Vitamin E Cancer Prevention Trial (SELECT). JAMA, 301(1), 39–​51.
of prostate cancer: the Selenium and Vitamin E Cancer Prevention Trial PMCID: PMC3682779.
(SELECT). JAMA, 306(14), 1549–​1556. Liu C, Russell RM, and Wang X. 2003. Exposing ferrets to cigarette smoke
Klein EA, Thompson IM, Lippman SM, et al. 2001. SELECT: the next prostate and a pharmacologic dose of beta-​carotene supplementation enhance in
cancer prevention trial. Selenum and Vitamin E Cancer Prevention Trial. vitro retinoic acid catabolism in lungs via induction of cytochrome P450
J Urol, 166(4), 1311–​1315. PMID: 11547064. enzymes. J Nutr, 133, 173–​179.
Kristal AR, Till C, Song X, et  al. 2014. Plasma vitamin d and prostate can- Loomis D, Guyton KZ, Grosse Y, et  al. 2016. Carcinogenicity of drink-
cer risk:  results from the selenium and vitamin e cancer preven- ing coffee, mate, and very hot beverages. Lancet, 17(7), 877–​ 878.
tion trial. Cancer Epidemiol Biomarkers Prev, 23(8), 1494–​ 1504. PMID: 27318851
PMCID: PMC4119495. Lovegrove C, Ahmed K, Challacombe B, et  al. 2015. Systematic review of
Kushi LH, Doyle C, McCullough M, et  al. 2012. American Cancer Society prostate cancer risk and association with consumption of fish and fish-​
Guidelines on nutrition and physical activity for cancer prevention: reduc- oils:  analysis of 495,321 participants. Int J Clin Pract, 69(1), 87–​105.
ing the risk of cancer with healthy food choices and physical activity. CA PMID: 25495842.
Cancer J Clin, 62(1), 30–​67. PMID: 22237782. Ma Y, Zhang P, Wang F, et al. 2011. Association between vitamin D and risk
Kyro C, Olsen A, Bueno-​de-​Mesquita HB, et al. 2014a. Plasma alkylresorcinol of colorectal cancer:  a systematic review of prospective studies. J Clin
concentrations, biomarkers of whole-​grain wheat and rye intake, in the Oncol, 29(28), 3775–​3782. PMID: 21876081.
European Prospective Investigation into Cancer and Nutrition (EPIC) Magalhaes B, Bastos J, and Lunet N. 2011. Dietary patterns and colorectal
cohort. Br J Nutr, 111(10), 1881–​1890. PMID: 24521535. cancer:  a case-​control study from Portugal. Eur J Cancer Prev, 20(5),
Kyro C, Olsen A, Landberg R, et al. 2014b. Plasma alkylresorcinols, biomark- 389–​395. PMID: 21558858.
ers of whole-​grain wheat and rye intake, and incidence of colorectal can- Maiani G, Caston MJ, Catasta G, et  al. 2009. Carotenoids:  actual knowl-
cer. J Natl Cancer Inst, 106(1), djt352. PMCID: PMC3906988. edge on food sources, intakes, stability and bioavailability and their
Lampe JW. 2009. Is equol the key to the efficacy of soy foods? Am J Clin Nutr, protective role in humans. Mol Nutr Food Res, 53 Suppl 2, S194–​218.
89(5), 1664S–​1667S. PMCID: PMC2677009. PMID: 19035552.
Lamprecht SA, and Lipkin M. 2001. Cellular mechanisms of calcium and vita- Mannisto S, Smith-​Warner SA, Spiegelman D, et al. 2004. Dietary carotenoids
min D in the inhibition of colorectal carcinogenesis. Ann NY Acad Sci, and risk of lung cancer in a pooled analysis of seven cohort studies.
952, 73–​87. Cancer Epidemiol Biomarkers Prev, 13, 40–​48.
Lappe JM, Travers-​Gustafson D, Davies KM, Recker RR, and Heaney RP. 2007. Manson JE, and Bassuk SS. 2015. Vitamin D research and clinical practice: at a
Vitamin D and calcium supplementation reduces cancer risk: results of a crossroads. JAMA, 313(13), 1311–​1312. PMID: 25695911.
randomized trial. Am J Clin Nutr, 85(6), 1586–​1591. Manson JE, Bassuk SS, Lee IM, et  al. 2012. The VITamin D and OmegA-​3
Larsson SC, Bergkvist L, and Wolk A. 2009. Long-​term dietary calcium intake TriaL (VITAL):  rationale and design of a large randomized controlled
and breast cancer risk in a prospective cohort of women. Am J Clin Nutr, trial of vitamin D and marine omega-​3 fatty acid supplements for the
89(1), 277–​282. PMID: 19056569. primary prevention of cancer and cardiovascular disease. Contemp Clin
Larsson SC, Orsini N, and Wolk A. 2015. Urinary cadmium concentration Trials, 33(1), 159–​171. PMCID: PMC3253961.
and risk of breast cancer: a systematic review and dose-​response meta-​ Martin LJ, Li Q, Melnichouk O, et  al. 2011. A randomized trial of dietary
analysis. Am J Epidemiol, 182(5), 375–​380. PMID: 26254432. intervention for breast cancer prevention. Cancer Res, 71(1), 123–​133.
Lauby-​Secretan B, Scoccianti C, Loomis D, et  al. 2016. Body Fatness and PMID: 21199800.
Cancer—​Viewpoint of the IARC Working Group. N Engl J Med, 375, McCann SE, Thompson LU, Nie J, et al. 2010. Dietary lignan intakes in rela-
794–​798. tion to survival among women with breast cancer: the Western New York
Lee I, Cook NR, Manson JE, Buring JE, and Hennekens CH. 1999. B-​carotene Exposures and Breast Cancer (WEB) Study. Breast Cancer Res Treat,
supplementation and incideence of cancer and cardiovascular disease: the 122(1), 229–​235. PMCID: PMC2883619.
women’s health study. J Natl Cancer Inst, 91(24), 2102–​2106. McCullough ML, Patel AV, Kushi LH, et al. 2011. Following cancer preven-
Lee JE, Hunter DJ, Spiegelman D, et al. 2007. Intakes of coffee, tea, milk, soda tion guidelines reduces risk of cancer, cardiovascular disease, and all-​
and juice and renal cell cancer in a pooled analysis of 13 prospective stud- cause mortality. Cancer Epidemiol Biomarkers Prev, 20(6), 1089–​1097.
ies. Int J Cancer. 121(10), 2246–​2253. PMID: 17583573. PMID: 21467238.
Lee JE, Li H, Chan AT, et  al. 2011a. Circulating levels of vitamin D and McCullough ML, Weinstein SJ, Freedman DM, et al. 2010. Correlates of circulat-
colon and rectal cancer:  the Physicians’ Health Study and a meta-​ ing 25-​hydroxyvitamin D: Cohort Consortium Vitamin D Pooling Project
analysis of prospective studies. Cancer Prev Res (Phila), 4(5), 735–​743. of Rarer Cancers. Am J Epidemiol, 172(1), 21–​35. PMCID: PMC2892536.
PMCID: PMC3412303. Menkes MS, Comstock GW, Vuilleumier JP, et al. 1986. Serum beta-​carotene,
Lee JE, Mannisto S, Spiegelman D, et  al. 2009. Intakes of fruit, vegetables, vitamins A and E, selenium, and the risk of lung cancer. N Engl J Med,
and carotenoids and renal cell cancer risk: a pooled analysis of 13 pro- 315(20), 1250–​1254. PMID: 3773937.
spective studies. Cancer Epidemiol Biomarkers Prev, 18(6), 1730–​1739. Menzel P. 2005. Hungry planet family food portraits.Available from: http://m
​ enzel-
PMID: 19505906. photo.photoshelter.com/​gallery/​Hungry-​Planet-​Family-​Food-​Portraits/​
 347

Diet and Nutrition 347


G0000zmgWvU6SiKM/​C0000k7JgEHhEq0w. Accessed on March Pereira MA, Jacobs DR, Jr, Pins JJ, et al. 2002. Effect of whole grains on insu-
4, 2016. lin sensitivity in overweight hyperinsulinemic adults. Am J Clin Nutr,
Messina MJ, and Wood CE. 2008. Soy isoflavones, estrogen therapy, 75(5), 848–​855.
and breast cancer risk:  analysis and commentary. Nutr J, 7, 17. Peterson JJ, Dwyer JT, Jacques PF, and McCullough ML. 2015. Improving the
PMCID: PMC2443803. estimation of flavonoid intake for study of health outcomes. Nutr Rev,
Meyerhardt JA, Niedzwiecki D, Hollis D, et al. 2007. Association of dietary 73(8), 553–​576. PMCID: PMC4581481.
patterns with cancer recurrence and survival in patients with stage III Petrick JL, Freedman ND, Graubard BI, et al. 2015. Coffee consumption and
colon cancer. JAMA, 298(7), 754–​764. PMID: 17699009. risk of hepatocellular carcinoma and intrahepatic cholangiocarcinoma by
Miller JC. 1994. Importance of glycemic index in diabetes. Am J Clin Nutr, sex:  The Liver Cancer Pooling Project. Cancer Epidemiol Biomarkers
59(3 Suppl), 747S-​752S. PMID: 8116560. Prev, 24(9), 1398–​1406. PMCID: PMC4576990.
Missmer SA, Smith-​Warner SA, Spiegelman D, et  al. 2002. Meat and dairy Pierce JP, Natarajan L, Caan BJ, et  al. 2007. Influence of a diet very
food consumption and breast cancer: a pooled analysis of cohort studies. high in vegetables, fruit, and fiber and low in fat on prognosis fol-
Int J Epidemiol, 31, 78–​85. lowing treatment for breast cancer:  the Women’s Healthy Eating
Moyer VA, and USPS Task Force. 2014. Vitamin, mineral, and multivitamin and Living (WHEL) randomized trial. JAMA, 298(3), 289–​ 298.
supplements for the primary prevention of cardiovascular disease and PMCID: PMC2083253.
cancer: U.S. Preventive Services Task Force recommendation statement. Prentice RL, Caan B, Chlebowski RT, et al. 2006. Low-​fat dietary pattern and
Ann Intern Med, 160(8), 558–​564. PMID: 24566474. risk of invasive breast cancer: the Women’s Health Initiative Randomized
Mozaffarian D, Hao T, Rimm EB, Willett WC, and Hu FB. 2011. Changes in Controlled Dietary Modification Trial. JAMA, 295(6), 629–​ 642.
diet and lifestyle and long-​term weight gain in women and men. N Engl J PMID: 16467232
Med, 364(25), 2392–​2404. PMCID: PMC3151731. Prentice RL, Kakar F, Hursting S, et  al. 1988. Aspects of the rationale
Mulholland HG, Murray LJ, Cardwell CR, and Cantwell MM. 2009. Glycemic for the Women’s Health Trial. J Natl Cancer Inst, 80(11), 802–​814.
index, glycemic load, and risk of digestive tract neoplasms:  a sys- PMID: 3292773.
tematic review and meta-​ analysis. Am J Clin Nutr, 89(2), 568–​ 576. Produce for Better Health Foundation. State of the Plate. 2015 study on
PMID: 19088152. America’s consumption of fruit and vegetables. Produce for Better Health
Murphy N, Norat T, Ferrari P, et  al. 2012. Dietary fibre intake and risks of Foundation, 2015. Web. <http://​www.PBHFoundation.org>
cancers of the colon and rectum in the European prospective inves- Qiao YL, Dawsey SM, Kamangar F, et al. 2009. Total and cancer mortality after
tigation into cancer and nutrition (EPIC). PLoS One, 7(6), e39361. supplementation with vitamins and minerals:  follow-​up of the Linxian
PMCID: PMC3382210. General Population Nutrition Intervention Trial. J Natl Cancer Inst,
Nagata C. 2010. Factors to consider in the association between soy isofla- 101(7), 507–​518. PMCID: PMC2664089.
vone intake and breast cancer risk. J Epidemiol, 20(2), 83–​89. PMCID: Rich-​Edwards JW, Ganmaa D, Pollak MN, et al. 2007. Milk consumption and
PMC3900805. the prepubertal somatotropic axis. Nutr J, 6, 28. PMCID: PMC2098760.
Nair SC, Toshkov IA, Yaktine AL, et  al. 1995. Dietary energy restriction-​ Richman EL, Kenfield SA, Chavarro JE, et al. 2013. Fat intake after diagnosis
induced modulation of protein kinase C zeta isozyme in the hamster pan- and risk of lethal prostate cancer and all-​cause mortality. JAMA Intern
creas. Mol Carcinog, 14(1), 10–​15. PMID: 7546218. Med, 173(14), 1318–​1326. PMCID: PMC3935610.
Nan H, Lee JE, Rimm EB, et  al. 2013. Prospective study of alcohol con- Rimm EB, Giovannucci EL, Stampfer MJ, et  al. 1992. Reproducibility and
sumption and the risk of colorectal cancer before and after folic acid validity of a expanded self-​administered semiquantitative food frequency
fortification in the United States. Ann Epidemiol, 23(9), 558–​ 563. questionnaire among male health professionals. Am J Epidemiol, 135,
PMCID: PMC3749268. 1114–​1126.
Neuhouser ML, Platz EA, Till C, et  al. 2013. Insulin-​like growth factors Rochester JR. 2013. Bisphenol A and human health: a review of the literature.
and insulin-​ like growth factor-​ binding proteins and prostate cancer Reprod Toxicol, 42, 132–​155. PMID: 23994667.
risk:  results from the prostate cancer prevention trial. Cancer Prev Res Rock CL, Doyle C, Demark-​Wahnefried W, et al. 2012. Nutrition and physical
(Phila), 6(2), 91–​99. PMCID: PMC3565024. activity guidelines for cancer survivors. CA Cancer J Clin, 62(4), 242–​
Ng K, Wolpin BM, Meyerhardt JA, et al. 2009. Prospective study of predictors 274. PMID: 22539238.
of vitamin D status and survival in patients with colorectal cancer. Br J Romagnolo DF, and Selmin OI. 2012. Flavonoids and cancer prevention:  a
Cancer, 101, 916–​923. review of the evidence. J Nutr Gerontol Geriatr, 31(3), 206–​ 238.
Nomura AM, Lee J, Stemmermann GN, and Combs GF, Jr. 2000. Serum PMID: 22888839.
selenium and subsequent risk of prostate cancer. Cancer Epidemiol Romaguera D, Vergnaud AC, Peeters PH, et al. 2012. Is concordance with World
Biomarkers Prev, 9(9), 883–​887. PMID: 11008904. Cancer Research Fund/​American Institute for Cancer Research guide-
O'Keefe SJD, Li JV, Lahti L, et al. 2015. Fat, Fiber and Cancer Risk in African lines for cancer prevention related to subsequent risk of cancer? Results
Americans and Rural Africans. Nat Commun, 6, 6342. from the EPIC study. Am J Clin Nutr, 96(1), 150–​163. PMID: 22592101.
Ollberding NJ, Wilkens LR, Henderson BE, Kolonel LN, and Le Marchand Ross MH, and Bras G. 1971. Lasting influence of early caloric restriction on
L. 2012. Meat consumption, heterocyclic amines and colorectal cancer prevalence of neoplasms in the rat. J Natl Cancer Inst, 47(5), 1095–​1113.
risk:  the Multiethnic Cohort Study. Int J Cancer, 131(7), E1125–​1133. PMID: 4330798.
PMCID: PMC3553660. Rothwell JA, Perez-​Jimenez J, Neveu V, et  al. 2013. Phenol-​Explorer 3.0:  a
Omenn GS, Goodman GE, Thornquist MD, et al. 1996. Effects of a combina- major update of the Phenol-​Explorer database to incorporate data on the
tion of beta carotene and vitamin A  on lung cancer and cardiovascular effects of food processing on polyphenol content. Database (Oxford),
disease. N Engl J Med, 334, 1150–​1155. 2013, bat070. PMCID: PMC3792339.
Ong TP, Moreno FS, and Ross SA. 2011. Targeting the epigenome with bioac- Rowlands MA, Gunnell D, Harris R, et al. 2009. Circulating insulin-​like growth
tive food components for cancer prevention. J Nutrigenet Nutrigenomics, factor peptides and prostate cancer risk: a systematic review and meta-​
4(5), 275–​292. PMCID: PMC3388269. analysis. Int J Cancer, 124(10), 2416–​2429. PMCID: PMC2743036.
Orlich MJ, Singh PN, Sabate J, et  al. 2015. Vegetarian dietary patterns and Saarinen NM, Tuominen J, Pylkkanen L, and Santti R. 2010. Assessment of
the risk of colorectal cancers. JAMA Intern Med, 175(5), 767–​ 776. information to substantiate a health claim on the prevention of prostate
PMCID: PMC4420687. cancer by lignans. Nutrients, 2(2), 99–​115. PMCID: PMC3257165.
Panel on Dietary Antioxidants and Related Compounds. 2000. Dietary ref- Scalbert A, Brennan L, Manach C, et  al. 2014. The food metabolome:  a
erence intakes for vitamin C, vitamin E, selenium, and carotenoids. window over dietary exposure. Am J Clin Nutr, 99(6), 1286–​ 1308.
Washington, DC: National Academies Press. PMID: 24760973.
Park Y, Hunter DJ, Spiegelman D, et al. 2005. Dietary fiber intake and risk of Schatzkin A, Lanza E, Corle D, et al. 2000. Lack of effect of a low-​fat, high-​
colorectal cancer: a pooled analysis of prospective cohort studies. JAMA, fiber diet on the recurrence of colorectal adenomas. N Engl J Med,
294(22), 2849–​2857. PMID: 16352792. 342(16), 1149–​1155.
Parkin DM. 2011. The fraction of cancer attributable to lifestyle and envi- Schatzkin A, Mouw T, Park Y, et al. 2007. Dietary fiber and whole-​grain con-
ronmental factors in the UK in 2010. Br J Cancer, 105(Suppl 2), S2–​5. sumption in relation to colorectal cancer in the NIH-​AARP Diet and
PMCID: PMC3252063. Health Study. Am J Clin Nutr, 85(5), 1353–​1360. PMID: 17490973.
Pelucchi C, La Vecchia C, Bosetti C, Boyle P, and Boffetta P. 2011. Exposure to Schatzkin A, Park Y, Leitzmann MF, Hollenbeck AR, and Cross AJ. 2008.
acrylamide and human cancer: a review and meta-​analysis of epidemio- Prospective study of dietary fiber, whole grain foods, and small intestinal
logic studies. Ann Oncol, 22(7), 1487–​1499. PMID: 21239401. cancer. Gastroenterology, 135(4), 1163–​1167. PMID: 18727930.
348

348 PART III:  The Causes of Cancer


Schwartz GG, and Skinner HG. 2012. A prospective study of total and ion- in the Women’s Health Initiative. Cancer Prev Res (Phila), 7(1), 42–​53.
ized serum calcium and time to fatal prostate cancer. Cancer Epidemiol PMID: 24403289.
Biomarkers Prev, 21(10), 1768–​1773. PMID: 22914529. Thomson CA, Van Horn L, Caan BJ, et al. 2014b. Cancer incidence and mortal-
Schwingshackl L, and Hoffmann G. 2015. Adherence to Mediterranean ity during the intervention and postintervention periods of the Women’s
diet and risk of cancer:  an updated systematic review and meta-​ Health Initiative dietary modification trial. Cancer Epidemiol Biomarkers
analysis of observational studies. Cancer Med, 4(12), 1933–​ 1947. Prev, 23(12), 2924–​2935. PMCID: PMC4257879.
PMID: 26471010. Thun MT, Peto R, Lopez AD, et al. 1997. Alcohol consumption and mortal-
Setchell KD, Brown NM, Zhao X, et  al. 2011. Soy isoflavone phase II ity among middle-​aged and elderly U.S. adults. N Engl J Med, 337(24),
metabolism differs between rodents and humans:  implications for 1705–​1714.
the effect on breast cancer risk. Am J Clin Nutr, 94(5), 1284–​1294. Tobias DK, Chen M, Manson JE, et al. 2015. Effect of low-​fat diet interventions
PMCID: PMC3192476. versus other diet interventions on long-​term weight change in adults: a
Shai I, Schwarzfuchs D, Henkin Y, et  al. 2008. Weight loss with a low-​ systematic review and meta-​analysis. Lancet Diabetes Endocrinol, 3(12),
carbohydrate, Mediterranean, or Low-​fat diet. N Engl J Med, 359(3), 968–​979. PMCID: PMC4667723.
229–​241. Toledo E, Salas-​Salvado J, Donat-​Vargas C, et  al. 2015. Mediterranean diet
Shin M, Holmes MD, Hankinson SE, et al. 2002. Intake of dairy products, cal- and invasive breast cancer risk among women at high cardiovascular risk
cium, and vitamin D and risk of breast cancer. J Natl Cancer Inst, 94(17), in the PREDIMED Trial: a randomized clinical trial. JAMA Intern Med,
1301–​1311. PMID: 12208895 175(11), 1752–​1760. PMID: 26365989.
Sieri S, Chiodini P, Agnoli C, et  al. 2014. Dietary fat intake and develop- Trichopoulou A, Costacou T, Bamia C, and Trichopoulos D. 2003. Adherence
ment of specific breast cancer subtypes. J Natl Cancer Inst, 106(5). to a Mediterranean diet and survival in a Greek population. N Engl J Med,
PMID: 24718872. 348(26), 2599–​2608.
Sinha R, Knize MG, Salmon CP, et al. 1998a. Heterocyclic amine content of Trichopoulou A, Martinez-​Gonzalez MA, Tong TY, et al. 2014. Definitions and
pork products cooked by different methods and to varying degrees of potential health benefits of the Mediterranean diet:  views from experts
doneness. Food Chem Toxicol, 36(4), 289–​297. PMID: 9651045. around the world. BMC Med, 12, 112. PMCID: PMC4222885.
Sinha R, Rothman N, Salmon CP, et al. 1998b. Heterocyclic amine content in Trock B, Lanza E, and Greenwald P. 1990. Dietary fiber, vegetables, and colon
beef cooked by different methods to varying degrees of doneness and cancer: critical review and meta-​analyses of the epidemiologic evidence.
gravy made from meat drippings. Food Chem Toxicol, 36(4), 279–​287. J Natl Cancer Inst, 82, 650–​661.
PMID: 9651044. US Department of Health and Human Services and US Department of
Smith-​Warner SA, Spiegelman D, Adami HO, et  al. 2001. Types of dietary Agriculture. 2015. 2015–​2020 dietary guidelines for Americans (8th ed.).
fat and breast cancer: a pooled analysis of cohort studies. Int J Cancer, December 2015. Available at http://​health.gov/​dietaryguidelines/​2015/​
92(5), 767–​774. guidelines/​.
Smith-​Warner SA, Spiegelman D, Ritz J, et  al. 2006. Methods for pooling US Dietary Guidelines Advisory Committee. 2015. Scientific report of the 2015
results of epidemiologic studies:  the Pooling Project of Prospective US Dietary Guidelines Advisory Committee. USDA. Available at: https://​
Studies of Diet and Cancer. Am J Epidemiol, 163(11), 1053–​ 1064. health.gov/​ d ietaryguidelines/​ 2 015-​ s cientific-​ r eport/​ p dfs/​ s cientific-​
PMID: 16624970. report-​of-​the-​2015-​dietary-​guidelines-​advisory-​committee.pdf
Smith-​Warner SA, Spiegelman D, Yaun SS, et  al. 1998. Alcohol and breast Van Blarigan EL, Kenfield SA, Yang M, et al. 2015. Fat intake after prostate
cancer in women:  a pooled analysis of cohort studies. JAMA, 279(7), cancer diagnosis and mortality in the Physicians’ Health Study. Cancer
535–​540. Causes Control, 26(8), 1117–​1126. PMCID: PMC4499006.
Stahelin HB, Rosel F, Buess E, and Brubacher G. 1984. Cancer, vitamins, and Van Blarigan EL, and Meyerhardt JA. 2015. Role of physical activity and
plasma lipids: prospective Basel study. J Natl Cancer Inst, 73(6), 1463–​ diet after colorectal cancer diagnosis. J Clin Oncol, 33(16), 1825–​1834.
1468. PMID: 6595456. PMCID: PMC4438267.
State of California. 2015. Notice of intent to list:  tetrachlorvinphos, Van Dam RM, and Hunter DJ. 2013. Biochemical indicators of dietary
parathion, malathion, glyphosate. Available from: http://​oehha. intake. In:  Willett WC (Ed.), Nutritional epidemiology (3rd ed.).
ca.gov/ ​ p roposition- ​ 6 5/ ​ c rnr/ ​ n otice- ​ i ntent- ​ l ist- ​ t etrachlorvinphos- New York: Oxford University Press.
​parathion-​malathion-​glyphosate. Vartanian LR, Schwartz MB, and Brownell KD. 2007. Effects of soft drink con-
Stevens VL, McCullough ML, Sun J, et al. 2011. High levels of folate from sumption on nutrition and health: a systematic review and meta-​analysis.
supplements and fortification are not associated with increased risk Am J Public Health, 97(4), 667–​675. PMCID: PMC1829363.
of colorectal cancer. Gastroenterology, 141(1), 98–​ 105, 105 e101. Vastag B. 2007. Soy and prostate cancer study results mixed. J Natl Cancer
PMID: 21586288. Inst, 99(18), 1364–​1365. PMID: 17848664.
Stolzenberg-​Solomon RZ, Hayes RB, Horst RL, et al. 2009. Serum vitamin D Velentzis LS, Cantwell MM, Cardwell C, et al. 2009. Lignans and breast cancer
and risk of pancreatic cancer in the prostate, lung, colorectal, and ovarian risk in pre-​and post-​menopausal women: meta-​analyses of observational
screening trial. Cancer Res, 69(4), 1439–​1447. PMCID: PMC3052200 studies. Br J Cancer, 100(9), 1492–​1498. PMCID: PMC2694431.
Sun Z, Chen T, Thorgeirsson SS, et al. 2013. Dramatic reduction of liver can- Vergnaud AC, Romaguera D, Peeters PH, et al. 2013. Adherence to the World
cer incidence in young adults: 28 year follow-​up of etiological interven- Cancer Research Fund/​American Institute for Cancer Research guide-
tions in an endemic area of China. Carcinogenesis, 34(8), 1800–​1805. lines and risk of death in Europe: results from the European Prospective
PMCID: PMC3731800. Investigation into Nutrition and Cancer cohort study. Am J Clin Nutr,
Szymanski KM, Wheeler DC, and Mucci LA. 2010. Fish consumption and 97(5), 1107–​1120. PMID: 23553166.
prostate cancer risk: a review and meta-​analysis. Am J Clin Nutr, 92(5), Virk-​Baker MK, Nagy TR, Barnes S, and Groopman J. 2014. Dietary acryl-
1223–​1233. PMID: 20844069. amide and human cancer: a systematic review of literature. Nutr Cancer,
Tannenbaum A. 1942. The genesis and growth of tumors. III: Effects of a high 66(5), 774–​790. PMCID: PMC4164905.
fat diet. Cancer Res, 2, 468–​475. Visvanathan K, Mondul A, Zeleniuch-​Jacquotte A, et  al. 2015. Circulating
Tannenbaum A, and Silverstone H. 1953. Nutrition in relation to cancer. Adv vitamin D concentrations and breast cancer risk: a pooled analysis of
Cancer Res, 1, 451–​501. 17 cohorts [abstract]. Paper presented at the Thirty-​Seventh Annual
Tannour-​Louet M, Lewis SK, Louet JF, et  al. 2014. Increased expression of CTRC-​ AACR San Antonio Breast Cancer Symposium, 2014, San
CYP24A1 correlates with advanced stages of prostate cancer and can Antonio, TX.
cause resistance to vitamin D3-​based therapies. FASEB J, 28(1), 364–​372. Vogt TM, Ziegler RG, Graubard BI, et al. 2003. Serum selenium and risk of
PMID: 24081904. prostate cancer in U.S. blacks and whites. Int J Cancer, 103(5), 664–​670.
Teegarden D, Romieu I, and Lelievre SA. 2012. Redefining the impact of nutri- PMID: 12494476.
tion on breast cancer incidence: is epigenetics involved? Nutr Res Rev, Wactawski-​Wende J, Kotchen JM, Anderson GL, et al. 2006. Calcium plus vita-
25(1), 68–​95. PMCID: PMC4090752. min D supplementation and the risk of colorectal cancer. N Eng J Med,
The Alpha-​Tocopherol Beta-​Carotene Cancer Prevention Study Group. 1994. 354(7), 684–​696.
The effect of vitamin E and beta carotene on the incidence of lung cancer Wang L, Lee IM, Zhang SM, et al. 2009. Dietary intake of selected flavonols,
and other cancers in male smokers. N Engl J Med, 330, 1029–​1035. flavones, and flavonoid-​rich foods and risk of cancer in middle-​aged and
The Women’s Health Initiative Study Group. 1998. Design of the Women’s older women. Am J Clin Nutr, 89(3), 905–​912. PMCID: PMC2667658.
Health Initiative clinical trial and observational study. Control Clin Trials, Wang XD, and Russell R. 1999. Procarcinogenic and anticarcinogenic effects
19(1), 61–​109. of beta-​carotene. Nutr Rev, 57, 263–​272.
Thomson CA, McCullough ML, Wertheim BC, et  al. 2014a. Nutrition and WCRF International. 2015. Cancer preventability estimates for diet, nutrition,
physical activity cancer prevention guidelines, cancer risk, and mortality body fatness, and physical activity. Available at:  http://​www.wcrf.org/​
 349

Diet and Nutrition 349


int/​cancer-​facts-​figures/​preventability-​estimates/​cancer-​preventability-​ Wilson KM, Vesper HW, Tocco P, et al. 2009. Validation of a food frequency
estimates-​diet-​nutrition. Accessed on October 20, 2016. questionnaire measurement of dietary acrylamide intake using hemo-
WCRF/​AICR. 1997. Food, nutrition, and the prevention of cancer:  a global globin adducts of acrylamide and glycidamide. Cancer Causes Control,
perspective. Washington, DC: AICR. 20(3), 269–​278. PMCID: PMC3147248.
WCRF/​AICR. 2007. Food, nutrition, physical activity, and the prevention of Wolever TMS, and Jenkins DJA. 1986. The use of the glycemic index in predicting
cancer: a global perspective. Washington DC: AICR. the blood glucose response to mixed meals. Am J Clin Nutr, 43, 167–​172.
WCRF/​AICR. 2010. Continuous update project report: food, nutrition, physi- Wolpin BM, Ng K, Bao Y, et al. 2012. Plasma 25-​hydroxyvitamin D and risk
cal activity, and the prevention of breast cancer. London; Washington, of pancreatic cancer. Cancer Epidemiol Biomarkers Prev, 21(1), 82–​91.
DC: WCRF/​AICR. PMCID: PMC3253914.
WCRF/​AICR. 2011. Continuous update project report: food, nutrition, physi- Wu X, Zhu X, and Xie M. 2015. Association between dietary cadmium expo-
cal activity and the prevention of colorectal cancer. London; Washington, sure and breast cancer risk:  an updated meta-​analysis of observational
DC:: WCRF/​AICR. studies. Med Sci Monit, 21, 769–​775. PMCID: PMC4371715.
WCRF/​AICR. 2013. Continuous update project report: food, nutrition, physical Xie W, Yang M, Chan J, et al. 2016. Association of genetic variations of seleno-
activity, and the prevention of endometrial cancer. London; Washington, protein genes, plasma selenium levels, and prostate cancer aggressiveness
DC: WCRF/​AICR. at diagnosis. Prostate, 76(7), 691–​699, PMID: 26847995.
WCRF/​AICR. 2014a. Continuous update project report:  diet, nutrition, Xu Y, Shao X, Yao Y, et al. 2014. Positive association between circulating 25-​
physical activity, and breast cancer survivors. London; Washington, hydroxyvitamin D levels and prostate cancer risk: new findings from an
DC: WCRF/​AICR. updated meta-​analysis. J Cancer Res Clin Oncol, 140(9), 1465–​1477.
WCRF/​AICR. 2014b. Continuous update project report: diet, nutrition, physi- PMID: 24838848.
cal activity, and prevention of prostate cancer. London; Washington, Yang M, Kenfield SA, Van Blarigan EL, et al. 2015. Dietary patterns after pros-
DC: WCRF/​AICR. tate cancer diagnosis in relation to disease-​specific and total mortality.
WCRF/​AICR. 2015a. Continuous update project report:  diet, nutrition, Cancer Prev Res (Phila), 8(6), 545–​551. PMCID: PMC4452958.
physical activity and prevention of kidney cancer. London; Washington, Yang TO, Crowe F, Cairns BJ, Reeves GK, and Beral V. 2015. Tea and coffee
DC: WCRF/​AICR. and risk of endometrial cancer: cohort study and meta-​analysis. Am J Clin
WCRF/​AICR. 2015b. Continuous update project report:  diet, nutrition, Nutr, 101(3), 570–​578. PMCID: PMC4340062.
physical activity and prevention of liver cancer. London; Washington, Yin L, Grandi N, Raum E, et  al. 2010. Meta-​analysis:  serum vitamin D and
DC: WCRF/​AICR. breast cancer risk. Eur J Cancer, 46, 2196–​2205. PMID: 20456946
Webb A, and McCullough ML. 2005. Dietary lignans: potential role in cancer Yoshizawa K, Willett WC, Morris SJ, et al. 1998. Study of prediagnostic sele-
prevention. Nutr Cancer, 51(2), 117–​131. nium level in toenails and the risk of advanced prostate cancer. J Natl
Weindruch R, and Walford RL. 1982. Dietary restriction in mice beginning Cancer Inst, 90(16), 1219–​1224.
at 1  year of age:  effect on life-​span and spontaneous cancer incidence. Yuan JM, Sun C, and Bustler LM. 2011. Tea and cancer prevention. Pharmacol
Science, 215(4538), 1415–​1418. PMID: 7063854. Res, 64(2), 123–​135. PMID: 21419224
Wells HF, Hodan F, and Buzby JC. 2008. Dietary assessment of major trends in Zamora-​Ros R, Agudo A, Lujan-​Barroso L, et  al. 2012. Dietary flavonoid
U.S. food consumption, 1970–​2005. Economic Information Bulletin No. and lignan intake and gastric adenocarcinoma risk in the European
33. US Department of Agriculture. Prospective Investigation into Cancer and Nutrition (EPIC) study. Am J
Welsh JA, Sharma AJ, Grellinger L, and Vos MB. 2011. Consumption of added Clin Nutr, 96(6), 1398–​1408. PMID: 23076618.
sugars is decreasing in the United States. Am J Clin Nutr, 94(3), 726–​734. Zamora-​Ros R, Fedirko V, Trichopoulou A, et al. 2013. Dietary flavonoid, lig-
PMCID: PMC3155936. nan and antioxidant capacity and risk of hepatocellular carcinoma in the
Willett W. 2013. Genetics in dietary analyses nutritional epidemiology. European prospective investigation into cancer and nutrition study. Int J
New York: Oxford University Press. Cancer, 133(10), 2429–​2443. PMID: 23649669.
Willett WC. 2010. The WHI joins MRFIT: a revealing look beneath the covers. Zamora-​Ros R, Sacerdote C, Ricceri F, et al. 2014. Flavonoid and lignan intake
Am J Clin Nutr, 91(4), 829–​830. PMID: 20181816. in relation to bladder cancer risk in the European Prospective Investigation
Willett WC. 2013. Nutritional epidemiology (3rd ed.). New  York:  Oxford into Cancer and Nutrition (EPIC) study. Br J Cancer, 111(9), 1870–​1880.
University Press. PMCID: PMC4453722.
Willett WC, and Colditz GA. 2013. Vitamin A and lung cancer. In: Willett WC (Ed.), Ziegler RG, Smith-​Warner SA, Fedirko V, et al: Circulating vitamin D and risk of
Nutritional epidemiology (3rd ed.). New York: Oxford University Press. breast and colorectal cancer: an international pooling project of 21 cohorts,
Willett WC, Sampson L, Stampfer MJ, et  al. 1985. Reproducibility and valid- 2015 Vitamin D Workshop. J Steroid Biochem and Mol Biol, in press
ity of a semiquantitative food frequency questionnaire. Am J Epidemiol, Zeisel SH. 2007. Nutrigenomics and metabolomics will change clinical nutrition
122, 51–​65. and public health practice: insights from studies on dietary requirements
Wilson KM, Kasperzyk JL, Rider JR et  al. 2011. Coffee consump- for choline. Am J Clin Nutr, 86(3), 542–​548. PMCID: PMC2430757.
tion and prostate cancer risk and progression in the Health Zhao M, Li X, Xing C, and Zhou B. 2013. Association of methylenetetrahydrofo-
Professionals Follow-​up Study. J Natl Cancer Inst, 103(11), 876–​884. late reductase C677T and A1298C polymorphisms with colorectal cancer
PMCID: PMC3110172. risk: a meta-​analysis. Biomed Rep, 1(5), 781–​791. PMCID: PMC3917732.
Wilson KM, Shui IM, Mucci LA, and Giovannucci E. 2015. Calcium and phos- Zhou Y, Tian C, and Jia C. 2012. A dose-​response meta-​analysis of cof-
phorus intake and prostate cancer risk: a 24-​y follow-​up study. Am J Clin fee consumption and bladder cancer. Prev Med, 55(1), 14–​22. PMID:
Nutr, 101(1), 173–​183. PMCID: PMC4266887. 22564775.
350
 351

20 Obesity and Body Composition

NANA KEUM, MINGYANG SONG, EDWARD L. GIOVANNUCCI,


AND A. HEATHER ELIASSEN

OVERVIEW MEASURES OF BODY FAT AND BODY COMPOSITION

In 2014, an estimated 1.9 billion adults were either overweight (BMI Individuals vary in their size and shape. Because of this variation, it
25–​29.9) or obese (BMI ≥30), worldwide. The so-​called obesity epi- is not surprising that no single measure of adiposity and body com-
demic began in high-​income, English-​speaking countries in the early position is ideal in all circumstances. The so-​called gold s​tandard
1970s, but soon spread globally; worldwide, more than one-​third (38%) approaches for measuring the amount and distribution of body fat in
of all adults and 600,000 children under age 5 are overweight or obese, clinical settings involve imaging with computed tomography (CT) or
as are two-​thirds (69%) of adults in the United States. Excessive body magnetic resonance imaging (MRI). While these provide the most
fat is a major cause of type 2 diabetes, hypertension, and cardiovascu- accurate measurements, the devices are expensive to transport and
lar and liver disease, among other disorders, and has been designated operate, limiting their use in large-​scale population studies. Various
a definite cause of at least 14 cancer sites:  breast (postmenopausal), anthropometric measures have been developed that use self-​reported
colorectum, endometrium, esophagus (adenocarcinoma), gallblad- or measured information on height and weight and/​or waist and hip
der, kidney (renal cell), pancreas, gastric cardia, liver, ovary, pros- circumference to assess different aspects of body composition. These
tate (advanced tumors), multiple myeloma, thyroid, and meningioma. are widely used because of their simplicity and low cost. It is impor-
A  causal association is considered possible for leukemia and lym- tant to understand the specific significance of these measures, as well
phoma. Estimates of the proportion of cancers attributable to over- as their limitations, since they serve as the exposure variable(s) in epi-
weight and/​or obesity globally range from 6% for rectal cancer to 33% demiologic studies.
for esophageal cancer in men and from 4% for rectal cancer to 34% for
endometrial cancer in women. Mechanisms by which adipose tissue
are thought to promote tumor growth include the endocrine and met- Body Mass Index (BMI)
abolic effects of fat on sex hormones, growth factors, and inflamma- BMI is the most commonly used measure of overall body fatness.
tion, as well as local chemical or mechanical injury of gastrointestinal BMI is defined as weight in kilograms (kg) divided by height in
organs. Because of the high prevalence of excess adiposity world- meters squared (m2). The advantages of BMI are that it provides a sin-
wide and the difficulty that most individuals have in losing substantial gle measure of adiposity that is independent of height and that can be
amounts of weight and maintaining that weight loss, population-​based compared across populations (Gallagher et al., 1996). BMI is highly
approaches will require policies and community actions that help indi- correlated (r = 0.82–​0.91) with absolute fat mass, calculated from
viduals avoid excessive weight gain throughout life. percent body fat measured by densitometry, even after accounting for
extraneous variations caused by age and sex (Spiegelman et al., 1992;
Willett, 2013). Densitometry has long been considered a gold standard
INTRODUCTION for measuring total body fat.
The World Health Organization (WHO) has established criteria to
The hypothesis that excess energy intake may contribute to cancer classify individuals according to their BMI (WHO, 2015). Adults (age
risk originated from experimental studies of animals in the 1940s, in >18 years) with a BMI <18.5 are considered underweight; those with
which mice fed ad libitum developed larger and more numerous mam- BMI 18.5–​<25 are judged to have normal weight; those with BMI
mary tumors than mice on energy-​restricted diets (Sellers et al., 2007; ≥25 are considered overweight, and those with BMI ≥30 are desig-
Tannenbaum, 1940a, 1940b). As noted in Chapter 19, this finding has nated obese. The latter category can further be separated into obese
consistently been replicated in a wide variety of mammary and other class I (30–​<35), class II (35–​<40) and class III (≥40). Currently the
tumor models (Birt et al., 1992; Nair et al., 1995; Ross and Bras, 1971; same categories are used for men and women and all ethnicities. It
Weindruch and Walford, 1982); a 30% restriction in energy intake will be important to determine whether the standard WHO BMI clas-
since birth reduces mammary tumors in laboratory animals by as much sifications for adults are universally applicable to diverse populations
as 90% (Boissonneault et al., 1986). in predicting cancer risk. The criteria used to define overweight and
Epidemiologic studies, beginning in the 1970s, reported that over- obesity in children are discussed later in the chapter.
weight women were more likely to develop breast and endometrial The two major disadvantages of BMI are that it does not specify
cancer (Blitzer et  al., 1976; de Waard and Baanders-​van Halewijn, where the fat is located, nor does it account for variations in lean body
1974). Large prospective studies subsequently observed associations mass. The location of fat is important, because accumulation around
between excess body weight and increased mortality from multiple the waist (abdominal obesity, or “apple-​shape”) is more closely related
types of cancer (Calle et al., 2003; Lew and Garfinkel, 1979). During to metabolic syndrome than is excess fat distributed on the hips and
the last 15–​20 years, the literature on cancer in relation to obesity and thighs (“pear-​shape”) (Lebovitz, 2003). Moreover, visceral adipose
body composition has expanded exponentially. This chapter will focus tissue (VAT) that accumulates inside the peritoneum is a more potent
primarily on epidemiologic studies published in the last 10–​15 years cause of insulin resistance and metabolic syndrome than subcutaneous
and on advances in understanding the relationship of obesity and body adipose tissue (SAT) located beneath the skin (Donohoe et al., 2011;
composition to cancer since the publication of the corresponding Riondino et al., 2014). The extent to which studies that rely on BMI
chapter in the third edition of Cancer Epidemiology and Prevention alone may misclassify visceral obesity is illustrated by MRI images
(Ballard-​Barbash et al., 2006). that show wide variation in fat deposits around organs such as the

351
352

352 PART III:  THE CAUSES OF CANCER


liver, pancreas, and kidney in individuals with the same BMI (Thomas one study has shown that the combination of BMI and WC explains
et al., 2012). a greater proportion of variability in VAT than either alone (Janssen
The dependence of BMI on assumptions about lean body mass is et al., 2002).
also important, particularly for the elderly, males, African Americans,
and highly muscled individuals. In the elderly, variations in weight
(and thus BMI) are substantially affected by the loss of lean body Timing of Weight Change
mass (Willett, 2013). Furthermore, for a given BMI, percent body fat The age at which study participants gain or lose weight is of great
is usually lower in men than in women (Gallagher et al., 1996; Garn interest in epidemiologic studies. Excess body weight in childhood
et al., 1986; Hu, 2008a), and lower in blacks but higher in Asians than reflects exposure during a critical period of development and suscep-
in whites (Deurenberg et al., 2002; Hu, 2008a). Consequently, varia- tibility. Weight gain in adulthood reflects the cumulative increase in
tions in the risk of adiposity-​related cancers in people with the same fat mass over several decades. Weight loss during the most recent 10–​
BMI level may partly reflect variation in lean body mass by age, sex, 15 years may indicate involuntary rather than voluntary weight loss.
and race. Involuntary weight loss can be caused either by the condition being
In many large epidemiologic studies, BMI is calculated using studied (reverse causation), or wasting due to age or illness. Either can
self-​reported height and weight, rather than technician-​measured mimic a protective effect of excess weight. This issue has not been
height and weight. Due to individuals’ tendency to overreport height thoroughly studied with respect to the conditions that cause it and
and underreport weight (Willett, 2013), BMI calculated from self-​ the time period over which it occurs, complicating studies of weight
reported data may be underestimated, which could lead to an under- change and disease.
estimation of obesity prevalence. However, self-​reported BMI is
highly correlated with measured BMI (r > 0.90) with a relatively Adult Weight Gain
small mean difference (Willett, 2013). Thus, measurement error Although there is wide variability among individuals, adults gain
in BMI does not introduce substantial bias in epidemiologic mea- weight at an average of 0.5 kilograms annually (Malhotra et al., 2013).
sures of association, at least for young and middle-​aged adults, and Weight gain in adults largely represents the accumulation of abdomi-
the accuracy of BMI based on self-​reported data is considered suf- nal fat, especially in men (Ballard-​Barbash, 1994). Because the weight
ficient to investigate the relationship between adiposity and cancer gain usually reflects increases in fat rather than lean body mass, it pro-
(Willett, 2013). vides a longitudinal marker of the trajectory in fat mass during the
decades when most adult cancers develop. People can usually remem-
ber their approximate weight at the end of adolescence (i.e., between
Waist Circumference (WC) and ages 18 and 25  years). This can be compared to their weight at the
Waist-​to-​Hip Ratio (WHR) time of enrollment into an epidemiologic study. Weight change during
Waist circumference (WC) and waist-​to-​hip ratio (WHR) are other other periods of life, such as before and after menopause or during
measures commonly used in large epidemiologic studies to reflect the previous 10  years, is also potentially informative. The potential
central adiposity and intra-​abdominal visceral fat. As noted earlier, for measurement error increases with the number of measurements.
abdominal obesity and visceral fat are metabolically more detrimen- Underreporting of weight tends to be larger when recalled for periods
tal than fat distributed on the hips and thighs (Donohoe et al., 2011; earlier in life than self-​reported current weight. The extent of underre-
Lebovitz, 2003; Riondino et al., 2014). porting is greater among women than men and increases with increas-
WC is measured at the midpoint between the lowest rib margin and ing current weight (Niedhammer et al., 2000; Palta et al., 1982; Perry
the iliac crest (i.e., top of hip), roughly in line with the umbilicus. et al., 1995; Stevens et al., 1990). Despite these problems, self-​reported
WHR further accounts for frame size by dividing WC by hip cir- information on weight and weight gain is informative in epidemiologic
cumference, which is typically measured at the widest circumference research (Hu, 2008a; Willett, 2013). Subjects who underreport their
around the buttocks. The US Department of Agriculture recommends weight are likely to do so for both current and past weight, partially
maintaining WC ≤40 inch (102 cm) for men and ≤35 inch (88 cm) for offsetting the error.
women; the recommendations for WHR are ≤0.95 for men and ≤0.88
for women (US Department of Agriculture, 1990). As is the case with Adiposity in Early Life
BMI, the recommended cutpoints are currently the same for men and Emerging evidence suggests that excess body fat in early life (i.e.,
women of all ethnicities. in childhood and adolescence, ages 2–​19  years) may be especially
Both WC and WHR are more pertinent to abdominal adiposity than important for the risk of developing certain cancers later in life.
BMI (Hu, 2008a) and thus are increasingly used in epidemiologic Anthropometric classification is more complicated for children and
research. Furthermore, WC is generally preferred over WHR for sev- adolescents than for adults, however, since the categories are based
eral reasons. First, WHR requires measures of both waist and hip cir- on age-​and sex-​specific distributions, rather than absolute cutoffs,
cumference and is therefore more burdensome for study participants to account for growth (Hu, 2008b; Krebs et al., 2007). In the United
and more subject to measurement error. Second, the interpretation of States, growth charts for children and adolescents are published by
WHR is complicated, since hip circumference reflects not only fat the Centers for Disease Control and Prevention (CDC) (Centers for
mass but also gluteal muscle mass and bone structure. A high WHR Disease Control and Prevention, 2015). National data for the year
may indicate increased abdominal obesity and/​ or reduced gluteal 2000 are used as the reference, with individuals at <5th percentile of
muscle mass (Hu, 2008a). Third, when WC and WHR were compared the BMI for age and sex classified as underweight, at 5th–​84.9th per-
with total abdominal fat as measured by CT, the correlation was stron- centile as normal weight, at 85th–​94.9th percentile as overweight, and
ger for WC (r = 0.90–​0.91) than for WHR (r = 0.60–​0.73) (Clasey ≥95th percentile as obese (Centers for Disease Control and Prevention,
et al., 1999). 2015). The CDC categories for overweight and obesity in children do
However, when used alone, WC also has limitations. Like BMI, not correspond exactly to the BMI categories for adults when applied
WC cannot distinguish visceral from subcutaneous fat (Willett, to adolescents. Some adolescents who would be classified as over-
2013), and individuals with the same WC may have very different weight by the adult BMI categorization (25≤ BMI <30) would not
amounts of VAT (Thomas et  al., 2012). Second, study participants meet the CDC criterion for overweight in children (85th–​94.9th per-
tend to underreport WC, especially since the location of the waist is centile for age and sex) (Hu, 2008b).
less clearly demarcated than is the calculation of BMI. Despite these
limitations, the correlation between self-​reported WC and technician-​ Assessment of Childhood Adiposity.  Since it is usually dif-
measured WC was generally high (r = 0.7–​0.9) in validation studies ficult for adults to recall their weight during childhood, an alternative
(Hu, 2008a). There is some evidence that BMI and WC measured approach is to use a visual aid, such as a pictogram, that depicts dif-
together may provide complementary information about VAT. At least ferent body shapes. For example, Stunkard and colleagues developed
 35

Obesity and Body Composition 353


a set of nine images, ranging from underweight to obese, from which United States, using CDC Growth Charts from 2000, the prevalence of
study participants can choose the image that corresponds most closely overweight children and adolescents aged 2–​19 years increased from
to the shape they recall from childhood (Stunkard et al., 1983). In a 9.2% in 1980 to 14.9% in 2012, and the prevalence of obesity tripled,
validation study of adults ages 71–​76 years, the correlation between from 5.5% to 16.9% (Figure 20–​2) (Fryar et  al., 2014a). Globally,
recalled body shape at early ages (e.g., 5, 10, and 20), assessed by the across a similar time period, the prevalence of overweight and obesity
pictogram, and measured BMI at approximately the same ages ranged increased from 10% to 14% (Ng et al., 2014). These trends potentially
between 0.60 and 0.75 (Must et al., 1993). Analyses of body size in affect cancer rates in two ways. First, childhood and/​or adolescent adi-
childhood based on pictograms have identified associations with can- posity may have a direct impact on carcinogenesis through metabolic
cer (Baer et  al., 2010; Fagherazzi et  al., 2013; Palmer et  al., 2007; and hormonal influences during a susceptible developmental period.
Suzuki et al., 2011; Weiderpass et al., 2004; Zhang X et al., 2015a). Second, obesity in early life correlates strongly with excess adiposity
in adulthood as populations age (Cunningham et al., 2014).

EPIDEMIOLOGY OF EXCESS BODY WEIGHT


TYPES OF CANCER CAUSED BY EXCESS ADIPOSITY
The prevalence of overweight and obesity has been increasing world-
wide, beginning in high-​income, English-​speaking countries in the Obesity is associated with increased risk of multiple types of cancer.
early 1970s and subsequently in many other industrialized and/​or Several organizations, including the International Agency for Research
middle-​income countries (Ng et al., 2014). Excess body fat also affects on Cancer (IARC), the World Cancer Research Fund/​American Institute
subgroups of the population in low-​income countries where overnutri- for Cancer Research (WCRF/​AICR), and the National Institutes of
tion coexists with famine. Globally, the prevalence of BMI levels ≥25 Health (NIH), have periodically evaluated the evidence regarding spe-
increased from 28.8% to 36.9% in men and from 29.8% to 38.0% in cific cancer sites in relation to excess body fat. The WCRF/​AICR and
women from 1980 to 2013 (Ng et al., 2014). the IARC classify the evidence as “definite” that obesity is causally
The patterns of weight gain vary in severity as well as timing among related to cancers at 10 anatomic sites: female breast (postmenopausal),
countries. In the United States, the prevalence of BMI ≥25 increased colorectum, endometrium, esophagus (adenocarcinomas), gallbladder,
from 56% in 1980 to 69% in 2012, with all of the increase in adults kidney (renal cell), pancreas, gastric cardia, liver, and ovary (Table
occurring in the obese category of BMI >30 (Figure 20–​1) (Fryar 20–1). For cancer of the prostate (advanced), the WCRF/​AICR classi-
et al., 2014b). The prevalence of obesity increased by approximately fies the evidence as “definite” (WCRF/​AICR, 2014b), while the most
8% per decade from 1980 to 2000. While the overall prevalence of recent IARC report includes multiple myeloma, thyroid cancer, and
obesity in adults appears to have stabilized at about 35.5% in men in meningioma (Lauby-​Secretan et al., 2016). Obesity is considered as a
the United States in the last 10 years (Flegal et al., 2002, 2010, 2012, “possible” causal factor for leukemia and lymphoma. The associations
2016), it has continued to increase in women, reaching 40.4% in 2014 with cervix, lung, melanoma, and bladder have not been classified.
(Flegal et al., 2016).The greatest proportional increase has been in the Compared to BMI, which reflects both lean and fat mass, adult
prevalence of severe obesity (Class III). weight gain largely reflects an increase in body fat independent of
In certain geographic regions, such as among men and women in BMI, and thus may better reflect adiposity and its metabolic conse-
some Pacific Islands and women in several Middle Eastern coun- quences. Weight gain commonly occurs throughout early and middle
tries, the estimated prevalence of obesity exceeds 50% (Ng et  al., adulthood in Western societies, and this measure is associated with
2014). Except in the United States, the increase has been greater for several cancers. As noted earlier, the distribution of body fat is also
overweight than for obesity. Currently, more than two-​thirds (69%) thought to affect the metabolic consequences of adiposity. Measured
of adults in the United States are overweight or obese, compared to by WC or WHR, higher abdominal adiposity in adulthood is associ-
over one-​third (38%) globally. In 2014, an estimated 1.9 billion adults ated with several cancers.
worldwide had a BMI ≥25. Nearly one-​third of these (about 600 mil- The relationship between obesity in early life (i.e., in utero or dur-
lion adults) were obese, including about 11% of men and 15% of ing childhood and adolescence) and subsequent cancer risk, inde-
women (WHO, 2015). pendent of adult weight, is of considerable research interest. Obesity
Secular increases in adiposity also have occurred among child-
ren and adolescents in the United States and internationally. In the
18

16
40
14
Age-adjusted Prevalence (percent)

35
Prevalence (percent)

12
30
10
25
8
20
6
15
4
10
2
5
0
0
74

80

94

00

02

04

06

08

10

12
19

19

19

20

20

20

20

20

20

20
74

2
98

99

00

00

00

00

00

01

01


19

71

76

88

99

01

03

05

07

09

11
–1

–1

–2

–2

–2

–2

–2

–2

–2

19

19

19

19

20

20

20

20

20

20
71

76

88

99

01

03

05

07

09

11
19

19

19

19

20

20

20

20

20

20

Years
Years
Overweight Obese
Overweight Obese

Figure  20–​2.  Prevalence of overweight and obesity among children and


Figure 20–​1.  Age-​adjusted prevalence of overweight and obesity among adolescents aged 2–​19 years: United States (1971–​2012).
adults aged 20–​74: United States (1971–​2012). Source: Fryar et al. (2014a).
Source: Fryar et al. (2014b).
354

354 PART III:  THE CAUSES OF CANCER


in childhood has similar metabolic consequences to exposure in Childhood and Premenopausal Adiposity. In contrast,
adulthood, but these occur during a critical developmental period. women who were most overweight during childhood and adolescence,
Given the long delay between the initiation of many exposures and compared to those who were lean, have 20%–​50% lower risk of both
the diagnosis of an invasive cancer, it is plausible that obesity that premenopausal (Baer et  al., 2010; Fagherazzi et  al., 2013; Palmer
begins in childhood or adolescence may independently affect cancer et  al., 2007; Suzuki et  al., 2011; Weiderpass et  al., 2004)  and post-
risk later in life. menopausal (Baer et al., 2010; Fagherazzi et al., 2013; Kawai et al.,
The evidence linking excess body weight and body composition to 2010; Palmer et al., 2007; Suzuki et al., 2011; van den Brandt et al.,
individual cancer sites is discussed in detail in the site-​specific chapters 1997) breast cancer. This association persists with adjustment for adult
in this volume. In the following, we summarize for specific cancers the BMI, suggesting that an independent, lifelong reduction in risk occurs
associations with adult adiposity, weight gain, body fat distribution, among girls who are overweight. The inverse association is apparent
and early life adiposity. for both ER+ and ER–​breast cancer (Baer et al., 2010). Although most
studies were conducted among Caucasian women (Ahlgren et  al.,
2004; Baer et al., 2010; Fagherazzi et al., 2013; Hilakivi-​Clarke et al.,
Cancers Definitely Caused by Excess Adiposity 2001; van den Brandt et al., 1997; Weiderpass et al., 2004), this inverse
association has been confirmed in African American (Bandera et al.,
2013; Palmer et  al., 2007), Hispanic (Sangaramoorthy et  al., 2011),
Female Breast (Postmenopausal)
and Asian (Kawai et al., 2010; Suzuki et al., 2011) women.
In a systematic literature review and dose-​response meta-​analysis
Adiposity Measured After Menopause.  Numerous obser- of cohort studies published in 2008 by the WCRF/​AICR, relative risk
vational studies have established that excess body fat affects breast (RR) of breast cancer decreased with higher BMI in premenopausal
cancer risk, and that this effect varies depending on the timing of women (RR = 0.93; 95% CI: 0.88, 0.98 with a 5 kg/​m2 increase in
adiposity in relation to menopausal status. Women with a high BMI BMI). Lower BMI was characterized as a “convincing” risk-​reduc-
after menopause have increased risk of postmenopausal breast can- ing factor (WCRF/​AICR, 2008). This conclusion was upheld in a
cer, specifically ER+/​PR+ tumors (positive for estrogen receptor subsequent meta-​analysis of 16 studies conducted the WCRF/​AICR
[ER] and progesterone receptor [PR]). The opposite relationship Continuous Update Project, which reported a significant 3% decrease
exists for premenopausal women, in whom a high BMI decreases in risk per 2 kg/​m2 increase in BMI (WCRF/​AICR, 2010a).
the risk of premenopausal breast cancer (WCRF/​AICR, 2010a) (see The inverse relationship between excess body fat and risk of pre-
Chapter 45). menopausal breast cancer does not alter the recommendation that
In postmenopausal women, a 5  kg/​m2 increase in BMI increases women should maintain a healthy body weight throughout life. Excess
breast cancer incidence by about 13% (95% CI:  1.08, 1.19) adiposity in childhood increases the likelihood of obesity in adulthood,
(Table 20–​1), based on a systematic review of prospective cohort stud- as well as increasing the risk of multiple obesity-​related diseases. It
ies in which height and weight were self-​reported or measured (WCRF/​ is extremely difficult for most people to lose substantial amounts of
AICR, 2010a). This positive relationship was confirmed in a secondary weight voluntarily and then maintain this weight loss.
analysis of postmenopausal women in the Women’s Health Initiative
(WHI) clinical trials, where weight and height were measured at base-
line and annually. The WHI trial protocol also required mammography,
Weight Gain During Adulthood.  Weight gain that occurs
after age 18  years is also consistently associated with higher risk
thereby minimizing the possibility of ascertainment bias (Neuhouser
of postmenopausal breast cancer (Table 20–​2) (Ahn et  al., 2007;
et al., 2015). Postmenopausal women with a BMI higher than 35 had
Alsaker et  al., 2013; Eliassen et  al., 2006; Feigelson et  al., 2004;
58% (95% CI: 1.40, 1.79) higher risk of invasive breast cancer, com-
Kawai et  al., 2010; Keum et  al., 2015; Krishnan et  al., 2013;
pared to women with a BMI less than 25.
Lahmann et  al., 2005; Palmer et  al., 2007; Radimer et  al., 2004;

Table 20–​1. Association Between Adiposity and Specific Cancers, RR (95% CI) per 5 kg/​m2 Increase in BMI

Cancer Type Association RR (95% CI) Reference

Breast (postmenopausal) Definite 1.13 (1.08, 1.19) (WCRF/​AICR, 2010a)


Colorectum Definite 1.10 (1.05, 1.16) (WCRF/​AICR, 2011)
Endometrium Definite 1.50 (1.42, 1.59) (WCRF/​AICR, 2013)
Esophagus (adenocarcinoma) Definite 1.48 (1.35, 1.62) (WCRF/​AICR, 2016b)
Gallbladder Definite 1.25 (1.15, 1.37) (WCRF/​AICR, 2015a)
Kidney (renal cell) Definite 1.30 (1.16, 1.46) (WCRF/​AICR, 2015b)
Pancreas Definite 1.10 (1.07, 1.14) (WCRF/​AICR, 2012)
Gastric cardia Definite 1.23 (1.07, 1.40) (WCRF/​AICR, 2016a)
Liver Definite 1.30 (1.16, 1.46) (WCRF/​AICR, 2015c)
Ovary Definite 1.06 (1.03, 1.11) (WCRF/​AICR, 2014c)
Prostate (advanced) Definite* 1.08 (1.04, 1.12) (WCRF/​AICR, 2014b)
Multiple myeloma Definite* 1.12 (1.07, 1.18) (Wallin and Larsson, 2011)
Thyroid Definite* 1.06 (1.02, 1.10) (Kitahara et al., 2016)
Meningioma Definite* Obese =1.51 (1.25, 1.82)^ (Niedermaier et al., 2015)
Leukemia Possible 1.13 (1.07, 1.19) (Larsson and Wolk, 2008)
Lymphoma Possible HL: Obese = 1.41 (1.14, 1.75)^ (Larsson and Wolk, 2011)
NHL: 1.07 (1.04, 1.10)
Cervix Inconsistent (Lacey et al., 2003)
Lung Inconsistent (WCRF/​AICR, 2007)
Melanoma Inconsistent (Sergentanis et al., 2013)
Bladder No association (WCRF/​AICR, 2007)

* Designations of “definite*” correspond to cancers determined to be causally related to obesity by either WCRF/​AICR or IARC, but not by both organizations.
^ Association with BMI was non-​linear, with no increased risk observed among overweight
 35

Obesity and Body Composition 355


Table 20–​2. Summary of Meta-​analysis Findings on the Association of Cancer Risk with Weight Gain (per 5 kg) from Young Adulthood
(Age 18–​25 Years) to Study Enrollment

Cancer Type RR (95% CI) Reference

Breast (postmenopausal) Overall: 1.08 (1.06, 1.10) (Keum et al., 2015)


MHT users: 1.01 (0.99, 1.02)
MHT non-​users: 1.11 (1.08, 1.13)
Endometrium Overall: 1.16 (1.12, 1.20) (Aune et al., 2015b; Keum et al., 2015)
MHT users: 1.09 (1.02, 1.16)
MHT non-​users: 1.39 (1.29, 1.49)
Ovary 1.02 (0.96, 1.09) (Aune et al., 2015a)
Colorectum Overall: 1.04 (0.99, 1.09) (Chen Q et al., 2014)
Women: 1.01 (0.96, 1.08)
Men: 1.09 (1.01, 1.18)
Pancreas 1.40 (1.13, 1.72)* (Genkinger et al., 2011)
Prostate Overall: 0.98 (0.94, 1.02) (Moller et al., 2013)
Localized: 0.96 (0.92, 1.00)
Advanced: 1.04 (0.99, 1.09)

Abbreviation: MHT = menopausal hormone therapy


*The estimate was calculated by comparing BMI gain of >10 kg/​m2 to BMI change ≤2 kg/​m2 in a pooled analysis of 14 cohort studies.

Sweeney et al., 2004; van den Brandt et al., 1997; White et al., 2012). Adult Weight Gain. Weight gain between early and middle
In a recent meta-​analysis, each 5 kg of adult weight gain was associ- adulthood (usually somewhere between ages 30 and 65 years) is con-
ated with an 11% (95% CI: 1.06, 1.13) increase in risk of postmeno- sistently associated with higher risk of colorectal cancer in observa-
pausal breast cancer (Keum et al., 2015). The association with adult tional studies (Table 20–​2) (Aleksandrova et al., 2013; Bassett et al.,
weight gain is stronger for tumors that are ER+/​PR+ than for ER–​/​ 2010; Hughes et al., 2011; Laake et al., 2010; Oxentenko et al., 2010;
PR–​tumors (Alsaker et al., 2013; Kawai et al., 2010; Tamimi et al., Renehan et al., 2012; Song et al., 2015b; Steins Bisschop et al., 2014;
2012; Vrieling et al., 2010). Weight gain in later adulthood, specifi- Thygesen et al., 2008). Like the association with BMI, this relation-
cally after menopause, is also associated with postmenopausal breast ship is stronger in men than in women (Bassett et al., 2010; Hughes
cancer risk (Eliassen et al., 2006). et  al., 2011; Laake et  al., 2010; Renehan et  al., 2012; Song et  al.,
For premenopausal breast cancer, although a high BMI at age 18 2015b) and for colon than for rectum cancer (Aleksandrova et  al.,
is inversely associated with risk, it is not clear how additional weight 2013; Hughes et al., 2011; Laake et al., 2010; Renehan et al., 2012;
gain before menopause affects this risk. An increased risk has been Steins Bisschop et al., 2014). It is especially stronger for tumors in the
reported for short-​term weight gain (i.e., over 4 years) during premen- distal rather than in the proximal colon (Bassett et al., 2010; Laake
opausal adulthood, particularly for ER+/​PR–​and ER–​/P ​ R–​tumors et al., 2010; Oxentenko et al., 2010). Weight gain at older ages (after
(Rosner et al., 2015). Other studies have not found an association with mid-​life) has not been associated with colorectal cancer risk, although
weight gain or loss before menopause (Lahmann et al., 2005; Michels the number of studies is limited (Karahalios et al., 2015; Song et al.,
et al., 2012; Palmer et al., 2007). 2015b).

Effect Modification by  Menopausal Hormone Body Fat Distribution.  While weight and weight gain are posi-
Therapy.  Numerous observational studies have suggested that the tively associated with colorectal cancer, the association with abdomi-
association between obesity and increased breast cancer risk among nal adiposity is particularly strong. Risk increases by 3% (95%
postmenopausal women may be limited to those who have never taken CI: 1.02, 1.04) per 2.5 cm increase in WC. This association persists in
menopausal hormone therapy (MHT) (reviewed in Kabat et al., 2015; analyses adjusted for BMI (WCRF/​AICR, 2011). Similarly, WHR is
Munsell et al., 2014; WCRF/​AICR, 2008). This finding was not con- positively associated with risk of colorectal cancer. Risk increases by
firmed by the WHI clinical trials, however (Neuhouser et al., 2015). 17% (95% CI: 1.09, 1.25) per 0.1 increase in WHR. Furthermore, the
association with adult weight gain is particularly strong in women who
Body Fat Distribution.  It is unclear whether the specific distri- also increase their WC (Aleksandrova et al., 2013; Song et al., 2015c),
bution of body fat affects breast cancer risk, beyond its contribution to supporting an adverse effect of abdominal adiposity on colorectal can-
total fat mass. In a meta-​analysis of seven cohorts of postmenopausal cer. The stronger association with adiposity for men than for women
women, higher WC was associated with an increased risk of breast may relate to gender differences in fat distribution, given that men are
cancer (RR = 1.07; 95% CI: 1.04, 1.10 per 8 cm increase) in analyses more likely to accumulate intraabdominal fat than women (Despres,
not adjusted for BMI (WCRF/​AICR, 2010a). However, in three stud- 2006; Geer and Shen, 2009).
ies, adjustment for BMI attenuated this association (RR = 1.04; 95%
CI: 1.00, 1.06). The contribution of fat mass to estrogen synthesis in Early Life Adiposity.  While adult obesity is an established risk
postmenopausal women is not thought to depend on its location in the factor for colorectal cancer, few studies have examined the association
body, unlike the greater effect of abdominal obesity on insulin and between body size in early life and colorectal cancer risk in adulthood.
other metabolic peptide hormones. In the most recent, and largest, analysis of two prospective cohorts,
body fatness in childhood and adolescence was associated with
Colorectum increased risk of colorectal cancer in women, but not men (Zhang X
et al., 2015a). Specifically, compared to the smallest body size, as mea-
Adult Adiposity. In a meta-​analysis published by the WCRF/​ sured by somatotype pictogram (Stunkard et al., 1983), women who
AICR, colorectal cancer risk increased by 10% (95% CI: 1.05, 1.16) were in the top category of body fatness in childhood had a 28% (95%
for every 5 kg/​m2 increase in adult BMI, with slightly stronger associa- CI: 1.04, 1.58) higher risk of developing colorectal cancer. The asso-
tions observed for men than for women (Table 20–​1) (WCRF/​AICR, ciation between childhood body size and colorectal cancer was inde-
2011). The association was also stronger for colon cancer (per 5 kg/​m2 pendent of adult BMI. Excess body fat in childhood is also positively
increase RR  =  1.16; 95% CI:  1.10, 1.22) than for rectal cancer associated with colorectal adenomas among women, independent of
(RR = 1.10; 95% CI: 1.05, 1.16). adult BMI (Nimptsch et al., 2011a).
356

356 PART III:  THE CAUSES OF CANCER

Endometrium increase (95% CI:  1.25, 1.35) in the risk of renal cell carcinoma
(WCRF/​AICR, 2015b).
Adult Adiposity.  The association of excess adiposity with endo-
metrial cancer is among the strongest and most consistent relationships Adult Weight Gain. Adult weight gain has been associated
between body fat and any cancer. Each 5 kg/​m2 increase in adult BMI with a higher risk of renal cell cancer in several studies (Adams et al.,
results in a 50% (95% CI: 1.42, 1.59) increase in the incidence of uter- 2008; Chow et al., 2000; Luo et al., 2007; van Dijk et al., 2004), espe-
ine cancer (Table 20–​1) (WCRF/​AICR, 2013). Mendelian randomiza- cially among individuals with a low BMI in adolescence (Adams
tion analyses have reported positive associations between genetic risk et al., 2008).
scores for BMI and the risk of endometrial cancer (Nead et al., 2015;
Prescott et al., 2015). Body Fat Distribution. WC is associated with higher risk of
renal cell carcinoma independent of BMI. Each 10 cm increase in WC
Adult Weight Gain.  Endometrial cancer is also strongly asso- is associated with an 11% increase in the risk of renal cell carcinoma
ciated with adult weight gain (Canchola et  al., 2010b; Chang et  al., (95% CI:  1.05, 1.19) in analyses adjusted for BMI (WCRF/​AICR,
2007; Dougan et al., 2015; Friedenreich et al., 2007; Han et al., 2014; 2015b). WHR also is positively associated with risk (per 0.1 increase,
Jonsson et al., 2003; Nagle et al., 2013; Park et al., 2010). This asso- RR  =  1.26; 95% CI:  1.18, 1.36), with no attenuation observed after
ciation is stronger in women who do not use MHT than in those who adjustment for BMI.
do (Table 20–​2) (Keum et  al., 2015)  and is stronger for endometri-
oid cancer than for other histologic subtypes (Nagle et  al., 2013). Pancreas
Corroborating the dominant effect of adiposity in later life, most stud-
ies have shown that the positive association of BMI at age 18–​20 years Adult Adiposity.  Higher BMI is positively associated with pan-
with endometrial cancer risk is substantially weakened in analyses that creatic cancer risk (per 5 kg/​m2 increase, RR = 1.10; 95% CI: 1.07,
adjust for current BMI (Canchola et  al., 2010b; Chang et  al., 2007; 1.14). This association is similar for men and women (WCRF/​
Dal Maso et al., 2011; Dougan et al., 2015; Levi et al., 1992; Swanson AICR, 2012).
et al., 1993; Xu et al., 2006; Yang et al., 2012).
Adult Weight Gain.  Although overweight and obesity throughout
Body Fat Distribution.  Abdominal adiposity, as measured by adulthood are associated with both increased risk of pancreatic cancer
WC, has been positively associated with endometrial cancer risk (per and younger age at diagnosis (Genkinger et  al., 2011; Stolzenberg-​
5 cm increase, RR = 1.13; 95% CI: 1.08, 1.18), as has WHR (per 0.1 Solomon et al., 2013), the influence of weight change on risk remains
increase, RR = 1.21; 95% CI: 1.13, 1.29) (WCRF/​AICR, 2013). The unclear (Genkinger et al., 2011; Isaksson et al., 2002; Patel et al., 2005;
association with WC was not attenuated by adjustment for BMI, sug- Stolzenberg-​Solomon et al., 2013). In a pooled analysis of 14 cohort
gesting that the distribution of the fat mass may independently affect studies, study participants who gained ≥10  kg/​m2 between ages 18–​
the risk of endometrial cancer. 21 years and age at enrollment had increased risk of pancreatic cancer
compared to those with BMI change ≤2 kg/​m2, as did those who lost
Esophagus (Adenocarcinoma) >2 kg/​m2 during this period (Genkinger et al., 2011). These associations
were adjusted for history of diabetes, and persisted even after excluding
Adult Adiposity. Like the association between adiposity and cases diagnosed within the first 5 years of follow-​up. Further studies are
endometrial cancer, the association with esophageal adenocarcinoma needed to better understand these associations.
is particularly strong. For every 5 kg/​m2 increase in BMI, the risk of
esophageal adenocarcinoma increases by 48% (95% CI: 1.35, 1.62) Body Fat Distribution. In the WCRF/​AICR report, WC was
(WCRF/​AICR, 2016b). This association has been confirmed by a positively associated with pancreatic cancer risk. Risk increased by
Mendelian randomization analysis of a genetic risk score for BMI in 11% (95% CI: 1.05, 1.18) per 10 cm increase in WC (WCRF/​AICR,
relation to the risk of esophageal adenocarcinoma (Thrift et al., 2014). 2012). A pooled analysis of cohort studies observed a similar, although
not statistically significant, increase in risk (RR = 1.16; 95% CI: 0.92,
Body Fat Distribution.  Abdominal adiposity is more strongly 1.46), but this was attenuated with adjustment for BMI (RR = 1.04;
associated with esophageal adenocarcinoma than fat elsewhere on the 95% CI:  0.73, 1.47) (Genkinger et  al., 2011). WHR was positively
body. Many studies have reported strong positive associations between associated with risk in both the WCRF/​AICR meta-​analysis (per 0.1
WC and risk. A recent study of esophageal adenocarcinoma in Europe increase, RR = 1.19; 95% CI: 1.09, 1.31) and in the pooled analysis
found that the association with WC remained strong after adjustment of cohort studies (RR  =  1.35; 95% CI:  1.03, 1.78). Adjustment for
for BMI, whereas the association with BMI was largely attenuated BMI did not attenuate this association in the pooled analysis. Further
by adjustment for WC (Steffen et al., 2015). The higher incidence of replication of these findings is needed to determine whether abdominal
esophageal cancer incidence in men than women may partly reflect the adiposity is a risk factor for pancreatic cancer, independent of general
greater prevalence of abdominal adiposity in men (Lagergren et  al., adiposity.
1999; Singh et al., 2013).
Gastric Cardia
Gallbladder The WCRF/​AICR summary meta-​analysis found a significant positive
association between increasing BMI and risk of gastric cardia cancer,
Adult Adiposity. A meta-​analysis of eight studies, included in but not non-​cardia gastric cancer. The relative risk increased by 23%
the WCRF/​AICR report, showed a significant positive association (95% CI: 1.07, 1.40) per 5 kg/​m2 increase (WCRF/​AICR, 2016a). The
between BMI and gallbladder cancer (WCRF/​AICR, 2015c). Risk association appeared to be non-​linear, with a greater increase in risk
increased by 25% (95% CI: 1.15, 1.37) per 5 kg/​m2 increase in BMI. observed at higher levels of BMI.
The association was similar in men and women. There are limited data
on the association of weight gain and body fat distribution with gall- Liver
bladder cancer. A meta-​analysis of 12 studies shows a positive association between
BMI and risk of hepatocellular carcinoma. The incidence rate
Kidney (Renal Cell) increased by 30% (95% CI: 1.16, 1.46) per 5 kg/​m2 increase in BMI in
men and women (WCRF/​AICR, 2015a). The meta-​analysis provided
Adult Adiposity.  A meta-​analysis of 23 studies found that each evidence of non-​linearity, with risk increasing more steeply at higher
5  kg/​m2 increase in BMI in adulthood was associated with a 30% BMI levels.
 357

Obesity and Body Composition 357


Ovary non-​Hodgkin lymphomas, significant positive associations with BMI
The combined evidence from 25 studies shows a modest positive were observed in a meta-​analysis (per 5kg/​m2 increase, RR = 1.14;
association between BMI and ovarian cancer risk, with a 6% (95% 95% CI: 1.04, 1.26) (Larsson and Wolk, 2011). More recent analyses of
CI: 1.02, 1.11) increase in risk per 5 kg/​m2 increase in BMI (WCRF/​ the UK Million Women Study reported associations between BMI and
AICR, 2014c). Whether risk is associated with body fat distribution is both Hodgkin lymphoma and non-​Hodgkin lymphomas (Murphy et al.,
not clear (WCRF/​AICR, 2014c). Weight gain in adulthood has been 2013). Replication is needed to confirm or refute these associations.
associated with risk of ovarian cancer in some (Canchola et al., 2010a;
Ma et al., 2013), but not most (Aune et al., 2015a; Fairfield et al., 2002;
Jonsson et al., 2003; Kotsopoulos et al., 2010; Lahmann et al., 2010; Cancers Inversely Related to Excess Adiposity
Schouten et al., 2003) prospective studies. Inverse associations have been reported between increasing adiposity
and cancers of the lung (sese Chapter 28), oral cavity (see Chapter 29),
Prostate (Advanced) and esophagus (squamous cell) (see Chapter 30). As yet, no biologi-
No significant association has been observed between BMI and the cally plausible mechanisms have been proposed to account for these
overall incidence of prostate cancer (per 5 kg/​m2 increase, RR = 1.00; associations, which could plausibly reflect confounding or reverse
95% CI: 0.97, 1.03). However, it may be that excess body weight causation (WCRF/​AICR, 2007). Cancer at all of these sites is strongly
increases the risk of advanced prostate cancer, but decreases the risk associated with tobacco use, and tumors could potentially interfere
of non-​advanced tumors. The incidence of advanced prostate cancer with eating and/​or breathing. Indeed, a genetic risk score for obesity in
increases by 8% (95% CI: 1.04, 1.12) per 5 kg/​m2 increase in BMI, as the GAME-​ON consortium was positively associated with lung can-
does the incidence of high-​grade prostate cancer (RR = 1.08; 95% CI: cer, suggesting that the observed inverse associations with BMI may
1.01, 1.15) (WCRF/​AICR, 2014b). Weight gain is also associated with be non-​causal (Gao et al., 2016).
increased risk of aggressive or fatal disease in some studies (Bassett
et  al., 2012; Keum et  al., 2015; Wright et  al., 2007), but not others
(Chamberlain et  al., 2011; Littman et  al., 2007; Moller et  al., 2013; CANCERS ASSOCIATED WITH HEIGHT
Rodriguez et al., 2007). Both WC and WHR are positively associated
with advanced prostate cancer (e.g., per 10 cm increase in WC, RR = Numerous studies have found that adult height is positively associated
1.12; 95% CI: 1.04, 1.21), but not with total prostate cancer risk (RR = with cancer risk. The UK Million Women Study represents the larg-
1.01; 95% CI: 0.90, 1.12) (WCRF/​AICR, 2014b). est prospective study on height and cancer. The analyses of height and
In contrast, an inverse association between excess body fat and cancer included 1,297,124 women and 97,376 incident cancer cases
non-​advanced prostate cancers has been observed in multiple stud- (Green et al., 2011). Total cancer risk increased by 16% for each 10 cm
ies. In the WCRF/​AICR meta-​analysis, the risk of non-​advanced dis- increase in adult height. A positive association of height with total can-
ease decreased by 5% (95% CI:  0.92, 0.98) per 5  kg/​m2 increase in cer incidence or mortality was observed for both men and women in a
BMI (WCRF/​AICR, 2014a). Subsequent studies have confirmed this meta-​analysis that included this study and 10 other prospective studies
inverse relationship with adult BMI (Moller et  al., 2016)  and adult (Green et al., 2011). The Million Women Study also examined the rela-
weight gain (Keum et al., 2015). tionship separately for 17 cancer sites; significant positive associations
were observed with 10 anatomic sites (colon, rectum, melanoma, breast,
Multiple Myeloma endometrium, ovary, kidney, central nervous system, non-​ Hodgkin
A meta-​analysis reported a significant positive association between lymphoma, leukemia), accounting for multiple testing. The increase
BMI and multiple myeloma, with a 12% (95% CI: 1.08, 1.16) increase in risk per 10  cm increase in adult height ranged from 14% for rec-
in risk per 5 kg/​m2 increase in BMI (Wallin and Larsson, 2011), and tal cancer to 32% for malignant melanoma. Subsequent meta-​analyses
the recent IARC report considers the evidence “sufficient” (Lauby-​ reported positive associations with cancers of the pancreas (Aune et al.,
Secretan et al., 2016). 2012) and thyroid (Jing et al., 2015). Height has also been associated
with advanced or high-​grade prostate cancer (Giovannucci et al., 1997;
Thyroid Zuccolo et al., 2008). Associations with adult height have generally not
In a large pooled analysis of 22 prospective studies of thyroid cancer, been seen with smoking-​related cancers (Green et al., 2011).
BMI was associated with an increased risk (per 5 kg/​m2, RR = 1.06; The presence of similar associations with height across different
95% CI: 1.02, 1.10), which was marginally stronger in men (RR = 1.17; cancer sites suggests that a common underlying mechanism might be
95% CI: 1.06, 1.28) than in women (RR = 1.04; 95% CI: 1.00, 1.09) involved. Adult height per se is unlikely to be the causative exposure.
(Kitahara et al., 2016). The recent IARC report considers the evidence It is more plausible that height reflects some underlying exposure that
“sufficient” for a causal association between adiposity and thyroid may increase both height and cancer risk. Adult height is determined
cancer (Lauby-​Secretan et al., 2016). by a complex interplay between inherited genetic traits and early-​life
environmental factors, including nutrition and/​or illness (Giovannucci
Meningioma et al., 2004). Insulin-​like growth factor-​1 (IGF-​1) is known to influ-
The IARC also considers the evidence “sufficient” for a causal associa- ence adult height (Albanes, 1998; Giovannucci et al., 2004) and has
tion between adiposity and meningioma (Lauby-​Secretan et al., 2016), mitogenic and anti-​apoptotic hormonal effects (see later discussion in
with a meta-​analysis of six cohort studies reporting a significantly this chapter for more detail) (Aaronson, 1991). IGF-​1 is influenced
increased risk for BMI ≥30 (RR  =  1.51; 95% CI:  1.25, 1.82). The by both genetic and nutritional factors. Thus, adult height has been
association with BMI was significantly non-​linear, with no significant hypothesized to reflect circulating exposure to IGF-​1 in early life,
association observed with BMI 25–​<30 (Niedermaier et al., 2015). which modifies cancer risk later in life.
Although early-​life IGF-​1 level is more strongly associated with
leg length than other components of height, evidence supporting the
Cancers Possibly Caused by Excess Adiposity superiority of leg length compared to total height in predicting cancer
Accumulating evidence suggests possibly causal associations between risk has not been consistent (Gunnell et al., 2001; Lawlor et al., 2003;
excess adiposity and leukemia (see Chapter 38) and lymphomas (see Mellemkjaer et al., 2012). Another potential mechanism involves the
Chapters 39, 40). In a meta-​analysis of nine cohort studies, a positive greater number of cells in taller individuals, increasing the opportunity
association was reported between BMI and leukemia, with a 13% (95% to acquire genetic and epigenetic alterations that drive cancer develop-
CI: 1.07, 1.19) increase in risk per 5 kg/​m2 increase in BMI (Larsson ment (Albanes and Winick, 1988; Trichopoulos and Lipworth, 1995).
and Wolk, 2008). For Hodgkin lymphoma, increased risk was observed It is worth noting that lower adult height was associated with
for BMI ≥30 (RR = 1.41; 95% CI: 1.14, 1.75), but the association was increased risk of non-​cardia gastric cancer in the NIH-​AARP Diet and
non-​linear, with no apparent increase in risk among the overweight. For Health Study (Camargo et al., 2015) and with head and neck cancer
358

358 PART III:  THE CAUSES OF CANCER


in a pooled analysis (Leoncini et  al., 2014). Biological mechanisms by CT scan, even though BMI or traditional circumference measures
underlying this inverse association are not clear, although short height may not capture this relationship (Kim et  al., 2012). The relation
may be a marker of poor health status (e.g. infectious illness, poor between smoking and visceral accumulation of adipose tissue, pos-
nutritional status) in early life. sibly mediated by an increase in cortisol, may account for the dose-​
dependent association between smoking and insulin resistance and
hyperinsulinemia (Cena et al., 2011).
EXPOSURE PARAMETERS THAT INFLUENCE RISK Though the biologic underpinnings of these complex relationships
are incompletely understood, it is clear that adiposity in smokers dif-
fers from that in never-​smokers. The effects of smoking may involve
Menopausal Hormone Therapy Use a mixture of traditional confounding, effect modification, and reverse
As mentioned, the associations between obesity and both breast and causation. Arguably, it may be unjustifiable to examine associations
endometrial cancers in postmenopausal women vary in some studies between measures such as BMI or WC in relation to cancer in mixed
depending on MHT use (reviewed in Munsell et  al., 2014; WCRF/​ study populations of smokers and non-​smokers. Analyses that stratify
AICR, 2008; and Kabat et  al., 2015). Adipose tissue is the primary on smoking status are critical for obtaining valid information. This
source of estrogens after menopause in women who are not taking is particularly true for cancer sites that are most strongly associated
MHT. For women who use exogenous hormones, however, MHT with smoking, such as lung cancer, where either smoking or smoking-​
could obscure the contribution of adipose tissue to circulating hor- induced lung disease may cause weight loss. It is also true for other
mone levels (Key et al., 2003). Thus, the effect of obesity on breast and cancers for which the metabolic consequences of smoking and excess
endometrial cancers would be easier to detect in women who do not adiposity may overlap. Renehan and colleagues have noted that stron-
use MHT. If all of the studies supported this finding, it would strongly ger associations are observed between adiposity and various cancers
implicate estrogen production by fat tissue as the principal mediator of in non-​smokers (preferably never smokers) than in smokers (Renehan
risk for endometrial and postmenopausal breast cancer. As mentioned, et al., 2008).
however, the WHI clinical trials report did not support the hypothesis
of effect modification by MHT (Neuhouser et al., 2015).
EFFECT OF WEIGHT LOSS

Smoking Although excess body weight and weight gain are considered
causally related to at least seven types of cancer, the evidence
Tobacco use, especially cigarette smoking, is a major modifier of the regarding whether weight loss reverses these risks is less certain.
association between adiposity and cancer risk. This influence is thought Observational studies of weight loss and cancer are limited by the
to involve multiple mechanisms that relate directly or indirectly to relatively small number of people who have voluntarily lost sub-
smoking (Song and Giovannucci, 2016). First, smoking-​related can- stantial amounts of weight and have maintained this weight loss.
cers in general (particularly lung cancer and upper aerodigestive can- It is difficult to distinguish voluntary from involuntary weight loss
cers) may be less likely to be obesity-​related cancers. Smokers have in observational studies (Birks et  al., 2012). Randomized trials of
proportionally more cancers of the lung and upper aerodigestive tract dietary or exercise interventions with durations of 4–​12  months
than non-​smokers, and thus have proportionately fewer obesity-​related have demonstrated favorable changes in metabolic biomarkers, but
cancers. Smoking is also a major cause of many illnesses (e.g., chronic have not yet confirmed any reduction in cancer risk. Surgical inter-
obstructive pulmonary disease) that can cause weight loss or limit ventions effectively reduce weight, improve metabolic biomarkers,
weight gain through pathophysiologic mechanisms. In epidemiologic and decrease the risk of type 2 diabetes and cardiovascular disease,
studies, this phenomenon would enrich the categories of lower weight but have not yet provided a sufficient sample size to evaluate site-​
individuals with unhealthy individuals who suffer more severe conse- specific cancer risk. Thus, the data currently available to evaluate
quences of long-​term smoking. The cancer risk for these individuals the extent to which weight loss affects the risk of specific cancer
with low body weight may be different from that of normal healthy sites remain sparse and inconclusive.
lean individuals. Controlling for smoking may not fully eradicate
confounding because smokers who are lean may be unhealthier than
smokers who maintain a higher body mass. Such a scenario would Intentional Weight Loss
result in intractable residual confounding.
More broadly, smoking has direct consequences for body weight. The results from observational studies of intentional weight loss and
These relationships may be quite complex. Over the short term, the cancer have been inconsistent. In the Iowa Women’s Health Study,
nicotine from smoking increases energy expenditure and may reduce researchers compared cancer risk among women who reported inten-
appetite. This relationship may explain why smokers tend to have a tional weight loss of >20 pounds to that in women who reported never
lower BMI than non-​smokers, though underlying illness also may achieving this level of voluntary weight loss. The incidence rate of
contribute. Furthermore, smoking cessation is frequently followed by all cancer sites combined was 11% lower (HR = 0.89; 95% CI: 0.79,
weight gain. For example, recent quitters in two large cohorts of men 1.00) among the women who reported this level of weight loss; breast
and women had a 5.17 pound weight gain over a 4-​year period, con- cancer risk was 19% lower (HR = 0.81; 95% CI: 0.66, 1.00); colon
trolling for other factors (Mozaffarian et al., 2011). On the other hand, cancer 9% lower (HR = 0.91; 95% CI: 0.66, 1.24); and endometrial
among smokers, those who smoke heavily tend to have a higher BMI cancer 4% lower (HR = 0.96; 95% CI: 0.61, 1.52). The risk was lower
than lighter smokers. This association is surprising given the effect of by 14% for all obesity-​related cancers combined (HR  =  0.86; 95%
smoking on energy expenditure and appetite suppression. A potential CI: 0.74, 1.01) (Parker and Folsom, 2003). In contrast, two other stud-
explanation for this association is that heavy smokers have multiple ies reported no significant association between intentional weight loss
unhealthy behaviors, including poor diet, heavy alcohol intake, and and risk of all-​cancer mortality (Williamson et al., 1999) except among
lower physical activity (Chiolero et  al., 2008). The combination of women with preexisting obesity-​related health conditions (Williamson
these may offset the mechanisms by which smoking usually limits et al., 1995).
weight gain. Other observational studies that have examined the relationship
A final consideration is that smoking is associated with central fat between weight loss and colon cancer also characterized weight loss
accumulation, so that smokers are at increased risk of metabolic syn- by a single category, due to the limited number of cases. A statisti-
drome and insulin resistance. Smoking appears to increase the WHR, cally significantly lower risk of colon cancer associated with weight
independently of BMI; the phenotype of a high WHR with relatively loss was reported in only one (Rapp et  al., 2008)  of nine studies
low BMI is more common in smokers than in non-​smokers (Chiolero (Aleksandrova et  al., 2013; Bassett et  al., 2010; Campbell et  al.,
et al., 2008). In fact, smoking may be associated with increased VAT 2007; Han et  al., 2014; Hughes et  al., 2011; Laake et  al., 2010;
 359

Obesity and Body Composition 359


Oxentenko et al., 2010; Renehan et al., 2012; Steins Bisschop et al., As the use of bariatric surgery continues to increase (Santry et al.,
2014). The study in which lower risk was observed (Rapp et  al., 2005) and larger numbers of participants can be followed for longer, it
2008) found that weight loss of more than 0.1 kg/​m2 BMI per year will become feasible to assess the long-​term effects of surgical inter-
over 5–​9  years was associated with 50% lower risk of colon can- vention on specific cancer sites.
cer in men after 9 years of follow-​up. No association was observed
in women, however. Recently, a detailed dose–​response analysis
of weight change in relation to risk of colorectal cancer used data Changes in Biomarkers with Weight Loss
from two large prospective studies that compared weight in young In randomized trials of dietary or exercise interventions with duration
adulthood to current weight reported throughout 24–​34  years of of 4–​12 months, weight loss has produced favorable changes in a wide
follow-​up (Song et  al., 2015b). Weight loss during adulthood was range of metabolic biomarkers, as mentioned earlier. Changes have
associated with a suggestively lower risk of colorectal cancer in been observed in the circulating levels of steroid hormones, inflam-
men, but not women. Compared to men who maintained a stable matory markers, and metabolic hormones, mostly in postmenopausal
weight, those who lost 4–​8  kg and ≥8.0  kg between age 21  years women (Byers and Sedjo, 2011). For example, the levels of estradiol,
and baseline enrollment (median age = 52 years) had 27% and 39% estrone, and testosterone decreased by 10%–​20% among individuals
lower risk of colorectal cancer, respectively. who lost >2% of body fat (Campbell et al., 2012; Fabian et al., 2013;
For postmenopausal breast cancer, weight loss has been associ- Kaaks et al., 2003; McTiernan et al., 2004a, 2004b; van Gemert et al.,
ated with a decreased risk in some (Eliassen et  al., 2006; Harvie 2015). In contrast, sex hormone binding globulin levels were increased
et al., 2005) but not all (Teras et al., 2011) prospective studies. In the after weight loss. These relationships appeared to be dose-​dependent,
Nurses’ Health Study, weight loss of ≥10 kg since menopause was with stronger associations with sex hormones with greater weight
associated with a suggestive 23% decrease in risk (95% CI:  0.56, loss. Reductions in inflammatory biomarkers have also been observed
1.08), although this was not statistically significant (Eliassen et al., after weight loss, with more consistent evidence for C-​reactive protein
2006). Weight loss of 10 kg or more among women who had never (CRP) and interleukin-​6 (IL-​6) (Esposito et  al., 2003; Fabian et  al.,
used MHT was significantly associated with lower risk (HR = 0.63; 2013; Harvie et al., 2011; Imayama et al., 2012; Madsen et al., 2008;
95% CI: 0.38, 1.05; p-​trend for weight loss = 0.04), as was sustained Ryan and Nicklas, 2004; Tchernof et al., 2002) than tumor necrosis
weight loss of ≥10 kg of weight after menopause (HR = 0.43; 95% factor-​α (TNF-​α) (Fabian et al., 2013; Ryan and Nicklas, 2004). An
CI:  0.21, 0.86; p-​trend for weight loss  =  0.01). Weight loss was increase in adiponectin concentrations and decrease in leptin levels
inversely associated with ER+/​PR+ tumors, whereas no associations have been observed with weight loss (Abbenhardt et al., 2013; Esposito
seen for ER–​/​PR–​ tumors. et al., 2003; Fabian et al., 2013; Madsen et al., 2008); some studies
suggest that >10% weight loss is needed to increase adiponectin levels
significantly (Fabian et  al., 2013; Madsen et  al., 2008). Weight loss
Bariatric Surgery as low as 2.5%–​5% of body weight has been shown to lower insulin
Bariatric surgery is the most effective therapeutic approach for patients levels and improve insulin sensitivity (Fabian et al., 2013; Kaaks et al.,
with morbid obesity. Bariatric surgery has been shown to improve 2003; Lofgren et  al., 2005; Mason et  al., 2011; Ryan and Nicklas,
metabolic biomarkers and reduce the risk of type 2 diabetes and car- 2004). The evidence for IGF-​1 is inconsistent (Kaaks et  al., 2003;
diovascular disease (Adams et al., 2012; Sjostrom et al., 2004, 2012). Mason et al., 2013).
Recent evidence from four observational studies (Adams et al., 2009;
Christou et al., 2004, 2008; McCawley et al., 2009) and one interven-
tion trial (Sjostrom et al., 2009) suggests that gastric bypass surgery MECHANISMS OF CARCINOGENESIS
may decrease risk of all cancers combined. At this point, the studies
of surgical weight loss have insufficient follow-​up to assess effects Various mechanisms have been proposed by which excess body fat
on individual cancer sites. In the largest observational study to date, could affect cancer risk (Figure 20–​3). These can be classified broadly
6,596 morbidly obese patients who underwent bariatric surgery had a as hormonal (endocrine) effects from both sex steroid hormones and
significantly lower total cancer incidence than their counterparts who peptide metabolic hormones and the effects of chronic inflammation.
received conventional care (HR = 0.76; 95% CI: 0.65, 0.89) (Adams Systemic and local inflammation can result from the hormonal effects
et al., 2009). Similarly, cancer mortality was 46% lower in the patients of adipose tissue, or it can be caused by the chemical or mechanical
treated with surgery than in the controls (HR = 0.54; 95% CI: 0.37, injury, as in the context of gastroesophageal reflux or chronic gall-
0.78). It is interesting that the reduction in mortality was seen for all stones. The effects of obesity on metabolic hormones are thought
cancers combined, whereas the reduction in incidence was limited to to affect multiple types of cancers. Effects on steroid hormones are
obesity-​related cancers (esophagus [adenocarcinoma only], colorec- thought to be more specific, affecting breast and endometrial can-
tal, pancreas, postmenopausal breast, corpus and uterus, kidney, cer (and perhaps prostate and colon as well); local inflammation has
non-​Hodgkin lymphoma, leukemia, myeloma, liver, and gallbladder) been implicated at some, but not all, cancer sites. Fat cells secrete and
(Adams et al., 2009). These results were supported by three other stud- receive endocrine signals and are a major site for the metabolism of
ies (Adams et  al., 2007; Flum and Dellinger, 2004; Sjostrom et  al., sex steroids. It is difficult to separate the metabolic and endocrine
2007) that reported reductions in all-​cause and all-​cancer mortality in functions of adipose tissue, since these pathways overlap.
morbidly obese patients who received gastric bypass surgery, com-
pared to those who did not.
The Swedish Obese Subjects cohort, a prospective, controlled Metabolic Peptide Hormones
intervention trial (Sjostrom et al., 2009), found that individuals who
had bariatric surgery had a hazard ratio of 0.67 (95% CI: 0.53, 0.85) Insulin and IGF-​1
for overall incident cancers when compared to participants who Insulin and IGF-​1 are closely related metabolic protein (peptide) hor-
received conventional treatment. Of note, an interaction with gender mones that are thought to mediate some of the effects of excess body
was observed:  bariatric surgery was associated with reduced cancer fat on cancer risk. Excess adiposity, particularly visceral fat (Donohoe
incidence in obese women (HR = 0.58; 95% CI: 0.44, 0.77) but not et  al., 2011; Wajchenberg, 2000), induces insulin resistance. The
in obese men (HR  =  0.97; 95% CI:  0.62, 1.52). These results were resulting hyperinsulinemia suppresses hepatic production of hormone
consistent with previous observational findings that lower cancer inci- binding proteins (e.g., insulin-​like growth factor binding proteins
dence and mortality associated with gastric bypass surgery were seen [IGFBPs]) (Kaaks et al., 2002a). This increases the concentration of
only in women (Adams et al., 2009), although surprisingly, in the latter bioavailable IGF-​1, especially that which is not bound to IGFBP-​3
study breast cancer risk did not differ between the surgery and control (van Kruijsdijk et al., 2009). The net consequence of excess adipos-
groups. ity on metabolic hormones is to increase concentrations of circulating
360

360 PART III:  THE CAUSES OF CANCER

Bioavailable
Sex Hormones

Aromatase

SHBG
Insulin
Proliferation

Excessive Free Fatty Acids


Insulin
Calorie Central Leptin Resistance Apoptosis
Intake Adiposity
Adiponectin
IGFBPs

Bioavailable Genomic
IGF-1 Instability

Inflammation

Figure 20–​3.  Proposed mechanisms by which excess adiposity affects cancer risk.

insulin and bioavailable IGF-​1 (Kaaks et al., 2002a). Normal and can- 2005; Wu et  al., 2011). This finding, if confirmed, suggests a com-
cerous cells express receptors for insulin and IGF-​1 (Pollak, 2008). mon carcinogenic pathway shared by insulin and IGF-​1, whereby high
Peptide hormones such as insulin and bioavailable IGF-​1 promote car- levels of either insulin or IGF-​1 may be sufficient to achieve maximal
cinogenesis by enhancing cell proliferation and inhibiting apoptosis effect. For example, colorectal cancer may possess hybrid insulin and
(van Kruijsdijk et al., 2009). It should be noted that IGF-​1 elicits stron- IGF-​1 receptors, which can be stimulated and saturated by either insu-
ger mitogenic and anti-​apoptotic responses than insulin. Insulin may lin or IGF-​1.
act primarily through enhancing the bioavailability of IGF-​1, rather Adult height is also associated with breast cancer risk (Zhang B et al.,
than through direct ligand activity (Nimptsch and Giovannucci, 2012). 2015), potentially implicating the IGF-​1 pathway in the pathogenesis
Cancer cells may reactivate the expression of insulin receptor isoform of breast as well as colorectal cancer. The Nurses’ Health Study was
A, which is normally expressed primarily in embryonic/​fetal tissue. the first prospective study to examine prediagnostic concentrations of
The A  isoform is more mitogenic than the B isoform of the insulin circulating IGF-​1 in relation to breast cancer risk. A  strong positive
receptor, which is typically expressed in adults (Pandini et al., 2003). association was observed only in women who were premenopausal
Yu and Rohan have comprehensively reviewed the role of the IGF at the time of blood collection (Hankinson et al., 1998). In contrast,
family on cancer development and progression (Yu and Rohan, 2000). a pooled analysis of 17 prospective studies reported increased risk
Based on epidemiologic evidence, the insulin and/​or IGF-​1 pathways of breast cancer in women with higher circulating IGF-​1 concentra-
appear to be particularly relevant to cancers of the colorectum, breast, tions, regardless of menopausal status at the time of blood collection.
endometrium, and prostate. In fact, the positive association was stronger among postmenopausal
Indirect evidence linking IGF-​1 to colorectal cancer comes from than premenopausal women (Endogenous Hormones Breast Cancer
the observation that taller individuals, who potentially had greater Collaborative Group, 2010). Both this analysis and a subsequent study
exposure to IGF-​1 during adolescent growth, are consistently associ- conducted in the European Prospective Investigation into Cancer and
ated with an increased risk of colorectal cancer (Green et al., 2011). Nutrition (EPIC) cohort (Kaaks et  al., 2014)  observed the positive
Epidemiologic studies based on direct measurement of IGF-​1 are lim- association with IGF-​1 only with ER+ breast cancer. This difference
ited to measurements made in adults. Recent meta-​analyses of obser- by ER status is supported by accumulating evidence of the presence of
vational studies found that higher levels of circulating IGF-​1 levels in synergistic cross-​talk in mammary cells between IGF-​1 and estrogen
adulthood are associated with an increased risk of both colorectal can- signaling pathways (Yee and Lee, 2000).
cer (Rinaldi et al., 2010) and high-​risk adenomas (Yoon et al., 2015a). The role of insulin in breast cancer etiology remains unclear. Given
Of note, adult IGF-​1 levels are an imperfect surrogate for adolescent that estrogens have a dominant influence on breast cancer (Colditz,
IGF-​1 levels. Whereas height is strongly correlated with IGF-​1 lev- 1998), it has been hypothesized that insulin may promote breast cancer
els in adolescence, it is only weakly correlated with levels in adults. by suppressing hepatic production of sex hormone binding globulins
However, Yoon and colleagues have inferred that both early-​life and (SHBG), thereby increasing the bioavailability of estrogens (Singh
later-​life IGF-​1 levels may contribute to the development and progres- et al., 1990). However, no overall association was observed between
sion of colorectal cancer (Yoon et al., 2015b). insulin or C-​peptide and breast cancer in a recent meta-​analysis of 10
Strong evidence supports a role for insulin in colorectal carcino- prospective studies (Autier et al., 2013) Six of these studies measured
genesis. Studies of this issue have generally measured C-​peptide, a insulin (Eliassen et al., 2007; Gunter et al., 2009; Kaaks et al., 2002b;
marker of insulin secretion, rather than insulin itself, because the lon- Kabat et  al., 2009; Mink et  al., 2002; Sieri et  al., 2012); five mea-
ger half-​life of C-​peptide provides a more accurate marker of insu- sured C-​peptide (Cust et al., 2009; Eliassen et al., 2007; Keinan-​Boker
lin activity. In a recent meta-​analysis of eight prospective studies, et al., 2003; Toniolo et al., 2000; Verheus et al., 2006). Contrary to the
higher C-​peptide levels were associated with an approximately 40% hypothesis that estrogen mediates the increased risk, the most recent
increased risk of colorectal cancer (Chen et al., 2013). Furthermore, prospective study that examined the relationship between C-​peptide
an elevated risk of colorectal neoplasia is associated with disorders and subtypes of breast cancer found a stronger association with ER–​
that involve hyperinsulinemia, such as insulin resistance (as marked than ER+ cancer (Ahern et al., 2013). Thus, the mechanism by which
by high HOMA-​IR or by hypertriglyceridemia) and diabetes mellitus hyperinsulinemia increases breast cancer risk remains unclear.
(Grundy, 1999; Yao and Tian, 2015; Yoon et al., 2015b). Interestingly, The relationship between excess body fat and endometrial cancer is
in some studies, individuals who have elevations in both C-​peptide and thought to be driven by primarily by estrogen (Schmandt et al., 2011),
IGF-​1 levels have the same level of increased risk of colorectal cancer although insulin may also play a role. Epidemiologic evidence sup-
as those who have elevated levels of only one biomarker (Wei et al., porting the link between insulin resistance and endometrial cancer
 361

Obesity and Body Composition 361


risk is comprehensively reviewed by Mu and Zhu (Mu et al., 2012). et al., 2008; Oh et al., 2007), was not more strongly associated with
A recent meta-​analysis found an approximately 2-​fold increased risk colorectal cancer risk (Aleksandrova et al., 2012).
of endometrial cancer among women with type 2 diabetes mellitus Similarly intriguing, a sex difference has been observed in an
compared with non-​diabetic women (Liao et al., 2014). In a Mendelian analysis of the Nurses’ Health Study and the Health Professionals
randomization study that provides evidence for causality, single nucle- Follow-​up Study, where the inverse association between adiponectin
otide polymorphisms (SNPs) for fasting insulin and SNPs for post-​ and colorectal cancer risk was confined to men (Song et al., 2013b).
challenge insulin were linked to an elevated endometrial cancer risk Of note, women generally had higher levels of adiponectin than men
(Nead et al., 2015). (median:  8.2 vs. 5.3 μg/​mL). The gender difference in plasma lev-
A recent meta-​analysis of studies of height and prostate cancer els appears to be independent of fat mass or distribution, and may
observed a modest positive association between adult attained height result from the influence of sex steroid hormones (Cnop et al., 2003;
and prostate cancer risk (Zuccolo et  al., 2008). A  pooled study of Lanfranco et al., 2004; Yarrow et al., 2012). Thus, the sex heteroge-
12 prospective studies that measured IGF-​1 concentrations in adults neity in the adiponectin–​colorectal cancer association may reflect a
found a positive relationship between IGF-​ 1 and prostate cancer distinct influence of altered estrogen and testosterone concentrations
(Roddam et  al., 2008). The results are inconsistent, however, with related to adiposity (Giovannucci, 2007; Koo and Leong, 2010).
respect to the association by stage and grade of prostate cancer. IGF-​ A Mendelian randomization study of SNPs associated with circulat-
1 appears to be more strongly associated with low-​grade than high-​ ing adiponectin levels (Dastani et al., 2012; Ling et al., 2009; Richards
grade tumors (Chan et  al., 2002; Nimptsch et  al., 2011b; Roddam et al., 2009) did not report consistent, strong associations with a variety
et  al., 2008), whereas the opposite is observed for both height and of outcomes, including colorectal cancer (Song et al., 2015a), although
obesity in adulthood. the SNPs explain only a small fraction of variation in adiponectin lev-
Only a few prospective studies have examined the association els. However, low circulating levels of adiponectin have been primar-
between prediagnostic levels of circulating IGF-​1 and insulin and pan- ily linked to the increased risk of KRAS-​mutant cancer (Inamura et al.,
creatic cancer risk, and the results are conflicting (Giovannucci and 2016), and a larger Mendelian randomization study would be needed
Michaud, 2007). However, numerous studies have investigated the to assess whether adiponectin-​related SNPs are associated with any
relationship between history of type 2 diabetes and pancreatic cancer particular subtypes of colorectal cancer.
risk. A recent meta-​analysis reported an elevated risk associated with For breast cancer, although retrospective case-​control studies have
type 2 diabetes, regardless of the duration of diabetes prior to cancer generally reported lower adiponectin levels in cases than in controls
diagnosis (RR = 1.5, 95% CI: 1.3, 1.8 for 5–​9 years; RR = 1.5, 95% (Macis et  al., 2014), this finding was replicated in only two (Macis
CI: 1.2, 2.0 for 10+ years) (Huxley et al., 2005). et  al., 2012; Tworoger et  al., 2007)  of the seven prospective studies
(Cust et al., 2009; Gaudet et al., 2010; Gunter et al., 2015; Macis et al.,
Adiponectin 2012; Ollberding et  al., 2013; Touvier et  al., 2013; Tworoger et  al.,
Another peptide hormone, adiponectin, possesses significant anti-​ 2007). However, the two studies differ in that adiponectin was associ-
inflammatory and insulin-​ sensitizing effects (Tilg and Moschen, ated with breast cancer risk in premenopausal women in one study
2006), and has been postulated to mediate the relationship between (Macis et  al., 2012), but only with postmenopausal breast cancer in
obesity and several cancers (Vansaun, 2013). Although predominantly the other (Tworoger et  al., 2007). Similarly, for endometrial cancer,
secreted by white adipose tissue (Maeda et  al., 1996), adiponec- prospective studies have produced inconsistent findings. While an
tin expression is reduced in obesity because of complex factors that early study reported that prediagnostic adiponectin level, independent
suppress transcription, including adipocyte hypertrophy, inflamma- of BMI and other metabolic biomarkers, was associated with a lower
tion, and oxidative stress (Kadowaki et  al., 2006; Kim et  al., 2015; risk of postmenopausal endometrial cancer (Cust et  al., 2007), this
Maury and Brichard, 2010). Lower levels of adiponectin have been finding was not replicated in three subsequent studies (Dallal et  al.,
linked to the development of metabolic syndrome and type 2 diabetes 2013; Luhn et al., 2013; Soliman et al., 2011), except for an inverse
(Kadowaki et al., 2014; Li et al., 2009; Matsuzawa, 2006). The insulin-​ association noted in one study only among non-​current MHT users
sensitizing effect of adiponectin may be mediated, at least in part, by only (Luhn et al., 2013).
an increase in fatty-​acid oxidation via activation of AMP-​activated For prostate cancer, higher adiponectin levels were associated with
protein kinase (AMPK) and peroxisome proliferator-​activated recep- lower risk of high-​grade or lethal prostate cancer in the Physicians’
tor (PPAR)-​ α, which bind to adiponectin receptors AdipoR1 and Health Study (Li et  al., 2010), and this finding was further corrob-
AdipoR2, respectively (Yamauchi et  al., 2002, 2003). Synthetic orated by genetic studies demonstrating that SNPs related to lower
AdipoR agonist has been shown to ameliorate diabetes and prolong adiponectin levels were associated with higher prostate cancer risk
the life span in mice who are either genetically obese or become so (Dhillon et al., 2011). However, such findings have not been consis-
on a high-​fat diet, suggesting that adiponectin can be a promising tently replicated in other studies (Baillargeon et al., 2006; Moore et al.,
therapeutic target for metabolic disorders (Okada-​Iwabu et al., 2013). 2009; Touvier et al., 2013). For pancreatic cancer, a pooled analysis of
Given the well-​established role of inflammation and insulin resistance five prospective cohort studies observed a common inverse association
in carcinogenesis, adiponectin has been hypothesized to lower obesity-​ between adiponectin and pancreatic cancer risk among smokers and
related cancer risk. In addition, experimental evidence suggests that non-​smokers (Bao et al., 2013). Higher levels of circulating adiponec-
adiponection may directly control malignant potential by regulating tin were inversely associated with multiple myeloma risk (Hofmann
metabolic, inflammatory, and cell cycle signaling pathways (Fujisawa et al., 2012), and also have been associated with a lower risk of pro-
et  al., 2008; Moon et  al., 2013; Sugiyama et  al., 2009). Despite the gression from monoclonal gammopathy of undetermined significance
strength of experimental data, epidemiologic studies have reported (MGUS) to myeloma (Fowler et al., 2011; Reseland et al., 2009).
inconsistent findings on the relationship of prediagnostic adiponectin For primary liver cancer, four prospective studies have consistently
with risk of several specific cancers. shown that high adiponectin is associated with increased risk among
Six prospective case-​control studies have studied the relationship patients with chronic hepatitis C (Arano et al., 2011), among patients
between adiponectin and colorectal cancer. Three of these reported no with hepatitis B or C infection (Chen CL et al., 2014; Michikawa et al.,
association (Chandler et al., 2015; Lukanova et al., 2006; Stocks et al., 2013), or among participants with low infection rate (Aleksandrova
2008), whereas three others reported mixed results (Aleksandrova et  al., 2014). This positive association appears to be stronger for
et al., 2012; Ho et al., 2012; Song et al., 2013b). In the Women’s Health non-​HMW adiponectin (Aleksandrova et al., 2014; Michikawa et al.,
Initiative, while high plasma adiponectin was associated with lower 2013). Although these findings seem to contradict those for other can-
colorectal cancer risk, this association disappeared after adjusting for cers, circulating adiponectin has been found to be elevated in patients
insulin levels, suggesting that insulin could be on the causal pathway with chronic hepatitis C or with high hepatitis B viral load (Chen CL
(Ho et al., 2012). This notion was not supported by EPIC, however, in et al., 2014; Wong et al., 2010), suggesting that adiponectin may rise
that the high molecular weight fraction of adiponectin, which is most as a result of reduced degradation by the liver due to virus-​induced
strongly associated with insulin sensitivity and diabetes (Heidemann inflammation and injury. Furthermore, higher adiponectin levels
362

362 PART III:  THE CAUSES OF CANCER


among patients with newly diagnosed or recurrent hepatocellular car- to testosterone in women not using MHT has been associated with
cinoma have been linked to increased mortality (Siegel et al., 2015). lower colorectal cancer risk after adjustment for BMI and C-​peptide
Interestingly, the phenomenon of adiponectin-​ associated increased levels (Lin et al., 2013). One potential explanation is that the increase
mortality, known as the “adiponectin paradox,” has also been reported in estrogens associated with obesity may partially offset the increased
in patients with colorectal cancer (Chong et al., 2015), prevalent car- risk from obesity-​related metabolic factors, such as hyperinsulinemia.
diovascular disease (Cavusoglu et al., 2006), and heart failure (Kistorp Adiposity in men is associated with lower levels of androgens (Kapoor
et al., 2005), and among the elderly (Karas et al., 2014; Kistorp et al., et al., 2005); higher circulating levels of testosterone are associated with
2005; Kizer et  al., 2012; Laughlin et  al., 2007; Poehls et  al., 2009; lower colorectal cancer (Lin et al., 2013). A higher ratio of estradiol to
Wannamethee et al., 2007). Although there is evidence suggesting that testosterone was associated with higher risk in men, which may reflect
increased adiponectin concentrations may represent a response to an increasing aromatization and lower testosterone levels (Lin et al., 2013).
underlying disease process, this paradox has not been explained and
warrants further studies to elucidate the complex pathophysiology of
adiponectin (Sattar and Nelson, 2008). Inflammation
Chronic inflammation creates a tissue microenvironment that stimu-
lates cellular proliferation, suppresses apoptosis, and generates free
Sex Steroid Hormones radicals that can damage DNA (see Chapter 25). While it is difficult to
Contributing to the role of adipose tissue as an endocrine organ, body measure local inflammation in population studies, there are numerous
fat is a major site for the synthesis of sex steroid hormones, and con- clinical examples in which chronic local inflammation predisposes to
tributes to the bioavailability of these hormones. Steroid hormone certain types of cancer (see Chapter 25). This section discusses several
synthesis occurs as a result of adipocyte expression of the aromatase examples in which tissue damage caused by the effects of obesity fos-
enzyme. In men and postmenopausal women, the primary source of ters neoplastic growth and malignant transformation. It also discusses
circulating estrogens is the conversion of androgens (testosterone and systemic markers of inflammation that have been measured in epide-
androstenedione) to estrogens (estradiol and estrone) by adipose tissue. miologic studies of cancer.
Less active forms of these hormones (androstenedione, estrone) are
converted to more active forms (testosterone, estradiol) by the enzyme Examples of Local Inflammation
17B-​hydroxysteroid dehydrogenase, expressed in adipocytes. Adipose
tissue contributes to increased bioavailability of sex hormones through Esophageal Adenocarcinoma. Esophageal adenocarcinoma
the inhibition by insulin of the synthesis of SHBG in the liver (Crave typically occurs in the lower one-​third of the esophagus, and has a
et al., 1995). Lower SHBG levels result in higher concentrations of free greater propensity to occur in men than in women (see Chapter 30).
estradiol and free testosterone, which are more readily taken up by cells Incidence rates have increased in parallel with the increasing preva-
in target organs. Combining the strengths of several nested case-​control lence of obesity. This malignancy usually arises from mucosa that has
studies of circulating hormones and breast cancer risk, a large collab- been damaged by chronic reflux of acid and bile from the stomach into
orative study of BMI and circulating hormones was conducted recently, the lower esophagus. A recent extensive meta-​analysis demonstrated
including more than 6000 postmenopausal women (Endogenous the importance of central adiposity in increasing risk of esophageal
Hormones Breast Cancer Collaborative Group, 2011). Compared to inflammation, metaplasia (Barrett’s esophagus), and neoplasia (Singh
thin women (BMI ≤22.5), obese women (BMI >30) had 47% higher et al., 2013). Obesity, particularly central obesity, is the strongest risk
circulating estradiol and estrone levels. In addition, obese women had factor for acid reflux through disruption of the normal physiology of
46% lower SHBG levels, contributing to significantly higher circulat- the gastroesophageal junction (Friedenberg et al., 2008). The relation-
ing free estradiol (89%) and free testosterone (72%) levels. ship between obesity and acid reflux likely largely explains the asso-
Steroid hormones play an important etiologic role in cancers of the ciation between obesity and esophageal adenocarcinoma. The male
breast, endometrium, ovary, and prostate. Circulating hormone levels propensity for an abdominal pattern of adiposity may at least partly
have been consistently associated with postmenopausal breast cancer account for the higher rates of esophageal adenocarcinoma in men
(Kaaks et al., 2005; Key et al., 2002; Zhang X et al., 2013). In a pooled (Friedenberg et al., 2008; Singh et al., 2013).
analysis of nine prospective studies, including 663 breast cancer cases
and 1765 controls, higher circulating levels of estrogens, including Gallbladder Cancer. Gallbladder cancer is associated with
estradiol, estrone, estrone sulfate, and free estradiol, were associ- excess adiposity (Larsson and Wolk, 2007; Park et al., 2014; Tan et al.,
ated with higher subsequent risk of breast cancer (e.g., top vs. bot- 2015), and with related metabolic abnormalities such as type 2 diabe-
tom quintile estradiol, RR = 2.0; 95% CI: 1.5, 2.7) (Key et al., 2002). tes (Gu et al., 2015), lipid disorders, and elevated fasting blood glu-
Adipose tissue may also affect breast cancer risk locally, given that cose (Borena et al., 2014; Rapp et al., 2006) (see Chapter 34). Obese
adipocytes in breast stromal tissue likely contribute to estrogen levels women are at particularly high risk of gallbladder cancer. The main
in the microenvironment. risk factor for gallbladder cancer is a history of gallstones (Randi et al.,
In endometrial carcinogenesis, the interplay between peptide and sex 2006) (see Chapter 34). Obesity, hyperinsulinemia, and dyslipidemia
steroid hormones appears to be critical (Kaaks et al., 2002a). Higher insu- are related to a cluster of factors that predispose to gallstones, including
lin and IGF-​1 levels in obese women increase ovarian androgen produc- biliary supersaturation with cholesterol, inflammation, hypersecretion
tion. In premenopausal women, ovarian androgen production interferes of mucin, slow colonic motility, and increased intestinal cholesterol
with ovulation, which decreases production of progesterone. Normally, absorption. Chronic inflammation from recurrent gallstones is the pre-
progesterone provides a check on estrogen-​induced proliferation in the sumptive mediator of obesity-​related gallbladder cancer.
uterus (Key and Pike, 1988). Therefore, decreases in progesterone pro-
duction in premenopausal women, and increases in bioavailable estro- Liver Cancer.  Obesity has been suggested to be the second larg-
gens, create the environment for endometrial carcinogenesis. est contributor to the recent increase in the incidence of hepatocel-
Sex steroid hormones play a role in colorectal cancer, and may partly lular carcinoma in developed countries, after hepatitis C infection
account for the stronger association with obesity observed in men. In (Caldwell et al., 2004; Starley et al., 2010) (see Chapter 33). Obesity
women, higher exposure to estrogen and progesterone, whether through plays an important role in the progression from non-​alcoholic fatty
pregnancy or the use of MHT, is associated with lower risk of colorectal liver disease to steatohepatitis to cirrhosis and hepatocellular carci-
cancer. For instance, in the Women’s Health Initiative trial, combined noma (Caldwell et al., 2004; Cohen et al., 2011; Hassan et al., 2015).
use of estrogen and progestin was associated with a 40% lower risk As the most common form of chronic liver disease, non-​alcoholic
of colorectal cancer (Chlebowski et  al., 2004). Although associations fatty liver disease affects about 30% of the US general population
with circulating levels of estradiol are inconsistent (Clendenen et  al., and up to 90% of those who are morbidly obese (Torres and Harrison,
2009; Gunter et al., 2008; Lin et al., 2013), a higher ratio of estradiol 2008). The more aggressive form of non-​alcoholic fatty liver disease,
 36

Obesity and Body Composition 363


non-​alcoholic steatohepatitis, has a prevalence of 5%–​7%; in this sub- increased lifetime exposure to insulin (Caprio et al., 1995), which may
group, up to 9% can progress to cirrhosis (Ong and Younossi, 2007). contribute to subsequent risk of colon cancer.
An estimated 40%–​60% of patients with cirrhosis from non-​alcoholic Mechanisms by which childhood/​adolescent obesity may influence
steatohepatitis develop major complications, including hepatocellu- breast cancer risk later in life could involve both the hormonal milieu
lar carcinoma over 5–​7 years of follow-​up (Adams et al., 2005; Hui in early life and the establishment of set points that persist with age.
et al., 2003). Although overweight and obese girls are likely to have earlier ages
at menarche compared to normal weight girls, they also are more
Systemic Markers of Inflammation likely to have anovulatory menstrual cycles (Rich-​Edwards et  al.,
Epidemiologic studies can more easily measure systemic markers of 1994; Stoll, 1998) and irregular cycle patterns (Tehard et al., 2005).
inflammation than inflammation at the tissue level. It is often unclear Adiposity at young ages is associated with slower adolescent growth
whether the increase in a systemic marker derives from local inflam- (Berkey et al., 1999). Excess adiposity in childhood is inversely asso-
mation or from a more generalized inflammatory process, perhaps ciated with at least two breast cancer risk factors in adulthood: circu-
involving the infiltration of immune cells into adipose tissue, espe- lating IGF-​1 levels (Poole et  al., 2011), and breast density (Samimi
cially visceral adipose. It is also uncertain whether a low-​grade gen- et al., 2008; Sellers et al., 2007). Among 6520 women in the Nurses’
eralized inflammatory state can predispose to cancers at remote sites. Health Study and Nurses’ Health Study II, those who were overweight
One of the most widely used inflammatory biomarkers is CRP, which at young ages had 14% lower adult plasma IGF-​1 levels, compared
is secreted from the liver and is an acute phase marker of inflamma- with leaner girls (Poole et al., 2011). The inverse associations observed
tion. CRP is increased in obese individuals (Visser et al., 1999), but between childhood body fatness and breast density suggest that the
also is increased by smoking, alcohol, physical inactivity, and various structure of breast tissue may be established as a result of early life
inflammatory conditions (Chan et al., 2015). Recognizing the complex adiposity and then may persist throughout life (Samimi et al., 2008;
determinants of CRP, some studies have examined pre-​diagnostic CRP Sellers et al., 2007).
levels in blood in relation to risk of various cancers. In a meta-​analysis
of circulating CRP and incident breast cancer risk, a statistically sig-
nificant positive association was found; each doubling of the CRP Microbiome
concentration was associated with a 7% increase in incident breast
cancer (Chan et al., 2015). In a meta-​analysis of colon cancer, circu- The explosion of research in host-​microbe interaction over recent
lating CRP was modestly associated with increased risk, although a years supports a potential role of the gut microbiota in human health
closely related inflammatory marker, IL-​6, was not (Zhou et al., 2014). and disease through regulation of host metabolism and immune func-
The interpretation of these results is unclear; some studies observed tion (Blumberg and Powrie, 2012; Brestoff and Artis, 2013; Guarner
increased risk with higher levels of the inflammatory markers only in and Malagelada, 2003; Kau et al., 2011; Sommer and Backhed, 2013;
the early years after blood samples were provided (Song et al., 2013a), Tremaroli and Backhed, 2012). Several lines of evidence indicate a
raising the possibility that the tumor itself may have affected the circu- bidirectional association. While perturbations in the gut microbiota
lating levels (i.e., reverse causation). may predispose individuals to obesity (Cox and Blaser, 2013), obe-
Because CRP and IL-​6 are affected by many factors besides excess sity also can alter gut microbiota, potentially contributing to obesity-​
body weight, it is difficult to ascribe any observed association strictly linked carcinogenesis. Maintenance of body weight strongly predicts
to obesity. This problem is illustrated in a large case-​control study of microbiota stability (Faith et al., 2013); obesity has been associated
prostate cancer nested in a prospective cohort. As expected, obese with lower diversity and altered composition of gut microbiota, spe-
men had substantially higher levels of CRP and IL-​6. However, IL-​ cifically a decrease in the phylum Bacteroidetes and an increase in
6 was associated with increased risk of prostate cancer only among Firmicutes (Furet et  al., 2010; Le Chatelier et  al., 2013; Ley et  al.,
men with normal BMI (Stark et al., 2009). An inverse relationship was 2005, 2006). An opposite change has been observed with weight
observed between IL-​6 and prostate cancer among men with increased loss (Aron-​Wisnewsky et  al., 2012; Furet et  al., 2010; Kong et  al.,
BMI. This finding suggests that determinants of elevated inflammatory 2013; Ley et  al., 2006; Zhang et  al., 2009). Furthermore, growing
markers unrelated to adiposity accounted for the increased risk. A pos- evidence implicates the altered microbial community and enrich-
sible explanation is that high IL-​6 or CRP among the leaner men may ment of certain microbes in cancer development (e.g., Fusobacterium
be more indicative of inflammation within the prostate gland, which nucleatum in colorectal cancer) (Castellarin et al., 2012; Kostic et al.,
directly influences risk, whereas high concentrations of inflammatory 2013). Microbes may amplify or mitigate carcinogenesis by affecting
markers among the heavier men reflected the greater amount of adi- genomic stability (Cuevas-​Ramos et  al., 2010; Guerra et  al., 2011;
pose tissue. Similarly, in colon cancer, an association with IL-​6 was Nougayrede et  al., 2006), altering the balance of host cell prolif-
observed only in leaner men, again suggesting that inflammation at eration and death (Rubinstein et  al., 2013; Sears, 2009), regulating
the tissue level, rather than systemic inflammation related to adiposity, pro-​inflammatory or immunosuppressive responses (Hu et al., 2013;
influences risk (Song et al., 2013a). Further, using a Mendelian ran- Irrazabal et  al., 2014; Kostic et  al., 2013; Warren et  al., 2013), and
domization approach, genetic determinants of elevated levels of CRP influencing host metabolism (Belcheva et al., 2014; Donohoe et al.,
were not associated with increased risk of cancer, possibly arguing 2014; Singh et al., 2014). Therefore, it has been hypothesized that the
against a causal role of systemic inflammation for cancer risk (Allin gut microbiota may affect multiple pathways by which obesity influ-
et al., 2010). ences cancer risk (Ohtani et al., 2014). Indeed, studies in liver cancer
Although it is not clear that obesity-​associated systemic inflam- have suggested that obesity-​induced alteration of the gut microbiota
mation would necessarily affect the risk of solid tumors, effects on resulted in enhanced production of DNA-​damaging secondary bile
immune system cancers are also plausible. Obesity has been associ- acid (e.g., deoxycholic acid) that promotes carcinogenesis through
ated with increased risk of multiple myeloma, a malignancy of plasma cellular senescence-​related pathways (Ohtani et al., 2014; Yoshimoto
cells (Teras et al., 2014); IL-​6, a pro-​inflammatory cytokine, is a potent et al., 2013). Given the potential role of secondary bile acids, further
growth factor for plasma cells and may contribute to the risk of mul- studies are needed to investigate whether metabolic change due to
tiple myeloma (Ziakas et al., 2013). microbial imbalance also mediates obesity-​related tumor promotion
in other organs.

Mechanisms Associated with Early Life Adiposity Site-​Specific Mechanisms: Hypertension


Excess body fat in childhood and/​or adolescence occurs during an in Renal Cell Cancer
important developmental period, and may influence lifelong metabolic Obesity is a risk factor for hypertension. Hypertension is associated
and hormonal patterns, as well as behaviors. Early life obesity has with higher risk of renal cell cancer in many studies (Chow et  al.,
been suggested to influence basal insulin levels, and thus contribute to 2000, 2010; Sanfilippo et  al., 2014; Weikert et  al., 2008), and has
364

364 PART III:  THE CAUSES OF CANCER


been postulated as a mechanism linking obesity to renal cell cancer. illustrated in the Health Professionals Follow-​up Study, where the PAF
However, epidemiologic data lend little support to this hypothesis. for colon cancer in men was 14% based on a single measure of BMI
Although individuals who are both obese and hypertensive have a and a cutpoint of 25, but increased to 30% when the BMI cutpoint was
higher risk of renal cell cancer than those who have only one of these reduced to 22.5 kg/​m2 and was based on an average of multiple mea-
conditions, the increased risk of renal cell cancer associated with BMI sures of BMI (Thygesen et al., 2008).
persists even after adjustment for diagnosis of hypertension or mea- Other methodologic factors also influence the estimates of PAF.
surement of blood pressure (Adams et  al., 2008; Chow et  al., 2000; Although BMI is widely used for PAF estimates because of its avail-
Sanfilippo et  al., 2014; Setiawan et  al., 2007). This indicates that ability, it is not an ideal measure of adiposity, as previously discussed.
hypertension-​independent mechanisms may mediate renal cell cancer More direct measures of visceral or central adiposity, such as VAT,
development in obese individuals. might be expected to provide stronger associations of the relative
risks and higher estimates of the PAF. For example, VAT based on CT
scan was strongly associated with risk of colorectal adenoma, even
ESTIMATED ATTRIBUTABLE FRACTION adjusting for BMI (Keum et  al., 2015). In study populations where
FOR  INCIDENCE AND SURVIVAL the majority of participants had BMI levels below 25 kg/​m2, VAT was
strongly and linearly associated with the risk of colorectal adenoma
Several studies have estimated the population attributable fraction (Keum et al., 2015). To date, there are limited data by which to esti-
(PAF), which incorporates both the prevalence of the exposure and the mate PAF using measures other than the traditional cutpoints for BMI.
strength of the association between the exposure and disease. While Relying on a single measure of BMI during adulthood conflates
the effects of obesity are relatively modest for specific types of cancer exposures that occur at different times of life. For example, a study of
(Table 20–​1), the prevalence of overweight and obesity are high, espe- breast cancer based on adult BMI cannot separate early life adiposity
cially in the United States and other high-​income countries. Therefore (which decreases breast cancer risk) from adult weight gain (which
the number of cancer cases attributable to overweight and obesity is increases risk). On a per kilogram increment basis, adult weight gain is
likely to be substantial. a much stronger risk factor for breast cancer than is adult BMI (Keum
The WCRF/​AICR has estimated the PAF for cancer sites designated et al., 2015). Thus, the use of a single measure of BMI may not accu-
as definitely caused by obesity (WCRF/​AICR, 2010b). The estimates rately reflect the relevant measure of adiposity at different time points.
for the United States range from 11% for gallbladder cancer to 34% The presence of effect modifiers may also differ between populations
for pancreatic cancer in men, and from 3% for colorectal to 49% for and may change over time, further constraining the estimates of PAF.
endometrial cancer in women. PAF estimates for the world were pub- For example, as described earlier, MHT may modify the effect of obe-
lished recently by the IARC (Arnold et al., 2015); the estimated frac- sity on breast cancer, just as smoking modifies the relation of adiposity
tions in men ranged from 6% for rectal cancer to 33% for esophageal to smoking-​related cancers. The prevalence and timing of these modi-
adenocarcinoma; for women, they ranged from 4% for rectal cancer to fying factors in the population will affect the strength of the association
34% for endometrial cancer. and the estimated PAF. For example, as the fraction of perimenopausal
Combining PAF estimates for specific cancer sites into an overall women who use MHT in the United States decreases, the estimated
PAF for obesity and cancer, the IARC estimated that 481,000 (3.6%) PAF for obesity and breast cancer would be expected to increase.
of all new cancers in 2012 in adults were attributable to high BMI A particularly important modifier to consider is smoking. As dis-
(Arnold et al., 2015). By sex, the PAF were 1.9% in men and 5.4% cussed earlier, the complex relationship between smoking and adi-
in women. Further, one-​quarter of these cases in 2012 were attributed posity makes it difficult to interpret the effects of adiposity. In the
to the global increase in BMI since 1982. If the population had main- Cancer Prevention Study II, a prospective mortality study begun by
tained the prevalence of overweight and obesity in 1982, instead of the American Cancer Society in 1982, the PAF of total cancer mortal-
increasing to today’s prevalence, an estimated 118,000 cases of cancer ity in the United States due to high BMI was 4.2% in men and 14.3%
could have been avoided. The magnitude of the PAF also varies by in women in the entire population, but these estimates increased to
human developmental indices (HDIs), since obesity is more common 14.2% and 19.8%, respectively, in the population limited to healthy
in countries with higher HDIs. For example, the IARC estimates that never-​smokers (Calle et  al., 2003). The inclusion of smokers quali-
5.4% of all incident cancers could be attributed to obesity in countries tatively changes the association observed among never smokers,
with very high HDI, 4.8% in those with high HDI, 1.6% in moderate either because of reverse causation (as hypothesized for lung cancer)
HDI countries, and 1.0% in those with low HDI. or intractable confounding (see earlier disucssion). Regardless of
In contrast to the estimates for obesity alone, the WCRF/​AICR the reason, given the strong effect modification, the implications are
Continuous Update Project estimates the PAF for the combined effects that the overall PAF in a population is arbitrary because it depends
of diet, physical activity, and body mass (WCRF/​AICR, 2009). The largely on the proportion of smokers in the population, and would not
estimates range from 19% to 26% in low-​to high-​income countries, apply to either the subgroup of smokers or to non-​smokers (Song and
respectively. While this approach would exaggerate the PAF due to Giovannucci, 2016).
body mass alone, these factors inherently overlap, since weight gain In summary, while the PAF is a widely used measure for commu-
reflects both diet and activity level. For example, the association nicating the impact of obesity on cancer, the calculation of PAF is
between colorectal cancer and a dietary pattern that stimulates insulin not straightforward, and the exact value can be influenced by many
secretion appears to be highly dependent on BMI and physical activity factors. Nevertheless, the high prevalence of obesity and its consistent
level (Fung et al., 2012). Thus, at least for some cancers, the effects associations with multiple types of cancer indicate that the disease bur-
of obesity cannot be isolated from those related to diet and physical den is high for cancer, as for other major diseases.
inactivity.
The published PAFs (besides those from the WCRF/​ AICR
Continuous Update Project) for cancer are likely to be substantial EXCESS ADIPOSITY AND CANCER SURVIVAL
underestimates of the true impact of excess adiposity on cancer risk for
numerous reasons. First, despite the categorical terms such as “over- The association between obesity and survival of patients with cancer
weight” and “obesity,” adiposity is a continuous variable. Thus, selec- is a relatively recent area of research, and one that is methodologi-
tion of the cutpoint is arbitrary. For example, a cutpoint of 25 kg/​m2 is cally difficult for several reasons. If adiposity is measured before
typically used because it is the lower bound of overweight, and may diagnosis, one cannot separate an effect on cancer incidence from an
be considered a potential target for population-​level interventions. Yet, effect on disease progression. However, if adiposity is measured after
the association may be linear even below 25 kg/​m2. Moreover, most diagnosis, it becomes impossible to exclude the possibility that weight
studies have relied on a single measure of adiposity, whereas BMI and loss resulted from reverse causation. Weight loss is a common and
other measures of adiposity may vary over time. The potential impor- generally unfavorable prognostic indicator in patients with advanced
tance of the cutpoint and of obtaining repeated measures of BMI are or metastatic cancer. Particularly for cancers that tend to have short
 365

Obesity and Body Composition 365


survival time (e.g., pancreatic cancer), the effects of local progression et al., 2012), obesity was associated with poorer survival in women
or metastatic disease may cause weight loss some time before diag- with ovarian cancer. A suggestive association was found for studies
nosis. Therefore, a measure of body mass at the time of diagnosis or that assessed BMI before, at the time of diagnosis or post-​diagnosis,
shortly thereafter is likely to be misleading. Further, low body weight or at commencement of chemotherapy. However, these were not sta-
may reflect loss of lean body mass prior to diagnosis. tistically significant, and there was considerable heterogeneity across
The relationship of obesity to survival from breast cancer has been studies.
more extensively studied than its relationship to survival of other can- One study collected clinical and pathologic data from 1543 patients
cers, because of the large number of cases and generally long survival. who underwent nephrectomy for renal cell cancer (Choi et  al., 2013).
A recent meta-​analysis summarized the results of 82 studies, encom- In contrast to most cancers, higher BMI measured after diagnosis was
passing 213,075 breast cancer survivors and 41,477 deaths (23,182 associated with improved survival. In the multivariable analysis, there
from breast cancer). Increased adiposity was associated with poorer was lower overall mortality (HR = 0.45; 95% CI: 0.29, 0.68) and cancer-​
overall and site-​specific survival for both pre-​and postmenopausal specific mortality (HR = 0.47; 95% CI: 0.29, 0.77) in obese (defined as
breast cancer, regardless of when BMI was ascertained (Chan et al., BMI ≥25) patients than in normal weight patients (BMI 18.5–<23). The
2014). Relative to normal weight women, the summary RRs for all-​ authors also conducted a meta-​analysis that corroborated these results.
cause mortality were as follows: 1.41, 95% CI: 1.29, 1.53 for obese While it is possible that renal cell cancers that develop in an obesogenic
(BMI >30.0); 1.07, 95% CI:  1.02, 1.12 for overweight (BMI 25.0–​ environment may be molecularly more indolent than other renal cell can-
<30.0); and 1.10, 95% CI:  0.92, 1.31 for underweight (BMI <18.5) cers (Hakimi et al., 2013), reverse causation is a likely explanation.
women. For breast cancer mortality, the associations were observed For esophageal cancer, higher body weight has tended to be associ-
regardless of menopausal status, but were non-​significantly stronger ated with better survival. This appears to be the case for either pre-​
for premenopausal (RRs for obesity: 1.75; 95% CI: 1.26, 2.41) than for diagnostic or post-​diagnostic BMI, and with squamous cell carcinoma
postmenopausal breast cancer (RR = 1.34; 95% CI: 1.18, 1.53). Breast or adenocarcinoma of the esophagus (Fahey et al., 2015; Hong et al.,
cancer mortality was increased when BMI was measured before diag- 2013; Zhang SS et al., 2013), but reverse causation cannot be ruled out.
nosis (14% increase per 5 kg/​m2 increment), <12 months after diagno- Although excess adiposity has clearly been associated with
sis (14%), and ≥12 months after diagnosis (29%). For total mortality, increased risk of numerous cancers, the influence of adiposity on
the corresponding increases in risk were 17%, 11%, and 8%. The asso- cancer survival has been less clear. The problem of reverse causation
ciation between obesity with breast cancer outcome did not appear to from the cancer of interest or comorbidities is extremely difficult to
differ by hormone receptor status of the tumor (Niraula et al., 2012). resolve. Some researchers have suggested the notion of an “obesity
Obesity has generally not been associated with prostate cancer paradox,” whereby those who develop cancer and remain overweight
incidence, but may be associated with poor survival. A meta-​analysis or obese have a better prognosis than those with lower weight. This so-​
based on 17 studies (Zhang X et al., 2015b), which included 3,569,926 called paradox is seen especially with cancers of the digestive tract and
men, found no association between obesity and prostate cancer inci- kidneys. The issue of reverse causation cannot be resolved, however,
dence (RR = 1.00; 95% CI: 0.95, 1.06), but a significant association without a better understanding of the natural history of weight loss
with prostate cancers categorized as aggressive (RR  =  1.14; 95% caused by either these cancers or comorbidities associated with them.
CI: 1.04, 1.25) and with prostate cancer mortality (RR = 1.24; 95% Furthermore, some evidence indicates that lean body mass is a critical
CI: 1.15, 2.33). These results were confirmed in another meta-​analysis factor for survival, and this is not measured well by BMI (Prado et al.,
of six post-​diagnosis survival studies, including 18,203 patients with 2015). After a diagnosis of cancer, future studies need to account for
932 prostate cancer deaths, where a 5 kg/​m2 increase in BMI was asso- body composition more precisely and not rely simply on BMI, which
ciated with 20% higher prostate cancer-​specific mortality (RR = 1.20; does not discriminate between lean tissue and adipose.
95% CI: 0.99, 1.46) (Cao and Ma, 2011). In 16 studies that followed
26,479 prostate cancer patients after primary treatment, a 5  kg/​m2
increase in BMI was significantly associated with 21% increased risk Impact of Excess Adiposity on Treatment Efficacy
of biochemical recurrence (RR = 1.21; 95% CI: 1.11, 1.31). Obesity has been associated with poorer outcomes; the reasons for
Four recent meta-​analyses have examined BMI in relation to colorec- this are numerous. Individuals who are obese are more likely to have
tal cancer survival, with similar results (Lee et al., 2015; Parkin et al., comorbid conditions and complications (McTiernan et al., 2010), mak-
2014; Schlesinger et al., 2014; Wu et al., 2014). Being underweight ing treatment less effective and more difficult to manage. These comor-
before diagnosis was associated with an approximately 60% higher bidities including surgical wound complications, lymphedema, and
all-​cause death rate compared to being normal weight; whereas being possible congestive heart failure (McTiernan et al., 2010). Additionally,
obese was associated with a 20%–​30% higher death rate. For post-​ obese cancer patients may be at risk for obesity-​related morbidities
diagnostic BMI, obesity generally was not associated with increased including type 2 diabetes, hypertension, cardiovascular disease, osteo-
risk of death, except for a suggested association with BMI ≥35 kg/​m2. arthritis, and pulmonary disease (Demark-​ Wahnefried et  al., 2012;
As observed for pre-​diagnostic underweight, being underweight after McTiernan et al., 2010). In some cases, obesity may be associated with
diagnosis was associated with higher overall mortality. a higher likelihood of unfavorable tumor characteristics (Griggs and
At least five studies have examined pre-​diagnostic BMI in relation Sabel, 2008), making the cancer more virulent and difficult to treat.
to mortality from pancreatic cancer (Majumder et al., 2015). When all Obesity is also a factor in suboptimal dosing of chemotherapy,
were combined, each 1 kg/​m2 increase in BMI was associated with a which decreases the effectiveness of the treatment and contributes
statistically significant 10% increase in mortality. Given that pancre- to poorer survival rates. Chemotherapy dosing for adults with can-
atic cancer has an extraordinarily high mortality rate, it is not feasible cer has traditionally been based on a patient’s estimated body surface
to study post-​diagnostic BMI in relation to outcome. area (BSA) (Freireich et  al., 1966), but in practice many clinicians
The international Ovarian Cancer Association Consortium related use ideal body weight, rather than actual body weight, to calculate
BMI levels shortly before diagnosis to survival among women with the BSA, or use a cap such as 2.0 m2 (Griggs et  al., 2012). Studies
invasive ovarian cancer (Nagle et al., 2015). The consortium included of clinical practice patterns show that up to 40% of obese patients
original data from 21 studies, 12,390 women with ovarian carci- receive suboptimal doses that are not based on actual body weight
noma, and 6715 total deaths documented during follow-​up. Women (Field et  al., 2008; Griggs and Sabel, 2008; Griggs et  al., 2005;
who had a BMI of 30–​34.9 had a 10% increase in the all-​cause death Lyman, 2009; Lyman et al., 2003, 2004; Shayne et al., 2007). While
rate; those with BMI ≥35 had a 12% increase. In a meta-​analysis this practice likely reflects clinicians’ concerns about over-​dosing
of 17 cohort studies (Bae et  al., 2014), obesity in early adulthood and chemotherapy-​related toxicity, there is no evidence to suggest
(HR = 1.67; 95% CI: 1.29, 2.16) and obesity 5 years before ovarian that toxicity is increased in obese patients receiving doses based on
cancer diagnosis (HR  =  1.35; 95% CI:  1.03, 1.76) were associated actual weight (Griggs et al., 2012). Under-​dosing of chemotherapy has
with increased patient mortality. In another meta-​analysis (Protani been well documented in breast cancer patients who are overweight or
36

366 PART III:  THE CAUSES OF CANCER


obese (Colleoni et al., 2005; Demark-​Wahnefried et al., 2012; Griggs between childhood adiposity and subsequent breast cancer risk, the
et al., 2007; Griggs and Sabel, 2008; Madamas et al., 2001), which in harms of childhood obesity and the high likelihood of childhood
turn leads to increased recurrence and mortality (Griggs et al., 2012; becoming adulthood obesity far outweigh the inverse association with
Lyman et al., 2004). Although data are more limited for other cancers, one type of cancer. Therefore, encouraging a prevention focus, for all
the evidence in breast cancer and the observed dose–​response rela- ages, on improvements in diet and physical activity is necessary to
tionship for most chemotherapy agents led the American Society of strive toward better energy balance (see Chapter 62.2 for more details).
Clinical Oncology (ASCO), in 2012, to establish clinical guidelines
for dosing obese cancer patients (Griggs et al., 2012). The ASCO panel
concluded that full weight-​based chemotherapy doses should be used FUTURE RESEARCH DIRECTIONS
to treat obese patients, although more research is necessary to better
understand chemotherapy pharmacokinetics in obese patients. While the associations between obesity and several cancers are clear,
there are still many areas where further research is necessary to
improve our understanding of the etiologic relationships and disease
INTERPRETATION OF TEMPORAL TRENDS burden. First, it is clear that obesity across the life course is impor-
tant, starting with childhood body size and following the trajectory
The secular trends of increasing prevalence of obesity have contrib- of weight gain over adulthood. However, understanding the complete
uted to temporal trends in the incidence of some types of cancer. For picture of cancer risk associated with a life course of excess weight
example, kidney cancer incidence has increased in many developed is essential given the large numbers of overweight children who are
countries over the same time period that obesity prevalence has risen growing into overweight adults. There is an urgent need for strategies
(e.g., in the United States from 1975 to 2007, age-​standardized rates to prevent this trajectory. More research is necessary on the underly-
doubled for women from 3.6 to 7.4 cases per 100,000, and nearly dou- ing biologic effects of childhood obesity, particularly to understand
bled for men from 8.0 to 14.5 cases per 100,000) (Ferlay et al., 2014). why childhood obesity is protective against breast cancer, in hopes of
Over this same time period, the trend in the incidence of pancreatic identifying an intervention without the health hazards of obesity for
cancer actually reversed, from a statistically significant decrease in girls who are lean.
incidence with the decrease in tobacco smoking from 1975 to 2000, Second, evidence is emerging that distribution of body fat (e.g., the
to a significant increase thereafter (Ryerson et al., 2016). Esophageal more metabolically active visceral fat), not just total amount of fat, is
adenocarcinoma incidence also has risen, with timing similar to the important in the etiology of some cancers. However, to better under-
rise in obesity prevalence. Given the strong association between adi- stand the role of visceral fat and its independence from BMI in cancer
posity and adenocarcinoma, the rising prevalence of obesity clearly etiology, we need better tools to assess this in epidemiology studies,
has contributed to the increase in incidence, although other factors also without relying on expensive MRI or relatively crude waist circumfer-
may have contributed (see Chapter 30). ence measures. The availability of a simple tool to assess visceral fat
For other cancers that have clear links to obesity, such as endome- could allow more accurate assessment of individual risk without the
trial and postmenopausal breast cancer, the incidence of disease over need for expensive testing. Further, a better assessment of lean mass
time is affected by obesity, but the picture is further complicated by versus adipose tissue would allow for enhanced understanding of the
secular changes in MHT use. The relationship of the obesity epidemic role of body mass at the time of and following a cancer diagnosis.
with colorectal cancer incidence over time is similarly complicated by Further research into tumor molecular markers specifically related
coincident increases in colonoscopy screening rates. to energy balance may contribute insights into the role of obesity and
Although the impact of the obesity epidemic is seen in the incidence energy balance in carcinogenesis. We propose that fatty acid syn-
of some cancers, given the relative recency of the obesity epidemic thase (FASN) may be a candidate biomarker, because in tumor cells,
and long latency of cancer, it is likely that the full effects of obesity on most fatty acids are synthesized de novo by FASN. FASN has been
cancer risk have not become apparent. A similar finding was observed suggested to act as a “metabolic oncogene,” conferring a selective
in the association between smoking and lung cancer risk, where early growth advantage to meet the excessive bioenergetic and structural
studies did not capture the full effects of smoking from adolescence demands of proliferating neoplastic cells (Menendez and Lupu, 2007).
through adulthood (Thun et al., 2013). Thus, while obesity is clearly Experimentally, FASN activity appears to promote tumor cell prolif-
associated with cancer risk, the impact of obesity across the life course, eration and survival and to allow neoplastic cells to attain autonomy
accumulating to decades of excess weight in many individuals, likely from the regulation of host metabolic status. In one analysis based
has not yet reached its peak in the population. on the Nurses’ Health Study, women who were overweight or obese
were at higher risk of FASN-​negative tumors but not at increased risk
of FASN-​positive tumors relative to normal-​weight women (Kuchiba
OPPORTUNITIES FOR PREVENTION et  al., 2012). The authors hypothesized that FASN-​ inactive cells
are dependent on excess energy balance for malignant progression,
The evidence of the harmful effect of adiposity on cancer risk and whereas FASN-​active cells may progress to cancer independent of
mortality is robust, and the biologic mechanisms underlying these energy balance status. In addition to incidence, tumor FASN status
associations are numerous. The critical question now is how best to may also have an influence on prognosis. For both colorectal (Ogino
reduce the number of cases of cancer that arise in the population as et  al., 2008)  and prostate cancer (Nguyen et  al., 2010), obesity was
a result of adiposity. Along with increased risks of cardiovascular, an adverse prognostic factor in individuals whose tumors expressed
endocrine, pulmonary, renal, gastrointestinal, and musculoskeletal FASN. Other tumor-​related metabolic factors may be important in
diseases, the association with cancer provides further impetus for determining how obesity will influence the development and progres-
intervention to reduce the incidence and prevalence of obesity in the sion of specific cancers. Further tumor tissue-​based studies should
population. Given that extensive evidence from the literature has docu- increase our understanding of the underlying mechanisms between
mented the difficulty for people to lose weight and keep weight off, the adiposity and cancer.
most important public health message is likely to be to avoid gaining Finally, while it is clear that weight gain should be avoided, the
weight across the life course. However, this message is not applicable direct association between weight loss and long-​term cancer risk
to the millions of people worldwide who are currently overweight and deserves further study, given the relatively sparse data on weight loss
obese, and the millions of overweight and obese children today who to date. In addition, new ideas to reduce this highly prevalent expo-
are likely to become overweight and obese adults. In these individuals, sure in the population are desperately needed, and may require policy
there should be attempts to prevent further weight gain (e.g., going changes similar to those used to address tobacco control. At a mini-
from overweight to obese), more research in ways to reduce BMI, mum, efforts should be undertaken to increase access to affordable
and efforts to find ways to ameliorate risk due to excess adiposity (for healthy foods and to enhance opportunities for physical activity for
example, diet, physical activity, drugs). Despite the inverse association the population.
 367

Obesity and Body Composition 367


References Bae HS, Kim HJ, Hong JH, et al. 2014. Obesity and epithelial ovarian cancer
survival:  a systematic review and meta-​analysis. J Ovarian Res, 7, 41.
Aaronson SA. 1991. Growth factors and cancer. Science, 254(5035), 1146–​
PMCID: PMC4022349.
1153. PMID: 1659742.
Baer HJ, Tworoger SS, Hankinson SE, and Willett WC. 2010. Body fatness at
Abbenhardt C, McTiernan A, Alfano CM, et al. 2013. Effects of individual and
young ages and risk of breast cancer throughout life. Am J Epidemiol,
combined dietary weight loss and exercise interventions in postmeno-
171(11), 1183–​1194. PMCID: PMC2915489.
pausal women on adiponectin and leptin levels. J Intern Med, 274(2),
Baillargeon J, Platz EA, Rose DP, et al. 2006. Obesity, adipokines, and pros-
163–​175. PMCID: PMC3738194.
tate cancer in a prospective population-​based study. Cancer Epidemiol
Adams KF, Leitzmann MF, Albanes D, et al. 2008. Body size and renal cell
Biomarkers Prev, 15(7), 1331-​1335. PMID: 16835332.
cancer incidence in a large US cohort study. Am J Epidemiol, 168(3),
Ballard-​Barbash R. 1994. Anthropometry and breast cancer:  body size—​a
268–​277. PMCID: PMC2727262.
moving target. Cancer, 74(3 Suppl), 1090–​1100. PMID: 8039144.
Adams LA, Lymp JF, St Sauver J, et al. 2005. The natural history of nonalcoholic
Ballard-​Barbash R, Friedenreich C, Slattery ML, and Thune I. 2006.
fatty liver disease:  a population-​based cohort study. Gastroenterology,
Obesity and body composition. In:  Schottenfeld D, and Fraumeni JF,
129(1), 113–​121. PMID: 16012941.
Jr. (Eds.), Cancer epidemiology and prevention (3rd ed., pp. 422–​448).
Adams TD, Davidson LE, Litwin SE, et  al. 2012. Health benefits of gas-
New York: Oxford University Press.
tric bypass surgery after 6  years. JAMA, 308(11), 1122–​ 1131.
Bandera EV, Chandran U, Zirpoli G, et al. 2013. Body size in early life and
PMCID: PMC3744888.
breast cancer risk in African American and European American women.
Adams TD, Gress RE, Smith SC, et al. 2007. Long-​term mortality after gastric
Cancer Causes Control, 24(12), 2231–​2243. PMCID: PMC3886722.
bypass surgery. N Engl J Med, 357(8), 753–​761. PMID: 17715409.
Bao Y, Giovannucci EL, Kraft P, et  al. 2013. A  prospective study of plasma
Adams TD, Stroup AM, Gress RE, et al. 2009. Cancer incidence and mortal-
adiponectin and pancreatic cancer risk in five US cohorts. J Natl Cancer
ity after gastric bypass surgery. Obesity (Silver Spring), 17(4), 796–​802.
Inst, 105(2), 95–​103. PMCID: PMC3545904.
PMCID: PMC2859193.
Bassett JK, Severi G, Baglietto L, et  al. 2012. Weight change and pros-
Ahern TP, Hankinson SE, Willett WC, et  al. 2013. Plasma C-​peptide, mam-
tate cancer incidence and mortality. Int J Cancer, 131(7), 1711–​1719.
mographic breast density, and risk of invasive breast cancer. Cancer
PMID: 22213024.
Epidemiol Biomarkers Prev, 22(10), 1786–​1796. PMCID: PMC3809910.
Bassett JK, Severi G, English DR, et al. 2010. Body size, weight change, and
Ahlgren M, Melbye M, Wohlfahrt J, and Sorensen TI. 2004. Growth patterns
risk of colon cancer. Cancer Epidemiol Biomarkers Prev, 19(11), 2978–​
and the risk of breast cancer in women. N Engl J Med, 351(16), 1619–​
2986. PMID: WOS:000283991600032.
1626. PMID: 15483280.
Belcheva A, Irrazabal T, Robertson SJ, et al. 2014. Gut microbial metabolism
Ahn J, Schatzkin A, Lacey JV, Jr., et al. 2007. Adiposity, adult weight change,
drives transformation of MSH2-​ deficient colon epithelial cells. Cell,
and postmenopausal breast cancer risk. Arch Intern Med, 167(19), 2091–​
158(2), 288–​299. PMID: 25036629.
2102. PMID: 17954804.
Berkey CS, Frazier AL, Gardner JD, and Colditz GA. 1999. Adolescence and
Albanes D. 1998. Height, early energy intake, and cancer:  evidence mounts
breast carcinoma risk. Cancer, 85(11), 2400–​2409. PMID: 10357411.
for the relation of energy intake to adult malignancies. BMJ, 317(7169),
Birks S, Peeters A, Backholer K, O’Brien P, and Brown W. 2012. A systematic
1331–​1332. PMCID: PMCPmc1114242.
review of the impact of weight loss on cancer incidence and mortality.
Albanes D, and Winick M. 1988. Are cell number and cell prolifera-
Obes Rev, 13(10), 868–​891. PMID: 22672203.
tion risk factors for cancer? J Natl Cancer Inst, 80(10), 772–​ 774.
Birt DF, Kris ES, Choe M, and Pelling JC. 1992. Dietary energy and fat
PMID: 3385783.
effects on tumor promotion. Cancer Res, 52(7 Suppl), 2035s-​2039s.
Aleksandrova K, Boeing H, Jenab M, et  al. 2012. Total and high-​molecular
PMID: 1544138.
weight adiponectin and risk of colorectal cancer: the European Prospective
Blitzer PH, Blitzer EC, and Rimm AA. 1976. Association between teen-​age
Investigation into Cancer and Nutrition Study. Carcinogenesis, 33(6),
obesity and cancer in 56,111 women: all cancers and endometrial carci-
1211–​1218. PMCID: PMC3388489.
noma. Prev Med, 5(1), 20–​31. PMID: 1264967.
Aleksandrova K, Boeing H, Nothlings U, et al. 2014. Inflammatory and meta-
Blumberg R, and Powrie F. 2012. Microbiota, disease, and back to health:  a
bolic biomarkers and risk of liver and biliary tract cancer. Hepatology,
metastable journey. Sci Transl Med, 4(137), 137rv137. PMID: 22674557.
60(3), 858–​871. PMCID: PMC4231978.
Boissonneault GA, Elson CE, and Pariza MW. 1986. Net energy effects of
Aleksandrova K, Pischon T, Buijsse B, et al. 2013. Adult weight change and risk
dietary fat on chemically induced mammary carcinogenesis in F344 rats.
of colorectal cancer in the European Prospective Investigation into Cancer
J Natl Cancer Inst, 76(2), 335–​338. PMID: 3080637.
and Nutrition. Eur J Cancer, 49(16), 3526–​3536. PMID: 23867126.
Borena W, Edlinger M, Bjorge T, et  al. 2014. A  prospective study on meta-
Allin KH, Nordestgaard BG, Zacho J, Tybjaerg-​Hansen A, and Bojesen SE.
bolic risk factors and gallbladder cancer in the metabolic syndrome
2010. C-​reactive protein and the risk of cancer: a mendelian randomiza-
and cancer (Me-​ Can) collaborative study. PLoS One, 9(2), e89368.
tion study. J Natl Cancer Inst, 102(3), 202–​206. PMID: 20056955.
PMCID: PMC3931760.
Alsaker MD, Janszky I, Opdahl S, Vatten LJ, and Romundstad PR. 2013.
Brestoff JR, and Artis D. 2013. Commensal bacteria at the interface of host
Weight change in adulthood and risk of postmenopausal breast can-
metabolism and the immune system. Nat Immunol, 14(7), 676–​ 684.
cer:  the HUNT study of Norway. Br J Cancer, 109(5), 1310–​1317.
PMCID: PMC4013146.
PMCID: PMC3778278.
Byers T, and Sedjo RL. 2011. Does intentional weight loss reduce cancer risk?
Arano T, Nakagawa H, Tateishi R, et al. 2011. Serum level of adiponectin and
Diabetes Obes Metab, 13(12), 1063–​1072. PMID: 21733057.
the risk of liver cancer development in chronic hepatitis C patients. Int J
Caldwell SH, Crespo DM, Kang HS, and Al-​Osaimi AM. 2004. Obesity and
Cancer, 129(9), 2226–​2235. PMID: 21170963.
hepatocellular carcinoma. Gastroenterology, 127(5 Suppl 1), S97–​103.
Arnold M, Pandeya N, Byrnes G, et al. 2015. Global burden of cancer attribut-
PMID: 15508109.
able to high body-​mass index in 2012: a population-​based study. Lancet
Calle EE, Rodriguez C, Walker-​Thurmond K, and Thun MJ. 2003. Overweight,
Oncol, 16(1), 36–​46. PMCID: PMC4314462.
obesity, and mortality from cancer in a prospectively studied cohort of
Aron-​Wisnewsky J, Dore J, and Clement K. 2012. The importance of the gut
U.S. adults. N Engl J Med, 348(17), 1625–​1638. PMID: 12711737.
microbiota after bariatric surgery. Nat Rev Gastroenterol Hepatol, 9(10),
Camargo MC, Rabkin CS, Appel N, and Hollenbeck AR. 2015. Variable associ-
590–​598. PMID: 22926153.
ation of height with gastric cancer by anatomical subsite. Cancer Causes
Aune D, Navarro Rosenblatt DA, Chan DS, et al. 2015a. Anthropometric factors
Control, 26(9), 1361. PMCID: PMCPmc4545309.
and ovarian cancer risk: a systematic review and nonlinear dose-​response
Campbell KL, Foster-​Schubert KE, Alfano CM, et al. 2012. Reduced-​calorie
meta-​analysis of prospective studies. Int J Cancer, 136(8), 1888–​1898.
dietary weight loss, exercise, and sex hormones in postmenopausal
PMID: 25250505.
women:  randomized controlled trial. J Clin Oncol, 30(19), 2314–​2326.
Aune D, Navarro Rosenblatt DA, Chan DS, et al. 2015b. Anthropometric fac-
PMCID: PMC3675691.
tors and endometrial cancer risk: a systematic review and dose-​response
Campbell PT, Cotterchio M, Dicks E, et  al. 2007. Excess body weight and
meta-​ analysis of prospective studies. Ann Oncol, 26(8), 1635–​ 1648.
colorectal cancer risk in Canada: associations in subgroups of clinically
PMID: 25791635.
defined familial risk of cancer. Cancer Epidemiol Biomarkers Prev, 16(9),
Aune D, Vieira AR, Chan DS, et al. 2012. Height and pancreatic cancer risk: a
1735–​1744. PMID: 17855691.
systematic review and meta-​analysis of cohort studies. Cancer Causes
Canchola AJ, Chang ET, Bernstein L, et al. 2010a. Body size and the risk of ovar-
Control, 23(8), 1213–​1222. PMID: 22689322.
ian cancer by hormone therapy use in the California Teachers Study cohort.
Autier P, Koechlin A, Boniol M, et al. 2013. Serum insulin and C-​peptide con-
Cancer Causes Control, 21(12), 2241–​2248. PMCID: PMC3120052.
centration and breast cancer:  a meta-​analysis. Cancer Causes Control,
Canchola AJ, Chang ET, Bernstein L, et al. 2010b. Body size and the risk of
24(5), 873–​883. PMID: 23408243.
endometrial cancer by hormone therapy use in postmenopausal women
368

368 PART III:  THE CAUSES OF CANCER


in the California Teachers Study cohort. Cancer Causes Control, 21(9), Clasey JL, Bouchard C, Teates CD, et  al. 1999. The use of anthropometric
1407–​1416. PMCID: PMC2925506. and dual-​energy X-​ray absorptiometry (DXA) measures to estimate total
Cao Y, and Ma J. 2011. Body mass index, prostate cancer-​specific mortality, abdominal and abdominal visceral fat in men and women. Obes Res, 7(3),
and biochemical recurrence:  a systematic review and meta-​ analysis. 256–​264. PMID: 10348496.
Cancer Prev Res (Phila), 4(4), 486–​501. PMCID: PMC3071449. Clendenen TV, Koenig KL, Shore RE, et  al. 2009. Postmenopausal lev-
Caprio S, Hyman LD, Limb C, et al. 1995. Central adiposity and its metabolic els of endogenous sex hormones and risk of colorectal cancer. Cancer
correlates in obese adolescent girls. Am J Physiol, 269(1 Pt 1), E118–​126. Epidemiol Biomarkers Prev, 18(1), 275–​281. PMCID: PMC2682428.
PMID: 7631766. Cnop M, Havel PJ, Utzschneider KM, et al. 2003. Relationship of adiponectin
Castellarin M, Warren RL, Freeman JD, et al. 2012. Fusobacterium nucleatum to body fat distribution, insulin sensitivity and plasma lipoproteins: evi-
infection is prevalent in human colorectal carcinoma. Genome Res, 22(2), dence for independent roles of age and sex. Diabetologia, 46(4), 459–​
299–​306. PMCID: PMC3266037. 469. PMID: 12687327.
Cavusoglu E, Ruwende C, Chopra V, et al. 2006. Adiponectin is an independent Cohen JC, Horton JD, and Hobbs HH. 2011. Human fatty liver dis-
predictor of all-​cause mortality, cardiac mortality, and myocardial infarc- ease:  old questions and new insights. Science, 332(6037), 1519–​1523.
tion in patients presenting with chest pain. Eur Heart J, 27(19), 2300–​ PMCID: PMC3229276.
2309. PMID: 16864609. Colditz GA. 1998. Relationship between estrogen levels, use of hormone
Cena H, Fonte ML, and Turconi G. 2011. Relationship between smoking and replacement therapy, and breast cancer. J Natl Cancer Inst, 90(11), 814–​
metabolic syndrome. Nutr Rev, 69(12), 745–​753. PMID: 22133198. 823. PMID: 9625169.
Centers for Disease Control and Prevention. 2015. Defining Childhood Obesity. Colleoni M, Li S, Gelber RD, et al. 2005. Relation between chemotherapy dose,
Available from:  http://​www.cdc.gov/​obesity/​childhood/​defining.html. oestrogen receptor expression, and body-​mass index. Lancet, 366(9491),
Accessed Jan 1, 2016. 1108–​1110. PMID: 16182899.
Chamberlain C, Romundstad P, Vatten L, Gunnell D, and Martin RM. 2011. Cox LM, and Blaser MJ. 2013. Pathways in microbe-​induced obesity. Cell
The association of weight gain during adulthood with prostate cancer Metab, 17(6), 883–​894. PMCID: PMC3727904.
incidence and survival: a population-​based cohort. Int J Cancer, 129(5), Crave JC, Lejeune H, Brebant C, Baret C, and Pugeat M. 1995. Differential
1199–​1206. PMID: 21064096. effects of insulin and insulin-​like growth factor I  on the production of
Chan DS, Bandera EV, Greenwood DC, and Norat T. 2015. Circulating C-​ plasma steroid-​binding globulins by human hepatoblastoma-​derived (Hep
reactive protein and breast cancer risk-​ systematic literature review G2) cells. J Clin Endocrinol Metab, 80(4), 1283–​1289. PMID: 7536204.
and meta-​ analysis of prospective cohort studies. Cancer Epidemiol Cuevas-​Ramos G, Petit CR, Marcq I, et al. 2010. Escherichia coli induces DNA
Biomarkers Prev, 24(10), 1439–​1449. PMID: 26224798. damage in vivo and triggers genomic instability in mammalian cells.
Chan DS, Vieira AR, Aune D, et  al. 2014. Body mass index and survival PNAS, 107(25), 11537–​11542. PMCID: PMC2895108.
in women with breast cancer-​ systematic literature review and meta-​ Cunningham SA, Kramer MR, and Narayan KM. 2014. Incidence of child-
analysis of 82 follow-​ up studies. Ann Oncol, 25(10), 1901–​ 1914. hood obesity in the United States. N Engl J Med, 370(5), 403–​411.
PMCID: PMC4176449. PMCID: PMC4017620.
Chan JM, Stampfer MJ, Ma J, et al. 2002. Insulin-​like growth factor-​I (IGF-​I) Cust AE, Kaaks R, Friedenreich C, et  al. 2007. Plasma adiponectin levels
and IGF binding protein-​3 as predictors of advanced-​stage prostate can- and endometrial cancer risk in pre-​and postmenopausal women. J Clin
cer. J Natl Cancer Inst, 94(14), 1099–​1106. PMID: 12122101. Endocrinol Metab, 92(1), 255–​263. PMID: 17062769.
Chandler PD, Buring JE, Manson JE, et al. 2015. Association between plasma Cust AE, Stocks T, Lukanova A, et  al. 2009. The influence of overweight
adiponectin levels and colorectal cancer risk in women. Cancer Causes and insulin resistance on breast cancer risk and tumour stage at diag-
Control, 26(7), 1047–​1052. PMCID: PMC4466025. nosis:  a prospective study. Breast Cancer Res Treat, 113(3), 567–​576.
Chang SC, Lacey JV, Jr., Brinton LA, et al. 2007. Lifetime weight history and PMID: 18330696.
endometrial cancer risk by type of menopausal hormone use in the NIH-​ Dal Maso L, Tavani A, Zucchetto A, et al. 2011. Anthropometric measures at
AARP diet and health study. Cancer Epidemiol Biomarkers Prev, 16(4), different ages and endometrial cancer risk. Br J Cancer, 104(7), 1207–​
723–​730. PMID: 17416763. 1213. PMCID: PMC3068501.
Chen CL, Yang WS, Yang HI, et al. 2014. Plasma adipokines and risk of hepa- Dallal CM, Brinton LA, Bauer DC, et al. 2013. Obesity-​related hormones and
tocellular carcinoma in chronic hepatitis B virus-​ infected carriers:  a endometrial cancer among postmenopausal women: a nested case-​control
prospective study in Taiwan. Cancer Epidemiol Biomarkers Prev, 23(8), study within the B~FIT cohort. Endocr Relat Cancer, 20(1), 151–​160.
1659–​1671. PMID: 24895413. PMCID: PMC4038326.
Chen L, Li L, Wang Y, et  al. 2013. Circulating C-​peptide level is a predic- Dastani Z, Hivert MF, Timpson N, et al. 2012. Novel loci for adiponectin lev-
tive factor for colorectal neoplasia:  evidence from the meta-​ analysis els and their influence on type 2 diabetes and metabolic traits: a multi-​
of prospective studies. Cancer Causes Control, 24(10), 1837–​ 1847. ethnic meta-​analysis of 45,891 individuals. PLoS Genet, 8(3), e1002607.
PMID: 23846284. PMCID: PMC3315470.
Chen Q, Wang J, Yang J, et al. 2014. Association between adult weight gain and de Waard F, and Baanders-​van Halewijn EA. 1974. A  prospective study in
colorectal cancer: a dose-​response meta-​analysis of observational studies. general practice on breast-​cancer risk in postmenopausal women. Int J
Int J Cancer, 136(12), 2880–​2889. PMID: 25395274. Cancer, 14(2), 153–​160. PMID: 4378317.
Chiolero A, Faeh D, Paccaud F, and Cornuz J. 2008. Consequences of smoking Demark-​Wahnefried W, Platz EA, Ligibel JA, et al. 2012. The role of obesity
for body weight, body fat distribution, and insulin resistance. Am J Clin in cancer survival and recurrence. Cancer Epidemiol Biomarkers Prev,
Nutr, 87(4), 801–​809. PMID: 18400700. 21(8), 1244–​1259. PMCID: PMC3415558.
Chlebowski RT, Wactawski-​Wende J, Ritenbaugh C, et al. 2004. Estrogen plus Despres JP. 2006. Abdominal obesity:  the most prevalent cause of the met-
progestin and colorectal cancer in postmenopausal women. N Engl J Med, abolic syndrome and related cardiometabolic risk. Eur Heart J, 8(B),
350(10), 991–​1004. PMID: 14999111. B4–​B12.
Choi Y, Park B, Jeong BC, et al. 2013. Body mass index and survival in patients Deurenberg P, Deurenberg-​Yap M, and Guricci S. 2002. Asians are different
with renal cell carcinoma: a clinical-​based cohort and meta-​analysis. Int J from Caucasians and from each other in their body mass index/​body fat
Cancer, 132(3), 625–​634. PMID: 22610826. per cent relationship. Obes Rev, 3(3), 141–​146. PMID: 12164465.
Chong DQ, Mehta RS, Song M et  al. 2015. Prediagnostic plasma adiponec- Dhillon PK, Penney KL, Schumacher F, et al. 2011. Common polymorphisms
tin and survival among patients with colorectal cancer. Cancer Prev Res in the adiponectin and its receptor genes, adiponectin levels and the risk
(Phila), 8(12), 1138–​1145. PMCID: PMC4670783. of prostate cancer. Cancer Epidemiol Biomarkers Prev, 20(12), 2618–​
Chow WH, Dong LM, and Devesa SS. 2010. Epidemiology and risk 2627. PMCID: PMC3700351.
factors for kidney cancer. Nature Rev Urology, 7(5), 245–​ 257. Donohoe CL, Doyle SL, and Reynolds JV. 2011. Visceral adiposity,
PMCID: PMC3012455. insulin resistance and cancer risk. Diabetol Metab Syndr, 3, 12.
Chow WH, Gridley G, Fraumeni JF, Jr., and Jarvholm B. 2000. Obesity, hyper- PMCID: PMC3145556.
tension, and the risk of kidney cancer in men. N Engl J Med, 343(18), Donohoe DR, Holley D, Collins LB, et al. 2014. A gnotobiotic mouse model
1305–​1311. PMID: 11058675. demonstrates that dietary fiber protects against colorectal tumorigenesis
Christou NV, Lieberman M, Sampalis F, and Sampalis JS. 2008. Bariatric sur- in a microbiota-​and butyrate-​dependent manner. Cancer Discov, 4(12),
gery reduces cancer risk in morbidly obese patients. Surg Obes Relat Dis, 1387–​1397. PMCID: PMC4258155.
4(6), 691–​695. PMID: 19026373. Dougan MM, Hankinson SE, Vivo ID, et al. 2015. Prospective study of body
Christou NV, Sampalis JS, Liberman M, et al. 2004. Surgery decreases long-​ size throughout the life-​course and the incidence of endometrial cancer
term mortality, morbidity, and health care use in morbidly obese patients. among premenopausal and postmenopausal women. Int J Cancer, 137(3),
Ann Surg, 240(3), 416–​423; discussion 423–​414. PMCID: PMC1356432. 625–​637. PMID: 25641700.
 369

Obesity and Body Composition 369


Eliassen AH, Colditz GA, Rosner B, Willett WC, and Hankinson SE. 2006. Fryar CD, Carroll MD, and Ogden CL. 2014b. Prevalence of overweight,
Adult weight change and risk of postmenopausal breast cancer. JAMA, obesity, and extreme obesity among adults:  United States, 1960–​1962
296(2), 193–​201. PMID: 16835425. through 2011–​2012. Atlanta, GA: CDC.
Eliassen AH, Tworoger SS, Mantzoros CS, Pollak MN, and Hankinson SE. Fujisawa T, Endo H, Tomimoto A, et al. 2008. Adiponectin suppresses colorec-
2007. Circulating insulin and c-​peptide levels and risk of breast can- tal carcinogenesis under the high-​fat diet condition. Gut, 57(11), 1531–​
cer among predominately premenopausal women. Cancer Epidemiol 1538. PMCID: PMC2582344.
Biomarkers Prev, 16(1), 161–​164. PMID: 17220346. Fung TT, Hu FB, Schulze M, et al. 2012. A dietary pattern that is associated
Endogenous Hormones Breast Cancer Collaborative Group. 2010. Insulin-​like with C-​peptide and risk of colorectal cancer in women. Cancer Causes
growth factor 1 (IGF1), IGF binding protein 3 (IGFBP3), and breast can- Control, 23(6), 959–​965. PMCID: PMC3572718.
cer risk: pooled individual data analysis of 17 prospective studies. Lancet Furet JP, Kong LC, Tap J, et  al. 2010. Differential adaptation of human gut
Oncol, 11(6), 530–​542. PMCID: PMC3113287. microbiota to bariatric surgery-​induced weight loss: links with metabolic
Endogenous Hormones Breast Cancer Collaborative Group. 2011. and low-​ grade inflammation markers. Diabetes, 59(12), 3049–​ 3057.
Circulating sex hormones and breast cancer risk factors in postmeno- PMCID: PMC2992765.
pausal women: reanalysis of 13 studies. Br J Cancer, 105(5), 709–​722. Gallagher D, Visser M, Sepulveda D, et  al. 1996. How useful is body mass
PMCID: PMC3188939. index for comparison of body fatness across age, sex, and ethnic groups?
Esposito K, Pontillo A, Di Palo C, et al. 2003. Effect of weight loss and lifestyle Am J Epidemiol, 143(3), 228–​239. PMID: WOS:A1996TR34900003.
changes on vascular inflammatory markers in obese women: a random- Gao C, Patel CJ, Michailidou K, et al. 2016. Mendelian randomization study
ized trial. JAMA, 289(14), 1799–​1804. PMID: 12684358. of adiposity-​related traits and risk of breast, ovarian, prostate, lung and
Fabian CJ, Kimler BF, Donnelly JE, et  al. 2013. Favorable modulation of colorectal cancer. Int J Epidemiol, 45(3), 896–​908. PMID: 27427428.
benign breast tissue and serum risk biomarkers is associated with > 10% Garn SM, Leonard WR, and Hawthorne VM. 1986. Three limitations of the
weight loss in postmenopausal women. Breast Cancer Res Treat, 142(1), body mass index. Am J Clin Nutr, 44(6), 996–​997. PMID: 3788846.
119–​132. PMCID: PMC3921968. Gaudet MM, Falk RT, Gierach GL, et  al. 2010. Do adipokines underlie the
Fagherazzi G, Guillas G, Boutron-​Ruault MC, Clavel-​Chapelon F, and Mesrine association between known risk factors and breast cancer among a
S. 2013. Body shape throughout life and the risk for breast cancer at cohort of United States women? Cancer Epidemiol, 34(5), 580–​586.
adulthood in the French E3N cohort. Eur J Cancer Prev, 22(1), 29–​37. PMCID: PMC2949443.
PMID: 22694827. Geer EB, and Shen W. 2009. Gender differences in insulin resistance, body
Fahey PP, Mallitt KA, Astell-​Burt T, Stone G, and Whiteman DC. 2015. Impact composition, and energy balance. Gend Med, 6 Suppl 1, 60–​ 75.
of pre-​diagnosis behavior on risk of death from esophageal cancer: a sys- PMCID: PMC2908522.
tematic review and meta-​analysis. Cancer Causes Control, 26(10), 1365–​ Genkinger JM, Spiegelman D, Anderson KE, et al. 2011. A pooled analysis of
1373. PMID: 26210679. 14 cohort studies of anthropometric factors and pancreatic cancer risk. Int
Fairfield KM, Willett WC, Rosner BA, et al. 2002. Obesity, weight gain, and J Cancer, 129(7), 1708–​1717. PMCID: PMC3073156.
ovarian cancer. Obstet Gynecol, 100(2), 288–​296. PMID: 12151152. Giovannucci E. 2007. Metabolic syndrome, hyperinsulinemia, and colon can-
Faith JJ, Guruge JL, Charbonneau M, et  al. 2013. The long-​ term stabil- cer: a review. Am J Clin Nutr, 86(3), s836–​842. PMID: 18265477.
ity of the human gut microbiota. Science, 341(6141), 1237439. Giovannucci E, and Michaud D. 2007. The role of obesity and related met-
PMCID: PMC3791589. abolic disturbances in cancers of the colon, prostate, and pancreas.
Feigelson HS, Jonas CR, Teras LR, Thun MJ, and Calle EE. 2004. Weight gain, Gastroenterology, 132(6), 2208–​2225. PMID: 17498513.
body mass index, hormone replacement therapy, and postmenopausal Giovannucci E, Rimm EB, Liu Y, and Willett WC. 2004. Height, predictors
breast cancer in a large prospective study. Cancer Epidemiol Biomarkers of C-​peptide and cancer risk in men. Int J Epidemiol, 33(1), 217–​225.
Prev, 13(2), 220–​224. PMID: 14973094. PMID: 15075172.
Ferlay J, Bray F, Steliarova-​Foucher E, and Forman D. 2014. Cancer incidence Giovannucci E, Rimm EB, Stampfer MJ, Colditz GA, and Willett WC. 1997.
in five continents, CI5plus. IARC CancerBase No. 9. Lyon: International Height, body weight, and risk of prostate cancer. Cancer Epidemiol
Agency for Research on Cancer. Biomarkers Prev, 6(8), 557–​563. PMID: 9264267.
Field KM, Kosmider S, Jefford M, et al. 2008. Chemotherapy dosing strategies Green J, Cairns BJ, Casabonne D, et  al. 2011. Height and cancer incidence
in the obese, elderly, and thin patient: results of a nationwide survey. J in the Million Women Study:  prospective cohort, and meta-​analysis of
Oncol Pract, 4(3), 108–​113. PMCID: PMC2794002. prospective studies of height and total cancer risk. Lancet Oncol, 12(8),
Flegal KM, Carroll MD, Kit BK, and Ogden CL. 2012. Prevalence of obesity 785–​794. PMCID: PMC3148429.
and trends in the distribution of body mass index among US adults, 1999–​ Griggs JJ, Culakova E, Sorbero ME, et  al. 2007. Effect of patient socioeco-
2010. JAMA, 307(5), 491–​497. PMID: 22253363. nomic status and body mass index on the quality of breast cancer adjuvant
Flegal KM, Carroll MD, Ogden CL, and Curtin LR. 2010. Prevalence and chemotherapy. J Clin Oncol, 25(3), 277–​284. PMID: 17159190.
trends in obesity among US adults, 1999–​2008. JAMA, 303(3), 235–​241. Griggs JJ, Mangu PB, Anderson H, et al. 2012. Appropriate chemotherapy dos-
PMID: 20071471. ing for obese adult patients with cancer:  American Society of Clinical
Flegal KM, Carroll MD, Ogden CL, and Johnson CL. 2002. Prevalence and Oncology clinical practice guideline. J Clin Oncol, 30(13), 1553–​1561.
trends in obesity among US adults, 1999–​2000. JAMA, 288(14), 1723–​ PMID: 22473167.
1727. PMID: 12365955. Griggs JJ, and Sabel MS. 2008. Obesity and cancer treatment:  weighing the
Flegal KM, Kruszon-​Moran D, Carroll MD, Fryar CD, and Ogden CL. 2016. evidence. J Clin Oncol, 26(25), 4060–​4062. PMID: 18757320.
Trends in obesity among adults in the United States, 2005 to 2014. JAMA, Griggs JJ, Sorbero ME, and Lyman GH. 2005. Undertreatment of obese women
315(21), 2284–​2291. PMID: 27272580. receiving breast cancer chemotherapy. Arch Intern Med, 165(11), 1267–​
Flum DR, and Dellinger EP. 2004. Impact of gastric bypass operation on sur- 1273. PMID: 15956006.
vival:  a population-​based analysis. J Am Coll Surg, 199(4), 543–​551. Grundy SM. 1999. Hypertriglyceridemia, insulin resistance, and the metabolic
PMID: 15454136. syndrome. Am J Cardiol, 83(9B), 25F–​29F. PMID: 10357572.
Fowler JA, Lwin ST, Drake MT, et  al. 2011. Host-​ derived adiponec- Gu J, Yan S, Wang B, et al. 2015. Type 2 diabetes mellitus and risk of gallblad-
tin is tumor-​ suppressive and a novel therapeutic target for multiple der cancer: a systematic review and meta-​analysis of observational stud-
myeloma and the associated bone disease. Blood, 118(22), 5872–​5882. ies. Diabetes Metab Res Rev, 32(1), 63–​72. PMID: 26111736.
PMCID: PMC3228502. Guarner F, and Malagelada JR. 2003. Gut flora in health and disease. Lancet,
Freireich EJ, Gehan EA, Rall DP, Schmidt LH, and Skipper HE. 1966. Quantitative 361(9356), 512–​519. PMID: 12583961.
comparison of toxicity of anticancer agents in mouse, rat, hamster, dog, mon- Guerra L, Guidi R, and Frisan T. 2011. Do bacterial genotoxins contribute to
key, and man. Cancer Chemother Rep, 50(4), 219–​244. PMID: 4957125. chronic inflammation, genomic instability and tumor progression? Febs J,
Friedenberg FK, Xanthopoulos M, Foster GD, and Richter JE. 2008. The 278(23), 4577–​4588. PMID: 21585655.
association between gastroesophageal reflux disease and obesity. Am J Gunnell D, Okasha M, Smith GD, et al. 2001. Height, leg length, and cancer risk: a
Gastroenterol, 103(8), 2111–​2122. PMID: 18796104. systematic review. Epidemiol Rev, 23(2), 313–​342. PMID: 12192740.
Friedenreich C, Cust A, Lahmann PH, et al. 2007. Anthropometric factors and risk Gunter MJ, Hoover DR, Yu H, et al. 2008. Insulin, insulin-​like growth factor-​I,
of endometrial cancer: the European prospective investigation into cancer endogenous estradiol, and risk of colorectal cancer in postmenopausal
and nutrition. Cancer Causes Control, 18(4), 399–​413. PMID: 17297555. women. Cancer Res, 68(1), 329–​337. PMCID: PMC4225702.
Fryar CD, Carroll MD, and Ogden CL. 2014a. Prevalence of overweight and Gunter MJ, Hoover DR, Yu H, et al. 2009. Insulin, insulin-​like growth factor-​I,
obesity among children and adolescents:  United States, 1963–​ 1965 and risk of breast cancer in postmenopausal women. J Natl Cancer Inst,
through 2011–​2012. Atlanta, GA: CDC. 101(1), 48–​60. PMCID: PMC2639294.
370

370 PART III:  THE CAUSES OF CANCER


Gunter MJ, Wang T, Cushman M, et  al. 2015. Circulating adipokines and Jing Z, Hou X, Liu Y, et  al. 2015. Association between height and thyroid
inflammatory markers and postmenopausal breast cancer risk. J Natl cancer risk: a meta-​analysis of prospective cohort studies. Int J Cancer,
Cancer Inst, 107(9). PMCID: PMC4651104. 137(6), 1484–​1490. PMID: 25693727.
Hakimi AA, Furberg H, Zabor EC, et al. 2013. An epidemiologic and genomic Jonsson F, Wolk A, Pedersen NL, et al. 2003. Obesity and hormone-​dependent
investigation into the obesity paradox in renal cell carcinoma. J Natl tumors:  cohort and co-​twin control studies based on the Swedish Twin
Cancer Inst, 105(24), 1862–​1870. PMCID: PMC3866155. Registry. Int J Cancer, 106(4), 594–​599. PMID: 12845658.
Han X, Stevens J, Truesdale KP, et al. 2014. Body mass index at early adult- Kaaks R, Bellati C, Venturelli E, et  al. 2003. Effects of dietary intervention
hood, subsequent weight change and cancer incidence and mortality. Int J on IGF-​I and IGF-​binding proteins, and related alterations in sex steroid
Cancer, 135(12), 2900–​2909. PMCID: PMC4192093. metabolism: the Diet and Androgens (DIANA) Randomised Trial. Eur J
Hankinson SE, Willett WC, Colditz GA, et  al. 1998. Circulating concentra- Clin Nutr, 57(9), 1079–​1088. PMID: 12947426.
tions of insulin-​like growth factor-​I and risk of breast cancer. Lancet, Kaaks R, Johnson T, Tikk K, et  al. 2014. Insulin-​like growth factor I  and
351(9113), 1393–​1396. PMID: 9593409. risk of breast cancer by age and hormone receptor status:  a prospec-
Harvie M, Howell A, Vierkant RA, et al. 2005. Association of gain and loss of tive study within the EPIC cohort. Int J Cancer, 134(11), 2683–​2690.
weight before and after menopause with risk of postmenopausal breast PMID: 24248481.
cancer in the Iowa women’s health study. Cancer Epidemiol Biomarkers Kaaks R, Lukanova A, and Kurzer MS. 2002a. Obesity, endogenous hormones,
Prev, 14(3), 656–​661. PMID: 15767346. and endometrial cancer risk:  a synthetic review. Cancer Epidemiol
Harvie MN, Pegington M, Mattson MP, et  al. 2011. The effects of intermit- Biomarkers Prev, 11(12), 1531–​1543. PMID: 12496040.
tent or continuous energy restriction on weight loss and metabolic disease Kaaks R, Lundin E, Rinaldi S, et al. 2002b. Prospective study of IGF-​I, IGF-​
risk markers: a randomized trial in young overweight women. Int J Obes binding proteins, and breast cancer risk, in northern and southern Sweden.
(Lond), 35(5), 714–​727. PMCID: PMC3017674. Cancer Causes Control, 13(4), 307–​316. PMID: 12074500.
Hassan MM, Abdel-​Wahab R, Kaseb A, et al. 2015. Obesity early in adulthood Kaaks R, Rinaldi S, Key TJ, et  al. 2005. Postmenopausal serum androgens,
increases risk but does not affect outcomes of hepatocellular carcinoma. oestrogens and breast cancer risk:  the European prospective investiga-
Gastroenterology, 149(1), 119–​129. PMID: 25836985. tion into cancer and nutrition. Endocr Relat Cancer, 12(4), 1071–​1082.
Heidemann C, Sun Q, van Dam RM, et  al. 2008. Total and high-​molecular-​ PMID: 16322344.
weight adiponectin and resistin in relation to the risk for type 2 diabetes in Kabat GC, Kim M, Caan BJ, et al. 2009. Repeated measures of serum glucose
women. Ann Intern Med, 149(5), 307–​316. PMCID: PMC3874083. and insulin in relation to postmenopausal breast cancer. Int J Cancer,
Hilakivi-​Clarke L, Forsen T, Eriksson JG, et al. 2001. Tallness and overweight 125(11), 2704–​2710. PMID: 19588485.
during childhood have opposing effects on breast cancer risk. Br J Kabat GC, Xue X, Kamensky V, et  al. 2015. Risk of breast, endometrial,
Cancer, 85(11), 1680–​1684. PMCID: PMC2363976. colorectal, and renal cancers in postmenopausal women in association
Ho GY, Wang T, Gunter MJ, et  al. 2012. Adipokines linking obesity with with a body shape index and other anthropometric measures. Cancer
colorectal cancer risk in postmenopausal women. Cancer Res, 72(12), Causes Control, 26(2), 219–​229. PMID: 25430815.
3029–​3037. PMCID: PMC3790260. Kadowaki T, Yamauchi T, Kubota N, et al. 2006. Adiponectin and adiponectin
Hofmann JN, Liao LM, Pollak MN, et al. 2012. A prospective study of circu- receptors in insulin resistance, diabetes, and the metabolic syndrome. J
lating adipokine levels and risk of multiple myeloma. Blood, 120(22), Clin Invest, 116(7), 1784–​1792. PMCID: PMC1483172.
4418–​4420. PMCID: PMC3507148. Kadowaki T, Yamauchi T, Okada-​Iwabu M, and Iwabu M. 2014. Adiponectin
Hong L, Zhang H, Zhao Q, et al. 2013. Relation of excess body weight and sur- and its receptors: implications for obesity-​associated diseases and longev-
vival in patients with esophageal adenocarcinoma: a meta-​analysis. Dis ity. Lancet Diabetes Endocrinol, 2(1), 8–​9. PMID: 24622657.
Esophagus, 26(6), 623–​627. PMID: 23317016. Kapoor D, Malkin CJ, Channer KS, and Jones TH. 2005. Androgens, insu-
Hu B, Elinav E, Huber S, et  al. 2013. Microbiota-​ induced activation of lin resistance and vascular disease in men. Clin Endocrinol (Oxf), 63(3),
epithelial IL-​ 6 signaling links inflammasome-​ driven inflamma- 239–​250. PMID: 16117808.
tion with transmissible cancer. PNAS, 110(24), 9862–​ 9867. PMID: Karahalios A, English DR, and Simpson JA. 2015. Weight change and risk of
WOS:000320930100063. colorectal cancer: a systematic review and meta-​analysis. Am J Epidemiol,
Hu FB. 2008a. Measurements of adiposity and body composition. In: Hu FB 181(11), 832–​845. PMID: 25888582.
(Ed.), Obesity epidemiology (1st ed., pp. 53–​83). New  York:  Oxford Karas MG, Benkeser D, Arnold AM, et al. 2014. Relations of plasma total and
University Press. high-​molecular-​weight adiponectin to new-​onset heart failure in adults
Hu FB. 2008b. Predictors and consequences of childhood obesity. In: Hu FB ≥65 years of age (from the Cardiovascular Health study). Am J Cardiol,
(Ed.), Obesity epidemiology (1st ed., pp. 416–​436). New  York:  Oxford 113(2), 328–​334. PMCID: PMC3968249.
University Press. Kau AL, Ahern PP, Griffin NW, Goodman AL, and Gordon JI. 2011. Human
Hughes LA, Simons CC, van den Brandt PA, et  al. 2011. Body size and nutrition, the gut microbiome and the immune system. Nature, 474(7351),
colorectal cancer risk after 16.3  years of follow-​up:  an analysis from 327–​336. PMCID: PMC3298082.
the Netherlands Cohort Study. Am J Epidemiol, 174(10), 1127–​1139. Kawai M, Minami Y, Kuriyama S, et al. 2010. Adiposity, adult weight change
PMID: 21984660. and breast cancer risk in postmenopausal Japanese women: the Miyagi
Hui JM, Kench JG, Chitturi S, et  al. 2003. Long-​term outcomes of cirrhosis Cohort Study. Br J Cancer, 103(9), 1443–​1447. PMCID: PMC2990597.
in nonalcoholic steatohepatitis compared with hepatitis C. Hepatology, Keinan-​Boker L, Bueno De Mesquita HB, Kaaks R, et  al. 2003. Circulating
38(2), 420–​427. PMID: 12883486. levels of insulin-​like growth factor I, its binding proteins -​1,-​2, -​3, C-​
Huxley R, Ansary-​ Moghaddam A, Berrington de Gonzalez A, Barzi peptide and risk of postmenopausal breast cancer. Int J Cancer, 106(1),
F, and Woodward M. 2005. Type-​ II diabetes and pancreatic can- 90–​95. PMID: 12794762.
cer:  a meta-​analysis of 36 studies. Br J Cancer, 92(11), 2076–​2083. Keum N, Greenwood DC, Lee DH, et al. 2015. Adult weight gain and adiposity-​
PMCID: PMC2361795. related cancers:  a dose-​response meta-​analysis of prospective observa-
Imayama I, Ulrich CM, Alfano CM, et al. 2012. Effects of a caloric restriction tional studies. J Natl Cancer Inst, 107(2). PMID: 25757865.
weight loss diet and exercise on inflammatory biomarkers in overweight/​ Key T, Appleby P, Barnes I, et al. 2002. Endogenous sex hormones and breast
obese postmenopausal women:  a randomized controlled trial. Cancer cancer in postmenopausal women: reanalysis of nine prospective studies.
Res, 72(9), 2314–​2326. PMCID: PMC3342840. J Natl Cancer Inst, 94(8), 606–​616. PMID: 11959894.
Inamura K, Song M, Jung S, et al. 2016. Prediagnosis plasma adiponectin in Key TJ, Appleby PN, Reeves GK, et al. 2003. Body mass index, serum sex hor-
relation to colorectal cancer risk according to KRAS mutation status. J mones, and breast cancer risk in postmenopausal women. J Natl Cancer
Natl Cancer Inst, 108(4). PMCID: PMC4668768. Inst, 95(16), 1218–​1226. PMID: 12928347.
Irrazabal T, Belcheva A, Girardin SE, Martin A, and Philpott DJ. 2014. The Key TJ, and Pike MC. 1988. The dose-​effect relationship between “unopposed”
multifaceted role of the intestinal microbiota in colon cancer. Mol Cell, oestrogens and endometrial mitotic rate:  its central role in explaining
54(2), 309–​320. PMID: 24766895. and predicting endometrial cancer risk. Br J Cancer, 57(2), 205–​212.
Isaksson B, Jonsson F, Pedersen NL, et al. 2002. Lifestyle factors and pancre- PMCID: PMC2246441.
atic cancer risk:  a cohort study from the Swedish Twin Registry. Int J Kim AY, Park YJ, Pan X, et al. 2015. Obesity-​induced DNA hypermethylation
Cancer, 98(3), 480–​482. PMID: 11920604. of the adiponectin gene mediates insulin resistance. Nat Commun, 6,
Janssen I, Heymsfield SB, Allison DB, Kotler DP, and Ross R. 2002. Body 7585. PMCID: PMC4506505.
mass index and waist circumference independently contribute to the pre- Kim JH, Shim KW, Yoon YS, et al. 2012. Cigarette smoking increases abdomi-
diction of nonabdominal, abdominal subcutaneous, and visceral fat. Am J nal and visceral obesity but not overall fatness:  an observational study.
Clin Nutr, 75(4), 683–​688. PMID: 11916754. PLoS One, 7(9), e45815. PMCID: PMC3454366.
 371

Obesity and Body Composition 371


Kistorp C, Faber J, Galatius S, et  al. 2005. Plasma adiponectin, body mass Lee J, Meyerhardt JA, Giovannucci E, and Jeon JY. 2015. Association
index, and mortality in patients with chronic heart failure. Circulation, between body mass index and prognosis of colorectal cancer:  a meta-​
112(12), 1756–​1762. PMID: 16157772. analysis of prospective cohort studies. PLoS One, 10(3), e0120706.
Kitahara CM, McCullough ML, Franceschi S, et al. 2016. Anthropometric fac- PMCID: PMC4374868.
tors and thyroid cancer risk by histological subtype: pooled analysis of Leoncini E, Ricciardi W, Cadoni G, et  al. 2014. Adult height and head and
22 prospective studies. Thyroid, 26(2), 306–​318. PMCID: PMC4754509. neck cancer: a pooled analysis within the INHANCE Consortium. Eur J
Kizer JR, Benkeser D, Arnold AM, et al. 2012. Associations of total and high-​ Epidemiol, 29(1), 35–​48. PMCID: PMCPmc4122122.
molecular-​weight adiponectin with all-​cause and cardiovascular mortality Levi F, La Vecchia C, Negri E, Parazzini F, and Franceschi S. 1992. Body mass
in older persons: the Cardiovascular Health Study. Circulation, 126(25), at different ages and subsequent endometrial cancer risk. Int J Cancer,
2951–​2961. PMCID: PMC3968250. 50(4), 567–​571. PMID: 1537622.
Kong LC, Tap J, Aron-​Wisnewsky J, et al. 2013. Gut microbiota after gastric Lew EA, and Garfinkel L. 1979. Variations in mortality by weight among
bypass in human obesity:  increased richness and associations of bac- 750,000 men and women. J Chronic Dis, 32(8), 563–​576. PMID: 468958.
terial genera with adipose tissue genes. Am J Clin Nutr, 98(1), 16–​24. Ley RE, Backhed F, Turnbaugh P, et al. 2005. Obesity alters gut microbial ecol-
PMID: 23719559. ogy. PNAS, 102(31), 11070–​11075. PMCID: PMC1176910.
Koo JH, and Leong RW. 2010. Sex differences in epidemiological, clinical and Ley RE, Turnbaugh PJ, Klein S, and Gordon JI. 2006. Microbial ecol-
pathological characteristics of colorectal cancer. J Gastroenterol Hepatol, ogy:  human gut microbes associated with obesity. Nature, 444(7122),
25(1), 33–​42. PMID: 19874446. 1022–​1023. PMID: 17183309.
Kostic AD, Chun E, Robertson L, et al. 2013. Fusobacterium nucleatum poten- Li H, Stampfer MJ, Mucci L, et al. 2010. A 25-​year prospective study of plasma
tiates intestinal tumorigenesis and modulates the tumor-​immune microen- adiponectin and leptin concentrations and prostate cancer risk and sur-
vironment. Cell Host Microbe, 14(2), 207–​215. PMCID: PMC3772512. vival. Clin Chem, 56(1), 34–​43. PMCID: PMC2858593.
Kotsopoulos J, Baer HJ, and Tworoger SS. 2010. Anthropometric measures and Li S, Shin HJ, Ding EL, and van Dam RM. 2009. Adiponectin levels and risk
risk of epithelial ovarian cancer:  results from the nurses’ health study. of type 2 diabetes: a systematic review and meta-​analysis. JAMA, 302(2),
Obesity (Silver Spring), 18(8), 1625–​1631. PMCID: PMC2888954. 179–​188. PMID: 19584347.
Krebs NF, Himes JH, Jacobson D, et al. 2007. Assessment of child and ado- Liao CY, Zhang DY, Mungo C, Tompkins DA, and Zeidan AM. 2014. Is
lescent overweight and obesity. Pediatrics, 120 Suppl 4, S193–​ 228. diabetes mellitus associated with increased incidence and disease-​
PMID: 18055652. specific mortality in endometrial cancer? A systematic review and meta-​
Krishnan K, Bassett JK, MacInnis RJ, et al. 2013. Associations between weight analysis of cohort studies. Gynecol Oncol, 135(1), 163–​171. PMID:
in early adulthood, change in weight, and breast cancer risk in postmeno- WOS:000343955900028.
pausal women. Cancer Epidemiol Biomarkers Prev, 22(8), 1409–​1416. Lin JH, Zhang SM, Rexrode KM, et  al. 2013. Association between sex hor-
PMID: 23720403. mones and colorectal cancer risk in men and women. Clin Gastroenterol
Kuchiba A, Morikawa T, Yamauchi M, et  al. 2012. Body mass index and Hepatol, 11(4), 419–​424 e411. PMCID: PMC3594467.
risk of colorectal cancer according to fatty acid synthase expres- Ling H, Waterworth DM, Stirnadel HA, et  al. 2009. Genome-​ wide link-
sion in the nurses’ health study. J Natl Cancer Inst, 104(5), 415–​420. age and association analyses to identify genes influencing adiponec-
PMCID: PMC3295745. tin levels:  the GEMS Study. Obesity (Silver Spring), 17(4), 737–​744.
Laake I, Thune I, Selmer R, et al. 2010. A prospective study of body mass index, PMCID: PMC4028785.
weight change, and risk of cancer in the proximal and distal colon. Cancer Littman AJ, White E, and Kristal AR. 2007. Anthropometrics and prostate can-
Epidemiol Biomarkers Prev, 19(6), 1511–​1522. PMID: 20501754. cer risk. Am J Epidemiol, 165(11), 1271–​1279. PMID: 17395597.
Lacey JV, Jr., Swanson CA, Brinton LA, et al. 2003. Obesity as a potential risk Lofgren IE, Herron KL, West KL, et al. 2005. Weight loss favorably modifies
factor for adenocarcinomas and squamous cell carcinomas of the uterine anthropometrics and reverses the metabolic syndrome in premenopausal
cervix. Cancer, 98(4), 814–​821. PMID: 12910527. women. J Am Coll Nutr, 24(6), 486–​493. PMID: 16373945.
Lagergren J, Bergstrom R, and Nyren O. 1999. Association between body mass Luhn P, Dallal CM, Weiss JM, et  al. 2013. Circulating adipokine levels and
and adenocarcinoma of the esophagus and gastric cardia. Ann Intern Med, endometrial cancer risk in the prostate, lung, colorectal, and ovarian
130(11), 883–​890. PMID: 10375336. cancer screening trial. Cancer Epidemiol Biomarkers Prev, 22(7), 1304–​
Lahmann PH, Cust AE, Friedenreich CM, et  al. 2010. Anthropometric mea- 1312. PMCID: PMC3819202.
sures and epithelial ovarian cancer risk in the European Prospective Lukanova A, Soderberg S, Kaaks R, Jellum E, and Stattin P. 2006. Serum adipo-
Investigation into Cancer and Nutrition. Int J Cancer, 126(10), 2404–​ nectin is not associated with risk of colorectal cancer. Cancer Epidemiol
2415. PMID: 19821492. Biomarkers Prev, 15(2), 401–​402. PMID: 16492937.
Lahmann PH, Schulz M, Hoffmann K, et al. 2005. Long-​term weight change and Luo J, Margolis KL, Adami HO, et al. 2007. Body size, weight cycling, and
breast cancer risk: the European prospective investigation into cancer and risk of renal cell carcinoma among postmenopausal women: the Women’s
nutrition (EPIC). Br J Cancer, 93(5), 582–​589. PMCID: PMC2361598. Health Initiative (United States). Am J Epidemiol, 166(7), 752–​ 759.
Lanfranco F, Zitzmann M, Simoni M, and Nieschlag E. 2004. Serum adiponec- PMID: 17615089.
tin levels in hypogonadal males:  influence of testosterone replacement Lyman GH. 2009. Impact of chemotherapy dose intensity on cancer patient
therapy. Clin Endocrinol (Oxf), 60(4), 500–​507. PMID: 15049966. outcomes. J Natl Compr Canc Netw, 7(1), 99–​108. PMID: 19176210.
Larsson SC, and Wolk A. 2007. Obesity and the risk of gallbladder cancer: a Lyman GH, Dale DC, and Crawford J. 2003. Incidence and predictors of
meta-​analysis. Br J Cancer, 96(9), 1457–​1461. PMCID: PMC2360167. low dose-​intensity in adjuvant breast cancer chemotherapy:  a nation-
Larsson SC, and Wolk A. 2008. Overweight and obesity and incidence of leuke- wide study of community practices. J Clin Oncol, 21(24), 4524–​4531.
mia: a meta-​analysis of cohort studies. Int J Cancer, 122(6), 1418–​1421. PMID: 14673039.
PMID: 18027857. Lyman GH, Dale DC, Friedberg J, Crawford J, and Fisher RI. 2004. Incidence
Larsson SC, and Wolk A. 2011. Body mass index and risk of non-​Hodgkin’s and predictors of low chemotherapy dose-​intensity in aggressive non-​
and Hodgkin’s lymphoma: a meta-​analysis of prospective studies. Eur J Hodgkin’s lymphoma: a nationwide study. J Clin Oncol, 22(21), 4302–​
Cancer, 47(16), 2422–​2430. PMID: 21733676. 4311. PMID: 15381684.
Lauby-​Secretan B, Scoccianti C, Loomis D, et al. 2016. Body fatness and can- Ma X, Beeghly-​Fadiel A, Shu XO, et  al. 2013. Anthropometric measures
cer: viewpoint of the IARC Working Group. N Engl J Med, 375(8), 794–​ and epithelial ovarian cancer risk among Chinese women:  results from
798. PMID: 27557308. the Shanghai Women’s Health Study. Br J Cancer, 109(3), 751–​755.
Laughlin GA, Barrett-​ Connor E, May S, and Langenberg C. 2007. PMCID: PMC3738128.
Association of adiponectin with coronary heart disease and mortal- Macis D, Gandini S, Guerrieri-​Gonzaga A, et  al. 2012. Prognostic effect of
ity:  the Rancho Bernardo study. Am J Epidemiol, 165(2), 164–​ 174. circulating adiponectin in a randomized 2 x 2 trial of low-​dose tamoxifen
PMCID: PMC2642645. and fenretinide in premenopausal women at risk for breast cancer. J Clin
Lawlor DA, Okasha M, Gunnell D, Smith GD, and Ebrahim S. 2003. Oncol, 30(2), 151–​157. PMCID: PMC3255561.
Associations of adult measures of childhood growth with breast can- Macis D, Guerrieri-​Gonzaga A, and Gandini S. 2014. Circulating adiponec-
cer:  findings from the British Women’s Heart and Health Study. Br J tin and breast cancer risk: a systematic review and meta-​analysis. Int J
Cancer, 89(1), 81–​87. PMCID: PMC2394232. Epidemiol, 43(4), 1226–​1236. PMCID: PMC4258781.
Le Chatelier E, Nielsen T, Qin J, et al. 2013. Richness of human gut micro- Madamas Y, Sawka C, Franssen E, and Bjarnason G. 2001. Are medical oncol-
biome correlates with metabolic markers. Nature, 500(7464), 541–​546. ogists biased in their treatment of the large woman with breast cancer?
PMID: 23985870. Breast Cancer Res Treat, 66(2), 123–​133. PMCID: PMC11437098.
Lebovitz HE. 2003. The relationship of obesity to the metabolic syndrome. Int Madsen EL, Rissanen A, Bruun JM, et al. 2008. Weight loss larger than 10% is
J Clin Pract Suppl, 134, 18–​27. PMID: 12793594. needed for general improvement of levels of circulating adiponectin and
372

372 PART III:  THE CAUSES OF CANCER


markers of inflammation in obese subjects:  a 3-​year weight loss study. Murphy F, Kroll ME, Pirie K, et al. 2013. Body size in relation to incidence of
Eur J Endocrinol, 158(2), 179–​187. PMID: 18230824. subtypes of haematological malignancy in the prospective Million Women
Maeda K, Okubo K, Shimomura I, et al. 1996. cDNA cloning and expression of Study. Br J Cancer, 108(11), 2390–​2398. PMCID: PMC3681016.
a novel adipose specific collagen-​like factor, apM1 (AdiPose Most abun- Must A, Willett WC, and Dietz WH. 1993. Remote recall of childhood height,
dant Gene transcript 1). Biochem Biophys Res Commun, 221(2), 286–​289. weight, and body build by elderly subjects. Am J Epidemiol, 138(1), 56–​
PMID: 8619847. 64. PMID: 8333427.
Majumder K, Gupta A, Arora N, Singh PP, and Singh S. 2015. Premorbid Nagle CM, Dixon SC, Jensen A, et  al. 2015. Obesity and survival among
obesity and mortality in patients with pancreatic cancer:  a systematic women with ovarian cancer: results from the Ovarian Cancer Association
review and meta-​analysis. Clin Gastroenterol Hepatol, 14(3), 355–​368. Consortium. Br J Cancer, 113(5), 817–​826. PMCID: PMC4559823.
PMID: 26460214. Nagle CM, Marquart L, Bain CJ, et  al. 2013. Impact of weight change and
Malhotra R, Ostbye T, Riley CM, and Finkelstein EA. 2013. Young adult weight cycling on risk of different subtypes of endometrial cancer. Eur J
weight trajectories through midlife by body mass category. Obesity Cancer, 49(12), 2717–​2726. PMID: 23583438.
(Silver Spring), 21(9), 1923–​1934. PMID: 23408493. Nair SC, Toshkov IA, Yaktine AL, et  al. 1995. Dietary energy restriction-​
Mason C, Foster-​Schubert KE, Imayama I, et al. 2011. Dietary weight loss and induced modulation of protein kinase C zeta isozyme in the hamster pan-
exercise effects on insulin resistance in postmenopausal women. Am J creas. Mol Carcinog, 14(1), 10–​15. PMID: 7546218.
Prev Med, 41(4), 366–​375. PMCID: PMC3185302. Nead KT, Sharp SJ, Thompson DJ, et al. 2015. Evidence of a causal association
Mason C, Xiao L, Duggan C, et al. 2013. Effects of dietary weight loss and between insulinemia and endometrial cancer: a Mendelian randomization
exercise on insulin-​like growth factor-​I and insulin-​like growth factor-​ analysis. J Natl Cancer Inst, 107(9). PMID: 26134033.
binding protein-​ 3 in postmenopausal women:  a randomized con- Neuhouser ML, Aragaki AK, Prentice RL, et al. 2015. Overweight, obesity, and
trolled trial. Cancer Epidemiol Biomarkers Prev, 22(8), 1457–​ 1463. postmenopausal invasive breast cancer risk: a secondary analysis of the
PMCID: PMC3732802. women’s health initiative randomized clinical trials. JAMA Oncol, 1(5),
Matsuzawa Y. 2006. Therapy Insight:  adipocytokines in metabolic syndrome 611–​621. PMID: 26182172.
and related cardiovascular disease. Nat Clin Pract Cardiovasc Med, 3(1), Ng M, Fleming T, Robinson M, et  al. 2014. Global, regional, and national
35–​42. PMID: 16391616. prevalence of overweight and obesity in children and adults during 1980–​
Maury E, and Brichard SM. 2010. Adipokine dysregulation, adipose tissue 2013: a systematic analysis for the Global Burden of Disease Study 2013.
inflammation and metabolic syndrome. Mol Cell Endocrinol, 314(1), 1–​ Lancet, 384(9945), 766–​781. PMID: 24880830.
16. PMID: 19682539. Nguyen PL, Ma J, Chavarro JE, et al. 2010. Fatty acid synthase polymorphisms,
McCawley GM, Ferriss JS, Geffel D, Northup CJ, and Modesitt SC. 2009. tumor expression, body mass index, prostate cancer risk, and survival. J
Cancer in obese women: potential protective impact of bariatric surgery. Clin Oncol, 28(25), 3958–​3964. PMCID: PMC2940394.
J Am Coll Surg, 208(6), 1093–​1098. PMID: 19476897. Niedermaier T, Behrens G, Schmid D, et  al. 2015. Body mass index, physi-
McTiernan A, Irwin M, and Vongruenigen V. 2010. Weight, physical activ- cal activity, and risk of adult meningioma and glioma: a meta-​analysis.
ity, diet, and prognosis in breast and gynecologic cancers. J Clin Oncol, Neurology, 85(15), 1342–​1350. PMID: 26377253.
28(26), 4074–​4080. PMCID: PMC2940425. Niedhammer I, Bugel I, Bonenfant S, Goldberg M, and Leclerc A. 2000.
McTiernan A, Tworoger SS, Rajan KB, et  al. 2004a. Effect of exercise on Validity of self-​reported weight and height in the French GAZEL cohort.
serum androgens in postmenopausal women:  a 12-​month randomized Int J Obes Relat Metab Disord, 24(9), 1111–​1118. PMID: 11033979.
clinical trial. Cancer Epidemiol Biomarkers Prev, 13(7), 1099–​1105. Nimptsch K, and Giovannucci E. 2012. Epidemiology of IGF-​1 and cancer.
PMID: 15247119. In: LeRoith D (Ed.), Insulin-​like growth factors and cancer: from basic
McTiernan A, Tworoger SS, Ulrich CM, et  al. 2004b. Effect of exercise on biology to therapeutics (Cancer drug discovery and development) (1st
serum estrogens in postmenopausal women:  a 12-​month randomized ed., pp. 1–​24). New York: Springer Science & Business Media.
clinical trial. Cancer Res, 64(8), 2923–​2928. PMID: 15087413. Nimptsch K, Giovannucci E, Willett WC, et  al. 2011a. Body fatness dur-
Mellemkjaer L, Christensen J, Frederiksen K, et al. 2012. Leg length, sitting ing childhood and adolescence, adult height, and risk of colorec-
height and postmenopausal breast cancer risk. Br J Cancer, 107(1), 165–​ tal adenoma in women. Cancer Prev Res (Phila), 4(10), 1710–​1718.
168. PMCID: PMCPmc3389429. PMCID: PMC3188350.
Menendez JA, and Lupu R. 2007. Fatty acid synthase and the lipogenic pheno- Nimptsch K, Platz EA, Pollak MN, et  al. 2011b. Plasma insulin-​like growth
type in cancer pathogenesis. Nat Rev Cancer, 7(10), 763–​777. factor 1 is positively associated with low-​grade prostate cancer in the
Michels KB, Terry KL, Eliassen AH, Hankinson SE, and Willett WC. 2012. Health Professionals Follow-​up Study 1993–​2004. Int J Cancer, 128(3),
Adult weight change and incidence of premenopausal breast cancer. Int J 660–​667. PMCID: PMC2948057.
Cancer, 130(4), 902–​909. PMCID: PMC3245343. Niraula S, Ocana A, Ennis M, and Goodwin PJ. 2012. Body size and breast cancer
Michikawa T, Inoue M, Sawada N, et  al. 2013. Plasma levels of adiponec- prognosis in relation to hormone receptor and menopausal status: a meta-​
tin and primary liver cancer risk in middle-​aged Japanese adults with analysis. Breast Cancer Res Treat, 134(2), 769–​781. PMID: 22562122.
hepatitis virus infection: a nested case-​control study. Cancer Epidemiol Nougayrede JP, Homburg S, Taieb F, et al. 2006. Escherichia coli induces DNA
Biomarkers Prev, 22(12), 2250–​2257. PMID: 24045928. double-​strand breaks in eukaryotic cells. Science, 313(5788), 848–​851.
Mink PJ, Shahar E, Rosamond WD, Alberg AJ, and Folsom AR. 2002. Serum PMID: 16902142.
insulin and glucose levels and breast cancer incidence:  the atheroscle- Ogino S, Nosho K, Meyerhardt JA, et  al. 2008. Cohort study of fatty acid
rosis risk in communities study. Am J Epidemiol, 156(4), 349–​ 352. synthase expression and patient survival in colon cancer. J Clin Oncol,
PMID: 12181105. 26(35), 5713–​5720. PMCID: PMC2630484.
Moller E, Adami HO, Mucci LA, et al. 2013. Lifetime body size and prostate Oh DK, Ciaraldi T, and Henry RR. 2007. Adiponectin in health and disease.
cancer risk in a population-​based case-​control study in Sweden. Cancer Diabetes Obes Metab, 9(3), 282–​289. PMID: 17391153.
Causes Control, 24(12), 2143–​2155. PMID: 24048969. Ohtani N, Yoshimoto S, and Hara E. 2014. Obesity and cancer: a gut microbial
Moller E, Wilson KM, Batista JL, et al. 2016. Body size across the life course connection. Cancer Research, 74(7), 1885–​1889. PMID: 24638983.
and prostate cancer in the Health Professionals Follow-​up Study. Int J Okada-​Iwabu M, Yamauchi T, Iwabu M, et al. 2013. A small-​molecule AdipoR
Cancer, 138(4), 853–​865. PMID: 26355806. agonist for type 2 diabetes and short life in obesity. Nature, 503(7477),
Moon HS, Liu X, Nagel JM, et  al. 2013. Salutary effects of adiponectin on 493–​499. PMID: 24172895.
colon cancer: in vivo and in vitro studies in mice. Gut, 62(4), 561–​570. Ollberding NJ, Kim Y, Shvetsov YB, et al. 2013. Prediagnostic leptin, adipo-
PMID: 22735569. nectin, C-​reactive protein, and the risk of postmenopausal breast cancer.
Moore SC, Leitzmann MF, Albanes D, et al. 2009. Adipokine genes and pros- Cancer Prev Res (Phila), 6(3), 188–​195. PMCID: PMC3595121.
tate cancer risk. Int J Cancer, 124(4), 869–​876. PMCID: PMC2879625. Ong JP, and Younossi ZM. 2007. Epidemiology and natural history of NAFLD
Mozaffarian D, Hao T, Rimm EB, Willett WC, and Hu FB. 2011. Changes in and NASH. Clin Liver Dis, 11(1), 1–​16, vii. PMID: 17544968.
diet and lifestyle and long-​term weight gain in women and men. N Engl J Oxentenko AS, Bardia A, Vierkant RA, et  al. 2010. Body size and incident
Med, 364(25), 2392–​2404. PMCID: PMC3151731. colorectal cancer: a prospective study of older women. Cancer Prev Res
Mu N, Zhu Y, Wang Y, Zhang H, and Xue F. 2012. Insulin resistance: a signifi- (Phila), 3(12), 1608–​1620. PMCID: PMC3005991.
cant risk factor of endometrial cancer. Gynecol Oncol, 125(3), 751–​757. Palmer JR, Adams-​Campbell LL, Boggs DA, Wise LA, and Rosenberg L. 2007.
PMID: 22449736. A prospective study of body size and breast cancer in black women. Cancer
Munsell MF, Sprague BL, Berry DA, Chisholm G, and Trentham-​Dietz A. Epidemiol Biomarkers Prev, 16(9), 1795–​1802. PMID: 17855697.
2014. Body mass index and breast cancer risk according to postmeno- Palta M, Prineas RJ, Berman R, and Hannan P. 1982. Comparison of self-​
pausal estrogen-​progestin use and hormone receptor status. Epidemiol reported and measured height and weight. Am J Epidemiol, 115(2), 223–​
Rev, 36, 114–​136. PMCID: PMC3873844. 230. PMID: 7058781.
 37

Obesity and Body Composition 373


Pandini G, Medico E, Conte E, et al. 2003. Differential gene expression induced patient data from 12 prospective studies. Ann Intern Med, 149(7), 461–​
by insulin and insulin-​like growth factor-​II through the insulin receptor 471, W483-​468. PMCID: PMC2584869.
isoform A. J Biol Chem, 278(43), 42178–​42189. PMID: 12881524. Rodriguez C, Freedland SJ, Deka A, et al. 2007. Body mass index, weight change,
Park M, Song da Y, Je Y, and Lee JE. 2014. Body mass index and biliary tract and risk of prostate cancer in the Cancer Prevention Study II Nutrition Cohort.
disease:  a systematic review and meta-​analysis of prospective studies. Cancer Epidemiol Biomarkers Prev, 16(1), 63–​69. PMID: 17179486.
Prev Med, 65, 13–​22. PMID: 24721739. Rosner B, Eliassen AH, Toriola AT, et  al. 2015. Short-​term weight gain and
Park SL, Goodman MT, Zhang ZF, et  al. 2010. Body size, adult BMI gain breast cancer risk by hormone receptor classification among pre-​and
and endometrial cancer risk: the multiethnic cohort. Int J Cancer, 126(2), postmenopausal women. Breast Cancer Res Treat, 150(3), 643–​ 653.
490–​499. PMCID: PMC2795089. PMCID: PMC4383816.
Parker ED, and Folsom AR. 2003. Intentional weight loss and incidence of Ross MH, and Bras G. 1971. Lasting influence of early caloric restriction on
obesity-​related cancers: the Iowa Women’s Health Study. Int J Obes Relat prevalence of neoplasms in the rat. J Natl Cancer Inst, 47(5), 1095–​1114.
Metab Disord, 27(12), 1447–​1452. PMID: WOS:000186844500002. Rubinstein MR, Wang X, Liu W et  al. 2013. Fusobacterium nucleatum pro-
Parkin E, O’Reilly DA, Sherlock DJ, Manoharan P, and Renehan AG. 2014. motes colorectal carcinogenesis by modulating E-​cadherin/​beta-​catenin
Excess adiposity and survival in patients with colorectal cancer: a system- signaling via its FadA adhesin. Cell Host Microbe, 14(2), 195–​ 206.
atic review. Obes Rev, 15(5), 434–​451. PMID: 24433336. PMCID: PMC3770529.
Patel AV, Rodriguez C, Bernstein L, et al. 2005. Obesity, recreational physi- Ryan AS, and Nicklas BJ. 2004. Reductions in plasma cytokine levels with
cal activity, and risk of pancreatic cancer in a large U.S. Cohort. Cancer weight loss improve insulin sensitivity in overweight and obese postmeno-
Epidemiol Biomarkers Prev, 14(2), 459–​466. PMID: 15734973. pausal women. Diabetes Care, 27(7), 1699–​1705. PMID: 15220249.
Perry GS, Byers TE, Mokdad AH, Serdula MK, and Williamson DF. 1995. The Ryerson AB, Eheman CR, Altekruse SF, et al. 2016. Annual Report to the Nation
validity of self-​reports of past body weights by U.S. adults. Epidemiology, on the Status of Cancer, 1975–​2012, featuring the increasing incidence of
6(1), 61–​66. PMID: 7888448. liver cancer. Cancer, 122(9), 1312–​1337. PMCID: PMC4840031.
Poehls J, Wassel CL, Harris TB, et al. 2009. Association of adiponectin with Samimi G, Colditz GA, Baer HJ, and Tamimi RM. 2008. Measures of energy
mortality in older adults:  the Health, Aging, and Body Composition balance and mammographic density in the Nurses’ Health Study. Breast
Study. Diabetologia, 52(4), 591–​595. PMCID: PMC2756826. Cancer Res Treat, 109(1), 113–​122. PMID: 17592770.
Pollak M. 2008. Insulin and insulin-​like growth factor signalling in neoplasia. Sanfilippo KM, McTigue KM, Fidler CJ, et al. 2014. Hypertension and obesity
Nat Rev Cancer, 8(12), 915–​928. PMID: 19029956. and the risk of kidney cancer in 2 large cohorts of US men and women.
Poole EM, Tworoger SS, Hankinson SE et  al. 2011. Body size in early Hypertension, 63(5), 934–​941. PMCID: PMC4098147.
life and adult levels of insulin-​ like growth factor 1 and insulin-​ like Sangaramoorthy M, Phipps AI, Horn-​Ross PL, Koo J, and John EM. 2011.
growth factor binding protein 3. Am J Epidemiol, 174(6), 642–​ 651. Early-​life factors and breast cancer risk in Hispanic women: the role of
PMCID: PMC3166705. adolescent body size. Cancer Epidemiol Biomarkers Prev, 20(12), 2572–​
Prado CM, Gonzalez MC, and Heymsfield SB. 2015. Body composition phe- 2582. PMCID: PMC3461314.
notypes and obesity paradox. Curr Opin Clin Nutr Metab Care, 18(6), Santry HP, Gillen DL, and Lauderdale DS. 2005. Trends in bariatric surgical
535–​551. PMID: 26335310. procedures. JAMA, 294(15), 1909–​1917. PMID: 16234497.
Prescott J, Setiawan VW, Wentzensen N, et al. 2015. Body mass index genetic Sattar N, and Nelson SM. 2008. Adiponectin, diabetes, and coronary heart dis-
risk score and endometrial cancer risk. PLoS One, 10(11), e0143256. ease in older persons: unraveling the paradox. J Clin Endocrinol Metab,
PMCID: PMC4659592. 93(9), 3299–​3301. PMID: 18772460.
Protani MM, Nagle CM, and Webb PM. 2012. Obesity and ovarian cancer sur- Schlesinger S, Siegert S, Koch M, et  al. 2014. Postdiagnosis body mass
vival: a systematic review and meta-​analysis. Cancer Prev Res (Phila), index and risk of mortality in colorectal cancer survivors: a prospective
5(7), 901–​910. PMID: 22609763. study and meta-​analysis. Cancer Causes Control, 25(10), 1407–​1418.
Radimer KL, Ballard-​Barbash R, Miller JS, et al. 2004. Weight change and the PMID: 25037235.
risk of late-​onset breast cancer in the original Framingham cohort. Nutr Schmandt RE, Iglesias DA, Co NN, and Lu KH. 2011. Understanding obesity
Cancer, 49(1), 7–​13. PMID: 15456630. and endometrial cancer risk:  opportunities for prevention. Am J Obstet
Randi G, Franceschi S, and La Vecchia C. 2006. Gallbladder cancer world- Gynecol, 205(6), 518–​525. PMCID: PMC4264838.
wide:  geographical distribution and risk factors. Int J Cancer, 118(7), Schouten LJ, Goldbohm RA, and van den Brandt PA. 2003. Height, weight,
1591–​1602. PMID: 16397865. weight change, and ovarian cancer risk in the Netherlands cohort study
Rapp K, Klenk J, Ulmer H, et  al. 2008. Weight change and cancer risk in a on diet and cancer. Am J Epidemiol, 157(5), 424–​433. PMID: 12615607.
cohort of more than 65,000 adults in Austria. Ann Oncol, 19(4), 641–​648. Sears CL. 2009. Enterotoxigenic Bacteroides fragilis: a rogue among symbi-
PMID: 18056917. otes. Clin Microbiol Rev, 22(2), 349–​369. PMCID: PMC2668231.
Rapp K, Schroeder J, Klenk J, et al. 2006. Fasting blood glucose and cancer risk Sellers TA, Vachon CM, Pankratz VS, et  al. 2007. Association of childhood
in a cohort of more than 140,000 adults in Austria. Diabetologia, 49(5), and adolescent anthropometric factors, physical activity, and diet with
945–​952. PMID: 16557372. adult mammographic breast density. Am J Epidemiol, 166(4), 456–​464.
Renehan AG, Flood A, Adams KF, et al. 2012. Body mass index at different PMID: 17548785.
adult ages, weight change, and colorectal cancer risk in the National Sergentanis TN, Antoniadis AG, Gogas HJ, et  al. 2013. Obesity and risk of
Institutes of Health-​AARP Cohort. Am J Epidemiol, 176(12), 1130–​1140. malignant melanoma: a meta-​analysis of cohort and case-​control studies.
PMID: 23186750. Eur J Cancer, 49(3), 642–​657. PMID: 23200191.
Renehan AG, Tyson M, Egger M, Heller RF, and Zwahlen M. 2008. Body-​mass Setiawan VW, Stram DO, Nomura AM, Kolonel LN, and Henderson BE. 2007.
index and incidence of cancer:  a systematic review and meta-​analysis Risk factors for renal cell cancer: the multiethnic cohort. Am J Epidemiol,
of prospective observational studies. Lancet, 371(9612), 569–​ 578. 166(8), 932–​940. PMID: 17656615.
PMID: 18280327. Shayne M, Culakova E, Poniewierski MS, et al. 2007. Dose intensity and hema-
Reseland JE, Reppe S, Olstad OK, et al. 2009. Abnormal adipokine levels and tologic toxicity in older cancer patients receiving systemic chemotherapy.
leptin-​induced changes in gene expression profiles in multiple myeloma. Cancer, 110(7), 1611–​1620. PMID: 17705197.
Eur J Haematol, 83(5), 460–​470. PMID: 19572994. Siegel AB, Goyal A, Salomao M, et al. 2015. Serum adiponectin is associated
Rich-​Edwards JW, Goldman MB, Willett WC, et  al. 1994. Adolescent body with worsened overall survival in a prospective cohort of hepatocellular
mass index and infertility caused by ovulatory disorder. Am J Obstet carcinoma patients. Oncology, 88(1), 57–​68. PMCID: PMC4275375.
Gynecol, 171(1), 171–​177. PMID: 8030695. Sieri S, Muti P, Claudia A, et al. 2012. Prospective study on the role of glucose
Richards JB, Waterworth D, O’Rahilly S, et  al. 2009. A genome-​wide asso- metabolism in breast cancer occurrence. Int J Cancer, 130(4), 921–​929.
ciation study reveals variants in ARL15 that influence adiponectin levels. PMID: 21413010.
PLoS Genet, 5(12), e1000768. PMCID: PMC2781107. Singh A, Hamilton-​Fairley D, Koistinen R, et al. 1990. Effect of insulin-​like
Rinaldi S, Cleveland R, Norat T, et al. 2010. Serum levels of IGF-​I, IGFBP-​3 growth factor-​type I (IGF-​I) and insulin on the secretion of sex hormone
and colorectal cancer risk:  results from the EPIC cohort, plus a meta-​ binding globulin and IGF-​I binding protein (IBP-​I) by human hepatoma
analysis of prospective studies. Int J Cancer, 126(7), 1702–​ 1715. cells. J Endocrinol, 124(2), R1–​3. PMID: 1690256.
PMID: 19810099. Singh N, Gurav A, Sivaprakasam S, et  al. 2014. Activation of Gpr109a,
Riondino S, Roselli M, Palmirotta R et al. 2014. Obesity and colorectal can- receptor for niacin and the commensal metabolite butyrate, suppresses
cer:  role of adipokines in tumor initiation and progression. World J colonic inflammation and carcinogenesis. Immunity, 40(1), 128–​ 139.
Gastroenterol, 20(18), 5177–​5190. PMCID: PMCPmc4017033. PMCID: PMC4305274.
Roddam AW, Allen NE, Appleby P, et  al. 2008. Insulin-​like growth factors, Singh S, Sharma AN, Murad MH, et  al. 2013. Central adiposity is associ-
their binding proteins, and prostate cancer risk:  analysis of individual ated with increased risk of esophageal inflammation, metaplasia, and
374

374 PART III:  THE CAUSES OF CANCER


adenocarcinoma: a systematic review and meta-​analysis. Clin Gastroenterol Suzuki R, Iwasaki M, Inoue M, et al. 2011. Body weight at age 20 years, sub-
Hepatol, 11(11), 1399–​1412 e1397. PMCID: PMC3873801. sequent weight change and breast cancer risk defined by estrogen and
Sjostrom L, Gummesson A, Sjostrom CD, et al. 2009. Effects of bariatric sur- progesterone receptor status:  the Japan public health center-​based pro-
gery on cancer incidence in obese patients in Sweden (Swedish Obese spective study. Int J Cancer, 129(5), 1214–​1224. PMID: 21064092.
Subjects Study):  a prospective, controlled intervention trial. Lancet Swanson CA, Potischman N, Wilbanks GD, et al. 1993. Relation of endometrial
Oncol, 10(7), 653–​662. PMID: 19556163. cancer risk to past and contemporary body size and body fat distribution.
Sjostrom L, Lindroos AK, Peltonen M, et  al. 2004. Lifestyle, diabetes, and Cancer Epidemiol Biomarkers Prev, 2(4), 321–​327. PMID: 8348055.
cardiovascular risk factors 10 years after bariatric surgery. N Engl J Med, Sweeney C, Blair CK, Anderson KE, Lazovich D, and Folsom AR. 2004. Risk
351(26), 2683–​2693. PMID: 15616203. factors for breast cancer in elderly women. Am J Epidemiol, 160(9), 868–​
Sjostrom L, Narbro K, Sjostrom CD, et al. 2007. Effects of bariatric surgery 875. PMID: 15496539.
on mortality in Swedish obese subjects. N Engl J Med, 357(8), 741–​752. Tamimi RM, Colditz GA, Hazra A, et al. 2012. Traditional breast cancer risk
PMID: 17715408. factors in relation to molecular subtypes of breast cancer. Breast Cancer
Sjostrom L, Peltonen M, Jacobson P, et al. 2012. Bariatric surgery and long-​ Res Treat, 131(1), 159–​167. PMCID: PMC3237947.
term cardiovascular events. JAMA, 307(1), 56–​65. PMID: 22215166. Tan W, Gao M, Liu N, et al. 2015. Body mass index and risk of gallbladder
Soliman PT, Cui X, Zhang Q, Hankinson SE, and Lu KH. 2011. Circulating cancer:  systematic review and meta-​analysis of observational studies.
adiponectin levels and risk of endometrial cancer:  the prospective Nutrients, 7(10), 8321–​8334. PMCID: PMC4632410.
Nurses’ Health Study. Am J Obstet Gynecol, 204(2), 167 e161–​165. Tannenbaum A. 1940a. The initiation and growth of tumors:  introduction.
PMCID: PMC3032826. I. Effects of underfeeding. Am J Cancer, 38, 335–​350.
Sommer F, and Backhed F. 2013. The gut microbiota-​ -​
masters of host Tannenbaum A. 1940b. Relationship of body weight to cancer incidence. Arch
development and physiology. Nat Rev Microbiol, 11(4), 227–​ 238. Pathol, 30, 509–​517.
PMID: 23435359. Tchernof A, Nolan A, Sites CK, Ades PA, and Poehlman ET. 2002. Weight
Song M, and Giovannucci E. 2016. Estimating the influence of obesity on can- loss reduces C-​reactive protein levels in obese postmenopausal women.
cer risk: stratification by smoking is critical. J Clin Oncol, 34(27), 3237–​ Circulation, 105(5), 564–​569. PMID: 11827920.
3239. PMID: 27458311. Tehard B, Kaaks R, and Clavel-​Chapelon F. 2005. Body silhouette, menstrual
Song M, Gong J, Giovannucci EL, et  al. 2015a. Genetic variants of adipo- function at adolescence and breast cancer risk in the E3N cohort study. Br
nectin and risk of colorectal cancer. Int J Cancer, 137(1), 154–​164. J Cancer, 92(11), 2042–​2048. PMCID: PMC1904482.
PMCID: PMC4405454. Teras LR, Goodman M, Patel AV, et al. 2011. Weight loss and postmenopausal
Song M, Hu FB, Spiegelman D, et  al. 2015b. Adulthood weight change breast cancer in a prospective cohort of overweight and obese US women.
and risk of colorectal cancer in the Nurses’ Health Study and Health Cancer Causes Control, 22(4), 573–​579. PMID: 21327461.
Professionals Follow-​up Study. Cancer Prev Res (Phila), 8(7), 620–​627. Teras LR, Kitahara CM, Birmann BM, et  al. 2014. Body size and multiple
PMCID: PMC4490957. myeloma mortality:  a pooled analysis of 20 prospective studies. Br J
Song M, Hu FB, Spiegelman D, et al. 2015c. Long-​term status and change of Haematol, 166(5), 667–​676. PMCID: PMC4134758.
body fat distribution, and risk of colorectal cancer: a prospective cohort Thomas EL, Frost G, Taylor-​Robinson SD, and Bell JD. 2012. Excess body
study. Int J Epidemiol, 45(3), 871–​883. PMID: 26403814. fat in obese and normal-​weight subjects. Nutr Res Rev, 25(1), 150–​161.
Song M, Wu K, Ogino S, et al. 2013a. A prospective study of plasma inflamma- PMID: 22625426.
tory markers and risk of colorectal cancer in men. Br J Cancer, 108(9), Thrift AP, Shaheen NJ, Gammon MD, et al. 2014. Obesity and risk of esopha-
1891–​1898. PMCID: PMC3658520. geal adenocarcinoma and Barrett’s esophagus: a Mendelian randomiza-
Song M, Zhang X, Wu K, et al. 2013b. Plasma adiponectin and soluble leptin tion study. J Natl Cancer Inst, 106(11). PMCID: PMC4200028.
receptor and risk of colorectal cancer: a prospective study. Cancer Prev Thun MJ, Carter BD, Feskanich D, et  al. 2013. 50-​year trends in smoking-​
Res (Phila), 6(9), 875–​885. PMCID: PMC3772620. related mortality in the United States. N Engl J Med, 368(4), 351–​364.
Spiegelman D, Israel RG, Bouchard C, and Willett WC. 1992. Absolute fat PMCID: PMC3632080.
mass, percent body fat, and body-​fat distribution: which is the real deter- Thygesen LC, Gronbaek M, Johansen C, et al. 2008. Prospective weight change
minant of blood pressure and serum glucose? Am J Clin Nutr, 55(6), and colon cancer risk in male US health professionals. Int J Cancer,
1033–​1044. PMID: 1595574. 123(5), 1160–​1165. PMCID: PMC3965300.
Stark JR, Li H, Kraft P, et  al. 2009. Circulating prediagnostic interleukin-​6 Tilg H, and Moschen AR. 2006. Adipocytokines:  mediators linking adipose
and C-​reactive protein and prostate cancer incidence and mortality. Int J tissue, inflammation and immunity. Nat Rev Immunol, 6(10), 772–​783.
Cancer, 124(11), 2683–​2689. PMCID: PMC2667697. PMID: 16998510.
Starley BQ, Calcagno CJ, and Harrison SA. 2010. Nonalcoholic fatty liver dis- Toniolo P, Bruning PF, Akhmedkhanov A, et al. 2000. Serum insulin-​like growth
ease and hepatocellular carcinoma:  a weighty connection. Hepatology, factor-​I and breast cancer. Int J Cancer, 88(5), 828–​832. PMID: 11072255.
51(5), 1820–​1832. PMID: 20432259. Torres DM, and Harrison SA. 2008. Diagnosis and therapy of nonalcoholic
Steffen A, Huerta JM, Weiderpass E, et al. 2015. General and abdominal obe- steatohepatitis. Gastroenterology, 134(6), 1682–​1698. PMID: 18471547.
sity and risk of esophageal and gastric adenocarcinoma in the European Touvier M, Fezeu L, Ahluwalia N, et  al. 2013. Association between predi-
Prospective Investigation into Cancer and Nutrition. Int J Cancer, 137(3), agnostic biomarkers of inflammation and endothelial function and can-
646–​657. PMID: 25598323. cer risk:  a nested case-​control study. Am J Epidemiol, 177(1), 3–​13.
Steins Bisschop CN, van Gils CH, Emaus MJ, et  al. 2014. Weight change PMCID: PMC3529490.
later in life and colon and rectal cancer risk in participants in the EPIC-​ Tremaroli V, and Backhed F. 2012. Functional interactions between the
PANACEA study. Am J Clin Nutr, 99(1), 139–​147. PMID: 24225355. gut microbiota and host metabolism. Nature, 489(7415), 242–​ 249.
Stevens J, Keil JE, Waid LR, and Gazes PC. 1990. Accuracy of current, 4-​ PMID: 22972297.
year, and 28-​year self-​reported body weight in an elderly population. Am Trichopoulos D, and Lipworth L. 1995. Is cancer causation simpler than
J Epidemiol, 132(6), 1156–​1163. PMID: 2260547. we thought, but more intractable. Epidemiology, 6(4), 347–​ 349.
Stocks T, Lukanova A, Johansson M, et  al. 2008. Components of the meta- PMID: WOS:A1995RF74700003.
bolic syndrome and colorectal cancer risk: a prospective study. Int J Obes Tworoger SS, Eliassen AH, Kelesidis T, et al. 2007. Plasma adiponectin con-
(Lond), 32(2), 304–​314. PMID: 17878894. centrations and risk of incident breast cancer. J Clin Endocrinol Metab,
Stoll BA. 1998. Teenage obesity in relation to breast cancer risk. Int J Obes 92(4), 1510–​1516. PMID: 17213279.
Relat Metab Disord, 22(11), 1035–​1040. PMID: 9822939. US Department of Agriculture. 1990. Dietary guidelines for Americans.
Stolzenberg-​Solomon RZ, Schairer C, Moore S, Hollenbeck A, and Silverman Washington, DC: US Department of Agriculture.
DT. 2013. Lifetime adiposity and risk of pancreatic cancer in the NIH-​ van den Brandt PA, Dirx MJ, Ronckers CM, van den Hoogen P, and Goldbohm
AARP Diet and Health Study cohort. Am J Clin Nutr, 98(4), 1057–​1065. RA. 1997. Height, weight weight change, and postmenopausal breast can-
PMCID: PMC3778860. cer risk:  The Netherlands Cohort Study. Cancer Causes Control, 8(1),
Stunkard AJ, Sorensen TI, and Schulsinger F. 1983. Use of the Danish Adoption 39–​47. PMID: 9051321.
Register for the study of obesity and thinness. In: Kety SS, Rowland LP, van Dijk BA, Schouten LJ, Kiemeney LA, Goldbohm RA, and van den Brandt
Sidman SW, and Mathysee SW (Eds.), The genetics of neurological and PA. 2004. Relation of height, body mass, energy intake, and physical
psychiatric disorders (pp. 115–​120). New York: Raven Press. activity to risk of renal cell carcinoma:  results from the Netherlands
Sugiyama M, Takahashi H, Hosono K, et  al. 2009. Adiponectin inhibits Cohort Study. Am J Epidemiol, 160(12), 1159–​1167. PMID: 15583368.
colorectal cancer cell growth through the AMPK/​mTOR pathway. Int J van Gemert WA, Schuit AJ, van der Palen J et al. 2015. Effect of weight loss,
Oncol, 34(2), 339–​344. PMID: 19148467. with or without exercise, on body composition and sex hormones in
 375

Obesity and Body Composition 375


postmenopausal women:  the SHAPE-​2 trial. Breast Cancer Res, 17(1), Weindruch R, and Walford RL. 1982. Dietary restriction in mice beginning
120. PMCID: PMC4557857. at 1  year of age:  effect on life-​span and spontaneous cancer incidence.
van Kruijsdijk RC, van der Wall E, and Visseren FL. 2009. Obesity and Science, 215(4538), 1415–​1418. PMID: 7063854.
cancer:  the role of dysfunctional adipose tissue. Cancer Epidemiol White KK, Park SY, Kolonel LN, Henderson BE, and Wilkens LR. 2012. Body
Biomarkers Prev, 18(10), 2569–​2578. PMID: 19755644. size and breast cancer risk: the Multiethnic Cohort. Int J Cancer, 131(5),
Vansaun MN. 2013. Molecular pathways: adiponectin and leptin signaling in E705–​716. PMCID: PMC4484854.
cancer. Clin Cancer Res, 19(8), 1926–​1932. PMCID: PMC3630242. WHO (World Health Organzation). 2016. Obesity and overweight fact sheet.
Verheus M, Peeters PH, Rinaldi S, et  al. 2006. Serum C-​peptide levels and http://​www.who.int/​mediacentre/​factsheets/​fs311/​en/​
breast cancer risk:  results from the European Prospective Investigation Willett W. 2013. Anthropometric measures and body composition. In: Willett
into Cancer and Nutrition (EPIC). Int J Cancer, 119(3), 659–​ 667. W (Ed.), Nutritional epidemiology (pp. 213–​259). New  York:  Oxford
PMID: 16572422. University Press.
Visser M, Bouter LM, McQuillan GM, Wener MH, and Harris TB. 1999. Williamson DF, Pamuk E, Thun M, et  al. 1995. Prospective study of inten-
Elevated C-​reactive protein levels in overweight and obese adults. JAMA, tional weight loss and mortality in never-​smoking overweight US white
282(22), 2131–​2135. PMID: 10591334. women aged 40–​64 years. Am J Epidemiol, 141(12), 1128–​1141. PMID:
Vrieling A, Buck K, Kaaks R, and Chang-​Claude J. 2010. Adult weight gain 7771451.
in relation to breast cancer risk by estrogen and progesterone recep- Williamson DF, Pamuk E, Thun M, et al. 1999. Prospective study of intentional
tor status:  a meta-​analysis. Breast Cancer Res Treat, 123(3), 641–​649. weight loss and mortality in overweight white men aged 40–​64 years. Am
PMID: 20711809. J Epidemiol, 149(6), 491–​503. PMID: 10084238.
Wajchenberg BL. 2000. Subcutaneous and visceral adipose tissue:  their Wong VW, Wong GL, Yu J, et al. 2010. Interaction of adipokines and hepatitis
relation to the metabolic syndrome. Endocr Rev, 21(6), 697–​ 738. B virus on histological liver injury in the Chinese. Am J Gastroenterol,
PMID: 11133069. 105(1), 132–​138. PMID: 19809411.
Wallin A, and Larsson SC. 2011. Body mass index and risk of multiple Wright ME, Chang SC, Schatzkin A, et al. 2007. Prospective study of adiposity
myeloma: a meta-​analysis of prospective studies. Eur J Cancer, 47(11), and weight change in relation to prostate cancer incidence and mortality.
1606–​1615. PMID: 21354783. Cancer, 109(4), 675–​684. PMID: 17211863.
Wannamethee SG, Whincup PH, Lennon L, and Sattar N. 2007. Circulating Wu K, Feskanich D, Fuchs CS, et  al. 2011. Interactions between plasma
adiponectin levels and mortality in elderly men with and without cardio- levels of 25-​ hydroxyvitamin D, insulin-​ like growth factor (IGF)-​
vascular disease and heart failure. Arch Intern Med, 167(14), 1510–​1517. 1 and C-​ peptide with risk of colorectal cancer. PLoS One, 6(12).
PMID: 17646605. PMID: WOS:000300676300005.
Warren RL, Freeman DJ, Pleasance S, et  al. 2013. Co-​ occurrence of Wu S, Liu J, Wang X, et al. 2014. Association of obesity and overweight with
anaerobic bacteria in colorectal carcinomas. Microbiome, 1(1), 16. overall survival in colorectal cancer patients: a meta-​analysis of 29 stud-
PMCID: PMC3971631. ies. Cancer Causes Control, 25(11), 1489–​1502. PMID: 25070668.
WCRF/​AICR (World Cancer Research Fund/​American Institute for Cancer Xu WH, Xiang YB, Zheng W, et  al. 2006. Weight history and risk of endo-
Research). 2007. Food, nutrition, physical activity, and the prevention of metrial cancer among Chinese women. Int J Epidemiol, 35(1), 159–​166.
cancer: a global perspective. Washington, DC: AICR. PMID: 16258056.
WCRF/​AICR. 2008. WCRF/​ AICR systematic literature review continuous Yamauchi T, Kamon J, Minokoshi Y, et al. 2002. Adiponectin stimulates glu-
update report:  the associations between food, nutrition and physical cose utilization and fatty-​acid oxidation by activating AMP-​activated pro-
activity and the risk of breast cancer. tein kinase. Nat Med, 8(11), 1288–​1295. PMID: 12368907.
WCRF/​AICR. 2009. Policy and action for cancer prevention. food, nutrition, Yamauchi T, Kamon J, Waki H, et al. 2003. Globular adiponectin protected ob/​
and physical activity: a global perspective. Washington DC: AICR. ob mice from diabetes and ApoE-​deficient mice from atherosclerosis. J
World Cancer Research Fund/​American Institute for Cancer Research. (2010). Biol Chem, 278(4), 2461–​2468. PMID: 12431986.
Continuous Update Project Report. Food, Nutrition, Physical Activity, Yang TY, Cairns BJ, Allen N, et al. 2012. Postmenopausal endometrial cancer
and the Prevention of Breast Cancer. risk and body size in early life and middle age: prospective cohort study.
WCRF/​AICR. 2010b. Policy and action for cancer prevention. executive sum- Br J Cancer, 107(1), 169–​175. PMCID: PMC3389421.
mary: food, nutrition, and physical activity with an added US perspective. Yao X, and Tian Z. 2015. Dyslipidemia and colorectal cancer risk:  a meta-​
Washington, DC: AICR. analysis of prospective studies. Cancer Causes Control, 26(2), 257–​268.
WCRF/​AICR. 2011. Continuous update project report. food, nutrition, physi- PMID: 25488827.
cal activity, and the prevention of colorectal cancer. Yarrow JF, Beggs LA, Conover CF, et  al. 2012. Influence of androgens on
WCRF/​AICR. 2012. Continuous update project report. food, nutrition, physi- circulating adiponectin in male and female rodents. PLoS One, 7(10),
cal activity, and the prevention of pancreatic cancer. e47315. PMCID: PMC3468547.
WCRF/​AICR. 2013. Continuous update project report. food, nutrition, physi- Yee D, and Lee AV. 2000. Crosstalk between the insulin-​like growth factors
cal activity, and the prevention of endometrial cancer. and estrogens in breast cancer. J Mammary Gland Biol Neoplasia, 5(1),
WCRF/​AICR. 2014a. Continuous update project prostate cancer SLR. 107–​115. PMID: 10791773.
WCRF/​AICR. 2014b. Continuous update project report: food, nutrition, physi- Yoon YS, Keum N, Zhang X, Cho E, and Giovannucci EL. 2015a. Circulating
cal activity, and prostate cancer. levels of IGF-​1, IGFBP-​3, and IGF-​1/​IGFBP-​3 molar ratio and colorec-
WCRF/​AICR. 2014c. Continuous update project report. food, nutrition, physi- tal adenomas:  a meta-​analysis. Cancer Epidemiol, 39(6), 1026–​1035.
cal activity, and the prevention of ovarian cancer. PMID: 26388613.
WCRF/​AICR. 2015a. Continuous update project report: food, nutrition, physi- Yoon YS, Keum N, Zhang X, Cho E, and Giovannucci EL. 2015b.
cal activity and liver cancer. Hyperinsulinemia, insulin resistance and colorectal adenomas:  a meta-​
WCRF/​AICR. 2015b. Continuous update project report: food, nutrition, physi- analysis. Metabolism, 64(10), 1324–​1333. PMID: 26169471.
cal activity and kidney cancer. Yoshimoto S, Loo TM, Atarashi K, et al. 2013. Obesity-​induced gut microbial
WCRF/​AICR. 2015c. Continuous update project report: food, nutrition, physi- metabolite promotes liver cancer through senescence secretome. Nature,
cal activity and gallbladder cancer. 499(7456), 97–​101. PMID: 23803760.
WCRF/​AICR. 2016a. Continuous update project report: diet, nutrition, physi- Yu H, and Rohan T. 2000. Role of the insulin-​like growth factor family in can-
cal activity and stomach cancer. cer development and progression. J Natl Cancer Inst, 92(18), 1472–​1489.
WCRF/​AICR. 2016b. Continuous update project report: diet, nutrition, physi- PMID: 10995803.
cal activity and oesophageal cancer. Zhang B, Shu XO, Delahanty RJ, et al. 2015. Height and breast cancer risk: evi-
Wei EK, Ma J, Pollak MN, et  al. 2005. A  prospective study of C-​peptide, dence from prospective studies and mendelian randomization. J Natl
insulin-​like growth factor-​I, insulin-​like growth factor binding protein-​1, Cancer Inst, 107(11), djv219. PMCID: PMC4643630.
and the risk of colorectal cancer in women. Cancer Epidemiol Biomarkers Zhang H, DiBaise JK, Zuccolo A, et  al. 2009. Human gut microbiota
Prev, 14(4), 850–​855. PMID: 15824155. in obesity and after gastric bypass. PNAS, 106(7), 2365–​ 2370.
Weiderpass E, Braaten T, Magnusson C, et  al. 2004. A  prospective study PMCID: PMC2629490.
of body size in different periods of life and risk of premenopausal Zhang SS, Yang H, Luo KJ, et  al. 2013. The impact of body mass index on
breast cancer. Cancer Epidemiol Biomarkers Prev, 13(7), 1121–​1127. complication and survival in resected oesophageal cancer:  a clinical-​
PMID: 15247122. based cohort and meta-​ analysis. Br J Cancer, 109(11), 2894–​ 2903.
Weikert S, Boeing H, Pischon T, et al. 2008. Blood pressure and risk of renal PMCID: PMC3844915.
cell carcinoma in the European prospective investigation into cancer and Zhang X, Eliassen AH, Tamimi RM, et al. 2015a. Adult body size and physical
nutrition. Am J Epidemiol, 167(4), 438–​446. PMID: 18048375. activity in relation to risk of breast cancer according to tumor androgen
376

376 PART III:  THE CAUSES OF CANCER


receptor status. Cancer Epidemiol Biomarkers Prev, 24(6), 962–​968. Ziakas PD, Karsaliakos P, Prodromou ML, and Mylonakis E. 2013. Interleukin-​
PMCID: PMC4452447. 6 polymorphisms and hematologic malignancy:  a re-​appraisal of evi-
Zhang X, Tworoger SS, Eliassen AH, and Hankinson SE. 2013. Postmenopausal dence from genetic association studies. Biomarkers, 18(7), 625–​631.
plasma sex hormone levels and breast cancer risk over 20 years of follow-​ PMID: 24059848.
up. Breast Cancer Res Treat, 137(3), 883–​892. PMCID: PMC3582409. Zuccolo L, Harris R, Gunnell D, et  al. 2008. Height and prostate can-
Zhang X, Zhou G, Sun B, et al. 2015b. Impact of obesity upon prostate cancer-​ cer risk:  a large nested case-​control study (ProtecT) and meta-​
associated mortality:  a meta-​analysis of 17 cohort studies. Oncol Lett, analysis. Cancer Epidemiol Biomarkers Prev, 17(9), 2325–​ 2336.
9(3), 1307–​1312. PMCID: PMC4315023. PMCID: PMC2566735.
Zhou B, Shu B, Yang J, et al. 2014. C-​reactive protein, interleukin-​6 and the risk
of colorectal cancer:  a meta-​analysis. Cancer Causes Control, 25(10),
1397–​1405. PMID: 25053407.
 37

21 Physical Activity, Sedentary Behaviors, and Risk of Cancer

STEVEN C. MOORE, CHARLES E. MATTHEWS, SARAH KEADLE,


ALPA V. PATEL, AND I-​MIN LEE

OVERVIEW refers to the specific kind of exercise, such as aerobic (e.g., walking
or running) or muscle-​ strengthening activities like weight lifting.
Current physical activity guidelines recommend that adults perform Frequency refers to the number of times the activity occurs, usually
at least 150 minutes per week of moderate-​intensity physical activ- in days per week. Duration refers to the time expended, usually in
ity (e.g., brisk walking), or 75 minutes per week of vigorous-​intensity hours per week. Intensity refers to the average energy cost of the activ-
activity (e.g., jogging), or an equivalent combination of these. In the ity; it is typically quantified using metabolic equivalents (METs).
United States and worldwide, many adults fail to meet these recom- The MET expresses an individual’s energy expenditure in terms of
mended activity levels, with deleterious consequences for health, multiples of energy expenditure at rest. An MET of 1 equals energy
including increased risk of some cancers. expenditure during quiet sitting; an MET of less than 3 corresponds
In this chapter, we review the epidemiologic evidence for links to energy expenditure during light-​intensity activities such as washing
between physical activity and cancer, emphasizing published meta-​ dishes; an MET of 3.0–​5.9 corresponds to energy expenditure during
analyses and the results of a recent large consortium-​based study. We walking or other moderate intensity activities; and an MET of 6.0+
find the evidence to be convincing that physical activity reduces risk corresponds to energy expenditure during vigorous-​intensity activities
of colon and female breast cancers, and probable that it reduces risk such as running. The MET values of at least 600 types of activities
of kidney and endometrial cancers. Moreover, physical activity has have been measured in controlled laboratory settings and compiled in
been associated with lower risk of cancers of the bladder, liver, gastric the Compendium of Physical Activities (https://​sites.google.com/​site/​
cardia, head and neck, esophagus (adenocarcinoma), and myeloma, compendiumofphysicalactivities).
myeloid leukemia, and non-​Hodgkin lymphoma. Mechanistic studies The volume or total amount of activity for a given individual rep-
indicate that an inadequate level of physical activity can increase levels resents cumulative energy expended during a specified time period,
of known cancer risk factors such as sex steroid hormones, insulin, and often per week. This is calculated by multiplying the intensity of each
inflammatory proteins. Physical activity likely has no substantial effect activity (in METs) by its duration (in hrs/​wk), and then summing
on risk of cancers of the ovary or lymphocytic leukemia. across all activities to obtain an overall MET-​hrs/​wk.
We also review evidence that sedentary behaviors (i.e., inactivity
while seated or reclining) may independently increase cancer risk.
Sedentary behaviors are most consistently associated with colon, EXPOSURE MEASUREMENT
endometrial, breast, and ovarian cancers; for other cancer sites, the
current data are inadequate to form conclusions. We also discuss how Habitual patterns of behavior can be estimated from records, such
physical activity can play a role in the treatment, rehabilitation, and as job classifications, or self-​reported information obtained by inter-
survivorship from cancer. view or self-​administered questionnaires. Epidemiologic studies also
Finally, we discuss future research directions, specifically the need increasingly incorporate personal devices such as accelerometers to
for studies that characterize the dose–​response relationships between track movement, or other objective measures of cardiorespiratory fit-
physical activity and cancer, that use new technologies to improve ness and muscular strength. Since there are as yet relatively few stud-
measurement of physical activity, and that evaluate sedentary behav- ies of cancer outcomes that have used these methods, this chapter
iors in relation to cancer risk. focuses primarily on studies of self-​reported or occupational indicators
of physical activity in relation to cancer.

DEFINITIONS AND CORE CONCEPTS Questionnaires


Physical activity is classically defined as “any bodily movement Nearly 100 different surveys/​ questionnaires have been developed
produced by skeletal muscles that results in energy expenditure” and validated to assess physical activity behaviors over the short (1–​
(Caspersen et  al., 1985). It includes both “exercise,” defined as 30  days) and long term (1–​10  years, lifetime) (Pereira et  al., 1997;
planned, structured, and repetitive activities done to improve health or Sallis and Saelens, 2000; van Poppel et al., 2010). These questionnaires
fitness (Physical Activity Guidelines Advisory Committee, 2008), and can be either self-​administered or completed using trained interviewers.
non-​exercise activities. A  variety of physical and social contexts or The time period of interest can be limited to the past year, or can cover
“domains” are used to categorize physical activity; these include activ- the entire life course, beginning in childhood and adolescence.
ities related to occupation, the household, education, transportation, Interviewer-​ administered questionnaires are commonly used in
and leisure time. The term “leisure-​time physical activity” reflects a case-​control studies where staff can make direct contact with partici-
broad class of activities that involve both exercise and recreational pants or their next of kin. The interviews can take from 10 minutes
pursuits. In practice, the terms “leisure time” and “recreational” are to 1 hour, depending on the length and complexity of the instrument.
sometimes used interchangeably, and recreational physical activity They typically collect more detailed information than can be obtained
may include exercise. Sedentary behaviors are behaviors done during with self-​administered questionnaires. Interviewers help to ensure that
waking hours that involve sitting or reclining and/​or a low level of respondents understand the questions and that the responses are logi-
energy expenditure (Lynch et al., 2010). cal (Sallis, 1997).
Physical activity and sedentary behaviors are characterized accord- Self-​administered questionnaires are widely used in prospective
ing to the type, frequency, duration, and intensity of activity. Type cohort studies, where collecting data by personal interview is cost

377
378

378 PART III:  THE CAUSES OF CANCER


prohibitive. Self-​administered questionnaires tend to be simpler and Health Interview Survey); and the National Health and Nutrition
more streamlined than interviewer-​ administered instruments; they Examination Survey (NHANES), which conducts both personal
generally assess the amount of time spent per week in only a few interviews and physical examinations (National Health and Nutrition
activity domains (e.g., leisure-​time, occupation), plus one or two addi- Examination Survey). All three surveys collect self-​reported informa-
tional questions to assess common daily activities (e.g., household tion on physical activity. In addition, NHANES uses accelerometers to
chores, sitting time). Self-​administered instruments tend to focus on measure movement over 7 days (Troiano et al., 2008).
the domain(s) that are reported most reliably and that pertain to spe- Different physical activity questionnaires obtain different informa-
cific hypotheses, such as the effects of leisure-​time physical activity tion. Because of this, estimates of the proportion of US adults who meet
on cancer risk. the recommended activity levels differ, especially when compared to
A limitation of both interviewer-​ based and self-​ administered accelerometry measurements (Carlson et al., 2009). According to the
questionnaires is that it is difficult for people to recall behaviors in BRFSS survey in 2011, 51.6% of US adults met the aerobic activity
past years (Matthews, 2002). The extent to which this contributes to guideline, 29.3% met the muscle-​strengthening guideline, and 20.6%
measurement error continues to be debated, however (Neilson et al., met both parts (Centers for Disease and Prevention, 2013). Similar
2008)  and likely varies by the type of activity. Leisure-​time and/​or prevalence estimates were reported in the 2013 NHIS, in which 49.4%,
vigorous activities are known to be more reliably reported than other 23.9%, and 20.4% of respondents, respectively, met the guidelines
aspects of daily life (Sallis and Saelens, 2000; van Poppel et  al., for aerobic activity, muscle strengthening, or both (National Center
2010), possibly because they represent discrete and structured enti- for Health Statistics, 2015). According to self-​reported data in the
ties (Matthews et al., 2012b). In contrast, lower-​intensity daily activi- NHANES 2005–​2006 collection cycle, 62.0% of respondents met the
ties (e.g., non-​exercise activity), often conducted in short bouts, are aerobic activity guideline (Tucker et al., 2011). However, only 9.6% of
reported less reliably (Matthews et al., 2012b). In prospective studies, participants met this guideline according to accelerometry measure-
errors in measuring physical activity that occurred many years before ments limited to activities that lasted at least 10 minutes, as speci-
the onset of disease are likely to be non-​differential with respect to dis- fied by the guidelines. This percentage increased to 44.6% when all
ease. Non-​differential misclassification of exposure typically attenu- measurements were included, without the restriction to ≥ 10-​minute
ates associations (Ferrari et al., 2007; Matthews et al., 2012b), thereby duration (Tucker et al., 2011).
underestimating relationships that are causal. Similar considerations Several factors likely contribute to the large discrepancy in the
apply to the assessment of sedentary behaviors. Structured sedentary proportion meeting the aerobic activity guideline (62.0% versus
behaviors, such as television viewing, appear to be reported more reli- 9.6%) when comparing self-​reported data (62.0%) to accelerometry
ably than sporadic, unstructured behaviors (Healy et al., 2011). measurements (9.6%). First, accelerometers only record continuous
movement and discount all periods of rest. For example, in a 2-​hour
tennis game, the accelerometer will register less than 2 hours of con-
Job Classifications/​Occupational Physical Activity tinuous movement, because of breaks during the game. In contrast,
Job titles provide some information about the amount of physical individuals may report the game as 2 full hours of activity. Second, the
activity or sedentary behavior while at work. For example, epidemi- algorithms used to process raw accelerometer information (i.e., activ-
ologists have compared the cancer risk among people with more sed- ity counts) into minutes of moderate-​to-​vigorous-​intensity activity
entary jobs (e.g., office and clerical workers) with workers whose jobs can influence prevalence estimates. When a different algorithm—​one
require physical activity (e.g., laborers, delivery workers) (Garabrant designed to capture a broader range of daily activities—​was applied
et al., 1984; Moradi et al., 1998). Employment records are especially to the NHANES data, it resulted in prevalence estimates that were
useful because they provide objective evidence about the jobs held more consistent with the estimates based on self-​report (Matthews
and the duration of each job. Limitations of job titles are that they do et al., 2005; Watson et al., 2014). Comparable data do not exist for
not specify the actual behavior(s) involved, the variations in activity muscle-​strengthening activities since the accelerometer cannot mea-
among individuals with the same title, and changes in employment sure these activities.
over time. While estimates of the proportion of people meeting the physical
activity guidelines may vary, the descriptive data consistently indicate
that Americans are sedentary during at least half of their waking day.
GUIDELINES FOR PHYSICAL ACTIVITY In the 2003–​2004 data collection cycle of NHANES, time expended
in sedentary behaviors was measured objectively by accelerometry
In 2008, the US government released “Guidelines for Physical Activity (Matthews et al., 2008). Among the more than 6000 participants with
for Americans” (US Department of Health and Human Services). at least one 10-​hour day of monitoring, 54.9% of the monitored time
These guidelines recommended the amount and intensity of aerobic was sedentary. Most people in the United States spent the majority of
and muscle-​strengthening activities considered optimal to maintain their time on behaviors that expend very little energy.
health in adults. For aerobic activity, adults were advised to engage Internationally, many other countries have also collected self-​
in at least 150 minutes per week of moderate-​intensity physical activ- reported information on physical activity. Self-​reported data collected
ity (e.g., brisk walking), or 75 minutes per week of vigorous-​intensity for the WHO global health observatory data repository show that
activity (e.g., jogging), or some combination thereof that expends an 31.1% of the adult population worldwide in 2008 did not meet the
equivalent amount of energy. For muscle-​strengthening activities, the guideline for aerobic activity (i.e., 68.9% did) (Hallal et  al., 2012).
guidelines recommend muscle-​strengthening activities that involve all The percentage failing to meet the guideline ranged from 17.0% in
major muscle groups on 2 or more days a week. Similar guidelines Southeast Asia to approximately 43% in the Americas and the eastern
have been adopted by other countries worldwide for general health Mediterranean region.
recommendations (World Health Organization, 2010), and specifically
for cancer prevention (World Cancer Research Fund and American
Institute for Cancer Research 2007). MECHANISMS OF CARCINOGENESIS

Physical activity is a physiologically complex behavior that involves


SURVEYS OF PHYSICAL ACTIVITY numerous highly coordinated biological responses. An acute bout of
physical activity increases oxygen consumption by up to 15-​fold
Several health surveys monitor physical activity in the United States. compared with the resting state, accelerates ATP turnover in skel-
These include the Behavioral Risk Factor Surveillance System etal muscle, increases secretion of glucose into the bloodstream and
(BRFSS), a telephone survey conducted by random-​ digit dialing uptake of glucose into muscles, and modulates the levels and activ-
(Behavioral Risk Factor Surveillance System); the National Health ity of multiple signaling factors such as insulin, growth factors, and
Interview Survey (NHIS), which uses personal interviews (National hormones (Hawley et  al., 2014). Because physical activity results
 379

Physical Activity, Sedentary Behaviors, and Risk of Cancer 379


in a variety of interrelated biological effects that are still not com- elevated levels of DNA-​damaging reactive oxygen species (Coussens
pletely understood (Hawley et  al., 2014), research on the mecha- and Werb, 2002) and chemokines that stimulate cellular proliferation
nisms by which physical activity may affect cancer risk continues and promote the survival of genetically damaged cells (Mantovani
to evolve. Research to date has focused primarily on the effects of et al., 2008). In epidemiologic studies, increased levels of circulating
physical activity on sex steroid hormones, insulin and insulin-​like pro-​inflammatory factors, such as C-​reactive protein (CRP), interleu-
growth factors, inflammation, and weight change (Friedenreich kin 6 (IL6), tumor necrosis factor ɑ (TNFɑ), and decreased levels of
et al., 2010; McTiernan, 2008). anti-​inflammatory factors like adiponectin have been associated with
increased cancer risk (Gunter et al., 2015; Ho et al., 2012; Nimptsch
et al., 2015; Ollberding et al., 2013; Toriola et al., 2013; Wang et al.,
Sex Steroid Hormones 2011). It is unclear, however, whether regular physical activity modi-
Sex steroid hormones have powerful proliferative and mutagenic fies the effects of local and/​or systemic inflammation. In randomized
effects. Excessive levels of estrogens, in particular, increase risk of clinical trials, assignment to exercise for 12 months has had inconsis-
breast and endometrial cancers (Chlebowski et al., 2003; Weiderpass tent effects on levels of C-​reactive protein, with some studies report-
et al., 1999; Yager and Davidson, 2006). A meta-​analysis of random- ing an inverse association (Campbell et  al., 2009), but others not
ized exercise intervention studies of 3–​12 months duration (Ennour-​ (Campbell et al., 2008; Kelley and Kelley, 2006). Randomized exer-
Idrissi et al., 2015) showed that exercise reduces concentrations of cise trials among postmenopausal women also had little effect on adi-
total estrogens (21 studies), free estradiol (5 studies), free testos- ponectin concentrations (Abbenhardt et al., 2013; Friedenreich et al.,
terone (9 studies), androstenedione (7 studies), and DHEA sulfate 2011). Overall, while inflammation may ultimately prove to mediate
(8 studies), and increases concentrations of sex hormone–​binding some the effects linking low physical activity to cancer risk, the cur-
globulin (14 studies). The effect of exercise on total estrogens, rent evidence is mixed.
while statistically significant, was relatively weak and appeared
to be mediated through exercise-​induced changes in body weight,
Prevention of Weight Gain
rather than through exercise itself. In contrast, its effects on free
estradiol (not bound to proteins) and sex hormone–​binding globu- Excess body weight increases the risk of more than a dozen different
lin are greater than its effect on total estrogens, and are indepen- types of cancer (World Cancer Research Fund and American Institute
dent of changes in body weight. The effects of physical activity on for Cancer Research, 2007) (Chapter 20). Several of the mechanisms
sex hormones have been documented in randomized clinical trials. thought to mediate the effects of excess body fat on cancer risk are
These, combined with definitive evidence that sex hormones influ- also affected by physical inactivity. For example, whereas excess
ence carcinogenesis, provide strong, albeit indirect, evidence that adiposity increases the peripheral aromatization of testosterone and
sex hormones mediate at least part of the effect of physical activity androstenedione in fat tissue and is a major source of estrogen in
on hormonally related cancers. postmenopausal women (Judd et al., 1982; Key et al., 2015), physi-
cal activity has been shown to reduce the concentrations of free
estradiol, free testosterone, and other sex hormones in randomized
Insulin and Other Growth Factors Related clinical trials. This hormonal effect is thought to contribute to the
to Insulin Resistance increased risk of endometrial and postmenopausal breast cancer in
obese women, and to the reductions in risk of these cancers in physi-
Insulin is a known mitogen (Ish-​Shalom et al., 1997; Mu et al., 2012); cally active women (Brown and Hankinson, 2015; Key et al., 2003).
administration of exogenous insulin promotes breast tumor growth in Excess adiposity and physical activity also have opposing effects on
animal models (Shafie and Hilf, 1981). Chronic hyperinsulinemia is hyperinsulinemia and perhaps inflammation (Calle and Kaaks, 2004;
also associated with reduced hepatic secretion of sex hormone–​binding Kahn and Flier, 2000).
globulin (Pugeat et al., 1991), which increases levels of bioavailable A meta-​analysis of 16 randomized exercise intervention studies,
steroidal sex hormones. In observational studies, higher levels of insu- including a total of 1737 participants, has shown that exercise, usu-
lin have been associated with increased risk of breast, endometrial, ally for a period of 3–​12 months, results in an average body weight
pancreatic, and colorectal cancers (Godsland, 2010; Gunter et  al., loss of 1.8 kg (Ennour-​Idrissi et al., 2015), a modest but discernible
2008, 2009; Kaaks and Lukanova, 2001; Stolzenberg-​Solomon et al., difference in weight. In longer-​term studies, young adults (age 18–​
2005). In two randomized clinical trials of physical activity, assign- 30 years) who maintain a high level of activity over 20 years gain less
ment to the exercise group for 12  months reduced circulating levels body weight compared to their less active counterparts. Active men
of insulin by 13% and 16% in postmenopausal women (Frank et al., gained 2.6 fewer kilograms and women gained 6.1 fewer kilograms
2005; Friedenreich et al., 2011). The ability of exercise to reduce insu- (Hankinson et al., 2010). In a study of middle-​aged women (mean age
lin levels may partly explain why physical activity is associated with 54.2 years), a high level of physical activity was associated with 0.12
lower risk of cancers thought to be insulin-​related. However, it is dif- fewer kilograms gained per 3 years (Lee et al., 2010). In sum, excess
ficult to separate the effects mediated by insulin from other factors body weight contributes to increased cancer risk, and physical activ-
closely related to insulin. ity at the levels typically performed in the general population helps to
Insulin-​like growth factor (IGF)-​ 1 is a hormone with high reduce weight gain.
sequence similarity to insulin, but with even stronger mitogenic
effects (Zapf et  al., 1984). Molecular epidemiological studies
have confirmed an association between high circulating levels of Other Mechanisms
IGF-​1 and modest increases in risk of cancers of the breast, pros-
tate, and colorectum (Endogenous Hormones and Breast Cancer The recent advent of new “omics”-​based technologies, such as pro-
Collaborative Group, 2010). However, randomized clinical trials of teomics and metabolomics, has broadened the opportunities to study
exercise suggest that exercise does not affect the circulating levels the endocrine functions of skeletal muscle. Myokines are cytokines
of IGF-​1 (McTiernan et al., 2005). Therefore IGF-​1 seems unlikely or proteoglycan peptides that are produced and released by muscle
to mediate the inverse relationship between physical activity and cells in response to muscular contraction. Different types of myo-
risk of these cancers. kines have different effects on fat metabolism. For example, irisin
stimulates development of brown fat, and brain-​derived neurotrophic
factor (BDNF) helps to modulate fatty acid oxidation (Pedersen and
Febbraio, 2012). Prolonged physical activity may also increase β-​
Inflammation oxidation of fatty acids within the mitochondria by increasing lipolysis
Chronic inflammation increases the risk of multiple types of cancer and fatty acid oxidation (Hawley et al., 2014). It is presently unclear
(Chapter  25). Inflammation creates a tissue microenvironment with whether or how these effects may relate to cancer.
380

380 PART III:  THE CAUSES OF CANCER

PHYSICAL ACTIVITY AND RISK shown in Figure 21–​1, but the results are discussed in the chapter in the
OF DEVELOPING CANCER context of each specific type of cancer.
Strengths of the 2016 pooled analysis are that it relied entirely on
Our review of physical activity in relation to cancer risk is based on data from prospective studies, eliminated several sources of hetero-
the findings of the most recent and comprehensive meta-​analyses geneity, and increased the number of cases identified prospectively
of the topic (Table 21–​1) and on a pooled analysis of 12 large pro- for some types of cancer. The latter factor was particularly impor-
spective cohorts participating in the National Cancer Institute Cohort tant for uncommon and rare types of cancer. For example, the 2016
Consortium (Moore et  al., 2016). The number of cases in both pooled analysis nearly doubled the number of cases in the literature
approaches, references for the meta-​analyses, and degree of overlap for esophageal adenocarcinoma (N  =  899 vs. 454)  and liver cancer
between the meta-​and pooled analyses are shown in Table 21–​1. (1384 vs. 628). The pooled analysis eliminated heterogeneity related
to study type (e.g., case-​control vs. prospective cohort), physical activ-
ity domain (leisure-​time vs. occupational activity), and the categories
Description of the Studies being contrasted (e.g. tertiles vs. quintiles).
All of the meta-​analyses listed in Table 21–​1 used a similar approach
to measure the risk ratios associated with physical activity and site-​ Results from the Meta-​and Pooled Analyses
specific cancers. In each, the highest level of physical activity cor-
responds to the top quintile, quartile, or tertile of activity in the Table 21–​2 shows the multivariable relative risk (RR) estimates and
populations studied, and the lowest level corresponds to the bottom 95% confidence intervals (CIs) for high versus low levels of physical
quintile, quartile, or tertile. Consequently, the relative risks from activity by cancer type as reported by the meta-​and pooled analy-
the meta-​analyses can be interpreted as approximately the contrast ses. The results from the pooled analysis are presented graphically in
between the top and bottom quartile of physical activity in both the Figure 21–​1. The similarity of findings from the meta-​analyses and
prospective and retrospective studies. the 2016 pooled analysis strengthens confidence that these results are
With one exception (Zhong et  al., 2014), the meta-​analyses com- reproducible.
bined all of the domains of physical activity, although data on
leisure-​time physical activity predominate. Occupational activity was
Summary of Evidence
specified in some studies. Several of the meta-​analyses published rel-
ative risk estimates for specific domains; we present these findings Based on these data, we characterized the evidence for a causal rela-
where appropriate. tionship between physical activity and reduced risk of various types of
The 2016 pooled analysis included 1.44 million participants, aged cancer into one of six levels: convincing, probable, limited–​suggestive,
19–​98  years (median 59  years). Subjects were drawn from 12 pro- uncertain–​may be confounded, inconsistent, and substantial effect
spective cohorts in the United States and Europe, and followed for a unlikely. The criteria used to define these evidence levels are shown in
median of 11 years. Overall, 186,932 incident cancers were diagnosed Table 21–​3. For three of the levels (convincing, probable, and limited–​
during follow-​up. suggestive), the criteria were adapted from those used by the World
The pooled analysis compared cancer risk in the 90th to the 10th Cancer Research Fund (WCRF) and the American Institute for Cancer
percentile of leisure-​time physical activity and found that greater Research (AICR) in their review entitled “Food, Nutrition, Physical
activity was associated with lower cancer risk for 13 of the 26 sites Activity, and the Prevention of Cancer: A Global Perspective” (World
or subsites examined. The overall summary figure for the analysis is Cancer Research Fund and American Institute for Cancer Research,

Table 21–​1. Case Numbers in Meta-​analyses that Examine Physical Activity and Cancer Associations and in a 2016 Pooled Analysis1

Cases in Meta-​ Cases Overlapping


Analysis, Prospective + Cases in Meta-​Analysis, Cases in Pooled Between Meta-​analysis and
Cancer Reference Case-​Control Prospective Studies Analysis Pooled Analysis

Colon Wolin et al., 2009 Not specified Not specified 14,160 6,612
Breast Wu et al., 2013 63,786 63,786 35,178 21,891
Kidney Behrens and Leitzmann, 2013 10,756 6104 4548 1,238
Endometrial Schmid et al., 2015 19,558 10,740 5346 3,309
Bladder Keimling et al., 2014 27,784 25,174 9073 1,831
Liver Behrens et al., 20132 628 628 1384 628
Esophageal Behrens et al., 2014 965 454 899 454
adenocarcinoma
Gastric cardia Behrens et al., 2014 844 436 790 313
Myeloma Jochem et al., 2014 1362 1362 2161 814
Myeloid leukemia N/​A 0 0 1692 0
Non-​Hodgkin lymphoma Jochem et al., 2014 9447 5243 6953 3,506
Head and neck Leitzmann et al., 20082 1249 1249 3985 1,249
Lung Brenner et al., 2016 20,169 14,306 19,133 8,605
Malignant melanoma N/​A 0 0 12,438 0
Pancreas Behrens et al., 2015 10,501 8091 4186 1,877
Prostate Liu et al., 2011 88,294 74,942 46,890 20,691
Rectum Robsahm et al., 2013 8698 8698 5531 2,463
Brain Niedermaier et al., 2015 2538 1194 2110 257
Thyroid Schmid et al., 2013 2250 1329 1829 770
Ovary Zhong et al., 2014 9459 2467 2880 801
Lymphocytic leukemia N/​A 0 0 2160 0

1
Moore et al. (2016). Association of leisure-​time physical activity with risk of 26 types of cancer in 1.44 million adults. JAMA Intern Med, 176(6), 816–​825.
2
Case numbers are based on the largest prospective cohort study, as no meta-​analyses or reviews have been published.
 381

Physical Activity, Sedentary Behaviors, and Risk of Cancer 381


Cancer # of Studies Cases HR (95% Cl) Ptrend Pheterogeneity
Esophageal adenocarcinoma 5 899 0.58 (0.37–0.89) 0.01 0.01

Gallbladder 6 382 0.72 (0.51–1.01) 0.06 0.29

Liver 10 1,384 0.73 (0.55–0.98) 0.04 0.02

Lung 12 19,133 0.74 (0.71–0.77) < 0.001 0.47

Kidney 11 4,548 0.77 (0.70–0.85) < 0.001 0.40

Small intestine 7 503 0.78 (0.60–1.00) 0.05 0.85

Gastric cardia 6 790 0.78 (0.64–0.95) 0.02 0.99


Endometrial 9 5,346 0.79 (0.68–0.92) 0.003 < 0.01

Esophageal squamous 6 442 0.80 (0.61–1.06) 0.12 0.78

Myeloid leukemia 10 1,692 0.80 (0.70–0.92) 0.002 0.78


Myeloma 9 2,161 0.83 (0.72–0.95) 0.008 0.36

Colon 12 14,160 0.84 (0.77–0.91) < 0.001 0.01


Head and neck 11 3,985 0.85 (0.78–0.93) < 0.001 0.45

Rectum 12 5,531 0.87 (0.80–0.95) 0.001 0.38


Bladder 12 9,073 0.87 (0.82–0.92) < 0.001 0.84

Breast 10 35,178 0.90 (0.87–0.93) < 0.001 0.30

Non-Hodgkin lymphoma 11 6,853 0.91 (0.83–1.00) 0.05 0.18

Thyroid 11 1,829 0.92 (0.81–1.06) 0.26 0.48

Gastric non-cardia 7 1,428 0.93 (0.73–1.19) 0.56 0.09

Soft tissue 10 851 0.94 (0.67–1.31) 0.70 0.03


Pancreas 10 4,186 0.95 (0.83–1.08) 0.40 0.14

Lymphocytic leukemia 10 2,160 0.98 (0.87–1.11) 0.73 0.99

Ovary 9 2,880 1.01 (0.91–1.13) 0.81 0.98

Brain 10 2,110 1.06 (0.93–1.20) 0.41 0.43

Prostate 7 46,890 1.05 (1.03–1.08) < 0.001 0.90

Malignant melanoma 12 12,438 1.27 (1.16–1.40) < 0.001 0.02

0.3 0.6 1 0.5


Hazard Ratio (90th vs 10th percentile of physical activity)

Figure 21–​1.  Summary multivariable hazard ratios (HR) and 95% confidence intervals (CI) for a higher (90th percentile) versus lower (10th percentile)
level of leisure-​time physical activity by cancer type in a pooled analysis of 12 prospective studies (Moore et al., 2016).

2007). Important considerations included the number of cases in pro- Colon Cancer


spective studies, the biological plausibility of the association (demon-
strated for many cancers in mechanisms section above), evidence for Cancers of the large bowel were the most commonly studied can-
a dose–​response relationship, and the likelihood of residual confound- cers in early epidemiologic research on physical activity and cancer
ing. Our findings are summarized in Table 21–​3. (Chapter  23, Lee and Oguma, 2006). The inverse relationship with
Under these criteria, there was convincing evidence that physical physical activity was consistently stronger for colon than for rectum
activity reduces the risk of colon and female breast cancers, and there cancer; we therefore discuss rectal cancer separately below, as part of
was probable evidence that activity protects against the development of “other cancers.”
kidney and endometrial cancers. There was some evidence that physi- The meta-​analysis by Wolin and colleagues (2009) included cases
cal activity may be inversely related to cancers of the bladder, liver, from 52 publications based on 28 cohort and 24 case-​control studies
gastric cardia, and head and neck, as well as esophageal adenocar- conducted in the United States, Europe, and Asia. The total number of
cinoma, myeloma, myeloid leukemia, and non-​Hodgkin lymphoma. cases was not specified. Overall, the most active people had 24% lower
The evidence was “uncertain–​may be confounded” for lung cancer risk of colon cancer than those who were least active (meta-​RR = 0.76;
and melanoma because of concerns that observed associations were 95% CI: 0.72, 0.81) (see Figure 21–​2). The findings were similar for
influenced by residual confounding by smoking (lung cancer) and men (RR = 0.76; 95% CI: 0.71, 0.82) and women (RR = 0.79; 95%
sun exposure (melanoma). The evidence was inconsistent for cancers CI: 0.71, 0.88).
of the pancreas, prostate, rectum, brain, and thyroid; for these sites, The inverse association was stronger in the meta-​analysis of case-​
unexplained heterogeneity in the associations was detected across the control (RR  =  0.69; 95% CI:  0.65, 0.74) than that of cohort studies
studies. Finally, for ovarian cancer, and lymphocytic leukemia, evi- (RR  =  0.83; 95% CI:  0.78, 0.88), most likely reflecting recall bias
dence indicates that a substantial effect of physical activity on risk is among participants in case-​control studies. Investigators also exam-
unlikely. ined domain-​specific physical activity in the meta-​analysis with signif-
We review the findings for each cancer in the following, presented icant risk reductions observed for both occupational (RR = 0.78; 95%
in order of the strength of the total evidence for a physical activity and CI:  0.74, 0.83) and leisure-​time physical activity (RR  =  0.77; 95%
risk association. CI: 0.72, 0.82).
382

382 PART III:  THE CAUSES OF CANCER


Table 21–​2. Multivariable Relative Risks (RRs) and 95% Confidence Intervals (CIs)
for High Versus Low Levels of Physical Activity by Cancer Type According to Recent
Meta-​analysis and Moore et al.

RR (95% CI) HR (95% CI)


Cancer Meta-​analysis1 Moore et al.2

Colon 0.76 (0.72, 0.81) 0.84 (0.77, 0.91)


Breast 0.87 (0.83, 0.92) 0.90 (0.87, 0.93)
Kidney 0.88 (0.79, 0.97) 0.77 (0.70, 0.85)
Endometrial 0.80 (0.75, 0.85) 0.79 (0.68, 0.92)
Bladder 0.85 (0.74, 0.98) 0.87 (0.82, 0.92)
Liver3 0.64 (0.49, 0.84) 0.73 (0.55, 0.98)
Esophageal adenocarcinoma 0.79 (0.66, 0.94) 0.58 (0.37, 0.89)
Gastric cardia 0.83 (0.69, 0.99) 0.78 (0.64, 0.95)
Myeloma 0.86 (0.68, 1.09) 0.83 (0.72, 0.95)
Myeloid leukemia Not available 0.80 (0.70, 0.92)
Non-​Hodgkin lymphoma 0.91 (0.82, 1.00) 0.91 (0.83, 1.00)
Head and neck3 0.89 (0.74, 1.06) 0.85 (0.78, 0.93)
Lung 0.76 (0.69, 0.85) 0.74 (0.71, 0.77)
Malignant melanoma Not available 1.27 (1.16, 1.40)
Pancreas 0.93 (0.88, 0.98) 0.95 (0.83, 1.08)
Prostate 0.90 (0.84, 0.95) 1.05 (1.03, 1.08)
Rectum 0.98 (0.88, 1.08) 0.87 (0.80, 0.95)
Brain 0.86 (0.76, 0.97) 1.06 (0.93, 1.20)
Thyroid 1.06 (0.79, 1.42) 0.92 (0.81, 1.06)
Ovary 0.92 (0.84, 1.00) 1.01 (0.91, 1.13)
Lymphocytic leukemia Not available 0.98 (0.87, 1.11)

1
Results incorporate multiple study designs (i.e., case-​control studies and prospective studies) and
multiple types of physical activity, including occupational and leisure-​time (recreational) physical
activity. References are provided in Table 21–​1.
2
Results based on only prospective studies and leisure-​time physical activity.
3
RR derived from the largest prospective cohort study in the literature as no meta-​analyses or reviews
have been published.

The meta-​analysis by Wolin et al. (2009) was unable to examine compared to those expending < 2 MET-​hours/​week (approximately
the dose–​response relationship in the combined studies because the 0.5 hours/​week).
methods used to measure physical activity varied. The research- The relative risk estimates reported by the individual studies were
ers did mention that most of the analyses of cohort studies tested adjusted for multiple potential confounders that varied across studies.
formally for a dose–​response relation, yet fewer than half reported The majority of studies included obesity as a potential confounder. The
a statistically significant inverse trend. Of the 10 case-​control findings suggested that the inverse relationship between physical activ-
studies that also tested for a trend across activity levels, four ity and colon cancer risk was independent of its effects on body weight.
reported a significant inverse trend in at least one subgroup. In the A meta-​analysis by Boyle et  al. (2012) investigated whether the
Nurses’ Health Study (Wolin et al., 2007), a 23% risk reduction—​ reduction in colon cancer risk associated with physical activity differed
comparable to the overall meta-​analysis results—​was observed for proximal and distal cancers. Cancers that arise in these two subsites
among women expending 21.5 MET-​hours/​week of leisure-​time differ with regard to their morphologic, molecular, and genetic charac-
physical activity (equivalent to 5–​6 hours/​week of brisk walking), teristics. Among the 12 cohort and case-​control studies included, risk

Table 21–​3. Summary of Evidence on Physical Activity and its Association with Risk of Cancer

Evidence Level Criteria for Evidence Level Cancers at Evidence Level


Convincing > 10,000 cases in prospective studies Colon, Female breast
Evidence for a dose–​response relationship
Biologically plausible
Low possibility of confounding
Probable > 5000 cases in prospective studies Kidney, Endometrium
Evidence for a dose–​response relationship
Biologically plausible
Some possibility of confounding
Limited–​suggestive > 500 cases in prospective studies Bladder, Liver, Esophageal adenocarcinoma,
Evidence for a relationship Gastric cardia, Myeloma, Myeloid leukemia,
Biologically plausible Non-​Hodgkin lymphoma, Head and neck
Some possibility of confounding
Uncertain–​may be confounded Evidence for a relationship Lung, Melanoma (increased risk)
Results seem likely to be confounded
Sensitivity analyses do not rule out confounding
Inconsistent Results vary by study type or over time Pancreas, Prostate, Rectum, Brain, Thyroid
Reasons for variability are unclear
Substantial effect unlikely > 2000 cases in prospective studies Ovary, Lymphocytic leukemia
Aggregate RR of 0.95 to 1.05
 38

Physical Activity, Sedentary Behaviors, and Risk of Cancer 383


Category # of Studies RR (95% Cl)

Overall 52 0.76 (0.72, 0.81)

Cohort 28 0.83 (0.78, 0.88)

Case-control 24 0.69 (0.65, 0.74)

Men 30 0.76 (0.71, 0.82)

Women 30 0.79 (0.71, 0.88)

Occupational 32 0.78 (0.74, 0.83)

LTPA 26 0.77 (0.72, 0.82)

0 1 2
Relative risk (95% Cl)
High vs. low physical activity

Figure 21–​2.  Colon cancer and its association with physical activity.

of proximal colon cancer was 27% lower among the most active peo- menopausal status, body mass index, or period of life during which
ple compared to the least active (random effects summary RR = 0.73; activity occurred. The point estimates were lower for women with
95% CI: 0.66, 0.81). This reduction in risk was indistinguishable from premenopausal breast cancer (RR  =  0.77; 95% CI:  0.69, 0.86)
the 26% lower risk of distal colon cancer (RR = 0.74; 95% CI: 0.68, than postmenopausal disease (RR = 0.87; 95% CI: 0.82, 0.92), and
0.80). As with the other analyses described earlier, the findings were in those with a BMI < 25  kg/​m2 (RR  =  0.72; 95% CI:  0.65, 0.90)
similar in men and in women, and the inverse relationship was stronger than in women who were overweight or obese (BMI ≥ 25  kg/​m2)
in case-​control than cohort studies. (RR  =  0.93; 95% CI:  0.87, 0.98). Finally, the pooled RR estimate
The results of the pooled analysis in 2016 strongly support the find- associated with physical activity was slightly lower for women age
ings of Wolin et  al. (2009) with respect to the overall reduction in ≥ 50 years (RR = 0.83; 95% CI: 0.76, 0.91) than for women age <
risk. The relative risk and 95% confidence interval estimates for high 25 years (RR = 0.90; 95% CI: 0.81, 1.02). However, no formal tests
versus low levels of leisure-​time activity were 0.84 (95% CI:  0.77, of heterogeneity were reported, and the 95% CI overlapped in most
0.91) for colon cancer. This is nearly identical to the hazard ratio of subgroup analyses.
0.83 observed in the meta-​analysis of prospective studies by Wolin Associations also varied according to estrogen receptor (ER) and
et al. (2009). progesterone receptor (PR) positivity, with lower point estimates for
In summary, based on the available evidence, there is convinc- ER–​/​PR–​cancers (RR = 0.77; 95% CI: 0.65, 0.90) (8 studies) than for
ing support for a causal relationship between physical activity and ER+/​PR+ cancers (RR  =  0.93; 95% CI:  0.87, 0.98) (7 studies). The
significance of this is uncertain, given the limited number of studies in
reduced risk of developing colon cancer. This conclusion is consist- these analyses and the lack of a mechanistic basis for the finding.
ent with those of several expert panels (IARC Handbooks of Cancer Relative risk estimates in studies were appropriately adjusted for
Prevention, 2002; Kushi et al., 2012; World Cancer Research Fund and potential confounders, including body mass index (BMI), menopau-
American Institute for Cancer Research, 2007). sal status and hormone therapy, reproductive history, and family his-
tory. Where available, the authors also presented relative risks from
analyses adjusted only for age and found substantively similar results
Female Breast Cancer (RR  =  0.85; 95% CI:  0.79, 0.90), indicating that adjustment for the
The only meta-​analysis that formally quantified the association between available confounders had little overall effect on the results.
physical activity and breast cancer risk was by Wu et  al. (2013). It Friedenrich et al. (2011) systematically reviewed the association of
included 63,786 breast cancer cases drawn from 31 prospective cohorts physical activity with breast cancer risk in 75 cohort and case control
in North America, Europe, and Asia. Overall, the most active women studies conducted worldwide. In this review, women who were the
had a 13% lower risk of developing breast cancer than the least active most physically active had, on average, 25% lower risk of developing
women (RR = 0.87; 95% CI: 0.83, 0.92; see Figure 21–​3). Associations breast cancer as compared with the least active women. These inverse
were similar for recreational activity (RR = 0.87; 95% CI: 0.83, 0.91; relationships were slightly stronger in the case control studies (RR 0.7)
25 studies) and occupational activity (RR = 0.84; 95% CI: 0.73, 0.96; than in the cohort studies (RR 0.8). Overall, the results were generally
7 studies). The analysis of recreational activity used restricted cubic consistent with those of Wu et al. (2013).
spline models to show that the dose–​response association was approxi- The 2016 pooled analysis reported a relative risk estimate of 0.90
mately linear (P for non-​linearity = 0.45); each additional increment of (95% CI: 0.87, 0.93) comparing breast cancer risk among women with
10 MET-​hours/​week correlated with a 3% lower risk of breast cancer. the highest versus lowest level of leisure-​time activity. This is similar
The associations were stronger for vigorous-​intensity physical activity to the hazard ratio of 0.87 reported from the meta-​analysis by Wu et al.
(RR = 0.85; 95% CI: 0.80, 0.90; 21 studies) than for moderate-​intensity (2013).
activity (RR = 0.95; 95% CI: 0.90, 0.99; 16 studies). In summary, the evidence is convincing that higher levels of physi-
Wu et  al. (2013) also examined whether the inverse associa- cal activity are associated with lower risk of breast cancer. Additional
tion between breast cancer and physical activity was modified by data on the benefits of vigorous-​intensity versus moderate-​intensity
384

384 PART III:  THE CAUSES OF CANCER


Category # of Studies RR (95% Cl)

Overall 31 0.87 (0.83, 0.92)

Premenopausal 6 0.77 (0.69, 0.86)

Postmenopausal 17 0.87 (0.82, 0.92)

ER-/PR- 7 0.77 (0.65, 0.90)

ER+/PR+ 8 0.93 (0.87, 0.98)

Occupational 7 0.84 (0.73, 0.96)

LTPA 25 0.87 (0.83, 0.91)

0 1 2
Relative risk (95% Cl)
High vs. low physical activity

Figure 21–​3.  Female breast cancer and its association with physical activity.

activities, and on how associations vary according to ER/​PR and other The risk of endometrial cancer risk was 20% lower for women in the
breast cancer subtypes, would help to refine understanding of this highest compared to the lowest category of physical activity, as shown
relationship. by the summary estimate of relative risk (RR = 0.80; 95% CI: 0.75,
0.85) in Figure 21–​4.
Exposure assessment in studies of endometrial cancer and physical
Kidney Cancer activity was based on self-​administered questionnaires (16 studies),
Behrens and Leitzmann (2013) identified a total of 19 studies (11 interview-​assisted questionnaires (10 studies), occupational job titles
cohort, 8 case control) from North America, Europe, and Asia that (4 studies), or a combination of job titles and interview-​assisted ques-
examined the association between physical activity and renal cell tionnaires (3 studies).
carcinoma, which accounts for approximately 90% of all kidney Physical activity was inversely associated with endometrial cancer
cancers. The analysis was based on 10,756 cases. Overall, physi- risk across all domains of activity. In 22 studies of recreational physi-
cal activity was inversely associated with risk of renal cell cancer cal activity there was a 16% reduction in the risk of endometrial cancer
(RR = 0.88; 95% CI: 0.79, 0.97). Results were similar for the pro- (RR = 0.84; 95% CI: 0.78, 0.91) comparing the highest to the lowest
spective studies (RR = 0.87; 95% CI: 0.79, 0.97) and case-​control category of activity; in 19 studies of occupational activity there was a
studies (RR  =  0.89; 95% CI:  0.74, 1.06). The point estimate was 19% reduction in risk (RR = 0.81; 95% CI: 0.75, 0.87), and in 7 studies
somewhat lower (RR = 0.78; 95% CI: 0.66, 0.92) in studies judged of household activity there was a 30% reduction in risk (RR = 0.70;
to be of higher quality according to a predetermined rubric than 95% CI:  0.47, 1.02). Ten studies specifically examined walking and
in the overall analysis. The dose–​response relationship was not reported an 18% reduction in risk (RR = 0.82; 95% CI: 0.69, 0.97).
assessed. The strength of association was similar for recreational No differences in risk were observed among studies with different
and occupational physical activity, and did not appear to vary intensity of physical activity. Two studies included light activity and
according to the age when the activity occurred. Adjustments for reported a 35% risk reduction (RR = 0.65; 95% CI: 0.49, 0.86), eight
smoking, BMI, hypertension, and diabetes had little effect on the assessed moderate-​to-​vigorous physical activity and reported a 17%
association. reduction (RR = 0.83; 95% CI: 0.71, 0.96) and eight included vigorous
In the 2016 pooled analysis, leisure-​ time physical activity was activity and reported a 20% reduction (RR = 0.80; 95% CI: 0.72, 0.90).
inversely associated with kidney cancer (RR  =  0.77; 95% CI:  0.70, The dose–​ response relationship was examined separately in nine
0.85). This association was stronger than that observed by Behrens studies that expressed activity in terms of MET-​hours per week. The
and Leitzmann (2013) when comparing high versus low levels of over- inverse association between the level of physical activity and risk of
all activity (RR = 0.88; 95% CI: 0.79, 0.97), and recreational activity endometrial cancer appeared to be non-​linear, with no further decrease
(RR = 0.88; 95% CI: 0.77, 1.00). The findings from the 2016 pooled in risk observed at activity levels higher than 20 MET-​hours per week.
analysis strengthen the evidence of an inverse association between As with other end points, the inverse relationship with endometrial
physical activity and kidney cancer. cancer appeared to be stronger in the case-​control studies (RR = 0.72;
95% CI: 0.64, 0.80) than in the prospective cohort studies (RR = 0.84;
95% CI: 0.78, 0.91), suggesting some influence from reporting bias.
Most studies (N = 29) were conducted in North America, or Europe
Endometrial Cancer (N = 29); three studies were from Asia and one from South America.
A meta-​analysis by Schmid et  al. (2015) identified 18 cohort stud- No significant differences were observed in the associations among
ies, 1 case-​cohort study, and 14 case-​control studies that examined geographic regions, based on the limited number of studies currently
the association between physical activity and endometrial cancer risk. available in middle and low-​income countries. There was no evidence
Collectively, these studies included 19,558 endometrial cancer cases. that the association differed by study size or quality.
 385

Physical Activity, Sedentary Behaviors, and Risk of Cancer 385


Category # of Studies RR (95% Cl)
Overall 33 0.80 (0.75, 0.85)

Cohort 19 0.84 (0.78, 0.91)


Case-control 14 0.72 (0.64, 0.80)

Premenopausal 4 0.74 (0.49, 1.13)


Postmenopausal 7 0.81 (0.67, 0.97)

Occupational 19 0.81 (0.75, 0.87)


LTPA 22 0.84 (0.78, 0.91)

Adjusted for adiposity 24 0.79 (0.73, 0.85)


Not adjusted for adiposity 9 0.83 (0.72, 0.97)

0 1 2
Relative risk (95% Cl)
High vs. low physical activity

Figure 21–​4.  Endometrial cancer and its association with physical activity.

One potential confounder in the analysis of endometrial cancer is different between the prospective studies (RR = 0.89; 95% CI: 0.80,
unopposed estrogen therapy for menopausal symptoms. Adjustment 1.00) and the case-​control studies (RR = 0.71; 95% CI: 0.43, 1.16)
for menopausal hormone therapy did not change the association (Pheterogeneity  =  0.11). Statistical modeling suggested a linear dose–​
between physical activity and endometrial cancer. There was no evi- response relationship. There was no evidence of heterogeneity by
dence of effect modification by parity, use of oral contraceptives, gender or BMI, nor was there obvious confounding by BMI or smok-
menopausal status, or the time period of physical activity (i.e., adoles- ing. Associations did not differ for recreational versus occupational
cence, midlife, or older age). activity.
Increased body weight is strongly associated with the risk of endo- The 2016 pooled analysis found an inverse relationship between
metrial cancer. Most of the studies (24 of 33)  in the meta-​analysis of bladder cancer risk and leisure-​time physical activity (RR = 0.87; 95%
physical activity controlled for BMI as a potential confounder, but nine CI: 0.82, 0.92), with a point estimate similar to that observed in the
studies did not, including some that considered BMI a potential mediator meta-​analysis (Keimling et al., 2014). However, when stratifying on
that should not be adjusted for. In the meta-​analysis, there was no differ- smoking, the association was stronger in former than in never smokers,
ence in the summary estimate between studies that controlled for BMI raising the possibility of residual confounding. Thus, despite the large
(RR = 0.79; 95% CI: 0.73, 0.85) and those that did not (RR 0.83; 95% number of cases in the published studies, the level of evidence was
CI: 0.72, 0.97). considered limited (suggestive).
In the 2016 pooled analysis, the relative risks and 95% confidence
intervals for a high versus low level of leisure-​time activity were 0.79
(95% CI:  0.68, 0.92) for endometrial cancer, similar to the meta-​ Liver Cancer and Gallbladder Cancer
analysis estimate. However, in contrast to the meta-​analysis, the pooled
study found that adjustment for BMI essentially eliminated the inverse The association between physical activity and risk of liver cancer and
association between physical activity and endometrial cancer (relative gallbladder cancer has been studied only recently. No meta-​analyses
risk of 0.98). When compared with previous studies, the pooled anal- have been conducted. The largest published study is based on the
ysis used a more thorough approach in adjusting for BMI, including National Institutes of Health (NIH)-​AARP Diet and Health Study, a
each of the subcategories of obesity (30.0–​34.9, 35.0–​39.9, 40+ kg/​m2) prospective cohort of more than 500,000 men and women residing in
separately. Finer adjustment may explain the greater attenuation in the North America (Behrens et al., 2013). A total of 628 cases of liver can-
2016 pooled analysis. Replication is needed to resolve this difference cer and 123 cases of gallbladder cancer were identified in the cohort
in results. over 10 years of follow-​up. Physical activity, defined by the number of
In summary, higher levels of physical activity are associated with days per week that subjects performed vigorous physical activity con-
an approximately 20% reduction in the risk of endometrial cancer. It tinuously for at least 20 minutes, was inversely associated with liver
is presently unclear, however, whether this association is confounded cancer (RR  =  0.64; 95% CI:  0.49, 0.84). The association with gall-
and/​or mediated by BMI. bladder cancer was similar but not statistically significant (RR = 0.63;
95% CI: 0.33, 1.21). The dose–​response relationship between physical
activity and liver cancer was approximately linear, with no apparent
effect modification by age, gender, BMI, diabetes, alcohol intake, or
Bladder Cancer coffee consumption.
A meta-​analysis by Keimling et  al. (2014) identified 15 studies The 2016 pooled analysis, which included the NIH-​AARP cohort,
(11 cohort, 4 case control) from North America, Europe, and Asia also found an inverse association between leisure-​time physical activ-
that examined the association between physical activity and blad- ity and risk of liver cancer (RR = 0.73; 95% CI: 0.55, 0.98). The num-
der cancer risk. The analysis, based on 27,784 cases, found an ber of cases in the pooled study (N  =  1384) more than doubled the
inverse association between physical activity and bladder cancer risk number in the AARP study (N = 628) (Behrens et al., 2013). The over-
(RR = 0.85; 95% CI: 0.74, 0.98). The findings were not significantly all evidence for causality was considered to be “limited–​suggestive.”
386

386 PART III:  THE CAUSES OF CANCER

Gastroesophageal Cancers combined. The RR estimates comparing high versus low physical activ-
ity were 0.86 (95% CI: 0.68, 1.09) for multiple myeloma, 0.97 (95%
Behrens et al. (2014) identified 24 studies (9 cohorts, 15 case control) CI: 0.84, 1.13) for leukemia, and 0.91 (95% CI: 0.82, 1.00) for non-​
from North America, Europe, Asia, and the Middle East that examined Hodgkin lymphoma. Results were generally similar between cohort
associations between physical activity and subtypes of gastroesopha- and case-​control studies, and for recreational and occupational activity.
geal cancer. The analyses included 965 cases of esophageal adenocar- In the 2016 pooled analysis, leisure-​ time physical activity was
cinoma, 844 cases of gastric cardia cancer, 1444 cases of esophageal inversely associated with risk of myeloma (RR = 0.83; 95% CI: 0.72,
squamous cell carcinoma, and 1571 cases of non-​cardia stomach can- 0.95), and myeloid leukemia (RR  =  0.80; 95% CI:  0.70, 0.92), and
cers. Physical activity was inversely associated with risk of esophageal there was a borderline significant association for non-​Hodgkin lym-
adenocarcinoma (RR = 0.79; 95% CI: 0.66, 0.94) and gastric cardia phoma (RR = 0.91; 95% CI: 0.83, 1.00). No association was observed
cancer (RR = 0.83; 95% CI: 0.69, 0.99), but not esophageal squamous with lymphocytic leukemia. The point estimates for these hematologic
cell carcinoma (RR = 0.94; 95% CI: 0.41, 2.16). In an analysis that cancers were similar but statistically more precise than those in the
combined all subtypes of gastroesophageal cancer, the point estimate meta-​analysis by Jochem et al. (2014).
for physical activity was lower in the case control studies (RR = 0.77; Based on these analyses, the evidence for a causal relationship
95% CI: 0.67, 0.89) than in the prospective studies (RR = 0.89; 95% with myeloid leukemia, myeloma, and non-Hodgkin lymphoma was
CI: 0.78, 1.01). The dose–​response relationship was non-​linear, with considered to be “limited–​suggestive.” Although the evidence for
diminishing benefits at high levels of physical activity. The inverse non-​Hodgkin lymphoma was based on more than 10,000 cases in
association was somewhat stronger in women than in men, and more the prospective studies, the association reached only borderline sig-
strongly associated with early-​life physical activity than that occurring nificance (p = 0.05) in both the meta-​analysis and the 2016 pooled
later in life. There was no apparent confounding by BMI, smoking, or analysis. For lymphocytic leukemia, the null findings indicate that a
alcohol use. substantial effect is unlikely.
In the 2016 pooled analysis, leisure-​ time physical activity was
inversely associated with risk of esophageal adenocarcinoma (RR = 0.58;
95% CI: 0.38, 0.89) and cancer of the gastric cardia (RR = 0.78; 95% Head and Neck Cancer
CI:  0.64, 0.95). These results were similar to, but somewhat stronger
than, those of the Behrens et al. (2014) meta-​analysis. The analysis was No meta-​ analyses have examined physical activity in relation to
based on 899 cases of esophageal adenocarcinoma and 790 cases of gas- head and neck cancer risk. In the NIH-​AARP Diet and Health Study
tric cardia cancer. The evidence for these two cancer endpoints was con- (Leitzmann et al., 2008), during up to 8 years of follow-​up, 1249 cases
sidered to be “limited–​suggestive.” Evidence for other gastroesophageal of head and neck cancer were diagnosed. In age-​and gender-​adjusted
endpoints was too sparse to draw conclusions. analyses, physical activity was inversely associated with head and neck
cancer risk (RR  =  0.62; 95% CI:  0.52, 0.74) comparing the top and
bottom quintiles. The association became statistically non-​significant
Hematologic Cancers (RR  =  0.89; 95% CI:  0.74, 1.06), however, after multivariate adjust-
ment for smoking, alcohol consumption, and other factors. The authors
A meta-​analysis by Jochem et al. (2014) included 23 studies (15 cohort, stated that the association in age-​and gender-​adjusted analyses likely
8 case control) from North America, Europe, and Asia that examined reflected some degree of residual confounding by smoking status in
the relationship between physical activity and risk of hematologic particular.
malignancies. The analysis was based on 1362 cases of myeloma, 1736 In the pooled analysis, the risk of head and neck cancers was
cases of leukemia (limited data available on subtypes), and 9447 cases inversely associated with leisure-​time physical activity (RR  =  0.85;
of non-​Hodgkin lymphoma. No statistically significant association 95% CI:  0.78, 0.93). This association was statistically significant
was observed between physical activity and all hematologic cancers even after adjusting for smoking status, and was similar for never

Category # of Studies RR (95% Cl)


Overall 27 0.76 (0.69, 0.85)

Cohort 21 0.79 (0.70, 0.89)


Case-control 6 0.64 (0.55, 0.74)

Men 17 0.82 (0.74, 0.90)


Women 10 0.83 (0.69, 0.99)

Never smokers 4 0.96 (0.78, 1.18)


Current smokers 2 0.66 (0.49, 0.89)

Adenocarcinoma 6 0.80 (0.72, 0.88)


Squamous 6 0.80 (0.71, 0.90)
Small cell 6 0.79 (0.66, 0.94)

0 1 2
Relative risk (95% Cl)
High vs. low physical activity

Figure 21–​5.  Lung cancer and its association with physical activity.


 387

Physical Activity, Sedentary Behaviors, and Risk of Cancer 387


(RR = 0.83; 95% CI: 0.68, 1.02), former (RR = 0.90; 95% CI: 0.79, which comprise most of the data on this issue, suggest that phys-
1.04), and current smokers (RR  =  0.85; 95% CI:  0.65, 1.10). These ically active participants have a 7% lower risk of pancreatic cancer
results do not support residual confounding by smoking. The evidence (RR  =  0.93; 95% CI:  0.88, 0.98) than inactive participants. Results
was designated “limited–​suggestive.” from case-​control studies show a larger reduction; risk was 22% lower
(RR = 0.78; 95% CI: 0.66, 0.94) among the most active versus least
active participants. A dose–​response analysis suggests that the asso-
Lung Cancer ciation departs somewhat from linearity, reaching a plateau at high
Brenner et  al. (2016) conducted a meta-​analysis of 27 studies (21 activity levels. The association had no evident heterogeneity by gen-
cohort, 6 case control) from North America, Europe, and Asia on the der, smoking, or BMI, and no difference in results for recreational as
association of recreational physical activity with risk of lung cancer. opposed to occupational activity. The strength of the association did
In total, the meta-​analyses included 20,169 cases. A significant 24% not appear to vary in relation to the timing of physical activity over
lower risk of being diagnosed with lung cancer was noted (RR = 0.76; the life span, though it was suggested that consistent physical activity
95% CI:  0.69, 0.85), comparing adults with higher versus lower over time may be more important than recent physical activity or phys-
levels of physical activity (see Figure 21–​5). A  subset of studies in ical activity during any given past period of life. Level of adjustment
the meta-​analysis stratified the analysis by smoking status. Physical for different confounders also had little effect. Overall, these results
inactivity was inversely associated with lung cancer in two studies indicate that the link between physical activity and lower risk of pan-
of current smokers (RR = 0.66; 95% CI: 0.49, 0.89) but not in four creatic cancer—​if a true association exists—​is comparatively weak.
studies that restricted the analysis to never smokers (RR = 0.96; 95% This represents a downgrading of the evidence since prior reviews
CI: 0.78, 1.18). (Schottenfeld and Fraumeni, 2006; World Cancer Research Fund and
In the 2016 pooled analysis, higher levels of leisure-​time physical American Institute for Cancer Research, 2007), largely owing to the
activity were associated with lower risk of lung cancer in the overall weak associations observed in more recent cohort studies.
population (RR  =  0.74, 95% CI:  0.71, 0.77), but not in an analysis In the 2016 pooled analysis, leisure-​time physical activity was not
restricted to never smokers (RR = 1.03; 95% CI: 0.89, 1.20). The null significantly associated with the risk of pancreatic cancer (RR = 0.95;
findings in never smokers underscore concern about residual con- 95% CI:  0.83, 1.08), although the point estimate was similar to
founding by smoking in the overall analysis. that from the prospective studies in the Behrens et  al. (2015) meta-​
In summary, higher levels of physical activity are consistently asso- analysis (RR = 0.93). These findings differ markedly from the results
ciated with 25% lower risk of developing lung cancer in prospective of case-​control studies (RR = 0.78). Consequently, the evidence level
studies. However, this association is susceptible to confounding by for an association between physical activity and pancreatic cancer is
smoking, which is associated with both physical inactivity and a large “inconsistent.”
(approximately 25-​fold) increase in lung cancer risk. The inverse asso-
ciation has not been observed in studies of never smokers. Unless it
Prostate Cancer
can be shown that physical activity decreases lung cancer risk in never
smokers, or changes the natural history of lung cancer in smokers, the A total of 43 studies (19 cohort, 24 case control) from North America
likelihood of confounding cannot be excluded. The overall level of and Europe have examined the association between physical activity
evidence for a causal association between physical activity and lung and prostate cancer (see Figure 21–​6), as summarized by Liu et  al.
cancer risk was designated “uncertain–​may be confounded.” (2011). The studies included a total of 88,294 cases. This meta-​analysis
found that a high level of total physical activity (leisure-​time physical
activity and occupational activity together) was inversely associated
Malignant Melanoma with prostate cancer risk (RR = 0.90; 95% CI: 0.84, 0.95). The asso-
No prior meta-​analyses or prospective cohort studies have examined ciation was weaker in prospective studies (RR = 0.94; 95% CI: 0.90,
the association between physical activity and risk of melanoma. Only 0.98) than in case-​control studies (RR  =  0.86; 95% CI:  0.75, 0.97),
one case-​control study with 386 cases had previously examined this however. Additionally, the associations were not consistent across
association (Shors et al., 2001); this study reported that people with domains of physical activity, with weaker associations for leisure-​time
a high versus low level of physical activity had a 30% lower risk of physical activity (RR  =  0.95; 95% CI:  0.89, 1.00) than for occupa-
melanoma. tional physical activity (RR = 0.81; 95% CI: 0.73, 0.91). The reasons
In contrast, the 2016 pooled analysis, based on 12,348 cases, found for the heterogeneity of associations across study designs and domains
that leisure-​time physical activity was associated with increased risk of of physical activity are not well understood. Some evidence suggests
melanoma (RR = 1.27; 95% CI: 1.16, 1.40). Not only was the pooled that men who engage in a high level of leisure-​time physical activity
analysis much larger than the single previous case-​control study, but are more frequently screened for prostate cancer, either by PSA or dig-
separate analyses of 8 of the 12 cohorts found at least a 20% higher ital rectal exam, than men who engage in little leisure-​time physical
risk among those who were physically active. activity (Moore et al., 2008). This may lead to higher detection rates
No biological mechanisms have been proposed to explain how among active men, and the resulting bias may obscure inverse associa-
physical activity might increase the risk of melanoma. An alternative tions that would have otherwise been present. The extent of detection
hypothesis is that physically active individuals spend more time out- bias could plausibly be different for leisure-​time physical activity than
doors and have greater exposure to the sun. Physical activity is fre- for occupational physical activity, leading to heterogeneity by type of
quently done outdoors in light clothing, increasing the risk of sunburn activity.
(Holman et al., 2014). The overall level of evidence for a causal rela- Fewer than half of the studies reported whether there was a sig-
tionship between physical activity and melanoma risk is “uncertain–​ nificant trend in prostate cancer risk with increasing physical activ-
may be confounded.” ity levels. Taken together with the disparate physical activity types
and measurements, it was not possible to determine whether a dose–​
response association exists.
Associations were stronger in studies where the mean age of men
Pancreatic Cancer was less than 65 years than in studies where the mean age was higher,
As reviewed by Behrens et al. (2015), a total of 30 studies (22 cohort, perhaps reflecting the greater range in activity levels among younger
8 case control) from North America, Europe and Asia have examined men. Whether this difference by age subgroup was statistically sig-
the association between physical activity and risk of incident pancre- nificant was not formally tested in the meta-​analysis, leaving open the
atic cancer. Taken together, these studies included 10,501 cases. The possibility of a chance finding. Associations were also much stronger
results differ markedly between prospective studies and case-​control in studies with longer follow-​up time (i.e., greater than 10 years) than
studies and so are presented separately. The prospective studies, in studies with less follow-​up time, perhaps reflecting that a relatively
38

388 PART III:  THE CAUSES OF CANCER


Category # of Studies RR (95% Cl)

Overall 43 0.90 (0.84, 0.95)

Cohort 19 0.94 (0.90, 0.98)

Case-control 24 0.86 (0.75, 0.97)

Localized cases 14 0.96 (0.87, 1.05)

Advanced cases 14 0.94 (0.80, 1.10)

Occupational 27 0.81 (0.73, 0.91)

LTPA 34 0.95 (0.89, 1.00)

0 1 2
Relative risk (95% Cl)
High vs. low physical activity

Figure 21–​6.  Prostate cancer and its association with physical activity.

long period of physical activity may be required to decrease prostate Brain Cancer


cancer risk.
In analyses of prostate cancer subtypes, physical activity was asso- As reviewed by Niedermaier et  al. (2015), a total of six studies (4
ciated with approximately the same decrease in risk for both localized cohort, 2 case control) from North America, Europe, and Australia
and advanced subtypes. The studies in the analysis included a wide (have examined physical activity in relation to risk of glioma (a het-
range of potential confounders. These confounders appeared to have erogeneous cancer that comprises approximately 80% of all brain can-
little effect on the association, as results did not vary between models cers). There were a total of 3057 glioma cases. The authors found that
that were more fully adjusted and those that were minimally adjusted. high versus low physical activity levels were inversely associated with
In contrast to previous studies, the 2016 pooled analysis showed risk of glioma (RR = 0.86; 95% CI: 0.76, 0.97). Preliminary evidence
that higher levels of leisure-​time physical activity were associated with indicates that the magnitude of the association may vary according to
increased, rather than decreased, risk of prostate cancer (RR = 1.06; the timing of physical activity during the life span, with physical activ-
95% CI: 1.03, 1.08). Inconsistency in the results for prostate cancer is ity during adolescence associated with greater reductions in risk than
unsurprising given the concerns about detection bias and the hetero- activity later in life (Moore et al., 2009).
geneity of associations across study designs and domains of physical The 2016 pooled analysis found no association between leisure-​
activity noted above in the Liu et al. (2011) meta-​analysis. time physical activity and brain cancer risk (RR = 1.06; 95% CI: 0.93,
In summary, while physical activity was associated with a mod- 1.20). This contradicts the prior finding of an inverse association for
estly lower risk of prostate cancer, especially in case-​control studies, gliomas (Niedermaier et  al., 2015), leading us to conclude that the
this was not confirmed by the pooled analysis of prospective studies. overall evidence for an association is “inconsistent.”
Consequently, the evidence that physical activity protects against pros-
tate cancer risk is considered “inconsistent” rather than “probable” or
“convincing.”
Thyroid Cancer
As reviewed by Schmid et al. (2013), a total of eight studies (5 cohort,
3 case control) from North America, Europe, and Asia have examined
Rectal Cancer the association between physical activity and risk of thyroid cancer. In
A meta-​ analysis by Robsahm et  al. (2013) compiled findings for total, their meta-​analysis included 2250 cases of thyroid cancer. The
11 cohort studies (case-​control studies were excluded) from North authors found that high versus low levels of physical activity had no
America, Europe, Asia, and Australia that examined physical activity association with thyroid cancer risk (RR = 1.06; 95% CI: 0.79, 1.42).
in relation to rectal cancer risk. There were 8698 rectal cancer cases In prospective studies, those with high versus low levels of physical
overall. In the meta-​analysis, the authors found that the relative risk activity had increased risk of thyroid cancer (RR = 1.28; 95% CI: 1.01,
of rectal cancer comparing most active with least active participants 1.63), whereas in case-​control studies, they had a statistically non-​
in cohort studies was 0.98 (95% CI = 0.88, 1.08). No association was significant decrease in risk (RR = 0.70; 95% CI: 0.48, 1.03). In the 2016
observed between physical activity and risk of developing rectal cancer. pooled analysis, no association was observed between thyroid cancer
In the 2016 pooled analysis, leisure-​ time physical activity was and leisure-​time physical activity (RR = 0.92; 95% CI: 0.81, 1.06). The
inversely associated with risk of rectal cancer (RR  =  0.87; 95% evidence for this association is considered to be inconsistent.
CI:  0.80, 0.95). This finding contradicts the null association previ-
ously reported by Robsahm et al. (2013), leaving considerable uncer-
tainty about the true relationship. We consider the current evidence
Ovarian Cancer
for an association between physical activity and rectal cancer to be Zhong et al. (2014) identified a total of 19 studies (9 cohort, 10 case
“inconsistent.” control) from North America, Europe, Asia, and Australia on the
 389

Physical Activity, Sedentary Behaviors, and Risk of Cancer 389


association between non-​occupational (mostly recreational) physical Gierach et  al., 2009; Patel et  al., 2015). Collectively, these findings
activity and ovarian cancer risk. There were 9459 cases in the analysis. suggest that the association between sitting time and endometrial can-
Overall, there was a borderline inverse association between physical cer incidence may be confounded or mediated by BMI.
activity and ovarian cancer risk (RR = 0.92; 95% CI: 0.84, 1.00). In pro- Seven other studies have examined leisure-​time sitting in relation to
spective studies, no association was observed between physical activity cancer mortality (Dunstan et al., 2010; Katzmarzyk et al., 2009; Kim
and ovarian cancer risk (RR = 1.03; 95% CI: 0.87, 1.20), in contrast to et al., 2013; Matthews et al., 2012a, 2014; Patel et al., 2010; Wijndaele
the findings of case-​control studies (RR = 0.86; 95% CI: 0.80, 0.93). et  al., 2010). Of these, three reported statistically significant positive
In the 2016 pooled analysis, leisure-​time physical activity was not associations between sitting time and cancer mortality in women (Kim
associated with ovarian cancer risk (RR = 1.01; 95% CI: 0.91, 1.13). et al., 2013; Matthews et al., 2012a; Patel et al., 2010), whereas only
Given that results from cohort studies are afforded greater weight than two reported positive associations in men (Kim et al., 2013; Matthews
case-​control studies due to their lower potential for recall and selec- et al., 2012a).
tion biases, the balance of evidence indicates that physical activity is Evidence linking sitting time to other individual cancer sites is lim-
unlikely to have a substantial effect on ovarian cancer risk. ited, although some associations have been reported. Two prospective
studies reported modestly higher risk of invasive breast cancer asso-
ciated with total sitting (George et  al., 2010)  or leisure-​time sitting
All Other Cancers (Patel et al., 2015). One prospective cohort (Patel et al., 2015) and one
Other cancers that have been examined with regard to an association case-​control study (Zhang et al., 2004) reported a higher risk between
with physical activity include esophageal squamous cell carcinoma leisure-​time sitting and ovarian cancer, whereas two studies of occupa-
(Behrens et al., 2014), gallbladder cancer (Behrens et al., 2013), can- tional sitting (Dosemeci et al., 1993; Lee et al., 2013) and one of total
cers of the small intestine (Cross et al., 2013), and soft tissue (Moore sitting time (Xiao et  al., 2013)  were null. A  statistically significant
et  al., 2016). Data on these cancers are too sparse to draw conclu- association has been reported with multiple myeloma and borderline
sions about whether physical activity is associated with decreases or associations with head and neck and gallbladder cancer in men and
increases in risk. women; associations with esophageal cancer in women and pancreatic
cancer in men have also been reported (Patel et al., 2015). In contrast,
two prospective cohort studies have reported an inverse association
with total prostate cancer, although there was no dose–​response rela-
SEDENTARY BEHAVIORS AND RISK tionship, and the results were attenuated when restricted to advanced
OF DEVELOPING CANCER disease (Lynch et  al., 2014; Patel et  al., 2015). In general, physical
activities of a moderate to vigorous intensity, BMI, and age have not
Sedentary behavior (i.e., sitting) has features that differ from general been shown to modify associations of sitting time with cancer risk.
physical inactivity, and has been independently associated with vari- In summary, there is some, but not sufficient evidence to support
ous risk factors for chronic disease such as weight gain, high choles- a positive association between sitting time and some types of cancer,
terol, and high fasting insulin levels, as well as other biomarkers (Ford including colon, endometrium, breast, and ovary. The exact amount
et al., 2005; Fung et al., 2000; Healy et al., 2008; Jakes et al., 2003). or domain of sitting time that may be associated with increased risk
Sitting time has also been associated with premature mortality, cardio- remains unclear; whether associations are modified by exercise or
vascular disease, type 2 diabetes mellitus, and obesity (Biswas et al., body weight requires further investigation. The role of body weight on
2014). Few studies have examined associations between sitting time the causal pathway for some cancers, such as endometrial cancer, also
and cancer with mixed results. warrants further exploration.
A recent meta-​analysis, including 21 prospective cohorts and 22
case-​control studies, examined sitting time in relation to site-​specific
cancer risk (Schmid and Leitzmann, 2014). Evidence was limited for PHYSICAL ACTIVITY, SEDENTARY BEHAVIORS,
most individual cancer sites, however. Only a few studies examined AND CANCER SURVIVORSHIP
any given site, often in relation to different domains of sitting time
(e.g., occupational, television, leisure-​time, or total). Approximately Currently, there are approximately 14.5  million individuals in the
half of those studies examined occupational sitting time. Overall, the United States living after a diagnosis of cancer. While cancer treat-
meta-​analysis found that sitting time was positively associated with ments have improved, traditional therapies have significant toxicity,
colon and endometrial cancer risk. and targeted therapies are expensive.
Following this meta-​ analysis, a recent study comprehensively In the late 1980s researchers began to investigate whether exer-
examined leisure time spent sitting in relation to total and site-​specific cise training during active treatment could mitigate the side effects
cancer incidence and found that women who reported sitting for more of treatment and help to preserve physical function (MacVicar et al.,
than 6 hours/​day versus fewer than 3 hours/​day during their leisure 1989). This research has accelerated in the last decade. Courneya and
time had a 10% higher risk of all cancers combined (Patel et al., 2015). Friedenreich (2007) have elegantly described where and how physical
The overall association in women was driven by site-​specific asso- activity fits within the cancer control framework (Figure 21–​7). Here
ciations with invasive breast cancer (RR = 1.10; 95% CI: 1.00, 1.21), we briefly describe the evidence regarding the benefits of physical
ovarian cancer (RR = 1.43; 95% CI: 1.10, 1.87), and multiple myeloma activity following a diagnosis of cancer. Points of intervention include
(RR = 1.65; 95% CI: 1.07, 2.54). Leisure-​time sitting was not associ- pre-​treatment, treatment, and survivorship (Figure 21–​7).
ated with cancer risk in men overall, although among obese men there Exercise prior to surgery, chemotherapy, or radiation is prescribed
was an 11% higher risk associated with high levels of sitting time. as “prehab” to enhance physical fitness and coping prior to initiating
Most previous studies reporting a positive association with colon cancer treatment. Cancer rehabilitation and health promotion programs
cancer specifically examined television time (Howard et  al., 2008; also use exercise during and after treatment to reduce fatigue, enhance
Steindorf et  al., 2000)  or occupational sitting (Boyle et  al., 2010; function, and improve the quality of life (Schmitz et al., 2010). There
George et al., 2010; Gerhardsson et al., 1986; Weiderpass et al., 2003), is ample evidence that exercise can be safe during this period. The
and only one study (Howard et al., 2008) reported an association with American College of Sports Medicine has developed exercise guide-
total sitting time. Associations appear to be strongest with television lines to enhance the safety and effectiveness of exercise (Schmitz
watching, but it is not clear whether this may reflect the greater preci- et al., 2010) during and after cancer treatment. The American Cancer
sion of measuring television time versus overall sitting, or effect modi- Society has published recommendations for physical activity (Rock
fication by unhealthy behaviors, such as excess snacking, that may et al., 2012), and practical information for cancer survivors (Demark-​
correlate with television watching. While some previous studies have Wahnefried et al., 2015).
reported positive associations with endometrial cancer, these asso- In the last decade, researchers have begun to investigate whether
ciations are attenuated by adjustment for BMI (Friberg et  al., 2006; physical activity before or after diagnosis can improve survival and
390

390 PART III:  THE CAUSES OF CANCER


DIAGNOSIS

CANCER CONTROL CATEGORIES

Prevention Detection Treatment Treatment Recovery/ Disease Prevention/ Palliation Survival


Preparation/ Effectiveness/ Rehabilitation Health Promotion
Coping Coping

Prescreening Screening Pretreatment Treatment Survivorship End of Life

PREDIAGNOSIS POSTDIAGNOSIS

CANCER-RELATED TIME PERIODS

Figure 21–​7.  Physical activity and cancer control framework.

reduce cancer recurrence. Recently, Schmid and Leitzmann (2014) simultaneously. We highlight two research areas of particular importance
reviewed 16 survivorship studies of breast and colorectal cancer for improving our understanding of physical activity and cancer in the
encompassing 49,095 participants and 8129 total deaths. They exam- future.
ined physical activity performed before and after diagnosis in relation
to cause-​specific mortality rates. Physical activity before diagnosis
was associated with significantly lower death rates from breast cancer Ascertaining the Type, Intensity, and Amount
(HR = 0.77; 95% CI: 0.66, 0.90), and colorectal cancer (HR = 0.75; of Physical Activity Needed to Reduce Cancer Risk
95% CI: 0.62, 0.91). For post-​diagnosis activity, meeting currently rec- and Establishing the Role of Sedentary Behaviors
ommended amounts of moderate to vigorous physical activity during in Cancer Incidence
the post-​diagnosis period was associated with 24% lower mortality for
breast cancer survivors, and 28% lower risk for those previously diag- Studies of physical activity and cancer risk have typically quanti-
nosed with colorectal cancer. For cancer-​specific death, post-​diagnosis fied the relative risk for high versus low levels of physical activity.
physical activity was associated with a 28% lower risk for breast can- However, to develop clear recommendations with respect to cancer
cer mortality (HR = 0.72; 95% CI: 0.60, 0.85), as well as a 39% lower risk, it is also important to define the type, intensity, and amount of
risk for colorectal cancer mortality (HR = 0.61; 95% CI: 0.40, 0.92). physical activity required for each level of benefit. For example, is
There is much less evidence for prostate cancer, although in some there a particular kind of physical activity (e.g. aerobic vs. strength-​
studies physical activity is associated with a lower risk of dying from oriented) that is especially beneficial with respect to reducing cancer
prostate cancer, and recent studies have suggested that physical activ- risk? Are the benefits comparable for light versus moderate versus vig-
ity post-​diagnosis can improve survival among men diagnosed with orous intensity activity?
prostate cancer (Bonn et al., 2014); still, much more work is needed. Especially important are efforts to define the dose–​response relation-
This emerging body of evidence suggests significant mortality bene- ships between sedentary behavior, physical activity, and cancer risk. The
fits from physical activity for cancer survivors; however, some caveats relationship of physical activity with health and longevity is typically
must be considered. Many of the observational studies reporting these characterized as curvilinear, with the greatest benefits resulting from the
results were not originally designed to address this question, and there transition from inactivity to the recommended minimum activity levels
is often limited information about surgical procedures and outcomes, (Arem et al., 2015; Moore et al., 2012). As activity levels increase, the
or detailed treatment information throughout the course of chemother- benefits are thought to diminish, ultimately reaching a plateau at activity
apy and radiation treatments, or information about compliance with levels two to three times that of the US recommended minimum levels.
long-​term hormonal treatment (e.g., aromatase inhibitors), all of which For cancer incidence, however, the dose–​response association may dif-
can have an impact on survival and may influence physical activity lev- fer; preliminary evidence suggests that benefits may accrue in a more
els of study participants. Thus, the potential for residual confounding linear relationship with activity levels (Moore et al., 2016). If replicated,
and/​or reverse causation calls for caution in interpreting these results. this would have important implications for the framing of physical activ-
In summary, the number of individuals who will live for many ity recommendations in cancer prevention and control efforts.
years after their cancer diagnosis has increased dramatically in recent Most of the existing studies of sedentary behaviors and cancer risk
decades, and physical activity has emerged as an important poten- have focused on relatively few types of cancer. Future studies should
tial component of our cancer control framework during the active examine a broader range of cancer sites. In addition, not all studies
treatment period and in the post-​treatment rehabilitation and health-​ explicitly account for the potential for sedentary behaviors to displace
promotion phases of cancer survivorship. physical activity, especially light-​intensity physical activity. Future
studies should evaluate the separate effects of sedentary behavior and
physical inactivity, while accounting for potential displacement of
OPPORTUNITIES FOR PREVENTION active behaviors. The effects of prolonged sitting, independent of total
sitting time, deserve further study.
Opportunities to prevent cancer by increasing physical activity lev-
els and decreasing sedentary behaviors are discussed in detail in
Chapter 62.2 of this volume. Application of Novel Technologies to Improve
Measurement of Physical Activity
FUTURE RESEARCH To date, most of the research on sedentary behavior, physical activity,
and cancer has used self-​reported rather than objective measurement.
The relationship of physical activity with cancer risk is an active area Recall of activity levels over months or years in the past is difficult, in
of research, with many aspects of this relationship being investigated the absence of contextual cues. Misclassification of exposure due to
 391

Physical Activity, Sedentary Behaviors, and Risk of Cancer 391


errors in recall can attenuate relative risk estimates in epidemiologic Arem H, Moore SC, Patel A, et  al. 2015. Leisure time physical activity and
studies. Newer technological approaches aim to minimize such error mortality:  a detailed pooled analysis of the dose-​response relationship.
by using Internet-​based 24-​hour recalls, or accelerometry to measure JAMA Intern Med, 175(6), 959–​967. PMCID: PMC4451435.
movement objectively. Behavioral Risk Factor Surveillance System. Available from: http://​www.cdc.
gov/​brfss/​. Accessed June 15, 2015.
Over the last 5  years, several automated Internet-​based 24-​hour Behrens G, Jochem C, Keimling M, et al. 2014. The association between physi-
recalls of physical activity behavior (Gomersall et al., 2011; Matthews cal activity and gastroesophageal cancer:  systematic review and meta-​
et  al., 2012b) have been validated with respect to measuring sitting analysis. Eur J Epidemiol, 29(3), 151–​170. PMID: 24705782.
time, including different activity intensities and the location and pur- Behrens G, Jochem C, Schmid D, et al. 2015. Physical activity and risk of pan-
pose of the activity (Gomersall et al., 2011; Hunt et al., 2013; Keadle creatic cancer: a systematic review and meta-​analysis. Eur J Epidemiol,
et al., 2014; Mace et al., 2014; Matthews et al., 2013). These recalls 30(4), 279–​298. PMID: 25773752.
have been shown to have less reporting error than many questionnaires Behrens G, and Leitzmann MF. 2013. The association between physical activ-
(Gomersall et al., 2011; Matthews et al., 2013), and can be adminis- ity and renal cancer: systematic review and meta-​analysis. Br J Cancer,
tered multiple times per year in order to assess habitual or usual sitting 108(4), 798–​811. PMCID: PMC3590672.
Behrens G, Matthews CE, Moore SC, et  al. 2013. The association between
and physical activity levels (Matthews et al., 2012b). To date, 24-​hour frequency of vigorous physical activity and hepatobiliary cancers in
physical activity recalls have not been employed in large-​scale popu- the NIH-​AARP Diet and Health Study. Eur J Epidemiol, 28(1), 55–​66.
lation studies due to logistical concerns. However, as epidemiologic PMCID: PMC3670752.
studies move toward collecting participant data using mobile technolo- Biswas A, Oh P, Faulkner G, et  al. 2014. Sedentary time and its association
gies, it will become increasingly feasible to integrate 24-​hour recalls with risk for disease incidence, mortality, and hospitalization in adults: a
into ongoing protocols for population-​based studies. systematic review and meta-​analysis. Ann Intern Med, 162(2), 123–​132.
Accelerometers are wearable motion sensors that measure accelera- PMID: 25599350.
tions in up to three different directions multiple times per second. These Bonn SE, Sjölander A, Lagerros YT, et  al. 2014. Physical activity and sur-
data are then integrated into estimates of overall energy expenditure vival among men diagnosed with prostate cancer. Cancer Epidemiol
Biomarkers Prev, 24(1), 57–​64. PMID: 25527697.
per day or week, time in distinct activity intensity categories (e.g., sed- Boyle T, Fritschi L, Heyworth J, and Bull F. 2010. Long-​ term sedentary
entary, moderate), and/​or time seated versus standing. Accelerometers work and the risk of subsite-​specific colorectal cancer. Am J Epidemiol,
have several advantages over self-​report, including that they minimize 173(10), 1183–​1191. PMID: 21421743.
recall and social desirability biases. Accelerometers perform well for Boyle T, Keegel T, Bull F, Heyworth J, and Fritschi L. 2012. Physical activity
estimating time in sedentary and light-​intensity activities, which is par- and risks of proximal and distal colon cancers: a systematic review and
ticularly difficult to measure from self-​reported data. They also place meta-​analysis. J Natl Cancer Inst, 104(20), 1548–​1561. PMID: 22914790.
no cognitive demands on participants, thus making the technology Brenner DR, Yannitsos DH, Farris MS, Johansson M, and Friedenreich CM.
attractive for studies in children. However, accelerometers are expen- 2016. Leisure-​time physical activity and lung cancer risk:  a systematic
sive, their application to large-​scale studies can be logistically com- review and meta-​analysis. Lung Cancer, 95, 17–​27. PMID: 27040847.
Brown SB, and Hankinson SE. 2015. Endogenous estrogens and the risk of
plicated, and they yield complex data that are challenging to process breast, endometrial, and ovarian cancers. Steroids, 99(Pt A), 8–​ 10.
(Lee and Shiroma, 2014). Despite these challenges, at least two studies PMID: 25555473.
with 10,000 or more participants with prospective follow-​up for dis- Calle EE, and Kaaks R. 2004. Overweight, obesity and cancer: epidemiologi-
ease outcomes are currently underway (Howard et al., 2015; Lee and cal evidence and proposed mechanisms. Nat Rev Cancer, 4(8), 579–​591.
Shiroma, 2014). In addition to research-​based accelerometers, com- PMID: 15286738.
mercial monitors to assess activity, such as the Fitbit and Apple Watch, Campbell KL, Campbell PT, Ulrich CM, et al. 2008. No reduction in C-​reactive
are now widely available in consumer markets and are used by millions protein following a 12-​month randomized controlled trial of exercise in
of people. Data from such commercial monitors could potentially be men and women. Cancer Epidemiol Biomarkers Prev, 17(7), 1714–​1718.
used to conduct large-​scale epidemiologic studies at a relatively low PMCID: PMC2493470.
Campbell PT, Campbell KL, Wener MH, et al. 2009. A yearlong exercise inter-
cost, provided that their reliability and validity can be confirmed. vention decreases CRP among obese postmenopausal women. Med Sci
Sports Exerc, 41(8), 1533–​1539. PMCID: PMC3850754.
Carlson SA, Densmore D, Fulton JE, Yore MM, and Kohl HW, 3rd. 2009.
CONCLUSION Differences in physical activity prevalence and trends from 3 U.S.  sur-
veillance systems: NHIS, NHANES, and BRFSS. J Phys Act Health, 6
In the last 5 years, the publication of numerous prospective studies and Suppl 1, S18–​S27. PMID: 19998846.
meta-​analyses, as well the large consortium-​based analysis, have shed Caspersen C, Powell K, and Christenson G. 1985. Physical activity, exer-
considerable light on the relationship between physical activity and cise, and physical fitness:  definitions and distinctions for health related
cancer risk. In addition to the associations for colon, postmenopausal research. Public Health Rep, 100, 12–​131. PMCID: PMC1424733.
breast, and endometrial cancers previously identified, physical activity Centers for Disease Control and Prevention. 2013. Adult participation in aero-
has now been associated with lower risk of multiple types of cancer, bic and muscle-​strengthening physical activities:  United States, 2011.
MMWR Morb Mortal Wkly Rep, 62(17), 326–​330. PMID: 23636025.
including, for example, gastroesophageal and hematologic malignan-
Chlebowski RT, Hendrix SL, Langer RD, et  al. 2003. Influence of estrogen
cies. The extensive breadth of findings highlights a key role for phys- plus progestin on breast cancer and mammography in healthy postmeno-
ical activity in population-​level cancer prevention and control efforts. pausal women: the Women’s Health Initiative Randomized Trial. JAMA,
Although much has been learned in recent years about physical 289(24), 3243–​3253. PMID: 12824205.
activity and cancer risk, many questions remain. Newly identified Courneya KS, and Friedenreich CM. 2007. Physical activity and cancer con-
associations should be confirmed, and the type, intensity, and amount trol. Semin Oncol Nursing, 23(4), 242–​252. PMID: 18022052.
of physical activity needed to reduce cancer risk should be clearly Coussens LM, and Werb Z. 2002. Inflammation and cancer. Nature, 420(6917),
defined. Newer technological methods should be implemented in can- 860–​867. PMCID: PMC2803035.
cer etiology studies. Finally, whether sedentary behaviors indepen- Cross AJ, Hollenbeck AR, and Park Y. 2013. A  large prospective study of
risk factors for adenocarcinomas and malignant carcinoid tumors
dently contribute to cancer risk should be thoroughly investigated in
of the small intestine. Cancer Causes Control, 24(9), 1737–​ 1746.
large prospective studies and consortia-​based studies. PMCID: PMC3737280.
Demark-​Wahnefried W, Rogers LQ, Alfano CM, et al. 2015. Practical clinical
References interventions for diet, physical activity, and weight control in cancer sur-
Abbenhardt C, McTiernan A, Alfano CM, et al. 2013. Effects of individual and vivors. CA Cancer J Clin, 65(3), 167–​189. PMID: 25683894.
combined dietary weight loss and exercise interventions in postmeno- Dosemeci M, Hayes R, Vetter R, et  al. 1993. Occupational physical activity,
pausal women on adiponectin and leptin levels. J Intern Med, 274(2), socioeconomic status, and risks of 15 cancer sites in Turkey. Cancer
163–​175. PMCID: PMC3738194. Causes Control, 4(4), 313–​321. PMID: 8347780.
392

392 PART III:  THE CAUSES OF CANCER


Dunstan DW, Barr ELM, Healy GN, et  al. 2010. Television viewing time Ho GY, Wang T, Gunter MJ, et  al. 2012. Adipokines linking obesity with
and mortality:  The Australian Diabetes, Obesity, and Lifestyle Study colorectal cancer risk in postmenopausal women. Cancer Res, 72(12),
(AusDiab). Circulation, 121(3), 384–​391. PMID: 20065160. 3029–​3037. PMCID: PMC3790260.
Endogenous Hormones and Breast Cancer Collaborative Group. 2010. Insulin-​ Holman DM, Berkowitz Z, Guy GP, Jr., Hartman AM, and Perna FM. 2014.
like growth factor 1 (IGF1), IGF binding protein 3 (IGFBP3), and breast The association between demographic and behavioral characteristics and
cancer risk:  pooled individual data analysis of 17 prospective studies. sunburn among U.S. adults: National Health Interview Survey, 2010. Prev
Lancet Oncol, 11(6), 530–​542. PMID: 20472501 Med, 63, 6–​12. PMCID: PMC4535173.
Ennour-​Idrissi K, Maunsell E, and Diorio C. 2015. Effect of physical activ- Howard RA, Freedman DM, Park Y, et  al. 2008. Physical activity, seden-
ity on sex hormones in women:  a systematic review and meta-​analysis tary behavior, and the risk of colon and rectal cancer in the NIH-​AARP
of randomized controlled trials. Breast Cancer Res, 17(1), 139. Diet and Health Study. Cancer Causes Control, 19(9), 939–​953. PMID:
PMCID: PMC4635995. 18437512.
Ferrari P, Friedenreich C, and Matthews CE. 2007. The role of measurement Howard VJ, Rhodes JD, Mosher A, et al. 2015. Obtaining accelerometer data in
error in estimating levels of physical activity. Am J Epidemiol, 166(7), a national cohort of black and white adults. Med Sci Sports Exerc, 47(7),
832–​840. PMID: 17670910. 1531–​1537. PMCID: PMC4404169.
Ford ES, Kohl HW, 3rd, Mokdad AH, and Ajani UA. 2005. Sedentary behavior, Hunt T, Williams MT, and Olds TS. 2013. Reliability and validity of the
physical activity, and the metabolic syndrome among U.S. adults. Obes multimedia activity recall in children and adults (MARCA) in people
Res, 13(3), 608–​614. PMID: 15833947. with chronic obstructive pulmonary disease. PLoS One, 8(11), e81274.
Frank LL, Sorensen BE, Yasui Y et al. 2005. Effects of exercise on metabolic PMCID: PMC3842291.
risk variables in overweight postmenopausal women: a randomized clini- IARC Handbooks of Cancer Prevention. 2002. Volume 6: Weight Control and
cal trial. Obes Res, 13(3), 615–​625. PMID: 15833948. Physical Activity. Lyon, France: IARC Press.
Friberg E, Mantzoros CS, and Wolk A. 2006. Physical activity and endometrial Ish-​Shalom D, Christoffersen CT, Vorwerk P, et  al. 1997. Mitogenic proper-
cancer: a population-​based prospective cohort study. Cancer Epidemiol ties of insulin and insulin analogues mediated by the insulin receptor.
Biomarkers Prev, 15(11), 2136–​2140. Diabetologia, 40(Suppl 2), S25–​S31. PMID: 9248698.
Friedenreich CM. 2011. Physical activity and breast cancer: review of the epi- Jakes RW, Day NE, Khaw KT, et al. 2003. Television viewing and low partici-
demiologic evidence and biologic mechanisms. Recent Results Cancer pation in vigorous recreation are independently associated with obesity
Res, 188, 125–​139. PMID: 21253795. and markers of cardiovascular disease risk:  EPIC-​Norfolk population-​
Friedenreich CM, Neilson HK, and Lynch BM. 2010. State of the epidemiolog- based study. Eur J Clin Nutr, 57(9), 1089–​1096. PMID: 12947427.
ical evidence on physical activity and cancer prevention. Eur J Cancer, Jochem C, Leitzmann MF, Keimling M, Schmid D, and Behrens G. 2014.
46(14), 2593–​2604. PMID: 20843488. Physical activity in relation to risk of hematologic cancers: a systematic
Friedenreich CM, Neilson HK, Woolcott CG, et al. 2011. Changes in insulin review and meta-​analysis. Cancer Epidemiol Biomarkers Prev, 23(5),
resistance indicators, IGFs, and adipokines in a year-​long trial of aerobic 833–​846. PMID: 24633143.
exercise in postmenopausal women. Endocr Relat Cancer, 18(3), 357–​ Judd HL, Shamonki IM, Frumar AM, and Lagasse LD. 1982. Origin of serum
369. PMCID: PMC3111235. estradiol in postmenopausal women. Obstet Gynecol, 59(6), 680–​686.
Fung TT, Hu FB, Yu J, et  al. 2000. Leisure-​time physical activity, television PMID: 7078905.
watching, and plasma biomarkers of obesity and cardiovascular disease Kaaks R, and Lukanova A. 2001. Energy balance and cancer:  the role of
risk. Am J Epidemiol, 152(12), 1171–​1178. PMID: 11130623. insulin and insulin-​like growth factor-​I. Proc Nutr Soc, 60(1), 91–​106.
Garabrant DH, Peters JM, Mack TM, and Bernstein L. 1984. Job activ- PMID: 11310428.
ity and colon cancer risk. Am J Epidemiol, 119(6), 1005–​ 1014. Kahn BB, and Flier JS. 2000. Obesity and insulin resistance. J Clin Invest,
PMID: 6731427. 106(4), 473–​481. PMCID: PMC380258.
George S, Irwin M, Matthews C, et al. 2010. Beyond recreational physical activ- Katzmarzyk PT, Church TS, Craig CL, and Bouchard C. 2009. Sitting time and
ity: examining occupational and household activity, transportation activ- mortality from all causes, cardiovascular disease, and cancer. Med Sci
ity, and sedentary behavior in relation to postmenopausal breast cancer Sports Exerc, 41(5), 998–​1005. PMID: 19346988.
risk. Am J Public Health, 100(11), 2288–​2295. PMCID: PMC2951936. Keimling M, Behrens G, Schmid D, Jochem C, and Leitzmann MF. 2014. The asso-
Gerhardsson M, Norell S, Kiviranta H, Pedersen N, and Ahlbom A. 1986. ciation between physical activity and bladder cancer: systematic review and
Sedentary jobs and colon cancer. Am J Epidemiol, 123(5), 775–​780. meta-​analysis. Br J Cancer, 110(7), 1862–​1870. PMCID: PMC3974090.
PMID: 3962961 Kelley GA, and Kelley KS. 2006. Effects of aerobic exercise on C-​reactive pro-
Gierach GL, Chang S, Brinton LA, et  al. 2009. Physical activity, sedentary tein, body composition, and maximum oxygen consumption in adults: a
behavior, and endometrial cancer risk in the NIH-​AARP Diet and Health meta-​analysis of randomized controlled trials. Metabolism, 55(11), 1500–​
Study. Int J Cancer, 124(9), 2139–​2147. PMCID: PMC2845165. 1507. PMID: 17046553.
Godsland IF. 2010. Insulin resistance and hyperinsulinaemia in the develop- Key TJ, Appleby PN, Reeves GK, et al. 2003. Body mass index, serum sex hor-
ment and progression of cancer. Clinical Science, 118(5-​6), 315–​332. mones, and breast cancer risk in postmenopausal women. J Natl Cancer
PMCID: PMC2782313. Inst, 95(16), 1218–​1226. PMID: 12928347.
Gomersall SR, Olds TS, and Ridley K. 2011. Development and evaluation of an Key TJ, Appleby PN, Reeves GK, et al. 2015. Steroid hormone measurements
adult use-​of-​time instrument with an energy expenditure focus. J Sci Med from different types of assays in relation to body mass index and breast
Sport, 14(2), 143–​148. PMID: 20932797. cancer risk in postmenopausal women: Reanalysis of eighteen prospec-
Gunter MJ, Hoover DR, Yu H, et al. 2008. A prospective evaluation of insulin and tive studies. Steroids, 99(Pt A), 49–​55. PMCID: PMC4502556.
insulin-​like growth factor-​I as risk factors for endometrial cancer. Cancer Kim Y, Wilkens L, Park S-​Y, et  al. 2013. Association between various sed-
Epidemiol Biomarkers Prev, 17(4), 921–​929. PMCID: PMC3090086. entary behaviours and all-​ cause, cardiovascular disease, and cancer
Gunter MJ, Hoover DR, Yu H, et al. 2009. Insulin, insulin-​like growth factor-​I, mortality:  the Multiethnic Cohort. Int J Epidemiol, 42(4), 1040–​1056.
and risk of breast cancer in postmenopausal women. J Natl Cancer Inst, PMCID: PMC3781003.
101(1), 48–​60. PMCID: PMC2639294. Kozey Keadle S, Lyden K, Hickey A, et al. 2014. Validation of a previous day
Gunter MJ, Wang T, Cushman M, et  al. 2015. Circulating adipokines and recall for measuring the location and purpose of active and sedentary
inflammatory markers and postmenopausal breast cancer risk. J Natl behaviors compared to direct observation. Int J Behav Nutr Phys Act, 11,
Cancer Inst, 107(9). PMCID: PMC4651104. 12. PMCID: PMC3922158.
Hallal PC, Bauman AE, Heath GW, et al. 2012. Physical activity: more of the Kushi LH, Doyle C, McCullough M, et  al. 2012. American Cancer Society
same is not enough. Lancet, 380(9838), 190–​191. PMID: 22818932. Guidelines on nutrition and physical activity for cancer prevention: reduc-
Hankinson AL, Daviglus ML, Bouchard C, et  al. 2010. Maintaining a high ing the risk of cancer with healthy food choices and physical activity. CA
physical activity level over 20  years and weight gain. JAMA, 304(23), Cancer J Clin, 62(1), 30–​67. PMID: 22237782.
2603–​2610. PMCID: PMC3864556. Lee A, Su D, Pasalich M, Wong Y, and Binns C. 2013. Habitual physical activ-
Hawley JA, Hargreaves M, Joyner MJ, and Zierath JR. 2014. Integrative biol- ity reduces risk of ovarian cancer: a case-​control study in southern China.
ogy of exercise. Cell, 159(4), 738–​749. PMID: 25417152. Prev Med, 57, S31–​S33.
Healy GN, Clark BK, Winkler EA, et al. 2011. Measurement of adults’ seden- Lee IM, Djousse L, Sesso HD, Wang L, and Buring JE. 2010. Physical
tary time in population-​based studies. Am J Prev Med, 41(2), 216–​227. activity and weight gain prevention. JAMA, 303(12), 1173–​ 1179.
PMCID: PMC3179387. PMCID: PMC2846540.
Healy GN, Wijndaele K, Dunstan DW, et al. 2008. Objectively measured sed- Lee IM, and Oguma Y. 2006. Physical activity. In:  Schottenfeld D, and
entary time, physical activity, and metabolic risk. Diabetes Care, 31(2), Fraumeni JF, Jr. (Eds.), Cancer Epidemiology and Prevention (3rd ed.,
369–​371. PMID: 18000181. pp. 449–​467). New York: Oxford University Press.
 39

Physical Activity, Sedentary Behaviors, and Risk of Cancer 393


Lee IM, and Shiroma EJ. 2014. Using accelerometers to measure physical Neilson HK, Robson PJ, Friedenreich CM, and Csizmadi I. 2008. Estimating
activity in large-​scale epidemiological studies: issues and challenges. Br activity energy expenditure:  how valid are physical activity question-
J Sports Med, 48(3), 197–​201. PMCID: PMC3947179. naires? Am J Clin Nutr, 87(2), 279–​291. PMID: 18258615.
Leitzmann MF, Koebnick C, Freedman ND, et al. 2008. Physical activity and Niedermaier T, Behrens G, Schmid D, et  al. 2015. Body mass index, physi-
head and neck cancer risk. Cancer Causes Control, 19(10), 1391–​1399. cal activity, and risk of adult meningioma and glioma: a meta-​analysis.
PMCID: PMC3507337. Neurology, 85(15), 1342–​1350. PMID: 26377253.
Liu Y, Hu F, Li D, et al. 2011. Does physical activity reduce the risk of prostate Nimptsch K, Aleksandrova K, Boeing H, et al. 2015. Association of CRP genetic
cancer? A systematic review and meta-​analysis. Eur Urol, 60(5), 1029–​ variants with blood concentrations of C-​reactive protein and colorectal
1044. PMID: 21802197. cancer risk. Int J Cancer, 136(5), 1181–​1192. PMID: 25043606.
Lynch B, Friedenreich C, Kopciuk K, et  al. 2014. Sedentary behavior and Ollberding NJ, Kim Y, Shvetsov YB, et al. 2013. Prediagnostic leptin, adipo-
prostate cancer risk in the NIH-​AARP Diet and Health Study. Cancer nectin, C-​reactive protein, and the risk of postmenopausal breast cancer.
Epidemiol Biomarkers Prev, 23(5), 882–​889. PMCID: PMC4012010. Cancer Prev Res (Phila), 6(3), 188–​195. PMCID: PMC3595121.
Lynch BM, Dunstan DW, Healy GN, et al. 2010. Objectively measured physi- Patel AV, Bernstein L, Deka A, et al. 2010. Leisure time spent sitting in relation
cal activity and sedentary time of breast cancer survivors, and asso- to total mortality in a prospective cohort of US adults. Am J Epidemiol,
ciations with adiposity:  findings from NHANES (2003–​2006). Cancer 172(4), 419–​429. PMCID: PMC3590043.
Causes Control, 21(2), 283–​288. PMID: 19882359. Patel AV, Hildebrand JS, Campbell PT, et al. 2015. Leisure-​time spent sitting
Mace CJ, Maddison R, Olds T, and Kerse N. 2014. Validation of a computer- and site-​specific cancer incidence in a large US cohort. Cancer Epidemiol
ized use of time recall for activity measurement in advanced-​aged adults. Biomarkers Prev, 24(9), 1350–​1359. PMID: 26126627.
J Aging Phys Act, 22(2), 245–​254. PMID: 23752221. Pedersen BK, and Febbraio MA. 2012. Muscles, exercise and obesity:  skel-
MacVicar MG, Winningham ML, and Nickel JL. 1989. Effects of aerobic inter- etal muscle as a secretory organ. Nat Rev Endocrinol, 8(8), 457–​465.
val training on cancer patients’ functional capacity. Nursing Res, 38(6), PMID: 22473333.
348–​351. PMID: 2587289. Pereira MA, Fitzgerald SJ, Gregg EW, et  al. 1997. A  collection of physi-
Mantovani A, Allavena P, Sica A, Balkwill F. 2008. Cancer-​related inflamma- cal activity for health-​ related research. Med Sci Sports Exerc, 29(6
tion. Nature, 454(7203), 436–​444. PMID: 18650914. Supplement), S1–​S203.
Matthews C, Cohen S, Fowke J, et  al. 2014. Physical activity, sedentary Physical Activity Guidelines Advisory Committee. 2008. Physical Activity
behavior, and cause-​specific mortality in black and white adults in the Guidelines Advisory Committee Report (pp. A1–​ A10). Washington,
Southern Community Cohort Study. Am J Epidemiol, 180(4), 394–​405. DC: U.S. Department of Health and Human Services.
PMCID: PMC4128770. Pugeat M, Crave JC, Elmidani M, et al. 1991. Pathophysiology of sex hormone
Matthews CE. 2002. Techniques for physical activity assessment:  self-​report binding globulin (SHBG): relation to insulin. J Steroid Biochem Mol Biol,
instruments. In: Welk G, and Dale D (Eds.), Physical Activity Assessments 40(4–​6), 841–​849. PMID: 1958579.
for Health-​ Related Research (pp. 107–​ 123). Champaign, IL:  Human Robsahm TE, Aagnes B, Hjartåker A, Langseth H, Bray FI, Larsen IK. 2013.
Kinetics. Body mass index, physical activity, and colorectal cancer by anatomical
Matthews CE, Ainsworth BE, Hanby C, et al. 2005. Development and testing of subsites: a systematic review and meta-​analysis of cohort studies. Eur J
a short physical activity recall questionnaire. Med Sci Sports Exerc, 37(6), Cancer Prev, 22(6), 492–​505. PMID: 23591454.
986–​994. PMID: 15947724. Rock CL, Doyle C, Demark-​Wahnefried W, et al. 2012. Nutrition and physical
Matthews CE, Chen KY, Freedson PS, et  al. 2008. Amount of time spent in activity guidelines for cancer survivors. CA Cancer J Clin, 62(4), 243–​
sedentary behaviors in the United States, 2003–​2004. Am J Epidemiol, 274. PMID: 22539238.
167(7), 875–​881. PMCID: PMC3527832. Sallis JF. 1997. A collection of physical activity questionnaires for health-​
Matthews CE, George SM, Moore S, et  al. 2012a. Amount of time spent in related research: seven-​day physical activity recall. In: Kriska AM, and
sedentary behaviors and cause-​specific mortality in US adults. Am J Clin Casperson CJ (Eds.), A Collection of Physical Activity Questionnaires.
Nutr, 95(2), 437–​445. PMCID: PMC3260070. Med Sci Sports Exerc (Vol. 29, pp. S89–​S103).
Matthews CE, Keadle SK, Sampson J et al. 2013. Validation of a previous-​day Sallis JF, and Saelens BE. 2000. Assessment of physical activity by self-​report: lim-
recall measure of active and sedentary behaviors. Med Sci Sports Exerc, itations and future directions. Res Q Exercise Sport, 71(Suppl 2), S1–​S14.
45(8), 1629–​1638. PMCID: PMC3717193. Schmid D, Behrens G, Jochem C, Keimling M, and Leitzmann M. 2013.
Matthews CE, Moore SC, George SM, Sampson J, and Bowles HR. 2012b. Physical activity, diabetes, and risk of thyroid cancer: a systematic review
Improving self-​reports of active and sedentary behaviors in large epidemio- and meta-​analysis. Eur J Epidemiol, 28(12), 945–​958. PMID: 24243033.
logic studies. Exerc Sport Sci Rev, 40(3), 118–​126. PMCID: PMC3388604. Schmid D, Behrens G, Keimling M, et al. 2015. A systematic review and meta-​
McTiernan A. 2008. Mechanisms linking physical activity with cancer. Nat Rev analysis of physical activity and endometrial cancer risk. Eur J Epidemiol,
Cancer, 8(3), 205–​211. PMID: 18235448. 30(5), 397–​412. PMID: 25800123.
McTiernan A, Sorensen B, Yasui Y et al. 2005. No effect of exercise on insulin-​ Schmid D, and Leitzmann M. 2014. Television viewing and time spent seden-
like growth factor 1 and insulin-​like growth factor binding protein 3 in tary in relation to cancer risk: a meta-​analysis. J Natl Cancer Inst, 106(7),
postmenopausal women:  a 12-​month randomized clinical trial. Cancer dju098. PMID: 24935969.
Epidemiol Biomarkers Prev, 14(4), 1020–​1021. PMID: 15824183. Schmid D, and Leitzmann MF. 2014. Association between physical activity and
Moore SC, Lee IM, Weiderpass E et al. 2016. Association of leisure-​time physi- mortality among breast cancer and colorectal cancer survivors: a systematic
cal activity with risk of 26 types of cancer in 1.44 million adults. JAMA review and meta-​analysis. Ann Oncol, 25(7), 1293–​1311. PMID: 24644304.
Intern Med, 176(6), 816–​825. PMID: 27183032. Schmitz KH, Courneya KS, Matthews C, et  al. 2010. American College of
Moore SC, Patel AV, Matthews CE, et al. 2012. Leisure time physical activity Sports Medicine roundtable on exercise guidelines for cancer survivors.
of moderate to vigorous intensity and mortality:  a large pooled cohort Med Sci Sports Exerc, 42(7), 1409–​1426. PMID: 20559064.
analysis. PLoS Med, 9(11), e1001335. PMCID: PMC3491006. Schottenfeld D, and Fraumeni JF, Jr. (Eds.) 2006. Cancer Epidemiology and
Moore SC, Peters TM, Ahn J, et al. 2008. Physical activity in relation to total, Prevention (3rd ed.). New York: Oxford University Press.
advanced, and fatal prostate cancer. Cancer Epidemiol Biomarkers Prev, Shafie SM, and Hilf R. 1981. Insulin receptor levels and magnitude of insulin-​
17(9), 2458–​2466. PMCID: PMC3443883. induced responses in 7,12-​dimethylbenz(a)anthracene-​induced mammary
Moore SC, Rajaraman P, Dubrow R, et al. 2009. Height, body mass index, and tumors in rats. Cancer Res, 41(3), 826–​829. PMID: 6780189.
physical activity in relation to glioma risk. Cancer Res, 69(21), 8349–​ Shors AR, Solomon C, McTiernan A, and White E. 2001. Melanoma risk in
8355. PMCID: PMC2783605. relation to height, weight, and exercise (United States). Cancer Causes
Moradi T, Nyren O, Bergstrom R et al. 1998. Risk for endometrial cancer in Control, 12(7), 599–​606. PMID: 11552707.
relation to occupational physical activity:  a nationwide cohort study in Steindorf K, Tobiasz-​Adamczyk B, Popiela T, et  al. 2000. Combined risk
Sweden. Int J Cancer, 76(5), 665–​670. PMID: 9610723. assessment of physical activity and dietary habits on the development of
Mu N, Zhu Y, Wang Y, Zhang H, and Xue F. 2012. Insulin resistance: a signifi- colorectal cancer:  a hospital-​based case-​control study in Poland. Eur J
cant risk factor of endometrial cancer. Gynecol Oncol, 125(3), 751–​757. Cancer Prev, 9(5), 309–​316. PMID: 11075883.
PMID: 22449736. Stolzenberg-​Solomon RZ, Graubard BI, Chari S et  al. 2005. Insulin, glu-
National Center for Health Statistics. (2015). Health, United States, 2014: With cose, insulin resistance, and pancreatic cancer in male smokers. JAMA,
Special Feature on Adults Aged 55–​64. Hyattsville, MD: NCHS. 294(22), 2872–​2878. PMID: 16352795.
National Health and Nutrition Examination Survey. Available from:  http://​ Toriola AT, Cheng TY, Neuhouser ML, et  al. 2013. Biomarkers of inflam-
www.cdc.gov/​nchs/​nhanes.htm Accessed June 15, 2015. mation are associated with colorectal cancer risk in women but are not
National Health Interview Survey. Available from:  http://​www.cdc.gov/​nchs/​ suitable as early detection markers. Int J Cancer, 132(11), 2648–​2658.
nhis.htm Accessed June 15, 2015. PMCID: PMC3609926.
394

394 PART III:  THE CAUSES OF CANCER


Troiano RP, Berrigan D, Dodd KW, et al. 2008. Physical activity in the United Wolin KY, Lee IM, Colditz GA, et al. 2007. Leisure-​time physical activity pat-
States measured by accelerometer. Med Sci Sports Exerc, 40(1), 181–​188. terns and risk of colon cancer in women. Int J Cancer, 121(12), 2776–​
PMID: 18091006. 2781. PMID: 17722094.
Tucker JM, Welk GJ, and Beyler NK. 2011. Physical activity in U.S. adults: com- Wolin KY, Yan Y, Colditz GA, and Lee IM. 2009. Physical activity and colon cancer
pliance with the physical activity guidelines for Americans. Am J Prev prevention: a meta-​analysis. Br J Cancer, 100(4), 611–​616. PMID: 19209175.
Med, 40(4), 454–​461. PMID: 21406280. World Cancer Research Fund, and American Institute for Cancer Research.
US Department of Health and Human Services. 2008 Physical Activity 2007. Food, nutrition, physical activity, and the prevention of cancer: a
Guidelines for Americans. Available from: http://​www.health.gov/​pagu- global perspective. Available from:  http://​www.dietandcancerreport.org/​
idelines/​Accessed September 6, 2013. Accessed June 15, 2015.
van Poppel MNM, Chinapaw MJM, Mokkink LB, van Mechelen W, and World Health Organization. 2010. Global Recommendations on Physical
Terwee CB. 2010. Physical activity questionnaires for adults: a system- Activity for Health. Geneva: WHO.
atic review of measurement properties. Sports Med, 40(7), 565–​600. Wu Y, Zhang D, and Kang S. 2013. Physical activity and risk of breast cancer: a
PMID: 20545381. meta-​analysis of prospective studies. Breast Cancer Res Treat, 137(3),
Wang T, Rohan TE, Gunter MJ, et  al. 2011. A  prospective study of inflam- 869–​882. PMID: 23274845.
mation markers and endometrial cancer risk in postmenopausal hor- Xiao Q, Yang H, Wentzensen N, Hollenbeck A, and Matthews C. 2013.
mone nonusers. Cancer Epidemiol Biomarkers Prev, 20(5), 971–​977. Physical activity in different periods of life, sedentary behavior and the
PMCID: PMC3096873. risk of ovarian cancer in the NIH-​AARP Diet and Health Study. Cancer
Watson KB, Carlson SA, Carroll DD, and Fulton JE. 2014. Comparison of Epidemiol Biomarkers Prev, 22(11), 2000–​2008. PMID: 23966580.
accelerometer cut points to estimate physical activity in US adults. J Yager JD, and Davidson NE. 2006. Estrogen carcinogenesis in breast cancer. N
Sports Sci, 32(7), 660–​669. PMCID: PMC4589136. Engl J Med, 354(3), 270–​282. PMID: 16421368.
Weiderpass E, Adami HO, Baron JA, et  al. 1999. Risk of endometrial can- Zapf J, Schmid C, and Froesch ER. 1984. Biological and immunological
cer following estrogen replacement with and without progestins. J Natl properties of insulin-​like growth factors (IGF) I and II. Clin Endocrinol
Cancer Inst, 91(13), 1131–​1137. PMID: 10393721. Metab, 13(1), 3–​30. PMID: 6202445.
Weiderpass E, Vainio H, Kauppinen T, et al. 2003. Occupational exposures and Zhang M, Xie X, Lee AH, and Binns CW. 2004. Sedentary behaviors and
gastrointestinal cancers among Finnish women. J Occup Environ Med, epithelial ovarian cancer risk. Cancer Causes Control, 15(1), 83–​89.
45(3), 305–​315. PMID: 12661188. PMID: 14970738.
Wijndaele K, Brage S, Besson H, et al. 2010. Television viewing time indepen- Zhong S, Chen L, Lv M, et al. 2014. Nonoccupational physical activity and risk
dently predicts all-​cause and cardiovascular disease mortality: the EPIC of ovarian cancer: a meta-​analysis. Tumour Biol, 35(11), 11065–​11073.
Norfolk Study. Int J Epidemiol, 40(1), 150–​159. PMID: 20576628. PMID: 25095977.
 395

22 Hormones and Cancer

ROBERT N. HOOVER, AMANDA BLACK, AND REBECCA TROISI

OVERVIEW to shed light on the role of metabolic pathways in cancer—​pathways


that often involve hormonal control. The recognition of the potentially
Hormones are highly biologically active endogenous compounds that central roles that two such hormones—​insulin and IGF-​1—​may play
control the growth, development, physiology, and homeostasis of in multiple malignancies is one example of this trend (Renehan et al.,
numerous organ systems. Because of this, they have long been thought 2004). Indeed, the very existence of this chapter is further evidence of
likely to play key roles in both normal and abnormal (malignant) an expanded interest beyond sex hormones. The first two editions of
growth. They are also noteworthy for being produced away from the this text had no such chapter, and the third edition had one, restricted
tissues that they control, and are thus secreted into circulating blood to to “Exogenous Hormones.”
reach their target organs. This combination of potent, targeted agents The current chapter is not intended to be a compilation of the state
of growth and development that can be measured in available biologic of our current knowledge of which hormones are, or might be, related
fluids has made them particularly susceptible and relevant to epide- to specific cancers or of cancers of hormone-​producing glands, as these
miologic investigation. In addition, as pharmacologic science has pro- will be summarized, along with the supporting data, in the relevant
gressed, medications containing hormones and hormone antagonists chapters on site-​specific cancers. Instead, we will discuss some of the
have come into widespread use, providing further opportunities for history of the attempts to identify and understand hormonal carcino-
epidemiologic insights into hormonal carcinogenesis. Based on the genesis via epidemiologic investigations, and what we might learn from
development of increasingly more accurate assays to measure sex hor- these efforts. We will begin with the sex hormones, since as noted, they
mones and their metabolites, this has been the most productive subject remain by far the most extensively investigated hormones in relation to
matter area to date, with major advances in understanding the hormonal cancer risk. Following this will be a brief discussion of other hormones
etiology of breast and gynecologic malignancies. It has also taught us that have come under exploration more recently, and likely represent
the challenges and complexities of these studies, such as our continu- the vanguard of a more concerted effort in the near future to exploit the
ing failure to uncover a role for androgens in prostate cancer etiology. opportunities provided by the various “-​omics” and other new molecular
One major lesson learned is that because of the biological importance epidemiologic tools now being developed. Finally, we will also discuss
of hormones, there are multiple mechanisms to regulate them and their how lessons learned thus far might guide us in designing new investiga-
effects. These include some things that can be measured and accounted tions in this era of interdisciplinary studies and rapid expansion of our
for, such as circulating binding proteins that inhibit the hormone’s understanding of the underlying biology of cancer. It is our hope that
ability to bind to the tissues it regulates. However, there are also potent when the time for the fifth edition of this text arrives, the authorship of
effects of other regulatory mechanisms that are not easily accounted this chapter will need to be extensively expanded in order to cover the
for in epidemiologic studies. Perhaps the most important of these are new knowledge brought about by using these new tools to study hor-
hormone receptors to which virtually all hormones bind in order to mones responsible for controlling so much of human physiology.
initiate their actions. A  variety of homeostatic mechanisms control
the activation and availability of these receptors, including increasing
activation in the presence of declining hormone levels, and decreas- SEX HORMONES
ing activation with rising levels. With the major and rapid advances
in molecular biology and molecular epidemiology, we are seeing a Estrogen, progesterone, and androgens (particularly testosterone) are
surge of new opportunities and creative new investigations across the hormones that have specific roles in reproduction and sexual develop-
entire range of human hormones, and anticipate major advances in our ment, and have thus been labeled as sex hormones. Studies of their
understanding of human hormonal carcinogenesis in the near future. effects on cancer risks have consequently been focused on reproduc-
To take full advantage of these opportunities, we will also need to learn tive organs—​specifically, the roles of these hormones in breast, endo-
how to account for the relevant regulatory mechanisms that evolution metrial, and ovarian cancers in women, and in prostate and testicular
has developed to control the effects of these potent biologic agents. cancers in men. While appropriate, this focus is also somewhat myo-
pic since all of these hormones are biologically active in more organs
than simply the reproductive ones, as evidenced by the presence of
INTRODUCTION estrogen receptors in the central nervous, cardiovascular, respira-
tory, gastrointestinal, urinary, and other organ systems (Levin, 2015;
Hormones are chemical signals secreted into the bloodstream that Millas and Liquidato, 2009). The observation of a possible protective
act on distant tissues, usually in a regulatory fashion (Melmed et al., effect of menopausal estrogen therapy on colon cancer risk (Chan and
2016). In humans, hormones have important roles in growth, develop- Giovannucci, 2010), and the recent appreciation that the risk of breast
ment, homeostasis, and maintenance of normal physiology. As such, it cancer associated with circulating levels of testosterone likely may not
would seem that they might also have a central role, for better or worse, be wholly explained by its role as a substrate for estrogen synthesis
in the process and regulation of abnormal growth and development, as indicate the need for broader and more biologically based assessments.
in malignancies. Despite this, with one notable exception, hormones
have garnered little interest in cancer epidemiology. The exception is
Breast Cancer
the area of sex hormones, which for decades has been a major focus
of investigations of cancers of the reproductive system. Currently, we As noted in the introduction, the history of our past attempts at using epi-
may be seeing the beginnings of a broader epidemiologic interest in demiologic approaches to understand hormonal carcinogenesis can teach
hormonal carcinogenesis in the context of the recent revolution in us valuable lessons about productive ways to pursue these goals in the
molecular science, opening possibilities for molecular epidemiology future. Perhaps the most instructive example is the study of breast cancer,

395
396

396 PART III:  THE CAUSES OF CANCER


which has a long history, filled with innovative methods and creative nature of these risk factors, and the compelling findings from animal
insights, as well as missed opportunities and flawed interpretations. In studies (Mceuen, 1939; Nandi et  al., 1995), estrogens were indicted
1700, Bernardino Ramazzini noted that “… cancerous tumors are very as the most likely responsible carcinogen. Geographic pathologists
often generated in the woman’s breast, and tumors of this sort are found also contributed, identifying major international differences in breast
in nuns more often than in other women. Now these are not caused by cancer risk, most notably between East Asian and Western countries
suppression of the menses but rather, in my opinion, by their celibate (Haenszel and Kurihara, 1968), differences that progressively disap-
life” (Ramazzini, 1700). It was over 100  years later that Rigoni-​Stern peared over two to three generations of migrants from low-​to high-​
quantified this observation by noting that the proportional mortality rates risk countries (Ziegler et al., 1993). These geographic differences and
for breast cancer in Verona, Italy, and its surrounding area were six times changes with migration were also attributed to hormonal differences,
higher in nuns compared to women in the general population (Rigoni-​ specifically circulating estrogen levels that were thought to be driven
Stern, 1842). One of the first to link breast cancer risk to ovarian function by major differences in dietary fat consumption (Lea, 1966).
was Beatson, who in 1896 posed the question, “Is cancer of the mamma The belief that hormones, and specifically estrogens, explain these
due to some ovarian irritation, as from some defective steps in the cycle risk factors was so strong that little was done to test these hypotheses.
of ovarian changes; and, if so, would the cell proliferation be brought to a Rather, they were combined to support a unifying hypothesis that vir-
standstill … were the ovaries to be removed” (Beatson, 1896). tually all established breast cancer risk factors were consistent with an
The concept of chemical “messengers” in the body had been recog- explanation of cumulative lifetime estrogen exposure being the cause
nized in the mid-​nineteenth century (Bernard, 1949), and persons with of breast cancer (Henderson and Feigelson, 2000; Marshall, 1993).
certain disorders were being treated with extracts of various endocrine Over time, many observations have been made that seem inconsistent
organs by late in the century (Borell, 1976). However, laboratory enthu- with this hypothesis, and we will return to this point later.
siasm to pursue studies of these compounds did not really take hold until
the early 1900s, when Ernest Starling coined the term “hormone”: “These Exogenous Hormones
chemical messengers, however, or hormones (from the Greek word Surprisingly understudied by the mid-​1970s were estrogen drugs. The
óρμáω, I  excite or arouse), as we might call them, have to be carried first synthetic estrogens were produced in 1938 (Dodds et al., 1938),
from the organ where they are produced to the organ which they affect and shortly thereafter were administered to women to treat various
by means of the blood stream and the continually recurring physiological disorders of pregnancy, menopausal symptoms, and other conditions.
needs of the organism must determine their repeated production and cir- Assessments of the long-​term consequences of these therapies were
culation through the body” (Henderson, 2005). Even with this stimulus, few, and most were methodologically flawed. Indeed, a publication in
it was not until 1929 that the estrogen molecule was identified and char- 1971 (Defares, 1971)  summarized four small follow-​up studies that
acterized (Tata, 2005). However, this was quickly followed by laboratory included 1130 women treated with estrogen drugs for menopause, and
animal experiments linking estrogen exposure to increased breast cancer noted that 74 cases of cancer would have been expected, but only 2
risk (Kordon, 2008). While this led to a substantial and sustained labora- were observed, concluding that exogenous estrogens were profoundly
tory commitment to understanding the role of estrogens in breast and protective for all cancer. Beginning in 1976 (Hoover et al., 1976), and
other cancers, epidemiologic advances had to wait for the maturation of over the course of the next 15 years, a series of epidemiologic stud-
the newly developing field of chronic disease epidemiology. ies of estrogen therapy (ET) for menopause was conducted. Most,
but not all, demonstrated a positive association of breast cancer with
Hormonally Linked Risk Factors duration of ET use. A combined analysis of studies conducted by the
In the 1950s and 1960s, Brian MacMahon (MacMahon et al., 1970), early 1990s found an overall association with breast cancer that was
Abraham Lilienfeld (Lilienfeld, 1956), Ernst Wynder (Wynder et al., particularly consistent and strong among those studies rated as hav-
1960), and several other pioneers in this field turned their attention to ing high-​quality methods (Steinberg et al., 1991). Larger studies and
breast cancer and discovered multiple important risk factors, includ- pooled analyses added to and refined our understanding of the role of
ing many associated with reproduction—​ nulliparity, parity within exogenous estrogen in carcinogenesis. Particularly noteworthy was the
the parous, age at menarche, age at first birth, lactation, and age at finding that stopping ET resulted in a rapid and marked decline in risk
menopause. About the same time, obesity among postmenopausal (Beral and Million Women Study Collaborators, 2003; Collaborative
women was also identified (Kelsey, 1979). Given the hormone-​related Group on Hormonal Factors in Breast Cancer, 1997). In fact, as

Duration of use and


time since last use Cases/Controls RR (FSE)* RR and 99% FCl*

Never-user 12467/23568 1.00 (0.021)


Last use < 5 years before diagnosis
Duration < 1 year 368/860 0.99 (0.085)
Duration 1–4 years 891/2037 1.08 (0.060)
Duration 5–9 years 588/1279 1.31 (0.079)
Duration 10–14 years 304/633 1.24 (0.108)
Duration ≥ 15 years 294/514 1.56 (0.128)
Last use ≥ 5 years before diagnosis
Duration < 1 year 437/890 1.12 (0.079)
Duration 1–4 years 566/1256 1.12 (0.068)
Duration 5–9 years 151/374 0.90 (0.115)
Duration ≥ 10 years 93/233 0.95 (0.145)

0 0.5 1.0 1.5 2.0

Figure 22–​1.  Relative risk (RR) of breast cancer for duration of use within categories of time since last use of HRT. *Source: Collaborative Group on
Hormonal Factors in Breast Cancer. Breast cancer and hormone replacement therapy: collaborative reanalysis of data from 51 epidemiological studies of 52,705
women with breast cancer and 108 411 women without breast cancer. Lancet 1997;350:1047–1059.
 397

Hormones and Cancer 397


depicted in Figure 22–​1 within only a few years of stopping, long-​term initiation, and remaining at about the same level during the trial and
users had risks equivalent to those who never used ET (Figure 22–​1). also for several years after cessation of treatment (Figure 22–​2).
The addition of progestins to ET (EPT) was also a learning experi- Based on the results of observational studies described in the pre-
ence. A year before the 1976 publication linking ET to breast cancer ceding, this finding of apparent protection in the ET group was unex-
risk, two studies (Ziel and Finkle, 1975; Smith et al., 1975) found a pected. On closer inspection, the reduced risk appeared to be driven by
very strong increased risk of endometrial cancer with use of ET. The subjects in the trial who began use 5 or more years after their meno-
combination of strong associations with two common cancers appear- pause, whereas those who began their use closer in time to menopause
ing over a short time period led to a decline in ET prescriptions. Given had risks similar to the placebo group (Prentice, 2009). It has been
that progestins were known to differentiate the endometrium, stopping noted that this pattern of reduced risk could be consistent with the
estrogen-​driven proliferation, formulations combining both an estro- observation that among women with breast cancer, treatments result-
gen and a progestin were introduced and became increasingly popu- ing in long-​term estrogen deprivation can cause “the eventual devel-
lar. Suggestions were made that in addition to lowering the risk of opment and evolution of anti-​hormonal resistant cell populations that
endometrial cancer related to ET, combined formulations would likely emerge with a vulnerability, as estrogen is no longer a survival signal,
also ameliorate any increase in breast cancer (Gambrell, 1984), even but is an apoptotic trigger” (Lewis-​Wambi and Jordan, 2009; Obiorah
though it had been established that progesterone had different biologic et al., 2014). In a very large cohort study that had enough women to
activity in the breast versus the endometrial epithelium (Key and Pike, assess those who had not begun use of ET until 5 years or more after
1988). In 1989, data from a Swedish cohort (Bergkvist et  al., 1989; menopause, these women were not at a reduced risk of breast cancer,
Persson et  al., 1999)  suggested that while endometrial cancer risk but also were not at increased risk, while those who began use within
was substantially lower among the EPT users versus ET users, breast 5  years of menopause were at a significantly increased risk (Beral
cancer was not, and might actually be higher among EPT users. This et al., 2011).
finding of increased breast cancer risk was subsequently replicated in There are also a number of other, more mundane reasons that the
larger data sets (Schairer et al., 2000), indicating significant heteroge- trial may not be able to optimally address the observational findings.
neity in risk between EPT and ET use. One relates to dose–​response. In the ET trial, the average follow-​up
In the mid-​1990s, the Women’s Health Initiative (Rossouw et  al., time at stopping was 6.8 years (Anderson et al., 2004). In several of
1995), a randomized clinical trial of hormone therapy in menopau- the larger observational studies, the excess risk does not appear until at
sal women, was initiated. The main goal of the trial was to determine least 5 to 8 years of exposure, and rises with further use (Collaborative
whether the suggestion from observational studies that hormone ther- Group on Hormonal Factors, 1997; Schairer et  al., 2000). Also, as
apy in menopausal women might provide protection from cardiovascu- noted earlier, most of the observational studies have found evidence of
lar disease was correct, and if so, whether the benefit outweighed the a strong interaction of estrogen use and breast cancer risk with body
risks of treatment, particularly the increase in breast cancer. Women mass index (BMI), with the risks being higher in, and frequently lim-
with an intact uterus were randomized to EPT or placebo, and women ited to, those with a BMI under 25 (Collaborative Group on Hormonal
who had undergone hysterectomy were randomized to ET or placebo. Factors, 1997; Schairer et al., 2000). With only 1110 women with a
The EPT trial was stopped when women had been on the medications BMI under 25 in the ET arm of the WHI, and an expectation of no, or
for an average of 5.2 years. The primary reason for stopping was an a very low level of, excess risk at 6 years of follow-​up, there is likely
excess risk of cardiovascular disease, rather than an anticipated defi- little statistical power to adequately address this question.
cit, among those taking the active drugs (Manson et al., 2003; Writing
Group for WHI, 2002). At the time of stopping, a significant increase in Endogenous Hormones
breast cancer with increasing duration of EPT use, similar to that seen With the plethora of seemingly hormone-​related risk factors for breast
in the observational studies, was observed. Since stopping, the EPT cancer, and with the prime explanatory candidate hypothesized to be
group has experienced a marked reduction in excess breast cancer risk estrogen, one would think that an early response would have been an
(Chlebowski et al., 2009), also similar to the findings in the observa- assessment of circulating levels of estrogen. This did not happen for
tional studies. The ET trial was also stopped early, primarily because of some time, likely due to concern over the quality of assays availa-
an excess risk of strokes in the exposed group. For the women in the ET ble, and particularly with the paucity of prospective cohort studies
group, however, there was no evidence of increased breast cancer risk with biospecimen collections. For whatever reason, it was not until
(Anderson et al., 2012). In fact, at the time of stopping, this group had 30  years after the elucidation of many hormonally related risk fac-
a non-​significant deficit of 20%; early in the post-​intervention period, tors, and 20  years after the first assessments of exogenous estrogen
this deficit remained and became statistically significant (Manson et al., exposure, that reasonable studies of prospectively collected endoge-
2013). Indeed, this association had a rather unusual pattern, as depicted nous estrogens began to appear. Most of the initial studies, although
in Figure 22–​2 with the deficit appearing almost immediately after trial relatively small, did show evidence of a positive association between

No. (%) of Events


HR P value Favors Favors
CEE Placebo (95% Cl) for Difference CEE Placebo

Invasive breast cancer


Intervention 104 (0.28) 135 (0.35) 0.79 (0.61–1.02)
Postintervention 47 (0.26) 64 (0.75) 0.75 (0.51–1.09) .76
Overall 151 (0.27) 199 (0.35) 0.77 (0.62–0.95)

0.50 0.67 1.00 1.50 2.00


HR (95% Cl)

Figure 22–​2.  Cumulative hazard of breast cancer associated with Conjugated Equine Estrogen (CEE), overall and post-​intervention. The hazard ratios
(HRs) are derived from proportional hazards models stratified by prior disease (for outcomes in which women were eligible for enrollment with and
without the prevalent condition), age, and dietary modification randomization group. The P values for differences between the intervention and post-​
intervention phases were calculated from models for the overall mean follow-​up period that also included a time-​dependent term for trial phase. For the
intervention and overall phases, time to event equals 0 on date of randomization. For the post-​intervention phase, time to event equals 0 on February
29, 2004. Source: JAMA 2011;305(13):1305–1314.
398

398 PART III:  THE CAUSES OF CANCER


estradiol and estrone levels and elevated breast cancer risk in post- in Chapter  45 of this volume, the first studies to use this method to
menopausal women that became convincing in larger studies and comprehensively evaluate these pathways in prospective cohort studies
pooled analyses (Key et  al., 2002). More recently, circulating estro- of breast cancer (Dallal et al., 2014; Falk et al., 2013; Fuhrman et al.,
gens in premenopausal women have been linked to increased breast 2012; Moore et al., 2016) found a strong relationship of decreasing risk
cancer risk (Endogenous Hormones and Breast Cancer Collaborative of postmenopausal breast cancer with an increasing proportion of estro-
Group et al., 2013), although at lower levels of risk than for postmeno- gen being metabolized down the 2-​OH pathway. This supported the
pausal women—​somewhat surprising given the much higher absolute mitogenic hypothesis for estrogen-​induced breast cancer. The fact that
estrogen concentrations in premenopausal women. There is also some low breast cancer–​risk populations in Asia have relatively high ratios of
evidence that the magnitude of this risk may differ by time in the men- 2-​OH to 16-​OH metabolites that decline in Asian migrants to the West
strual cycle when estrogen was measured (Fortner et al., 2013). Thus (Falk et al., 2005; Moore et al., 2016), along with more favorable ratios
far, these observations on premenopausal estrogen levels have come being associated with increased levels of exercise (Dallal et al., 2016;
from assessing all breast cancers following hormone measurement. Matthews et al., 2004), suggest that this risk factor may be modifiable.
Very recently, one study was able to assess the association of premen-
opausal estrogen levels separately for breast cancers occurring before Unifying Hypothesis?
and after menopause (Fortner et al., 2013). A doubling of premeno- As previously noted, very early in the pursuit of the epidemiology
pausal estradiol resulted in a 10% increase in risk for breast cancers of breast cancer, risk factors seemed to indict hormones, specifically
occurring in premenopausal women, but a 40% reduced risk for those estrogens, leading to the unifying hypothesis that cumulative lifetime
occurring in postmenopausal women. It is clear that what is needed is exposure to estrogen explained the disease. Unfortunately, this was so
much more focus on studies of the effects of premenopausal hormonal widely accepted that it seemed to discourage any analytic attempts to
profiles in order to understand the underlying biology. robustly test the hypothesis. Over many decades, a number of well-​
Before there were even prospective epidemiologic studies of cir- replicated observations have challenged this assumption.
culating estrogens, both epidemiologists and laboratory investigators
were interested in a potential role for estrogen metabolism, and thus • Obesity, associated with increased levels of estrogen, is a postmeno-
metabolites in carcinogenesis. The most prominent early hypothesis pausal breast cancer risk factor, but only near the time of diagnosis,
from epidemiologists was the estrogen-​fraction hypothesis (Cole and and not when it only existed, even for long periods, at some time in
MacMahon, 1969). Noting that estriol, a metabolite of the estrogens the past (Ziegler, 1997).
estrone and estradiol, was a much “weaker” estrogen than its parents, • Long-​term use of exogenous estrogen therapy by postmenopausal
it speculated that women with larger ratios of estriol to estradiol and women increases risk, but only among current or very recent users,
estrone would be at lower breast cancer risk. Since there were no pro- with even very long-​ term users who have stopped for several
spective studies in which to test such a hypothesis, they tried to assess years having the same risk as never users (Collaborative Group on
its credibility by comparing measurements in populations with vary- Hormonal Factors in Breast Cancer, 1997).
ing breast cancer risks, including women living in high-​and low-​risk • The anticipated additional increased risk associated with exogenous
countries, and among women by their parity, ages at first birth, at men- hormones is not apparent in obese women who take these drugs
arche, and menopause, and other risk factors (MacMahon et al., 1970). (Collaborative Group on Hormonal Factors in Breast Cancer, 1997).
While some of these comparisons were consistent with the hypothesis, • Circulating estrogens in premenopausal women (when estrogen lev-
some were not, and there were some differences between pre-​and els are at their highest) are associated with only modest increases
postmenopausal studies. In addition, as one of the originators of the in breast cancer risk (Endogenous Hormones and Breast Cancer
hypothesis commented, “… as knowledge of the biology of the estro- Collaborative Group et al., 2013) compared to effects in postmeno-
gen fractions developed, the idea that the estrogen fractions compete pausal women (Key et al., 2002).
for binding sites became less attractive” (MacMahon, 2006). • With respect to the international differences in risk, the actual dif-
At about the same time as the estrogen fraction hypothesis emerged, ferences in circulating estrogens between low-​and high-​risk popula-
laboratory investigators, after decades of work on potential mechanisms tions do not appear to explain much of the observed differences in
of carcinogenesis, had also turned their attention to estrogen metabo- breast cancer rates (Hoover, 2012). Moreover, in new data from a
lism. It was noted that the parent estrogens were rapidly metabolized, previously unstudied Asian population (Mongolians), we see very
with the initial step being hydroxylation at the 16, 2, or 4 position of the low rates of breast cancer, but higher estrogen levels than those in
steroid ring, followed by a cascade of metabolites down each of these the West (Troisi et al., 2014).
pathways. It was found that the 16-​hydroxy metabolites had an affinity • Doubt has even been raised about the risk factor that was thought
for the estrogen receptor and thus were strong mitogens stimulating cell to most clearly show a long-​ term estrogen dose effect—​ age at
proliferation. On the other hand, metabolites down the 2 and 4 pathways menarche—​where seemingly a few years more of menstrual cycling
(known as the catechol estrogens) were not strong mitogens, but could would end up contributing to risk many years later. Breast can-
be oxidized to form quinones that interact directly with DNA to form cer incidence in Bangladesh is five times lower than in the United
a variety of adducts that could enhance mutational events (Yager and Kingdom, and South Asians in Britain have a rate double that in
Davidson, 2006). This was particularly true for the 4-​OH pathway. As a South Asia, a pattern that has also been observed with East Asian
result, two distinct hypotheses arose for how estrogen could cause breast populations studied in the past (Houghton et al., 2014). However, in
cancer and maybe other malignancies as well—​a mitogenic hypothesis a recent study of Bangladeshi migrants, the age at menarche was the
and a mutagenic hypothesis (Yager and Davidson, 2006). Each of these same for girls living in Bangladesh as for those who had migrated to
hypotheses developed a constituency within the laboratory community, Britain, and both were the same as for British girls (Houghton et al.,
leading to several decades of experiments and spirited debates on which 2014). What was different was the pubertal transition (Figure 22–​3).
hypothesis was correct, or at least more important than the other. Until As depicted in Figure 22–​3 the interval from adrenarche (when the
recently, for the most part, the epidemiology community had not been adrenal cortex secretes increased levels of androgens) to thelarche
involved in assessing these hypotheses. Indeed, for about a decade, (the onset of breast development) decreased, and that from thelar-
many epidemiologists were largely unaware of the relevant laboratory che to menarche increased with increasing Western acculturation.
work. Even after becoming aware of it, there was very little investi- This is just one study, and conclusions may change with further
gation in human populations because there were no reliable assays to investigations, but it has stimulated investigators to speculate about
measure the relevant metabolites in humans. Eventually, two of the whether the historic age at menarche observation might not operate
metabolites within these pathways were measured (2-​hydroxyestrone by increasing lifetime estrogen exposure, but through early molec-
and 16-​alpha-​hydroxyestrone), with inconsistent results with respect to ular events impacting cancer susceptibility. This is the direction
risk (Cauley et al., 2003; Muti et al., 2000; Ursin et al., 1999). Several that much of the search for the underlying mechanism of the effect
years later, a comprehensive method to assess a range of metabolites of another risk factor, age at first birth, has been taking recently.
across these pathways was finally developed (Xu et al., 2007). As noted Initially, the focus was on possible hormonal changes associated
 39

Hormones and Cancer 399


factor differences for pre-​versus postmenopausal disease. Circulating
estrogen levels, a well-​ established risk factor for postmenopausal
White British 1.6 3.9 breast cancer, is likely associated, but to a much smaller degree, with
premenopausal disease. Likewise, obesity is associated with increased
risk of postmenopausal breast cancer, but actually appears to be pro-
tective for premenopausal disease (Hunter and Willett, 1993). In fact,
British-Bangladeshi 2.2 2.5 the expectation that pre-​and postmenopausal breast cancers should
have a common epidemiology and share the same risk factors could be
the problem. Certainly all aspects of reproductive biology and lacta-
tion have been driven by powerful evolutionary pressures over millen-
nia, while postmenopausal hormonal physiology was likely not subject
Bangladeshi 3.5 1.8
to such pressures. Thus, perhaps one should actually expect there to be
consequential differences between pre-​and postmenopausal hormonal
Years between milestones epidemiology, and exploiting these differences may give us clues to
etiology. For example, normal postmenopausal breast lobules contain
Adrenarche to Thelarche Thelarche to Menarche 4–​5 times the number of cells expressing estrogen receptors than those
present in premenopausal lobules (Bernstein and Press, 1998). This
likely at least partially reflects the frequently observed phenomenon
Figure 22–​3.  Juvenile and pubertal tempo. of an increase in activation of receptors in response to a decline in
the presence of their ligands (De Meyts and Rousseau, 1980). If this
with this risk factor (MacMahon et al., 1970), but most recent efforts relatively simple explanation is responsible for the shift in ER status
have turned to studies of significant molecular changes within the of tumors with age, some focus on a more comprehensive understand-
breast (Sivaraman and Medina, 2002). ing of the epidemiology of ER expression might be useful. Another
emerging possibility is one that has a more general implication as
While the described findings are not consistent with a cumulative well. That is, as our knowledge of hormones and their actions expand,
lifetime exposure hypothesis for estrogen carcinogenesis, they do pro- we are seeing the breadth of hormones that contribute and interact
vide impressive support for estrogen acting relatively late in breast in regulation of specific organs and their functions. One of the more
carcinogenesis, likely as a tumor promoter. In addition, some of these recent of these to be appreciated for the breast is the interrelationship
findings—​lack of additive effects of obesity and menopausal hormone between estrogens and their receptors and the insulin/​IGF system of
therapy, smaller effects for pre-​versus postmenopausal circulating hormones and their receptors. Recent prospective studies have shown
estrogen—​could be viewed as reflecting a potential (upper) threshold stronger associations of circulating levels of IGF-​1 with postmeno-
for estrogen’s influence. This would be consistent with estrogen acting pausal breast cancer risk in the presence of elevated androgens or
mainly via receptors, with saturation of available receptors at some level estrogens (Tworoger et al., 2011). In addition, there is impressive lab-
of concentration. But if we have learned anything from the last 60 years oratory evidence of “cross-​talk” between these two hormonal systems,
of breast cancer research, perhaps we simply should be working to more suggesting that assessment of interactions between these two systems
fully characterize and understand the known risk factors, rather than in human risk might be fruitful (De Marco et al., 2015). However, to
attempting to develop comprehensive explanatory hypotheses. do this comprehensively will likely require not only measurement of
the hormones, but also their receptors, for which practical methods of
Breast Cancer Subtypes measurement in large epidemiologic studies will need to be developed.
It should be noted that the discussions of hormones and breast cancer in Receptors are not the only likely significant modifiers of estroge-
this chapter have focused on the development of our knowledge of its nicity. Sex hormone binding globulin (SHBG) binds these hormones,
etiology over many decades, and therefore we have described studies particularly androgens and estrogens, and can thus modify their impact
of overall breast cancer. More recently, molecular markers in tumors on multiple biologic actions (Thaler et al., 2015). Fortunately, SHBG
have been used to form subgroups that have distinct therapeutic, and can be measured reasonably well in serum and can be evaluated for its
likely etiologic, implications. The first and most widely used catego- impact on the effects of hormones. Indeed, as our colleagues in basic
rization distinguishes between tumors that express estrogen receptors science pursue advances in molecular biology, it has become clear that
and those that do not. As might be expected, most etiologic studies steroid hormones can be bound by numerous proteins in the cell mem-
that address both types find that the estrogen-​related risk factors tend brane (Caldwell et al., 2016). As is frequently the case, it will be up to
to be much stronger in, and sometimes limited to, the estrogen receptor epidemiologists to determine which aspects of this continuing elucida-
positive (ER+) malignancies (Press et al., 2011; discussed comprehen- tion of the complexity of biologic processes need to be accommodated
sively in the reproductive factors section of Chapter 45 in this volume). in order to come to meaningful conclusions relevant to disease risk and
Studies of recent time trends in incidence have noted a steady decline prevention opportunities at the population level.
in incidence of ER–​tumors and an increase in ER+ tumors, trends While exploring new possibilities for hormones that might play a
that are predicted to continue at least through 2030 (Anderson et al., role in breast cancer etiology, we need to also include other sex hor-
2011; Rosenberg et al, 2015). Discerning the reasons for these patterns mones that seem obvious, but have not yet been thoroughly investi-
would seem to be a high priority. gated. The most notable of these are progesterone, testosterone and
Until recently, the classification of subtypes for therapeutic pur- prolactin. Given that the proliferative action of progesterone in breast
poses has used classical immunochemistry (e.g., estrogen, progester- duct tissue has been known for decades (Graham and Clarke, 1997),
one, and HER2 receptors) in combination with clinical and pathologic and its carcinogenic action as a component of menopausal hormone
variables. The recent introduction of high-​throughput gene-​expression therapy has been known for some time (Schairer et al., 2000), remark-
analyses has introduced a whole new and complex method for predict- ably little has been done to assess its role at physiologic levels in
ing tumor response to treatments (Blows et al., 2010; Dai et al., 2015). breast cancer etiology (Endogenous Hormones and Breast Cancer
How many prognostic categories this will result in and how relevant Collaborative Group, 2013; Fortner et al., 2013; Kaaks et al., 2014).
any of this will be to etiology are unknown, but worth keeping track of. With respect to testosterone, while not pursued as extensively as
estrogen, prospective studies have fairly consistently found circulating
Future Prospects levels to be positively associated with both pre-​(Kaaks et al., 2014) and
Some of the seemingly conflicting evidence for hormonal effects in postmenopausal breast cancer (Key et al., 2002). Until very recently,
breast cancer risk could be clues to a deeper understanding of hor- the association in postmenopausal women has frequently been writ-
monal carcinogenesis, but for the most part has not been aggressively ten off as only reflecting testosterone’s role as the molecular source
pursued with this in mind. A good example of this is the striking risk for the synthesis of estrogen in peripheral fat. However, this seems
40

400 PART III:  THE CAUSES OF CANCER


Table 22–​1. The Effect on Basal Serum Prolactin Levels of Pregnancy, underlying mechanisms in human populations in order to determine
Parity, and the Elapsed Time after the Last Delivery if and how we can use this information for prevention. We have been
able to recommend no or minimal use of menopausal hormone ther-
Serum Prolactin ng/​ml apy, have emphasized the need to reduce obesity, and have developed
Variable No. (mean = SE) P Value* certain hormone-​targeted chemoprevention interventions for women
whose risk/​benefit ratio supports such treatment. The last example
Pregnancy
is unfortunately also an example of how, even when we understand
Nulliparous 19 8.85 ß 1.39 enough about a specific aspect of hormonal carcinogenesis to develop
a preventive intervention, the pervasive level of biologic activities of
Parous 29 4.77 ß 0.43 < 0.01
hormones can discourage meaningful uptake of such an intervention.
No. of Clinical trials of tamoxifen and raloxifene, medications that can block
pregnancies estrogen receptors, have documented reductions in breast cancer risk
of 40%–​50% (Fisher et al., 1998; Vogel et al., 2006). They also have
1 10 5.18 ß 0.47 documented consequential adverse biologic effects. Statistical model-
2 14 4.61 ß 0.48 ing of benefit/​risk ratios have identified groups of women for whom
3 5 4.40 ß 0.35 NS benefits outweigh risks (Gail, 2012), and such prevention efforts have
been widely promoted for these groups. These efforts have been widely
Time since last rejected by the women involved, who have made quite different per-
delivery (mo) sonal risk/​benefit judgments (Hoerger et al., 2016; Smith et al., 2016).
Because the hormonal component for breast cancer is so pro-
12–​36 10 4.15 ß 0.42
found, there has been hope that with a deeper understanding of the
37–​78 10 4.87 ß 0.53 relevant underlying normal and abnormal hormonal mechanisms, we
79–​150 9 4.83 ß 0.25 NS might learn how to use this knowledge for more effective preventive
strategies.
NS = not significant.

Prostate Cancer
less likely to explain its association with premenopausal breast cancer,
where circulating estrogen levels primarily reflect ovarian production. Given the role of testosterone, or androgens generally, in the growth,
For postmenopausal disease, in studies that include both estrogen and function, and maintenance of the prostate gland, as well as how prom-
testosterone in their models, both hormones remain associated, but at inently androgens are associated with tumor activity once the disease
reduced levels (Key et al., 2002). Clarifying the role and mechanism(s) develops, it has been surprising and mystifying to clinicians, epide-
of action of testosterone’s effect on breast cancer risk would seem to miologists, and laboratory scientists alike that circulating levels of
be a high priority for both epidemiology and laboratory investigators. these hormones have not been consistently related to prostate cancer
Prolactin, with its major role in normal breast physiology, has also risk (Roddam et al., 2008). Attention has turned to the possibility that
long been thought of as a candidate hormone for impact on breast can- other hormones such as estrogen, or the balance of estrogen and tes-
cer risk. This enthusiasm was enhanced when it was determined that tosterone, may be important. In men, most testosterone is converted to
normal levels of circulating prolactin were reduced by about 50% fol- dihydrotestosterone (DHT) by 5a reductase, but may also be enzymat-
lowing a first birth, and further reduced by smaller amounts with each ically converted to estrogens by aromatase (CYP19). While it has been
successive birth (Musey et al., 1987; Table 22–​1). This led to specula- hypothesized that higher levels of intraprostatic DHT may be associated
tion that this lowering might explain the age at first birth and parity risk with increased prostate cancer risk (Platz and Giovannucci, 2004), it is
factors for breast cancer. Unfortunately, as noted in Chapter 45, studies also possible that if 5a reductase activity is inhibited, excess testoster-
to date have not consistently linked circulating prolactin to breast can- one may instead be metabolized to estradiol. The bulk of the evidence
cer risk (Tikk et al., 2014). Some of this inconsistency may relate to shows no association of estradiol concentrations and prostate cancer
measurement issues. There are in fact a number of different isoforms risk, and findings for the association of the estradiol:testosterone ratio
of prolactin, with substantial differences in size and shape. From a and prostate cancer risk have been mixed. A recent study of aggressive
teleological perspective, this would seem to imply that these prolactin prostate cancer in the PLCO cohort study simultaneously assessed the
variants might have different biologic roles. Some have attempted to effects of testosterone and the estradiol:testosterone ratio and found
identify “bioactive” forms of prolactin using various immunoassays. a marked attenuation of an association with testosterone, but min-
The most recent effort found some evidence of increased risk with imal effect on the inverse association for the estradiol:testosterone
a bioactivity assay in a subgroup of postmenopausal women whose ratio (Black et al., 2014). Estrogen’s metabolites have drawn attention
blood was sampled relatively close in time to breast cancer diagnosis recently in prostate cancer epidemiology, as they have in breast can-
(Tworoger et al., 2015). A more definitive resolution of the role of pro- cer epidemiology, because of improvements in assay methodologies
lactin will probably require a comprehensive, agnostic assessment of used to measure them. A recent meta-​analysis (Roddam et al., 2008)
all of its isoforms, a technology not yet available. showed an increased risk of prostate cancer with higher urinary 16a-​
hydroxyestrone and a protective effect of excretion of urine with a
Summary higher 2:16a-​hydroxyestrone ratio, neither of which was observed in
While the saga of attempts to identify hormonal risk factors for breast a recent study of aggressive disease (Black et al., 2014). In contrast,
cancer, and the underlying hormones and mechanisms involved in car- the latter study reported a positive association between serum 2:16a-​
cinogenesis, has spanned centuries, revealing associations that have hydroxyestrone ratio and aggressive prostate cancer (Black et al.,
markedly increased our understanding of some of the elements of 2014). The inverse association for estradiol:testosterone ratio, together
breast cancer etiology, as yet only a few provide practical opportuni- with the increasing risk with an increasing proportion of estrogen
ties for prevention. On the other hand, much remains unknown, despite being metabolized down the 2-​OH pathway, may indicate a potential
many provocative observations. This is itself a commentary on the level role for estrogen metabolism in the development of aggressive pros-
of knowledge that we need in order to translate findings in hormonal tate cancer. In breast cancer model systems, 16a-​hydroxyestrone can
carcinogenesis, as well as in other metabolic processes, into practi- bind covalently to the ER and has been shown to induce abnormal
cal prevention. In much of chemical carcinogenesis, it is sufficient to cell proliferation (Bradlow et al., 1985; Suto et al., 1993), whereas 2-​
identify the carcinogen and design ways of decreasing exposure to it. hydroxyestrone has a weak binding capacity to the estrogen receptor
Since hormones are essential for life and generally are regulated in and has been associated with normal cell differentiation and apoptosis
a fairly tight manner, for the most part we need to understand their (Gupta et al., 1998; Telang et al., 1997).
 401

Hormones and Cancer 401


In contrast, strong estrogenic effects (relative to androgen or antago- variants identified has been prostate cancer. Currently, over 100 such
nistic metabolites of estrogen) may give protection against aggressive susceptibility loci have been identified, likely explaining a meaningful
prostate cancer. Previous work to establish independent associations of proportion of the familial risk. These numbers are also likely to rise
androgens and estrogens with prostate cancer risk may have resulted further with ongoing genetic studies.
in inconclusive findings because these hormones interact as part of As for the vast majority of all the newly identified genetic variants
a complex pathway. Future work should focus on mechanistic stud- associated with cancer risks, we know little about the biologic path-
ies in prostate cancer, which are currently lacking. Given the track ways through which the variants related to prostate cancer risk oper-
record for studies of this malignancy—​of positive findings that are not ate. It will take some time for our laboratory colleagues to address
replicable—​these recent findings clearly require replication before any these questions. However, in the interim, we will have the ability to
further pursuit. investigate how this major role of genetics in prostate cancer etiology
The accumulation of increasing numbers of studies focused on controls, relates to, and/​or interacts with other potential risk factors in
various hypotheses to explain the lack of any consistent association prostate carcinogenesis. Certainly, investing in such efforts with can-
between circulating androgens and prostate cancer risk has not yet led didate hormonal exposures may help to clarify the current enigmatic
to an answer. This has resulted in speculation that perhaps circulating state of our knowledge.
levels of testosterone may not reflect its concentration in the prostate. Our attempts to understand the hormonal etiology of prostate can-
As previously noted, testosterone is one of the hormones for which cer provide a valuable counterpoint to the epidemiologic elucidation
paracrine and autocrine production is particularly noteworthy. While of the role of estrogen and other hormones in breast and gynecologic
the textbook examples of this often focus on testosterone’s role in the malignancies. Hormones are central to normal growth, development,
testes, paracrine production also occurs in the prostate. It would seem and homeostasis, and thus are also likely to be biologically involved
to be a high priority, using modern measurement techniques, to assess in pathologic alterations of these processes. In some instances, these
the levels of correlation between circulating and prostate tissue levels might be easily identified using epidemiologic tools at hand. In other
of androgens and estrogens overall, and for their various metabolic instances, we may need to have a much more detailed understanding of
subtypes. the possible underlying biology involved in order to develop the tools
In addition, as was discussed for the estrogens, it is likely that and methods required to assess effects in human populations. With the
an adequate understanding of the role of androgens in prostate car- recent marked advances in molecular science, and the development of
cinogenesis is also going to require much more information about, molecular epidemiologic methods, we are hopefully entering an era
and measurement of, androgen receptors. In fact, the complexity of where this will be possible.
receptor activity is enhanced in the prostate gland. Both epithelial
and stromal tissues have receptors that have differential expression
that can result in different growth factors expressed by the stroma. ENDOCRINE DISRUPTORS
These actions can mediate the effects of androgen on the epithelium,
potentially influencing malignant initiation. Similar interactions can There is one area of hormonal carcinogenesis that has garnered
also affect the progression of an established malignancy, including the substantial attention in both scientific and lay communities—​ the
induction of direct apoptotic effects, effectively moving androgens potential impact of endocrine-​disrupting agents in the environment
from a cause to a potential treatment of prostate cancer (Berry et al., and the risk of various cancers (Soto and Sonnenschein, 2010). Of
2008; Nieto et al., 2014; Stamatiou and Pierris, 2013). particular concern are exposures to these agents in very early life,
Part of our difficulty in understanding the role, if any, of hormones when most hormone-​driven development of many organs is promi-
in prostate cancer risk may also lie in our ignorance of almost any risk nent. To date, most of the concerns have come from laboratory animal
factors for prostate carcinogenesis. It is often pointed out that many experiments (Fenton et al., 2012; Newbold et al., 1987), augmented
decades ago we knew of three well-​established risk factors—​age, race, by various malformations and malignancies identified in wildlife
and family history—​and this describes our level of knowledge today raised in water sources highly polluted with endocrine-​disrupting
as well. For a long period of time, part of the enthusiasm for pursuing chemicals (Bernanke and Köhler, 2009; Hamlin and Guillette, 2010).
a hormonal etiology was not only the central role of hormones in the Epidemiologically robust studies of cancer risks in humans following
normal function of the prostate, but also the belief that prostate cancer high-​level exposure to suspect endocrine disruptors have been few
showed substantial international variation, similar to breast and other and limited, and have generally not found consistently compelling
hormonally related cancers. This belief was based on major interna- evidence of risk (Soto and Sonnenschein, 2010; Teitelbauma et al.,
tional differences in prostate cancer incidence and mortality, with low 2015). The one notable exception has been in utero exposure to high
rates in Asian and African men, higher rates in Asian migrants to the doses of the synthetic estrogen drug diethylstilbestrol (DES). The
United States, substantially higher rates in white men in the United first evidence of adverse effects of in utero exposure to DES was the
States, and the highest prostate cancer rates in the world in African-​ identification of a cluster of seven cases of the rare tumor clear-​cell
American men. Recent observations have called these conclusions into adenocarcinoma of the vagina in adolescent girls whose mothers had
question. With the improvement of cancer registration in Africa, esti- used the drug in their pregnancies (Herbst et al. 1971; Ulfelder, 1980).
mates of prostate cancer there have risen (Korir et al., 2015). Indeed, a More systematic studies have estimated the attack rate of this tumor
systematic prostate cancer screening study in Ghana produced prostate in the exposed through age 39 to be about 1.6/​1000 women (Melnick
cancer rates similar to those among African-​Americans (Hsing et al., et al., 1987; Troisi et al., 2007). Other studies of this exposure have
2014). Coupled with these observations has been an increasing appre- also identified evidence of increased risk of pre-​malignant lesions of
ciation of the major role of genetics in prostate cancer etiology. While the uterine cervix (Hatch et al., 2001) and adenocarcinomas of the
a family history of prostate cancer has been a well-​recognized risk fac- breast (Troisi et al., 2007). While these are important leads, it should
tor, the magnitude of the role of genetics in this tumor was not appre- also be recognized that the overwhelming adverse impact on women
ciated until recently. A seminal study within the Nordic Twin Cohort of this fetal exposure has been in anatomic and physiologic abnormal-
reported in 2000 that the estimate of heritability for prostate cancer ities resulting in very high rates of infertility and abnormal conditions
was 42%, higher than for any other common cancer (Lichtenstein and complications of pregnancy, as depicted in Figure 22–​4 (Hoover
et  al., 2000). A  more recent update of the study (Hjelmborg et  al., et al., 2011). Both the neoplastic and adverse gynecologic outcomes
2014)  with increased numbers of cases and improved methods esti- risks are higher among women whose mothers received higher doses
mated heritability at 58% (95% CI: 52, 63). Concurrently, over the past and/​or received them early in gestation, as evidenced by the increased
10 years, with the new capability to agnostically interrogate the entire prevalence of vaginal epithelial changes in these women, an anatomic
genome for common genetic variants associated with cancer risk, the marker of high-​dose and/​or early exposure. Overall it is estimated
number of such identified variants has risen from less than 6 to more that 27.5% of women in these high-​risk categories experienced one
than 600—​and counting (Gusev et al., 2016). Initially surprising, but or more serious adverse outcomes by age 55 years as a result of their
in line with the heritability estimates, the cancer with the most such exposure (Hoover et al., 2011).
402

402 PART III:  THE CAUSES OF CANCER

Infertility§§

Spontaneous abortion†§§

Ectopic†§§

Second trimester loss†

Preterm birth†§§

Pre-eclampsia†

Stillbirth‡†

Neonatal deathत

Early menopause

ClN2+§
VEC +
VEC –
Breast cancer ≥ 40§

0.4 1 10 40
HR and 95% Cl (log-scale)

Figure 22–​4.  Health Outcomes According to Diethylstilbestrol (DES) Exposure Status and, in the DES-​Exposure Group, the Presence or Absence of
Vaginal Epithelial Changes (VEC) at a Young Age. Hazard ratios differed significantly between VEC-​positive and VEC-​negative subgroups of the DES-​
exposed group for infertility, spontaneous abortion, ectopic pregnancy, preterm delivery, and neonatal death (P < 0.01), as well as for grade 2 or higher cer-
vical intraepithelial neoplasia (CIN 2+ and invasive breast cancer at an age of 40 years or older (P < 0.05). Total numbers of women vary among outcomes,
primarily reflecting whether all, pregnant, or parous women were included in the analysis, but also owing to some missing responses to the questionnaires
ascertaining the outcome and, to a lesser extent, to specific questions. Data for spontaneous abortion, ectopic pregnancy, and loss of second-​trimester preg-
nancy were computed with age as the time metric and with adjustment for date of birth and cohort. Data for preterm delivery, pre-​eclampsia, stillbirth,
and neonatal death were restricted to pregnant women and were adjusted for number of pregnancies. † The analysis was restricted to gravid women and
adjusted for number of pregnancies. ‡ The analysis was restricted to parous women and adjusted for number of births. § p-​value < 0.05. §§ p-​value < 0.01.
Source: Hoover RN, Hyer M, Pfeiffer RM, et al. Adverse health outcomes in women exposed in utero to diethylstilbestrol. N Engl J Med 2011;365:1304–1314.

Many fewer males exposed in utero to DES have been systemati- outcomes, but there have been relatively few relevant outcomes overall,
cally studied, limiting our ability to assess the scope of resulting adverse and the sample size of the cohort is likely not large enough to address
effects. Overall, they have experienced more urogenital anomalies, the issues raised by the animal experiments.
including cryptorchidism (Palmer et  al., 2009), than those unexposed
to DES. Relevant to this, they appear to have incurred about a two-​fold
increased risk of testicular cancer over the adolescent–​young-​adult peak EMERGING OPPORTUNITIES
of this cancer (Strohsnitter et al., 2001). However, this observation fell
short of nominal statistical significance (RR 2.04; 95% CI = 0.82, 4.20). In the following we briefly summarize research on some hormones
Prenatal exposure to high doses of DES has become a key compo- that have received less attention in epidemiologic studies but for which
nent of laboratory research into the carcinogenicity of in utero and other there is increasing enthusiasm that such efforts are warranted.
early-​life exposures to endocrine disruptors (Newbold, 1995). Some of
these studies have found evidence of trans-​generational effects (i.e., an
Anti-​Müllerian Hormone
increase in malignancies in animals not exposed, but whose mothers
were exposed when they were in utero; Newbold et al., 2006). This has Anti-​Müllerian hormone (AMH), also called Müllerian inhibiting sub-
been interpreted as consistent with the mechanism of action being epi- stance (MIS), is produced by the granulosa cells of the ovarian follicles
genetic changes caused by the exposure (in this instance, changes in the and serves as a biomarker of ovarian reserve. It is structurally related
ova of the exposed mother). With respect to studies in humans, there has to inhibin and activin and belongs to the transforming growth factor
been only one study of offspring of mothers exposed when they were β (TGF-​β) family. Available evidence indicates a positive association
in utero (Titus et al., 2008). This cohort (n = 793) of primarily young between AMH concentrations and breast cancer, and no overall associ-
women has not identified any excess in potentially exposure-​related ations with ovarian and prostate cancer. Findings from two prospective
 403

Hormones and Cancer 403


studies (Dorgan et  al., 2009; Nichols et  al., 2015)  demonstrated an Other Hormone Regulators of Energy Metabolism
increased risk of breast cancer with higher compared with lower or
undetectable premenopausal AMH concentrations. In both studies, the Leptin, adiponectin, resistin, and ghrelin are important regulators
risk was most pronounced among women who were older at diagno- of energy homeostasis, and glucose and lipid metabolism. They are
sis compared with those who were younger, and in one study, the risk involved in cell growth and proliferation and inflammation, as well
was limited to ER+ tumors (Nichols et al., 2015). AMH declines as as tumor angiogenesis. Leptin, resistin, and ghrelin are associated
women approach menopause, so higher concentrations could indicate with insulin resistance; in contrast, adiponectin mediates insulin sen-
an older age at menopause. However, adjusting for total and bioavaila- sitivity. This biological activity, in addition to their utility in assess-
ble estrogen and testosterone did not materially change the association ing tumor progression, has raised interest in whether the balance of
of AMH with breast cancer risk (Dorgan et al., 2009). In a study with these hormones plays a role in cancers associated with obesity such
prospectively collected AMH concentrations, there was no association as colorectal, pancreatic, hepatic, ovarian, and postmenopausal breast
with risk of invasive serous ovarian cancer overall, but results varied cancer.
by age at which AMH was measured: in women who were older com- While leptin and resistin were not associated with postmenopausal
pared with younger, higher AMH was associated with a decreased risk, breast cancer in a study nested within the Women’s Health Initiative
whereas in women who were younger compared with older, higher (Gunter et al., 2015), there was suggestion of an inverse trend in risk
AMH was associated with an increased risk (Schock et  al., 2014). with increasing adiponectin levels, the latter consistent with results of
AMH measured prior to diagnosis was not associated with prostate meta-​analyses (Liu et al., 2013; Macis et al., 2014; Ye et al., 2014).
cancer in the PLCO (Sklavos et al., 2014). However, the positive association was not independent of circulating
insulin, suggesting that the effects of adiponectin on breast cancer
risk are partly mediated by insulin. In fact, insulin, estradiol, and C-​
reactive protein together appeared to explain the association of BMI
Insulin, Growth Hormone, and Insulin-​Like
with breast cancer risk in this study.
Growth Factor Elevated leptin levels have been associated with increased risk of
IGF-​1 is similar in chemical structure to insulin, as its name implies. endometrial and ovarian cancer (Wu et al., 2014). Reduced levels of
Insulin increases bioactive IGF-​ 1 through various mechanisms, adiponectin have been associated with an increased risk of endometrial
including regulation of growth hormone (GH). Additionally, insulin cancer (Cust et al., 2007; Luhn et al., 2013; Wu et al., 2014; Zheng
reduces hepatic release of IGF binding protein-​1 (IGFBP-​1), which et  al., 2015), with risk appearing to be higher among obese women
binds with high-​affinity to IGF-​1, thereby reducing its bioactive form. than non-​obese women and among post-​or perimenopausal women
IGF-​1 effects on cellular proliferation, together with strong antipop- compared with premenopausal women (Cust et al., 2007; Zheng et al.,
totic activity, results in hyperproliferation, and thus IGF-​1 is an active 2015). In one study, much of the association between adiponectin and
player in cancer angiogenesis and metastasis. endometrial cancer was attenuated after adjustment for other obesity-​
A systematic review and meta-​analysis of IGFs and IGFBPs and related physiological factors such as C-​peptide, IGFBP-​1, IGFBP-​2,
cancer (Renehan et al., 2004) concluded that higher circulating total SHBG, estrone, or free testosterone (Cust et al., 2007).
concentrations of IGF-​1 were associated with prostate, colorectal, and Colorectal cancer risk also was assessed in relation to hormones
premenopausal breast cancer. Dose–​response associations were con- in postmenopausal women who participated in the Women’s Health
firmed for prostate and premenopausal breast cancer but not for colon Initiative. Leptin, but not adiponectin or resistin, remained positively
cancer. Except for a possible inverse association of lung cancer with associated with colorectal cancer risk after adjustment for insulin (Ho
higher concentrations of IGFBP-​3, there were no protective effects et  al., 2012). Results of a meta-​analysis were consistent with these
with any of the other cancer sites. findings for leptin, which showed a positive association with colorec-
A modest positive association of IGF-​1 concentrations and breast tal cancer in prospective studies, while adiponectin was also shown
cancer risk was demonstrated in a large, pooled analysis (Endogenous to be inversely associated with colorectal cancer risk (Joshi et  al.,
Hormones and Breast Cancer Collaborative Group, 2010), which was 2014). Limited data suggest a positive association between resistin and
independent of IGFBP-​3, its main binding protein. The association colorectal cancer risk (Joshi and Lee, 2014). In addition to their role in
between IGF-​1 and breast cancer risk did not vary by timing of blood obesity and metabolic syndrome, cytokines are of interest in carcino-
collection in regard to menopause, but was only observed for ER+ genesis because of their possible impact on the microbiome, given that
tumors and not ER–​tumors. IGFBP-​3 was not associated with pre- several are secreted in the gut.
menopausal breast cancer, and a weak positive association of IGFBP-​ Leptin concentrations have been positively associated (Stolzenberg-​
3 with breast cancer risk in postmenopausal women was attenuated Solomon et  al., 2015), and adiponectin has been inversely associ-
with adjustment for IGF-​1. While independent associations between ated, with pancreatic cancer risk (Stolzenberg-​Solomon et al., 2008).
IGF-​ 1 and breast cancer, when demonstrated, have been modest, Adiponectin concentrations have been inversely associated with pros-
there is some evidence of stronger associations with postmenopausal tate cancer risk in several nested case-​control studies (Liao et  al.,
breast cancer risk in the presence of elevated androgens or estrogens 2015), but one study found no association with aggressive prostate
(Tworoger et al., 2011). tumors (Stevens et al., 2014). A meta-​analysis of hepatocellular car-
The associations of greater circulating concentrations of IGF-​1 and cinoma showed an association with high adiponectin levels, but it is
lower concentrations of IGFBP-​3, and perhaps IGFBP-​2, have been unclear whether values were a result of the tumor or played a causal
found for risk of colorectal cancer and high-​risk adenoma, but not low-​ role (Song and Gu, 2015).
risk tubular adenomas (Giovannucci, 2001). IGF-​1 is positively asso-
ciated with prostate cancer risk, but an association for low IGFBP-​1
is inconsistent, and there is little evidence of associations with IGF-​2, Thyroid Hormones
IGFBP-​1, and IGFBP-​2 (Rowlands et al., 2009). There are limited data While several studies have assessed the associations of thyroid condi-
on IGFs and other cancers, particularly using pre-​diagnostic biosamples. tions or the use of thyroid hormones and cancer risk, few have pro-
Studies have provided no evidence of an association of IGFs and ovar- spectively collected biological measures of thyroid hormones to assess
ian cancer (Tworoger et al., 2007) but have suggested the possibility of causality. Results for thyroid status and breast cancer based on cross-​
reduced testicular cancer risk with high IGF-​1 levels (Chia et al., 2008). sectional studies have been mixed. Two prospective studies with thy-
Results from a meta-​analysis of studies assessing the effect of exog- roid measurements also had discrepant findings, with one showing
enous insulin use on cancer risk in diabetics suggested positive asso- associations of a diagnosis of hypothyroidism, thyroid medication use,
ciations for pancreas, liver, kidney, stomach, and respiratory cancer, and low thyroxine (T4) with an elevated breast cancer risk (Kuijpens
and a decreased risk for prostate cancer (Karlstad et al., 2013). The et  al., 2005), and one showing a positive association of triiodothy-
data, however, were limited by lack of information on dose, duration, ronine (T3), which is more potent than T4, with breast cancer risk
and potential confounders. (Tosovic et al., 2010) and mortality (Tosovic et al., 2013).
40

404 PART III:  THE CAUSES OF CANCER


In a nested case-​control study in the United Kingdom (Boursi et al., cycle; secretion of insulin and several other hormones is profoundly
2015), colorectal cancer risk was increased in untreated hypothyroid- influenced by recency of food intake; other hormones have appreciable
ism and in those with hyperthyroidism compared with having no thy- diurnal variation, such as prolactin, for which secretion occurs episodi-
roid condition, while long-​term thyroid hormone replacement therapy cally, peaking during sleep and substantially declining by late morn-
was protective, particularly in women. ing; and still other hormones vary substantially by levels of physical
Higher serum thyroid stimulating hormone (TSH), which activates or psychological stress. In some studies, these and other issues can
T3 and T4, has been associated with a decreased risk of prostate cancer be taken into account by timed and/​or multiple blood collections.
compared to men with lower levels, and hypothyroidism based on T4 However, often this is not possible, including in the common practice
and TSH measures was associated with a lower risk compared with of using large established prospective cohorts. In these circumstances,
men classified as euthyroid (Mondul et al., 2012). The causal associa- it is hoped that biologic collections would have been conducted in a
tion of thyroid hormones and thyroid cancer is surprisingly unclear. systematic manner, or at least that the timing and circumstances of the
A  prospective study in South Korea showed that T4 was positively collections would have been recorded, allowing for some control in the
associated with thyroid cancer risk in women, but only in measure- analysis, but frequently even this is not possible. The result of inad-
ments collected within 3 years before diagnosis (Cho et al., 2014). equate control for these determinants of hormonal exposures would
likely result in random misclassification of exposure and the resulting
biasing of any association toward the null. This has been speculated
Cortisol as the reason for the unexpectedly weak associations of prolactin and
Despite wide interest in the possible association of hormones indica- estrogen with breast cancer risk in premenopausal women.
tive of chronic stress and cancer risk, research has been limited by the A recurring measurement dilemma in designing studies of hormone-​
lack of a valid, reproducible biomarker. In a Danish study of women, related cancers is when, if ever, to use a case-​control design. Certainly
hormone-​dependent cancers were not associated with the ratio of cor- these can be done more quickly and can more easily achieve larger
tisol to estrogen, but neither was the ratio associated with self-​reported sample sizes with considerable cost-​efficiency compared to de novo
stress (Rod et al., 2010). A positive association between the ratio of cohort studies. However, they are rarely conducted because of two con-
cortisol to dehydroepiandrosterone sulfate (DHEAS) and cancer mor- cerns. If hormones are localized or stored in certain organs, a malig-
tality, adjusted for smoking and concurrent illness at diagnosis, was nancy in the organ may disrupt homeostasis and thus release more
found in a study of Vietnam-​era male veterans (Phillips et al., 2010). hormones into circulation. Alternatively, if the tumor can manufacture
the hormone, an established capability of breast cancers, then circulat-
ing levels may not represent those during carcinogenesis. All of this
Melatonin certainly supports the cohort approach. However, as noted previously,
case-​control studies of early-​stage breast cancer have found associa-
Associations of night-​shift work with cancer incidence have raised tions with circulating estrogens similar to those from prospective stud-
interest in disruption in secretion of the hormone melatonin, which is ies. Perhaps in a practical sense, doing such studies on early-​stage
linked to circadian rhythm. Evidence using this biomarker is limited cancers could inform the level of enthusiasm for developing compre-
to studies of breast and prostate cancers. A meta-​analysis of five pro- hensive cohort studies, particularly for less common malignancies.
spective studies with concentrations of the primary urinary metabolite A much less well-​recognized measurement issue is whether (and
of melatonin, 6-​sulfatoxymelatonin (aMT6s), showed an inverse asso- if so, how) to accommodate paracrine signaling, a form of local cell-​
ciation with breast cancer risk (Basler et al., 2014), although whether cell communication that uses the same pathways as hormonal signals
this association holds in premenopausal women remains unclear (Melmed et al., 2016). Because of this shared response machinery, tar-
(Brown et al., 2015). Icelandic men with morning aMT6s levels below get cells respond similarly to the signals from the blood and from adja-
the median compared with those with levels above the median had cent cells. Indeed there are several analytes, most notably testosterone
no difference in prostate cancer risk overall, but an increased risk for and IGF-​1, that function both as hormones and paracrine signals. The
advanced disease (Sigurdardottir et al., 2015). amounts of paracrine factors produced per cell are very small. In addi-
tion, because the paracrine factor’s point of action is so constrained
(often one cell), elaborate cellular mechanisms have evolved to con-
LESSONS LEARNED strain its diffusion. Thus any attempts to measure it require tissue sam-
ples and often single-​cell assays. While this is not particularly practical
We have tried to briefly describe the evolution of attempts to under- in epidemiologic studies, it also would not likely reflect total organ
stand the roles that hormones play in cancer etiology. This includes doses. While some studies of tumor specimens use these techniques to
many efforts over an extended period of time focused on several ste- address specific molecular questions, for both practical and scientific
roid sex hormones, and more recent opportunities and initiatives for a reasons it seems that answering epidemiologic questions relating to
diverse set of other hormones. While important discoveries and mean- normal tissue exposure will, for the foreseeable future, need to rely on
ingful progress have been made, there is much that remains to be dis- circulating hormone measurements.
covered; with the remarkable recent advances in molecular science, The central role of hormones in so many critical physiologic path-
as well as in epidemiologic and statistical methods, the time may be ways has resulted in a number of homeostatic mechanisms to regu-
right to greatly expand our understanding of hormonal carcinogene- late their production, dissemination, and action. This has impacted
sis in human populations. To that end, our past efforts have taught us epidemiologic approaches in at least two ways. For one, this level of
some useful lessons that we need to remember in starting ambitious control results in the typical distributions of exposure in studies being
new initiatives. constrained to the “normal range,” with little opportunity to study the
effect of the extremes (low or high) of exposure. In some instances,
therapeutic interventions have restored these opportunities (e.g.,
Measurement oophorectomy, sex hormone therapy). However, even without this,
On first impression, it would seem that understanding hormonal effects because of the powerful biologic actions of some hormones, there are
would be well suited to epidemiologic approaches. Active biologic also opportunities to observe important dose–​response effects within
agents that are secreted into the bloodstream to travel to the organs the normal range, as seen with estrogens and IGFs.
they regulate should be easily sampled in populations and related The second way in which the homeostatic mechanisms of hormones
to subsequent biological effects, including carcinogenesis. In some impact epidemiologic approaches is the value of, and sometimes the
instances this is the case, but often the amount of hormone secreted is necessity for, taking account of these physiologic mechanisms of
influenced by a variety of other factors. A few examples of this include control when trying to get a clear picture of how hormones act in
the following: in premenopausal women, concentrations of estrogens, neoplasia. As described earlier, one of these mechanisms involves
androgens, and progesterone vary substantially over the menstrual binding proteins in the circulation that can modify the availability
 405

Hormones and Cancer 405


of the hormones they bind and thus impact the total amount of hor- epithelium at the time of the pregnancy—​specifically, terminal dif-
mones available for the organs through which they travel (Thaler, ferentiation of the ductal epithelium (Russo and Russo, 1995). This
2015). Fortunately, these proteins can be measured and evaluated for seems to have become accepted as the definitive explanation for many
their impact. Various chemical modifications of hormones also rou- working on breast cancer, even though it has not been proven, and
tinely occur, particularly as they pass through the liver. One of the there are a number of other hypotheses that seem as plausible (Medina,
more common modifications is conjugation, particularly sulfation and 2004, 2005).
glucuronidation. These changes are generally viewed as protecting The tendency to embrace attractive hypotheses before they are tested
cells from genotoxic and cytotoxic effects, as well as allowing excre- has extended to the pursuit of very early-​life exposures and cancer
tion via either urine or stool. However, it is also speculated that these risk. This is easy to understand, since these are very difficult hypothe-
metabolites may provide a reserve supply of hormones that could be ses to test directly. However, often there are opportunities to assess the
made available, some of which can induce ER-​mediated signaling general credibility of such hypotheses. A provocative example of both
pathways (Chang, 2011). of these points was the hypothesis that breast cancer risk in women
Finally, there is one measurement challenge in hormonal epidemi- is related to the levels of circulating estrogens they were exposed to
ology that may be more important than all of those mentioned in the while they were in utero (Trichopoulos, 1990). An appreciable portion
preceding: that is, that the biological effects of hormones depend not of this hypothesis was based on two observations:  that offspring of
only on the effects of the hormones themselves, but also on that of pre-​eclamptic pregnancies had a lower risk of breast cancer than those
their receptors—​agents that are not easily measured or evaluated using from normal pregnancies (Ekbom et al., 1992), and that among female
common epidemiologic methods (Eyster, 2016; Katzenellenbogen migrants from Asia to the United States, those who were born in Asia
et al., 2000; Zhu et al., 2006). In terms of their mechanisms of action, had lower breast cancer rates than those living in the United States for
there are two types of hormones—​those that do not enter cells, but a comparable period of time but who were born in the United States
bind with receptors at the cell surface and regulate gene expression (Ziegler et al., 1993). Since pre-​eclampsia was known to be associated
through second messengers via this interaction, and those that enter with lower levels of estrogen, and Asians in Asia were known to have
cells and bind to intracellular receptors in the nucleus of target cells, lower levels of estrogen than Western women, the hypothesis that low
resulting in gene expression. Thus, the effects of hormones are not levels of estrogens during pregnancy might be protective for breast
only a result of the “dose” of the hormone but also the presence, num- cancer seemed quite logical. It took about a decade before replacing
ber, and activity of relevant receptors. As noted in the discussion of assumptions with facts severely undercut this hypothesis. Specifically,
the exogenous sex hormones, there is a general observation that the the lower levels of estrogen in pre-​eclampsia were based on urinary
number of active receptors is often inversely related to the circulat- measurements:  pre-​eclampsia affects liver function through its hall-
ing level of their ligands, representing a sort of host-​cell mechanism mark, hypertension, resulting in a decreased capacity of the liver
to prevent too much or too little hormonal stimulation. One can see to conjugate estrogens so they can be excreted. The lower levels of
how this might lead to an alteration of our usual expectations for evi- excretion and a metabolic backup of estrogen actually result in slightly
dence of dose–​response relationships. But it is probably even more higher levels in circulation (Troisi et al., 2003). In addition, compara-
complicated than that. Multiple co-​activators and co-​repressors exist tive measurement of circulating estrogen levels in normal pregnan-
that can affect the activity of receptors and thus their ability to activate cies in the United States and China determined that, contrary to the
gene transcription (Shibata, 1997). Indeed, transcriptional activity can circumstance in non-​ pregnant women, circulating estrogens were
occur with unliganded receptors that “are able to have a life on their actually higher in Asian versus Western pregnant women (Lipworth
own” (Stellato, 2016). et al., 1999).
While all of the complexities of receptor activity need not be So while creatively developing hypotheses has been and will con-
accounted for in assessing the relationship between a hormonal ligand tinue to be a major aspect of hormone research in cancer etiology, for
and cancer risk, it could represent a key interactive piece in trying to a research area where relevant measurements are often lacking and/​or
understand hormonal carcinogenesis. Being able to measure activated difficult to do, the approach to new hypotheses should probably be to
hormone receptors in malignant tumors has been a key element in clar- robustly challenge them before they become dogma.
ifying etiologically distinct types of breast cancer. The ability to assess In addition, mechanistic hypotheses to explain the biology of hor-
receptor status of normal tissue might well be an important contributor monally related risk factors are almost solely based on the assump-
to understanding organ-​specific hormonal carcinogenesis. tion that hormonal carcinogenesis is the result of a hormone’s direct
action on susceptible cells to stimulate pathways resulting in increased
growth and/​or DNA damage. While this is undoubtedly the case for
some malignancies, there are other scenarios worth considering.
Hypotheses Specifically, as noted, hormone receptors exist in many organs that are
While there are remarkable new opportunities to pursue the carcin- not hormone-​dependent. It may be that alterations in cancer risk in one
ogenic potential of hormones and their metabolites, perhaps equally organ may not be due to direct hormonal action, but may be mediated
important in this pursuit is to question old “knowledge.” The use of through the effects on another organ system. A leading candidate for
quotation marks here is purposeful, since we are not referring nec- such a scenario would be hormonal effects on the immune system. The
essarily to specific facts or results, but more often to the ways these sex hormones, particularly estrogen, have major effects on immune
results have been interpreted and/​or combined. The frequent result of function, and are linked to some of the significant gender differences
this activity is one or more hypotheses to explain the observations. in autoimmune disease risk (Khan and Ansar, 2016; Kovats, 2015). As
This is a critical piece of the scientific method. Unfortunately, when we begin to learn more about the role of immune function and the risks
attractive and logical hypotheses are not quickly robustly tested, they of various cancers, the more attention we will need to devote to fac-
may become accepted as fact. This seems to occur relatively frequently tors influencing such function. A prime candidate might be malignant
in studies of hormones, perhaps because for many hypotheses in this melanoma. The more we learn about the role of the immune system
area, the populations, technologies, or science required to test them in its etiology, prognosis, and treatment (Guennoun et al., 2016), the
simply do not exist at the time they are developed. The example of more relevant its somewhat enigmatic relationships with sex hormone
cumulative lifetime estrogen exposure being a “unifying hypothesis” variables (gender differences in age-​incidence, pregnancy, exogenous
explaining virtually all of breast cancer risk factors was discussed in hormones) seem to become.
our section on breast cancer. Another example is the attempt to explain
the underlying biologic mechanism for the protective effect on breast
cancer risk of a woman having her first birth at a relatively young age. New Knowledge and Opportunities
Initial attempts to explain the effect on the basis of a change in the Recent dramatic advances in etiologic research in general, along with
post-​pregnancy hormonal profile of the mother were not supportive. several preceding decades of research on reproductive hormones,
The next most popular hypothesis was a molecular change in breast
406

406 PART III:  THE CAUSES OF CANCER


have taught us a number of lessons about how we might make more Basler M, Jetter A, Fink D, Seifert B, Kullak-​Ublick GA, Trojan A. 2014.
progress in trying to understand the role of hormones in carcinogen- Urinary excretion of melatonin and association with breast cancer: meta-​
esis in human populations. The advent of new technologies allowing analysis and review of the literature. Breast Care (Basel), 9(3), 182–​187.
extensive measurement of broad categories of biologically important PMCID: PMC4132247
Beatson GT. 1896. On the treatment of inoperable cases of carcinoma of the
analytes (genes, RNAs, metabolites, microbes, etc.) have opened up mamma:  suggestions for a new method of treatment, with illustrative
whole new vistas of opportunities to use molecular epidemiologic cases. Lancet, 148, 162–​165.
approaches to better understand cancer etiology, prevention, and con- Beral V, Million Women Study Collaborators. 2003. Breast cancer and
trol. In some instances we can actually interrogate agnostically large hormone-​replacement therapy in the Million Women Study. Lancet, 362
categories of molecules for their impact on cancer risks. The poster (9382), 419–​427. PMID: 12927427
child for this is genomics where, by using these agnostic approaches, Beral V, Reeves G, Bull D, Green J; Million Women Study Collaborators.
in less than 10  years we have been able to increase the number of 2011. Breast cancer risk in relation to the interval between menopause
known common genetic variants associated with cancer risk from less and starting hormone therapy. J Natl Cancer Inst, 103(4), 296–​305.
than 6 to greater than 600. The challenge here is that most of those PMID: 21278356
Bergkvist L, Adami HO, Persson I, Hoover R, Schairer C. 1989. The risk of
identified have unknown functions, and it is likely to take some time breast cancer after estrogen and estrogen-​progestin replacement. N Engl J
before their mechanisms of action in carcinogenesis can be identi- Med, 321(5), 293–​297. PMID: 2546079
fied and exploited. However, our laboratory colleagues are more than Bernanke J, Köhler HR. 2009. The impact of environmental chemicals on wild-
happy to take on these tasks, since they know the answers will be life vertebrates. Rev Environ Contam Toxicol, 198, 1–​47. PMID: 19253040
directly applicable to human cancer risk. Bernard, C. 1949. An introduction to the study of experimental medicine
For the other “-​omics,” it is likely to be some time before we know, (trans. H. C. Greene), introduction by L. J. Henderson. New York: Henry
and can measure, all possible analytes, and thus be able to comprehen- Schuman.
sively and agnostically test to identify those associated with specific Bernstein L, Press MF. 1998. Does estrogen receptor expression in normal
cancer risks. However, until such a time when this becomes possible, breast tissue predict breast cancer risk? J Natl Cancer Inst, 90(1), 5–​7.
PMID: 9428773
we can systematically assess all of the increasing families of analytes Berry PA, Maitland NJ, Collins AT. 2008. Androgen receptor signalling in pros-
that have been identified and for which reliable measurement is pos- tate: effects of stromal factors on normal and cancer stem cells. Mol Cell
sible. This seems to have led to important new leads in studies of Endocrinol, 288(1-​2), 30–​37. PMID: 18403105
micro-​RNAs (Cordero et  al., 2015), anti-​inflammatory agents (Lang Black A, Pinsky PF, Grubb RL, 3rd, et al. 2014. Sex steroid hormone metabo-
Kuhs et al., 2015), microbiomics (Bultman et al., 2016) and metabo- lism in relation to risk of aggressive prostate cancer. Cancer Epidemiol
lomics (Fages et  al., 2015). Fortunately, but not widely appreciated, Biomarkers Prev, 23(11), 2374–​2382. PMCID: PMC4221438
these same approaches can also expand our opportunities to more Blows FM, Driver KE, Schmidt MK, et  al. 2010. Subtyping of breast can-
comprehensively pursue already known or highly suspect candidate cer by immunohistochemistry to investigate a relationship between
carcinogenic exposures. For example, as described in the discussion subtype and short and long term survival:  a collaborative analysis
of data for 10,159 cases from 12 studies. PLoS Med, 7(5), e1000279.
of sex hormones, the recent advances in our ability to comprehen- PMCID: PMC2876119
sively measure estrogen metabolites has led to confirmation in human Borell M. 1976. Brown-​ Séquard’s organotherapy and its appearance in
populations of a decades-​old hypothesis from laboratory studies—​a America at the end of the nineteenth century. Bull Hist Med, 50(3), 309–​
finding that may have prevention implications. By the nature of their 320. PMID: 791406
role in organ development and function, hormones have been leading Boursi B, Haynes K, Mamtani R, Yang YX. 2015. Thyroid dysfunction, thy-
candidates for a potential role in carcinogenesis, and our enhanced roid hormone replacement and colorectal cancer risk. J Natl Cancer Inst,
opportunities to understand and measure their functions at a metabo- 107(6). PMCID: PMC4838062
lite level should lead to powerful new insights in studies of the long list Bradlow HL, Hershcopf RJ, Martucci CP, Fishman J. 1985. Estradiol 16 alpha-​
of candidate hormones and their metabolites outlined in this chapter. hydroxylation in the mouse correlates with mammary tumor incidence
and presence of murine mammary tumor virus: a possible model for the
One other lesson learned is appropriate to mention in the context of hormonal etiology of breast cancer in humans. Proc Natl Acad Sci USA,
these new opportunities—​that is, the wisdom of, bordering on neces- 82(18), 6295–​6299. PMCID: PMC391040
sity for, truly interdisciplinary studies in order to exploit these oppor- Brown SB, Hankinson SE, Eliassen AH, et al. 2015. Urinary melatonin con-
tunities. Unfortunately, for several decades hormonal carcinogenesis centration and the risk of breast cancer in Nurses’ Health Study II. Am J
was not a pursuit characterized by true collaborations between basic Epidemiol, 181(3), 155–​162. PMCID: PMC4312424
scientists and population scientists. Quite the contrary; more notable Bultman SJ. 2016. The microbiome and its potential as a cancer preventive
were animated disagreements (Cole and MacMahon 1970; Fishman intervention. Semin Oncol, 2016,43(1), 97–​106. PMCID: PMC4789109
et al., 1986; Lipsett, 1971; Smith and Smith, 1970; Sumoff et al., 1975). Caldwell JD, Gebhart VM, Jirikowski GF. 2016. Estradiol’s interesting life at
More recently these features have subsided, but while, by necessity, the cell’s plasma membrane. Steroids, 111, 4–​11. PMID: 27018128
Cauley JA, Zmuda JM, Danielson ME, et al. 2003. Estrogen metabolites and
both laboratory scientists and epidemiologists interested in hormonal the risk of breast cancer in older women. Epidemiology, 14(6), 740–​744.
carcinogenesis work closely with analytic chemists, there is still very PMID: 14569192
little active collaboration with each other. Given the rapidly increasing Chan AT, Giovannucci EL. 2010. Primary prevention of colorectal cancer.
opportunities to understand hormonal carcinogenesis at the molecular Gastroenterology, 138(6), 2029–​2043. PMCID: PMC2947820
level in human populations, the relevant disciplines will need to work Chang M. 2011. Dual roles of estrogen metabolism in mammary carcinogen-
together to maximally exploit these opportunities. esis. BMB Rep, 44(7), 423–​434. PMID: 21777512
Chia VM, Quraishi SM, Graubard BI, et  al. 2008. Insulin-​like growth factor
1, insulin-​like growth factor-​binding protein 3, and testicular germ-​cell
References
tumor risk. Am J Epidemiol, 167(12), 1438–​1445. PMID: 18408226
Anderson GL, Chlebowski RT, Aragaki AK, et  al. 2012. Conjugated equine Chlebowski RT, Kuller LH, Prentice RL, et al. for the WHI Investigators. 2009.
oestrogen and breast cancer incidence and mortality in postmenopausal Breast cancer after use of estrogen plus progestin in postmenopausal
women with hysterectomy:  extended follow-​up of the Women’s Health women. N Engl J Med, 360(6), 573–​587. PMCID: PMC3963492
Initiative randomised placebo-​controlled trial. Lancet Oncol, 13(5), 476–​ Cho YA, Kong SY, Shin A, et  al. 2014. Biomarkers of thyroid function and
486. PMCID: PMC3412626 autoimmunity for predicting high-​risk groups of thyroid cancer: a nested
Anderson GL, Limacher M, Assaf AR, et al. 2004. Effects of conjugated equine case-​control study. BMC Cancer, 14, 873. PMCID: PMC4289269
estrogen in postmenopausal women with hysterectomy:  the Women's Cole P, MacMahon B. 1969. Estrogen fractions during early reproductive life in
Health Initiative randomized controlled trial. JAMA, 291(14), 1701–​1712. the aetiology of breast cancer. Lancet, 1(7595), 604–​606. PMID: 4180132
PMID: 15082697 Cole P, MacMahon B. 1970. Urinary oestrogen profiles and aetiology of breast
Anderson WF, Katki HA, Rosenberg PS. 2011. Incidence of breast can- cancer. Lancet, 2(7664), 153. PMID: 4194538
cer in the United States:  current and future trends. J Natl Cancer Inst, Collaborative Group on Hormonal Factors in Breast Cancer. 1997. Breast can-
103(18),1397–​1402. PMCID: PMC3176776 cer and hormone replacement therapy:  collaborative reanalysis of data
from 51 epidemiological studies of 52,705 women with breast cancer and
 407

Hormones and Cancer 407


108,411 women without breast cancer. Lancet, 350(9089), 1047–​1059. Graham JD, and Clarke CL. 1997. Physiological action of progesterone in tar-
PMID: 10213546 get tissues. Endocrine Rev, 18(4), 502–​519. PMID: 9267762
Cordero F, Ferrero G, Polidoro S, et al. 2015. Differentially methylated microR- Guennoun A, Sidahmed H, Maccalli C, Seliger B, Marincola FM, Bedognetti
NAs in prediagnostic samples of subjects who developed breast cancer in D. 2016. Harnessing the immune system for the treatment of mela-
the European Prospective Investigation into Nutrition and Cancer (EPIC-​ noma:  current status and future prospects. Expert Rev Clin Immunol
Italy) cohort. Carcinogenesis, 36(10), 1144–​1153. PMID: 26168820 12(8), 879–​893. PMID: 27070898
Cust AE, Kaaks R, Friedenreich C, et  al. 2007. Plasma adiponectin levels Gunter MJ, Wang T, Cushman M, et  al. 2015. Circulating adipokines and
and endometrial cancer risk in pre-​and postmenopausal women. J Clin inflammatory markers and postmenopausal breast cancer risk. J Natl
Endocrinol Metab, 92(1), 255–​263. PMID: 17062769 Cancer Inst, 107(9). PMCID: PMC4651104
Dai X, Li T, Bai Z, Yang Y, Liu X, Zhan J, Shi B. 2015. Breast cancer intrinsic Gupta M, McDougal A, Safe S. 1998. Estrogenic and antiestrogenic activities
subtype classification, clinical use and future trends. Am J Cancer Res, of 16alpha-​and 2-​hydroxy metabolites of 17 beta-​estradiol in MCF-​7 and
5(10), 2929–​2943. PMCID: PMC4656721 T47D human breast cancer cells. J Steroid Biochem Mol Biol, 67(5–​6),
Dallal CM, Brinton LA, Matthews CE, et al. 2016. Association of active and 413–​419. PMID: 10030690
sedentary behaviors with postmenopausal estrogen metabolism. Med Sci Gusev A, Shi H, Kichaev G, et al. 2016. Atlas of prostate cancer heritability in
Sports Exerc, 48(3), 439–​448. PMCID: PMC4861700 European and African-​American men pinpoints tissue-​specific regulation.
Dallal CM, Tice JA, Buist et al. 2014. Estrogen metabolism and breast cancer Nat Commun, 7, 10979. PMCID: PMC4829663
risk among postmenopausal women: a case-​cohort study within B~FIT. Haenszel W, Kurihara M. 1968. Studies of Japanese migrants. I. Mortality from
Carcinogenesis, 35(2), 346–​355. PMCID: PMC3908751 cancer and other diseases among Japanese in the United States. J Natl
Defares JG. 1971. Is cancer preventible? Lancet, 297(7690), 135–​136. Cancer Inst, 40(1), 43–​68. PMID: 5635018
De Marco P, Cirillo F, Vivacqua A, Malaguarnera R, Belfiore A, Maggiolini Hamlin HJ, Guillette LJ, Jr. 2010. Birth defects in wildlife: the role of envi-
M. 2015. Novel aspects concerning the functional cross-​talk between ronmental contaminants as inducers of reproductive and developmental
the insulin/​igf-​i system and estrogen signaling in cancer cells. Front dysfunction. Syst Biol Reprod Med, 56(2), 113–​121. PMID: 20377310
Endocrinol (Lausanne), 6, 30. PMCID: PMC4351617 Hatch EE, Herbst AL, Hoover RN, et  al. 2001. Incidence of squamous neo-
De Meyts P, Rousseau GG. 1980. Receptor concepts: a century of evolution. plasia of the cervix and vagina in women exposed prenatally to dieth-
Circ Res, 46 (6 part 2), I3–​I9. PMID: 6991151 ylstilbestrol (United States). Cancer Causes Control, 12(9),837–​845.
Dodds EC, Goldberg L, Robinson R. 1938. Oestrogenic activity of certain syn- PMID: 11714112
thetic compounds. Nature, 141, 247–​248. Henderson BE, Feigelson HS. 2000. Hormonal carcinogenesis. Carcinogenesis,
Dorgan JF, Stanczyk FZ, Egleston BL, et  al. 2009. Prospective case–​control 21(3), 427–​433. PMID: 10688862
study of serum mullerian inhibiting substance and breast cancer risk. J Henderson J. 2005. Ernest Starling and ‘Hormones’: an historical commentary.
Natl Cancer Inst, 101(21),1501–​1509. PMCID: PMC2773186 J Endocrinol, 184(1), 5–​10. PMID: 15642778
Ekbom A, Trichopoulos D, Adami HO, Hsieh CC, Lan SJ. 1992. Evidence of Herbst AL, Ulfelder H, Poskanzer DC. 1971. Adenocarcinoma of the
prenatal influences on breast cancer risk. Lancet, 340(8826), 1015–​1018. vagina: association of maternal stilbestrol therapy with tumor appearance
PMID: 1357410 in young women. N Engl J Med, 284(15), 878–​881. PMID: 5549830
Endogenous Hormones and Breast Cancer Collaborative Group. 2010. Insulin-​ Hjelmborg JB, Scheike T, Holst K, et al. 2014. The heritability of prostate can-
like growth factor 1 (IGF1), IGF binding protein 3 (IGFBP3), and breast cer in the Nordic Twin Study of Cancer. Cancer Epidemiol Biomarkers
cancer risk:  pooled individual data analysis of 17 prospective studies. Prev, 23(11), 2303–​2310. PMCID: PMC4221420
Lancet Oncol, 11(6), 530–​542. PMCID: PMC3113287 Ho GY, Wang T, Gunter MJ, et  al. 2012. Adipokines linking obesity with
Endogenous Hormones and Breast Cancer Collaborative Group, Key TJ, colorectal cancer risk in postmenopausal women. Cancer Res, 72(12),
Appleby PN, et al. 2013. Sex hormones and risk of breast cancer in pre- 3029–​3037. PMCID: PMC3790260
menopausal women: a collaborative reanalysis of individual participant Hoerger M, Scherer LD, Fagerlin A. 2016. Affective forecasting and medica-
data from seven prospective studies. Lancet Oncol, 14(10), 1009–​1019. tion decision making in breast-​cancer prevention. Health Psychol, 35(6),
PMCID: PMC4056766 594–​603. PMCID: PMC4868645
Eyster KM (Ed.). 2016. Estrogen receptors:  methods and protocols. Houghton LC, Cooper GD, Bentley GR, et al. 2014. A migrant study of puber-
New York: Humana Press. tal timing and tempo in British-​Bangladeshi girls at varying risk for breast
Fages A, Duarte-​Salles T, Stepien M, et al. 2015. Metabolomic profiles of hepa- cancer. Breast Cancer Res, 16(6), 469. PMCID: PMC4303203
tocellular carcinoma in a European prospective cohort. BMC Med, 13, Hoover R, Gray LA, Sr, Cole P, MacMahon B. 1976. Menopausal estrogens and
242. PMCID: PMC4581424 breast cancer. N Engl J Med, 295(8), 401–​405. PMID: 180409
Falk RT, Brinton LA, Dorgan JF, et al. 2013. Relationship of serum estrogens Hoover RN. 2012. That recognised risk factors can explain past and present
and estrogen metabolites to postmenopausal breast cancer risk: a nested international differences in breast cancer incidence: misconceptions 5. Br
case-​control study. Breast Cancer Res, 15(2), R34. PMCID: PMC4053199 J Cancer, 107(3), 408–​410. PMCID: PMC3405203
Falk RT, Fears TR, Xu X, et al. 2005. Urinary estrogen metabolites and their Hoover RN, Hyer M, Pfeiffer RM, et  al. 2011. Adverse health outcomes in
ratio among Asian American women. Cancer Epidemiol Biomarkers women exposed in utero to diethylstilbestrol. N Engl J Med, 365(14),
Prev, 14(1), 221–​226. PMID: 15668498 1304–​1314. PMID: 21991952
Fenton SE, Reed C, Newbold RR. 2012. Perinatal environmental exposures Hsing AW, Yeboah E, Biritwum R, et  al. 2014. High prevalence of screen
affect mammary development, function, and cancer risk in adulthood. detected prostate cancer in West Africans: implications for racial dispar-
Annu Rev Pharmacol Toxicol, 52, 455–​479. PMCID: PMC3477544 ity of prostate cancer. J Urol, 192(3), 730–​735. PMCID: PMC4332806
Fisher B, Costantino JP, Wickerham DL, et  al. 1998. Tamoxifen for preven- Hunter DJ, Willett WC. 1993. Diet, body size, and breast cancer. Epidemiol
tion of breast cancer:  report of the National Surgical Adjuvant Breast Rev, 15(1), 110–​132. PMID: 8405195
and Bowel Project P-​1 study. J Natl Cancer Inst, 90(18), 1371–​1388. Joshi RK, Kim WJ, Lee SA. 2014. Association between obesity-​related adi-
PMID: 9747868 pokines and colorectal cancer:  a case-​control study and meta-​analysis.
Fishman J, Hershcopf RJ, Bradlow HL. 1986. Biochemical epidemiology of World J Gastroenterol, 20(24), 7941–​7949. PMCID: PMC4069321
breast cancer. In Dao TL (Ed.), Tumor markers and their significance in Joshi RK, Lee SA. 2014. Obesity related adipokines and colorectal cancer: a
the management of breast cancer. New York: Alan R. Liss. review and meta-​analysis. Asian Pac J Cancer Prev, 15(1), 397–​405.
Fortner RT, Eliassen AH, Spiegelman D, Willett WC, Barbieri RL, Hankinson PMID: 24528064
SE. 2013. Premenopausal endogenous steroid hormones and breast can- Kaaks R, Tikk K, Sookthai D, et al. 2014. Premenopausal serum sex hormone
cer risk:  results from the Nurses’ Health Study II. Breast Cancer Res, levels in relation to breast cancer risk, overall and by hormone receptor
15(2), R19. PMCID: PMC3672790 status -​results from the EPIC cohort. Int J Cancer, 134(8), 1947–​1957.
Fuhrman BJ, Schairer C, Gail MH. 2012. Estrogen metabolism and risk of PMID: 24155248
breast cancer in postmenopausal women. J Natl Cancer Inst, 104(4), Karlstad O, Starup-​Linde J, Vestergaard P, et al. 2013. Use of insulin and insulin
326–​339. PMCID: PMC3283536 analogs and risk of cancer: systematic review and meta-​analysis of obser-
Gail MH. 2012. Using multiple risk models with preventive interventions. Stat vational studies. Curr Drug Saf, 8(5), 333–​348. PMCID: PMC3899599
Med, 31(23), 2687–​2696. PMCID: PMC3926659 Katzenellenbogen BS, Choi I, Delage-​Mourroux R, et  al. 2000. Molecular
Gambrell RD, Jr. 1984. Proposal to decrease the risk and improve the prognosis mechanisms of estrogen action:  selective ligands and receptor pharma-
of breast cancer. Am J Obstet Gynecol, 150(2), 119–​132. PMID: 6476033 cology. J Steroid Biochem Mol Biol, 74(5), 279–​285. PMID: 11162936
Giovannucci E. 2001. Insulin, insulin-​ like growth factors and colon can- Kelsey JL. 1979. A  review of the epidemiology of human breast cancer.
cer:  a review of the evidence. J Nutr, 131(11 Suppl), 3109S–​3120S. Epidemiol Rev, l, 74–​109. PMID: 398270
PMID: 11694656
408

408 PART III:  THE CAUSES OF CANCER


Key T, Appleby P, Barnes I, Reeves G, Endogenous Hormones and Breast Melmed S, Polonsky KS, Reed Larsen P, Kronenberg HM (Eds.). 2016.
Cancer Collaborative Group. 2002. Endogenous sex hormones and breast Williams textbook of endocrinology (13th ed.). Philadelphia: Elsevier.
cancer in postmenopausal women: reanalysis of nine prospective studies. Melnick S, Cole P, Anderson D, Herbst A. 1987. Rates and risks of
J Natl Cancer Inst, 94(8), 606–​616. PMID: 11959894 diethylstilbestrol-​
related clear-​cell adenocarcinoma of the vagina and
Key TJ, Pike MC. 1988. The role of oestrogens and progestagens in the epide- cervix: an update. N Engl J Med, 316(9), 514–​516. PMID: 3807995
miology and prevention of breast cancer. Eur J Cancer Clin Oncol, 24(1), Millas I, Liquidato BM. 2009. Estrogen receptors alpha and beta in non-​target
29–​43. PMID: 3276531 organs for hormone action: review of the literature. Braz J Morphol Sci,
Khan D, Ansar Ahmed S. 2016. The immune system is a natural target for estro- 26(3-​4), 193–​197.
gen action: opposing effects of estrogen in two prototypical autoimmune Mondul AM, Weinstein SJ, Bosworth T, Remaley AT, Virtamo J, Albanes D.
diseases. Front Immunol, 6, 635. PMCID: PMC4701921 2012. Circulating thyroxine, thyroid-​ stimulating hormone, and hypo-
Kordon EC. 2008. MMTV-​ induced pregnancy-​ dependent mammary thyroid status and the risk of prostate cancer. PLoS One, 7(10), e47730.
tumors:  early history and new perspectives. J Mammary Gland Biol PMCID: PMC3484141
Neoplasia, 13(3), 289–​297. PMID: 18661103 Moore SC, Matthews CE, Ou Shu X, et  al. 2016. Endogenous estrogens,
Korir A, Okerosi N, Ronoh V, Mutuma G, Parkin M. 2015. Incidence of can- estrogen metabolites and breast cancer risk in postmenopausal Chinese
cer in Nairobi, Kenya (2004–​2008). Int J Cancer, 137(9), 2053–​2059. women. J Natl Cancer Inst, 108(10). PMID: 27193440
PMID: 26139540 Musey VC, Collins DC, Musey PI, Martino-​Saltzman D, Preedy JR. 1987.
Kovats S. 2015. Estrogen receptors regulate innate immune cells and signaling Long-​term effect of a first pregnancy on the secretion of prolactin. N Engl
pathways. Cell Immunol, 294(2), 63–​69. PMCID: PMC4380804 J Med, 316(5), 229–​234. PMID: 3099198
Kuijpens JL, Nyklíctek I, Louwman MW, Weetman TA, Pop VJ, Coebergh Muti P, Bradlow HL, Micheli A, et al. 2000. Estrogen metabolism and risk of
JW. 2005. Hypothyroidism might be related to breast cancer in post-​ breast cancer: a prospective study of the 2:16alpha-​hydroxyestrone ratio
menopausal women. Thyroid, 15(11), 1253–​1259. PMID: 16356089 in premenopausal and postmenopausal women. Epidemiology, 11(6),
Lang Kuhs KA, Hildesheim A, Trabert B, et al. 2015. Association between reg- 635–​640. PMID: 11055622
ular aspirin use and circulating markers of inflammation: a study within Nandi S, Guzman RC, Yang J. 1995. Hormones and mammary carcinogenesis
the prostate, lung, colorectal, and ovarian cancer screening trial. Cancer in mice, rats, and humans:  a unifying hypothesis. Proc Natl Acad Sci
Epidemiol Biomarkers Prev, 24(5), 825–​832. PMCID: PMC4417370 USA, 92(9), 3650–​3657. PMCID: PMC42019
Lea AJ. 1966. Dietary factors associated with death-​rates from certain neo- Newbold RR, Bullock BC, McLachlan JA. 1987. Testicular tumors in mice
plasms in man. Lancet, 2(7458), 332–​333. PMID: 4161509 exposed in utero to diethylstilbestrol. J Urol, 138(6), 1446–​ 1450.
Levin ER. 2015. Extranuclear steroid receptors are essential for steroid hor- PMID: 3682076
mone actions. Annu Rev Med, 66, 271–​280. PMID: 25587652 Newbold R. 1995. Cellular and molecular effects of developmental exposure
Lewis-​Wambi JS, Jordan VC. 2009. Estrogen regulation of apoptosis:  how to diethylstilbestrol:  implications for other environmental estrogens.
can one hormone stimulate and inhibit? Breast Cancer Res, 11(3), 206. Environ Health Perspect, 103(Suppl 7), 83–​87. PMCID: PMC1518878
PMCID: PMC2716493 Newbold RR, Padilla-​Banks, Jefferson WN. 2006. Adverse effects of the model
Liao Q, Long C, Deng Z, Bi X, Hu J. 2015. The role of circulating adiponectin environmental estrogen diethylstilbestrol are transmitted to subsequent
in prostate cancer: a meta-​analysis. Int J Biol Markers, 30(1), e22–​e31. generations. Endocrinol, 147(6 Suppl), S11–​S17. PMID: 16690809
PMID: 25450645 Nichols HB, Baird DD, Stanczyk FZ, et al. 2015. Anti-​Mullerian hormone con-
Lichtenstein P, Holm NV, Verkasalo PK, et al. 2000. Environmental and heri- centrations in premenopausal women and breast cancer risk. Cancer Prev
table factors in the causation of cancer:  analyses of cohorts of twins Res, 8(6), 528–​534. PMCID: PMC4452465
from Sweden, Denmark, and Finland. N Engl J Med, 343(2), 78–​85. Nieto CM, Rider LC, Cramer SD. 2014. Influence of stromal-​epithelial inter-
PMID: 10891514 actions on androgen action. Endocr Relat Cancer, 21(4), T147–​T160.
Lilienfeld AM. 1956. The relationship of cancer of the female breast to artificial PMID: 24872510
menopause and martial status. Cancer, 9(5), 927–​934. PMID: 13364877 Obiorah IE, Fan P, Jordan VC. 2014. Breast cancer cell apoptosis with phy-
Lipsett MB. 1971. Estrogen profiles and breast cancer. Lancet, 2(7738), 1378. toestrogens is dependent on an estrogen-​deprived state. Cancer Prev Res
PMID: 4108297 (Phila), 7(9), 939–​949. PMID: 24894196
Lipworth L, Hsieh CC, Wide L, et al. 1999. Maternal pregnancy hormone levels Palmer JR, Herbst AL, Noller KL, et al. 2009. Urogenital abnormalities in men
in an area with a high incidence (Boston, USA) and in an area with a low exposed to diethylstilbestrol in utero: a cohort study. Environ Health, 8,
incidence (Shanghai, China) of breast cancer. Br J Cancer, 79(1), 7–​12. 37. PMCID: PMC2739506
PMCID: PMC2362176 Persson I, Weiderpass E, Bergkvist L, Bergström R, Schairer C. 1999. Risks
Liu LY, Wang M, Ma ZB, et al. 2013. The role of adiponectin in breast cancer: a of breast and endometrial cancer after estrogen and estrogen-​progestin
meta-​analysis. PLoS One, 8(8), e73183. PMCID: PMC3749999 replacement. Cancer Causes Control, 10(4), 253–​ 260. PMID:
Luhn P, Dallal CM, Weiss JM, et  al. 2013. Circulating adipokine levels and 10482483
endometrial cancer risk in the prostate, lung, colorectal, and ovarian can- Phillips AC, Carroll D, Gale CR, Lord JM, Arlt W, Batty GD. 2010. Cortisol,
cer screening trial. Cancer Epidemiol Biomarker Prev, 22(7), 1304–​1312. DHEA sulphate, their ratio, and all-​cause and cause-​specific mortality
PMCID: PMC3819202 in the Vietnam Experience Study. Eur J Endocrinol, 163(2), 285–​292.
Macis D, Guerrieri-​Gonzaga A, Gandini S. 2014. Circulating adiponectin and PMID: 20498139
breast cancer risk: a systematic review and meta-​analysis. Int J Epidemiol, Platz EA, Giovannucci E. 2004. The epidemiology of sex steroid hormones and
43(4), 1226–​1236. PMCID: PMC4258781 their signaling and metabolic pathways in the etiology of prostate cancer.
MacMahon B. 2006. Epidemiology and the causes of breast cancer. Int J J Steroid Biochem Mol Biol, 92(4), 237–​253. PMID: 15663987
Cancer, 118(10), 2373–​2378. PMID: 16358260 Prentice RL, Manson JE, Langer RD, et al. 2009. Benefits and risks of post-
MacMahon B, Cole P, Lin TM, et al. 1970. Age at first birth and breast cancer menopausal hormone therapy when it is initiated soon after menopause.
risk. Bull World Health Organ, 43(2), 209–​221. PMCID: PMC2427645 Am J Epidemiol, 170(1), 12–​23. PMID: 19468079
Manson JE, Chlebowski RT, Stefanick ML, et al. 2013. Menopausal hormone Press DJ, Sullivan-​Halley J, Ursin G, et al. 2011. Breast cancer risk and ovar-
therapy and health outcomes during the intervention and extended post- iectomy, hysterectomy, and tubal sterilization in the women’s contracep-
stopping phases of the Women’s Health Initiative randomized trials. tive and reproductive experiences study. Am J Epidemiol, 173(1), 38–​47.
JAMA, 310(13), 1353–​1368. PMCID: PMC3963523 PMCID: PMC3025644
Manson JE, Hsia J, Johnson KC, et al. 2003. Estrogen plus progestin and the Ramazzini B. De morbis artificum diatriba [diseases of workers]. 1713. 2001.
risk of coronary heart disease. N Engl J Med, 349(6), 523–​534. Am J Public Health, 91(9), 1380–​1382. PMID: 11527762
Marshall E. 1993. Epidemiology. Search for a killer: focus shifts from fat to Renehan AG, Zwahlen M, Minder C, O’Dwyer ST, Shalet SM, Egger M. 2004.
hormones. Science, 259(5095), 618–​621. PMID: 8430308 Insulin-​like growth factor (IGF)-​I, IGF binding protein-​3, and cancer
Matthews CE, Fowke JH, Dai Q, et al. 2004. Physical activity, body size, and risk: systematic review and meta-​regression analysis. Lancet, 363(9418),
estrogen metabolism in women. Cancer Causes Control, 15(5), 473–​481. 1346–​1353. PMID: 15110491
PMID: 15286467 Rigoni-​ Stern D. 1842. Fatti statistici relativi alle malattie cancerose che
McEuen CS. 1939. Observations on rats treated with the sex hormones estrin servorino di base alle poche cose dette dal dott. Rigoni Stern il de 23
and testosterone. Am J Cancer, 36(4), 551–​566. PMID: 12264172 settenbre alla Sotto-​sezione di chirurgia del IV Congresso degli scienzati
Medina D. 2005. Mammary developmental fate and breast cancer risk. Endocr Italiani. Giornale per servire ai progressi della patologia e della terapeu-
Relat Cancer, 12(3), 483–​495. PMID: 16172188 tica, 2, 507–​517.
Medina D. 2004. Breast cancer:  the protective effect of pregnancy. Clinical Rod NH, Kristensen TS, Diderichsen F, Prescott E, Jensen GB, Hansen AM.
Cancer Res, 10(1 pt 2), 380S–​384S. 2010. Cortisol, estrogens and risk of ischaemic heart disease, cancer and
 409

Hormones and Cancer 409


all-​cause mortality in postmenopausal women: a prospective cohort study. Tata JR. 2005. One hundred years of hormones. EMBO Reports, 6(6), 490–​496.
Int J Epidemiol, 39(2), 530–​538. PMID: 20022927 PMCID: PMC1369102
Roddam AW, Allen NE, Appleby P, Key TJ. 2008. Endogenous sex hormones Teitelbaum SL, Belpoggi F, Reinlib L. 2015. Environmental impact on breast
and prostate cancer: a collaborative analysis of 18 prospective studies. J development and disease:  advancing research on endocrine disrupt-
Natl Cancer Inst, 100(3), 170–​183. PMID: 18230794 ing chemicals in breast cancer: Expert panel recommendations. Reprod
Rosenberg PS, Barker KA, Anderson WF. 2015. Estrogen receptor status and Toxicol, 54, 141–​147. PMCID: PMC4464965
the future burden of invasive and in situ breast cancers in the United Telang NT, Katdare M, Bradlow HL, Osborne MP. 1997. Estradiol metabo-
States. J Natl Cancer Inst, 107(9). PMCID: PMC4836802 lism:  an endocrine biomarker for modulation of human mammary
Rossouw JE, Finnegan LP, Harlan WR, Pinn VW, Clifford C, McGowan JA. carcinogenesis. Environ Health Perspect, 105(Suppl 3), 559–​ 564.
1995. The evolution of the Women’s Health Initiative: perspectives from PMCID: PMC1469897
the NIH. J Am Med Womens Assoc, 50(2), 50–​55. PMID: 7722207 Thaler MA, Seifert-​Klauss V, Luppa PB. 2015. The biomarker sex hormone-​
Rowlands MA, Gunnell D, Harris R, Vatten LJ, Holly JM, Martin RM. 2009. binding globulin:  from established applications to emerging trends in
Circulating insulin-​like growth factor peptides and prostate cancer risk: a clinical medicine. Best Pract Res Clin Endocrinol Metab, 29(5), 749–​
systematic review and meta-​analysis. Int J Cancer, 124(10), 2416–​2429. 760. PMID: 26522459
PMCID: PMC2743036 Tikk K, Sookthai D, Johnson T, et  al. 2014. Circulating prolactin and breast
Russo J, Russo IH. 1995. Hormonally induced differentiation:  a novel cancer risk among pre-​and postmenopausal women in the EPIC cohort.
approach to breast cancer prevention. J Cell Biochem Suppl, 22, 58–​64. Ann Oncol, 25(7), 1422–​1428. PMID: 24718887
PMID: 8538211 Titus-​Ernstoff L, Troisi R, Hatch EE, et al. 2008. Offspring of women exposed
Schairer C, Lubin J, Troisi R, Sturgeon S, Brinton L, Hoover R. 2000. in utero to diethylstilbestrol (DES):  a preliminary report of benign and
Menopausal estrogen and estrogen-​progestin replacement therapy and malignant pathology in the third generation. Epidemiology, 19(2), 251–​
breast cancer risk. JAMA, 283(4), 485–​491. PMID: 10659874 257. PMID: 18223485
Schock H, Lundin E, Vääräsmäki M, et al. 2014. Anti-​Mullerian hormone and Tosovic A, Bondeson AG, Bondeson L, Ericsson UB, Malm J, Manjer J. 2010.
risk of invasive serous ovarian cancer. Cancer Causes Control, 25(5), Prospectively measured triiodothyronine levels are positively associated
583–​589. PMID: 24562905 with breast cancer risk in postmenopausal women. Breast Cancer Res,
Shibata H. 1997. Role of co-​activators and co-​repressors in the mechanism 12(3), R33. PMCID: PMC2917028
of steroid/​thyroid receptor action. Recent Prog Horm Res, 52, 141–​164. Tosovic A, Bondeson AG, Bondeson L, Ericsson UB, Manjer J. 2013.
PMID: 9238851 Triiodothyronine levels in relation to mortality from breast cancer and all
Sigurdardottir LG, Markt SC, Rider JR, et al. 2015. Urinary melatonin levels, causes: a population-​based prospective cohort study. Eur J Endocrinol,
sleep disruption, and risk of prostate cancer in elderly men. Eur Urol, 168(4), 483–​490. PMID: 23258272
67(2), 191–​194. PMCID: PMC4318783 Trichopoulos D. 1990. Hypothesis:  does breast cancer originate in utero?
Sivaraman L, Medina D. 2002. Hormone-​induced protection against breast can- Lancet, 335(8695):939–​940.
cer. J Mammary Gland Biol, 7(1), 77–​92. PMID: 12160088 Troisi R, Ganmaa D, dos Santos Silva I, et al. 2014. The role of hormones
Sklavos MM, Zhou CK, Pinto LA, Cook MB. 2014. Prediagnostic circulating in the differences in the incidence of breast cancer between Mongolia
anti-​Müllerian hormone concentrations are not associated with prostate and the United Kingdom. PLoS One, 9(12), e114455. PMCID:
cancer risk. Cancer Epidemiol Biomarkers Prev, 23(11), 2597–​2602. PMC4275167
PMCID: PMC4221416. Troisi R, Hatch EE, Titus-​Ernstoff L, et al. 2007. Cancer risk in women pre-
Smith DC, Prentice R, Thompson DJ, Herrmann WL. 1975. Association of natally exposed to diethylstilbestrol. Int J Cancer, 121(2), 356–​ 360.
exogenous estrogen and endometrial carcinoma. N Engl J Med, 293(23), PMID: 17390375
1164–​1167. PMID: 1186789 Troisi R, Potischman N, Roberts JM, et al. 2003. Maternal serum oestrogen and
Smith OW, Smith GV. 1970. Urinary oestrogen profiles and aetiology of breast androgen concentrations in preeclamptic and uncomplicated pregnancies.
cancer. Lancet, 1(7657), 1152–​1155. PMID: 4192103 Int J Epidemiol, 32(3), 455–​460. PMID: 12777436
Smith SG, Sestak I, Forster A, et al. 2016. Factors affecting uptake and adher- Tworoger SS, Lee IM, Buring JE, Pollak MN, Hankinson SE. 2007. Insulin-​
ence to breast cancer chemoprevention:  a systematic review and meta-​ like growth factors and ovarian cancer risk: a nested case-​control study
analysis. Ann Oncol, 27(4), 575–​590. PMCID: PMC4803450 in three cohorts. Cancer Epidemiol Biomarkers Prev, 16(8), 1691–​1695.
Song RR, Gu XL. 2015. Serum adiponectin levels may be associated with the PMID: 17684148
pathogenesis of hepatocellular carcinoma. Tumour Biol, 36(4), 2983–​ Tworoger SS, Rice MS, Rosner BA, Feeney YB, Clevenger CV, Hankinson
2992. PMID: 25492487 SE. 2015. Bioactive prolactin levels and risk of breast cancer: a nested
Soto AM, Sonnenschein C. 2010. Environmental causes of cancer:  endo- case-​control study. Cancer Epidemiol Biomarkers Prev, 24(1), 73–​80.
crine disruptors as carcinogens. Nat Rev Endocrinol, 6(7), 363–​370. PMCID: PMC4294963
PMCID: PMC3933258 Tworoger SS, Rosner BA, Willett WC, Hankinson SE. 2011. The combined
Stamatiou K, Pierris N. 2013. Could testosterone have a therapeutic role in influence of multiple sex and growth hormones on risk of postmenopausal
prostate cancer? Urol J, 10(1), 747–​754. PMID: 23504676 breast cancer: a nested case-​control study. Breast Cancer Res, 13(5), R99.
Steinberg KK, Thacker SB, Smith SJ, et al. 1991. A meta-​analysis of the effect PMCID: PMC3262212
of estrogen replacement therapy on the risk of breast cancer. JAMA, Ulfelder H. 1980. The stilbestrol disorders in historical perspective. Cancer,
265(15), 1985–​1990. PMID: 1826136 45(12), 3008–​3011. PMID: 6992969
Stellato C. 2016. The “busy life” of unliganded estrogen receptors. Proteomics, Ursin G, London S, Stanczyk FZ, et  al. 1999. Urinary 2-​hydroxyestrone/​
16(2), 288–​300. PMID: 26508451 16alpha-​hydroxyestrone ratio and risk of breast cancer in postmenopausal
Stevens VL, Jacobs EJ, Sun J, Gapstur SM. 2014. No association of plasma women. J Natl Cancer Inst, 91(12), 1067–​1072. PMID: 10379970
levels of adiponectin and c-​peptide with risk of aggressive prostate cancer Vogel VG, Constantino JP, Wickerham DL, et al. 2006. Effects of tamoxifen vs
in the Cancer Prevention Study II Nutrition Cohort. Cancer Epidemiol raloxifene on the risk of developing invasive breast cancer and other dis-
Biomarkers Prev, 23(5), 890–​892. PMID: 24585409 ease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR)
Stolzenberg-​Solomon RZ, Newton CC, et al. 2015. Circulating leptin and risk P-​2 trial. JAMA, 295(23), 2727–​2741. PMID: 16754727
of pancreatic cancer: a pooled analysis from 3 cohorts. Am J Epidemiol, Writing Group for the Women’s Health Initiative. 2002. Risks and benefits
182(3), 187–​197. PMCID: PMC4517697 of estrogen plus progestin in healthy postmenopausal women: principal
Stolzenberg-​Solomon RZ, Weinstein S, Pollak M, et  al. 2008. Prediagnostic results From the Women’s Health Initiative randomized controlled trial.
adiponectin concentrations and pancreatic cancer risk in male smokers. JAMA, 288(3), 321–​333. PMID: 12117397
Am J Epidemiol, 168(9), 1047–​1055. PMCID: PMC2720773 Wu M-​M, Chen H-​C, Chen C-​L, et al. 2014. A prospective study of cancer risk
Strohsnitter WC, Noller KL, Hoover RN, et  al. 2001. Cancer risk in men in relation to adiposity factors: Cumulative incidence and association with
exposed in utero to diethylstilbestrol. J Natl Cancer Inst, 93(7), 545–​551. plasma adipokine levels. Plos One, 9(8), e104630. PMCID: PMC4130554
PMID: 11287449 Wynder EL, Bross IJ, Hirayama T. 1960. A study of the epidemiology of cancer
Sumoff B, Fishman J, Bradlow HL, Hellman L. 1975. Hormone profiles in of the breast. Cancer, 13, 559–​601. PMID: 13846284
hormone-​dependent cancers. Cancer Res, 35(11 Pt 2), 3365–​ 3373. Xu X, Roman JM, Issaq HJ, Keefer LK, Veenstra TD, Ziegler RG. 2007.
PMID: 1104149 Quantitative measurement of endogenous estrogens and estrogen metabo-
Suto A, Bradlow HL, Wong GY, Osborne MP, Telang NT. 1993. Experimental lites in human serum by liquid chromatography-​tandem mass spectrome-
down-​regulation of intermediate biomarkers of carcinogenesis in mouse try. Anal Chem, 79(20), 7813–​7821. PMID: 17848096
mammary epithelial cells. Breast Cancer Res Treat, 27(3), 193–​202. Yager JD, Davidson NE. 2006. Estrogen carcinogenesis in breast cancer. N
PMID: 8312577 Engl J Med, 354(3), 270–​282. PMID: 16421368
410

410 PART III:  THE CAUSES OF CANCER


Ye J, Jia J, Dong S, Zhang C, et al. 2014. Circulating adiponectin levels and the Ziegler RG. 1997. Anthropometry and breast cancer. J Nutr, 127(5 Suppl),
risk of breast cancer: a meta-​analysis. Eur J Cancer Prev, 23(3), 158–​165. 924S–​928S. PMID: 9164265
PMID: 23929213. doi: 10.1371/​journal.pone.0129824. eCollection 2015. Ziegler RG, Hoover RN, Pike MC, et al. 1993. Migration patterns and breast
Zheng Q, Wu H, Cao J. 2015. Circulating adiponectin and risk of endometrial cancer risk in Asian-​American women. J Natl Cancer Inst, 85(22), 1819–​
cancer. PLoS One, 10(6):e0129824. PMID: 26030130 1827. PMID: 8230262
Zhu BT, Han GZ, Shim JY, Wen Y, Jiang XR. 2006. Quantitative structure-​ Ziel HK, Finkle WD. 1975. Increased risk of endometrial carcinoma among
activity relationship of various endogenous estrogen metabolites for human users of conjugated estrogens. N Engl J Med, 293(23), 1167–​ 1170.
estrogen receptor alpha and beta subtypes: insights into the structural deter- PMID: 171569
minants favoring a differential subtype binding. Endocrinology, 147(9),
4132–​4150. PMID: 16728493
 41

23 Pharmaceutical Drugs Other Than Hormones

MARIE C. BRADLEY, MICHAEL A. O’RORKE, JANINE A. COOPER, SØREN FRIIS,


AND LAUREL A. HABEL

OVERVIEW Pharmaceutical Drugs
The FDA defines a drug as a substance (other than food) that is
Drug prescribing is one of the most common medical interventions. Yet,
intended to affect the structure or any function of the body and is used
current regulatory programs for drug safety are not designed to identify
in the diagnosis, cure, mitigation, treatment, or prevention of disease
adverse events that are very rare or have a long latency, such as most
(FDA, 2015). Some drugs are enzyme inhibitors, others interfere with
cancers. Meta-​analyses of randomized clinical trials of medications
molecular transport mechanisms, and some bring about their effects
can sometimes provide information on shorter-​term risk of common
entirely through physicochemical properties, or by interfering with
cancer types, though large observational studies with long follow-​up
specific proteins essential for signal transduction (Hill, 2010). Most
are needed to examine the majority of drug–​cancer associations. Over
drug effects are achieved by the interaction of a drug with receptors in
the last few decades, a number of new methods have been developed
target tissues. The intensity and duration of the effects (i.e., pharma-
to address several types of confounding and bias of particular con-
codynamics) are usually determined by the number of drug molecules
cern in pharmacoepidemiology studies, and better data sources have
present, their receptor site binding affinity, and whether the binding
become available. Of the approximately 20 medications with sufficient
is reversible or irreversible (Hill, 2010). Considerable inter-​individual
evidence to be classified by the International Agency for Research on
variation in drug response has been documented (Hill, 2010). These
Cancer (IARC) as human carcinogens, most are anti-​neoplastic agents
are induced by differences in pharmacokinetic processes (absorp-
or immunosuppressants. Substantial data from studies in humans indi-
tion, distribution, metabolism, and excretion) and pharmacodynamic
cate that use of aspirin and other nonsteroidal anti-​inflammatory drugs
effects, as well as by cofactors such as age, gender, comorbidity, drug
(NSAIDs) protects against colorectal cancer and possibly a number of
interactions, and genetic factors.
other common cancers. In the last decade, many other non-​hormonal
drugs have been examined for their suggested carcinogenic or chemo-
preventive effects, but additional human data are needed. In the future, Pharmaceutical Drugs as Chemical Carcinogens
new drugs, databases, and analytic methods are all likely to contribute
to an increase in the number of pharmacoepidemiology studies and Although drugs enter the body in typically higher concentrations
reported drug–​cancer associations. Given their potential public health than most other chemicals in the environment that are known or sus-
importance, it will be essential to carefully examine identified drug–​ pected to be carcinogens, only a couple dozen medications have been
cancer associations within multiple, well-​conducted studies. Before established as human carcinogens (Table 23–​1). However, several
translating findings into regulatory action or clinical care, it also will additional drugs are suspected to be carcinogenic based on labora-
be essential that both the benefits and harms of a medication are care- tory data, but lack sufficient or consistent human data (Table 23–​2).
fully assessed. In addition, a number of drugs have been found or suggested to pos-
sess chemopreventive effects (Friis et al., 2015a; Kelloff et al., 2006).
Carcinogens, including carcinogenic drugs, may act at any stage of
INTRODUCTION carcinogenesis, including initiation, promotion, progression, invasion,
or metastasis (Hanahan and Weinberg, 2011). Mechanisms influenc-
ing carcinogenesis include mutation, DNA repair, cell proliferation,
Global Trends in Drug Utilization differentiation, angiogenesis, immunosurveillance, and apoptosis. In
The use of drugs is increasing dramatically worldwide and each year addition, candidate carcinogenic and chemopreventive agents, includ-
hundreds of new pharmaceutical products are approved by the US Food ing drugs, can affect DNA methylation patterns, endogenous synthesis
and Drug Administration (FDA) and the European Medicines Agency of carcinogens, absorption or metabolism of carcinogenic chemicals,
(EMA) (CenterWatch, 2016). Currently, more than 4000 new medica- generation of free radicals and activated forms of oxygen, or covalent
tions are being evaluated in clinical trials worldwide (CenterWatch, binding of carcinogens to DNA (Erson and Petty, 2006). While many
2016). Within the European Union alone, there were 81 new medica- carcinogens have induction periods of 20 years or more, several drugs
tions recommended by the EMA for marketing authorization in 2013, appear to affect cancer development much earlier after initiation. For
of which 38 contained a de novo active substance (EMA, 2014). example, some immunosuppressants increase the risk of lymphomas
Drug use varies substantially by country. For example, compared to after only a few years of treatment (Kotlyar et al., 2015), and aspirin
the United Kingdom, France, and Spain, the United States and Denmark appears to reduce colorectal cancer risk after 5 years (Cuzick et al.,
generally have higher medication usage, although not for all diseases 2015; Friis et al., 2015b).
(Richards, 2010). The use of specific drugs also varies markedly.
For example, atorvastatin is the preferred statin in the United States,
whereas simvastatin is the most commonly prescribed statin (and the Drug Safety
top prescribed drug) in the United Kingdom (Figures 23–1 and 23–2). All drugs are evaluated in Phase I–​III clinical trials for efficacy and
International variations in drug use can be attributed to differences in safety before they are approved for marketing by regulatory authori-
healthcare system factors, such as the use of restrictive formularies, ties, such as the EMA in the European Union, the FDA in the United
reimbursement initiatives such as drug copayments, regulatory policies States, and similar agencies in other countries (CenterWatch, 2016).
influencing prescribing practices, clinical culture, and the incidence When a drug is first marketed, its efficacy and safety have typically
and prevalence of various diseases and conditions (Richards, 2010). been tested for a maximum of 3–​4  years in relatively small and

411
412

412 Part III: The Causes of Cancer


Levothroxine 119.9
Acetaminophen/hydrocodone 119.2
Lisinopril 103.7
Metoprolol 85.3
Atorvastatin 80.7
Amlodipine 78.3
Metformin 76.9
Omeprazole 75

Drug Name
Simvastatin 72.8
Albuterol 67.1
Amoxicillin 54.5
Fluticasone 50.8
Gabapentin 50.7
Alprazolam 49.4
Hydrochlorothiazide 49.1
Azithromycin 47
Furosemide 46.5
Tramadol 44.2
Sertraline 44.2
Losartan 39.5
0 20 40 60 80 100 120 140
Number of dispensed item (millions)

Figure 23–​1.  Top 20 dispensed prescriptions in the United States in 2014. Source: IMS Health, National Prescription Audit, 2014.

homogeneous patient populations. Thus, pre-​marketing trials will miss spontaneous reporting systems (Dal Pan et  al., 2012). In the United
rare and long-​term adverse drug reactions (ADRs), such as cancer, that States, the Sentinel Initiative was launched in 2008 to enable the FDA
occur after long exposure duration or latency. to evaluate potential safety issues quickly and securely using elec-
tronic health records, administrative and insurance claims databases,
and registries covering approximately 100 million individuals (Avorn
Passive Surveillance of Adverse Drug Events and Schneeweiss, 2009). However, because many of the contributing
In many countries, drug regulatory authorities rely heavily on spontaneous healthcare systems have high enrollee turnover and databases without
ADR reporting systems to monitor drug safety (Dal Pan et al., 2012). In ready linkage to cancer registries, the Sentinel Initiative is not cur-
the spontaneous reporting systems, suspected ADRs are usually reported rently an optimal setting for studies of drug use and cancer risk.
to a central agency by healthcare professionals, manufacturers, or directly In the European Union, the “Exploring and Understanding Adverse
by patients. The most important function of these systems is the early Drug Reactions” (EU_​ADR) project uses integrative mining of various
identification of drug safety signals (Harmark and van Grootheest, 2008), electronic healthcare data for approximately 30 million EU citizens to
which may generate hypotheses, requirements for manufacturers to spon- conduct large-​scale monitoring of drug safety signals and rare ADRs.
sor post-​marketing safety studies, or regulatory warnings and changes of Detected signals are evaluated by semantic mining of the current lit-
product information labels or leaflets. A major limitation of the spontane- erature and computational analysis of pharmacological and biologi-
ous reporting systems is the high level of under-​reporting by healthcare cal information on drugs, molecular targets, and pathways (Coloma
professionals and manufacturers. In addition, the programs are generally et al., 2011).
unable to estimate rates of ADRs because the sizes of the underlying A small number of non-​regulatory-​based surveillance programs
patient populations are unknown (i.e., no denominators). They are also have been developed to identify drug–​cancer signals for hypothesis
unlikely to detect ADRs with only moderate associations with a particular generation. These include the Kaiser Permanente Drug Surveillance
type of drug and ADRs with long latencies, such as cancer. Program, which has been exploring drug–​cancer signals for com-
monly prescribed pharmaceuticals since the 1970s by linking the
electronic pharmacy records of health plan enrollees to cancer regis-
Active Surveillance of Adverse Drug Events try and other health records (Friedman et al., 2009a), and a recently
developed Danish surveillance program, linking pharmacy records
Both the United States and the European Union have recently initi- to cancer registry and other national healthcare data (Pottegård
ated systems for active surveillance of ADRs to complement their et al., 2016a).

Figure 23–​2.  Top 20 dispensed prescriptions in the United Kingdom in 2014. Attribution statement: This chart contains public sector information licensed
under the Open Government licence v3.0 http://www.nationalarchives.gov.uk/doc/open-government-licence/version/3/ Sources: Business Services
Organisation (Northern Ireland), ISD National Services Scotland (Scotland), Welsh Assembly Government (Wales) and the Health and Social Care
Information Centre (England).
 413

Pharmaceutical Drugs Other Than Hormones 413


Table 23–​1.  Non-​Hormone Drugs Evaluated by the IARC Monograph Program* and Classified as Carcinogenic to Humans

Cancer Sites with Sufficient Evidence of


Group 1 Agents Carcinogenicity in Humans Established Mechanisms

Anti-​neoplastic Agents
Busulfan Acute myeloid leukemia Genotoxicity (alkylating agent)
Chlorambucil Acute myeloid leukemia Genotoxicity (alkylating agent)
Chlornaphazine Bladder Genotoxicity (alkylating agent, metabolism to 2-​
naphthylamine derivatives)
Cyclophosphamide Acute myeloid leukemia, bladder Genotoxicity (metabolism to alkylating agents)
Etoposide (Group 2A in 2000) —​ Genotoxicity, translocations involving MLL gene
Etoposide in combination with cisplatin and Acute myeloid leukemia Genotoxicity, translocations involving MLL gene
bleomycin (etoposide)
Melphalan Acute myeloid leukemia Genotoxicity (alkylating agent)
MOPP combined chemotherapy Acute myeloid leukemia, lung Genotoxicity
Semustine (methyl-​CCNU) Acute myeloid leukemia Genotoxicity (alkylating agent)
Thiotepa Leukemia Genotoxicity (alkylating agent)
Treosulfan Acute myeloid leukemia Genotoxicity (alkylating agent)
Immunosuppressive Agents
Azathioprine Non-​Hodgkin lymphoma, skin Genotoxicity, immunosuppression
Cyclosporin Non-​Hodgkin lymphoma, skin, multiple other sites Immunosuppression
Other Carcinogenic Agents
Analgesic mixtures containing phenacetin Renal pelvis, ureter Increased cancer risk cannot be explained by other
components of the analgesic mixtures, i.e., aspirin,
codeine phosphate, or caffeine
Aristolochic acid (Group 2A in 2002) —​ Genotoxicity, DNA adducts in animals are the same as
those found in humans exposed to plants, A:T →
T:A transversions in TP53; RAS activation
Methoxsalen plus ultraviolet A radiation Skin Genotoxicity following photo-​activation
Phenacetin (Group 2A in 1987) Renal pelvis, ureter Genotoxicity, cell proliferation
Plants containing aristolochic acid Renal pelvis, ureter Genotoxicity, DNA adducts in humans, A:T → T:A
transversions in TP53 in human tumours

* IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, Volume 100A (2012), http://​monographs.iarc.fr/​ENG/​Monographs/​vol100A/​.
MOPP = Chlormethine (mechlorethamine), vincristine (oncovin), procarbazine, and prednisone
Source: Table adapted from Grosse et al., Lancet Oncol 2009 10(1): 13–​14.

Targeted Assessments of Drug Carcinogenicity specific cancer types are rare, necessitating special requirements and
diligence in the choice of study designs and data sources.
In addition to regulatory agencies, the International Agency for
Research on Cancer (IARC) and the US National Toxicology
Program (NTP) conduct regular assessments of the evidence of the Study Designs and Data Sources
potential carcinogenicity of medical exposures, including drugs.
The IARC has conducted systematic evaluations of human carcino- Many case-​control and cohort studies have examined drug–​cancer
gens since the 1970s; the evidence for carcinogenicity is evaluated associations using interviews or postal surveys. Increasingly, phar-
by expert committees and is based on comprehensive literature macoepidemiologic studies are being conducted using large health-
reviews of data from animal experiments, bioassays, mechanistic care databases, either from healthcare insurers or providers or from
studies, epidemiologic studies, or clinical trials (Cogliano et  al., national databases and registries, such as those in the United Kingdom
2011). Of the over 900 potential carcinogens evaluated, slightly and other northern European countries (Strom et  al., 2012). While
over 100 have been classified as carcinogenic to humans (IARC, large healthcare databases linkable to cancer registries generally pro-
2012), including approximately 20 non-​ hormonal medications vide high-​quality data on prescription drugs and enable studies of rare
(Table 23–​1). Another nearly 40 non-​hormonal medications have exposures and outcomes (such as drug–​cancer associations), these
been classified as probably or possibly carcinogenic to humans, databases typically do not hold information on non-​prescription drugs
because epidemiological or clinical evidence has not been defin- or on lifestyle factors (e.g., smoking), introducing potential residual
itive, or because carcinogenicity has been demonstrated only in confounding (Strom et al., 2012). In addition, some databases include
experimental animals (Table 23–​2). populations with high turnover or non-​continuous registration, com-
plicating studies of long-​term effects. Further, drug formularies are
common in many healthcare systems and can result in nearly exclusive
use of one or two drugs within a class (e.g., one type of statin). Such
METHODOLOGIC ISSUES IN STUDIES OF DRUGS formularies limit the ability to examine multiple drugs within a class
AND CANCER or to examine the same specific drug in large, collaborative studies.
Most healthcare databases are not designed for research, but rather
Although a randomized clinical trial (RCT) is the optimal design for for administrative or clinical purposes (Strom, 2005). When designing
studies of both drug efficacy and safety (Grossman and Mackenzie, studies, it is important to understand the strengths and limitations of
2005), large observational studies with long-​term follow-​up are gener- the source databases (e.g., claims vs. electronic health records) and to
ally needed to examine drug–​cancer associations. Since findings from recognize that special procedures and expertise are often needed to
such pharmacoepidemiologic studies can have clinical, regulatory, and create research-​quality data sets (Ross et al., 2014). Given the diver-
economic implications, consideration of methodologic issues is of par- sity of data sources and coding, collaborative studies using multiple
amount importance. There are several types of confounding, selection, healthcare databases may benefit from using a common data model
and information biases of particular concern in studies of drugs. In to help standardize and harmonize pharmacy and other data elements
addition, the prevalence of most drug use and the incidence of most (Curtis et al., 2012; Ross et al., 2014).
41

414 Part III: The Causes of Cancer


Table 23–​2.  Non-​Hormonal Drugs Evaluated by the IARC Monograph Program and Classified as Probably (Group 2A) or Possibly (Group 2B)
Carcinogenic to Humans

Summary of Evidence on Carcinogenicity


Monograph
Drug Group Human Datac Animal Datac Volumes Year

α-​Blockers, Cardiac Glycosides, and Diuretics


Digoxin 2B L I 108 2015
Hydrochlorothiazide 2B L L 50, 108 2015
Phenoxybenzamine hydrochloride 2B ND S 24, Suppl 7b 1987
Triamterene 2B I S 108 2015
Analgesic
Phenazopyridine hydrochloride 2B I S 24, Suppl 7b 1987
Antibiotic and Antifungal Agents
Chloramphenicol 2A L I 50 1990
Griseofulvin 2B I S 79 2001
Metronidazole 2B I S 13, Suppl 7b 1987
Anticonvulsants
Phenytoin 2B I S 66 1996
Primidone 2B I S 108 2015
Antidiabetic
Pioglitazone 2A L S 108 2015
Anti-​HIV
Zalcitabine 2B I S 76 2000
Zidovudine 2B I S 76 2000
Anti-​inflammatory
Sulfasalazine 2B I S 108 2015
Anti-​neoplastic Agents
Adriamycin 2A I S 10, Suppl 7b 1987
Azacitidine 2A ND S 50 1990
Aziridine 2B ND L 9, Suppl 7b, 71 1999
Bleomycin 2B I L 26, Suppl 7b 1987
Chlorozotocin 2A ND S 50 1990
Cisplatin 2A I S Suppl 7b 1987
Dacarbazine 2B I S 26, Suppl 7b 1987
Daunomycin 2B ND S 10, Suppl 7b 1987
Lomustine 2A I S Suppl 7b 1987
Merphalan 2B ND S 9, Suppl 7b 1987
Mitomycin C 2B ND S 10, Suppl 7b 1987
Procarbazine hydrochloride 2A I S 26, Suppl 7b 1987
Streptozotocin 2B ND S 17, Suppl 7b 1987
Teniposide 2A L I 76 2000
Anxiolytics and Antidepressants
Mitoxantrone 2B L I 76 2000
Oxazepam 2B I S 66 1996
Sedatives and Hypnotics
Chloral 2A I S 63, 106 2014
Chloral hydrate 2A I S 84, 106 2014
Phenobarbital 2B I S 79 2001
Thyroid Medications
Methylthiouracil 2B I S 79 2001
Propylthiouracil 2B I S 79 2001
Thiouracil 2B I S 79 2001
Miscellaneous
Pentosan polysulfate sodium 2B I S 108 2015

a
http://​monographs.iarc.fr/​ENG/​Classification/​
b
IARC Monographs Supplement No 7. Overall Evaluations of Carcinogenicity: An Updating of IARC Monographs Volumes 1 to 42.
c
I = inadequate, L = limited, S = sufficient, ND = no data

Confounding An example is the use of diabetes drugs in relation to breast cancer


risk, since diabetes is associated with an elevated risk of breast cancer.
One of the most important potential confounders in studies of drug use Methods to address this type of confounding include restricting the
is the indication (or contraindication) for the treatment. Confounding population to patients with the condition being treated (e.g., restrict
by indication can occur when the condition treated by the medication studies of diabetes drugs to patients with diabetes), although this does
is also a risk factor for the outcome of interest (Signorello et al., 2002). not preclude confounding by disease severity when this is associated
 415

Pharmaceutical Drugs Other Than Hormones 415


with the outcome (Salas et al., 1999). An optimal strategy, when pos- both the exposure and the outcome (Hammer et al., 2009). Efforts to
sible, is to compare risk among users of the drug of interest to risk maximize response rates or to obtain study data without patient contact
among users of another drug prescribed for the same indication (i.e., (and with waived consent) reduce this bias. Studies using large health
an active comparator drug) and, ideally, with the same severity of dis- databases may limit selection bias, provided they are highly represen-
ease (Yoshida et al., 2015). Many patients, especially the elderly, often tative of the target population (Tanaka et al., 2015). In cohort studies,
use more than one type of drug. For example, patients with diabe- selection bias may be present if losses to follow-​up are differential
tes are often treated with multiple diabetes drugs (sequentially and/​or with respect to both drug exposure and cancer risk.
concurrently) as their disease progresses. Similar combination therapy In addition, selection bias may occur when most users of a drug
is also seen in patients with hypertension or depression. Such poly- are prevalent users. Prevalent users are not a random sample of all
pharmacy adds substantial complexity to studies of drug efficacy and users, and they may under-​represent those at early risk of the disease
safety, as the additional drugs may be time-​varying confounders if they (Ray, 2003). Although risk of cancer is generally thought to increase
are also associated with the outcome of interest. Confounding can also with longer exposure and after a minimum latency (i.e., under-​
occur when patients using a particular drug differ from the general representation of those at early risk is usually not an issue), restrict-
population (of healthy individuals or patients) on overall health sta- ing studies to new users allows for complete assessment of drug use
tus, life-​style factors, or drug adherence, introducing “healthy user” or and for adjustment of covariates prior to drug exposure, thus avoiding
“healthy adherer” effects (Shrank et al., 2011). potential selection bias due to depletion of susceptibles and avoiding
There is typically greater potential for confounding in studies of adjusting for a factor that may have been affected by the medication
drug use and cancer outcomes than in studies of cancer risk. While (i.e., on the causal pathway). However, restricting the cohort to first
most risk factors are only modestly associated with cancer risk, dis- initiators of a drug can substantially limit sample size, and thus the
ease characteristics (e.g., stage, lymph node involvement, tumor size) statistical precision of the study. It can also limit follow-​up time and
are generally strong risk factors for both recurrence and progression. the ability to examine long-​term drug use, which are important con-
Thus, disease characteristics can be important confounders if they are siderations for cancer outcomes. For example, in the first report of the
also associated with use of the drug of interest. Likewise, in studies Women’s Health Initiative (Rossouw et al., 2002), based on an average
of cancer mortality, disease recurrence and progression can be strong of 5 years of follow-​up, risk of breast cancer associated with estrogen
confounders if they have an impact on drug use (Chubak et al., 2013). plus progestin (vs. placebo) was not elevated among women who had
Standard methods can be used to improve comparability between never used postmenopausal hormone therapy (PMT) prior to baseline
exposed and unexposed groups (Jager et  al., 2008), including restric- (i.e., new users). However, risk was increased over 2-​fold for women
tion, matching, stratification, and multiple regression techniques. with up to 5 years of prior PMT use, and over 4.5-​fold for women with
Among more recent methods is propensity score (PS) analysis, which 5–​10 years of prior use (i.e., prevalent users).
uses information on a large number of covariates to predict the prob-
ability of use of the specific drug(s) of interest and consequently to help
balance observed and unobserved characteristics between treatment Information Bias
groups (Austin, 2011). Analogous to the PS approach, the disease risk Information bias typically results from error in the measurement of an
score (DRS) calculates a summary measure as a function of confound- exposure, covariate, or outcome and can either be differential (system-
ers, but it is based on the probability of disease, rather than the exposure atic) or non-​differential (random) between comparison groups (Tanaka
(Arbogast and Ray, 2011). The DRS is particularly appealing in analyses et  al., 2015). Non-​differential misclassification will usually bias the
of multiple drugs or multiple levels of drug exposure in relation to can- exposure–​ outcome association toward the null, whereas differential
cer risk, in which the PS approach is unwieldy. Confounding can also be misclassification can lead to bias in either direction (Jepsen et al., 2004).
addressed by sensitivity analyses incorporating external data to estimate
associations adjusted for unmeasured confounders (Schneeweiss, 2006). Outcome Misclassification
Marginal structural modeling can be used when the confounder is also Cancer registries are considered the gold standard for information on
an intermediate variable in the causal pathway between the exposure cancer diagnoses, although the time period covered, data collected,
(drug) and the outcome (Robins et al., 2000). While this is common in and quality control procedures vary by registry program (Dreyer and
studies of drug efficacy, it can also be an issue in studies of unintended Velentgas, 2012). In addition to histologic subtypes, many registries
drug effects, such as drug safety or chemoprevention. Marginal struc- are increasingly recording information on clinical cancer stage and
tural models can also be used to control for confounding by recurrence tumor markers (e.g., estrogen receptor [ER] status), which allow for
in studies of drug use and cancer mortality (Chubak et al., 2013). improved examination of the specificity of drug–​cancer associations
Instrumental variable (IV) analysis is a method that can potentially and thus better evaluation of biologic plausibility. In some survey-​
be used to mitigate unmeasured confounding. A  good IV is a vari- based observational studies, linkages to cancer registries are not fea-
able that is strongly associated with the exposure of interest, is not sible, and the information on cancer outcome must be obtained from
associated with other risk factors (including other drugs) for the out- self-​reports, ideally with validation by review of available medical
come, and is only associated with the outcome through its association records. If conducted within healthcare databases not linkable to can-
with the exposure. However, examples of well-​suited IVs are few, and cer registries, the information can be obtained by diagnosis codes. The
the underlying assumptions are easily violated (Martens et al., 2006). accuracy of self-​report and diagnosis codes has been found to vary by
Mendelian randomization (MR) is a variation of IV analysis that is cancer type and stage (Bergmann et al., 1998). Algorithms for health-
being used increasingly in epidemiology. The principle in the MR care databases with varying sensitivity, specificity, and predictive
approach is that a genetic variant (or combination of variants) is used value have been developed to identify new cancer diagnoses or cancer
as an IV to estimate a causal effect between a potential risk factor and recurrences (Chubak et al., 2012).
an outcome of interest (Verduijn et al., 2010). For example, MR has Misclassification can also occur because cancer is a long-​term out-
been used to examine the association between body mass index (BMI) come, and there is always a period of time before an existing cancer is
and cancer risk using the FTO gene as an IV (Brennan et al., 2009). detected. Detection bias can occur if individuals receiving the drug of
However, the MR method is limited by the need for very large sample interest are more or less likely to be seen by a healthcare provider or to
sizes, as well as the difficulty in identifying genetic variants that meet undergo cancer screening prior to clinical attention and cancer diagno-
the other IV criteria (Bochud and Rousson, 2010). sis. This can also occur in studies of drugs use and cancer recurrence,
if the users of the drug of interest are more or less likely to have routine
medical visits or to undergo disease surveillance. Detection bias can
Selection Bias be addressed using standard methods (e.g., restriction, stratification,
Selection bias occurs when factors involved in the selection of the multivariable adjustment), although appropriate adjustment for cancer
study population, or that influence study participation, are related to screening can present challenges (Weiss, 2003).
416

416 Part III: The Causes of Cancer


When use of a drug is influenced by an early sign or symptom of time when the exposure of interest is measured. For example, in stud-
undiagnosed cancer, such reverse causation, also called protopathic ies using electronic health records, a drug will look protective if cases
bias, can lead to spurious drug–​cancer associations (Faillie, 2015). For have health records of drug use for a shorter time than controls. This
example, some undiagnosed cancers may cause pain, which can result bias can be avoided by matching cases and controls on, or adjusting
in the use of analgesics. Other undiagnosed cancers may compromise for, follow-​up time. In studies comparing drugs prescribed for the
the immune system and lead to the use of antibiotics or other anti-​ same condition, a first-​line drug may appear protective when the com-
infectives. Pancreatic cancer can interfere with insulin production, and parison drug is second-​or third-​line therapy if longer disease dura-
anti-​diabetic therapies are sometimes initiated prior to cancer diagno- tion is associated with increased risk of the outcome. Such bias can be
sis. A common approach to minimize this bias is to disregard drug use avoided by comparing therapies given for the same stage of disease, or
within a period (e.g., 1 year) prior to cancer diagnosis or to conduct by matching on, or adjusting for, duration or stage of disease.
sensitivity analyses using different intervals, since the period when the
cancer may cause signs or symptoms but remains undetected is usually
unknown. PHARMACOEPIDEMIOLOGY STUDIES OF DRUG–​
CANCER ASSOCIATIONS
Exposure Misclassification
Drugs can be taken for a single acute condition (e.g., antibiotic for an As noted earlier, only about 20 non-hormonal medications have been
infection), sporadically or intermittently over a lifetime (e.g., aspirin classified as human carcinogens by the IARC (Table 23–​1) (IARC,
for headache or muscle pain), or chronically for years (e.g., statin for 2012). The majority of these agents derive from two therapeutic cat-
high cholesterol), which emphasizes the need for thoughtful data col- egories: anti-​neoplastic and immunosuppressant therapy. About twice
lection and exposure definitions in accordance with existing knowl- as many drugs have been classified as probably or possibly carcino-
edge or hypotheses about the influence of the timing, strength, and genic to humans (Table 23–​2) (Friis et al., 2015a).
duration of drug use on the cancer outcome(s) of interest. Over the last decade, an increasing number of pharmacoepidemi-
Self-​reported information can result in either non-​differential or dif- ology studies have explored hypotheses on the use of specific drugs
ferential (e.g., recall bias) misclassification of medication histories. and cancer risk or prevention. Further, a growing number of studies
Drug names can be unfamiliar and difficult to remember, especially if have examined whether medications used to treat a variety of common
individuals take a number of different medications or there is substan- conditions may be related to an increased or decreased risk of cancer
tial switching or stopping of drug use. Accuracy of self-​report can be recurrence or mortality.
improved by providing study participants with lists of drug names, pic- In the following, we summarize briefly the literature on non-​
tures of tablets, and indications. Use of life calendars with important hormonal drugs known or suspected of increasing or decreasing can-
events such as marriage, births, and new jobs can improve the accuracy cer risk or progression; focusing on those drugs that have received
of the timing of drug use. the most attention during the last 10 years. Hormonal agents, such as
Electronic pharmacy records are usually considered the gold stan- estrogens, are addressed in Chapter 22.
dard in observational pharmacoepidemiologic research, since they are
often required for patient management and/​or billing, and frequently
include strength and amount dispensed. However, many healthcare
Anti-​neoplastic Agents
databases do not include drugs given during hospitalizations and most
do not record non-​prescription drugs. While some pharmacy databases Anti-​neoplastic drugs are agents used primarily to inhibit or prevent the
record the actual dispensing of the drug prescription, outpatient phar- growth and spread of cancer cells. These drugs include cytotoxic agents
macy databases will generally not have information on whether or not and hormonal therapies, as well as the more recently developed tar-
filled prescription drugs were actually consumed by the patients. This geted or non-​specific immunotherapies. Whether used as single agents
misclassification can be reduced by defining exposure as filling more or in combination therapy, these medications have yielded substantial
than one prescription during a certain time period (e.g., 6 months). In improvements in survival, attributable to an enhanced understanding
addition, healthcare databases do not have prescription information for of the mechanisms of anti-​tumor activity and micrometastases (Chu
the period prior to enrollment in the healthcare or insurance plan or and Sartorelli, 2012), and the recognition of tumor heterogeneity and
prior to the establishment of the pharmacy database. molecular subtypes (Hoelder et al., 2012). Unfortunately, most cyto-
In general, the etiologically relevant exposure period for a specific toxic agents also cause damage to healthy cells, especially in rapidly
drug–​cancer association is unknown, and may also vary by specific proliferating tissues, such as hematopoietic tissue and gastrointestinal
agent, dose, or patient characteristics (Gottlieb and Altman, 2014). epithelia, which may induce secondary malignancies. The exact carci-
Assessment of drug use outside the etiologically relevant exposure nogenetic pathways of secondary neoplasms following chemotherapy
period may attenuate measures of association and reduce the likeli- have not been fully elucidated (Travis et al., 2013), despite the long-​
hood of observing a true association. In order to identify the critical known mutagenic and carcinogenic effects of many anti-​neoplastic
exposure period, information on drug use should ideally be collected drugs (Dedrick and Morrison, 1992). A  mutual mechanism is DNA
according to different aspects of time (e.g., age of first use, time of first damage (genotoxicity), leading to either clonal loss of chromosomes
use, time of last use, duration of use), allowing for the examination of or gene translocations (Sill et al., 2011).
multiple exposure windows. Cytotoxic agents can be grouped into distinct categories based on
Drug use is a time-​varying exposure, and there are several time-​ their pharmacodynamic mechanisms and include alkylating agents,
related biases to consider in pharmacoepidemiology studies that can antimetabolites, cytotoxic antibiotics, plant alkaloids, and plati-
greatly distort the measures of association (Suissa and Azoulay, 2012). num compounds (WHO, 2015). At least 11 of these drugs have been
Immortal time bias occurs in cohort studies when unexposed time is classified as carcinogenic to humans (Grosse et  al., 2009; IARC,
misclassified as exposed time. For example, if an individual is not 2012) (Table 23–​1). Of these, the majority are alkylating drugs, includ-
using a drug at cohort entry and later initiates drug use but the entire ing busulfan, chlorambucil, cyclophosphamide, melphalan, semustine
follow-​up time is classified as exposed, the time until start of drug use (methyl-​CCNU), thiotepa, and treosulfan, which all exhibit genotoxic
is “immortal” since the individual obviously must survive the unex- activity, commonly through alkylation of purine bases in DNA (Sill
posed time in order to become exposed. When this “immortal” time is et al., 2011). Typically, alkylating drugs induce acute myeloid leukemia
incorrectly attributed to exposed time of the drug of interest, the rela- (Table 23–​1) (IARC, 2012), but these agents have also been related to
tive risk estimates will be reduced and the drug may appear more ben- the development of some solid tumors, notably lung cancer (Swerdlow
eficial than it is. This bias can be avoided by the use of time-​varying et al., 2011), bladder cancer (Travis et al., 1995) and myelodysplastic/​
exposure variables or by starting follow-​up at or after drug initiation. myeloproliferative neoplasms, particularly in patients with a prior his-
Time-​window bias occurs in case-​control studies when cases and con- tory of Hodgkin lymphoma (Sill et al., 2011). In addition, a number of
trols have different opportunities for exposure or different periods of other cytotoxic agents have been classified as probably carcinogenic to
 417

Pharmaceutical Drugs Other Than Hormones 417


humans (group 2A carcinogens), including azacitidine, cisplatin, pro- griseofulvin exhibits antimitotic activity, inhibits proliferation,
carbazine, and teniposide (IARC, 2012) (Table 23–​2). and activates apoptosis (Mauro et  al., 2013; Panda et  al., 2005;
When examining the risk of secondary cancer following chemother- Rathinasamy et al., 2010).
apy, a particular difficulty is that these malignancies typically have a Metronidazole is an antiprotozoal and antibacterial drug classified
very long latency and thus are inadequately captured in most observa- as a group 2B carcinogen (Table 23–​2). A number of epidemiologic
tional studies and clinical trials. However, a few observational studies studies conducted prior to 2005 examined metronidazole use and risk
have followed patients over several decades, corroborating a substan- of cancer overall or at specific sites and reported mainly null find-
tial excess risk of secondary malignancies following chemotherapy ings (see previous edition of this textbook). More recently, metro-
(Olsen et al., 2009). Another challenge is to isolate the carcinogenic nidazole has been shown to induce growth of breast and colorectal
role of one particular antineoplastic agent, notably when evaluating cancer cell lines (Sadowska et  al., 2013a, 2013b). In a large cohort
combination therapies or when patients also undergo other therapy study of antibiotics and risk of breast cancer, Friedman et al. (2006)
with carcinogenic potential (e.g., radiotherapy). found no increased risk with use of metronidazole equivalent to at least
Several traditional anti-​neoplastic drugs have been supplemented or 100 days of treatment.
superseded by newer molecularly targeted drugs (Hoelder et al., 2012), The antiviral drugs zalcitabine and zidovudine have been catego-
and research continues to seek ways to reduce dosage or develop safer, rized as group 2B carcinogens (Table 23–​2). Recently, however, zido-
more efficient anti-​neoplastic agents (Travis et al., 2013). Obviously, vudine has been suggested to possess chemopreventive properties by
the carcinogenic potential of these new drugs, and combinations inducing apoptosis and delaying cell cycle progression (Fang and
thereof, should be monitored and evaluated closely (Boffetta and Beland, 2009), although clinical trials have shown limited evidence of
Islami, 2013; IARC, 2012). The anti-​neoplastic mechanisms of new anti-​cancer effects (Chow et al., 2009).
drugs are often, but not always (e.g., immunotherapeutic agents), simi-
lar to those of the traditional drugs they are replacing. Focusing on
relevant mechanisms (i.e., mechanistic analyses) may therefore con- Antihypertensive Drugs and Statins
tribute to improved identification of individuals susceptible to drug-​ Among drugs used to treat disorders of the cardiovascular system,
induced cancers (Boffetta and Islami, 2013; IARC, 2012). antihypertensives and statins have received the most attention in rela-
tion to cancer endpoints (Singh and Bangalore, 2012). Considering
the common use and proven tolerability of these agents, discovery of
Anti-​infective Agents any carcinogenic or chemopreventive effects would be of profound
Anti-​infective agents are a broad category of drugs that include anti- clinical and public health importance. Given that these agents are
bacterials, antifungals, antiprotozoals, and antivirals. They include used to treat both hypertension and heart diseases, which are asso-
some of the most widely prescribed medications in both children and ciated with obesity, cardiometabolic abnormalities, and lifestyle fac-
adults (Figures 23–​1 and 23–​2). In adults, assessment of lifetime use tors, such as tobacco use, which may increase cancer incidence and
of anti-​infective agents is challenging, and most studies focus on a mortality, potential confounding is of concern. In addition, studies of
relatively limited exposure period prior to cancer diagnosis. A small cardiovascular drugs should also account for the use of concomitant
number of anti-​infective drugs have been classified by IARC as prob- medications associated with cancer risk, notably aspirin. Finally, the
ably (group 2A) or possibly carcinogenic to humans (group 2B) presence of other diseases (e.g., colitis ulcerosa and other inflamma-
(Table 23–​2) (Friis et al., 2015a). tory conditions, and diabetes mellitus) associated with risk of both car-
Chloramphenicol, an antibiotic classified as group 2A, has been diovascular disease and cancer should also be considered as potential
shown to induce abnormal cell differentiation and inhibit apoptosis confounders.
in activated T-​cell models, which may contribute to leukemogenesis
(Yuan and Shi, 2008). To our knowledge, however, there are no new Angiotensin Converting Enzyme Inhibitors and
human data addressing specifically the probable carcinogenicity of Angiotensin-​II Receptor Blockers
chloramphenicol use. Recent epidemiological studies of the associa- Some studies suggest that the renin-​angiotensin-​aldosterone system
tion between use of antibiotics and cancer risk have mainly focused on may play a role in cancer development (Singh and Bangalore, 2012).
breast and prostate cancer. A meta-​analysis of five case-​control stud- Angiotensin converting enzyme inhibitors (ACEIs) block the con-
ies reported a slightly increased risk for breast cancer associated with version of angiotensin I to angiotensin II, a hormone that stimulates
ever use of any antibiotics and a borderline dose–​response relation- neovascularization and promotes angiogenesis and thus may stimu-
ship based on the number of prescriptions (Sergentanis et al., 2010). late cancer development (Yoshiji et  al., 2001). Consequently, ACEI
However, residual confounding was likely, and the risk pattern for use may reduce cancer risk. A number of observational studies have
individual categories of antibiotics could not be investigated. Some found a reduced incidence of overall cancer (Chiang et al., 2014) or
large cohort studies have also reported an association between use of of cancer at specific sites, including the colorectum (Dai et al., 2015),
various types of antibiotics and increased breast cancer risk (Friedman esophagus (Sjøberg et al., 2007) and breast (Li et al., 2013). However,
et al., 2006; Kilkkinen et al., 2008). Other studies have examined the a meta-​analysis of observational studies (Yoon et al., 2011) and a sec-
association between antibiotic use and prostate cancer risk, and find- ondary analysis of RCTs (Bangalore et al., 2011) reported no associa-
ings have been inconsistent, albeit mainly null (Daniels et al., 2009; tion between ACEI use and risk of overall cancer.
Tamim et  al., 2010). Most studies may have missed associations if Angiotensin-​II receptor blockers (ARBs) share many properties of
antibiotic use requires long-​term chronic treatment to influence cancer ACEIs and are used for similar indications, including hypertension and
risk. However, a nationwide cohort study of 8445 patients with endo- heart failure (Brayfield, 2014). ARBs selectively block the angiotensin
carditis, who are typically on long-​term therapy with antibiotics, found II type 1 receptor, which may explain the anti-​angiogenic and anti-​
only weak associations for several cancers, including cancers of the proliferative effects observed in animal models of sarcoma, pancre-
prostate, stomach, and breast, which might be explained by non-​causal atic, and breast cancer (George et al., 2010). This blockage, however,
factors (Thomsen et al., 2013a). induces overstimulation of angiotensin II type 2 receptors that may
Griseofulvin is an antifungal drug classified as a group 2B car- result in cellular proliferation, angiogenesis, and tumor progression
cinogen (Table 23–​2). A large nested case-​control study reported an (Walther et al., 2003), which has led to concerns about the potential
increased risk of breast cancer with griseofulvin use, with a sugges- carcinogenicity of these agents.
tive dose–​response relationship (Friedman et al., 2009b). However, Concern about the carcinogenic potential of ARBs was raised
the study could not adjust for risk factors other than age and a after a secondary analysis of five RCTs of ARBs and cardiovascu-
crude measure of hormone use. More recently, there has been inter- lar disease (Sipahi et al., 2010) reported a slightly increased cancer
est in the role of adjuvant griseofulvin in the treatment of patients risk among ARB users. This prompted the FDA (2011) and EMA
with cancer, with in vitro cancer cell line studies suggesting that (2011) to conduct safety reviews of the issue. However, two large
418

418 Part III: The Causes of Cancer


meta-​analyses (ARB Trialists Collaboration, 2011; Bangalore et al., not allow examination of long-​term use (Numbere et al., 2015). Other
2011)  showed no elevated cancer risk with ARBs. In the largest observational studies have reported an inverse association between
meta-​analysis (Bangalore et al., 2011), the main limitation was that ß-​blocker use and cancer risk. In a large nested case-​control study,
follow-​up averaged just 3.5 years (range, 1–​9 years) for all 70 trials Assimes et al. (2008) reported an approximate 10% reduction in over-
combined. all cancer risk among ever users of ß-​blockers compared to ever users
Relatively little data are available on the potential association of of thiazide diuretics. This finding was driven primarily by a reduced
ARBs or ACEIs with cancer recurrence or survival, and the few avail- risk of colorectal cancer and to a lesser degree by risk reductions in
able studies have provided conflicting results. A cohort study of breast other gastrointestinal cancers, head and neck cancer, and hematologi-
cancer patients found an increased recurrence risk associated with cal cancer. Most recently, a meta-​analysis of RCTs suggested that use
post-​diagnosis use of ACEIs, although there was no evidence of a of non-​selective ß-​blockers may reduce the risk of hepatocellular car-
dose–​response relationship (Ganz et al., 2011). In contrast, a review of cinoma in patients with liver cirrhosis (Thiele et  al., 2015). A  large
10 other studies reported that ACEI use was associated with improved case control study in the UK CPRD (Hagberg et al., 2016), however,
outcomes in patients with breast or other cancers (McMenamin et al., found no association between ß-​blocker use and primary liver cancer.
2012). A more recent observational study found little evidence for an Designing robust observational studies to examine liver cancer is chal-
association between the use of ARBs and cancer-​specific mortality lenging due to the considerable potential for unmeasured confounding,
among patients with breast, colorectal, or prostate cancer (Cardwell especially when using healthcare databases that do not routinely col-
et al., 2014a). lect information on many important risk factors for liver cancer.
Several recent studies have examined a possible protective effect of
Calcium Channel Blockers ß-​blockers on the progression of cancer. Some observational studies
Calcium channel blockers (CCBs) decrease intracellular calcium, have reported substantial reductions in risk of metastasis and cancer-​
which has been suggested to increase cancer risk by inhibiting apop- specific death with the use of ß-​blockers, particularly propranolol or
tosis (Wu et al., 2014). An early observational study by Pahor et al. other non-​selective agents, among patients with cancers of the breast
(1996), which first raised concerns about CCB use and cancer risk, (Barron et al., 2011), prostate (Grytli et al., 2014), lung (Wang et al.,
reported an increased risk of fatal cancers in older patients using 2013), or ovary (Watkins et al., 2015), and with malignant melanoma
nifedipine, a CCB. However, this study was prone to serious meth- (Lemeshow et al., 2011). However, other studies have not consistently
odologic shortcomings, and other observational studies (Assimes supported these findings. Post-​diagnosis ß-​blocker use was associated
et al., 2008; Boudreau et al., 2008; Devore et al., 2015; Grimaldi-​ with a modestly reduced risk of breast cancer recurrence (without a
Bensouda et al., 2016) reported no association between CCB use and dose–​response relationship) in one study (Ganz et  al., 2011), but a
cancer risk overall or at specific sites, even among long-​term (>7.5 marginally increased risk of recurrence in another (Boudreau et  al.,
years) users. Particular emphasis has been placed on the association 2014). In addition, studies conducted in the UK CPRD found little
between CCB use and breast cancer risk. A recent meta-​analysis of evidence of an association between post-​diagnostic ß-​blocker use and
17 observational studies indicated a slightly increased risk of breast outcomes for breast (Cardwell et al., 2013), prostate (Assayag et al.,
cancer associated with long-​term (>10 years) use of CCBs (Li W 2014; Cardwell et al., 2014b), melanoma, or colorectal cancer (Hicks
et al., 2014), but other meta-​analyses (Chen et al., 2014; Saltzman et al., 2013; McCourt et al., 2014).
et al., 2013) and large observational studies (Assimes et al., 2008;
Devore et  al., 2015) found no association. In addition to observa- Diuretics
tional studies, a number of large RCTs, such as the ALLHAT trial An association between the use of diuretics and cancer risk was first
(ALLHAT Officers and Coordinators for the ALLHAT Collaborative proposed in 1988 (McLaughlin et  al., 1988). Indeed, the diuretic
Research Group, 2002) and large meta-​analyses of trial participants hydrochlorothiazide is a cyclic imide that can be converted in the
(Bangalore et  al., 2011), have found no association between CCB stomach to a mutagenic nitroso derivative (Gold and Mirvish, 1977).
use and incidence or mortality of cancer overall; however, these A meta-​analysis of antihypertensive therapy found an increased risk
studies may have been limited by short follow-​up periods. of renal cell cancer (RCC) in female, but not male, users of diuretics
A recent Canadian cohort study reported improved survival among (Corrao et al., 2007). Large across-​study heterogeneity, however, may
breast and lung cancer patients using CCBs (Holmes et  al., 2013). have compromised the pooling of results. Moreover, it is important
However, studies of CCB use and cancer prognosis have mostly pro- to acknowledge that hypertension per se may be associated with an
vided null results. Within the COmmonly used Medications and Breast increased risk of RCC and thus that confounding by indication also
Cancer Outcomes (COMBO) cohort, specifically designed to study remains a possibility. The ALLHAT trial comparison of diuretic use
the influence of drugs and comorbidities on breast cancer prognosis, with use of other antihypertensives provided no evidence that diuret-
Boudreau et al. (2014) observed no association between CCB use and ics increased cancer risk (ALLHAT Officers and Coordinators for the
breast cancer recurrence or risk of contralateral breast cancer. Chen ALLHAT Collaborative Research Group., 2002). More recently, two
et al. (2015) reported similar findings in regard to CCB use and contra- meta-​analyses of RCTs have corroborated a null association between
lateral breast cancer among women diagnosed with estrogen receptor diuretic use and overall cancer risk (Bangalore et al., 2011). Null asso-
(ER)-​positive invasive breast cancer. ciations have also been reported in observational studies of specific
cancer types, for example, kidney (Fryzek et al., 2005) and colorectal
ß-​blockers cancer (Boudreau et al., 2008). However, diuretic use has been associ-
Animal studies have shown that ß-​adrenergic signaling regulates mul- ated with an increased risk of basal cell carcinoma (particularly among
tiple cellular processes involved in carcinogenesis, particularly in overweight or obese individuals) (McDonald et al., 2014) and squa-
pancreatic and breast cancer, and ß-​blockers have also been shown mous cell carcinoma (Schmidt et al., 2015), the latter driven by results
to disrupt migratory activity and inhibit angiogenesis of cancer cells for potassium-​sparing agents alone or in combination with low-​ceiling
(Pasquier et  al., 2011). Concern about the carcinogenicity of ß-​ diuretics. Hydrochlorathiazide, a photosensitizing low ceiling diuretic,
blockers emerged when a clinical trial showed an increase in cancer-​ has also been associated with lip cancer (Friedman et al., 2012).
related deaths among men assigned to use of the ß-​blocker atenolol,
compared to men assigned to a diuretic or placebo (MRC Working Statins
Party, 1992). A  subsequent large meta-​analysis of clinical trial data Hydroxy-​3-​methylglutaryl coenzyme A (HMG-​CoA) reductase inhib-
found no association between ß-​blocker use and cancer risk or cancer-​ itors (statins) are among the most frequently prescribed drugs in the
related mortality (Bangalore et  al., 2011). Similarly, a large case-​ world, and therapy is generally “lifelong.” Statins inhibit the rate-​
control study nested in the UK Clinical Practice Research Datalink limiting step of cholesterol biosynthesis and are indicated for lowering
(CPRD) reported no association between ß-​blocker use and the risk of serum cholesterol and prevention of coronary artery and vascular dis-
common cancers (lung, colorectal, breast, and prostate); however, the ease (Maron et al., 2000). In addition to reducing cholesterol produc-
exposure period prior to cancer diagnosis was relatively short and did tion, statins target lipid metabolism, prevent inflammation, and induce
 419

Pharmaceutical Drugs Other Than Hormones 419


apoptosis, thereby modulating several cellular processes, including There is increasing interest in the potential influence of statin use on
those essential to carcinogenesis (Altwairgi, 2015; Matusewicz et al., cancer prognosis and mortality. A meta-​analysis of RCTs did not show
2015). Statins also impair prenylation, an essential process in the post-​ reductions in overall cancer mortality with statin use, but several of the
translational modification of many proteins, including the Ras protein trials were limited by short follow-​up and insufficient power to detect
family, which are involved in tumorigenesis and cancer progression. a significant difference in cancer outcomes (Cholesterol Treatment
In addition, statins target mevalonate synthesis, a precursor to several Trialists Collaboration, 2012). In contrast, a recent systematic review
major cellular products that are important in cell growth, signal trans- and meta-​analysis of 39 cohort studies and two case-​control studies
duction, differentiation, and survival (Altwairgi, 2015). showed about a 20% decrease in risk for both all-​cause and cancer-​
Some epidemiologic studies have reported an association between specific death, with both post-​diagnostic and pre-​diagnostic statin use
low cholesterol and increased cancer incidence (Liao and Farmer, appearing beneficial (Zhong et al., 2015b). Most observational studies
2013), although the underlying causality of the relationship is unclear. of statin use and cancer mortality have focused on breast (Mansourian
If lowering cholesterol induces cancer, statin use should be associ- et  al., 2016; Zhong et  al., 2015b) or prostate cancer (Raval et  al.,
ated with increased cancer risk. However, if undiagnosed cancer low- 2016). Studies of statin use and breast cancer recurrence or mortality
ers serum cholesterol for an extended period prior to a clinical cancer have quite consistently found a reduced risk of recurrence or cause-​
diagnosis, statin use could appear protective since there would be specific death associated with statin use (Mansourian et al., 2016). The
less use among cases. Both possibilities should be considered when largest study, a Danish nationwide study including over 18,000 Danish
designing studies and interpreting results of studies of statin use and women diagnosed with stage I–​III breast cancer, reported a substantial
cancer risk. reduction in risk of breast cancer recurrence associated with statin use
Two large RCTs reported excess incidences of breast and gas- during a median follow-​up period of 6.8 years (Ahern et al., 2011). The
trointestinal cancer, respectively, among patients assigned to statin results for prostate cancer are more heterogeneous, with some studies
therapy (Sacks et  al., 1996; Shepherd, 2002). However, the larg- suggesting null associations, and others a 20%–​40% reduction in risk
est randomized statin trial, the Heart Protection Study, found no of prostate cancer recurrence, or prostate cancer-​specific or all-​cause
increased risk of cancer or cancer mortality after a mean of 11 years mortality (Raval et al., 2016; Yu et al., 2014). The most recent meta-​
of follow-​up (Heart Protection Study Collaborative Group, 2011). analysis of 34 observational studies (Raval et  al., 2016)  indicated a
One randomized trial reported a significant increase in cancer risk clear beneficial effect of both pre-​and post-​diagnostic use of statins on
among patients assigned to a combination of simvastatin and ezeti- prostate cancer mortality, as well as a reduction in biochemical recur-
mibe. This raised concern that the additional reduction in LDL cho- rence among patients treated with radiotherapy. However, this meta-​
lesterol induced by the combination therapy might increase cancer analysis and an earlier one that included eight cohort studies (Scosyrev
risk (Rossebo et al., 2008). However, findings from subsequent stud- et al., 2013) failed to find a beneficial effect of statin use on biochemi-
ies, including the Heart Protection Study (Heart Protection Study cal recurrence among patients treated with radical prostatectomy.
Collaborative et  al., 2011), did not support this hypothesis (Peto
et al., 2008).
Numerous observational studies have examined associations Aspirin and Other Nonsteroidal
between statin use and risk of cancer. Overall, the studies suggest a null
Anti-​inflammatory Drugs
association between statin use and risk of smoking-​related cancers,
including cancers of the lung (Hippisley-​Cox and Coupland, 2010), There is convincing evidence that use of aspirin and other non-​steroidal
bladder (Zhang XL et al., 2013), kidney (Zhang et al., 2014), and pan- anti-​inflammatory drugs (NSAIDs) reduce the risk of colorectal cancer
creas (Cui et al., 2012; Simon et al., 2016). Meta-​analyses of breast and and possibly a number of other cancers (see Chapter 62.5). While the
skin cancers have also reported null associations (Undela et al., 2012). precise mechanisms by which these drugs may prevent cancer have
In contrast, a number of observational studies and meta-​analyses have not been elucidated, the inhibition of cyclooxygenase (COX) enzymes
found that statin use is associated with slight to modest reductions in and prostaglandin activity appears to be important (Wang and Dubois,
risk of gastric (Ma et al., 2014), colorectal (Lytras et al., 2014), pros- 2006). Prostaglandins are involved in a number of mechanisms that
tate (notably advanced) (Bansal et  al., 2012; Tan et  al., 2016), liver may promote tumor growth and progression, such as cell proliferation,
(Singh et  al., 2013a), hematologic malignancies (Yi et  al., 2014), promotion of angiogenesis, suppression of immune responses, inhibi-
and esophageal cancer (particularly in patients with Barrett’s esopha- tion of apoptosis, and stimulation of invasion and motility (Wang and
gus) (Singh et al., 2013b). Dubois, 2004, 2006). Aspirin and non-​aspirin NSAIDs may affect car-
Reasons underlying the apparent discrepancy between the results cinogenesis by inhibiting prostaglandin endoperoxide synthase-​2 or
of numerous laboratory studies, demonstrating promising chemopre- type 2 of the COX enzyme (COX-​2), the rate-​limiting enzyme in the
ventive effects of statins (Altwairgi, 2015), and those of clinical and prostaglandin pathway (Usman et al., 2015; Wang and Dubois, 2004).
epidemiological studies are unclear. A possible explanation is differ- There is also increasing evidence that the antiplatelet activity of low-​
ences between dosage levels of statins applied in the laboratory setting dose aspirin may have a distinct role in reducing cancer risk, as well
and the recommended doses of statins used in patient treatment (Friis as risk of metastasis, through inhibition of tumor angiogenesis (Thun
and Olsen, 2006). Another explanation might be the extensive hepatic et al., 2012; Usman et al., 2015).
“first-​pass metabolism” of most statins, resulting in low concentration Over the past decade, numerous studies have explored the potential
of statins in non-​hepatic organs and tissues (Friis and Olsen, 2006). chemopreventive effects of NSAIDs, in particular aspirin. As many
Several US groups have also suggested that “healthy-​user bias” may NSAIDs are available without a prescription, this has implications
account for a considerable part of the observed inverse associations for the choice of the optimal data source for epidemiological studies
between statin use and risk of some cancer types, whereby statin use (i.e., self-​reporting versus prescription records). There is evidence that
may be associated with more “health-​aware” behavior, such as par- the accuracy of patient-​reported drug use and prescriptions records of
ticipation in cancer screening and other preventive behaviors associ- NSAIDs, and other drugs used to treat musculoskeletal disorders, dif-
ated with a reduced risk of manifest cancer (Setoguchi et al., 2005). fer by the type of drug and the reporting period (Grimaldi-​Bensouda
However, surveys in Denmark and the United Kingdom indicate et al., 2012).
that statin users are fairly representative of the general population, Multiple systematic reviews and meta-​ analyses have reported
and not a selected population of “healthier” individuals as reported that the use of aspirin or non-​aspirin NSAIDs reduces the risk of
in the United States (Hippisley-​Cox and Coupland, 2010; Thomsen colorectal cancer (Algra and Rothwell, 2012; Bibbins-​ Domingo
et al., 2013b). A recent study with clinical laboratory data (Friedman and US Preventive Services Task Force, 2016; Cuzick et  al., 2009;
et al., 2016) suggests that the inverse association commonly observed Rothwell et  al., 2012a). The meta-​analyses indicate that long-​term
between statins and liver cancer may not be causal, but due to con- (> 5  years), consistent use of low-​dose aspirin is associated with a
founding by indication (elevated cholesterol levels) and contraindica- significant reduction in the risk of colorectal cancer (Cuzick et  al.,
tion (elevated liver enzymes). 2015). However, the optimal dose and duration is still unclear, and
420

420 Part III: The Causes of Cancer


other important questions remain unresolved, including the optimal esophageal, pancreas, colorectal, and stomach) (Rothwell et al., 2011).
age at initiation and whether other factors might help maximize the Of note, this improved prognosis was apparent after 5 years of follow-​
benefits and minimize the harms of treatment (Drew et al., 2016). For up (7 trials) and the mortality risk remained reduced at 20 years of
example, there is increasing evidence that the chemopreventive effect follow-​up among individuals who were assigned to the original aspirin
of aspirin varies substantially with body mass and level of inflamma- intervention arm compared to those in the control arm (three trials)
tory response (Movahedi et al., 2015), as well as with genetic markers (Rothwell et al., 2011). There is also some evidence that the anti-​can-
(Drew et al., 2016). cer effect of aspirin may extend to the prognosis of breast and pros-
The potential use of aspirin for prevention of colorectal cancer tate cancer (Elwood et  al., 2016; Huang et  al., 2015); however, the
is limited by the risk for gastrointestinal bleeding and, less often, results are not consistent (Cardwell et al., 2014c; Zhong et al., 2015a).
cerebral bleeding (Dehmer et al., 2016; Drew et al., 2016) (see also Randomized controlled trials of aspirin in the treatment of cancer will
Chapter  62.5), and these potential harms will need to be balanced provide further information on cancer recurrence and mortality. For
against the preventive benefits against cancer and cardiovascular example, the Add-​Aspirin Trial (phase III double-​blind placebo-​con-
disease. trolled randomized trial) aims to examine the effect of aspirin exposure
There is also increasing evidence that aspirin use is associated after standard primary therapy on early-​stage common solid tumors
with a decreased risk of upper gastrointestinal cancer (Sahin et  al., (Langley et al., 2013).
2014). A recent meta-​analysis found that long-​term (≥4 years) and
low frequency (1–​4.5 times/​week) aspirin use was associated with
a substantial reduction in gastric cancer risk (Ye et al., 2013). Other
Bisphosphonates
meta-​analyses have supported this inverse association (Sahin et  al.,
2014), although there is evidence of possible confounding by contra- Bisphosphonates (BPs) are potent inhibitors of osteoclast-​mediated
indication or publication bias. Consistent risk reductions with use of bone resorption, and are commonly prescribed in the management of
aspirin have also been reported for esophageal cancer (Xiaohua et al., postmenopausal and glucocorticoid-​ associated osteoporosis, meta-
2014; see also Chapter 62.5). static bone disease, and other bone-​related conditions (Drake et  al.,
In the largest meta-​analysis of NSAID use and risk of breast cancer 2008). In vitro and in vivo studies have suggested that BPs, particu-
performed to date, which included case-​control (n  =  23) and cohort larly those containing nitrogen, exhibit antitumor activity by promot-
studies (n = 24) and two randomized clinical trials, the pooled rela- ing apoptosis and by inhibiting angiogenesis, tumor cell adhesion, and
tive risk estimates were consistent with a slightly protective effect of invasion (Clezardin, 2013).
NSAID use against breast cancer (de Pedro et  al., 2015). Although However, in 2009, based on an accumulating number of spontane-
based on fewer studies, the meta-​analysis also indicated that the pro- ous reports from the United States, Europe, and Japan, the FDA issued
tective effect of aspirin pertained specifically to hormone receptor–​ a warning of a potentially increased risk of esophageal cancer associ-
positive breast tumors (de Pedro et al., 2015). Meta-​analyses of a large ated with the use of oral BPs (Wysowski, 2009). This sparked a flurry
number of studies have also reported a 5%–​10% reduced risk of pros- of epidemiological studies. Although results across meta-​ analyses
tate cancer, or advanced prostate cancer, associated with aspirin use have been somewhat conflicting, the most recent (Wright et al., 2015),
(Liu et al., 2014). based on five retrospective observational studies, did not find a signif-
Regarding other non-​gastrointestinal cancer sites, the evidence is icantly elevated risk of either esophageal or gastric cancer. However,
either inconsistent or nonexistent for a chemopreventive effect of use there was considerable heterogeneity in population characteristics
of aspirin or other NSAIDs. For cancers of the lung (Hochmuth et al., and definitions of drug exposure, and information was limited on the
2016; Jiang et al., 2015), pancreas (Cui et al., 2014), ovary (Baandrup effects of type, dose, or duration of BP use, as well as potential risk
et al., 2013; Trabert et al., 2014), endometrium (Verdoodt et al., 2016), variation by histologic subtype of esophageal cancer (i.e., adenocarci-
and non-​melanoma skin cancer (Muranushi et  al., 2015, 2016), the noma or squamous cell carcinoma).
majority of studies have reported inverse associations with aspirin or A recent pooled analysis of eight epidemiological studies (Bonovas
non-​aspirin NSAID use. Largely null results have been observed for et  al., 2013)  reported a statistically significant inverse association
bladder cancer (Daugherty et al., 2011; Zhang H et al., 2013) and mel- between BP use and colorectal cancer risk. Long-​term (3 or more
anoma (Hu et al., 2014). A recent meta-​analysis reported an increased years) use of BPs was associated with a 27% reduction in colorectal
risk of kidney cancer with use of non-​aspirin NSAIDs (Choueiri et al., cancer risk compared with non-​use. Some studies have also reported
2014). Ongoing randomized controlled trials of aspirin in the preven- a decreased incidence of postmenopausal breast cancer with BP use
tion of specific cancers will provide further information on aspirin use (see Chapters  45 and 62.5). However, confounding by indication is
and cancer risk. For example, the ASPirin in Reducing Events in the likely given the association between lower levels of endogenous
Elderly (ASPREE) is examining the risk of cancer as a secondary out- estrogen and both lower bone mineral density and lower breast can-
come (ASPREE Investigator Group, 2013). cer risk (Chlebowski and Col, 2012). Recent cohort (Alford et  al.,
The US Preventive Services Task Force (USPSTF) has recently 2015; Newcomb et al., 2015) and case-​control studies (Rennert et al.,
issued recommendations advising the use of aspirin to prevent cardio- 2014)  have also reported reductions in risk of endometrial cancer,
vascular disease and colorectal cancer in middle-​aged individuals with particularly among postmenopausal women (Newcomb et al., 2015);
modestly increased risk of cardiovascular disease (Bibbins-​Domingo however, additional studies with larger sample sizes and more compre-
and US Preventive Services Task Force, 2016; Dehmer et al., 2016). hensive adjustment for confounding are warranted.
The USPSTF systematic review was based on the multiple lines of Recently, there has been increasing interest in a potential therapeu-
evidence that aspirin use reduces the risk of colorectal adenoma and tic role of adjuvant BPs in patients with breast cancer. A meta-​analy-
cancer; however, the recommendation is not an explicit endorsement sis of individual patient data from RCTs of more than 18,000 women
of aspirin use for the sole purpose of colorectal cancer prevention. conducted by the Early Breast Cancer Trialists’ Collaborative Group
Several questions remain to be addressed before recommendations for reported a reduction in breast cancer bone metastases and improved
primary prevention of cancer with aspirin can be established, includ- survival with adjuvant BP use among postmenopausal but not premen-
ing the optimal dose of aspirin for cancer prevention and potential opausal women (Early Breast Cancer Trialists’ Collaborative Group
effect modification according to genetic, medical, and lifestyle factors et  al., 2015). Subsequently, a European consensus panel suggested
(Drew et al., 2016). that BPs have a clinically important role in early breast cancer in a
Recent systematic reviews and meta-​analyses also suggest that aspi- postmenopausal adjuvant setting (Hadji et al., 2016). Outcome stud-
rin use may reduce cancer progression and mortality (Chubak et al., ies have also been conducted among other cancer patients frequently
2016b; Elwood et al., 2016; Rothwell et al., 2011, 2012a, 2012b). A prescribed BPs. At present, there is some evidence that post-​diagnos-
pooled analysis of large cardiovascular trials found that daily aspirin tic BP use improves survival in localized or metastatic prostate (Vale
use was associated with a 34% reduction in mortality from all cancers et al., 2016) or colorectal cancer (Hicks et al., 2015). A large Cochrane
and a 54% reduction in death from gastrointestinal cancers (including review (Mhaskar et al., 2012) summarizing the findings from 20 RCTs
 421

Pharmaceutical Drugs Other Than Hormones 421


found no indication of improved survival among patients with multiple prevent cancer or cancer recurrence have been initiated (Cuzick et al.,
myeloma. Notably, the across-​study heterogeneity was high (I2 =55%) 2015). Use of sulfonylurea, another class of first-​line therapy and the
in this analysis, and several of the included studies may not have fol- comparator drug for many studies of metformin, has been reported
lowed standard clinical trial methods. to increase the risk of cancer in some, but not other studies (Thakkar
et al., 2013).
Several second-​and third-​ line therapies have also been evalu-
Digoxin ated. Some studies have indicated that insulin use may be associ-
Digoxin, a cardiac glycoside derived from natural steroid compounds, ated with an increased risk of several cancer types (Carstensen et al.,
is widely prescribed for atrial fibrillation, congestive heart failure, and 2012; Janghorbani et  al., 2012; Singh et  al., 2013c, 2013d). Insulin
related conditions (Brayfield, 2014). Early observational studies sug- analogues, in particular, have been suggested to increase cancer risk
gested that digoxin use might protect against cancer, notably breast (McFarland and Cripps, 2010). However, this association was not
cancer (Stenkvist et  al., 1979). It was hypothesized that the steroid supported in a recent update from the ORIGIN trial, where an insulin
core of cardiac glycosides might reduce ER signaling analogous to analogue, glargine, was compared with standard care among 12,537
tamoxifen (Stenkvist, 1999). However, recently, the IARC has catego- patients with diabetes for a median follow-​up of 6.2 years (Bordeleau
rized digoxin as possibly carcinogenic to humans (Group 2B) (Grosse and Gerstein, 2014). Also, a previous meta-​analysis of 11 RCTs or
et al., 2013; IARC, 2015) because it has been associated consistently observational studies reported null effects of insulin analogues on can-
with increased risk of both male and female breast cancer. Recent cer risk (Tang et al., 2012).
findings from the Women’s Health Study and a nationwide Danish The thiazolidinediones also have been reported to increase the risk
case-​control study indicated an increased risk of invasive breast cancer of cancer, either overall or at specific sites, although findings have
associated with current digoxin use (Ahern et al., 2014; Biggar et al., been conflicting (Monami et al., 2014). Some of the most consistent
2011; Grosse et al., 2013; IARC, 2015). The association was strongest findings have been reported for pioglitazone, a thiazolidinedione, and
for ER-​positive tumors (Biggar et  al., 2013), and laboratory studies bladder cancer, with modestly increased risk occurring after 2+ years
have demonstrated that digoxin acts like a phytoestrogen, binding to of use (Ferwana et al., 2013). Pioglitazone has recently been classi-
ERs and stimulating the growth of ER-​sensitive tumors (Biggar et al., fied by the IARC as probably carcinogenic to humans (Group  2A)
2013). Digoxin use has also been associated with an increased risk (IARC, 2015).
of uterine and colorectal cancer (Grosse et  al., 2013; IARC, 2015). The recently marketed incretin mimetics (dipeptidyl peptidase-​4
However, the findings for digoxin are difficult to interpret, due to pos- inhibitors and glucagon-​like peptide-​1 analogues) have also been sug-
sible confounding factors, particularly obesity and alcohol consump- gested to possess a carcinogenic potential (Egan et al., 2014). Three
tion (Grosse et  al., 2013). Moreover, some animal (Menger et  al., meta-​analyses yielded null findings for associations between use of
2012) and epidemiologic studies have indicated that digoxin also pos- incretin mimetics and short-​term incidence of pancreatic or other can-
sesses anti-​neoplastic effects. Consistent with the possible estrogenic cers (Alves et al., 2012; Dejgaard et al., 2009; Monami et al., 2011).
effect, digoxin use has been suggested to reduce the risk of prostate However, additional studies with longer follow-​ up are warranted.
cancer. One study based on data from the Health Professionals Follow-​ Several international research initiatives have recently been launched
up Study demonstrated an inverse association between digoxin use to clarify issues related to diabetes and cancer.
and prostate cancer risk, especially after more than 10  years of use
(Platz et al., 2011). An additional US population-​based cohort study
reported a 40% reduced risk of prostate cancer among digoxin users Drugs Used in Acid–​Related Disorders
(Wright et al., 2014). In contrast, a recent case-​control study showed Proton pump inhibitors (PPIs) and histamine-​2 receptor antagonists
no association between digoxin use and prostate cancer risk (Kaapu (H2RAs) inhibit or reduce gastric acid secretion and are commonly
et al., 2015). prescribed for the treatment of gastrointestinal disorders such as dys-
A few studies have also examined the association between digoxin pepsia, gastroesophageal reflux disease, and peptic ulcers. Inhibition
use and cancer prognosis. A  large population-​ based cohort study of acid secretion may increase bacterial growth, leading to the pro-
in the United Kingdom presented little evidence of an increase in duction of N-​nitrosamines, which may promote carcinogenesis (Laine
breast cancer–​specific mortality with post-​diagnostic use of digoxin et al., 2000; Stockbruegger, 1985). Inhibition of acid secretion can also
(Karasneh et al., 2015), and a recent cohort study showed no associa- induce hypergastrinemia, and high gastrin levels have been suggested
tion between digoxin and prostate cancer–​specific mortality (Flahavan to induce neoplasia in the gastrointestinal tract (Laine et al., 2000).
et al., 2014). In recent years, several studies have examined the association
between PPI/​H2RA use and cancer risk across a number of gastro-
Drugs Used in Diabetes intestinal sites including esophageal, gastric, and colorectal cancer
(Ahn et al., 2013). Some PPI/​H2RA drugs are now available over the
Diabetes mellitus is associated with an increased or decreased risk of counter, and thus some use will not be captured by electronic pre-
several cancers (Vigneri et  al., 2009). Over the last decade, numer- scription records. Prophylactic therapy with PPI for Barrett’s esoph-
ous studies and meta-​analyses of antidiabetic drugs and cancer risk agus has been of particular interest. A recent meta-​analysis (including
have been published. In addition to potential confounding by indi- 7 observational studies) reported that PPI use was associated with a
cation (i.e., diabetes or diabetes severity), challenges to these stud- decreased risk of disease progression to esophageal adenocarcinoma
ies include the cross-​over and treatment escalation often required to and/​or high-​grade dysplasia (Singh et al., 2014). However, there was
manage the progressive hyperglycemia occurring with type 2 diabetes, substantial heterogeneity in the results. Furthermore, only two of
which accounts for up to 90% of diabetes in older adults (ages when the studies examined H2RAs, and most studies of PPI/​H2RA drugs
most cancers occur). Some antidiabetic drugs are also associated with did not adjust for potential confounding factors, such as obesity and
weight gain, an independent risk factor for several cancers (Renehan smoking. The results of ongoing phase III RCTs of acid suppression
et al., 2015). in patients with Barrett’s esophagus will likely provide more robust
Metformin, a first-​line therapy and the most commonly prescribed evidence. For example, the multi-​center Aspirin and Esomeprazole
antidiabetic drug, has been hypothesized to possess chemopreventive Chemoprevention in Barrett’s Metaplasia (AspECT) trial—​the larg-
properties. While several observational studies have reported inverse est RCT of Barrett’s metaplasia performed to date—​will assess the
associations for cancer at multiple sites (e.g., breast, colon, prostate, influence of varying doses of esomeprazole, with or without aspi-
and lung), many studies have suffered from time-​related biases that rin, on cancer progression in patients with Barrett’s metaplasia (Das
may at least in part explain some of these protective associations et al., 2009).
(Suissa and Azoulay, 2012). Given findings from in vivo and in vitro Several studies have investigated the risk of gastric cancer associated
studies as well as observational studies, several RCTs of metformin to with use of PPIs/​H2RAs. A recent meta-​analysis of 11 observational
42

422 Part III: The Causes of Cancer


studies (9 case-​control and 2 cohort studies) reported that both PPI (10 cancer (Chubak et al., 2011; Coogan et al., 2009). However, the results
studies) and H2RA (3 studies) use were associated with an increased are inconsistent (Cronin-​ Fenton et  al., 2011), and a recent meta-​
risk of gastric cancer (Ahn et al., 2013). Of note, site-​specific analy- analysis of observational studies evaluating SSRI use and colon cancer
ses showed a significant increased risk for gastric non-​cardia cancer, risk reported an overall null association (Lee et al., 2012). Failure to
but no association with gastric cardia cancer. Use of PPI/​H2RA for account for colorectal cancer screening history and lack of adjustment
less than 5 years was significantly associated with an increased risk of for concomitant drug use are important limitations of several of the
gastric cancer, with a non-​significant positive association observed for included studies, as both medical surveillance and drug use may differ
long-​term use (greater than 5 years). However, this review could not between AD users and non-​users in the general population. Regarding
exclude protopathic bias (reverse causation), and no data were avail- other cancer sites, there is little evidence of an association with SSRI
able on underlying gastric conditions. Furthermore, the results of the use. A recent large case-​control study in Kaiser Permanente Northern
meta-​analysis were heavily influenced by one case-​control study that California (KPNC) found little evidence of an association between
did not adjust for weight, smoking status, or alcohol consumption, and fluoxetine, paroxetine, or other SSRIs and testicular cancer, although
only one of the included studies adjusted for NSAID use. In a meta-​ a weak association could not be ruled out for individual histological
analysis of six randomized controlled trials, the authors reported no types (Friedman et al., 2014). One study reported that SSRI use was
association between PPI use and risk of gastric atrophic changes or associated with a reduced risk of lung cancer (Toh et al., 2007). The
enterochromaffin-​like (ECL) cell hyperplasia over 3 years (Eslami and authors of that study attempted to account for confounding by indi-
Nasseri-​Moghaddam, 2013). Although four of the six studies were cation by adjusting for the severity of depression using information
prone to bias, a sensitivity analysis restricted to the two higher-​quality on the length of depression history, use of other AD types, and over-
studies showed similar results to the overall analysis with the exception all healthcare utilization. However, adjustment for these factors and
of PPI dosage, where ECL-​cell score was significantly increased with restriction to individuals with more than 5 years of depression history
esomeprazole (20 mg and 40 mg, but not 10 mg) compared to placebo. had only a modest impact on the association (Toh et al., 2007). The
A Cochrane review, which included seven randomized controlled tri- authors also acknowledged the association of smoking and depression.
als, reported no clear evidence that long-​term use of PPIs (greater than Smoking is associated with an increased risk of depression and, con-
6 months) was associated with the progression of corpus gastric atro- sequently, certain unmeasured aspects of smoking might be associated
phy or intestinal metaplasia (Song et al., 2014). However, PPI users with AD use.
exhibited an increased risk of simple or focal ECL-​cell hyperplasia. In a Canadian study, TCA use was associated with an elevated
Relatively few studies have examined PPI use and risk of colorectal risk of prostate cancer, although results may have been influenced by
cancer to date. A meta-​analysis of four large case-​control studies with detection bias (Tamim et al., 2008). Some studies indicate that long-​
an average follow-​up period of less than 5 years showed no associa- term use of TCA may increase the risk of non-​Hodgkin lymphoma
tion (Chen et al., 2011). A Cochrane review of six randomized con- (Dalton et al., 2008); however, the results are conflicting (Bahl et al.,
trolled trials (5 studies of cimetidine, 1 study of ranitidine) concluded 2004). In a recent study, Walker et al. (2011) found an inverse associa-
that H2RA use as adjunct therapy for resected colorectal cancer was tion between TCA and risk of glioma or colorectal cancer, with the risk
associated with improved survival (Deva and Jameson, 2012). A com- of both decreasing with increasing TCA use. A second recent study
bined follow-​up analysis of data from three adenoma prevention trials also observed an inverse association between TCA use and glioma risk
(n = 2915 participants) based on self-​reported information on drug use (Pottegård et al., 2016b).
other than aspirin showed no association between H1RA or H2RA use Limited research has been undertaken on ADs and cancer outcomes.
and adenoma risk during a maximum follow-​up of 4 years, although It has been suggested that SSRI use (particularly paroxetine and fluox-
H2RA use was associated with a reduced risk of adenomas in one of etine) may inhibit the effects of the endocrine therapy tamoxifen, thus
the trial populations (Robertson et al., 2005). increasing the risk of breast cancer recurrence in concurrent tamoxi-
fen/​SSRI users. A  large study of breast cancer patients treated with
tamoxifen found little evidence for an association between concurrent
Psychotropic Drugs SSRI use overall, or paroxetine or fluoxetine specifically, and risk of
recurrence (Haque et al., 2016). A second study (Chubak et al., 2016a)
Antidepressants also found no association between breast cancer recurrence or mortal-
Antidepressants (ADs) are widely used to alleviate mood disorders, ity and use of different types of AD (SSRIs/​TCAs/​other). However, and
such as major depression and anxiety, and work by altering either the although the number of users was small, concurrent users of tamoxi-
re-​uptake of common neurotransmitters, such as serotonin, or enzymes fen and paroxetine appeared to have an increased risk of breast cancer
involved in their synthesis (Stafford et al., 2001). Immune system acti- recurrence compared to tamoxifen-​treated patients not using an AD.
vation and inflammation have been suggested to be involved in the
pathophysiology of both depression and cancer, and chronic depres- Benzodiazepines
sion has been associated with increased cancer risk, progression, and Benzodiazepines are commonly used anxiolytic and hypnotic medi-
impaired survival, although the underlying mechanisms are unclear cations (Brayfield, 2014). Early animal studies suggested that these
(Currier and Nemeroff, 2014). It has been proposed that ADs may agents may be carcinogenic and increase the risk of a number of can-
attenuate any such effect (Currier and Nemeroff, 2014); however, cers (IARC,1996), observations that are also inferred by some obser-
observational studies (Dalton et  al., 2008)  and a recent screening vational studies (Halapy et al., 2006; Rosenberg et al., 1995). Recently,
study of commonly used drugs for carcinogenicity suggest that some a US cohort study reported that use of hypnotic drugs, including ben-
ADs may actually increase the risk of cancer (Friedman et al., 2009c). zodiazepines, was associated with an up to two-​fold increased risk of
Preclinical studies have also shown that ADs may influence various cancer overall (Kripke et al., 2012). However, this study has been criti-
processes in the cell cycle such as growth proliferation, metastasis, and cized due to its inappropriate and biased methods of selecting controls
apoptosis (Frick and Rapanelli, 2013). (Pottegård et al., 2013a). A nationwide nested case-​control study using
Some observational studies have shown an increased risk of breast data from Danish registries found no association between long-​term
cancer associated with long-​ term use of tricyclic antidepressants benzodiazepine use and overall cancer risk, but did observe marginally
(TCAs) or selective serotonin reuptake inhibitors (SSRIs) (Cummings increased risks at a number of sites including the stomach, esophagus,
et al., 2002; Moorman et al., 2003; Sharpe et al., 2002). However, a liver, lung, pancreas, and kidney. The authors interpreted these slight
recent well-​conducted meta-​analysis of 18 studies found no overall risk elevations as residual confounding by lifestyle factors, such as
increased risk of breast cancer among AD users, irrespective of AD alcohol and tobacco use, which are known to have a higher prevalence
type (Eom et al., 2012). among individuals using psychotropic drugs, including benzodiazepines
SSRIs are purported to have antiproliferative effects on colon cancer (Pottegård et al., 2013a). Prompted by these findings, a team at Kaiser
cells (Kannen et al., 2012), and some observational studies have also Permanente Northern California examined the association of benzo-
suggested that SSRI use is associated with a reduced risk of colorectal diazepine use and liver cancer risk. Like the Danish study, the Kaiser
 423

Pharmaceutical Drugs Other Than Hormones 423


Permanente study revealed a slight excess risk in liver cancer associated use of sildenafil, a PDE5 inhibitor. Another recent study conducted
with the use of some types of benzodiazepines (diazepam/​temazepam in Sweden (Loeb et al., 2015) also reported a statistically significant
and oxazepam). However, again, since alcohol use, particularly heavy increased risk of melanoma associated with the use of PDE5 inhibi-
use, is associated with both benzodiazepine use and liver cancer, the tors, including sildenafil and other PDE5 inhibitors. However, the
association was likely due to confounding by alcohol, and the database association was limited to early stage disease and no dose–​response
did not have sufficient information on lifestyle variables to evaluate this relationship was apparent, suggesting non-​causal explanations, such as
interpretation (Friedman et al., 2015). For other ethnic populations, one increased disease surveillance among users of PDE5 inhibitors.
population-​based cohort study in Taiwan reported a 19% higher risk of
cancer overall, and a 45% elevated risk of liver cancer, among benzodi- Immunosuppressants
azepine users compared to non-​users (Kao et al., 2012). Immunosuppressants are a class of drugs primarily used to avert rejec-
To our knowledge, no studies have examined benzodiazepine use tion in solid organ transplant recipients (i.e., kidney, heart, liver, and
in relation to cancer survival, despite the fact that the use of these bone marrow), but may also form the cornerstone of management of
agents increases substantially following cancer diagnosis (Andersen many chronic inflammatory and autoimmune diseases (Forsythe and
et al., 2015). Paterson, 2014). There are four major types of immunosuppressants,
including corticosteroids (e.g., prednisolone and dexamethasone),
cytotoxic agents (e.g., cyclophosphamide, azathioprine, and myco-
Miscellaneous Drugs phenolate mofetil), T-​cell suppressive agents (e.g., cyclosporine and
tacrolimus), and antibodies (e.g., rituximab).
Tumor Necrosis Factor Inhibitors Experimental and clinical data have established that suppression of
Concerns have been raised about the new biological response modi- the immune system may induce several malignancies (see Chapter 25),
fiers (e.g., tumor necrosis factor [TNF] inhibitors) (Singh et al., 2009). including skin cancers (Pedersen et al., 2014), lymphoma (Khan et al.,
TNF inhibitors have been suspected to increase the risk of certain can- 2013), myeloid leukemia and myelodysplastic syndromes (Lopez et al.,
cers, including lymphoma in children and adolescents. A large meta-​ 2014), bladder (Yan et al., 2014), and colorectal cancer (Gong et al., 2013).
analysis of 61 RCTs of TNF inhibitors did not indicate an increased The IARC Monographs Program (IARC, 2012)  recently assessed
risk of cancer, either overall or at specific sites, associated with use of all available experimental and clinical data and concluded that the
TNF inhibitors (Lopez-​Olivo et al., 2012). This was compatible with immunosuppressants azathioprine and cyclosporin are definitely car-
findings from other meta-​analyses (Zhang D et al., 2013), except for cinogenic in humans (Table  23–​1). Specifically, sufficient evidence
one meta-​analysis that reported a non-​statistically significant increase has shown that immunosuppressants induce non-​ melanoma skin
in the risk of B-​cell lymphomas associated with TNF inhibitors (Wong cancer and non-​Hodgkin lymphoma. Moreover, epidemiologic stud-
et al., 2012). TNF inhibitors were introduced on the market in 2002, ies have indicated that long duration and high-​intensity exposure to
and thus, studies have generally only been able to evaluate effects of immunosuppressants is associated with an increased risk of several
shorter-​term treatment (Wong et  al., 2012). A recent Danish nation- other cancer types (Gutierrez-​Dalmau and Campistol, 2007). On bal-
wide register-​based study showed that among 56,156 patients with ance, it would seem that the benefits of immunosuppressants outweigh
inflammatory bowel disease, long-​term (> 5 years) use of TNF inhibi- their carcinogenicity (Rama and Grinyo, 2010); however, each patient
tors was associated with a relative risk for cancer overall of 1.33 (95% eligible for immunosuppressive therapy should undergo a thorough
CI: 0.88, 2.03) (Nyboe Andersen et al., 2014). Continued monitoring benefit–​risk evaluation (Friis et al., 2015a).
of TNF inhibitors is warranted. Only relatively few studies have examined the association
between immunosuppressive therapy and risk of cancer recurrence
Warfarin or second primary malignancy. A recent systematic review (Shelton
In vitro and in vivo studies have shown that warfarin interferes with et  al., 2016), including 16 studies (12 cohort, 3 case-​series, and
cancer cell growth and development, inhibits angiogenesis, alters host-​ 1 case-​control) and a total of 11,702 patients, reported that thiopu-
immune responses and prevents metastatic spread (Hejna et al., 1999). rines, methotrexate, and TNF inhibitors were not associated with an
Only a few clinical studies have investigated the association between increased risk of cancer recurrence. Limitations of this review, how-
warfarin use and cancer risk, and the results are inconsistent (Schulman ever, included substantial diversity in the included cancer types, dif-
et  al., 1995; Taliani et  al., 2003). Epidemiological studies have also ferences in the ascertainment of cancer recurrence, and small sample
reported mixed findings. A large case-​control study in the United States sizes (3 studies included less than 20 cases). Another recent meta-​
(Blumentals et al., 2004) reported no association between warfarin use analysis of 4621 non-​Hodgkin lymphoma patients from nine RCTs
and bladder cancer, whereas a Canadian case-​control study found that found no increased risk of second primary cancers among users of
warfarin reduced the risk of prostate cancer (Tagalakis et  al., 2007). rituximab (Fleury et al., 2016).
Recently, a large Danish nationwide case-​control study found no asso-
ciation between long-​term use of warfarin or vitamin K antagonists
(VKA) and overall cancer risk, but the results indicated a protective FUTURE DIRECTIONS
effect of VKA against prostate cancer (Pottegård et al., 2013b).
Several studies have investigated whether warfarin may improve New Drug Agents and Exposures
survival in cancer patients, but findings have been equivocal (O’Rorke
et  al., 2015). In addition, an extension of the above Canadian study The development of new medications with de novo active substances
of bladder cancer indicated that long-​term warfarin use (> 4 years) and new targets and mechanisms is expected to continue. In addition,
may be associated with an impaired cancer prognosis (Tagalakis and the increasingly global manufacturing of and marketplace for pharma-
Tamim, 2010), and a nested case-​control study found no evidence that ceuticals has the potential to result in rapid and large-​scale exposure
long-​term warfarin use improved prostate cancer survival (Tagalakis to medications. Thus, increasing proportions of children and adults
et al., 2013), whereas short-​term use indicated an increased mortality. worldwide will likely be exposed to a diversity of both older and
newer medications. It is imperative to continue close monitoring of
Phosphodiesterase Type 5 Inhibitors both short-​and long-​term drug effects, including potential increases or
The target for several oral erectile dysfunction drugs, phosphodi- decreases in cancer risk.
esterase type 5 (PDE5), is part of a signaling pathway that may be
involved in the development of malignant melanoma. PDE5 inhibitors
have thus been suggested to increase the risk of cutaneous melanoma. New Classifications of Cancer and Cancer Subtypes
In 2014, a cohort study among US male health professionals (Li WQ There is a growing appreciation of the molecular complexity of cancer
et al., 2014) reported an increased risk of melanoma associated with such that, even for patients with the same cancer and disease stage,
42

424 Part III: The Causes of Cancer


there are heterogeneous subgroups with varying risk factor profiles may act as confounders or effect modifiers, and by identifying cancer
and clinical outcomes (Ogino et  al., 2012). Most cancer registries subtypes that may exhibit stronger or weaker associations with use of
record information on histologic (or morphologic) subtype; however, the drug(s) of interest (Gottlieb and Altman, 2014).
details of specificity, accuracy, and completeness vary. In addition, as
cancer therapies increasingly target specific tumor subtypes, the test-
ing of tumor tissue for genetic mutations and gene and protein expres- Translational Research: Discovery, Validation,
sion is increasingly routine in clinical practice. Such routine testing and Clinical Utility
will facilitate studies of medication use and risk of specific molecular
New drugs, molecular testing, databases, and analytic methods are
subtypes of cancer.
all likely to increase the number of pharmacoepidemiology studies
and findings. In the future, new drug–​cancer signals and hypotheses
Incorporating Germline Genetics into will continue to arise from animal and in vitro studies, clinical trials,
and epidemiologic studies, as well as from complementary bioinfor-
Drug–​Cancer Studies matics, such as systems pharmacology. However, while “Big Data”
Germline genetic variants (e.g., single nucleotide polymorphisms promises to provide important insights into the causes and outcomes
[SNPs]), may help identify subgroups of patients most likely to of diseases, better drug targets and improved disease prediction, it can
benefit—​or to develop adverse effects—​from chemopreventive and also be subject to “Big Errors” (Khoury and Ioannidis, 2014). Thus,
therapeutic agents (Mendes, 2015). For example, aspirin was recently given the potential public health importance of drug–​cancer signals, it
found to be beneficial against colorectal cancer only among individu- will be essential to carefully examine these findings within multiple,
als with at least one T allele associated with expression of the MYC well-​conducted studies that address potential selection bias and con-
oncogene (Nan et al., 2013). founding, as well as biologic plausibility (e.g., latency, cumulative
exposure). For some replicated drug–​cancer associations, it may be
possible to subsequently conduct RCTs. However, observational stud-
New Databases and Collaborative Opportunities ies will be the only option for evaluating the majority of drug–​cancer
Studies of low-​prevalence medications, tumor subtypes, and genetic vari- associations. Before translating findings into regulatory action or clini-
ants all require extremely large study populations. A growing number of cal care, it will also be essential that both the benefits and harms of a
large healthcare databases with relatively easy linkage to cancer registries medication are carefully considered.
are available for research. However, the level of data quality is inconsis-
tent across all health data sets and registries. Frameworks for improving References
data quality have been suggested and may prove useful (Arts et al., 2002). Abernathy D, Altman R, Brouwer K, et  al. 2011. Quantitative and systems
For example, the Surveillance, Epidemiology, and End Results (SEER) pharmacology in the post-​genomic era: new approaches to discovering
Program and the IARC have provided guidelines to improve the quality drugs and understanding therapeutic mechanisms. [Internet]. Available
of cancer data. Ideally, high-​quality population-​based data should also be at:  https://​www.nigms.nih.gov/​training/​documents/​systemspharmawps-
available on genetic, lifestyle, and environmental factors. orger2011.pdf Accessed on April 24, 2016.
Many of the large studies discussed in this chapter were unable to Ahern TP, Pedersen L, Tarp M, et al. 2011. Statin prescriptions and breast can-
cer recurrence risk: a Danish nationwide prospective cohort study. J Natl
adequately assess the drugs of interest, and even larger, collaborative
Cancer Inst, 103(19), 1461–​1468. PMCID: PMC3186780.
studies are clearly warranted. Such collaborations could follow the Ahern TP, Tamimi RM, Rosner BA, and Hankinson SE. 2014. Digoxin use
example of the international research consortia for studying the genet- and risk of invasive breast cancer:  evidence from the Nurses’ Health
ics of cancer and other complex diseases. The aims of these consortia Study and meta-​analysis. Breast Cancer Res Treat, 144(2), 427–​435.
are to share and analyze the wealth of data from a number of already PMCID: PMC4010120.
established cohort or case-​control data sets (ENGAGE, 2012). While Ahn JS, Eom CS, Jeon CY, and Park SM. 2013. Acid suppressive drugs
such consortia can raise issues related to data harmonization, as well and gastric cancer:  a meta-​analysis of observational studies. World J
as sharing of extremely large healthcare data sets, new methods and Gastroenterol, 19(16), 2560–​2568. PMCID: PMC3646149.
strategies are under development to address privacy issues and other Alford SH, Rattan R, Buekers TE, and Munkarah AR. 2015. Protective effect
of bisphosphonates on endometrial cancer incidence in data from the
challenges (Toh et al., 2014).
Prostate, Lung, Colorectal and Ovarian (PLCO) cancer screening trial.
Cancer, 121(3), 441–​447. PMID: 25533883.
Algra AM, and Rothwell PM. 2012. Effects of regular aspirin on long-​term
Advances in Pharmacoepidemiology cancer incidence and metastasis:  a systematic comparison of evidence
and Complementary Methods from observational studies versus randomised trials. Lancet Oncol, 13(5),
518–​527. PMID: 22440112.
In the past 20  years, numerous advances in epidemiologic methods ALLHAT Officers and Coordinators for the ALLHAT Collaborative Research
have been developed, many of which are particularly relevant to phar- Group 2002. Major outcomes in high-​risk hypertensive patients random-
macoepidemiology, such as propensity scores, marginal structural ized to angiotensin-​ converting enzyme inhibitor or calcium channel
models, and methods for external adjustment. Conceivably, such meth- blocker vs diuretic: The Antihypertensive and Lipid-​Lowering Treatment
odologic advances will continue, as will the development of new tools to Prevent Heart Attack Trial (ALLHAT). JAMA, 288(23), 2981–​2997.
PMID: 12479763.
to facilitate their use.
Altwairgi AK. 2015. Statins are potential anticancerous agents (review). Oncol
Translational bioinformatics also has the potential to complement Rep, 33(3), 1019–​1039. PMID: 25607255.
pharmacoepidemiology. Systems pharmacology is an emerging field Alves C, Batel-​Marques F, and Macedo AF. 2012. A meta-​analysis of seri-
that applies systems biology approaches, combining large-​scale exper- ous adverse events reported with exenatide and liraglutide:  acute
imental studies with model-​based computational analyses, often lever- pancreatitis and cancer. Diabetes Res Clin Pract, 98(2), 271–​ 284.
aging publicly available databases, such as PharmGKB and DrugBank, PMID: 23010561.
to study drug activities, targets, and effects (Abernathy et al., 2011). Andersen LT, Suppli NP, Dalton SO, et al. 2015. New and chronic use of hyp-
In the systems pharmacology framework, a drug interacts with multi- notics after diagnosis with early breast cancer:  a retrospective cohort
ple targets that lie within interconnected biological pathways related study. Acta Oncol, 54(5), 704–​711. PMID: 25752966.
ARB Trialists Collaboration. 2011. Effects of telmisartan, irbesartan, valsartan,
to one or more diseases (Berger and Iyengar, 2011). Computational
candesartan, and losartan on cancers in 15 trials enrolling 138,769 indi-
analyses of these drug–​disease networks are being increasingly con- viduals. J Hypertens, 29(4), 623–​635. PMID: 21358417.
ducted to predict and understand why current drugs work, fail, and Arbogast PG, and Ray WA. 2011. Performance of disease risk scores, pro-
result in deleterious outcomes, like cancer. In addition to mechanistic pensity scores, and traditional multivariable outcome regression in the
insight, systems pharmacology can potentially help guide drug–​cancer presence of multiple confounders. Am J Epidemiol, 174(5), 613–​620.
studies by identifying other medications and health conditions that PMID: 21749976.
 425

Pharmaceutical Drugs Other Than Hormones 425


Arts DG, De Keizer NF, and Scheffer GJ. 2002. Defining and improv- Brennan P, McKay J, Moore L, et al. 2009. Obesity and cancer: Mendelian ran-
ing data quality in medical registries:  a literature review, case study, domization approach utilizing the FTO genotype. Int J Epidemiol, 38(4),
and generic framework. J Am Med Inform Assoc, 9(6), 600–​ 611. 971–​975. PMCID: PMC2734066.
PMCID: PMC349377. Cardwell CR, Coleman HG, Murray LJ, Entschladen F, and Powe DG. 2013.
Assayag J, Pollak MN, and Azoulay L. 2014. Post-​diagnostic use of beta-​ Beta-​blocker usage and breast cancer survival: a nested case-​control study
blockers and the risk of death in patients with prostate cancer. Eur J within a UK clinical practice research datalink cohort. Int J Epidemiol,
Cancer, 50(16), 2838–​2845. PMID: 25204805. 42(6), 1852–​1861. PMID: 24536096.
Assimes TL, Elstein E, Langleben A, and Suissa S. 2008. Long-​term use of Cardwell CR, McMenamin UC, Hicks BM, et al. 2014a. Drugs affecting the
antihypertensive drugs and risk of cancer. Pharmacoepidemiol Drug Saf, renin-​angiotensin system and survival from cancer: a population based
17(11), 1039–​1049. PMID: 18780400. study of breast, colorectal and prostate cancer patient cohorts. BMC Med,
ASPREE Investigator Group. 2013. Study design of ASPirin in Reducing 12, 28. PMCID: PMC3926686.
Events in the Elderly (ASPREE): a randomized, controlled trial. Contemp Cardwell CR, Coleman HG, Murray LJ, O’Sullivan JM, and Powe DG. 2014b.
Clin Trials, 36(2), 555–​564. PMCID: PMC3919683. Beta-​blocker usage and prostate cancer survival: a nested case-​control
Austin PC. 2011. An introduction to propensity score methods for reducing the study in the UK Clinical Practice Research Datalink cohort. Cancer
effects of confounding in observational studies. Multivariate Behav Res, Epidemiol, 38(3), 279–​285. PMID: 24786858.
46(3), 399–​424. PMCID: PMC3144483. Cardwell CR, Flahavan EM, Hughes CM, et al. 2014c. Low-​dose aspirin and
Avorn J, and Schneeweiss S. 2009. Managing drug-​risk information:  what survival in men with prostate cancer: a study using the UK Clinical
to do with all those new numbers. N Engl J Med, 361(7), 647–​649. Practice Research Datalink. Cancer Causes Control, 25(1), 33–​ 43.
PMID: 19635948. PMID: 24310109.
Baandrup L, Faber MT, Christensen J, et  al. 2013. Nonsteroidal anti-​ Carstensen B, Witte DR, and Friis S. 2012. Cancer occurrence in Danish dia-
inflammatory drugs and risk of ovarian cancer:  systematic review and betic patients: duration and insulin effects. Diabetologia, 55(4), 948–​958.
meta-​ analysis of observational studies. Acta Obstet Gynecol Scand, PMID: 22120574.
92(3), 245–​255. PMID: 23240575. CenterWatch. 2016. FDA approved drugs 2016. [Internet] Available at: http://​
Bahl S, Cotterchio M, Kreiger N, and Klar N. 2004. Antidepressant medication www.centerwatch.com/​drug-​information/​fda-​approved-​drugs/​year/​2015.
use and non-​Hodgkin’s lymphoma risk: no association. Am J Epidemiol, Accessed on March 19, 2016.
160(6), 566–​575. PMID: 15353417. Chen L, Malone KE, and Li CI. 2015. Use of antihypertensive medications
Bangalore S, Kumar S, Kjeldsen SE, et al. 2011. Antihypertensive drugs and not associated with risk of contralateral breast cancer among women
risk of cancer:  network meta-​analyses and trial sequential analyses of diagnosed with estrogen receptor-​positive invasive breast cancer. Cancer
324,168 participants from randomised trials. Lancet Oncol, 12(1), 65–​82. Epidemiol Biomarkers Prev, 24(9), 1423–​1426. PMCID: PMC4560607.
PMID: 21123111. Chen Q, Zhang Q, Zhong F, et al. 2014. Association between calcium chan-
Bansal D, Undela K, D’Cruz S, and Schifano F. 2012. Statin use and risk of nel blockers and breast cancer: a meta-​analysis of observational studies.
prostate cancer:  a meta-​analysis of observational studies. PLoS One, Pharmacoepidemiol Drug Saf, 23(7), 711–​718. PMID: 24829113.
7(10), e46691. PMCID: PMC3462187. Chen S, Song X, Gao X, et al. 2011. Proton pump inhibitors and the risk of
Barron TI, Connolly RM, Sharp L, Bennett K, and Visvanathan K. 2011. Beta-​ colorectal cancer. J Clin Gastroenterol, 45(2), 177. PMID: 20697292.
blockers and breast cancer mortality:  a population-​based study. J Clin Chiang YY, Chen KB, Tsai TH, and Tsai WC. 2014. Lowered cancer risk with
Oncol, 29(19), 2635–​2644. PMID: 21632503. ACE inhibitors/​ARBs: a population-​based cohort study. J Clin Hypertens
Berger SI, and Iyengar R. 2011. Role of systems pharmacology in understand- (Greenwich), 16(1), 27–​33. PMID: 24304931.
ing drug adverse events. Wiley Interdiscip Rev Syst Biol Med, 3(2), 129–​ Chlebowski RT, and Col N. 2012. Bisphosphonates and breast cancer preven-
135. PMCID: PMC3057924. tion. Anticancer Agents Med Chem, 12(2), 144–​150. PMID: 21864227.
Bergmann MM, Calle EE, Mervis CA, et  al. 1998. Validity of self-​reported Cholesterol Treatment Trialists Collaboration. 2012. Lack of effect of lower-
cancers in a prospective cohort study in comparison with data from state ing LDL cholesterol on cancer:  meta-​analysis of individual data from
cancer registries. Am J Epidemiol, 147(6), 556–​562. PMID: 9521182. 175,000 people in 27 randomised trials of statin therapy. PLoS One, 7(1),
Bibbins-​Domingo K, US Preventive Services Task Force. 2016. Aspirin use e29849. PMCID: PMC3261846.
for the primary prevention of cardiovascular disease and colorectal can- Choueiri TK, Je Y, and Cho E. 2014. Analgesic use and the risk of kidney can-
cer:  U.S. Preventive Services Task Force Recommendation Statement. cer: a meta-​analysis of epidemiologic studies. Int J Cancer, 134(2), 384–​
Ann Intern Med, 164(12), 836–​845. PMID: 27064677. 396. PMCID: PMC3815746.
Biggar RJ, Andersen EW, Kroman N, Wohlfahrt J, and Melbye M. 2013. Breast Chow WA, Jiang C, and Guan M. 2009. Anti-​HIV drugs for cancer therapeu-
cancer in women using digoxin:  tumor characteristics and relapse risk. tics: back to the future? Lancet Oncol, 10(1), 61–​71. PMID: 19111246.
Breast Cancer Res, 15(1), R13. PMCID: PMC3672748. Chu E, and Sartorelli A. 2012. Cancer chemotherapy. In: Katzung B, Masters
Biggar RJ, Wohlfahrt J, Oudin A, Hjuler T, and Melbye M. 2011. Digoxin use S, and Trevor A (Eds.), Basic and clinical pharmacology (12th ed., pp.
and the risk of breast cancer in women. J Clin Oncol, 29(16), 2165–​2170. 949–​976). New York: McGraw-​Hill.
PMID: 21422417. Chubak J, Boudreau DM, Rulyak SJ, and Mandelson MT. 2011. Colorectal
Blumentals WA, Foulis PR, Schwartz SW, and Mason TJ. 2004. Does warfarin cancer risk in relation to antidepressant medication use. Int J Cancer,
therapy influence the risk of bladder cancer? Thromb Haemost, 91(4), 128(1), 227–​232. PMCID: PMC2962879.
801–​805. PMID: 15045143. Chubak J, Boudreau DM, Wirtz HS, McKnight B, and Weiss NS. 2013. Threats
Bochud M, and Rousson V. 2010. Usefulness of Mendelian randomization in to validity of nonrandomized studies of postdiagnosis exposures on can-
observational epidemiology. Int J Environ Res Public Health, 7(3), 711–​ cer recurrence and survival. J Natl Cancer Inst, 105(19), 1456–​1462.
728. PMCID: PMC2872313. PMCID: PMC3787908.
Boffetta P, and Islami F. 2013. The contribution of molecular epidemiology to Chubak J, Bowles EJ, Yu O, et  al. 2016a. Breast cancer recurrence in rela-
the identification of human carcinogens:  current status and future per- tion to antidepressant use. Cancer Causes Control, 27(1), 125–​ 136.
spectives. Ann Oncol, 24(4), 901–​908. PMID: 23136234. PMID: 26518198.
Bonovas S, Nikolopoulos G, and Bagos P. 2013. Bisphosphonate use and risk Chubak J, Pocobelli G, and Weiss NS. 2012. Tradeoffs between accuracy mea-
of colorectal cancer:  a systematic review and meta-​analysis. Br J Clin sures for electronic health care data algorithms. J Clin Epidemiol, 65(3),
Pharmacol, 76(3), 329–​337. PMCID: PMC3769661. 343–​349 e342. PMCID: PMC3264740.
Bordeleau L, and Gerstein HC. 2014. Response to Zanders et al. The association Chubak J, Whitlock EP, Williams SB, et al. 2016b. Aspirin for the prevention
of basal insulin glargine and/​or n-​3 fatty acids with incident cancers in of cancer incidence and mortality:  systematic evidence reviews for the
patients with dysglycemia. Diabetes Care, 37(10), e223. PMID: 25249683. U.S. Preventive Services Task Force. Ann Intern Med, 164(12), 814–​825.
Boudreau DM, Koehler E, Rulyak SJ, et al. 2008. Cardiovascular medication PMID: 27064482.
use and risk for colorectal cancer. Cancer Epidemiol Biomarkers Prev, Clezardin P. 2013. Mechanisms of action of bisphosphonates in oncology:  a
17(11), 3076–​3080. PMCID: PMC2675612. scientific concept evolving from antiresorptive to anticancer activities.
Boudreau DM, Yu O, Chubak J, et al. 2014. Comparative safety of cardiovas- Bonekey Rep, 2, 267. PMCID: PMC3722725.
cular medication use and breast cancer outcomes among women with Cogliano VJ, Baan R, Straif K, et  al. 2011. Preventable exposures associ-
early stage breast cancer. Breast Cancer Res Treat, 144(2), 405–​416. ated with human cancers. J Natl Cancer Inst, 103(24), 1827–​ 1839.
PMCID: PMC3988288. PMCID: PMC3243677.
Brayfield A. 2014. Martindale:  The complete drug reference (38th ed.). Coloma PM, Schuemie MJ, Trifiro G, et  al. 2011. Combining electronic
London: Pharmaceutical Press. healthcare databases in Europe to allow for large-​ scale drug safety
426

426 Part III: The Causes of Cancer


monitoring: the EU-​ADR Project. Pharmacoepidemiol Drug Saf, 20(1), Drake MT, Clarke BL, and Khosla S. 2008. Bisphosphonates: mechanism of
1–​11. PMID: 21182150. action and role in clinical practice. Mayo Clin Proc, 83(9), 1032–​1045.
Coogan PF, Strom BL, and Rosenberg L. 2009. Antidepressant use and colorec- PMCID: PMC2667901.
tal cancer risk. Pharmacoepidemiol Drug Saf, 18(11), 1111–​ 1114. Drew DA, Cao Y, and Chan AT. 2016. Aspirin and colorectal cancer: the prom-
PMCID: PMC2783290. ise of precision chemoprevention. Nat Rev Cancer, 16(3), 173–​ 186.
Corrao G, Scotti L, Bagnardi V, and Sega R. 2007. Hypertension, antihyperten- PMID: 26868177.
sive therapy and renal-​cell cancer: a meta-​analysis. Curr Drug Saf, 2(2), Dreyer N, and Velentgas P. 2012. Registries. In:  Strom B, Kimmel S, and
125–​133. PMID: 18690958. Hennessy S (Eds.), Pharmacoepidemiology (5th ed., pp. 331). Chichester,
Cronin-​Fenton DP, Riis AH, Lash TL, et  al. 2011. Antidepressant use and UK: Wiley-​Blackwell.
colorectal cancer risk: a Danish population-​based case-​control study. Br J Early Breast Cancer Trialists’ Collaborative G, Coleman R, Powles T, et  al.
Cancer, 104(1), 188–​192. PMCID: PMC3039807. 2015. Adjuvant bisphosphonate treatment in early breast cancer:  meta-​
Cui X, Xie Y, Chen M, et  al. 2012. Statin use and risk of pancreatic can- analyses of individual patient data from randomised trials. Lancet,
cer:  a meta-​analysis. Cancer Causes Control, 23(7), 1099–​ 1111. 386(10001), 1353–​1361. PMID: 26211824.
PMID: 22562222. Egan AG, Blind E, Dunder K, et al. 2014. Pancreatic safety of incretin-​based
Cui XJ, He Q, Zhang JM, et al. 2014. High-​dose aspirin consumption contrib- drugs:  FDA and EMA assessment. N Engl J Med, 370(9), 794–​797.
utes to decreased risk for pancreatic cancer in a systematic review and PMID: 24571751.
meta-​analysis. Pancreas, 43(1), 135–​140. PMID: 24263109. Elwood PC, Morgan G, Pickering JE, et  al. 2016. Aspirin in the treatment
Cummings SR, Duong T, Kenyon E, et al. 2002. Serum estradiol level and risk of cancer:  Reductions in metastatic spread and in mortality:  a system-
of breast cancer during treatment with raloxifene. JAMA, 287(2), 216–​ atic review and meta-​analyses of published studies. PLoS One, 11(4),
220. PMID: 11779264. e0152402. PMCID: PMC4838306.
Currier MB, and Nemeroff CB. 2014. Depression as a risk factor for can- European Medicines Agency (EMA). 2011. European Medicines Agency
cer:  from pathophysiological advances to treatment implications. Annu concludes that benefit-​ risk balance of angiotensin-​ II-​
receptor antag-
Rev Med, 65, 203–​221. PMID: 24215332. onists remains positive (pp. 1–​ 2). London:  European Medicines
Curtis LH, Weiner MG, Boudreau DM, et  al. 2012. Design considerations, Agency [Internet]. Available at:  http://​www.ema.europa.eu/​ema/​index.
architecture, and use of the Mini-​ Sentinel distributed data system. jsp?curl=pages/​news_​and_​events/​news/​2011/​10/​news_​detail_​001365.
Pharmacoepidemiol Drug Saf, 21(Suppl 1), 23–​31. PMID: 22262590. jsp&mid=WC0b01ac058004d5c1. Accessed on May 8, 2016.
Cuzick J, Otto F, Baron JA, et  al. 2009. Aspirin and non-​ steroidal anti-​ European Medicines Agency (EMA). 2014. Annual report 2013 (pp. 1–​64).
inflammatory drugs for cancer prevention:  an international consensus Available at:  http://​www.ema.europa.eu/​docs/​en_​GB/​document_​library/​
statement. Lancet Oncol, 10(5), 501–​507. PMID: 19410194. Annual_​report/​2014/​04/​WC500165986.pdf. Accessed July 11, 2016.
Cuzick J, Thorat MA, Bosetti C, et al. 2015. Estimates of benefits and harms of ENGAGE. 2012. European Network for Genetic and Genomic
prophylactic use of aspirin in the general population. Ann Oncol, 26(1), Epidemiology:  Final publishable summary report (pp. 5–​ 31).
47–​57. PMCID: PMC4269341. Available at:  http://​www.euengage.org/​Publishable%20Summary%20-​
Dai YN, Wang JH, Zhu JZ, Lin JQ, Yu CH, and Li YM. 2015. Angiotensin-​ %20ENGAGE_​Final_​Publishable_​Summary_​Report.pdf. Accessed July
converting enzyme inhibitors/​angiotensin receptor blockers therapy and 11, 2016.
colorectal cancer: a systematic review and meta-​analysis. Cancer Causes Eom CS, Park SM, and Cho KH. 2012. Use of antidepressants and the risk of
Control, 26(9),1245–​1255. PMID:26081426 breast cancer:  a meta-​analysis. Breast Cancer Res Treat, 136(3), 635–​
Dal Pan G, Lindquist M, and Gelperin K. 2012. Postmarketing spontane- 645. PMID: 23139055.
ous pharmacovigilance reporting systems. In:  Strom B, Kimmel S, Erson A, and Petty E. 2006. Molecular and genetic events in neoplastic
and Hennessy S (Eds.), Pharmacoepidemiology (5th ed., pp. 99–​117). transformation. In:  Schottenfeld D, and Fraumeni JJ (Eds.), Cancer
Chichester, UK: Wiley-​Blackwell. epidemiology and prevention (3rd ed., pp. 47–​64). New  York:  Oxford
Dalton SO, Poulsen AH, Nørgaard M, et  al. 2008. Tricyclic antidepres- University Press.
sants and non-​ Hodgkin lymphoma. Epidemiology, 19(4), 546–​ 549. Eslami L, and Nasseri-​Moghaddam S. 2013. Meta-​analyses:  does long-​term
PMID: 18552589. PPI use increase the risk of gastric premalignant lesions? Arch Iran Med,
Daniels NA, Chen YH, and Bent S. 2009. Antibiotic and anti-​inflammatory 16(8), 449–​458. PMID: 23906249.
use and the risk of prostate cancer. BMC Res Notes, 2, 57. Faillie JL. 2015. Indication bias or protopathic bias? Br J Clin Pharmacol,
PMCID: PMC2672937. 80(4), 779–​780. PMCID: PMC4594717.
Das D, Chilton AP, and Jankowski JA. 2009. Chemoprevention of oesophageal Fang JL, and Beland FA. 2009. Long-​term exposure to zidovudine delays
cancer and the AspECT trial. Recent Results Cancer Res, 181, 161–​169. cell cycle progression, induces apoptosis, and decreases telomer-
PMID: 19213566. ase activity in human hepatocytes. Toxicol Sci, 111(1), 120–​ 130.
Daugherty SE, Pfeiffer RM, Sigurdson AJ, et  al. 2011. Nonsteroidal antiin- PMCID: PMC2734309.
flammatory drugs and bladder cancer: a pooled analysis. Am J Epidemiol, FDA. 2011. Angiotensin receptor blockers (ARBs):  drug safety communica-
173(7), 721–​730. PMCID: PMC3105281. tion. [Internet] Available at:  http://​www.fda.gov/​Safety/​MedWatch/​
de Pedro M, Baeza S, Escudero MT, et al. 2015. Effect of COX-​2 inhibitors SafetyInformation/​SafetyAlertsforHumanMedicalProducts/​ucm219185.
and other non-​steroidal inflammatory drugs on breast cancer risk: a meta-​ htm. Accessed on March 20, 2016.
analysis. Breast Cancer Res Treat, 149(2), 525–​536. PMID: 25589172. FDA. 2015. FDA glossary of terms. [Internet] Available at:  http://​www.fda.
Dedrick RL, and Morrison PF. 1992. Carcinogenic potency of alkylating agents gov/​Drugs/​InformationOnDrugs/​ucm079436.htm. Accessed on March
in rodents and humans. Cancer Res, 52(9), 2464–​2467. PMID: 1568217. 5, 2016.
Dehmer SP, Maciosek MV, and Flottemesch TJ. 2015. Aspirin use to pre- Ferwana M, Firwana B, Hasan R, et al. 2013. Pioglitazone and risk of bladder
vent cardiovascular disease and colorectal cancer:  a decision analy- cancer: a meta-​analysis of controlled studies. Diabet Med, 30(9), 1026–​
sis. Technical report. (US Preventive Services Task Force Evidence 1032. PMID: 23350856.
Syntheses, Trans.) [Internet]. Available at:  http://​www.uspreventiveser- Flahavan EM, Sharp L, Bennett K, and Barron TI. 2014. A cohort study of
vicestaskforce.org/​Page/​Document/​decision-​analysis-​for-​aspirin/​aspirin-​ digoxin exposure and mortality in men with prostate cancer. BJU Int,
to-​prevent-​cardiovascular-​disease-​and-​cancer. Accessed on June 5, 2016. 113(2), 236–​245. PMID: 23937513.
Dehmer SP, Maciosek MV, Flottemesch TJ, et al. 2016. Aspirin for the primary Fleury I, Chevret S, Pfreundschuh M, et al. 2016. Rituximab and risk of sec-
prevention of cardiovascular disease and colorectal cancer:  a decision ond primary malignancies in patients with non-​Hodgkin lymphoma:  a
analysis for the U.S. Preventive Services Task Force. Ann Intern Med, systematic review and meta-​ analysis. Ann Oncol, 27(3), 390–​ 397.
164(12), 777–​786. PMID: 27064573. PMID: 26681685.
Dejgaard A, Lynggaard H, Råstam J, and Krogsgaard Thomsen M. 2009. Forsythe P, and Paterson S. 2014. Ciclosporin 10 years on: indications and effi-
No evidence of increased risk of malignancies in patients with diabe- cacy. Vet Rec, 174(Suppl 2), 13–​21. PMCID: PMC3995292.
tes treated with insulin detemir:  a meta-​analysis. Diabetologia, 52(12), Frick LR, and Rapanelli M. 2013. Antidepressants:  influence on cancer and
2507–​2512. PMID: 19838665. immunity? Life Sci, 92(10), 525–​532. PMID: 23369745.
Deva S, and Jameson M. 2012. Histamine type 2 receptor antagonists as adju- Friedman GD, Achacoso N, Fireman B, Habel LA. 2016. Statins and reduced
vant treatment for resected colorectal cancer. Cochrane Database Syst risk of liver cancer:  Evidence for confounding. J Natl Cancer Inst,
Rev, 8(8), CD007814. PMID: 22895966. 108(10), pii: djw109. PMID: 27381455.
Devore EE, Kim S, Ramin CA, et al. 2015. Antihypertensive medication use Friedman GD, Achacoso N, and Habel LA. 2015. Confounding by alcohol
and incident breast cancer in women. Breast Cancer Res Treat, 150(1), use: benzodiazepines and risk of liver cancer. Br J Clin Pharmacol, 79(5),
219–​229. PMCID: PMC4381798. 872–​873. PMCID: PMC4415724.
 427

Pharmaceutical Drugs Other Than Hormones 427


Friedman GD, Asgari MM, Warton EM, Chan J, Habel LA. 2012. Clinical Practice Research Datalink. Pharmacotherapy, 36(2), 187–​195.
Antihypertensive drugs and lip cancer in non-​ Hispanic whites. Arch PMID: 26846893.
Intern Med, 172(16), 1246–​1251. Halapy E, Kreiger N, Cotterchio M, and Sloan M. 2006. Benzodiazepines and
Friedman GD, Jiang SF, Udaltsova N et al. 2009a. Epidemiologic evaluation of risk for breast cancer. Ann Epidemiol, 16(8), 632–​636. PMID: 16406246.
pharmaceuticals with limited evidence of carcinogenicity. Int J Cancer, Hammer GP, du Prel JB, and Blettner M. 2009. Avoiding bias in observational
125(9), 2173–​2178. PMCID: PMC2759691. studies: part 8 in a series of articles on evaluation of scientific publica-
Friedman GD, Jiang SF, Udaltsova N et al. 2009b. Pharmaceuticals that cause tions. Dtsch Arztebl Int, 106(41), 664–​668. PMCID: PMC2780010.
mammary gland tumors in animals:  findings in women. Breast Cancer Hanahan D, and Weinberg RA. 2011. Hallmarks of cancer: the next generation.
Res Treat, 116(1), 187–​194. PMCID: PMC2695837. Cell, 144(5), 646–​674. PMID: 21376230.
Friedman GD, Udaltsova N, Chan J, Quesenberry CP, Jr., and Habel LA. 2009c. Haque R, Shi J, Schottinger JE, et al. 2016. Tamoxifen and antidepressant drug
Screening pharmaceuticals for possible carcinogenic effects: initial posi- interaction in a cohort of 16,887 breast cancer survivors. J Natl Cancer
tive results for drugs not previously screened. Cancer Causes Control, Inst, 108(3), pii: djv337. PMID: 26631176.
20(10), 1821–​1835. PMCID: PMC3010483. Harmark L, and van Grootheest AC. 2008. Pharmacovigilance: methods, recent
Friedman GD, Oestreicher N, Chan J, et  al. 2006. Antibiotics and risk of developments and future perspectives. Eur J Clin Pharmacol, 64(8), 743–​
breast cancer: up to 9 years of follow-​up of 2.1 million women. Cancer 752. PMID: 18523760.
Epidemiol Biomarkers Prev, 15(11), 2102–​2106. PMID: 17119034. Heart Protection Study Collaborative Group, Bulbulia R, Bowman L, et  al.
Friedman GD, Schwalbe J, Achacoso N, et  al. 2014. Antidepressants 2011. Effects on 11-​ year mortality and morbidity of lowering LDL
and testicular cancer. Cancer Causes Control, 25(2), 251–​ 258. cholesterol with simvastatin for about 5 years in 20,536 high-​risk indi-
PMCID: PMC3922298. viduals:  a randomised controlled trial. Lancet, 378(9808), 2013–​2020.
Friis S, Kesminiene A, Espina C et al. 2015a. European Code against Cancer PMCID: PMC3242163.
4th ed.:  Medical exposures, including hormone therapy, and cancer. Hejna M, Raderer M, and Zielinski CC. 1999. Inhibition of metastases by anti-
Cancer Epidemiol, 39(Suppl 1), S107–​119. PMID: 26390952. coagulants. J Natl Cancer Inst, 91(1), 22–​36. PMID: 9890167.
Friis S, and Olsen JH. 2006. Statin use and cancer risk:  an epidemiologic Hicks BM, Murray LJ, Hughes C, and Cardwell CR. 2015. Post-​diagnostic
review. Cancer Invest, 24(4), 413–​424. PMID: 16777695. oral bisphosphonate use and colorectal cancer mortality:  a population-​
Friis S, Riis AH, Erichsen R, Baron JA, and Sørensen HT. 2015b. Low-​dose based cohort study within the UK Clinical Practice Research Datalink. Br
aspirin or nonsteroidal anti-​inflammatory drug use and colorectal cancer J Cancer, 113(1), 123–​126. PMCID: PMC4647537.
risk:  a population-​based, case-​control study. Ann Intern Med, 163(5), Hicks BM, Murray LJ, Powe DG, Hughes CM, and Cardwell CR. 2013. Beta-​
347–​355. PMID: 26302241. blocker usage and colorectal cancer mortality: a nested case-​control study
Fryzek JP, Poulsen AH, Johnsen SP, et al. 2005. A cohort study of antihyper- in the UK Clinical Practice Research Datalink cohort. Ann Oncol, 24(12),
tensive treatments and risk of renal cell cancer. Br J Cancer, 92(7), 1302–​ 3100–​3106. PMID: 24050955.
1306. PMCID: PMC2361985. Hill S. 2010. Pharmacology. In: Tomlin M (Ed.), Pharmacology and pharma-
Ganz PA, Habel LA, Weltzien EK, Caan BJ, and Cole SW. 2011. Examining the cokinetics: a basic reader (1st ed., pp. 1–​11). London: Springer.
influence of beta blockers and ACE inhibitors on the risk for breast can- Hippisley-​Cox J, and Coupland C. 2010. Unintended effects of statins in men
cer recurrence: results from the LACE cohort. Breast Cancer Res Treat, and women in England and Wales: population based cohort study using
129(2), 549–​556. PMCID: PMC3145014. the QResearch database. BMJ, 340, c2197. PMCID: PMC2874131.
George AJ, Thomas WG, and Hannan RD. 2010. The renin-​angiotensin sys- Hochmuth F, Jochem M, and Schlattmann P. 2016. Meta-​analysis of aspirin use
tem and cancer: old dog, new tricks. Nat Rev Cancer, 10(11), 745–​759. and risk of lung cancer shows notable results. Eur J Cancer Prev, 25(4),
PMID: 20966920. 259–​268. PMID: 26067033.
Gold B, and Mirvish SS. 1977. N-​Nitroso derivatives of hydrochlorothiazide, Hoelder S, Clarke PA, and Workman P. 2012. Discovery of small molecule
niridazole, and tolbutamide. Toxicol Appl Pharmacol, 40(1), 131–​136. cancer drugs: successes, challenges and opportunities. Mol Oncol, 6(2),
PMID: 867427. 155–​176. PMCID: PMC3476506.
Gong J, Zhu L, Guo Z, et al. 2013. Use of thiopurines and risk of colorectal Holmes S, Griffith EJ, Musto G, and Minuk GY. 2013. Antihypertensive medi-
neoplasia in patients with inflammatory bowel diseases: a meta-​analysis. cations and survival in patients with cancer: a population-​based retrospec-
PLoS One, 8(11), e81487. PMCID: PMC3842949. tive cohort study. Cancer Epidemiol, 37(6), 881–​885. PMID: 24075077.
Gottlieb A, and Altman RB. 2014. Integrating systems biology sources Hu H, Xie Y, Yang G, Jian C, and Deng Y. 2014. Nonsteroidal anti-​inflammatory
illuminates drug action. Clin Pharmacol Ther, 95(6), 663–​ 669. drug use and the risk of melanoma: a meta-​analysis. Eur J Cancer Prev,
PMCID: PMC4029855. 23(1), 62–​68. PMID: 23549150.
Grimaldi-​Bensouda L, Klungel O, Kurz X, et al. 2016. Calcium channel block- Huang XZ, Gao P, Sun JX, et  al. 2015. Aspirin and nonsteroidal anti-​
ers and cancer: a risk analysis using the UK Clinical Practice Research inflammatory drugs after but not before diagnosis are associated with
Datalink (CPRD). BMJ Open, 6(1), e009147. PMCID: PMC4716173. improved breast cancer survival: a meta-​analysis. Cancer Causes Control,
Grimaldi-​Bensouda L, Rossignol M, Aubrun E, et al. 2012. Agreement between 26(4), 589–​600. PMID: 25701248.
patients’ self-​report and physicians’ prescriptions on nonsteroidal anti-​ IARC. 1996. Benzodiazepines and related compounds and phenytoin.
inflammatory drugs and other drugs used in musculoskeletal disorders: the Pharmaceuticals Monogr Eval Carcinog Risks Hum, 66 [Internet].
international Pharmacoepidemiologic General Research eXtension data- Available at:  http://​monographs.iarc.fr/​ENG/​Monographs/​vol66/​.
base. Pharmacoepidemiol Drug Saf, 21(7), 753–​759. PMID: 22315129. Accessed on March 4, 2016.
Grosse Y, Baan R, Straif K et al. 2009. A review of human carcinogens—​Part IARC. 2012. Pharmaceuticals Monogr Eval Carcinog Risks Hum, 100(Pt A),
A: pharmaceuticals. Lancet Oncol, 10(1), 13–​14. PMID: 19115512. 1–​401. PMCID: PMC4781347.
Grosse Y, Loomis D, Lauby-​ Secretan B, et  al. 2013. Carcinogenicity of IARC. 2015. Some drugs and herbal products. Monogr Eval Carcinog Risks
some drugs and herbal products. Lancet Oncol, 14(9), 807–​ 808. Hum, 108, 1–​438.
PMID: 24058961. Jager KJ, Zoccali C, Macleod A, and Dekker FW. 2008. Confounding: what it
Grossman J, and Mackenzie FJ. 2005. The randomized controlled trial: gold is and how to deal with it. Kidney Int, 73(3), 256–​260. PMID: 17978811.
standard, or merely standard? Perspect Biol Med, 48(4), 516–​ 534. Janghorbani M, Dehghani M, and Salehi-​Marzijarani M. 2012. Systematic
PMID: 16227664. review and meta-​analysis of insulin therapy and risk of cancer. Horm
Grytli HH, Fagerland MW, Fossa SD, and Tasken KA. 2014. Association Cancer, 3(4), 137–​146. PMID: 22528451.
between use of beta-​blockers and prostate cancer-​specific survival:  a Jepsen P, Johnsen SP, Gillman MW, and Sørensen HT. 2004. Interpretation of
cohort study of 3561 prostate cancer patients with high-​risk or metastatic observational studies. Heart, 90(8), 956–​960. PMCID: PMC1768356.
disease. Eur Urol, 65(3), 635–​641. PMID: 23351721. Jiang HY, Huang TB, Xu L, et al. 2015. Aspirin use and lung cancer risk: a pos-
Gutierrez-​Dalmau A, and Campistol JM. 2007. Immunosuppressive ther- sible relationship? Evidence from an updated meta-​analysis. PLoS One,
apy and malignancy in organ transplant recipients: a systematic review. 10(4), e0122962. PMCID: PMC4388842.
Drugs, 67(8), 1167–​1198. PMID: 17521218. Kaapu KJ, Ahti J, Tammela TL, Auvinen A, and Murtola TJ. 2015. Sotalol,
Hadji P, Coleman RE, Wilson C, et al. 2016. Adjuvant bisphosphonates in early but not digoxin is associated with decreased prostate cancer risk:  a
breast cancer: consensus guidance for clinical practice from a European population-​based case-​control study. Int J Cancer, 137(5), 1187–​1195.
Panel. Ann Oncol, 27(3), 379–​390. PMID: 26681681. PMID: 25656312.
Hagberg KW, Sahasrabuddhe VV, McGlynn KA, and Jick SS. 2016. Does Kannen V, Hintzsche H, Zanette DL, et  al. 2012. Antiproliferative effects
angiotensin-​converting enzyme inhibitor and beta-​ blocker use reduce of fluoxetine on colon cancer cells and in a colonic carcinogen mouse
the risk of primary liver cancer? A  case-​control study using the UK model. PLoS One, 7(11), e50043. PMCID: PMC3507893.
428

428 Part III: The Causes of Cancer


Kao CH, Sun LM, Su KP, et al. 2012. Benzodiazepine use possibly increases Martens EP, Pestman WR, de Boer A, Belitser SV, and Klungel OH. 2006.
cancer risk:  a population-​based retrospective cohort study in Taiwan. J Instrumental variables: application and limitations. Epidemiology, 17(3),
Clin Psychiatry, 73(4), e555–​560. PMID: 22579162. 260–​267. PMID: 16617274.
Karasneh RA, Murray LJ, McMenamin UC, Hughes CM, and Cardwell Matusewicz L, Meissner J, Toporkiewicz M, and Sikorski AF. 2015. The
CR. 2015. Digoxin use after diagnosis of breast cancer and survival:  a effect of statins on cancer cells: review. Tumour Biol, 36(7), 4889–​4904.
population-​based cohort study. Breast Cancer Res Treat, 151(3), 661–​ PMID: 26002574.
669. PMID: 25975954. Mauro V, Carette D, Pontier-​Bres R, et al. 2013. The anti-​mitotic drug griseo-
Kelloff GJ, Lippman SM, Dannenberg AJ, et al. 2006. Progress in chemopre- fulvin induces apoptosis of human germ cell tumor cells through a con-
vention drug development: the promise of molecular biomarkers for pre- nexin 43-​dependent molecular mechanism. Apoptosis, 18(4), 480–​491.
vention of intraepithelial neoplasia and cancer—​a plan to move forward. PMID: 23329179.
Clin Cancer Res, 12(12), 3661–​3697. PMID: 16778094. McMenamin UC, Murray LJ, Cantwell MM, and Hughes CM. 2012.
Khan N, Abbas AM, Lichtenstein GR, Loftus EV, Jr., and Bazzano LA. 2013. Angiotensin-​ converting enzyme inhibitors and angiotensin receptor
Risk of lymphoma in patients with ulcerative colitis treated with thiopu- blockers in cancer progression and survival: a systematic review. Cancer
rines: a nationwide retrospective cohort study. Gastroenterology, 145(5), Causes Control, 23(2), 221–​230. PMID: 22116540.
1007–​1015 e1003. PMID: 23891975. McCourt C, Coleman HG, Murray LJ, et  al. 2014. Beta-​blocker usage after
Khoury MJ, and Ioannidis JP. 2014. Medicine: big data meets public health. malignant melanoma diagnosis and survival:  a population-​based nested
Science, 346(6213), 1054–​1055. PMCID: PMC4684636. case-​control study. Br J Dermatol, 170(4), 930–​938. PMID: 24593055.
Kilkkinen A, Rissanen H, Klaukka T, et  al. 2008. Antibiotic use pre- McDonald E, Freedman DM, Alexander BH, et al. 2014. Prescription diuretic
dicts an increased risk of cancer. Int J Cancer, 123(9), 2152–​ 2155. use and risk of basal cell carcinoma in the nationwide U.S.  radiologic
PMID: 18704945. technologists cohort. Cancer Epidemiol Biomarkers Prev, 23(8), 1539–​
Kotlyar DS, Lewis JD, Beaugerie L, et al. 2015. Risk of lymphoma in patients 1545. PMCID: PMC4119543.
with inflammatory bowel disease treated with azathioprine and 6-​ McFarland MS, and Cripps R. 2010. Diabetes mellitus and increased risk of
mercaptopurine:  a meta-​analysis. Clin Gastroenterol Hepatol, 13(5), cancer: focus on metformin and the insulin analogs. Pharmacotherapy,
847–​858 e844; quiz e848–​850. PMID: 24879926. 30(11), 1159–​1178. PMID: 20973689.
Kripke DF, Langer RD, and Kline LE. 2012. Hypnotics’ association with McLaughlin JK, Blot WJ, and Fraumeni JF, Jr. 1988. Diuretics and renal cell
mortality or cancer: a matched cohort study. BMJ Open, 2(1), e000850. cancer. J Natl Cancer Inst, 80(5), 378. PMCID: PMC3357203.
PMCID: PMC3293137. Mendes E. 2015. Personalized cancer prevention: American Cancer Society’s
Laine L, Ahnen D, McClain C, Solcia E, and Walsh JH. 2000. Review arti- cutting-​edge cancer science. [Internet] Available at:  http://​www.cancer.
cle:  potential gastrointestinal effects of long-​ term acid suppression org/​research/​acsresearchupdates/​cancerprevention/​personalized-​cancer-​
with proton pump inhibitors. Aliment Pharmacol Ther, 14(6), 651–​668. prevention. Accessed on February 10, 2016.
PMID: 10848649. Menger L, Vacchelli E, Adjemian S, et  al. 2012. Cardiac glycosides exert
Langley R, Wilson R, Ring A, et al. 2013. Evidence for the use of aspirin as a treat- anticancer effects by inducing immunogenic cell death. Sci Transl Med,
ment for cancer and the planned Add-​Aspirin trial. Paper presented at the 4(143), 143ra199. PMID: 22814852.
10th NCRI Cancer Conference, The BT Convention Centre, Liverpool, UK. Mhaskar R, Redzepovic J, Wheatley K, et al. 2012. Bisphosphonates in mul-
Lee HK, Eom CS, Kwon YM, et al. 2012. Meta-​analysis: selective serotonin tiple myeloma:  a network meta-​analysis. Cochrane Database Syst Rev,
reuptake inhibitors and colon cancer. Eur J Gastroenterol Hepatol, 5(5), CD003188. PMID: 22592688.
24(10), 1153–​1157. PMID: 22735609. Monami M, Dicembrini I, and Mannucci E. 2014. Thiazolidinediones and can-
Lemeshow S, Sørensen HT, Phillips G et al. 2011. Beta-​blockers and survival cer: results of a meta-​analysis of randomized clinical trials. Acta Diabetol,
among Danish patients with malignant melanoma:  a population-​based 51(1), 91–​101. PMID: 23851465.
cohort study. Cancer Epidemiol Biomarkers Prev, 20(10), 2273–​2279. Monami M, Dicembrini I, Martelli D, and Mannucci E. 2011. Safety of dipep-
PMCID: PMC3652234. tidyl peptidase-​4 inhibitors: a meta-​analysis of randomized clinical trials.
Li CI, Daling JR, Tang MT et al. 2013. Use of antihypertensive medications and Curr Med Res Opin, 27(Suppl 3), 57–​64. PMID: 22106978.
breast cancer risk among women aged 55 to 74 years. JAMA Intern Med, Moorman PG, Grubber JM, Millikan RC, and Newman B. 2003.
173(17), 1629–​1637. PMCID: PMC4112594. Antidepressant medications and their association with invasive breast
Li W, Shi Q, Wang W, et al. 2014. Calcium channel blockers and risk of breast cancer and carcinoma in situ of the breast. Epidemiology, 14(3), 307–​314.
cancer:  a meta-​analysis of 17 observational studies. PLoS One, 9(9), PMID: 12859031.
e105801. PMCID: PMC4153551. Movahedi M, Bishop DT, Macrae F, et al. 2015. Obesity, aspirin, and risk of
Li WQ, Qureshi AA, Robinson KC, and Han J. 2014. Sildenafil use and colorectal cancer in carriers of hereditary colorectal cancer:  a prospec-
increased risk of incident melanoma in US men:  a prospective cohort tive investigation in the CAPP2 Study. J Clin Oncol, 33(31), 3591–​3597.
study. JAMA Intern Med, 174(6), 964–​970. PMCID: PMC4178948. PMID: 26282643.
Liao J, and Farmer JA. 2013. Aggressive statin therapy and the risk of malig- MRC Working Party. 1992. Medical Research Council trial of treatment of
nancy. Curr Atheroscler Rep, 15(4), 316. PMID: 23435673. hypertension in older adults: principal results. MRC Working Party. BMJ,
Liu Y, Chen JQ, Xie L, et al. 2014. Effect of aspirin and other non-​steroidal anti-​ 304(6824), 405–​412. PMCID: PMC1995577.
inflammatory drugs on prostate cancer incidence and mortality: a system- Muranushi C, Olsen CM, Green AC, and Pandeya N. 2016. Can oral nonste-
atic review and meta-​analysis. BMC Med, 12, 55. PMCID: PMC4021622. roidal antiinflammatory drugs play a role in the prevention of basal cell
Loeb S, Folkvaljon Y, Lambe M, et  al. 2015. Use of phosphodiesterase type carcinoma? A systematic review and metaanalysis. J Am Acad Dermatol,
5 inhibitors for erectile dysfunction and risk of malignant melanoma. 74(1), 108–​119 e101. PMID: 26433247.
JAMA, 313(24), 2449–​2455. PMID: 26103029. Muranushi C, Olsen CM, Pandeya N, and Green AC. 2015. Aspirin and non-
Lopez-​Olivo MA, Tayar JH, Martinez-​Lopez JA, et al. 2012. Risk of malignan- steroidal anti-​inflammatory drugs can prevent cutaneous squamous cell
cies in patients with rheumatoid arthritis treated with biologic therapy: a carcinoma:  a systematic review and meta-​analysis. J Invest Dermatol,
meta-​analysis. JAMA, 308(9), 898–​908. PMID: 22948700. 135(4), 975–​983. PMID: 25521453.
Lopez A, Mounier M, Bouvier AM, et al. 2014. Increased risk of acute myeloid Nan H, Morikawa T, Suuriniemi M, et  al. 2013. Aspirin use, 8q24 single
leukemias and myelodysplastic syndromes in patients who received thio- nucleotide polymorphism rs6983267, and colorectal cancer accord-
purine treatment for inflammatory bowel disease. Clin Gastroenterol ing to CTNNB1 alterations. J Natl Cancer Inst, 105(24), 1852–​1861.
Hepatol, 12(8), 1324–​1329. PMID: 24582568. PMCID: PMC3866156.
Lytras T, Nikolopoulos G, and Bonovas S. 2014. Statins and the risk of colorec- Newcomb PA, Passarelli MN, Phipps AI, et al. 2015. Oral bisphosphonate use
tal cancer: an updated systematic review and meta-​analysis of 40 studies. and risk of postmenopausal endometrial cancer. J Clin Oncol, 33(10),
World J Gastroenterol, 20(7), 1858–​1870. PMCID: PMC3930985. 1186–​1190. PMCID: PMC4372855.
Ma Z, Wang W, Jin G, Chu P, and Li H. 2014. Effect of statins on gastric cancer Numbere B, Fleming KM, Walker A, and Card TR. 2017. Adrenergic blockers
incidence:  a meta-​analysis of case control studies. J Cancer Res Ther, and the risk for common solid cancers: a case-​control study. Eur J Cancer
10(4), 859–​865. PMID: 25579519. Prev. 26(1), 86–93. PMID: 26649549.
Mansourian M, Haghjooy-​Javanmard S, Eshraghi A, et al. 2016. Statin use and Nyboe Andersen N, Pasternak B, Basit S, et  al. 2014. Association between
risk of breast cancer recurrence and death: a systematic review and meta-​ tumor necrosis factor-​alpha antagonists and risk of cancer in patients
analysis of observational Studies. J Pharm Pharm Sci, 19(1), 72–​81. with inflammatory bowel disease. JAMA, 311(23), 2406–​ 2413.
PMID: 27096694. PMID: 24938563.
Maron DJ, Fazio S, and Linton MF. 2000. Current perspectives on statins. O’Rorke MA, Murray LJ, Hughes CM, Cantwell MM, and Cardwell CR.
Circulation, 101(2), 207–​213. PMID: 10637210. 2015. The effect of warfarin therapy on breast, colorectal, lung, and
 429

Pharmaceutical Drugs Other Than Hormones 429


prostate cancer survival:  a population-​ based cohort study using the Ross TR, Ng D, Brown JS, et al. 2014. The HMO Research Network Virtual
Clinical Practice Research Datalink. Cancer Causes Control, 26(3), 355–​ Data Warehouse: a public data model to support collaboration. EGEMS
366. PMID: 25534917. (Wash DC), 2(1), 1049. PMCID: PMC4371424.
Ogino S, Fuchs CS, and Giovannucci E. 2012. How many molecular subtypes? Rossebo AB, Pedersen TR, Boman K, et  al. 2008. Intensive lipid lowering
Implications of the unique tumor principle in personalized medicine. with simvastatin and ezetimibe in aortic stenosis. N Engl J Med, 359(13),
Expert Rev Mol Diagn, 12(6), 621–​628. PMCID: PMC3492839. 1343–​1356. PMID: 18765433.
Olsen JH, Møller T, Anderson H, et  al. 2009. Lifelong cancer incidence in Rossouw JE, Anderson GL, Prentice RL, et  al. 2002. Risks and benefits of
47,697 patients treated for childhood cancer in the Nordic countries. J estrogen plus progestin in healthy postmenopausal women:  principal
Natl Cancer Inst, 101(11), 806–​813. PMID: 19470947. results from the Women’s Health Initiative randomized controlled trial.
Pahor M, Guralnik JM, Ferrucci L, et al. 1996. Calcium-​channel blockade and JAMA, 288(3), 321–​333. PMID: 12117397.
incidence of cancer in aged populations. Lancet, 348(9026), 493–​497. Rothwell PM, Fowkes FG, Belch JF, et al. 2011. Effect of daily aspirin on long-​
PMID: 8757150. term risk of death due to cancer: analysis of individual patient data from
Panda D, Rathinasamy K, Santra MK, and Wilson L. 2005. Kinetic suppres- randomised trials. Lancet, 377(9759), 31–​41. PMID: 21144578.
sion of microtubule dynamic instability by griseofulvin: implications for Rothwell PM, Price JF, Fowkes FG, et al. 2012a. Short-​term effects of daily
its possible use in the treatment of cancer. Proc Natl Acad Sci U S A, aspirin on cancer incidence, mortality, and non-​vascular death: analysis
102(28), 9878–​9883. PMCID: PMC1174985. of the time course of risks and benefits in 51 randomised controlled trials.
Pasquier E, Ciccolini J, Carre M, et  al. 2011. Propranolol potentiates the Lancet, 379(9826), 1602–​1612. PMID: 22440946.
anti-​angiogenic effects and anti-​ tumor efficacy of chemotherapy Rothwell PM, Wilson M, Price JF, et al. 2012b. Effect of daily aspirin on risk
agents:  implication in breast cancer treatment. Oncotarget, 2(10), 797–​ of cancer metastasis: a study of incident cancers during randomised con-
809. PMCID: PMC3248157. trolled trials. Lancet, 379(9826), 1591–​1601. PMID: 22440947.
Pedersen EG, Pottegård A, Hallas J, et al. 2014. Risk of non-​melanoma skin Sacks FM, Pfeffer MA, Moye LA, et  al. 1996. The effect of pravastatin on
cancer in myasthenia patients treated with azathioprine. Eur J Neurol, coronary events after myocardial infarction in patients with average cho-
21(3), 454–​458. PMID: 24330255. lesterol levels. Cholesterol and Recurrent Events Trial investigators. N
Peto R, Emberson J, Landray M, et  al. 2008. Analyses of cancer data Engl J Med, 335(14), 1001–​1009. PMID: 8801446.
from three ezetimibe trials. N Engl J Med, 359(13), 1357–​ 1366. Sadowska A, Kretowski R, Szynaka B, Cechowska-​Pasko M, and Car H. 2013a.
PMID: 18765432. Metronidazole decreases viability of DLD-​1 colorectal cancer cell line.
Platz EA, Yegnasubramanian S, Liu JO, et al. 2011. A novel two-​stage, trans- Cancer Biother Radiopharm, 28(8), 615–​622. PMCID: PMC3777553.
disciplinary study identifies digoxin as a possible drug for prostate cancer Sadowska A, Prokopiuk S, Miltyk W, et al. 2013b. Metronidazole affects breast
treatment. Cancer Discov, 1(1), 68–​77. PMCID: PMC3227223. cancer cell lines. Adv Med Sci, 58(1), 90–​95. PMID: 23640948.
Pottegård A, Friis S, Andersen M, and Hallas J. 2013a. Use of benzodiazepines Sahin IH, Hassan MM, and Garrett CR. 2014. Impact of non-​steroidal anti-​
or benzodiazepine related drugs and the risk of cancer:  a population-​ inflammatory drugs on gastrointestinal cancers: current state-​of-​the sci-
based case-​ control study. Br J Clin Pharmacol, 75(5), 1356–​ 1364. ence. Cancer Lett, 345(2), 249–​257. PMID: 24021750.
PMCID: PMC3635606. Salas M, Hofman A, and Stricker BH. 1999. Confounding by indication:  an
Pottegård A, Friis S, and Hallas J. 2013b. Cancer risk in long-​term users of vita- example of variation in the use of epidemiologic terminology. Am J
min K antagonists: a population-​based case-​control study. Int J Cancer, Epidemiol, 149(11), 981–​983. PMID: 10355372.
132(11), 2606–​2612. PMID: 23065946. Saltzman BS, Weiss NS, Sieh W, et  al. 2013. Use of antihypertensive medi-
Pottegård A, Friis S, Christensen RD, Habel LA, Gagne JJ, Hallas J. 2016a. cations and breast cancer risk. Cancer Causes Control, 24(2), 365–​371.
Identification of associations between prescribed medications and can- PMID: 23224328.
cer: A nationwide screening study. Ebio Medicine, 7, 73–79. PMID: Schmidt SA, Schmidt M, Mehnert F, Lemeshow S, and Sørensen HT. 2015. Use
27322460. of antihypertensive drugs and risk of skin cancer. J Eur Acad Dermatol
Pottegård A, García Rodríguez LA, Rasmussen L, et al. 2016b. Use of tricyclic Venereol, 29(8), 1545–​1554. PMID: 25640031.
antidepressants and risk of glioma: a nationwide case-​control study. Br J Schneeweiss S. 2006. Sensitivity analysis and external adjustment for unmea-
Cancer, 114(11): 1265–​1268. PMID: 27115466. sured confounders in epidemiologic database studies of therapeutics.
Rama I, and Grinyo JM. 2010. Malignancy after renal transplantation:  the Pharmacoepidemiol Drug Saf, 15(5), 291–​303. PMID: 16447304.
role of immunosuppression. Nat Rev Nephrol, 6(9), 511–​ 519. Schulman S, Rhedin AS, Lindmarker P, et  al. 1995. A comparison of six
PMID: 20736984. weeks with six months of oral anticoagulant therapy after a first episode
Rathinasamy K, Jindal B, Asthana J, et  al. 2010. Griseofulvin stabilizes of venous thromboembolism. Duration of Anticoagulation Trial Study
microtubule dynamics, activates p53 and inhibits the proliferation of Group. N Engl J Med, 332(25), 1661–​1665. PMID: 7760866.
MCF-​7 cells synergistically with vinblastine. BMC Cancer, 10, 213. Scosyrev E, Tobis S, Donsky H, et  al. 2013. Statin use and the risk of bio-
PMCID: PMC2885362. chemical recurrence of prostate cancer after definitive local therapy:  a
Raval AD, Thakker D, Negi H, Vyas A, and Salkini MW. 2016. Association meta-​analysis of eight cohort studies. BJU Int, 111(3 Pt B), E71–​77.
between statins and clinical outcomes among men with prostate cancer: a PMID: 23017100.
systematic review and meta-​ analysis. Prostate Cancer Prostatic Dis, Sergentanis TN, Zagouri F, and Zografos GC. 2010. Is antibiotic use a risk
19(2), 222. PMID: 26857024. factor for breast cancer? A meta-​analysis. Pharmacoepidemiol Drug Saf,
Ray WA. 2003. Evaluating medication effects outside of clinical trials:  new-​ 19(11), 1101–​1107. PMID: 20845408.
user designs. Am J Epidemiol, 158(9), 915–​920. PMID: 14585769. Setoguchi S, Avorn J, and Schneeweiss S. 2005. Statins and the risk of colorec-
Renehan AG, Zwahlen M, and Egger M. 2015. Adiposity and cancer risk: new tal cancer. N Engl J Med, 353(9), 952–​954. PMID: 16136709.
mechanistic insights from epidemiology. Nat Rev Cancer, 15(8), 484–​ Sharpe CR, Collet JP, Belzile E, Hanley JA, and Boivin JF. 2002. The effects of
498. PMID: 26205341. tricyclic antidepressants on breast cancer risk. Br J Cancer, 86(1), 92–​97.
Rennert G, Rennert HS, Pinchev M, and Lavie O. 2014. The effect of bisphos- PMID: PMC2746543.
phonates on the risk of endometrial and ovarian malignancies. Gynecol Shelton E, Laharie D, Scott FI, et  al. 2016. Cancer recurrence following
Oncol, 133(2), 309–​313. PMID: 24556062. immune-​suppressive therapies in patients with immune-​mediated dis-
Richards M. 2010. Extent and causes of international variation in drug eases: a systematic review and meta-​analysis. Gastroenterology, 151(1),
usage: a report for the Secretary of State for Health. [Internet]. Available 97–​109. PMID: 27039969.
at:  https://​www.gov.uk/​government/​uploads/​system/​uploads/​attachment_​ Shepherd J, Blauw GJ, Murphy MB, et  al. 2002. PROspective Study of
data/​file/​216249/​dh_​117977.pdf Accessed on April 5, 2016. Pravastatin in the Elderly at Risk. Pravastatin in elderly individuals at risk
Robertson DJ, Burke CA, Schwender BJ, et al. 2005. Histamine receptor antag- of vascular disease (PROSPER):  a randomised controlled trial. Lancet,
onists and incident colorectal adenomas. Aliment Pharmacol Ther, 22(2), 360(9346), 1623–​1630. PMID: 12457784.
123–​128. PMID: 16011670. Shrank WH, Patrick AR, and Brookhart MA. 2011. Healthy user and related
Robins JM, Hernan MA, and Brumback B. 2000. Marginal structural models biases in observational studies of preventive interventions: a primer for
and causal inference in epidemiology. Epidemiology, 11(5), 550–​560. physicians. J Gen Intern Med, 26(5), 546–​550. PMCID: PMC3077477.
PMID: 10955408. Signorello LB, McLaughlin JK, Lipworth L, et al. 2002. Confounding by indi-
Rosenberg L, Palmer JR, Zauber AG, et al. 1995. Relation of benzodiazepine cation in epidemiologic studies of commonly used analgesics. Am J Ther,
use to the risk of selected cancers: breast, large bowel, malignant mela- 9(3), 199–​205. PMID: 11941379.
noma, lung, endometrium, ovary, non-​ Hodgkin’s lymphoma, testis, Sill H, Olipitz W, Zebisch A, Schulz E, and Wolfler A. 2011. Therapy-​related
Hodgkin’s disease, thyroid, and liver. Am J Epidemiol, 141(12), 1153–​ myeloid neoplasms:  pathobiology and clinical characteristics. Br J
1160. PMID: 7771453. Pharmacol, 162(4), 792–​805. PMCID: PMC3042191.
430

430 Part III: The Causes of Cancer


Simon MS, Desai P, Wallace R, et  al. 2016. Prospective analysis of associa- Tamim HM, Mahmud S, Hanley JA, et al. 2008. Antidepressants and risk of
tion between statins and pancreatic cancer risk in the Women’s Health prostate cancer:  a nested case-​control study. Prostate Cancer Prostatic
Initiative. Cancer Causes Control, 27(3), 415–​423. PMID: 26857832. Dis, 11(1), 53–​60. PMID: 17684479.
Singh A, and Bangalore S. 2012. Which, if any, antihypertensive agents cause Tan P, Wei S, Tang Z, et al. 2016. LDL-​lowering therapy and the risk of prostate
cancer? Curr Opin Cardiol, 27(4), 374–​380. PMID: 22565138. cancer: a meta-​analysis of 6 randomized controlled trials and 36 observa-
Singh JA, Christensen R, Wells GA, et  al. 2009. A network meta-​analysis tional studies. Sci Rep, 6, 24521. PMCID: PMC4830970.
of randomized controlled trials of biologics for rheumatoid arthritis:  a Tanaka S, Tanaka S, and Kawakami K. 2015. Methodological issues in observa-
Cochrane overview. CMAJ, 181(11), 787–​796. PMCID: PMC2780484. tional studies and non-​randomized controlled trials in oncology in the era
Singh S, Garg SK, Singh PP, Iyer PG, and El-​Serag HB. 2014. Acid-​suppressive of big data. Jpn J Clin Oncol, 45(4), 323–​327. PMID: 25589456.
medications and risk of oesophageal adenocarcinoma in patients with Tang X, Yang L, He Z, and Liu J. 2012. Insulin glargine and cancer risk in
Barrett’s oesophagus: a systematic review and meta-​analysis. Gut, 63(8), patients with diabetes:  a meta-​ analysis. PLoS One, 7(12), e51814.
1229–​1237. PMCID: PMC4199831. PMCID: PMC3526637.
Singh S, Singh AG, Singh PP, Murad MH, and Iyer PG. 2013b. Statins are Thakkar B, Aronis KN, Vamvini MT, Shields K, and Mantzoros CS. 2013.
associated with reduced risk of esophageal cancer, particularly in patients Metformin and sulfonylureas in relation to cancer risk in type II dia-
with Barrett’s esophagus:  a systematic review and meta-​analysis. Clin betes patients: a meta-​analysis using primary data of published studies.
Gastroenterol Hepatol, 11(6), 620–​629. PMCID: PMC3660516. Metabolism, 62(7), 922–​934. PMID: 23419783.
Singh S, Singh PP, Singh AG, et  al. 2013d. Anti-​diabetic medications and Thiele M, Albillos A, Abazi R, et  al. 2015. Non-​selective beta-​blockers may
risk of pancreatic cancer in patients with diabetes mellitus: a systematic reduce risk of hepatocellular carcinoma: a meta-​analysis of randomized
review and meta-​analysis. Am J Gastroenterol, 108(4), 510–​519; quiz trials. Liver Int, 35(8), 2009–​2016. PMID: 25581713.
520. PMID: 23399556. Thomsen RW, Farkas DK, Friis S, et  al. 2013a. Endocarditis and risk of
Singh S, Singh PP, Singh AG, Murad MH, and Sanchez W. 2013a. Statins cancer:  a Danish nationwide cohort study. Am J Med, 126(1), 58–​67.
are associated with a reduced risk of hepatocellular cancer:  a system- PMID: 23260503.
atic review and meta-​ analysis. Gastroenterology, 144(2), 323–​ 332. Thomsen RW, Nielsen RB, Nørgaard M, et al. 2013b. Lifestyle profile among
PMID: 23063971. statin users. Epidemiology, 24(4), 619–​620. PMID: 23732738.
Singh S, Singh PP, Singh AG, Murad MH, and Sanchez W. 2013c. Anti-​diabetic Thun MJ, Jacobs EJ, and Patrono C. 2012. The role of aspirin in cancer preven-
medications and the risk of hepatocellular cancer:  a systematic review tion. Nat Rev Clin Oncol, 9(5), 259–​267. PMID: 22473097.
and meta-​analysis. Am J Gastroenterol, 108(6), 881–​ 891; quiz 892. Toh S, Rodriguez LA, and Hernandez-​Diaz S. 2007. Use of antidepressants
PMID: 23381014. and risk of lung cancer. Cancer Causes Control, 18(10), 1055–​1064.
Sipahi I, Debanne SM, Rowland DY, Simon DI, and Fang JC. 2010. Angiotensin-​ PMID: 17682831.
receptor blockade and risk of cancer: meta-​analysis of randomised con- Toh S, Shetterly S, Powers JD, and Arterburn D. 2014. Privacy-​preserving ana-
trolled trials. Lancet Oncol, 11(7), 627–​636. PMCID: PMC4070221. lytic methods for multisite comparative effectiveness and patient-​centered
Sjöberg T, García Rodríguez LA, and Lindblad M. 2007. Angiotensin-​ outcomes research. Med Care, 52(7), 664–​668. PMID: 24926715.
converting enzyme inhibitors and risk of esophageal and gastric cancer: a Trabert B, Ness RB, Lo-​Ciganic WH, et  al. 2014. Aspirin, nonaspirin non-
nested case-​control study. Clin Gastroenterol Hepatol, 5(10):1160–​1166. steroidal anti-​ inflammatory drug, and acetaminophen use and risk
PMID: 17916544 of invasive epithelial ovarian cancer:  a pooled analysis in the Ovarian
Song H, Zhu J, and Lu D. 2014. Long-​term proton pump inhibitor (PPI) use and Cancer Association Consortium. J Natl Cancer Inst, 106(2), djt431.
the development of gastric pre-​malignant lesions. Cochrane Database PMCID: PMC3924755.
Syst Rev, 12(12), CD010623. PMID: 25464111. Travis LB, Curtis RE, Glimelius B, et al. 1995. Bladder and kidney cancer fol-
Stafford RS, MacDonald EA, and Finkelstein SN. 2001. National patterns of lowing cyclophosphamide therapy for non-​Hodgkin’s lymphoma. J Natl
medication treatment for depression, 1987 to 2001. Prim Care Companion Cancer Inst, 87(7), 524–​530. PMID: 7707439.
J Clin Psychiatry, 3(6), 232–​235. PMCID: PMC181191. Travis LB, Demark Wahnefried W, Allan JM, Wood ME, and Ng AK. 2013.
Stenkvist B. 1999. Is digitalis a therapy for breast carcinoma? Oncol Rep, 6(3), Aetiology, genetics and prevention of secondary neoplasms in adult can-
493–​496. PMID: 10203580. cer survivors. Nat Rev Clin Oncol, 10(5), 289–​301. PMID: 23529000.
Stenkvist B, Bengtsson E, Eriksson O, et  al. 1979. Cardiac glycosides and Undela K, Srikanth V, and Bansal D. 2012. Statin use and risk of breast can-
breast cancer. Lancet, 1(8115), 563. PMID: 85158. cer:  a meta-​analysis of observational studies. Breast Cancer Res Treat,
Stockbruegger RW. 1985. Bacterial overgrowth as a consequence of reduced 135(1), 261–​269. PMID: 22806241.
gastric acidity. Scand J Gastroenterol Suppl, 111, 7–​16. PMID: 2861652. Usman MW, Luo F, Cheng H, Zhao JJ, and Liu P. 2015. Chemopreventive
Strom B. 2005. Overview of automated databases. In:  Strom B (Ed.), effects of aspirin at a glance. Biochim Biophys Acta, 1855(2), 254–​263.
Pharmacoepidemiology (4th ed., pp. 217–​222). Chichester, UK:  John PMID: 25842298.
Wiley & Sons. Vale CL, Burdett S, Rydzewska LH, et  al. 2016. Addition of docetaxel or
Strom B, Kimmel S, and Hennessy S. 2012. Part III:  Sources of data for bisphosphonates to standard of care in men with localised or meta-
pharmacoepidemiologic studies. In: Strom B, Kimmel S, and Hennessy static, hormone-​ sensitive prostate cancer:  a systematic review and
S (Eds.), Pharmacoepidemiology (5th ed., p.  135). Chichester, meta-​ analyses of aggregate data. Lancet Oncol, 17(2), 243–​ 256.
UK: Wiley-​Blackwell. PMCID: PMC4737894.
Suissa S, and Azoulay L. 2012. Metformin and the risk of cancer: time-​related Verdoodt F, Friis S, Dehlendorff C, Albieri V, and Kjær SK. 2016. Non-​
biases in observational studies. Diabetes Care, 35(12), 2665–​ 2673. steroidal anti-​inflammatory drug use and risk of endometrial cancer:  a
PMCID: PMC3507580. systematic review and meta-​analysis of observational studies. Gynecol
Swerdlow AJ, Higgins CD, Smith P, et al. 2011. Second cancer risk after che- Oncol, 140(2), 352–​358. PMID: 26701413.
motherapy for Hodgkin’s lymphoma: a collaborative British cohort study. Verduijn M, Siegerink B, Jager KJ, Zoccali C, and Dekker FW. 2010. Mendelian
J Clin Oncol, 29(31), 4096–​4104. PMID: 21969511. randomization: use of genetics to enable causal inference in observational
Tagalakis V, and Tamim H. 2010. The effect of warfarin use on clinical stage studies. Nephrol Dial Transplant, 25(5), 1394–​1398. PMID: 20190244.
and histological grade of prostate cancer. Pharmacoepidemiol Drug Saf, Vigneri P, Frasca F, Sciacca L, Pandini G, and Vigneri R. 2009. Diabetes and
19(5), 436–​439. PMID: 20437456. cancer. Endocr Relat Cancer, 16(4), 1103–​1123. PMID: 19620249.
Tagalakis V, Tamim H, Blostein M, et  al. 2007. Use of warfarin and risk of Walker AJ, Card T, Bates TE, and Muir K. 2011. Tricyclic antidepressants and
urogenital cancer: a population-​based, nested case-​control study. Lancet the incidence of certain cancers: a study using the GPRD. Br J Cancer,
Oncol, 8(5), 395–​402. PMID: 17466896. 104(1), 193–​197. PMCID: PMC3039809.
Tagalakis V, Tamim H, Blostein M, Hanley JA, and Kahn SR. 2013. Risk Walther T, Menrad A, Orzechowski HD, et al. 2003. Differential regulation of
of prostate cancer death in long-​term users of warfarin:  a population-​ in vivo angiogenesis by angiotensin II receptors. FASEB J, 17(14), 2061–​
based case-​control study. Cancer Causes Control, 24(6), 1079–​1085. 2067. PMID: 14597675.
PMID: 23494728. Wang D, and Dubois RN. 2004. Cyclooxygenase-​2: a potential target in breast
Taliani MR, Agnelli G, Prandoni P, et al. 2003. Incidence of cancer after a first cancer. Semin Oncol, 31(1 Suppl 3), 64–​73. PMID: 15052544.
episode of idiopathic venous thromboembolism treated with 3  months Wang D, and Dubois RN. 2006. Prostaglandins and cancer. Gut, 55(1), 115–​
or 1 year of oral anticoagulation. J Thromb Haemost, 1(8), 1730–​1733. 122. PMCID: PMC1856377.
PMID: 12911585. Wang HM, Liao ZX, Komaki R, et al. 2013. Improved survival outcomes with
Tamim HM, Hajeer AH, Boivin JF, and Collet JP. 2010. Association between the incidental use of beta-​blockers among patients with non-​small-​cell
antibiotic use and risk of prostate cancer. Int J Cancer, 127(4), 952–​960. lung cancer treated with definitive radiation therapy. Ann Oncol, 24(5),
PMID: 20039321. 1312–​1319. PMCID: PMC3629895.
 431

Pharmaceutical Drugs Other Than Hormones 431


Watkins JL, Thaker PH, Nick AM, et  al. 2015. Clinical impact of selective Yi X, Jia W, Jin Y, and Zhen S. 2014. Statin use is associated with reduced risk
and nonselective beta-​blockers on survival in patients with ovarian cancer. of haematological malignancies:  evidence from a meta-​analysis. PLoS
Cancer, 121(19), 3444–​3451. PMCID: PMC4575637. One, 9(1), e87019. PMCID: PMC3909054.
Weiss NS. 2003. Adjusting for screening history in epidemiologic studies of Yoon C, Yang HS, Jeon I, Chang Y, and Park SM. 2011. Use of angiotensin-​
cancer: why, when, and how to do it. Am J Epidemiol, 157(11), 957–​961. converting-​enzyme inhibitors or angiotensin-​receptor blockers and cancer
PMID: 12777356. risk:  a meta-​analysis of observational studies. CMAJ, 183(14), E1073–​
WHO. (2015). WHO Collaborating Centre for Drug Statistics Methodology, Oslo. 1084. PMCID: PMC3185099.
Guidelines for ATC classification and DDD assignment. [Internet]. Available Yoshida K, Solomon DH, and Kim SC. 2015. Active-​comparator design and
at:  http://​www.whocc.no/​atc_​ddd_​publications/​guidelines/​. Accessed on new-​user design in observational studies. Nat Rev Rheumatol, 11(7), 437–​
June 5, 2016. 441. PMCID: PMC4486631.
Wong AK, Kerkoutian S, Said J, Rashidi H, and Pullarkat ST. 2012. Risk of Yoshiji H, Kuriyama S, Kawata M, et al. 2001. The angiotensin-​I-​converting
lymphoma in patients receiving antitumor necrosis factor therapy: a meta-​ enzyme inhibitor perindopril suppresses tumor growth and angiogen-
analysis of published randomized controlled studies. Clin Rheumatol, esis: possible role of the vascular endothelial growth factor. Clin Cancer
31(4), 631–​636. PMID: 22147207. Res, 7(4), 1073–​1078. PMID: 11309359.
Wright E, Schofield PT, and Molokhia M. 2015. Bisphosphonates and Yu O, Eberg M, Benayoun S, et al. 2014. Use of statins and the risk of death in
evidence for association with esophageal and gastric cancer:  a sys- patients with prostate cancer. J Clin Oncol, 32(1), 5–​11. PMID: 24190110.
tematic review and meta-​ analysis. BMJ Open, 5(12), e007133. Yuan ZR, and Shi Y. 2008. Chloramphenicol induces abnormal differentiation
PMCID: PMC4680000. and inhibits apoptosis in activated T cells. Cancer Res, 68(12), 4875–​
Wright JL, Hansten PD, and Stanford JL. 2014. Is digoxin use for cardiovas- 4881. PMID: 18559535.
cular disease associated with risk of prostate cancer? Prostate, 74(1), 97–​ Zhang D, Xiong B, Li X, Xu T, and Yu M. 2013. Meta-​analysis: serious adverse
102. PMCID: PMC3935420. events in Crohn’s disease patients treated with TNF-​alpha inhibitors.
Wu JW, Boudreau DM, Park Y, Simonds NI, and Freedman AN. 2014. Hepatogastroenterology, 60(126), 1333–​1342. PMID: 23340167.
Commonly used diabetes and cardiovascular medications and cancer Zhang H, Jiang D, and Li X. 2013. Use of nonsteroidal anti-​inflammatory drugs
recurrence and cancer-​specific mortality: a review of the literature. Expert and bladder cancer risk: a meta-​analysis of epidemiologic studies. PLoS
Opin Drug Saf, 13(8), 1071–​1099. PMID: 24999107. One, 8(7), e70008. PMCID: PMC3716767.
Wysowski DK. 2009. Reports of esophageal cancer with oral bisphosphonate Zhang XL, Geng J, Zhang XP, et  al. 2013. Statin use and risk of blad-
use. N Engl J Med, 360(1), 89–​90. PMID: 19118315. der cancer:  a meta-​analysis. Cancer Causes Control, 24(4), 769–​776.
Xiaohua Y, Zhenjiang Y, Weidong L, Pengcheng X, and Sidong C. 2014. PMID: 23361339.
The non-​linear threshold association between aspirin use and esopha- Zhang XL, Liu M, Qian J, et al. 2014. Statin use and risk of kidney cancer: a
geal adenocarcinoma:  results of a dose-​ response meta-​ analysis. meta-​analysis of observational studies and randomized trials. Br J Clin
Pharmacoepidemiol Drug Saf, 23(3), 278–​284. PMID: 24436316. Pharmacol, 77(3), 458–​465. PMCID: PMC3952720.
Yan L, Chen P, Chen EZ, Gu A, and Jiang ZY. 2014. Risk of bladder cancer in Zhong S, Zhang X, Chen L, et al. 2015a. Association between aspirin use and
renal transplant recipients: a meta-​analysis. Br J Cancer, 110(7), 1871–​ mortality in breast cancer patients: a meta-​analysis of observational stud-
1877. PMCID: PMC3974078. ies. Breast Cancer Res Treat, 150(1), 199–​207. PMID: 25677744.
Ye X, Fu J, Yang Y et al. 2013. Frequency-​risk and duration-​risk relationships Zhong S, Zhang X, Chen L, et  al. 2015b. Statin use and mortality in cancer
between aspirin use and gastric cancer:  a systematic review and meta-​ patients:  Systematic review and meta-​analysis of observational studies.
analysis. PLoS One, 8(7), e71522. PMCID: PMC3728206. Cancer Treat Rev, 41(6), 554–​567. PMID: 25890842.
432
 43

24 Infectious Agents

SILVIA FRANCESCHI, HASHEM B. EL-​SERAG, DAVID FORMAN,


ROBERT NEWTON, AND MARTYN PLUMMER

OVERVIEW GLOBAL BURDEN AND GENERAL FEATURES

Eight volumes in the International Agency for Research on Cancer We present estimates of the global burden of cancer attributable to
(IARC) Monographs on the Evaluation of Carcinogenic Risks to infection for 2012 (Plummer et al., 2016). The methodology is based
Humans have been dedicated to infectious agents (Monographs 59, on estimating the population attributable fraction (PAF) from the prev-
61, 64, 67, 70, 90, 100B, 104; IARC, 1994a, 1994b, 1995, 1996, alence of infections in cancer cases. The PAF is the proportion of can-
1997, 2007, 2012a, 2014). The current classification of infectious cer cases that would not have occurred if the exposure—​in this case,
agents is reproduced in Table 24–1, along with the cancer sites or an infectious agent—​had not been present in the population. The PAF
types for which there was “sufficient evidence” or “limited evi- shows the relative importance of any exposure as a cause of cancer in a
dence” of carcinogenicity in humans according to the IARC crite- given population. It does not correspond to the number of cancers that
ria (Monograph preamble) (IARC 2012b). Eleven infectious agents, can be prevented in practice, but may nevertheless be used as a guide
comprising seven viruses, three parasites, and one bacterium, have to priorities in future cancer control.
been classified as carcinogenic to humans (group 1): hepatitis B virus HIV-​1 causes a particular difficulty with PAF calculations as it
(HBV); hepatitis C virus (HCV); 13 types of human papillomavirus causes cancer indirectly through immunosuppression, increasing the
(HPV); human immunodeficiency virus type 1 (HIV-​1); human T-​cell risk of cancer due to other oncogenic viruses. In order to avoid double
leukemia virus type 1 (HTLV-​1); Epstein-​Barr virus (EBV); Kaposi counting, we do not attribute cases to HIV-​1 but to the other infec-
sarcoma herpesvirus (KSHV); Helicobacter pylori; Opisthorchis tious agents. HIV-​infected people have a distinct profile of cancer
viverrini; Clonorchis sinensis; and Schistosoma haematobium. There incidence (Robbins et  al., 2015), with a high proportion of cancers
is sufficient evidence for all of these agents that they cause cancer in caused by other viral infections. As an example, in the United States,
humans in at least one cancer site, and that some of them cause can- HIV-​infected people have an estimated fraction of infection-​associated
cer in many sites. Infectious agents that did not reach the evidence cancer that is 10-​fold higher than the general population (de Martel
threshold to be classified as carcinogenic to humans according to et al., 2015b).
IARC’s criteria are also shown in Table 24–1. These agents have, Out of 14  million incidence cases of cancer that are estimated to
at best, limited evidence of carcinogenicity in humans, and varying have occurred worldwide in 2012 (Ferlay et al., 2013), 2.2 million were
degrees of evidence for carcinogenicity in experimental animals that caused by infectious agents classified in group 1. Table 24–2 shows
may allow them to be classified as “probably carcinogenic” (group the breakdown of these 2.2 million cases by agent. H. pylori causes
2A) or “possibly carcinogenic” (group 2B) to humans. Agents clas- 770,000 cases and so is the single most important agent worldwide
sified in group 2B may have limited evidence of carcinogenicity in when both sexes are combined. HPV causes 640,000 cases worldwide,
humans, or sufficient evidence of carcinogenicity in experimental mainly of cervical cancer, but also other anogenital cancers and a frac-
animals without sufficient evidence in humans. Some HPV types tion of cancer of the head and neck. HBV causes 420,000 cases of
(HPV 30, 34, 69, 85, 97)  are classified in group 2B because they liver cancer. HCV causes 170,000 cases, mainly liver cancer, but also
are closely related to other types in group 1. The agents marked as non-​Hodgkin lymphoma. Epstein-​Barr virus causes Hodgkin lym-
“not classifiable” (group 3) were considered in the same monograph phoma, Burkitt lymphoma, and nasopharyngeal carcinoma with a total
volume as the other infectious agents, typically because they were of 120,000 cases. The remaining agents cause less than 60,000 cases.
closely related, but did not have sufficient evidence for their own Although one out of six cancers worldwide is caused by infec-
evaluation. tion, this proportion varies widely by geographic region. Table 24–3
It should be noted that the classification scale used by the IARC shows the number of incident cases by region, as well as the num-
Monographs is not based on the strength of the association, but the ber attributable to infection with the corresponding PAF. The low-
weight of evidence from studies in humans and animals and on mecha- est PAF is seen in North America, where only 4.0% of cancers are
nisms. In particular, the HPV types in group 1 may differ by an order caused by infections. The highest PAF is seen in Sub-​Saharan Africa,
of magnitude in risk for cervical cancer (IARC, 2007). where infections cause 31.3% of cancers. A  more detailed view of
One way to judge the relative importance of carcinogenic infec- the variation in PAF by country is shown in Figure 24–1. The United
tions is by estimating the global burden of cancers due to these States, Canada, Australia, New Zealand, and several countries in
infections. From this perspective, an important distinction emerges Western and Northern Europe have a PAF below 5% for infectious
between infections that are found worldwide and those that have a agents. Conversely, several countries in Sub-​Saharan Africa as well
limited geographical distribution. The limited infections are HTLV-​1, as Mongolia have a PAF over 40%. Table 24–3 also shows the varia-
O.  viverrini, C.  sinensis, and S.  haematobium. HTLV-​I is primarily tion when countries are grouped by their human development index
vertically transmitted through breastfeeding and is therefore limited (HDI) (Bray et al., 2012; United Nations Development Programme,
to endemic populations. The others are helminths with complex life 2013) into four categories: very high, high, medium, and low HDI.
cycles involving human and non-​human hosts, so that exposure is The PAF shows a gradient with HDI with a high PAF in the low
only possible under specific geographical conditions. The geographi- (25.3%) and medium (23.0%) HDI countries, lower PAF in the
cally limited infectious agents do not make a major contribution to the high HDI countries (13.2%), and the lowest found in the very high
global burden of cancer even if they are important causes of cancer in HDI countries (7.6%). Countries with very high HDI have the most
endemic populations. resources to prevent the circulation of HBV and HCV through unsafe

433
43

434 PART III:  THE CAUSES OF CANCER


Table 24–​1. Classification of Infectious Agents by the IARC Monograph Program

Evidence of Carcinogenicity in Humans (Animals)

Infectious Agent Volume Sufficient Limited

Group 1: Carcinogenic to humans
Hepatitis B virus 59, 100B Liver Non-​Hodgkin lymphoma
Hepatitis C virus 59, 100B Liver; Non-​Hodgkin lymphoma
Helicobacter pylori 61, 100B Stomach; MALT lymphoma
Opisthorchis viverrini 61, 100B Bile duct
Clonorchis sinensis 61, 100B Bile duct
Schistosoma haematobium 61, 100B Urinary bladder
Human papillomavirus type 16 64, 90, 100B Uterine cervix; anus; vulva; vagina; penis; oral Larynx
(HPV 16) cavity; tonsil; pharynx
HPV 18 64, 90, 100B Uterine cervix Anus; penis; vulva
HPV 33 64, 90, 100B Uterine cervix Anus; vulva
HPV 31,35,39,45,51,52,56,58,59 64, 90, 100B Uterine cervix
Human immunodeficiency virus-​I 67, 100B Lymphoma; Kaposi sarcoma; uterine Liver and bile duct; skin; Vulva; vagina;
(HIV-​I) cervix; anus; eye penis
Human T-​cell leukemia virus 67, 100B Adult T-​cell leukemia/​ lymphoma
type I (HTLV-​I)
Epstein-​Barr virus 70, 100B Lymphoma; nasopharynx Stomach
Kaposi sarcoma herpesvirus 70, 100B Kaposi sarcoma; primary effusion lymphoma
Group 2A: Probably carcinogenic to humans
HPV 68 100B Uterine cervix
Merkel cell polyomavirus 104 Skin
Plasmodium falciparum 104 Burkitt lymphoma
Group 2B: Possibly carcinogenic to humans
Schistosoma japonicum 61 Liver and bile duct;
colon and rectum
HIV-​2 67 Kaposi sarcoma; lymphoma
HPV 26,53,66,67,70,73,82 100B Uterine cervix
HPV 30,34,69,85,97 100B
HPV 5, 8 100B Skin1
JC polyomavirus 104 (Brain)
BK polyomavirus 104 (Sarcoma; brain)
Group 3: Not classifiable with regard to carcinogenicity to humans
Hepatitis D virus 59
Opisthorchis felineus 61
Schistosoma mansoni 61
HTLV-​II 67
HPV 6, 11 90, 100B
HPV genus beta and gamma 90, 100B
SV40 polyomavirus 104

1
Limited evidence in individuals with epidermodysplasia verruciformis.

injection practices and contaminated blood and blood products, and for a large proportion of cancers when the population age structure is
also to implement screening programs to prevent HPV from causing skewed toward younger ages. The relatively early age at onset of cervi-
cervical cancer. They are similarly well placed to prevent HPV infec- cal cancer may also explain why HPV is the dominant infectious cause
tion in young people through as yet expensive HPV vaccines. Most of cancer in low HDI countries.
very high HDI countries have avoided extensive iatrogenic transmis-
sion of HBV, HCV, and HIV, although Japan and some Southern
Detection and Attribution
European countries were historically notable exceptions.
Figure 24–2 shows the number of infection-​attributable cancers by Good-​quality biomarkers of infection are important for accurately esti-
HDI category, as well as the breakdown by agent. Half of the cases of mating risk and ascribing a causal role to infectious agents in cancer.
infection-​attributable cancer occur in medium HDI countries that rep- In their review of causes of cancer, Doll and Peto (1981) hypothesized
resent 50% of the world’s population. HBV is an important cause of that 10% of US cancer deaths were due to infection, with substan-
cancer in medium HDI countries and is relatively much more impor- tial uncertainty surrounding this estimate. This review was made at a
tant than HCV. This is reversed in high HDI and very high HDI coun- time before H. pylori and HCV had been identified, and before HPV
tries where HCV is relatively more important than HBV. H. pylori is had been confirmed as a major cause of cervical cancer. Progress in
an important infectious cause of cancer across medium, high, and very the epidemiological study of infections and cancer has been highly
high HDI countries. In low HDI countries, H. pylori is responsible for dependent on improvements in biomarkers of infection. Improvements
a relatively small proportion of infection-​attributable cancers. Gastric in sensitivity come from the ability to detect a lower burden of infec-
cancer reaches its highest incidence at much older ages than cervi- tion. The most sensitive biomarkers are DNA detection methods that
cal cancer and HBV-​associated liver cancer and so cannot account use polymerase chain reaction (PCR) to amplify few DNA or RNA
 435

Infectious Agents 435
Table 24–​2. Number of New Cancer Cases Occurring in 2012
1
Chronicity
Attributable to Infection by Infectious Agent
A universal feature of carcinogenic infectious agents is that chronic
Infectious Agent N % infection is a necessary condition for cancer to develop, with a latent
period between first infection and cancer incidence usually lasting
Helicobacter pylori 770,000 35.4 decades in immunocompetent individuals. For some agents, short-​
Human papillomavirus 640,000 29.5 term infections are also possible, but these do not entail any can-
Hepatitis B virus 420,000 19.2 cer risk. For example, the majority of HPV infections of the cervix
Hepatitis C virus 170,000 7.8 are transient and clear within 1 year, with a minority persisting and
Epstein-​Barr virus 120,000 5.5 progressing to cancer. Likewise, HBV and HCV may either cause a
Kaposi sarcoma herpesvirus 44,000 2.0 short-​term infection or lead to a chronic carrier state. The risk of an
Schistosoma haematobium 7,000 0.3 HBV infection leading to a chronic carrier state depends strongly on
Human T-​cell leukemia virus type I 3,000 0.1 age at exposure, with children at very high risk but adults at low risk.
Opisthorchis viverrini, 1,300 < 0.1 Conversely, a majority of HCV infections lead to a chronic carrier
Clonorchis sinensis state at any age.
Total infectious agents 2,200,000 100.0 Although chronicity is a universal feature of carcinogenic infec-
tions, it is attained by a wide variety of different mechanisms. For
example, HBV, HCV, H.  pylori, and helminth infections all cause a
1
Numbers are rounded to two significant digits.
chronic inflammatory response, but HPV achieves persistence by
evading immune surveillance. S. haematobium lives only 3–​5 years in
copies of the infectious agent. However, PCR detection methods may the human host without multiplying, but chronic exposure is attained
not be feasible, especially in large epidemiological studies, if the in endemic areas by repeated reinfection.
affected tissue is not readily accessible for sampling. In contrast, sero- The importance of length of exposure for cancer risk has long been
logical measurements of antibodies are easier to obtain and to use in recognized for strong carcinogens such as tobacco and asbestos (Doll,
prospective studies but may have poor sensitivity. The importance of 1978; Peto, 1979) and mathematical risk models have been developed
good biomarkers can be illustrated with H. pylori and non-​cardia gas- to explain the relationship between exposure time and risk based on the
tric cancer (see section in this chapter on H. pylori). multistage model of carcinogenesis (Armitage and Doll, 1954). Most
Improvements in specificity for infection biomarkers come of the infectious agents classified in group 1 are acquired in childhood,
from two sources. The first is to distinguish between agents that especially in endemic areas, leading to the maximum lifelong expo-
may be closely related, but differ in their carcinogenic potential. sure and hence maximum risk. The exceptions are HCV and HIV-​1,
This is shown by the different types of HPV, which are represented which are often acquired in adulthood. Although mucosal HPV types
across the whole spectrum of classifications from group 1 to group are sexually transmitted, they are highly transmissible, so first exposure
3 (Table 24–2). The second way to improve sensitivity is to distin- occurs soon after sexual debut. The risk of cervical cancer is higher
guish between transforming infections and those in which the agent among women with earlier age at first intercourse, and this relationship
is merely present. For example, EBV is ubiquitous in humans. Its is consistent with the multistage model (Plummer et al., 2012).
causal involvement in lymphomas and nasopharyngeal carcinoma
has been demonstrated by its latency pattern and the corresponding
gene products expressed in tumor cells (IARC, 2012a). Similarly, in HELICOBACTER PYLORI
head and neck cancer, the detection of HPV DNA in tumor cells by
in situ hybridization or detection of HPV E6/​E7 RNA as a marker of
transforming infection shows that HPV-​attributable cancer is rare in Nature of the Exposure
the oral cavity, larynx, and hypopharynx (Combes and Franceschi, H. pylori is a spiral Gram-​negative bacterium, with unipolar flagella,
2014). evolved and uniquely adapted to live within the highly acidic envi-
ronment of the human gastric mucosa (Wroblewski et  al., 2010).
Table 24–3. Number of New Cancer Cases1 Occurring in 2012 Although observed in the human stomach since at least the begin-
Attributable to Infectious Agents by Geographic Region and Human ning of the twentieth century (Krienitz, 1906), H. pylori (originally
Development Index termed Campylobacter pylori) was not characterized and cultured
until the early 1980s (Warren and Marshall, 1983); shortly after, it
Number was shown to be strongly associated with the etiology of both gastritis
Number of New Attributable PAF2 and peptic ulcer disease (Marshall and Warren, 1984). Although there
Region Cases in 2012 to Infection (%) are now over 30 Helicobacter species identified (http://​www.bacterio.
net/​helicobacter.html), most of which do not colonize humans, only
Sub-​Saharan Africa 630,000 200,000 31.3
H.  pylori has been intensively studied and clearly identified as a
Northern Africa and 540,000 70,000 13.1
Western Asia human pathogen.
Central Asia 1,500,000 290,000 19.4 The genome of H.  pylori has been fully sequenced (Tomb et  al.,
Eastern Asia 4,900,000 1,100,000 22.8 1997), and the organism exhibits considerable genomic diversity, with
South and Central America 1,100,000 160,000 14.4 infected humans sometimes harboring multiple strains (Suerbaum
North America 1,800,000 72,000 4.0 and Josenhans, 2007). The two major pathogenicity factors are CagA,
Europe 3,400,000 250,000 7.2 encoded by the cagA gene within the cag pathogenicity island (Censini
Oceania 160,000 7,600 4.9 et al., 1996), and VacA, encoded by the vacA gene (Cover et al., 1994).
CagA is highly immunogenic and elicits potent host antibody responses,
Human development index while VacA is a highly cytotoxic protein. Whereas there are genetic dif-
Very high 5,700,000 430,000 7.6 ferences leading to CagA negative and positive strain types (Suerbaum
High 2,200,000 290,000 13.2 and Josenhans, 2007), the vacA gene is conserved in all H. pylori strains,
Medium 5,200,000 1,200,000 23.0 although it is subject to allelic variation (Atherton et  al., 1995). The
Low 940,000 240,000 25.3 combination of CagA and VacA activity is the principal known mechan-
WORLD 14,000,000 2,200,000 15.4 ism giving rise to strain variation in H. pylori pathogenicity.
Although primary acquisition of H. pylori infection can occur in
1
Numbers are rounded to two significant digits. adults, it most typically arises in children and, once established, usu-
2
Population attributable fraction. ally persists for life in the absence of treatment (Malaty and Graham,
436

PAF < 5%
PAF in 5%−15%
PAF in 15%−25%
PAF in 25%−40%
PAF > 40%

Figure 24–​1.  Geographic variation in cancers attributable to infectious agents using the population attributable fraction (PAF). Source: Modified from
Plummer M, de Martel C, Vignat J, Ferlay J, Bray F and Franceschi S, Global burden of cancers attributable to infections in 2012: a synthetic analysis.
Lancet Glob Health, 4(9) (2016), e609-e616, with permission from the authors.

1200
Helicobacter pylori
Human papillomavirus
1000 Hepatitis B virus
No. of cancer cases attributable to

Hepatitis C virus
Other infectious agents
infection (thousands)

800

600

400

200

0
Low HDI Medium HDI High HDI Very High HDI
1,303,000 3,553,000 1,042,000 1,129,000
DEVELOPMENT STATUS
Population size (thousands)

Figure 24–​2.  Number of new cancer cases in 2012 attributable to infection by infectious agent. Source: Modified from Plummer M, de Martel C, Vignat
J, Ferlay J, Bray F and Franceschi S, Global burden of cancers attributable to infections in 2012: a synthetic analysis. Lancet Glob Health, 4(9), (2016),
e609-e616, with permission from the authors.
 437

Infectious Agents 437
1994). Humans are the only known reservoir of H. pylori (Oderda, reviewed the evidence for this association (IARC, 2012a), after a prior
1999), and person-​to-​person contact, especially within the family, is evaluation in 1994 (IARC, 1994b), and confirmed the categorization of
believed to be the primary route of transmission (Brown, 2000). There H. pylori infection as carcinogenic to humans (Group 1) (Table 24–1).
is evidence for both oral–​oral and fecal–​oral routes of transmission; The IARC evaluation specifically indicated gastric carcinoma, exclud-
in developing countries, consumption of contaminated water may ing the cardia region of the stomach adjacent to the esophagus (non-​
also act as an additional source of infection (Brown, 2000). cardia gastric carcinoma), and low-​grade B-​cell mucosa-​associated
Although there are many means of identifying H.  pylori infection lymphoid tissue (MALT) gastric lymphoma as the two forms of gastric
from gastric biopsy material, including direct histological visualization, cancer caused by chronic infection with H. pylori.
the most commonly used noninvasive test in epidemiological studies Recent estimates indicate that there are approximately 823,000
has been serology for H.  pylori immunoglobin G (IgG) antibodies new cases of non-​cardia gastric cancer diagnosed per annum (Forman
using enzyme-​linked immunosorbent assay (ELISA) (Megraud and and Sierra, 2014), making it one of the most frequent cancer diag-
Lehours, 2007). Several other noninvasive tests are available, including noses worldwide, while there is also an annual burden of approxi-
the urea breath test (UBT), stool antigen test (SAT), and immunoblot mately 20,000 gastric MALT lymphomas (Plummer et  al., 2016). It
procedures (Megraud and Lehours, 2007). The latter have been espe- is estimated that a total of 770,000 of these two (93.6%) cancers are
cially useful in the determination of specific H. pylori antigens (notably attributable to H. pylori infection and, considering all infection-​related
CagA) and in situations when sensitivity to past infections, which may cancers (2.2 million), this contributes the largest burden (Table 24–2).
have resolved, is of importance (Peleteiro et al., 2010). Tests making
use of urine and saliva samples have been developed but are rarely Non-​Cardia Gastric Cancer
employed in epidemiological settings (Megraud and Lehours, 2007).
There are many options for treatment of H.  pylori infection, usu- Mechanism.  The dominant model for carcinogenesis in the non-​
ally involving a combination of two or three antibiotics in combination cardia stomach is the one originally proposed by Correa in 1975 (Correa
with a proton pump inhibitor for periods of 7–​14 days (Li et al., 2015). and Piazuelo, 2012), involving a series of sequential pathological
There is an expected eradication rate of around 80% generally associ- transformations of the gastric mucosa from gastritis through atrophy,
ated with such regimens, although this depends on compliance with intestinal metaplasia, dysplasia, and ultimately cancer (Figure 24–3).
the treatment and the antibiotic resistance profile within the population H.  pylori is thought to act as the initial driver of this process, with
(Gisbert and Greenberg, 2014). the inflammation resulting from infection resulting in chronic gas-
tritis. Over subsequent decades, inflammation and accompanying
oxidative stress, upregulated by bacterial virulence factors, create an
Epidemiology of H. pylori Infection environment promoting DNA damage and leading to genetic instabil-
Infection with H. pylori is quite common, with many adult populations ity, metaplasia, and ultimately neoplastic transformation (Hardbower
having a prevalence of over 50%, although it may be as low as 10% et al., 2014). This model is conceived as the process underlying the
in some while exceeding 70% in others (Brown, 2000; Peleteiro et al., more common, “intestinal,” histological form of non-​cardia gastric
2014). As prevalence has been declining in recent decades and is also cancer (Correa and Piazuelo, 2012). The other “diffuse” histological
dependent on age, comparisons between prevalence surveys need to form is also associated with H. pylori infection and associated chronic
make due allowance for both time period and age range of the study. gastritis but, in this case, there are different genetic sequelae, particu-
However, in age-​stratified population-​based samples of adults taken larly involving e-​cadherin genes (Peek and Blaser, 2002).
since 2000, there remains substantial variation in prevalence, ranging,
for example, from 15% in Australians (1–​59 years) to 79% in Latvians Epidemiological Evidence. There have been several correla-
(17–​99 years) (Peleteiro et al., 2014). The association with age is evi- tional studies providing evidence in support of the association between
dent in most populations studied and, although not invariably so, after non-​cardia gastric cancer and H. pylori infection. These include geo-
the first decade of life, there is an age-​related increase in prevalence up graphical comparisons, where populations at high gastric cancer risk
until around 60 years, after which there tends to be a plateau or slight have been shown to have a high prevalence of infection (EUROGAST,
reduction in prevalence. However, with infection acquisition predomi- 1993; Peleteiro et al., 2014; Figure 24–4) and temporal studies, where
nantly occurring in childhood (Malaty and Graham, 1994), the observed the decline in gastric cancer incidence (or mortality) within a pop-
age relationship is believed to result primarily from a cohort effect with ulation is preceded by an equivalent decline in H.  pylori prevalence
those born more recently having lower levels of acquisition as children (Sonnenberg, 2013). There have also been a large number of case-​
compared with those born further back in time (Banatvala et al., 1993). control studies to investigate the association (Cavaleiro-​Pinto et  al.,
Taking account of the age and period effects, there appears to be a 2011). There are, however, substantive problems in the sensitivity of
generally higher prevalence of infection in Central and South America, standard ELISA procedures to detect H. pylori retrospectively in cases
parts of Asia and Eastern Europe, in comparison with North America, with a prior diagnosis of gastric cancer. This is due to atrophic gastritis,
Australia, and Western Europe (Peleteiro et al., 2014). a precancerous lesion that reduces the bacterial load of H. pylori in the
In general, poor socioeconomic status is strongly related to a high stomach and subsequently can reduce IgG antibody titers (Peleteiro
prevalence of infection (Brown, 2000). Socioeconomic-​related factors et  al., 2014). More reliable epidemiological evidence has been that
that contribute to this relationship include hygienic conditions, house- derived from prospective cohorts, including case-​control comparisons
hold density/​crowding, and the number of young children in the house- nested within cohorts, which make use of serum samples collected
hold (Goodman et  al., 1996). This is consistent with the fecal–​oral, prior to cancer diagnosis. Studies in which immunoblot assays have
person-​to-​person mode of bacterial transmission and provides a coher- been employed in preference to ELISA have also provided impor-
ent explanation for the consistent decline in infection prevalence over tant evidence because of their improved assay sensitivity (Plummer
time observed in those countries where sanitation has improved over et al., 2015).
recent decades (Peleteiro et  al., 2014). The long-​term persistence of A pooled analysis of 12 nested case-​control studies, including 762
childhood-​acquired infection into adult life means that improvements cases of non-​cardia gastric cancer and 2250 controls, derived an odds
in hygiene will rapidly impact on infection prevalence in the young but ratio of 2.97 (95% confidence interval [CI] = 2.34–​3.77) for H. pylori
have little immediate effect on adults. infection assessed by ELISA increasing to 5.93 (95% CI = 3.41–​10.3)
when restricted to cases diagnosed at least 10 years after blood sampling
(Helicobacter and Cancer Collaborative Group, 2001). Comparisons
Association with Cancer of the Stomach making use of immunoblot assays applied to prospective studies show
(Gastric Cancer) substantially elevated risks with a pooled odds ratios of 17.0 (95%
CI = 11.6–​25.0) (Plummer et al., 2015). Alongside these results are those
The cancer most intensively studied in relation to H. pylori infection from cohort studies in Japan (Take et  al., 2011; Uemura et  al., 2001;
and with clear evidence of association is gastric cancer. The IARC Yanaoka et al., 2009) and Taiwan (Lee et al., 2013), all showing reduced
438

438 PART III:  THE CAUSES OF CANCER

Multifocal
Intestinal Intestinal
atrophic
Normal Non- metaplasia metaplasia
gastritis Low-grade High-grade Invasive
gastric atrophic of the of the
without dysplasia dysplasia adenocarcinoma
mucosa gastritis complete incomplete
intestinal
type type
metaplasia

Figure  24–​3. Consecutive steps for development of gastric adenocarcinoma of the intestinal type. Source: Reprinted from Park JY, Forman D,
Greenberg ER, and Herrero R, Helicobacter pylori eradication in the prevention of gastric cancer: are more trials needed? Curr Oncol Rep 15 (2013),
517–​527, with permission from Springer International Publishing AG.

risks of gastric cancer following H. pylori eradication and confirming a with CagA-​negative strains (Cavaleiro-​Pinto et al., 2011). A cross-​sec-
primary role for infection as a risk factor for this form of cancer. tional analysis using PCR to detect H. pylori in gastric biopsies found
that cagA-​positive strains are associated with severity of precancerous
Parameters That Influence Risk.  Several environmental expo- lesions of the stomach, while cagA-​negative strains were associated
sures have been considered as potential cofactors in the relationship only with chronic gastritis (Plummer et al., 2007). It is conceivable that
between H. pylori infection and non-​cardia gastric cancer, including most non-​cardia gastric cancers have a CagA-​positive infection as their
tobacco smoking, salt and dietary antioxidant intake, coinfection with primary cause. Although environmental, host, and bacterial factors, and
helminths (reviewed in Wroblewski et al., 2010), and possibly Epstein-​ their joint effects (Figueiredo et al., 2002), may modify the effect of H.
Barr virus infection (see related section in this chapter). Both host pylori to a considerable extent, none has yet been able to offer a satis-
genetic polymorphisms, especially in the interleukin 1 family (Persson factory explanation for the reason that certain populations, sometimes
et al., 2011), and variation in bacterial pathogenic factors have also been labeled as “enigmatic,” have apparently a low cancer risk despite a high
investigated in terms of their impact on cancer risk. Of the latter, most prevalence of H. pylori infection (Ghoshal et al., 2010).
attention has been paid to CagA strain variation. Meta-​analyses of con-
ventional ELISA studies show raised risks of non-​cardia gastric cancer Intervention Studies.  Given the importance of H. pylori infection
associated with CagA-​positive H. pylori strain infections in comparison as the dominant risk factor for non-​cardia gastric cancer, eradication

KOR KOR
40
Age-standardized incidence rate/100.000 (Log scale)

30 Age 20 years JPN


Age 60 years

20
CHL CHL

LTV LTV

10

CZE DEU CZE MEX MEX


ARG

AUS AUS
FRA FRA

USA USA
SWE SWE

0 20 40 60 80 100
Prevalence of Helicobacter pylori infection (%)

Figure 24–​4.  Gastric cancer incidence retrieved from GLOBOCAN, 2008 (Ferlay et al.) as a function of the prevalence of Helicobacter pylori infection in
different countries considering the data from samples evaluated mostly in the late 1990s/​early 2000s, according to age groups (strata with median age clos-
est to 20 and 60 years). ARG: Argentina; AUS: Australia; CHL: Chile; CZE: Czech Republic; DEU: Germany; FRA: France; JPN: Japan; KOR: Republic
of Korea; LTV: Latvia; MEX: Mexico; SWE:Sweden; USA: United States of America. Source: Reprinted from Peleteiro B, Bastos A, Ferro A and Lunet N,
Prevalence of Helicobacter pylori infection worldwide: a systematic review of studies with national coverage, Dig Dis Sci 59 (2014), 1698–​1709, with permission
from Springer International Publishing AG.
 439

Infectious Agents 439
No of events/total
Study H pylori control Risk ratio Weight Risk ratio
eradication (95% Cl) (%) (95% Cl)

Correa 2000 3/437 2/415 4.0 1.42 (0.24 to 8.48)


Wong 2004 7/817 11/813 14.2 0.63 (0.25 to 1.63)
Leung 2004–Zhou 2008 2/276 7/276 5.2 0.29 (0.06 to 1.36)
Saito 2005 2/379 3/313 4.0 0.55 (0.09 to 3.27)
You 2004–Ma 2012 34/1130 52/1128 70.2 0.65 (0.43 to 1.00)
Wong 2012 3/225 1/258 2.5 3.04 (0.32 to 28.99)
Total 51/3294 76/3203 100.0 0.66 (0.46 to 0.95)
Test for heterogeneity: τ2 = 0.00, χ2 = 3.62, df = 5,
P = 0.60, l2 = 0% 0.1 0.2 0.5 1 2 5 10
Favors Favors
Test for overall effect: z = 2.27, P = 0.02
eradication control

Figure 24–​5.  Forest plot of randomized controlled trials of Helicobacter pylori eradication therapy: effect on subsequent occurrence of gastric cancer.
Source: Reprinted from Ford AC, Forman D, Hunt RH, Yuan Y, and Moayyedi P, Helicobacter pylori eradication therapy to prevent gastric cancer in
healthy asymptomatic infected individuals: systematic review and meta-​analysis of randomised controlled trials, BMJ 348 (2014), 3174, with permission
from BMJ Publishing Group Ltd.

of the infection through the use of prescribed antibiotic regimens pro- recommended the development of further “screen and treat” programs
vides a means of cancer prevention. The large numbers and lengthy in high cancer-​risk populations, they also advised that such programs
time required for follow-​up, together with logistical and ethical con- be implemented in conjunction with a scientifically valid assess-
cerns relating to such an intervention in the general population, how- ment of their effectiveness and possible adverse consequences (IARC
ever, makes randomized controlled trials of this subject particularly Helicobacter pylori Working Group, 2014).
challenging. Results from six such studies have now been reported,
and these have been combined in a Cochrane Collaboration systematic Gastric MALT Lymphoma
review and meta-​analysis (Ford et  al., 2014). All these studies com- B-​cell gastric MALT lymphoma is a relatively rare form of cancer
pared the incidence of subsequent gastric cancer in individuals who representing a component of gastric non-​Hodgkin lymphoma. A sys-
have tested positive for H. pylori and who were randomized to receive tematic review (Asenjo and Gisbert, 2007) has shown that H. pylori
either eradication therapy or placebo/​no treatment and followed up for infection has been diagnosed in around 80% of MALT lymphomas,
at least 2 years. Based on approximately 6500 individuals randomized, increasing to 90% when the diagnosis is of low-​grade disease. There
and 127 cancers diagnosed, the summary relative risk for those in the are very few case-​control studies of the association between infection
eradication group was 0.66 (95% CI = 0.46–​0.95). This can be trans- and the risk of MALT lymphomas and, while they show a positive
lated into a number needed to treat to prevent one gastric cancer of 124 effect (reviewed in IARC, 2012a), the decisive evidence for the rela-
(95% CI = 78–​843) (Figure 24–5). A Japanese trial of a slightly differ- tionship is provided by clinical trials showing that when H. pylori is
ent group of 544 patients, who had all undergone endoscopic resection eradicated in patients with such lymphomas, in low-​grade form, com-
for treatment of early gastric cancer, were randomized to receive either plete remission is obtained in 60%–​80% that may be sustained up until
H. pylori eradication therapy or standard care after resection. After 3-​ 10 years (Wundisch et al., 2012).
year follow-​up, there was a 65% reduction in the risk of metachronous
gastric cancer in the intervention group (9 versus 24 cancers) (Fukase Cardia Gastric Cancer
et al., 2008). This finding has not, however, been reproduced in other For gastric cancer occurring in the proximal region of the stomach,
trials in Japan (Maehata et al., 2012) and Korea (Choi et al., 2014). adjoining the esophagus (cardia gastric cancer), meta-​ analysis of
results from case-​control studies shows no overall association with H.
Screening Strategies.  The randomized trial evidence, in com- pylori infection (odds ratio [OR] 1.08; 95% CI = 0.83–​1.40), although
bination with the observational epidemiological results and the cur- there is an important heterogeneity between those studies conducted
rent mechanistic understanding of gastric carcinogenesis, provides in low gastric cancer–​risk settings (OR = 0.78; 95% CI = 0.63–​0.97,
compelling support for the protective effect of H.  pylori eradication based on 16 studies) and those in high-​risk settings (OR = 1.98; 95%
in gastric cancer prevention. In addition, evaluations of population-​ CI = 1.38–​2.83, based on 14 studies; Cavaleiro-​Pinto et al., 2011). The
based screening for H. pylori and eradication of the infection in those latter positive result comprised studies in China, Japan, and Korea, and
found positive have been shown to be cost-​effective under a large this heterogeneity provides some support to the hypothesis of there
range of assumptions about both effectiveness and costs (Areia et al., being two distinct subtypes of cardia gastric cancer, one etiologically
2013). There is, therefore, a compelling basis for such an interven- similar to non-​cardia gastric cancer and more common in populations
tion, especially in populations with a high non-​cardia gastric cancer with a high incidence of gastric cancer overall, and the other resem-
risk. Nevertheless there remain substantive concerns regarding the bling esophageal adenocarcinoma (Hansen et al., 2007). A fraction of
generalizability of existing trials and the possibility of deleterious cancer of the cardia in Asia has therefore been attributed to H. pylori
consequences of population-​ wide antibiotic administration. Aside in Plummer et al. (2016).
from the possibility of diseases potentially protected by the presence
of H.  pylori (see discussion later in this chapter), the promotion of
enhanced antibiotic resistance, together with the unknown impact of Association with Other Cancers
alterations to the human microbiome, is of serious public health con-
cern (Park et  al., 2013). Ongoing randomized trials (summarized in Esophageal Cancer
IARC Helicobacter pylori Working Group, 2014) are likely to address Meta-​analyses of studies of squamous cell cancer of the esophagus do
some of these issues; currently, although an IARC Working Group not indicate any association with H.  pylori infection (OR  =  1.1; 95%
40

440 PART III:  THE CAUSES OF CANCER


CI = 0.78–​1.55, based on 9 studies; Islami and Kamangar, 2008). Infection from using gastric cancer to earlier endpoints would be critical in accel-
with H. pylori has, however, been identified as a possible protective factor erating knowledge from eradication trials.
for adenocarcinoma of the esophagus. One meta-​analysis has estimated a An evident weakness in management of H.  pylori infection is the
statistically significant reduced odds ratio of 0.56 (95% CI = 0.46–​0.68) absence of a more effective therapy than the current multiple antibiotic/​
for this association, based on 13 studies (Islami and Kamangar, 2008). proton pump inhibitor regimens. The complexity of the regimens avail-
When stratified, the reduced risk was confined to the association with able and the required duration of administration make them suboptimal,
CagA-​positive strains (OR = 0.41; 95% CI = 0.28–​0.62), with no rela- especially in the context of population-​based treatment programs and at
tionship for CagA-​negative strains (OR  =  1.08; 95% CI  =  0.76–​1.53; a time when there is heightened concern regarding antibiotic resistance.
Islami and Kamangar, 2008). This is consistent with H.  pylori infec- In this respect, while effective vaccine development has not progressed
tion having an inverse risk of association with gastroesophageal reflux rapidly, the emergence of a candidate prophylactic vaccine reaching
disease (GERD) (Xie et al., 2013) and Barrett’s esophagus (Rubenstein phase 3 trials (Zeng et al., 2015) is potentially exciting.
and Chen, 2014), important precursors of esophageal adenocarcinoma.
A  proposed mechanism for these effects relates to H.  pylori–​induced
atrophy reducing the acid secretory capacity of the gastric mucosa, as MUCOSAL HUMAN PAPILLOMAVIRUS
the acidity of the gastric juice is its main damaging component (McColl
et al., 2008). It should be noted, however, that a meta-​analysis of 12 stud- Nature of the Exposure
ies showed no association between H. pylori eradication (in contrast to
prevalence) and the risk of GERD (Yaghoobi et al., 2010). Human papillomaviruses are small non-​enveloped icosahedral viruses
(50–​60 nm) that contain a circular double-​stranded DNA genome. They
Colorectal and Pancreatic Cancers belong to the Papillomaviridae family, which includes 16 different gen-
Results suggestive of a positive relationship with H. pylori infection era classified into species and types (Figure 24–6). HPV species have
have been reported for colorectal cancer (meta-​analysis OR = 1.4; 95% 60%–​70% genomic nucleotide similarity. Conversely, types share less
CI = 1.1–​1.8 based on 11 studies; Zumkeller et al., 2006) and pancre- than 90% homology in the L1 region. At a finer phylogenetic level, types
atic cancer (meta-​analysis OR = 1.38; 95% CI = 1.08–​1.75 based on 6 have evolved into variant lineages and sublineages (Cullen et al., 2015).
studies; Trikudanathan et al., 2011), and pathogenic mechanisms have This section deals exclusively with the alpha genus, that is, mucosal
been advanced for both associations. However, the modest effect size HPV types (hereafter referred to as HPVs) (IARC, 2007, 2012a) that
attaches some uncertainty to these results. mainly infect the anogenital tract. The alpha-​9 (HPV 16–​related) and
alpha-​7 (HPV 18–​related) species groups contain the majority of known
Other Diseases Associated with Infection carcinogenic (hereafter referred to as high risk, hr) types (Table 24–4).
The most important hrHPV in terms of frequency in invasive cervical
Apart from associations with cancer, H.  pylori infection has been cancer (ICC) is HPV 16, then HPV 18, 45, 31, 33, 52, and 58, followed
established as having a major causative role in both duodenal and gas- by HPV 35, 39, 51, 56, and 59. The classification of rarer or weakly car-
tric ulcer, and antibiotic-​based eradication of the infection is now the cinogenic types (HPV 26, 53, 66, 67, 68, 70, 73, and 82) is challenging.
major therapeutic modality for treating these potentially fatal condi- The HPV genome is divided into three regions:  the long control
tions (Ford et al., 2006). H. pylori infection has also been proposed as a region (LCR), which regulates viral gene expression and replication;
risk factor for non-​ulcer dyspepsia (Moayyedi et al., 2006), and effec- the early (E) region, which encodes proteins required for viral expres-
tive prevention of this common condition is an important consideration sion, replication, and survival; and the late (L) region, which encodes
in cost-​effectiveness models of H. pylori “screen and treat” programs. the viral capsid. When overexpressed in eukaryotic cells, L1 can self-​
There are also a wide range of extra-​intestinal conditions, including assemble in virus-​like particles (VLPs), the basis of current prophylac-
those of metabolic, cardiovascular, dermatological, and hematological tic HPV vaccines (see Chapter 62.3 on HPV vaccination).
origins that have been investigated for their association with H. pylori Molecular tests are available to detect HPV DNA or RNA in
infection with largely inconsistent results. It has also been suggested exfoliated cells or tissue biopsies, and serological tests to detect
that infection may protect against a number of extra-​intestinal condi- serum antibodies against L or E proteins. Molecular tests have been
tions, most notably childhood asthma (Parsonnet, 2014). used more extensively than serological tests due to their demon-
strated value in cervical cancer screening (Ronco et al., 2014) (see
Chapter  63 on screening). More than 100 HPV tests have been
Future Research developed (Poljak et al., 2012), of which the most widely used has
Almost 90% of non-​cardia gastric cancers can be attributed to H. pylori been Hybrid Capture version 2 (HC2). However, PCR-​based assays
infection (Plummer et al., 2015). The strength of the association has led are increasingly used not only in research, but also in clinical set-
some to propose that infection might be considered “a (close to) neces- tings, to detect individual HPV types. Test sensitivity and specificity
sary cause of non-​cardia gastric cancer” (Brenner et al., 2004). This is vary substantially across molecular assays, hampering comparison
unlikely to be correct, as there are other independent causes of at least between studies. From a practical viewpoint, analytical sensitivity
a small proportion of these cancers (e.g., Epstein-​Barr virus; see related (minimal detectable copy number) should be distinguished from
section in this chapter). Less controversial, however, is the call from the clinical sensitivity (i.e., a test’s ability to correctly detect clini-
same authors for a change in research emphasis away from considering cally relevant infections and HPV-​associated precancerous lesions)
the certainty and magnitude of the association to questions of (1) the (Snijders et al., 2003). Additional tests are being considered to detect
cofactors responsible for cancer development among the large number hrHPV oncogenic activity (e.g., p16 overexpression) (Carozzi et al.,
of infected people, and (2) how knowledge of the association can be 2008), and distinguish productive infections from transforming
used to formulate cancer prevention protocols (Brenner et al., 2004). infection (e.g., E6/​E7 mRNA, and hypermethylation of host or viral
These two questions represent the two major epidemiological issues cells) (Steenbergen et al., 2014).
for future research as, in combination, they would address the current Serum antibodies (anti-​HPV), mainly against L1, have been used to
uncertainties about using knowledge of the relationship as a means of better reflect past and present (cumulative) infection with individual
cancer control. It remains quite unclear, for example, how to implement HPVs. However, L1 antibodies are detectable in only about half of
a “screen and treat” program to maximize benefits and to minimize any women within 18  months of an HPV DNA-​positive test (Robbins
risks. The optimal age group for intervention is unknown and, apart et al., 2014a). A fluorescent bead-​based multiplex platform has been
perhaps from serum pepsinogen (Miki, 2011), biomarkers or other indi- widely used in epidemiologic studies to measure seropositivity to
cators are not yet available that could help in stratifying the population various HPV proteins (including L and E proteins) (Robbins et  al.,
into risk groups. It also has not yet been established when the patho- 2014a). The sensitivity and target (immunodominant epitopes or all
logical “point of no return” in H.  pylori carcinogenesis occurs, after IgG) vary across serological assays, and none is commercially avail-
which eradication would be ineffective in cancer prevention. A move able (Crosbie et al., 2013).
 41

Infectious Agents 441
13
2
species 8
Genus 54 1
c91 10
Alpha-papillomavirus 6 94 43
77 78 42 7 40 12
29 3 28 10 32 6
7
66 5330 11
74
PcPV
68 39 56 55 CCPV RhPV1
70 44 9
59 c85 13 52 67
15 5 45
18 58
71 51 69 26 33 35
14 52 31
c90 11 16
73
34 3
61 72
81 49 75
3 c62 76 38 23
83 c87 22
c89
c86 2
84 2 9
37
4 27 17
57 5
4 BPV2 80
15 96
BPV1
92 4
EEPV 25
Delta-papillomavirus 1 20
19 Beta-papillomavirus
21
RPV 14
93 1
24
5
2 DPV
36
12
47
OvPV1 8
3 95
65
OvPV2 4 1
BPV5
50 2
Epsilon-papillomavirus
EcPV1 48
Zeta-papillomavirus 88
60 3 Gamma-papillomavirus
POPV 4
COPV 1 63 HaOPV 5
MmPV BPV6
CRPV FdPV BPV3
1 2 BPV4
Pi-papillomavirus
PsPV
Eta-papillomavirus FcPV 41
Omikron-papillomavirus
PePV Mu-papillomavirus Xi-papillomavirus
Theta-papillomavirus Lambda-papillomavirus
Kappa-papillomavirus Nu-papillomavirus
Lota-papillomavirus

Figure 24–​6.  Phylogenetic tree containing the sequences of the 118 papillomavirus types. Source: Modified from de Villiers EM, Fauquet C, Broker TR,
Bernard HU, and zur Hausen H, Classification of papillomaviruses, Virology 324 (2004), 17–​27, with permission from Elsevier. We would like to acknowl-
edge Arthur Bouvard for reworking this figure.

Epidemiology of HPV Infection HPV infection is the most common sexually transmitted infection
(STI) worldwide, and the majority of sexually active individuals of
Vast information is available on the prevalence of HPV types in cer- both sexes will acquire it at some time during their life (IARC, 2007).
vico-​vaginal samples in more than a million women with normal Around 300 million women in the world are estimated to have cervico-​
cytology (Bruni et al., 2010) and over 100,000 women with HPV-​pos- vaginal HPV infection at a given point in time (Bruni et  al., 2010),
itive cytological/​histological abnormalities of different severity (Guan corresponding to an average world prevalence of 12%. The highest
et al., 2012). Although the quantity and quality of data vary by region, prevalence has been reported in Sub-​Saharan Africa (24%), Eastern
a relatively comprehensive and consistent view of the world distribu- Europe (21%), and Latin America (16%). Globally HPV 16 is the most
tion of HPVs is available. common type (> 20% of HPV-​positive women with normal cytology)
and HPV 18, 31, 52, and 58 are among the 10 most common types,
with small variations across regions. HPV 6 is the most frequent
Table 24–​4. Classification of Selected Mucosal Human Papillomavirus low-​risk type. Multiple-​type infections are common (about a third of
Types by Alpha-​Species and Carcinogenicity According to IARC HPV-​positive women), but there is no evidence of synergy or antag-
Monograph 100B (2012) onism between HPV types in multiple-​type combinations (Vaccarella
et al., 2010).
Probably Possibly HPV prevalence rises rapidly after sexual debut and becomes rela-
Carcinogenic Carcinogenic tively low after 35 years of age, especially in more developed coun-
Carcinogenic to to Humans to Humans tries (Bruni et al., 2010). In some countries in Latin America and Asia
Alpha Species Humans (Group 1) (Group 2A) (Group 2B) (Zhao et al., 2012), a small second HPV prevalence peak in middle-​
aged women has been observed. However, HPV prevalence was similar
5 51 26 69* 82 across age groups in some African and Asian populations (Franceschi
6 56 30* 53 66 et al., 2006; Gage et al., 2012).
7 18, 45, 39, 59 68 70 85* 97* The IARC HPV Prevalence Surveys (Crosbie et al., 2013) include
9 16, 31, 33, 35, 52, 58 67 approximately 33,000 women aged 15–​64  years from 29 different
11 34* 73 areas, mainly in less developed countries (Figure 24–7). All sur-
veys used a standardized protocol to help overcome the problems
*Based on phylogenic analogy to HPV types with sufficient or limited evidence in of heterogeneity related to HPV detection methods and population
humans. recruitment.
42

442 PART III:  THE CAUSES OF CANCER


HPV Prevalence (%)
N 0 5 10 15 20 25 30 35 40 45 50 55
Guinea 833
Mongolia 969
Rwanda 2508
Vanuatu 987
Nigeria 932
Bhutan 2505
Fiji 1244
Poland 834
China, Shenzhen 1027
Argentina 978
India 1891
China, Shenyang 685
China, Shanxi 662
Chile 955
Colombia 1834
Georgia 1309
Korea 863
Mexico 1340
Vietnam, Ho Chi Minh 922
Italy, Turin 1013
Thailand, Lampang 1035
Nepal 932
Iran 825
Netherlands 3304 HPV 16 or 18
Algeria 759 Other high-risk type
Thailand, Songkla 706
Low-risk type only
Spain 911
Pakistan 899
Vietnam, Hanoi 994

Figure 24–​7.  Age-​adjusted prevalence of cervical human papillomavirus (HPV) DNA in sexually active women aged 15–​69 years by HPV type. Source:
Modified from Crosbie EJ, Einstein MH, Franceschi S, and Kitchener HC, Human papillomavirus and cervical cancer, Lancet 382 (2013), 889–​899, with
permission from Elsevier.

was on average 9.8 months (Rositch et al., 2013) and the longest dura-


Natural History of HPV Infection
tions were found for HPV 16, 31, 33 and 52 (≥ 11 months). Longer
Mucosal HPVs are very efficiently transmitted through direct skin-​ persistence of infection with a given HPV type reduces the probability
to-​skin or skin-​to-​mucosa contact, mainly during sexual intercourse, of subsequent clearance. Prevalent infections in older women are less
including non-​penetrative intercourse. HPVs can reach basal cells likely to regress (Figure 24–8) not because of a woman’s age, but as
through micro-abrasions in the epithelium of the anogenital tract they are more likely to be long-​duration infections (Maucort-​Boulch
and head and neck (see corresponding sections in this chapter). Rare et  al., 2010). In fact, age is not associated with HPV persistence in
non-​sexual routes of transmission include perinatal transmission and, incident infections.
possibly, transmission through fingertips, medical procedures, and HPV persistence is a prerequisite for progression to differ-
fomites (IARC, 2007). ent grades of cervical intra-​ epithelial neoplasia (CIN) that are
HPV prevalence includes a mix of new incident infections and per- distinguished by depth of epithelial infiltration (Schiffman and
sistent infections that have accumulated over time due to lack of viral Wentzensen, 2013). Of note, CIN1 is only an insensitive histopath-
clearance (Castle et  al., 2009; Munoz et  al., 2004)  The vast major- ological sign of HPV infection, whereas CIN2 includes a heter-
ity of new HPV infections at any age become undetectable over 6–​ ogeneous group of lesions that are equivocal in cancer potential.
18 months. In a large meta-​analysis, the duration of hrHPV infection CIN3 represents the most clinically relevant lesion and is the best

Age (years) N infections OR (95% CI) FSE Relative Risk & 95% FCI

Prevalent infections
< 20 753 1.00 0.098
20–29 2600 1.09 (0.89–1.33) 0.035
30–39 525 1.30 (1.02–1.66) 0.076 Figure  24–​8. Odds ratios (ORs) for persis-
40–49 146 1.25 (0.88–1.77) 0.147 tence of human papillomavirus infections of
> = 50 62 2.14 (1.32–3.47) 0.224 any type, by age and prevalent versus incident
Incident infections infection. CI: confidence interval; FSE: floating
< 20 649 1.15 (0.80–1.66) 0.167 standard error; FCI: floating confidence inter-
val. Source: Reprinted from Maucort-​ Boulch D,
20–29 2100 1.14 (0.93–1.40) 0.040
Plummer M, Castle PE, Demuth F,
30–39 447 1.24 (0.96–1.61) 0.089 Safaeian M, Wheeler CM, and Schiffman M,
40–49 179 0.77 (0.55–1.08) 0.142 Predictors of human papillomavirus persistence
> = 50 53 0.93 (0.55–1.58) 0.250 among women with equivocal or mildly abnormal
cytology, Int J Cancer 126 (2010), 684–​691, with
0.0 1.0 2.0 3.0 permission from John Wiley & Sons.
 43

Infectious Agents 443
surrogate endpoint for efficacy against ICC in screening or vaccine Cervix
trials. The probability of progression from persistent infection with The identification of hrHPVs as the necessary cause of ICC
hrHPV to CIN2 or worse (CIN2+) has been reported in many cohort (Walboomers et  al., 1999)  followed a century of epidemiologic
studies. Three-​year incidence rates of CIN2+ after persistent infec- study that had highlighted the association of ICC with multiple
tion were 40.8%, 17.5%, and 10.0% for HPV 16, 18, and an “aver- sexual partners (IARC, 2007, 2012a). Since the 1980s, numerous
age” of other hrHPV, respectively (Castle et al., 2009). Cytologically case-​control studies have shown particularly strong associations
normal women with type-​specific persistent hrHPV infection have of ICC with hrHPVs reaching relative risks (RR) in the hundreds.
a substantial risk of developing CIN3+, mainly depending on type; Prospective studies suggested that a single HPV measurement con-
for example, 12-​year absolute risk in a large Danish study was 47% fers a 10-​to 30-​fold increase in ICC risk if not detected and treated
(95% CI = 35%–​58%) for HPV 16 but 6.0% (4%–​8%) for hrHPVs (IARC, 2007, 2012a).
other than HPV 16, 18, 31, or 33 (Kjaer et al., 2010). Laser capture The proportions of HPV 16 and 18 among HPV-​positive samples are
micro-​dissection showed that, despite frequent co-​presence of more approximately three and two times greater, respectively, in ICC com-
than one type, a single high-​grade CIN lesion is caused by a single pared to normal cytology, in all world regions (Figure 24–9) (Guan
HPV type that can proceed to monoclonal ICC through multiple et al., 2012). Other hrHPVs mainly contribute to CIN2, and sometimes
transforming, carcinogenetic steps (Quint et al., 2012). CIN3, but become much less frequent in ICC (Figure 24–10). ICC
The natural history of HPVs has been much more frequently includes two major histological types: squamous-​cell carcinoma (about
studied in women than men. However, HPV prevalence appears 70%–​80% of all ICC) and adenocarcinoma. Contrary to squamous-​cell
to be higher in men’s genitalia, mainly because it does not decline carcinoma, in which HPV 16 is predominant, the contributions of HPV
with age (Giuliano et al., 2011). Conversely, HPV antibody level is 16 and HPV 18 are similar in adenocarcinoma. Precancerous lesions
higher in women at any age (Markowitz et al., 2009). These obser- of adenocarcinoma are called atypical glandular cells and adenocarci-
vations point to the possibility that HPV infection of the keratin- noma in situ, rather than CIN (Guan et al., 2013).
ized epithelium of the male genital tract is less likely to induce an
immune response than in the mucosal epithelium of the cervix and Anogenital Other Than Cervix
vagina (Lu et al., 2012). Meta-​analyses (de Vuyst et  al., 2009), large case series (Alemany
Data on the natural history of HPVs in anatomic sites other than the et al., 2015; de Sanjosé et al., 2013; Miralles-​Guri et al., 2009), and a
cervix are limited for both sexes and will be briefly described in the few case-​control studies (IARC, 2007) have shown that hrHPV infec-
sections reviewing the corresponding cancer sites. tion, most often HPV 16, is a precursor state to cancer of the anus,
vulva, vagina, and penis.
The fraction of anal cancer (mainly squamous-​ cell carcinoma)
Types of Cancer Caused by HPV attributable to hrHPVs is similar to that for ICC. Anal cancer is rare
HPVs are powerful carcinogens and are implicated in the etiology of in the general population in both sexes (< 2 per 100,000) but is 20-​
cancer in the anogenital tract and head and neck, causing approxi- fold more frequent in men who have sex with men (MSM), especially
mately 640,000 cases per year (see section on global burden and gen- among HIV-​infected MSM (de Vuyst et al., 2009). In fact, the major-
eral features in this chapter, as well as chapters in Part IV, “Cancers by ity of information on HPVs and anal intra-​epithelial neoplasia derives
Tissue of Origin,” elsewhere in this volume). from HIV-​infected MSM (Machalek et al., 2012).

65%
60%
55%
50%
45%
40%
HPV Positivity

35%
30%
25%
20%
15%
10%
5%
0%
Normal ASCUS LSIL HSIL CIN1 CIN2 CIN3 ICC ICC: Normal ICC: CIN3
ratio ratio
HPV 16 20.5 ± 3.7 22.0 ± 2.9 24.9 ± 3.1 47.5 ± 5.7 27.5 ± 4.4 39.7 ± 5.0 58.2 ± 4.1 62.7 ± 2.3 3.06 1.08
HPV 18 8.4 ± 1.1 9.1 ± 1.8 8.4 ± 1.4 9.6 ± 1.6 9.1 ± 1.7 10.0 ± 1.0 8.0 ± 1.4 15.6 ± 2.9 1.85 1.94
HPV 45 4.9 ± 0.9 5.8 ± 1.5 4.1 ± 1.0 4.6 ± 1.3 4.1 ± 1.3 5.0 ± 1.7 3.7 ± 0.9 5.3 ± 0.7 1.08 1.44

Figure 24–​9.  Positivity (±1.96 SE) for human papillomavirus (HPV) types 16, 18, and 45 as a proportion of HPV-​positive samples, by cervical disease
grade. ASCUS:  atypical squamous cells of undetermined significance; LSIL:  low-​grade squamous intraepithelial lesion; HSIL:  high-​grade squamous
intraepithelial lesion; CIN: cervical intraepithelial neoplasia grade; ICC: invasive cervical cancer. Source: Reprinted from Guan P, Howell-​Jones R, Li N,
Bruni L, de Sanjosé S, Franceschi S, and Clifford GM, Human papillomavirus types in 115,789 HPV-​positive women: a meta-​analysis from cervical infection to
cancer, Int J Cancer 131 (2012), 2349–​2359, with permission from John Wiley & Sons.
4

444 PART III:  THE CAUSES OF CANCER

15%

HPV Positivity 12%

9%

6%

3%

0%
Normal ASCUS LSIL HSIL CIN1 CIN2 CIN3 ICC ICC: Normal ICC: CIN3
ratio ratio
HPV 33 4.8 ± 0.8 5.8 ± 1.6 6.1 ± 1.3 8.4 ± 1.2 6.2 ± 2.0 8.3 ± 1.3 9.0 ± 1.2 4.5 ± 0.8 0.94 0.50
HPV 58 6.3 ± 1.0 7.9 ± 2.1 7.1 ± 1.2 7.6 ± 1.6 9.8 ± 2.5 12.2 ± 4.3 8.9 ± 2.7 4.4 ± 2.2 0.70 0.50
HPV 31 8.0 ± 2.1 9.1 ± 1.4 9.1 ± 1.5 11.1 ± 1.6 11.4 ± 4.0 11.6 ± 2.8 11.6 ± 1.4 4.0 ± 0.4 0.50 0.34
HPV 52 8.0 ± 1.9 10.2 ± 2.5 8.0 ± 2.3 9.5 ± 2.1 14.1 ± 3.6 16.5 ± 5.2 10.1 ± 3.0 3.6 ± 0.9 0.44 0.35
HPV 35 3.4 ± 0.7 5.6 ± 1.6 4.7 ± 1.4 5.6 ± 1.6 4.1 ± 1.5 4.9 ± 2.1 3.5 ± 1.0 1.7 ± 0.3 0.51 0.48

Figure 24–​10.  Positivity (±1.96 SE) for human papillomavirus (HPV) types 33, 58, 31, 52, 35 as a proportion of HPV-​positive samples, by cervical dis-
ease grade. ASCUS: atypical squamous cells of undetermined significance; LSIL: low-​grade squamous intraepithelial lesion; HSIL: high-​grade squamous
intraepithelial lesion; CIN: cervical intraepithelial neoplasia grade; ICC: invasive cervical cancer. Source: Reprinted from Guan P, Howell-​Jones R, Li N,
Bruni L, de Sanjosé S, Franceschi S, and Clifford GM, Human papillomavirus types in 115,789 HPV-​positive women: a meta-​analysis from cervical infection to
cancer, Int J Cancer 131 (2012), 2349–​2359, with permission from John Wiley & Sons.

The relatively low fraction attributable to HPV in the vulva and vulvar intra-​epithelial neoplasias are often detected during cervical
penis (≤ 50%) is largely accounted for by the coexistence of histo- cancer screening (de Vuyst et al., 2009).
logical types that substantially differ in their association with HPV
infection. Basaloid and warty carcinomas of the vulva (de Vuyst et al., Head and Neck
2009) and penis (Miralles-​Guri et al., 2009) are much more frequently Head and neck carcinomas (HNCs) are the malignancies for which
HPV-​positive and are detected in younger patients than other histologi- an association with HPVs has been discovered most recently (IARC,
cal types (mainly keratinized squamous-​cell carcinoma). HPV-​positive 2007) and the fraction attributable to the infection is most uncertain

100
Oropharynx Oral cavity Larynx Hypopharynx
90

80

70

60

50
%

40

30

20

10

0
PCR p16 in situ hybridization E6/E7 mRNA

Figure 24–​11.  Prevalence of human papillomavirus molecular markers and 95% confidence intervals by head and neck cancer site. Source: Reprinted from
Combes JD and Franceschi S, Role of human papillomavirus in non-​oropharyngeal head and neck cancers, Oral Oncol 50 (2014), 370–​379, with permission from
Elsevier.
 45

Infectious Agents 445
and different across world regions (see global burden and general fea- immortalization, transformation, inhibition of apoptosis, and accumu-
tures in this chapter). lation of genomic instability. E6 and E7 oncoproteins are principally
Epidemiological studies have mainly evaluated the presence of responsible for HPV neoplastic effects (i.e., inactivation of p53, induc-
HPV in oral cells obtained by mouth brushing and gargle (Chung tion of hTERT, and binding to PDZ for E6; inactivation of pRb and
et al., 2014). A large population-​based survey of oral HPV infection related pocket proteins, and activation of E2Fs for E7). HPV E6 and
is available only for the United States, and it showed a significantly E7 genes must be present in every cancer cell to maintain tumor phe-
higher HPV prevalence in men (10.1%) than in women (3.6%), and notype. Low-​risk types, notably HPV 6 and 11, do not share the car-
in current smokers (also after adjustment for gender) (Gillison et al., cinogenic mechanisms of hrHPV.
2012). Sexual transmission (including orogenital intercourse and pos- In addition to cell-​cycle deregulation, hrHPVs have also developed
sibly deep kissing) is the preponderant mode of HPV acquisition in the sophisticated techniques of immune evasion in which E6 and E7 are
head and neck (Chung et al., 2014). involved, for example, via suppression of innate immune response
HPVs were seldom found in the tonsil, regardless of whether (Crosbie et al., 2013). Cell-​mediated immunity is implicated in viral
archival tissue specimens or fresh cell/​tissue samples had been used clearance, but it fails in a small percentage of infections. Precancerous
(Palmer et  al., 2014). However, evidence incriminating HPV is far lesion regression is associated with infiltrating CD8+ cytotoxic T-​cell
stronger for the tonsil and oropharyngeal cancer (collectively referred response, whereas regulatory T-​ cell infiltrates, which maintain an
to as OPC) than other HNCs (Combes and Franceschi, 2014; IARC, immune-​tolerant environment, are associated with progressive lesions
2012a; Mehanna et al., 2013). HPVs, in the vast majority HPV 16, are (Crosbie et al., 2013).
two to four times more frequently detected in OPC than other HNCs, Mechanisms are best understood for HPV 16, 18, 31, and 33 with
and the difference is greater (> 10-​fold) using markers of HPV malig- respect to infection of the cervix (IARC, 2012a). While the epithe-
nant transformation (Figure 24–11) (Combes and Franceschi, 2014; lium of the entire anogenital tract and head and neck can be infected
Ndiaye et al., 2014). An increase in incidence rates of OPC has been by HPV, the transformation zone (TZ) of the cervix is especially sus-
reported in the past two decades in several high-​income countries ceptible to carcinogenesis. The TZ is an area of metaplastic tissue
despite decreasing rates for other HNCs (Chaturvedi et  al., 2013). between the squamous epithelium of the vagina and the columnar epi-
The fraction of HPV-​positive OPC also increased over the same per- thelium of the endocervical canal (Schiffman and Wentzensen, 2013).
iod (Chaturvedi et al., 2013; Mehanna et al., 2013). The size and location (exo-​or endo-​cervix) of the TZ is influenced
A bulk of epidemiologic evidence supports the notion that two by changes in sex hormone levels and pregnancies (see section on
main distinct entities exist for OPC, one driven by HPVs and the cofactors). The recent discovery of a population of cells of embry-
other by the use of tobacco and alcohol (Marur et  al., 2010). These onic origin may, if confirmed by other investigators, help explain the
entities also show differences in genetic alterations (Rietbergen unique vulnerability to HPV carcinogenesis of the TZ (Herfs et al.,
et  al., 2014)  and prognosis (i.e., better survival in patients with 2012). These cells have a unique cuboidal morphology and express
HPV-​positive OPC) (Marur et  al., 2010). The fraction of OPC attrib- specific biomarkers and, although not permissive for the entire HPV
utable to HPV varies by region and, in some regions, by gender life cycle, can harbor the virus for extended periods of time (Herfs
(Figure 24–12) (Combes et  al., 2014). However, the estimate of the et al., 2012). It has been proposed that these cells are the source of
attributable fraction is hampered by lack of accurate data on the site most, if not all, CIN2–​3 and ICC, and do not regenerate after ablation
of origin of HNCs and the frequent co-​presence of HPVs and tobacco of the transformation zone.
exposure. In the head and neck, the tissue most susceptible to HPV carcino-
genesis is the thin epithelium of the deep crypts of the palatine and
lingual tonsils. Despite the characteristic abundance of lymphocytes
Mechanisms of Carcinogenesis in the tonsils, the virus escapes immune elimination during malignant
In vitro and in vivo models have elucidated how hrHPVs cause can- transformation and progression. Precancerous lesions are ill defined
cer (IARC, 2007, 2012a). HPV carcinogenic mechanisms involve and rarely reported in the tonsils, even in the proximity of HPV-​
random integration of the viral genome in the human genome, cell positive OPC (Pai and Westra, 2009). HPV 16-​E6-​mRNA was also

20

18 HPV–OPC
HPV+OPC
Age-standardized incidence rate/100 000

16

14

12

10

6 Figure  24–​12.  Age-​standardized (world)


incidence rates of oropharyngeal cancer
4 (OPC) per 100,000 persons stratified by
country, gender, and HPV status and cor-
2 responding male (M)  /​female (F)  ratios.
Source: Reprinted from Combes JD, Chen
0 AA, and Franceschi S, Prevalence of human
UK Australia US France
papillomavirus in cancer of the oropharynx
M F Ratio M F Ratio M F Ratio M F Ratio by gender, Cancer Epidemiol Biomarkers
HPV+OPC 2.8 0.9 3.3 2.8 0.6 4.4 4.8 1.0 4.9 7.2 1.7 4.3 Prev 23 (2014), 2954–​2958, with permis-
HPV–OPC 1.9 0.6 2.9 2.8 1.0 2.9 2.5 1.2 2.0 10.6 1.0 10.2 sion from the American Association for
Total OPC 4.7 1.5 3.1 5.6 1.6 3.5 7.3 2.2 3.3 17.8 2.7 6.6 Cancer Research.
46

446 PART III:  THE CAUSES OF CANCER


rarely detected in the mucosa surrounding HPV-​positive OPC, which habits and, if introduced, the early impact of HPV vaccination and
suggests the absence of a field cancerization effect so common around HPV-​based screening in different populations. Decreases in the pro-
tobacco-​associated OPC (Rietbergen et al., 2014). portion of CIN2–​3 and ICC associated with HPV 16 and 18 would also
be informative regarding the impact of HPV vaccination (see Chapter
62.3 on HPV vaccination).
Parameters That Influence Risk Little is known on the probability of undetectable latent HPV
The probability of HPV persistence and progression to cancer in the infection and on the degree of naturally acquired immunity or vac-
anogenital tract is strongly influenced by the immune status, notably cine-​induced protection against reinfection with the same HPV type
HIV infection (see Chapter  25 on immunologic factors). Additional (Safaeian et al., 2010). This information would be essential to opti-
risk factors are mainly related to an increased probability of HPV mize the age range to be targeted by HPV vaccination and HPV
infection (e.g., number of sexual partners of the individual and also screening. How aggressive searching for and treating precancerous
number of partners of his or her sexual partners). Anogenital inter- lesions should be is an especially important question for cancer of the
course and orogenital intercourse are risk factors for anal cancer and anus and vulva, while the characterization of precancerous stages is
OPC, respectively. Condom use provides only partial protection due a priority for OPC.
the extension of HPV-​infected area. Risk factors associated with the Finally, the genomic variation of hrHPV that determines infection
probability of HPV persistence and progression have been nearly persistence and malignant transformation is insufficiently understood.
exclusively studied in the cervix and include high parity and recent Newly developed, next-​generation, whole-​genome sequencing permits
oral contraceptive use (Plummer et al., 2012). Large pooled analyses much larger studies and more complete HPV genotype-​phenotype
have shown that all these preponderantly hormone-​related risk factors examinations than previously possible (Cullen et al., 2015).
are associated with an increased ICC risk also after careful adjustment
for lifetime number of sexual partners and socioeconomic factors HEPATITIS B VIRUS
(International Collaboration of Epidemiological Studies of Cervical
Cancer, 2006, 2007). Hepatitis B virus (HBV) is a DNA virus within the Hepadnaviridae
In fact, the cervix is very sensitive to changes in levels of endogenous family, also including the woodchuck and the duck HBV that has a nar-
and exogenous sex hormones during a woman’s life. The rise in ovar- row host range as well as a striking tropism for hepatocytes (reviewed
ian activity after menarche stimulates the transformation of columnar in IARC, 2012a). The outer lipid bilayer envelopes the spherical HBV
reserve cells into squamous epithelium in the TZ. However, the tem- virion of the hepatitis B surface antigen (HBsAg). Within the inner
poral proximity of first sexual intercourse to menarche per se does not nucleocapsid core (HBcAg) is a partially double-​ stranded, circu-
increase the risk of HPV infection and ICC, contrary to the hypothesis lar genome, as well as a DNA polymerase. Several HBV genotypes
of a special vulnerability of the “immature” cervix (Plummer et  al., (A–​H) with distinct geographical and ethnic distributions have been
2012). Young age at first sexual intercourse may rather be a proxy of identified based on differences of 8% or more in their whole-​genome
long-​duration exposure to HPV infection and hence high probability sequence. For example, genotypes A and D predominate in Africa,
of malignant transformation. In addition, incidence rates of ICC in Europe, and India; genotypes B and C predominate in Asia; geno-
unscreened populations have been shown to stop rising after approxi- type E predominates in West Africa; and genotype F predominates in
mately 45 years of age (i.e., when peri-​menopausal hormonal decline Central and South America. In the United States, HBV genotypes A
starts) (Plummer et al., 2012). and D are more common in African Americans and whites, whereas
Full-​term pregnancies and oral contraceptives involve exposure to HBV genotypes B and C are more common among persons of Asian
high levels of both estrogens and progesterone, but which hormonal ancestry (El-​Serag, 2012).
pattern is associated with ICC is unclear. A few studies in women
(Rinaldi et al., 2011) and HPV16-​carrying transgenic mice (Chung
et al., 2010) suggested a role for high estrogen levels. In these mice, Natural History of HBV Infection
ICC could even be controlled by treatment with raloxifene, an estro-
gen-​receptor antagonist (Spurgeon et al., 2014). In humans, estro- As shown in Table 24–5, in acute HBV infection, HBsAg is usually
gen signaling in cervical cancer was shown to be very different from the first marker detected, appearing within the first few weeks of infec-
that in breast cancer (i.e., associated with estrogen receptors alpha tion. Anti-​HBcAG also appears during the early phase of infection,
in stromal fibroblasts rather than in tumor cells) (den Boon et  al., with IgM-​specific antibody being a characteristic of acute infection
2015). and declining within about 6 months, whereas anti-HBc IgG generally
Tobacco consumption represents another cofactor of HPV infection persists indefinitely as a marker of past infection. As HBV is cleared,
in the anogenital tract. However, the role of tobacco in HPV-​associated antibody against HBsAg becomes detectable about 8 months after
OPC is unclear. A departure from a multiplicative model (i.e., the OR infection and subsequent to the disappearance of HBsAg. HBeAg is
for tobacco use being smaller among HPV-​positive individuals, or present at about the same time as HBsAg and then is lost after a few
the OR for HPV infection being smaller among smokers) has been days to weeks, with subsequent seroconversion to antibody against
suggested by case-​control studies (Combes and Franceschi, 2014). HBeAg.
However, the lack of prospective studies prevents the comparison of Chronic carrier state of infection is marked by the presence of
the absolute risk of HPV-​associated OPC in combination with or in the HBsAg for more than 6 months. In chronic HBV infection, HBsAg,
absence of tobacco use. along with antibodies to HBc, remains persistently detectable, and
anti-​HBsAg does not develop. About 10%–​15% of HBV chronic car-
riers lose their HBeAg per year and seroconvert to anti-​HBe seropos-
itivity (Evans et al., 1997). HBsAg can disappear at a very low rate of
Future Research < 1% annually. HBV can become integrated into the host genome over
Despite amazing progress, knowledge gaps remain in several aspects the course of chronic infection and represents an important step toward
of HPV infection and related cancer. An HPV epidemic due to the hepatocarcinogenesis (IARC, 2012a).
consequences of the sexual revolution in women born after the 1940s The probability of chronic HBV infection is inversely related to
or 1950s has been inferred from trends in different STIs, CIN3 (Peto the age at which infection occurs (IARC, 2012a). Around 80%–​90%
et al., 2004), and age-​period-​cohort models of ICC (Vaccarella et al., of infants born to infected mothers become chronic carriers, as is
2013), but, by now, high-​throughput molecular and serological tests the case in endemic areas in Asia, whereas less than 10% of adoles-
should make direct measurement of HPV trends in population-​based cents and adults infected through sexual and parenteral routes will
surveys possible. Ideally, these tests might also be used on archi- develop a persistent infection, as is the case in areas of low HBV
val samples. Trends in HPV prevalence by anatomic site and gen- prevalence. In Sub-​Saharan Africa, HBV transmission mainly occurs
der would be essential to monitor the influence of changes in sexual in childhood.
 47

Infectious Agents 447
Table 24–​5. Typical Serological Patterns in Hepatitis B Virus Infection

Anti-​HBc

Infection Status HBsAg IgM Total HBeAg Anti-​HBe Anti-​HBs

Acute infection* + + + + − −
Chronic infection with high levels of viral replication + − + + − −
Chronic infection with low levels of viral replication† + − + − + −
Recovery from acute infection before development of anti-​HBs − + + − + −
Low titer; possible false positive − − + − − −
High titer; possible “low-​level carrier” − − + − + −
Recovery from acute infection, indicating immunity − − + − + +
Vaccine response‡ − − − − − +
Susceptible to HBV infection − − − − − −

Anti-​HBs: antibody to hepatitis B surface antigen; HBV: hepatitis B virus; HBsAg: hepatitis B surface antigen; anti-​HBc: antibody to hepatitis B core antigen; HBeAg:
hepatitis B envelope antigen; anti-​HBe: antibody to hepatitis B envelope antigen.
* Reactivated chronic disease may have this pattern with sensitive anti-​HBc IgM assays.

Some patients may be seronegative for HBeAg and anti-​HBe.

In unvaccinated individuals, a high titer may represent immunity or be nonspecific; low titers are often nonspecific.

About 70%–​90% of chronic HBV carriers remain asymptomatic Hepatocellular Carcinoma


(IARC, 2012a). However, 10%–​30% develop chronic persistent or Chronic HBV infection accounts for approximately 50% of total HCC
chronic active hepatitis. Of those carriers with chronic active hep- cases, and virtually all of childhood HCC (IARC, 2012a). It is the
atitis, approximately 1%–​ 2% per year will progress to cirrhosis. dominant risk factor in most areas of Asia and Sub-​Saharan Africa
Hepatocellular carcinoma (HCC) develops at an annual rate of 1%–​6% that have high incidence of HCC (de Martel et al., 2015a), with the
among those with cirrhosis. The latent period from infection to HCC exception of Japan, where the major risk factor for HCC is chronic
ranges from 30 to 50 years. HCV infection. HBsAg seroprevalence among persons with HCC
(i.e., a proxy of HBV attributable fraction) varies widely; for exam-
Epidemiology of HBV Infection ple, it is less than 10% in HCC in Sweden and the United States,
and between 10% and 20% in Japan and most European countries
HBV is primarily a blood-​borne infection that is transmitted by per-​ except Greece and Russia (> 40%). The largest proportion of HBV-​
cutaneous and mucosal routes. The major modes of infection are via associated HCC can be found in Asia, primarily in China and Korea
sexual, parenteral, and perinatal exposure. Multiple partners and a (> 70%), and in some Sub-​Saharan African countries (de Martel et al.,
history of sexually transmitted diseases appear to increase the risk of 2015a). HBV is estimated to cause 420,000 cases of HCC each year
infection (IARC, 1994a). Transfusion as a cause of HBV and HCV (Table 24–2).
has decreased dramatically in the past two decades since screening A meta-​analysis of case-​control and prospective studies indicated
has become increasingly sensitive and more cost-​effective. Parenteral that the lifetime relative risk (RR) for HCC is 15–​20 among HBsAg-​
exposure often can occur via injection drug use and occasionally dur- positive individuals, compared with HBsAg-​negative individuals (Cho
ing tattooing and body piercing. Medical personnel are at risk of HBV et  al., 2011). Cohort studies estimate HCC incidence rates among
transmission by contaminated needlesticks or other injuries with sharp subjects with chronic HBV infection to be 0.02–​0.2 per 100 person-​
instruments. Since HIV and HBV have similar modes of transmission, years in inactive carriers, 0.3–​0.6 per 100 person-​years for those with
coinfection is quite common. chronic HBV infection without cirrhosis, and 2.2–​3.7 per 100 person-​
Perinatal infection, usually by contact with maternal blood during years for those with compensated cirrhosis (El-​Serag, 2012; Fattovich
delivery and possibly in utero, is an important transmission route in et al., 2008).
high-​endemic areas, particularly in China and other parts of Southeast
Asia, where up to half of the chronic infections are due to neonatal
transmission (IARC, 2012a). Horizontal transmission of HBV also Mechanisms of Carcinogenesis
occurs in childhood. In developing countries, the use of nondispos-
As much as 80% of HCC is observed to occur in conjunction with cir-
able, contaminated needles and syringes still plays a role in HBV
rhosis (Okuda et al., 1982). Chronic HBV-​induced necro-​inflammatory
transmission (Dondog et al., 2011). In non-​endemic areas, transmis-
disease can lead to HCC through continuous cycles of hepatocyte
sion occurs mainly during adulthood and adolescence, with sexual
necrosis and regeneration that promote acquired mutations existing
contact and parenteral exposure as the predominant modes of HBV
in the liver cells (reviewed in IARC, 2012a). By virtue of its ability
infection.
to integrate into the host genome, HBV can initiate and potentially
Approximately 5% of the world population (350–​ 400  million
cause mutation, as well as promote hepatocarcinogenesis via constant
people) has chronic HBV infection (McGowan et  al., 2015). In the
immune-​mediated inflammation and cell turnover (Tabor, 1994). The
United States, 0.33% of the population has been estimated to be HBV
smallest HBV protein HBx is essential for viral replication and can
chronic carriers, based on data from NHANES III, conducted from
adversely modulate the turnover of cellular proteins involved in cell-​
1988 to 1994 (McQuillan et al., 1999). Men are more likely to have
cycle regulation.
been infected than are women. Non-​Hispanic blacks (12.8%) have
higher levels of ever infection than do non-​Hispanic whites (2.8%)
and Mexican Americans (4.8%).
Parameters That Influence Risk
Several demographic, viral, clinical, and environmental factors
Types of Cancer Caused by HBV
increase HCC risk among individuals with chronic HBV infection.
Chronic HBV is a firmly established cause of HCC. The estimated Male HBV carriers are at a higher risk of HCC (as well as of cir-
number of cancers caused by HBV is 420,000 per year (Table rhosis) than are female carriers (Evans et al., 2002). Although some
24–2). Positive associations have been also reported between of the gender differences may be due to higher prevalence of other
HBV and both cholangiocarcinoma and non-​Hodgkin lymphoma cofactors in men (e.g., high alcohol or tobacco consumption), endog-
(IARC, 2012a). enous hormones may also play a role. Cohort studies in Shanghai and
48

448 PART III:  THE CAUSES OF CANCER


Taiwan found a 50% to fourfold increase in rate of HCC for men in not produce a functional product (null genotypes) slightly increased
the highest tertile of testosterone level (Yuan et  al., 1995). A study risk for HCC. Polymorphisms in tumor necrosis factor-​α (TNFα), IL-​
in Taiwan found an association between fewer CAG repeats in the 1B, and IL-​10 have also been associated with the risk for HCC (Yang
androgen receptor gene and the incidence of HCC in male HBV car- et al., 2011).
riers (Yu et al., 2002). Studies of HBV infection in transgenic mice HCC risk scores for HBV-​infected patients are based on age, sex,
demonstrated that the androgen pathway can increase the transcrip- level of alanine aminotransferase, viral HBV load, HBeAg status, and
tion of HBV genes (Yeh and Chen, 2010). the presence of cirrhosis, or on the presence of cirrhosis and core pro-
The risk of HCC is increased in patients with high levels of HBV moter mutations (Abu-​Amara et al., 2015). HCC risk scoring systems
replication, determined by tests for HBeAg and levels of HBV DNA. for patients receiving antiviral therapy with adequate viral suppression
In a study among 11,893 Taiwanese men followed over a mean of require further evaluation (Wong and Janssen, 2015) and possibly in
8.5 years, the incidence rate of HCC was 1169 per 100,000 person-​ different populations.
years among men who were HBsAg-​positive and HBeAg-​positive, 324
per 100,000 person-​years for those who were only HBsAg-​positive, Prevention
and 39 per 100,000 person-​years for those who were HBsAg-​negative
(Yang et al., 2002). A community-​based Taiwanese prospective study, HBV vaccination is the main method of primary HCC prevention.
the Risk Evaluation of Viral Load Elevation and Associated Liver However, in patients already infected with chronic HBV, antiviral
Disease/​Cancer–​HBV (REVEAL-​HBV), reported that in a cohort of treatment is the main method of secondary prevention. Currently,
3653 HbsAg-​positive participants, the incidence of cirrhosis and HCC there are five nucleoside analogues (NA) approved for the treatment
increased in proportion to the serum level of HBV DNA, from < 300 of patients with chronic HBV that can suppress HBV-​DNA suc-
(undetectable) to ≥ 1,000,000 copies/​mL. Studies with sensitive ampli- cessfully in almost all patients. Lamivudine monotherapy has long
fication assays have shown that HBV DNA can persist in serum or been used and is associated with ALT normalization, HBV-​DNA
liver, as an occult HBV infection, among persons who have serological suppression, HBeAg seroconversion, and histological improvement
recovery from transient HBV infection (Huo et al., 1998). The stud- in the long term. However, the development of resistant strains of
ies arrived, however, at inconsistent findings (Shi et al., 2012), and in HBV has been a major problem for lamivudine, whereas telbivudine,
aggregate there is no convincing evidence that occult HBV is an inde- adefovir, entecavir, and tenofovir disoproxil have shown low rates
pendent risk factor for HCC or a cofactor with HCV infection in most of drug resistance. Entecavir and tenofovir disoproxil are currently
regions of the world (Lok et al., 2011). the recommended first-​line medications (World Health Organization
In studies performed in Asia, there is a greater association between [WHO], 2015).
genotype C infection and severe liver disease, cirrhosis, and HCC A meta-​analysis of cohort studies and clinical trials indicated
than genotype B; in Western Europe and North America, individuals beneficial effects of medium-​term NAs in the prevention of HCC
with genotype D had a greater incidence of severe liver disease or in chronic HBV, although treatment does not completely eliminate
HCC than those with genotype A (Huang and Lok, 2011). Mutations the infection and HCC risk (Papatheodoridis et al., 2010). The three
in the region of the HBV genome that encode the basal core promoter, studies including untreated controls detected HCC significantly
such as T1762 and A1764 (Liu et  al., 2006), have been associated less frequently in treated than in untreated patients (2.8% vs. 6.4%,
with increased incidence of HCC, whereas those in the precore region respectively, P = 0.003). The rate of HCC was significantly higher
(G1896A) have been associated with decreased incidence of HCC in lamivudine-​resistant than in NA-​naïve patients. Achievement of
(Yang et al., 2008). virological response at rescue therapy did not significantly reduce
Meta-​analyses of studies from various countries (Donato et  al., HCC risk in lamivudine-​resistant patients, suggesting that the main-
1998; Shi et al., 2005) reported additive effects of HBV and HCV tenance of long-​term viral suppression is needed for cancer preven-
on HCC risk, but a sub-​additive effect was seen in more recent stud- tion (Papatheodoridis et al., 2010).
ies (2000–​2009), cohort studies, and studies conducted in areas Less data exist on HCC risk reduction with entecavir or tenofovir
in which HBV and HCV infection were not common (Cho et  al., than with lamivudine. In a large retrospective nationwide chronic
2011). A  sub-​additive effect may be explained by a negative cor- HBV cohort from Taiwan, the investigators included 21,595 matched
relation between the detection of anti-​HCV and HBsAg in the same patients in the treatment and control groups. Although most patients
individuals, suggesting virus interference (Donato et  al., 1998; were treated with lamivudine, 5748 patients received 0.5 mg/​ day
Thomas et al., 2000). entecavir. The entecavir-​treated cohort had a significantly lower 7-​
The consumption of alcohol is known to be hepatotoxic, although year incidence of HCC (7.3%; 95% CI = 6.8%–​7.9%) than controls
not necessarily mutagenic (IARC, 2012d). Several studies suggest that (22.7%; 95% CI = 22.1%–​23.3%) (Wu et al., 2014). In Japan, inves-
a more than additive effect of heavy alcohol consumption and chronic tigators found a significantly reduced risk of HCC in the entecavir-​
HBV infection exists with respect to the development of HCC (Donato treated group compared to a matched historical cohort of untreated
et  al., 2002). A population-​based cohort study performed in Korea patients. Five-​year incidence rates of HCC were 3.7% and 13.7%,
found that among individuals with chronic HBV infection, the risk for respectively (Hosaka et al., 2013).
HCC increased significantly among subjects with an alcohol intake of
50 g/​day or more, with a relative risk of 1.2 for 50–​99 g/​day and of 1.5
for > 100 g/​day (Jee et al., 2004). Most studies have found a small but HEPATITIS C VIRUS
positive significant interaction between increased smoking and HBV
infection for HCC (Chuang et al., 2010; IARC, 2012d). Hepatitis C virus (HCV) is a small, enveloped RNA virus classified
Aflatoxin B1 (AFB1) is a mycotoxin produced by fungi of the as a separate genus in the Flaviviridae family, which includes yel-
Aspergillus species that grows on foods such as corn and peanuts low fever, dengue, and hepatitis G virus (reviewed in IARC, 2012a).
stored in warm, damp conditions. AFB1 causes a mutation at serine The positive-​sense, single-​stranded viral genome codes for the core
249 in the tumor suppressor gene TP53 that was detected in 30%–​ nucleocapsid protein, two envelope (E) glycoproteins (E1, E2), and six
60% of HCC tumor samples collected from individuals in aflatoxin-​ nonstructural (NS) proteins (NS2, NS3, NS4A, NS4B, NS5A, NS5B).
endemic areas, most of whom had HBV infections (IARC, 2012c). With respect to the nonstructural proteins, NS2, NS3, and NS4A have
AFB1 in the pathogenesis of HCC is primarily mediated by its effects protease functions, with NS3 also demonstrating RNA helicase activ-
on chronic HBV infection. ity; NS5B functions as an RNA-​dependent RNA polymerase and is
Host genetic factors might account for some of the variation in important in genome replication. There are at least 6 HCV genotypes
the risk of developing cirrhosis or HCC. A meta-​analysis of variants which differ in 30%–​35% of nucleotides. There are also several sub-
of glutathione S-​transferase (GST) genes, phase II isoenzymes that types; HCV subtypes 1a and 1b are the most common in the United
protect against the endogenous oxidative stress causing deletions in States and Europe, whereas in Japan > 70% of HCV-​infected individu-
GSTM1 and GSTT1, indicated that forms of GSTM1 or GSTT1 that do als carry subtype 1b.
 49

Infectious Agents 449
Natural History of HCV Infection cirrhosis is established, HCC develops at an annual rate of 1%–​8%,
thus making cirrhosis an important precancerous condition. Most
In acute HCV infection, virus RNA is detectable within 2 weeks of cases of HCV-​related HCC occur among patients with advanced fibro-
exposure. A few weeks later, serum alanine aminotransferase (ALT) sis or cirrhosis (de Mitri et al., 1995; Haydon et al., 1995). The inci-
levels begin to rise, with symptoms occurring soon after. Antibodies dence of cirrhosis 25–​30 years after HCV infection ranges from 15%
against HCV become detectable about 8–​12 weeks after exposure, but to 35% (Freeman et  al., 2001), and is highest among recipients of
HCV RNA is detectable by reverse-​transcriptase PCR (RT-​PCR) 5–​6 HCV-​contaminated blood products and hemophiliacs.
weeks earlier (reviewed in IARC, 2012a). The relative burden of HCV in HCC, in absolute terms and in relation to
Most newly acquired infections (75%–​85%) become chronic, as the burden of HBV, greatly varies worldwide. Egypt has the highest pro-
demonstrated by the persistence of HCV RNA, the main biomarker portion of HCC attributable to HCV (80% HCV only and 8% co-​infected
of active infection, for more than 6 months. In general, persistence of with HBV) (de Martel et al., 2015a). With the exception of Greece, where
infection is less frequent in women, younger persons, and individuals HBV is dominant, European countries show a preponderance of HCV
with symptomatic acute infection (Armstrong et al., 2006) and more over HBV (HCV close to 60% of all HCC in Italy and Spain) and also
frequent in African Americans and in persons with immunodeficiency. a substantial fraction of viral marker negative cases (from 21% in Italy
to 82% in Sweden). A  preponderance of HCV is noted in most South
American countries and in the United States. In Asia, a large predomi-
Epidemiology of HCV Infection nance of HCV infection in HCC is found in Japan (65%) and Pakistan
The World Health Organization (WHO) estimates that approximately (54%). Mongolia and Taiwan also show a rather high prevalence of HCV
3% of the world’s population is infected with HCV, and this varies con- though inferior to that of HBV. In the last decade or two, the relative con-
siderably among the different regions (Mohd et al., 2013). The highest tribution of HCV to HCC burden is declining in some countries impacted
reported level of infection is in Egypt (Mohamoud et al., 2013), with by the earliest HCV epidemics, notably in Japan, but increasing in others
estimates of 20%–​30%, as well as in Pakistan (Umar et al., 2010) and (e.g., the United States, Taiwan, and Egypt) (de Martel et al., 2015a).
Mongolia (Dondog et al., 2011). Intermediate prevalence, up to 10%,
is found in some parts of Italy (Hahne et al., 2013) and of China (Gao Non-​Hodgkin Lymphoma
et  al., 2011). In the United States, HCV infection rose dramatically A number of extrahepatic malignancies have been associated with
through the 1970s and 1980s and showed a disproportionally high HCV infection, especially non-​Hodgkin lymphoma. Case-​control stud-
burden among Americans born from 1945 to 1965. Anti-​HCV preva- ies and cohort studies found approximately two-​fold increased risk of
lence in this birth cohort was 3.2% (95% CI = 2.8%–​3.8%), substan- non-​Hodgkin lymphoma in HCV-​seropositive individuals (Giordano
tially higher than among adults born before 1945 or after 1965 (0.9%) et  al., 2007; IARC, 2012a). No clear or consistent difference was
(Smith et al., 2014). Within the 1945–​1965 cohort, anti-​HCV preva- found by histologic subtype or according to nodal or extra-​nodal pre-
lence was significantly higher among non-​Hispanic blacks (adjusted sentation. Remission of non-​Hodgkin lymphoma in HCV-​seropositive
OR = 2.3), income below poverty threshold (adjusted OR = 4.7), and cases subsequent to treatment of the infection has also been reported.
among those who reported blood transfusion before 1992 (adjusted The fraction of non-​Hodgkin lymphoma attributable to HCV infection
OR = 2.3) or intravenous drug use (adjusted OR = 98.4) (Smith et al., varies greatly by country but may be upward of 10% in areas where the
2014). Based on phylogenetic studies, HCV began to infect large num- prevalence of the infection is high (Dal Maso and Franceschi, 2006).
bers of young adults in Japan in the 1920s, in southern Europe in the
1940s, and in North America in the 1960s and 1970s (Mizokami and
Orito, 1999; Tanaka et al., 2006). High HCV prevalence has also been Mechanisms of Carcinogenesis
recently reported in the Russian Federation (3% overall, and > 8% in As a nonintegrating virus, HCV is not likely to act as a cancer initiator.
the 15–​44 age group) (Saraswat et al., 2015). As up to 90% of HCV-​associated HCC arises within a cirrhotic liver
Prior to the screening of the blood supply for anti-​HCV, which (Alter and Seeff, 2000), HCV infection may lead to cancer through a
began in 1990 in the United States and Europe but much later in low-​ promoting process of recurring cycles of cell death and regeneration,
income countries, transfusion of contaminated blood and blood prod- as would occur with cirrhosis. HCV is also thought to induce liver
ucts was an important source of transmission (Alter and Seeff, 2000). steatosis by impairing lipid metabolism. The mechanisms by which
Injecting drug use represents the most frequently reported risk fac- lymphoma is induced by HCV is ill understood, but a role for the con-
tor for HCV infection in the United States and many other countries tinuous activation of B-​cells by chronic HCV infection is probable
(Alter and Seeff, 2000; Nelson et al., 2011). Intranasal cocaine use, (reviewed in IARC, 2012a).
tattooing, body piercing, and sharing of razors are also possible routes
of HCV transmission (Alter and Seeff, 2000).
Other percutaneous exposures associated with the transmission of Parameters That Influence Risk
HCV include hemodialysis, organ transplantation, and accidental nee-
dle sticks in the health-​care setting (Alter and Seeff, 2000). In Egypt, Among HCV-​infected individuals, risk factors for HCC include dem-
transmission likely resulted from insufficiently sterilized syringes ographic characteristics, viral factors, and lifestyle factors (El-​Serag
during the mass schistosomiasis intervention efforts (Frank et  al., et al., 2014). There is strong evidence for a synergistic effect between
2000). Transmission of HCV by the sexual route (Terrault, 2002) and heavy ingestion of alcohol and HBV/​HCV infection, and this is greater
from mother to child (Roberts and Yeung, 2002) is rare although also for HCV (these factors presumably operating together to promote cir-
possible. rhosis) (Donato et  al., 2002; Hutchinson et  al., 2005). Among alco-
hol drinkers, HCC risk increased in a linear fashion with daily intake
> 60 g (Donato et  al., 2002). Concomitant HCV infection led to an
Types of Cancer Caused by HCV additional two-​fold increase in risk of HCC (Figure 24–13). The rela-
tionship between cigarette smoking and HCC has been established
Chronic HCV is a firmly established cause of HCC and non-​Hodgkin in several studies among subgroups defined by HBV or HCV status
lymphoma. The estimated number of cancers caused by HCV is (IARC, 2012d).
170,000 per year (Table 24–2). Positive association have been also Insulin resistance is associated with increased risk of hepatic steato-
reported for cholangiocarcinoma (IARC, 2012a). sis, advanced fibrosis, and HCC among patients with HCV infection.
There is a significant (68%) increase in diabetes among HCV-​infected
Hepatocellular Carcinoma individuals, compared with noninfected individuals, based on retro-
HCV infection accounts for approximately 20% of total HCC cases spective and prospective studies (El-​Serag et  al., 2006). However, a
worldwide. The risk of HCC in persons with HCV infection is substan- few studies have found that HCV and diabetes synergize to increase the
tial, with RRs > 10 found in several cohort studies. Once HCV-​related risk of HCC (Arase et al., 2013; El-​Serag et al., 2006). Drinking coffee
450

450 PART III:  THE CAUSES OF CANCER

With HCV infection and minimal side effects. For example, the combination of pan-​geno-
20 With HBV infection typic nucleotide polymerase inhibitor (sofosbuvir) with NS5A inhibi-
Without HBV and HCV infection tor (ledipasvir or daclatasvir), and the combination of NS3/​4A protease
15
inhibitor (ABT-​450/​ritanovir) and a non-​nucleoside polymerase inhib-
10 itor (dasabuvir) with and without ribavirin have been approved and
recently have been used in the clinical setting with remarkable success.
5 However, the public health impact of HCV treatment has yet to be
Odds ratio

realized. Data from the United States and Europe show that fewer than
20% of HCV-​infected patients are ever treated (Kanwal and El-​Serag,
2014) due to a combination of lack of screening for HCV carriage,
and limited availability and affordability of DAAs. Modeling studies
found that treatment of half versus all infected persons would reduce
1 the number of HCC cases by 30% versus 60%, respectively, over the
next decade (Davis et al., 2010). Initiatives such as systematic screen-
ing for HCV of the highest-​risk birth cohorts (1945–​1965) have been
launched in the United States to increase HCV detection and linkage
40 60 80 100 120 140 to care (Smith et al., 2014).
Alcohol intake (g/day)

Figure  24–​13. Odds ratios for hepatocellular carcinoma, according to EPSTEIN-​BARR VIRUS


alcohol intake and the presence of hepatitis B virus (HBV) or hepatitis C
virus (HCV) infection. The plot was obtained by fitting spline regression Nature of the Exposure
models on data obtained in Brescia, Italy, 1995–​2000. Epstein-​Barr virus (EBV), or human herpesvirus 4, is a gamma herpes-
Reprinted from El-​Serag, H.B., Epidemiology of viral hepatitis and hepatocel- virus of the Lymphocryptovirus genus and was the first human tumor
lular carcinoma, Gastroenterology 142 (2012), 1264–​1273.e.1, with permis- virus to be identified (Epstein et al., 1964). Like all herpesviruses, it has
sion from Elsevier. (Modified from Donato F, Tagger A, Gelatti U, Parrinello a linear double-​stranded DNA genome, within a lipid envelope, which
G, Boffetta P, Albertini A, Decarli A, Trevisi P, Ribero ML, Martelli C, Porru exists mainly in episomal form within infected cells. EBV exhibits
S, and Nardi G, Am J Epidemiol 155 [2002], 323–​331, with permission from
Oxford Journals.)
substantial heterogeneity across the genome, and there are two major
types (EBV-​1 and EBV-​2), differing in relation to genes that code for
nuclear antigens. EBV-​2 immortalizes B cells less efficiently than
EBV-​1 in vitro (reviewed in IARC, 2012a). While almost all primates
(though not decaffeinated coffee or tea) and the use of statin medications have EBV-​like viruses, humans are the only natural host for EBV, in
(El-​Serag et al., 2009) have been associated with a reduction in HCC whom it establishes latent infection within lymphocytes, primarily B
risk among people with and without viral hepatitis (Bravi et al., 2013). lymphocytes but also T lymphocytes or, on occasion, epithelial cells.
Viral factors are less influential on HCC risk for HCV than HBV. In culture, EBV is the most potent transforming human virus,
Most studies from the United States and Europe have not found a cor- although more than 90% of those infected develop no disease.
relation between viral load counts and risk of HCC. Studies from the Transmitted via saliva, EBV enters the epithelium of Waldeyer’s ton-
United States reported that genotype 3 infection is associated with sillar ring in the oropharynx and from there it infects naïve B cells
higher risk of cirrhosis and HCC than other genotypes (Kanwal et al., in lymphoid tissue. Primary infection provokes cellular immune
2014). A  meta-​analysis of 21 studies reported that patients infected responses, which rapidly remove EBV-​infected cells—​EBV can per-
with HCV genotype 1b had an almost two-​fold greater risk of devel- sist only in resting memory B cells in which no viral proteins are
oping HCC than patients with other HCV genotypes (Raimondi et al., expressed in immunocompetent individuals. These memory B cells
2009). However, genotypes 1 are reported to respond more favorably can return to the tonsil, where they occasionally trigger viral replica-
to antiviral treatment than other genotypes. Persons co-​infected with tion; the resulting virus may be released into saliva for onward trans-
HIV have faster progression to cirrhosis and decompensated liver dis- mission (Thorley-​Lawson and Allday, 2008).
ease, and the risk increase is correlated with the severity of immuno- Detection of antibodies against EBV in biological fluids has been
suppression (Clifford et al., 2008; Kramer et al., 2007). the major means of diagnosis, and characteristic patterns of serologi-
cal response denote primary infection, latent infection, reactivation, or
specific disease states (reviewed in IARC, 2012a). EBV DNA can be
Prevention detected in the blood or saliva of healthy carriers, although it is more
Successful HCV treatment has been reported to slow liver disease pro- often found in those with EBV-​related disease. In such individuals,
gression and reduce the risk of cirrhosis and HCC. Randomized con- it can also be identified in tumor tissue and occasionally as cell-​free
trolled studies and several non-​randomized studies of HCV-​infected DNA in sera.
patients with and without cirrhosis indicate a 57%–​75% reduction in
risk of HCC in patients who received interferon-​based therapy and
achieved a sustained viral response (SVR) or virologic cure (Morgan
Epidemiology of EBV Infection
et al., 2013). Successful HCV treatment reduces but does not eliminate Worldwide, more than 90% of adults have antibodies against EBV,
HCC risk. Approximately 1.5% of patients with SVR develop HCC, although the age at which primary infection occurs varies substan-
compared with 6.2% of those who did not respond. SVR was asso- tially. In Uganda for example, by age 1 year, 80% of children are
ciated with a similar reduction in the risk for HCC in patients with infected, compared to only about 45% in rural United States. Primary
cirrhosis: approximately 4.2% of patients with SVR developed HCC infection during early childhood is usually subclinical or associated
compared to 17.8% of those without SVR (hazard ratio [HR] = 0.23; with mild respiratory symptoms. However, about 50% of those among
95% CI  =  0.16–​0.35). Persons with cirrhosis or those of older age, whom infection is delayed into older age develop infectious mono-
or persons with high levels of α-​fetoprotein, low platelet counts, high nucleosis (IM) (IARC, 1997). At all ages, transmission is primarily
stages of fibrosis, or diabetes, remain at risk for HCC (Arase et  al., oral, although infection via contaminated blood transfusion has also
2013; Chang et al., 2012). However, disabling side effects are associ- been demonstrated. Early age at infection with EBV is the norm in
ated with interferon-​based treatments. low-​and middle-​income settings and is associated with lower soci-
The introduction of new but costly interferon-​free direct-​acting anti- oeconomic status and crowding. Delayed infection is more common
viral agents (DAAs) in 2014 has brought cure rates of more than 90% in high-​ income countries. Balfour et  al. (2013) followed college
 451

Infectious Agents 451
students in the United States (37% of whom were EBV-​negative) and et al., 2005). Type III comprises over 95% of NPC in high and interme-
reported that 75% of primary EBV infections were associated with diate incidence areas. The development of IgA to EBV viral capsid or
IM. Although the disease (125,000 new cases per year in the USA) EBV-​DNAs in serum or saliva correlates with an approximate 20-​fold
is usually self-​limiting, 10% of patients have fatigue that persists for increased NPC risk (Cohen et al., 2013).
6  or more months, and about 1% of cases result in severe compli-
cations including encephalitis, hepatitis, severe hemolytic anemia, or Gastric Cancer
thrombocytopenia (Cohen et al., 2013). A meta-​analysis of international literature showed that about 9% of
gastric cancer contains EBV (Murphy et  al., 2009). According to a
subsequent pooled analysis (Camargo et  al., 2011), EBV-​positivity
Types of Cancer Caused by EBV in gastric cancer cases was significantly higher in males, young sub-
EBV is a firmly established cause of Burkitt lymphoma, Hodgkin jects, non-​antral subsites, diffuse-​type histology, and in studies from
lymphoma, nasopharyngeal carcinoma, immune-​suppression-​related the Americas. Findings from the The Cancer Genome Atlas Research
non-​Hodgkin lymphoma, and extra-​nodal NK/​T-​cell lymphoma (nasal Network (2014) threw additional light on the issue, recognizing EBV-​
type). The estimated number of cancers caused by EBV is 120,000 positive gastric cancer as one of the four subtypes of a new molecular
per year (Table 24–2). There is limited evidence of causality for a pro- classification of gastric adenocarcinoma, but the relationship between
portion of gastric cancer and for lympho-​epithelioma-​like carcinoma EBV and H.  pylori, the main cause of cancer and precancerous
(IARC, 2012a). lesions in the stomach, is unclear. Serologic profiles suggest a role
for H.  pylori in both EBV-​positive and EBV-​negative gastric cancer
(Camargo et al., 2015), and EBV may contribute with H. pylori to the
Burkitt Lymphoma induction of severe inflammation and cancer progression (Cardenas-​
There are three distinctive clinical variants of the aggressive B-​cell
Mondragon et  al., 2015). When EBV was evaluated by the IARC
non-​Hodgkin lymphoma, Burkitt lymphoma (BL): so-​called endemic
monograph program, epidemiological evidence for a causal role in
BL (eBL, found in areas of Falciparum malaria endemicity), sporadic
EBV-​positive gastric cancer was considered insufficient, despite posi-
(the predominant type in non-​malarial areas), and immunodeficiency-​
tive mechanistic data (IARC, 2012a).
related. It was the first human tumor associated with an infection
(Burkitt, 1962), the first shown to have a chromosomal translocation
that activates an oncogene, and the first childhood tumor success- Immune-​Suppression-​Related Non-​Hodgkin
fully treated with chemotherapy alone (Molyneux et al., 2012). It is Lymphoma
also the fastest-​growing human tumor, with a cell doubling time of Immune suppression, whether inherited, iatrogenic, or HIV-​related,
24–​48 hours, and the most common childhood cancer in areas where is associated with an increased risk of EBV-​ related lymphomas.
Falciparum malaria is holoendemic, particularly across parts of Sub-​ In 1969, McKhann and Penn reported independently on post-​trans-
Saharan Africa and Papua New Guinea. plant lymphoproliferative disorders, which are now recognized to be
A direct causal role for EBV in eBL is indicated by its consistent either polyclonal EBV-​driven lymphoproliferations or true monoclo-
presence in tumors; the fact that infection of B cells precedes tumori- nal malignancies. The risk is highest among previously EBV-​negative
genesis; that EBV induces immortalization of B cells in culture; children seroconverting after organ transplant possibly via the graft
and that high anti-​EBV antibody titers precede disease development (IARC, 1997). Immunosuppression caused by HIV is also strongly
(reviewed in IARC, 2014). associated with a range of EBV-​driven lymphomas. In addition to BL
and HL, diffuse large B-​cell lymphoma (DLBCL) is strongly asso-
Hodgkin Lymphoma ciated with HIV (IARC, 2012a). The risk of DLBCL, especially in
The proportion of EBV-​positive Hodgkin lymphoma (HL) varies with the central nervous system, is strongly correlated with the severity of
age, sex, geographical area, and histological subtype (reviewed in immunosuppression (CD4+ count) (IARC, 2012a). The introduction
IARC, 2012a). EBV is more commonly associated with nodular scle- of combined antiretroviral treatment (cART) was therefore associated
rosis and mixed cellular subtypes in all world regions (Stein et  al., with a striking decline in EBV-​associated non-​Hodgkin lymphoma
2008). The bimodal age distribution of HL has a different shape in incidence in HIV-​infected individuals. A record-​linkage study between
more developed versus less developed countries. In less developed HIV/​AIDS registries and cancer registries in the United States sug-
countries, the first peak of HL incidence is seen well before the age of gested, however, that also in the late-​cART era 12% of all cancers in
15, and virtually all cases of pediatric HL are EBV-​related. In more HIV-​infected individuals (with or without AIDS) were still attributable
developed countries the first peak of HL is seen between the ages of to EBV (de Martel et al., 2015b).
15 and 35. In both areas, a second peak is often seen in older adults.
In addition to the presence of monoclonal EBV genomes in malignant Other Cancers
cells, epidemiological evidence for an association with HL includes EBV is also an established cause of the rare extranodal NK/​T-​cell
a roughly four-​fold increase in risk of disease among those who lymphoma (nasal type), found most commonly in Central and South
have been previously diagnosed with IM, together with a substantial America and in East Asia. A rare form of carcinoma with histological
increase in risk of disease among those with higher titers of antibod- similarities to NPC has also been linked to infection with EBV (IARC,
ies against EBV. Increasing prevalence of detectable EBV-​DNA in 1997, 2012a). Such lymphoepithelial-​like carcinomas can occur at
serum has also been associated with risk of HL (IARC, 1997, 2012a). multiple organ sites with epithelial linings, but most frequently in the
Finally, in the HIV-​positive population, the vast majority of HL is salivary gland and in the stomach.
EBV-​related and mainly consists of mixed cellular subtypes (Stein
et al., 2008).
Mechanisms of Carcinogenesis
Nasopharyngeal Carcinoma Infection with EBV has been shown to induce a number of steps in
A striking association of nasopharyngeal carcinoma (NPC) with the process of malignant transformation. In particular, EBV immortal-
EBV was first identified in 1966 (Old et  al., 1966). NPC is a rare izes B cells in culture and EBV-​encoded gene products induce cell
cancer worldwide but shows exceptionally high incidence rates (10 proliferation, block apoptosis, induce genomic instability, and cause
to 30 per 100,000 men) in Southern China, Singapore, and Malaysia. cell migration. Several of these products are also associated with main-
Intermediate incidence rates are found in the Arctic, Middle East, tenance of malignant cell growth and tumor progression. A  number
Southeast Asia, and North Africa (Chang and Adami, 2006). EBV of EBV gene products (i.e., EBV latent membrane proteins 1 and 2
genome or gene products have been detected in virtually all cases of [LMP1, LMP2]) and EBV nuclear antigens 1, 2, 3 (EBNA-​1–​3), are
undifferentiated non-​keratinizing squamous cell carcinoma (histologic expressed in EBV-​associated cancers according to distinct patterns
type III), irrespective of the geographic origin (IARC, 2012a; Barnes (Latency I, II, and III).
452

452 PART III:  THE CAUSES OF CANCER

Parameters That Influence Risk Kaposi’s sarcoma (KS) lesions, macrophages, dendritic cells, and oro-
pharyngeal glandular epithelium. Lytic reactivation of latently infected
The best-​understood cofactor of EBV is immunosuppression, as the cells results in viral progeny and is normally tightly controlled by
impairment of cell-​mediated immunity allows the spread of reactivated the immune system. This appears to be an important step in disease
EBV from memory B cells, which can result in various lymphopro- pathogenesis.
liferative disorders (IARC, 2012a) (see Chapter 25 on immunologic Diagnosis of the infection is by PCR or serological assays to
factors). Infections and chronic inflammation can also reactivate detect KSHV antibodies, usually against latent or lytic antigens orf73
EBV-​infected cells. In addition, the role of other genetic or environ- and K8.1, respectively (reviewed in IARC, 2012a). ELISAs have
mental risk factors in the etiology of many EBV-​associated cancers recently been developed using recombinant proteins and peptides.
is suggested by extreme geographic variations in their incidence. An Concordance between serological assays remains, however, moderate.
association between Falciparum malaria and endemic BL has been While antibody titers are very high in KS patients, titers are so low in
demonstrated in numerous ecological studies that have identified asymptomatic people that establishing a clear assay cutoff is difficult.
strong geographical and temporal correlations. More recently, case-​ Comparison of studies using different assays or cutoffs is therefore
control studies have demonstrated increased risk of eBL with increas- problematic.
ing antibody titers against Plasmodium falciparum (P. falciparum).
The parasitic infection expands the B-​cell pool from which eBL can
emerge, as well as reactivating latent EBV infection (Molyneux et al., Epidemiology of KSHV Infection
2012). The association between IM and HL suggests that age at infec-
tion may be an important determinant of risk for certain EBV-​related KSHV is the human herpesvirus that displays the most marked geo-
malignancies. Various genetic factors have been linked to the devel- graphical variation in prevalence. Prevalence is highest in Sub-​Saharan
opment of NPC among EBV-​infected people, together with consump- Africa (50%–​80% of men and women in most studies), intermediate
tion of salted fish during weaning in China, and other preserved food in Mediterranean countries (10%–​30%), and generally low (less than
preparation (e.g., “harissa” in Tunisia) (IARC, 2012a). Although the 10%) in other parts of the world (Engels et al., 2007). However, there
association of NPC with tobacco use is firmly established, the strength exist certain high-​risk populations with higher prevalence than in the
of association is weaker for undifferentiated than differentiated NPC general population in those regions, such as among Amerindians in
(Polesel et al., 2011). South America and among men who have sex with men (MSM) in
Europe, Australia, and the United States (reviewed by Minhas and
Wood, 2014).
Future Research The main route of transmission of KSHV is via saliva and, in coun-
tries where prevalence is high, primary infection begins in childhood
The importance of EBV as a cause of IM and several cancer types, and continues into adult life. In these settings, prevalence increases
especially in immunosuppressed individuals, and the lack of specific with increasing age and is higher if family members—​ especially
antiviral treatment have prompted a National Institutes of Health mothers—​are infected (Dedicoat et al., 2004; Whitby et al., 2000). In
(NIH) working group to issue recommendations for future research lower prevalence settings, transmission is rare among children but, in
and EBV vaccine studies (Cohen et al., 2013). A candidate vaccine adults, is probably still via saliva. There is no clear evidence of het-
containing soluble glycoprotein gp350 in adjuvants reduced the rate erosexual transmission and no relationship between KSHV and being
of IM in EBV-​negative adults, but did not affect the rate of EBV a sex worker (Malope et al., 2008) or being HPV-​positive (de Sanjosé
infection (Sokal et al., 2007). Which combination of viral proteins et al., 2009). Conversely, among MSM, KSHV infection is very fre-
would be needed for a vaccine to prevent EBV-​associated malignan- quent and is strongly associated with recent history of oral and anal sex
cies is a research priority. The likely candidates are EBV proteins (Engels et al., 2007). In addition, KSHV can be detected in blood, and
expressed in these malignancies (LMP1, LMP2, and EBNA-​1–​3). transmission can also occur via blood transfusion and organ donation
The identification of good surrogate markers for cancer develop- (Minhas and Wood, 2014).
ment and immune correlates of protection against EBV and disease
are additional concerns (Cohen et  al., 2013). The potential for an
EBV vaccine to prevent malignancy could be directly evaluated Types of Cancer Caused by KSHV
in transplant recipients, particularly EBV-​ seronegative children.
However, the prevention of IM may be the most powerful trigger KSHV is a necessary but not sufficient cause of KS and primary effu-
of further work on EBV vaccine in high-​income countries and, sion lymphoma (PEL), and probably also of multicentric Castleman’s
therefore, a better evaluation of the burden of IM would be essential disease (IARC, 2012a). The estimated number of cancers caused by
to estimate the cost-​benefit of a vaccine against EBV infection or KSHV is 44,000 per year (Table 24–2).
related diseases.
Kaposi’s Sarcoma
KS is a locally aggressive, endothelial tumor that usually presents
KAPOSI’S SARCOMA-​ASSOCIATED HERPESVIRUS with cutaneous lesions in the form of multiple patches or nodules
(Mentzel et  al., 2013). It can also involve mucosal sites, lymph
Nature of the Exposure nodes, and visceral organs, especially in immunosuppressed indi-
viduals. The evidence linking KSHV to the development of KS
Kaposi’s sarcoma-​associated herpesvirus (KSHV), or human herpes- (including HIV-​associated, classical, endemic, and transplant-​asso-
virus 8 (HHV-​8), is a gamma-​2 herpesvirus (rhadinovirus)—​the first ciated variants of the disease) is overwhelming, with over 100 epi-
to be identified that affects humans (Chang et al., 1994; Moore et al., demiological studies (both prospective and case-​ control, across
1996). Morphological characteristics are typical of herpesviruses, con- diverse populations) showing consistent and strong associations.
sisting of a 100–​150 nm particle with a lipid envelope and a double-​ All cohort studies have, however, included only immunosuppressed
stranded DNA genome within, comprising a single contiguous region individuals, mainly HIV-​infected people. Dose–​response relation-
containing all the viral genes, with terminal repeat regions of variable ships are seen between antibody titers against KSHV and risk of
length on either side. There are at least four major subtypes of KSHV KS, as well as with viral load in blood and risk of KS (reviewed by
with a distinctive geographical distribution:  Africa (mainly subtype IARC, 2012a).
B), Mediterranean (C), northern Europe and North and South America Even before the HIV epidemic (see section on exposure parameters
(A), and the Far East (D). that influence risk), KS had a much greater geographical variation in
Humans are the natural host for KSHV, in whom it can establish incidence than most other malignancies. In Sub-​Saharan Africa, it
lifelong latent infection within B cells (reviewed in IARC, 2012a). was common (> 6 per 1,000) in a belt that stretched westward and
Other cell types infected include endothelial and spindle cells of southward from Uganda into Malawi, but was relatively rare in South
 453

Infectious Agents 453
Africa (Cook-​Mozaffari et al., 1998). In western Uganda, for instance, KSHV endemic settings (Ziegler et  al., 1997; Ziegler et  al., 2003).
it represented up to 9% of all cancers in men. KS was also endemic, Furthermore, those who acquire KSHV subsequent to HIV infection
although much rarer, in countries around the Mediterranean, particu- have substantially higher risk of KS than those who are infected prior
larly in Italy, Greece, and the Middle East, but was almost nonexis- to acquiring HIV (Martin et al., 1998).
tent elsewhere, except in immigrants from these endemic countries Other (as yet ill-​defined) cofactors may have the most effect on
(Franceschi and Geddes, 1995). Variation in KS incidence before the KSHV transmission or KS development in the absence of concurrent
HIV epidemic correlates well with the subsequently discovered world- HIV infection or other forms of immune suppression (Dedicoat and
wide distribution of KSHV infection. However, infection prevalence is Newton, 2003). These include malaria (Franceschi and Geddes, 1995;
similar in the two sexes, whereas KS incidence is consistently higher Serraino et al., 2003), volcanic soils (Ziegler, 1993), and exposure to
in men than in women (Bohlius et al., 2014; Franceschi and Geddes, certain plants (Whitby et al., 2007).
1995). In Sub-​Saharan Africa, KS is also found among children.
After the outbreak of the HIV epidemic in the 1980s, KS frequency
increased steeply until cART was introduced (i.e., in 1996 in high-​ Future Research
income countries) (IARC, 1996, 2012a, 2014). In the United States, Although cART can prevent and control the disease, even if started
KS incidence peaked in 1990–​1995 (1117 per 100,000 people with in the presence of very low CD4 levels, KS management remains a
AIDS) but declined in HIV-​infected people by 25% per year in 1996–​ significant clinical problem in HIV-​infected people (Nguyen et  al.,
2000 and by 6% in 2000–​2010 (Robbins et al., 2014b). Similar tem- 2008). A systematic review (Gbabe et  al., 2014) showed that cART
poral patterns were reported in Europe, although even in HIV-​infected plus chemotherapy may be beneficial in reducing disease progression
people with restored immunity, KS remained more frequent than in compared to cART alone in patients with severe or progressive KS, but
the general population (Franceschi et al., 2010; Hleyhel et al., 2013). that there was no significant difference between the use of liposomal
HIV-​associated KS hit Sub-​Saharan African populations of both doxorubicin, liposomal daunorubicin, and paclitaxel. In Sub-​Saharan
sexes more heavily than populations in Western countries, and cART, Africa, advanced stage at KS diagnosis and/​or late referral for treat-
though clearly efficacious, has been scaled up only since 2004 and ment is frequent and the availability of chemotherapy very limited
is often started at a very low CD4 level (Bohlius et al., 2014). In the (Krown, 2011). Better understanding of KS pathogenesis is leading
general population of some countries (e.g., Uganda, in 1991–​1995), to rational drug design, with a number of promising approaches under
KS was reported to be the most common cancer in men and the second investigation (reviewed by Mesri et al., 2010).
most common cancer in women (after cervical cancer). It subsequently However, the absence of curative drugs or a safe and effective
declined modestly from 1996 to 2010 by 2.1% per year in men and vaccine against KSHV highlights the need to better understand the
0.3% in women (Wabinga et al., 2014). genetic and environmental factors that impact on transmission. It is
an enigma as to why KSHV is not more widespread in human popu-
Primary Effusion Lymphoma and Multicentric lations, but rather is sustained at much higher prevalence in parts of
Castleman’s Disease Africa compared to the rest of the world. Although prevalent in West
Primary effusion lymphoma (PEL) is a rare subgroup of B-​cell malig- Africa, KSHV is rare among black Americans—​transmission follow-
nancies that presents with body cavity (pleural, peritoneal, or pericar- ing migration from Africa was clearly reduced and the strain of KSHV
dial) effusions usually, but not always, in the context of HIV infection present in North America is not that found in Africa, but that found
or other forms of immune suppression. KSHV is identified in all cases in Europe. A better understanding of cofactors for transmission may
and indeed the presence of the virus in tumor cells is now part of provide novel insights into how best to reduce the burden of KSHV in
the diagnostic criteria for PEL (IARC, 2012a). Castleman’s disease is settings where infection is prevalent.
not strictly a malignancy, but rather a polyclonal lymphoproliferation
that can progress to lymphoma. Because of its rarity, evidence for an
association with KSHV comes mainly from case-​series, which are HUMAN T-​CELL LEUKEMIA VIRUS TYPE 1
consistent in linking the virus to the disease. A  number of tumors,
notably multiple myeloma, have been linked to KSHV in some stud- Human T-​cell leukemia virus type 1 (HTLV-​1) is a retrovirus belonging
ies, but for none of these is the evidence sufficient to suggest causality to the family retroviridae, genus deltaretrovirus. Discovered in 1980
(IARC, 2012a). (Poiesz et al., 1980), it has an RNA genome that is reverse transcribed
into DNA and then integrated into cellular DNA, predominantly of
CD4+ T cells. There are seven genotypes identified to date, five of
Mechanisms of Carcinogenesis which are found only in Sub-​Saharan Africa and one of which is found
only in Asia; the human virus probably arose via interspecies trans-
In KS, primary endothelial cells undergo spindle cell formation, which
mission of related simian viruses (Verdonck et al., 2007). An estimated
express markers of the lymphatic endothelium. KSHV infection has
10–​20 million individuals worldwide are infected and 3–​8 million of
been shown to induce most steps in the process of malignant transfor-
those are in Sub-​Saharan Africa. More than 90% of those infected
mation, including cell proliferation, inhibition of apoptosis, genomic
will remain asymptomatic carriers throughout their lives (Gessain and
instability, and cell migration with associated tumor progression. At
Cassar, 2012).
least one KSHV gene product is expressed in all infected tumor cells
Estimation of the global prevalence of HTLV-​1 is based primar-
in all KSHV-​associated cancers and in vitro (IARC, 2012a; Mesri
ily on data from screening of healthy blood donors; for large parts
et al., 2010).
of the world, information is sparse or absent. Highest prevalence has
been reported in Japan, parts of Africa, the Caribbean Islands, areas of
Parameters That Influence Risk Central and South America, and Northern Oceania. The southwestern
islands of Japan are particularly affected, with prevalence estimates of
A number of cofactors have been suggested, but only for HIV and up to 20% (IARC, 2012a), followed by the Caribbean and countries
other causes of immune suppression is there evidence that KS risk in Sub-​Saharan Africa (up to 5%) (Fox et al., 2015). Contrary to HIV,
is increased, significantly and consistently in all populations (see HTLV-​1 predominantly exists as a cell-​associated provirus, and it is
Chapter 25 on immunologic factors). transmitted from human to human through infected lymphocytes. The
The higher incidence of KS in men despite a similar KSHV preva- predominant route of acquisition is through mother-​to-​child transmis-
lence in men and women points to the male sex as a cofactor for KS sion at birth and more commonly through breastfeeding (especially
onset (Bohlius et  al., 2014; Franceschi and Geddes, 1995). There is ≥ 6-​month breastfeeding), and has been correlated with mother–​child
also circumstantial evidence to suggest that later age at infection with concordance of HLA types and proviral load (Biggar et  al., 2006).
KSHV is associated with increased risk of KS, although this cannot Other routes include sexual intercourse, blood transfusion, organ
explain the development of tumors among young children seen in transplantation, and contaminated needle reuse.
45

454 PART III:  THE CAUSES OF CANCER


HTLV-​1 is the causative agent of adult T-​cell leukemia/​lymphoma (IARC, 1994b, 2012a). Infection occurs via exposure to contaminated
(ATLL) with an estimated 3000 cases per year (Table 24–2). It also fresh water and is most frequent in parts of Africa (mainly along the
causes the progressive, chronic, disabling HTLV-​1-​associated myelop- Nile River) and the Middle East. On release from freshwater snails,
athy/​tropical spastic paraparesis (HAM/​TSP), as well as other inflam- a free-​living form of S. haematobium can enter a human host through
matory conditions such as infective dermatitis and uveitis (Martin the skin, eventually migrating to live in the veins that drain the bladder.
et  al., 2014). ATLL is an aggressive malignancy of peripheral T Eggs are released and pass in urine back into water and then to the snail.
cells, with short survival in its acute form. Risk of disease increases Infection can be ubiquitous in endemic regions and begins in child-
with age, time since infection, and is higher among male carriers of hood. Many infections are asymptomatic. However, egg deposition in
HTLV-​1 compared to female carriers. It occurs almost exclusively in the bladder can cause significant inflammatory responses leading to
areas where infection is endemic, such as Japan, the Caribbean, and oxidative stress and nitric oxide–​induced genotoxicity, resulting in the
West Africa; in other areas, cases are generally among immigrants development of an estimated 7,000 cases of bladder cancer worldwide
from endemic populations. HTLV-​1 is considered a necessary cause in 2012 (Table 24–2), mainly squamous cell carcinoma (Bedwani et al.,
of ATLL, and presence of infection is part of the diagnostic criteria. 1998). Tobacco smoking is a cofactor of S. haematobium infection in
The host immune system is thought to play a critical role in control bladder cancer. Formerly a disease of rural areas, urban schistosomia-
of infection in the majority of carriers. Mechanistically, HTLV-​1 can sis from man-​made reservoir and irrigation systems is an increasing
transform CD4-​positive T lymphocytes via the action of the Tax pro- problem in many countries. Despite large-​scale pharmacological inter-
tein in vitro (reviewed in IARC, 2012a). ventions, particularly using praziquantel, evidence for the impact of
The prognosis of HTLV-​1 related diseases is poor and no vaccine repeated treatment for schistosomiasis on the prevention and control of
is yet available, highlighting the importance of prevention. Screening bladder cancer remains limited (IARC, 1994b, 2012a).
of blood products has also been shown to be effective in reducing
parenteral transmission, and use of condoms should be considered
by HTLV-​ 1 serologic-​discordant couples. Prevention of HTLV-​ 1 Opisthorchis viverrini and Clonorchis sinensis
transmission in neonates appears to be especially important because The liver flukes Opisthorchis viverrini (O. viverrini) and Clonorchis
of the association of ATLL with childhood infection (IARC, 1996). sinensis (C.  sinensis) are food-​borne trematode worms of the same
Limiting the duration of breastfeeding is of particular value. An inter- genus, but different species. It is estimated that about 45 million peo-
vention program to screen and counsel HTLV-​1-​seropositive women ple are infected, primarily in China, Thailand, Vietnam, Laos, Korea,
began in Japan in the late 1980s and has been shown to prevent 90% and Cambodia (IARC, 1994b, 2012a). C.  sinensis is approximately
of maternal transmission to infants (Hino, 1990). However, such a three times more frequent than O. viverrini. In endemic areas they are
policy can be adopted only where safe alternatives to breastfeeding an important cause of cholangiocarcinoma, resulting in approximately
are available. 1300 cases each year (Table 24–2). Infection in humans is via con-
sumption of contaminated undercooked or raw freshwater fish that are
used as traditional snacks or sauces. Other fish-​eating mammals can
MERKEL CELL POLYOMAVIRUS be affected, and domestic dogs and cats are an important reservoir of
infection. On release from snails, the parasites can enter many fish
Merkel cell polyomavirus (MCV) is a recently discovered virus (Feng species, and from there, infect humans. They eventually inhabit the
et al., 2008) that affects epithelial-​neuroendocrine cells located in the intrahepatic bile ducts and secrete eggs back into the environment
basal membrane of the skin (see IARC, 2014, for a review). MCV-​ via feces. As with schistosomiasis, infection is often asymptomatic
DNA is detected by PCR using different primers specific to genes and the development of cancer appears to be related to decade-​long
encoding MCV large T-​antigen (LT), small T-​antigen (sT), and capsid chronic inflammation and the resulting genotoxic oxidative stress.
viral proteins. Case-​series showed that MCV-​DNA can be detected Cholangiocarcinoma is usually late stage at diagnosis and is rarely
in the vast majority of cases of Merkel cell carcinoma, a rare neu- curable. Improvement of sanitation, aquaculture control, and dietary
roectodermal tumor that tends to recur locally and can also produce education could diminish the transmission of liver flukes.
metastases. Quantitative analyses revealed that > 90% of Merkel cell
carcinomas bear > 0.6 viral copies per cell. Clonal integration of viral
DNA in the cell genome of MCV-​positive carcinomas has been con- OTHER SUSPECTED CARCINOGENIC AGENTS
sistently reported. Among various MCV antibodies, seropositivity for
MCV LT and sT is specifically associated with cancer, while very A few other infectious agents have been suspected to be carcinogens
rare in healthy controls. Case-​control studies showed elevated and in humans, but epidemiological evidence was considered insufficient
consistent odds ratios for serological markers of MCV infection and, by IARC (Table 24–1).
especially, for markers of early gene expression. Immunosuppressed P. falciparum, like HIV, is not carcinogenic per se, but it can
persons are at > 10 times increased risk of Merkel cell carcinoma, and increase the carcinogenicity of other infections, notably EBV. There
sun exposure is suspected to contribute to MCV carcinogenicity (see are estimated to be globally about 198 million cases of malaria each
Chapter 25 on immunologic factors). year, most caused by one of four species (WHO, 2014). The major-
ity of malaria deaths are associated with P. falciparum, and 90% of
those occur in children < 5 years, primarily in Sub-​Saharan Africa.
PARASITES The life cycle has several stages and involves both humans and the
Anopheles mosquito. Co-​infection of P. falciparum with EBV seems
Three parasites are well-​established carcinogens in humans (reviewed to cause dysregulation of the virus, thereby facilitating the develop-
in IARC, 2012a). ment of BL (reviewed in IARC, 2014). Numerous ecological studies
link BL to malaria, both geographically (Burkitt, 1962) and over time
and, more recently, case-​control studies among children in Uganda
Schistosoma haematobium and Malawi demonstrated increased risks of the tumor with increasing
Schistosomes are parasitic trematode worms that live in the blood ves- titers of antibodies against malaria—​indeed the highest risk of BL was
sels of human hosts. There are six species that cause significant pathol- among those with high titers of antibodies against both malaria and
ogy in man, varying in their geographic distribution, intermediate EBV (Carpenter et al., 2008; Mutalima et al., 2008). Clear evidence
host, and location in human tissues; an estimated 200 million people for other malaria species and for other cancer sites is lacking (IARC,
are infected globally. They have been linked to various human tumors, 2014).
including cancers of the bladder, cervix, prostate, liver and colorectum, Two cutaneous HPV types, HPV5 and 8 are often detected in squa-
but only for Schistosoma haematobium (S.  haematobium) in relation mous cell carcinomas of the skin in Epidermodysplasia verruciformis
to bladder cancer is the evidence sufficiently robust to claim causality patients. HPV5 and 8 belong to the ß-​genus that, together with other
 45

Infectious Agents 455
HPV genera, includes many more HPV types than the mucosal α-​genus Areia M, Carvalho R, Cadime AT, Rocha GF, and Dinis-​Ribeiro M. 2013.
(IARC, 2012a). Cutaneous HPVs are also more ubiquitous in the gen- Screening for gastric cancer and surveillance of premalignant lesions: a
eral population than mucosal types and have been found in squamous systematic review of cost-​ effectiveness studies. Helicobacter, 18(5),
cell carcinomas of the skin and its precursor, actinic keratosis, also in 325–​337.
Armitage P, and Doll R. 1954. The age distribution of cancer and a multi-​stage
immunocompetent individuals. However, viral DNA is not integrated theory of carcinogenesis. Br J Cancer, 8(1), 1–​12.
within the host genome, is often present at low copy number, and is Armstrong GL, Wasley A, Simard EP, et al. 2006. The prevalence of hepatitis C
not necessary for expression of the malignant phenotype (Tommasino, virus infection in the United States, 1999 through 2002. Ann Intern Med,
2014). Transgenic mice expressing beta-​HPV E6/E7 proteins develop 144(10), 705–​714.
skin squamous cell carcinomas on ultraviolet (UV) light exposure as Asenjo LM, and Gisbert JP. 2007. [Prevalence of Helicobacter pylori infection
these proteins hamper the repair of UV-​induced DNA damage (Howley in gastric MALT lymphoma: a systematic review]. Rev Esp Enferm Dig,
and Pfister, 2015). Finally, contrary to mucosal HPV, a clear identifi- 99(7), 398–​404.
cation of high-​risk species/​types is lacking for cutaneous HPV types, Atherton JC, Cao P, Peek RM, Jr., et al. 1995. Mosaicism in vacuolating cyto-
whose truly causal role in skin cancer remains uncertain (IARC, 2012a). toxin alleles of Helicobacter pylori:  association of specific vacA types
with cytotoxin production and peptic ulceration. J Biol Chem, 270(30),
An especially interesting possibility is the involvement of the gut 17771–​17777.
microbiome in the onset of colorectal cancer (CRC), the most frequent Balfour HH, Jr., Odumade OA, Schmeling DO, et al. 2013. Behavioral, viro-
cancer in nonsmokers worldwide. The wide range in gut microbiota logic, and immunologic factors associated with acquisition and severity
composition and associated metabolic functions among individuals of primary Epstein-​Barr virus infection in university students. J Infect
provides a rationale for proposing the existence of associations with Dis, 207(1), 80–​88. PMCID: Pmc3523797.
disease processes, including CRC (Doll and Peto, 1981). A case-​con- Banatvala N, Mayo K, Megraud F, et  al. 1993. The cohort effect and
trol study based on 16S rRNA sequencing of fecal samples showed Helicobacter pylori. J Infect Dis, 168(1), 219–​221.
that CRC cases had decreased microbial diversity compared to con- Barnes L, Eveson JW, Reichart P, and Sidransky D (Eds.). 2005. WHO clas-
trols, lower levels of Clostridia, and significantly increased levels of sification of tumours. Pathology and genetics of head and neck tumours
(3rd ed.). Lyon, France: IARC.
Porphyromonas and Fusobacterium (Ahn et al., 2013). The CRC sub- Bedwani R, Renganathan E, El Kwhsky F, et  al. 1998. Schistosomiasis and
type associated with Fusobacterium may have molecularly distinct the risk of bladder cancer in Alexandria, Egypt. Br J Cancer, 77(7),
features (i.e., high degree of CpG island methylation, and high rate of 1186–​1189.
mutations overall, though not of the TP53 gene) (Tahara et al., 2014). Biggar RJ, Ng J, Kim N, et al. 2006. Human leukocyte antigen concordance
Of note, CRC occurrence is known to be influenced by host genetics and the transmission risk via breast-​feeding of human T cell lymphotropic
and factors such as obesity, nutrition, and exercise that also influence virus type I. J Infect Dis, 193(2), 277–​282. PMID: 16362892.
and can be influenced by gut microbiota. Bohlius J, Valeri F, Maskew M, et al. 2014. Kaposi’s Sarcoma in HIV-​infected
The discovery in human bilis and the biliary tract of Helicobacter patients in South Africa: Multicohort study in the antiretroviral therapy
species other than H. pylori raised the possibility of an association of era. Int J Cancer, 135(11), 2644–​2652. PMCID: Pmc4415728.
Bravi F, Bosetti C, Tavani A, Gallus S, and La VC. 2013. Coffee reduces risk for
these bacteria with cancer of the gallbladder and extrabiliary tract, a hepatocellular carcinoma: an updated meta-​analysis. Clin Gastroenterol
group of malignancies whose incidence varies substantially across the Hepatol, 11(11), 1413–​1421.
world (de Martel et al., 2009). A case-​control study showed an associa- Bray F, Jemal A, Grey N, Ferlay J, and Forman D. 2012. Global cancer tran-
tion of extra-​hepatic cholangiocarcinoma with PCR-​detected H.  bilis sitions according to the Human Development Index (2008–​ 2030):  a
but not H. hepaticus or Salmonella typhi in Mexico, a high-​incidence population-​based study. Lancet Oncol, 13(8), 790–​801.
area for these malignancies (Segura-​Lopez et al., 2015). An association Brenner H, Arndt V, Stegmaier C, Ziegler H, and Rothenbacher D. 2004. Is
of seropositivity for H.  pylori with biliary tract cancer was reported Helicobacter pylori infection a necessary condition for noncardia gastric
in the Finnish Alpha-​Tocopherol, Beta-​Carotene Cancer Prevention cancer? Am J Epidemiol, 159(3), 252–​258.
prospective study (Murphy et al., 2014). On account of the low speci- Brown LM. 2000. Helicobacter pylori: epidemiology and routes of transmis-
sion. Epidemiol Rev, 22(2), 283–​297.
ficity of H. pylori serology, these findings may reflect cross-​reactivity Bruni L, Diaz M, Castellsagué X, et al. 2010. Cervical human papillomavirus
with non-​pylori Helicobacter species, which could contribute to cancer prevalence in 5 continents: meta-​analysis of 1 million women with nor-
onset through chronic inflammation and epithelial DNA damage. mal cytological findings. J Infect Dis, 202(12), 1789–​1799.
Other associations between infections and cancer may emerge in the Burkitt D. 1962. A children’s cancer dependent on climatic factors. Nature,
future, driven by the discovery of more accurate biomarkers for candi- 194, 232–​234. PMID: 13874900.
date agents and a finer molecular characterization of cancer types into Camargo MC, Kim KM, Matsuo K et al. 2016. Anti-​Helicobacter pylori anti-
subsites that may differ from an etiologic point of view. body profiles in Epstein-​Barr virus (EBV)-​positive and EBV-​negative
gastric cancer. Helicobacter, 21(2), 153–​157. PMCID: PMC5003173.
Camargo MC, Murphy G, Koriyama C, et al. 2011. Determinants of Epstein-​
ACKNOWLEDGMENT Barr virus-​positive gastric cancer: an international pooled analysis. Br J
Cancer, 105(1), 38–​43. PMCID: PMC3137422.
The authors would like to thank Dr. Jin Young Park for her review of Cardenas-​Mondragon MG, Torres J, Flores-​Luna L, et al. 2015. Case-​control
the H. pylori section and Dr. Gary Clifford for suggestions on the HPV study of Epstein-​Barr virus and Helicobacter pylori serology in Latin
American patients with gastric disease. Br J Cancer, 112(12), 1866–​
section. Mrs. Susan Gamon provided technical assistance. 1873. PMCID: PMC4580389.
Carozzi F, Confortini M, Dalla Palma P, et al. 2008. Use of p16-​INK4A over-
References
expression to increase the specificity of human papillomavirus testing: a
Abu-​Amara M, Cerocchi O, Malhi G, et al. 2016. The applicability of hepa- nested substudy of the NTCC randomised controlled trial. Lancet Oncol,
tocellular carcinoma risk prediction scores in a North American patient 9(10), 937–​945.
population with chronic hepatitis B infection. Gut, 65(8), 1347–​1358. Carpenter LM, Newton R, Casabonne D, et  al. 2008. Antibodies against
PMID: 25934760. malaria and Epstein-​Barr virus in childhood Burkitt lymphoma: a case-​
Ahn J, Sinha R, Pei Z, et al. 2013. Human gut microbiome and risk for colorectal control study in Uganda. Int J Cancer, 122(6), 1319–​1323. PMID:
cancer. J Natl Cancer Inst, 105(24), 1907–​1911. PMCID: Pmc3866154. 18000823.
Alemany L, Saunier M, Alvarado-​Cabrero I, et al. 2015. Human papillomavirus Castle PE, Rodriguez AC, Burk RD, et  al. 2009. Short term persistence of
DNA prevalence and type distribution in anal carcinomas worldwide. Int human papillomavirus and risk of cervical precancer and cancer: popula-
J Cancer, 136(1), 98–​107. tion based cohort study. BMJ, 339, b2569.
Alter HJ, and Seeff LB. 2000. Recovery, persistence, and sequelae in hepatitis Cavaleiro-​Pinto M, Peleteiro B, Lunet N, and Barros H. 2011. Helicobacter
C virus infection: a perspective on long-​term outcome. Semin Liver Dis, pylori infection and gastric cardia cancer:  systematic review and meta-​
20(1), 17–​35. analysis. Cancer Causes Control, 22(3), 375–​387.
Arase Y, Kobayashi M, Suzuki F, et al. 2013. Effect of type 2 diabetes on risk Censini S, Lange C, Xiang Z, et  al. 1996. cag, a pathogenicity island of
for malignancies includes hepatocellular carcinoma in chronic hepatitis Helicobacter pylori, encodes type I-​specific and disease-​associated viru-
C. Hepatology, 57(3), 964–​973. lence factors. Proc Natl Acad Sci U S A, 93(25), 14648–​14653.
456

456 PART III:  THE CAUSES OF CANCER


Chang ET, and Adami HO. 2006. The enigmatic epidemiology of nasopharyn- de Vuyst H, Clifford GM, Nascimento MC, Madeleine MM, and Franceschi S.
geal carcinoma. Cancer Epidemiol Biomarkers Prev, 15(10), 1765–​1777. 2009. Prevalence and type distribution of human papillomavirus in carci-
PMID: 17035381. noma and intraepithelial neoplasia of the vulva, vagina and anus: a meta-​
Chang KC, Hung CH, Lu SN, et al. 2012. A novel predictive score for hepato- analysis. Int J Cancer, 124(7), 1626–​1636.
cellular carcinoma development in patients with chronic hepatitis C after Dedicoat M, and Newton R. 2003. Review of the distribution of Kaposi’s sarcoma-​
sustained response to pegylated interferon and ribavirin combination associated herpesvirus (KSHV) in Africa in relation to the incidence of
therapy. J Antimicrob Chemother, 67(11), 2766–​2772. Kaposi’s sarcoma. Br J Cancer, 88(1), 1–​3. PMCID: Pmc2376771.
Chang Y, Cesarman E, Pessin MS, et  al. 1994. Identification of herpesvirus-​ Dedicoat M, Newton R, Alkharsah KR, et al. 2004. Mother-​to-​child transmis-
like DNA sequences in AIDS-​ associated Kaposi’s sarcoma. Science, sion of human herpesvirus-​8 in South Africa. J Infect Dis, 190(6), 1068–​
266(5192), 1865–​1869. PMID: 7997879. 1075. PMID: 15319855.
Chaturvedi AK, Anderson WF, Lortet-​Tieulent J, et al. 2013. Worldwide trends den Boon JA, Pyeon D, Wang SS, et al. 2015. Molecular transitions from pap-
in incidence rates for oral cavity and oropharyngeal cancers. J Clin Oncol, illomavirus infection to cervical precancer and cancer:  Role of stromal
31(36), 4550–​4559. estrogen receptor signaling. Proc Natl Acad Sci U S A, 112(25), E3255–​
Cho LY, Yang JJ, Ko KP, et al. 2011. Coinfection of hepatitis B and C viruses 3264. PMCID: PMC4485108.
and risk of hepatocellular carcinoma:  systematic review and meta-​ Doll R. 1978. An epidemiological perspective of the biology of cancer. Cancer
analysis. Int J Cancer, 128(1), 176–​184. Res, 38(11 Pt 1), 3573–​3583.
Choi J, Kim SG, Yoon H, et al. 2014. Eradication of Helicobacter pylori after Doll R, and Peto R. 1981. The causes of cancer: quantitative estimates of avoid-
endoscopic resection of gastric tumors does not reduce incidence of meta- able risks of cancer in the United States today. J Natl Cancer Inst, 66(6),
chronous gastric carcinoma. Clin Gastroenterol Hepatol, 12(5), 793–​800. 1191–​1308.
Chuang SC, Lee YC, Hashibe M, et  al. 2010. Interaction between cigarette Donato F, Boffetta P, and Puoti M. 1998. A meta-​analysis of epidemiological
smoking and hepatitis B and C virus infection on the risk of liver cancer: a studies on the combined effect of hepatitis B and C virus infections in
meta-​analysis. Cancer Epidemiol Biomarkers Prev, 19(5), 1261–​1268. causing hepatocellular carcinoma. Int J Cancer, 75(3), 347–​354.
Chung CH, Bagheri A, and D’Souza G. 2014. Epidemiology of oral human Donato F, Tagger A, Gelatti U, et al. 2002. Alcohol and hepatocellular carci-
papillomavirus infection. Oral Oncol, 50(5), 364–​369. noma: the effect of lifetime intake and hepatitis virus infections in men
Chung SH, Franceschi S, and Lambert PF. 2010. Estrogen and ERalpha: cul- and women. Am J Epidemiol, 155(4), 323–​331.
prits in cervical cancer? Trends Endocrinol Metab, 21(8), 504–​511. Dondog B, Lise M, Dondov O, Baldandorj B, and Franceschi S. 2011. Hepatitis
Clifford GM, Rickenbach M, Polesel J, et al. 2008. Influence of HIV-​related B and C virus infections in hepatocellular carcinoma and cirrhosis in
immunodeficiency on the risk of hepatocellular carcinoma. AIDS, 22(16), Mongolia. Eur J Cancer Prev, 20(1), 33–​39.
2135–​2141. El-​Serag HB. 2012. Epidemiology of viral hepatitis and hepatocellular carci-
Cohen JI, Mocarski ES, Raab-​Traub N, Corey L, and Nabel GJ. 2013. The noma. Gastroenterology, 142(6), 1264–​1273.
need and challenges for development of an Epstein-​Barr virus vaccine. El-​Serag HB, Hampel H, and Javadi F. 2006. The association between diabetes
Vaccine, 31 (Suppl 2), B194–​196. PMCID: Pmc3636506. and hepatocellular carcinoma: a systematic review of epidemiologic evi-
Combes JD, Chen AA, and Franceschi S. 2014. Prevalence of human papil- dence. Clin Gastroenterol Hepatol, 4(3), 369–​380.
lomavirus in cancer of the oropharynx by gender. Cancer Epidemiol El-​Serag HB, Johnson ML, Hachem C, and Morgana RO. 2009. Statins are
Biomarkers Prev, 23(12), 2954–​2958. associated with a reduced risk of hepatocellular carcinoma in a large
Combes JD, and Franceschi S. 2014. Role of human papillomavirus in non-​ cohort of patients with diabetes. Gastroenterology, 136(5), 1601–​1608.
oropharyngeal head and neck cancers. Oral Oncol, 50(5), 370–​379. El-​Serag HB, Kramer J, Duan Z, and Kanwal F. 2014. Racial differences in the
Cook-​Mozaffari P, Newton R, Beral V, and Burkitt DP. 1998. The geographical progression to cirrhosis and hepatocellular carcinoma in HCV-​infected
distribution of Kaposi’s sarcoma and of lymphomas in Africa before the veterans. Am J Gastroenterol, 109(9), 1427–​1435.
AIDS epidemic. Br J Cancer, 78(11), 1521–​1528. PMCID: PMC2063225. Engels EA, Atkinson JO, Graubard BI, et  al. 2007. Risk factors for human
Correa P, and Piazuelo MB. 2012. The gastric precancerous cascade. J Dig Dis, herpesvirus 8 infection among adults in the United States and evidence
13(1), 2–​9. PMCID: PMC3404600. for sexual transmission. J Infect Dis, 196(2), 199–​207. PMID: 17570106.
Cover TL, Tummuru MK, Cao P, Thompson SA, and Blaser MJ. 1994. Epstein MA, Achong BG, and Barr YM. 1964. Virus particles in cultured lym-
Divergence of genetic sequences for the vacuolating cytotoxin among phoblasts from Burkitt’s lymphoma. Lancet, 283, 702–​703.
Helicobacter pylori strains. J Biol Chem, 269(14), 10566–​10573. EUROGAST. 1993. An international association between Helicobacter pylori
Crosbie EJ, Einstein MH, Franceschi S, and Kitchener HC. 2013. Human papil- infection and gastric cancer. The EUROGAST Study Group. Lancet,
lomavirus and cervical cancer. Lancet, 382(9895), 889–​899. 341(8857), 1359–​1362.
Cullen M, Boland JF, Schiffman M, et al. 2015. Deep sequencing of HPV16 Evans AA, Chen G, Ross EA, et al. 2002. Eight-​year follow-​up of the 90,000-​
genomes: A new high-​throughput tool for exploring the carcinogenicity person Haimen City cohort:  I.  Hepatocellular carcinoma mortality, risk
and natural history of HPV16 infection. Papillomavirus Res, 1, 3–​11. factors, and gender differences. Cancer Epidemiol Biomarkers Prev,
PMCID: PMC4669577. 11(4), 369–​376.
Dal Maso L, and Franceschi S. 2006. Hepatitis C virus and risk of lymphoma Evans AA, Fine M, and London WT. 1997. Spontaneous seroconversion in hep-
and other lymphoid neoplasms: a meta-​analysis of epidemiologic studies. atitis B e antigen-​positive chronic hepatitis B: implications for interferon
Cancer Epidemiol Biomarkers Prev, 15(11), 2078–​2085. therapy. J Infect Dis, 176(4), 845–​850.
Davis GL, Alter MJ, El-​Serag H, Poynard T, and Jennings LW. 2010. Aging Fattovich G, Bortolotti F, and Donato F. 2008. Natural history of chronic hepa-
of hepatitis C virus (HCV)-​ infected persons in the United States:  a titis B: special emphasis on disease progression and prognostic factors. J
multiple cohort model of HCV prevalence and disease progression. Hepatol, 48(2), 335–​352.
Gastroenterology, 138(2), 513–​521, 521. Feng H, Shuda M, Chang Y, and Moore PS. 2008. Clonal integration of a
de Martel C, Maucort-​ Boulch D, Plummer M, and Franceschi S. 2015a. polyomavirus in human Merkel cell carcinoma. Science, 319(5866),
Worldwide relative contribution of hepatitis B and C viruses in hepatocel- 1096–​1100.
lular carcinoma. Hepatology, 62(4), 1190–​1200. Ferlay J, Shin HR, Bray F, et al. (Writers). (2010). GLOBOCAN 2008 v1.2,
de Martel C, Plummer M, Parsonnet J, van Doorn LJ, and Franceschi S. 2009. Cancer Incidence and Mortality Worldwide:  IARC CancerBase No. 10
Helicobacter species in cancers of the gallbladder and extrahepatic biliary [Internet].
tract. Br J Cancer, 100(1), 194–​199. Ferlay J, Soerjomataram I, Ervik M, et  al. 2013. GLOBOCAN 2012 v1.0,
de Martel C, Shiels MS, Franceschi S, et  al. 2015b. Cancers attributable to Cancer Incidence and Mortality Worldwide:  IARC CancerBase No.
infections among adults with HIV in the United States. AIDS, 29(16), 11 [Internet]. Available from:  http://​globocan.iarc.fr Accessed June
2173–​2181. 20, 2015.
de Mitri MS, Poussin K, Baccarini P, et al. 1995. HCV-​associated liver cancer Figueiredo C, Machado JC, Pharoah P, et  al. 2002. Helicobacter pylori and
without cirrhosis. Lancet, 345(8947), 413–​415. interleukin 1 genotyping: an opportunity to identify high-​risk individuals
de Sanjosé S, Alemany L, Ordi J, et al. 2013. Worldwide human papillomavi- for gastric carcinoma. J Natl Cancer Inst, 94(22), 1680–​1687.
rus genotype attribution in over 2000 cases of intraepithelial and invasive Ford AC, Delaney BC, Forman D, and Moayyedi P. 2006. Eradication therapy
lesions of the vulva. Eur J Cancer, 49(16), 3450–​3461. for peptic ulcer disease in Helicobacter pylori positive patients. Cochrane
de Sanjosé S, Mbisa G, Perez-​Alvarez S, et al. 2009. Geographic variation in Database Syst Rev 2, CD003840.
the prevalence of Kaposi sarcoma-​associated herpesvirus and risk factors Ford AC, Forman D, Hunt RH, Yuan Y, and Moayyedi P. 2014. Helicobacter
for transmission. J Infect Dis, 199(10), 1449–​1456. pylori eradication therapy to prevent gastric cancer in healthy asymp-
de Villiers EM, Fauquet C, Broker TR, Bernard HU, and zur HH. 2004. tomatic infected individuals: systematic review and meta-​analysis of ran-
Classification of papillomaviruses. Virology, 324(1), 17–​27. domised controlled trials. BMJ, 348, g3174.
 457

Infectious Agents 457
Forman D, and Sierra M. 2014. The current and projected global burden of Herfs M, Yamamoto Y, Laury A, et al. 2012. A discrete population of squamo-
gastric cancer. In: Helicobacter pylori eradication as a strategy for pre- columnar junction cells implicated in the pathogenesis of cervical cancer.
venting gastric cancer (pp. 5–​15). Lyon, France: International Agency Proc Natl Acad Sci U S A, 109(26), 10516–​10521.
for Research on Cancer (IARC Working Group Reports No. 8). Hino S. 1990. Milk-​borne transmission of HTLV-​1 from carrier mothers and its
Fox JM, Mutalima N, Molyneux E, et  al. 2016. Seroprevalence of HTLV-​1 prevention. Hematol Rev, 3, 223–​233.
and 2 amongst mothers and children in Malawi within the context of a Hleyhel M, Belot A, Bouvier AM, et al. 2013. Risk of AIDS-​defining cancers
systematic review and meta-​analysis of HTLV seroprevalence in Africa. among HIV-​1-​infected patients in France between 1992 and 2009: results
Trop Med Int Health, 21(3), 312–​324. PMID: 26700941. from the FHDH-​ANRS CO4 cohort. Clin Infect Dis, 57(11), 1638–​1647.
Franceschi S, and Geddes M. 1995. Epidemiology of classic Kaposi’s sarcoma, PMID: 23899679.
with special reference to mediterranean population. Tumori, 81(5), 308–​ Hosaka T, Suzuki F, Kobayashi M, et al. 2013. Long-​term entecavir treatment
314. PMID: 8804445. reduces hepatocellular carcinoma incidence in patients with hepatitis B
Franceschi S, Herrero R, Clifford GM, et al. 2006. Variations in the age-​specific virus infection. Hepatology, 58(1), 98–​107.
curves of human papillomavirus prevalence in women worldwide. Int J Howley PM, and Pfister HJ. 2015. Beta genus papillomaviruses and skin can-
Cancer, 119(11), 2677–​2684. cer. Virology, 479–​480, 290–​296. PMCID: PMC4424140.
Franceschi S, Lise M, Clifford GM, et al. 2010. Changing patterns of cancer Huang Y, and Lok AS. 2011. Viral factors and outcomes of chronic HBV infec-
incidence in the early-​and late-​HAART periods: the Swiss HIV Cohort tion. Am J Gastroenterol, 106(1), 93–​95.
Study. Br J Cancer, 103(3), 416–​422. Huo TI, Wu JC, Lee PC, et  al. 1998. Sero-​clearance of hepatitis B surface
Frank C, Mohamed MK, Strickland GT, et al. 2000. The role of parenteral anti- antigen in chronic carriers does not necessarily imply a good prognosis.
schistosomal therapy in the spread of hepatitis C virus in Egypt. Lancet, Hepatology, 28(1), 231–​236.
355(9207), 887–​891. Hutchinson SJ, Bird SM, and Goldberg DJ. 2005. Influence of alcohol on
Freeman AJ, Dore GJ, Law MG, et al. 2001. Estimating progression to cirrhosis the progression of hepatitis C virus infection:  a meta-​ analysis. Clin
in chronic hepatitis C virus infection. Hepatology, 34(4 Pt 1), 809–​816. Gastroenterol Hepatol, 3(11), 1150–​1159.
Fukase K, Kato M, Kikuchi S, et al. 2008. Effect of eradication of Helicobacter IARC. 1994a. Hepatitis viruses. IARC Monogr Eval Carcinog Risks Hum,
pylori on incidence of metachronous gastric carcinoma after endoscopic 59, 1–​286.
resection of early gastric cancer:  an open-​label, randomised controlled IARC. 1994b. Schistosomes, liver flukes and Helicobacter pylori. IARC
trial. Lancet, 372(9636), 392–​397. Monogr Eval Carcinog Risks Hum, 61, 1–​270.
Gage JC, Ajenifuja KO, Wentzensen NA, et al. 2012. The age-​specific preva- IARC. 1995. Human papillomaviruses. IARC Monogr Eval Carcinog Risks
lence of human papillomavirus and risk of cytologic abnormalities in rural Hum, 64, 1–​409.
Nigeria: Implications for screen-​and-​treat strategies. Int J Cancer, 130(9), IARC. 1996. Human immunodeficiency viruses and human T-​cell lympho-
2111–​2117. tropic viruses. IARC Monogr Eval Carcinog Risks Hum, 67, 1–​424.
Gao X, Cui Q, Shi X, et  al. 2011. Prevalence and trend of hepatitis C virus IARC. 1997. Epstein-​Barr virus and Kaposi’s sarcoma herpesvirus/​human her-
infection among blood donors in Chinese mainland: a systematic review pesvirus 8. IARC Monogr Eval Carcinog Risks Hum, 70, 1–​524.
and meta-​analysis. BMC Infect Dis, 11, 88. IARC. 2007. Human papillomaviruses. IARC Monogr Eval Carcinog Risks
Gbabe OF, Okwundu CI, Dedicoat M, and Freeman EE. 2014. Treatment of Hum, 90, 1–​670.
severe or progressive Kaposi’s sarcoma in HIV-​infected adults. Cochrane IARC. 2012a. Biological agents. IARC Monogr Eval Carcinog Risks Hum,
Database Syst Rev, 9, Cd003256. PMID: 25313415. 100B, 1–​475. PMID: 23189750.
Gessain A, and Cassar O. 2012. Epidemiological aspects and world distribution IARC. 2012b. Preamble to the IARC Monographs. In:  IARC Monogr Eval
of HTLV-​1 infection. Front Microbiol, 3, 388. PMCID: PMC3498738. Carcinog Risks Hum, 100B (pp. 11–​34). PMID: 23189750.
Ghoshal UC, Chaturvedi R, and Correa P. 2010. The enigma of Helicobacter IARC. 2012c. Chemical agents and related occupations. IARC Monogr Eval
pylori infection and gastric cancer. Indian J Gastroenterol, 29(3), 95–​100. Carcinog Risks Hum, 100F, 1–​599.
PMCID: PMC3217495. IARC. 2012d. Personal habits and indoor combustions. IARC Monogr Eval
Gillison ML, Broutian T, Pickard RK, et  al. 2012. Prevalence of oral HPV Carcinog Risks Hum, 100E, 1–​575.
infection in the United States, 2009–​2010. JAMA, 307(7), 693–​703. IARC. 2014. Malaria and some polyomaviruses (SV40, BK, JC, AND
Giordano TP, Henderson L, Landgren O, et al. 2007. Risk of non-​Hodgkin lym- MERKEL CELL VIRUSES). IARC Monogr Eval Carcinog Risks Hum,
phoma and lymphoproliferative precursor diseases in US veterans with 104, 1–​353.
hepatitis C virus. JAMA, 297(18), 2010–​2017. IARC Helicobacter pylori Working Group. 2014. Helicobacter pylori eradica-
Gisbert JP, and Greenberg ER. 2014. Potential regimens for the mass eradication of tion as a strategy for preventing gastric cancer. Lyon, France: International
Helicobacter pylori infection. In: Helicobacter pylori eradication as a strat- Agency for Research on Cancer (IARC Working Group Reports No. 8).
egy for preventing gastric cancer (pp. 95–​110). Lyon, France: International International Collaboration of Epidemiological Studies of Cervical Cancer.
Agency for Research on Cancer (IARC Working Group Reports No. 8). 2006. Carcinoma of the cervix and tobacco smoking:  collaborative
Giuliano AR, Lee JH, Fulp W, et al. 2011. Incidence and clearance of genital reanalysis of individual data on 13,541 women with carcinoma of the
human papillomavirus infection in men (HIM):  a cohort study. Lancet, cervix and 23,017 women without carcinoma of the cervix from 23 epide-
377(9769), 932–​940. miological studies. Int J Cancer, 118(6), 1481–​1495.
Goodman KJ, Correa P, Tengana Aux HJ, et al. 1996. Helicobacter pylori infec- International Collaboration of Epidemiological Studies of Cervical Cancer.
tion in the Colombian Andes: a population-​based study of transmission 2007. Cervical cancer and hormonal contraceptives: collaborative reanal-
pathways. Am J Epidemiol, 144(3), 290–​299. ysis of individual data on 16,573 women with cervical cancer and 35,509
Guan P, Clifford GM, and Franceschi S. 2013. Human papillomavirus types in women without cervical cancer from 24 epidemiological studies. Lancet,
glandular lesions of the cervix: a meta-​analysis of published studies. Int J 370, 1609–​1621.
Cancer, 132(1), 248–​250. Islami F, and Kamangar F. 2008. Helicobacter pylori and esophageal cancer
Guan P, Howell-​Jones R, Li N, et  al. 2012. Human papillomavirus types in risk: a meta-​analysis. Cancer Prev Res (Phila), 1(5), 329–​338.
115,789 HPV-​positive women: a meta-​analysis from cervical infection to Jee SH, Ohrr H, Sull JW, and Samet JM. 2004. Cigarette smoking, alcohol
cancer. Int J Cancer, 131(10), 2349–​2359. drinking, hepatitis B, and risk for hepatocellular carcinoma in Korea. J
Hahne SJ, Veldhuijzen IK, Wiessing L, et  al. 2013. Infection with hepatitis Natl Cancer Inst, 96(24), 1851–​1856.
B and C virus in Europe:  a systematic review of prevalence and cost-​ Kanwal F, and El-​Serag HB. 2014. Hepatitis C virus treatment: the unyielding
effectiveness of screening. BMC Infect Dis, 13, 181. chasm between efficacy and effectiveness. Clin Gastroenterol Hepatol,
Hansen S, Vollset SE, Derakhshan MH, et al. 2007. Two distinct aetiologies of 12(8), 1381–​1383.
cardia cancer; evidence from premorbid serological markers of gastric Kanwal F, Kramer JR, Ilyas J, Duan Z, and El-​Serag HB. 2014. HCV genotype 3
atrophy and Helicobacter pylori status. Gut, 56(7), 918–​925. is associated with an increased risk of cirrhosis and hepatocellular cancer in
Hardbower DM, Peek RM, Jr., and Wilson KT. 2014. At the bench: Helicobacter a national sample of U.S. veterans with HCV. Hepatology, 60(1), 98–​105.
pylori, dysregulated host responses, DNA damage, and gastric cancer. J Kjaer SK, Frederiksen K, Munk C, and Iftner T. 2010. Long-​term absolute risk
Leukoc Biol, 96(2), 201–​212. of cervical intraepithelial neoplasia grade 3 or worse following human
Haydon GH, Jarvis LM, Simmonds P, and Hayes PC. 1995. Association papillomavirus infection: role of persistence. J Natl Cancer Inst, 102(19),
between chronic hepatitis C infection and hepatocellular carcinoma. 1478–​1488.
Lancet, 345(8954), 928–​929. Kramer JR, Giordano TP, and El-​Serag HB. 2007. Effect of human immunode-
Helicobacter and Cancer Collaborative Group. 2001. Gastric cancer and ficiency virus and antiretrovirals on outcomes of hepatitis C: a systematic
Helicobacter pylori: a combined analysis of 12 case control studies nested review from an epidemiologic perspective. Clin Gastroenterol Hepatol,
within prospective cohorts. Gut, 49(3), 347–​353. 5(11), 1321–​1328.
458

458 PART III:  THE CAUSES OF CANCER


Krienitz W. 1906. Ueber das Auftreten von Spirochäten verschiedener Form im Miki K. 2011. Gastric cancer screening by combined assay for serum anti-​
Mageninhalt bei Carcinoma ventriculi. Dtsch Med Wochenschr, 32, 872. Helicobacter pylori IgG antibody and serum pepsinogen levels:  “ABC
Krown SE. 2011. Treatment strategies for Kaposi sarcoma in sub-​Saharan method.” Proc Jpn Acad Ser B Phys Biol Sci, 87(7), 405–​414.
Africa: challenges and opportunities. Curr Opin Oncol, 23(5), 463–​468. Minhas V, and Wood C. 2014. Epidemiology and transmission of Kaposi’s
PMCID: PMC3465839. sarcoma-​associated herpesvirus. Viruses, 6(11), 4178–​ 4194.
Lee YC, Chen TH, Chiu HM, et al. 2013. The benefit of mass eradication of PMCID: Pmc4246215.
Helicobacter pylori infection: a community-​based study of gastric cancer Miralles-​Guri C, Bruni L, Cubilla AL, et al. 2009. Human papillomavirus prev-
prevention. Gut, 62(5), 676–​682. alence and type distribution in penile carcinoma. J Clin Pathol, 62(10),
Li BZ, Threapleton DE, Wang JY, et al. 2015. Comparative effectiveness and 870–​878.
tolerance of treatments for Helicobacter pylori:  systematic review and Mizokami M, and Orito E. 1999. Molecular evolution of hepatitis viruses.
network meta-​analysis. BMJ, 351, h4052. PMCID: PMC4541168. Intervirology, 42(2–​3), 159–​165.
Liu CJ, Chen BF, Chen PJ, et al. 2006. Role of hepatitis B viral load and basal Moayyedi P, Soo S, Deeks J, et al. 2006. Eradication of Helicobacter pylori for
core promoter mutation in hepatocellular carcinoma in hepatitis B carri- non-​ulcer dyspepsia. Cochrane Database Syst Rev 2, CD002096.
ers. J Infect Dis, 193(9), 1258–​1265. Mohamoud YA, Mumtaz GR, Riome S, Miller D, and Abu-​Raddad LJ. 2013.
Lok AS, Everhart JE, Di Bisceglie AM, et al. 2011. Occult and previous hepatitis The epidemiology of hepatitis C virus in Egypt: a systematic review and
B virus infection are not associated with hepatocellular carcinoma in United data synthesis. BMC Infect Dis, 13, 288. PMCID: PMC3702438.
States patients with chronic hepatitis C. Hepatology, 54(2), 434–​442. Mohd HK, Groeger J, Flaxman AD, and Wiersma ST. 2013. Global epidemiol-
Lu B, Viscidi RP, Wu Y, et al. 2012. Prevalent serum antibody is not a marker ogy of hepatitis C virus infection: new estimates of age-​specific antibody
of immune protection against acquisition of oncogenic HPV16 in men. to HCV seroprevalence. Hepatology, 57(4), 1333–​1342.
Cancer Res, 72(3), 676–​685. Molyneux EM, Rochford R, Griffin B, et al. 2012. Burkitt’s lymphoma. Lancet,
Machalek DA, Poynten M, Jin F, et al. 2012. Anal human papillomavirus infec- 379(9822), 1234–​1244. PMID: 22333947.
tion and associated neoplastic lesions in men who have sex with men: a Moore PS, Kingsley LA, Holmberg SD, et  al. 1996. Kaposi’s sarcoma-​
systematic review and meta-​analysis. Lancet Oncol, 13(5), 487–​500. associated herpesvirus infection prior to onset of Kaposi’s sarcoma.
Maehata Y, Nakamura S, Fujisawa K, et  al. 2012. Long-​ term effect of AIDS, 10(2), 175–​180. PMID: 8838705.
Helicobacter pylori eradication on the development of metachronous gas- Morgan RL, Baack B, Smith BD, et al. 2013. Eradication of hepatitis C virus
tric cancer after endoscopic resection of early gastric cancer. Gastrointest infection and the development of hepatocellular carcinoma:  a meta-​
Endosc, 75(1), 39–​46. analysis of observational studies. Ann Intern Med, 158(5 Pt 1), 329–​337.
Malaty HM, and Graham DY. 1994. Importance of childhood socioeconomic Munoz N, Mendez F, Posso H, et al. 2004. Incidence, duration, and determi-
status on the current prevalence of Helicobacter pylori infection. Gut, nants of cervical human papillomavirus infection in a cohort of Colombian
35(6), 742–​745. women with normal cytological results. J Infect Dis, 190(12), 2077–​2087.
Malope BI, MacPhail P, Mbisa G, et al. 2008. No evidence of sexual transmis- Murphy G, Michel A, Taylor PR, et  al. 2014. Association of seropositiv-
sion of Kaposi’s sarcoma herpes virus in a heterosexual South African ity to Helicobacter species and biliary tract cancer in the ATBC study.
population. AIDS, 22(4), 519–​526. PMID: 18301065. Hepatology, 60(6), 1963–​1971. PMCID: Pmc4216769.
Markowitz LE, Sternberg M, Dunne EF, McQuillan G, and Unger ER. 2009. Murphy G, Pfeiffer R, Camargo MC, and Rabkin CS. 2009. Meta-​analysis
Seroprevalence of human papillomavirus types 6, 11, 16, and 18 in the shows that prevalence of Epstein-​Barr virus-​positive gastric cancer differs
United States: National Health and Nutrition Examination Survey 2003–​ based on sex and anatomic location. Gastroenterology, 137(3), 824–​833.
2004. J Infect Dis, 200(7), 1059–​1067. PMCID: Pmc3513767.
Marshall BJ, and Warren JR. 1984. Unidentified curved bacilli in the stomach of Mutalima N, Molyneux E, Jaffe H, et al. 2008. Associations between Burkitt
patients with gastritis and peptic ulceration. Lancet, 1(8390), 1311–​1315. lymphoma among children in Malawi and infection with HIV, EBV
Martin F, Taylor GP, and Jacobson S. 2014. Inflammatory manifestations of and malaria:  results from a case-​control study. PLoS One, 3(6), e2505.
HTLV-​1 and their therapeutic options. Expert Rev Clin Immunol, 10(11), PMCID: Pmc2423475.
1531–​1546. PMID: 25340428. Ndiaye C, Mena M, Alemany L, et al. 2014. HPV DNA, E6/​E7 mRNA, and
Martin JN, Ganem DE, Osmond DH, et al. 1998. Sexual transmission and the p16INK4a detection in head and neck cancers: a systematic review and
natural history of human herpesvirus 8 infection. N Engl J Med, 338(14), meta-​analysis. Lancet Oncol, 15(12), 1319–​1331.
948–​954. PMID: 9521982. Nelson PK, Mathers BM, Cowie B, et al. 2011. Global epidemiology of hepa-
Marur S, D’Souza G, Westra W, and Forastiere A. 2010. HPV-​associated head titis B and hepatitis C in people who inject drugs: results of systematic
and neck cancer: a virus-​related cancer epidemic. Lancet Oncol, 11(8), reviews. Lancet, 378(9791), 571–​583.
781–​789. Nguyen HQ, Magaret AS, Kitahata MM, et al. 2008. Persistent Kaposi sarcoma
Maucort-​Boulch D, Plummer M, Castle PE, et al. 2010. Predictors of human in the era of highly active antiretroviral therapy: characterizing the predic-
papillomavirus persistence among women with equivocal or mildly tors of clinical response. AIDS, 22(8), 937–​945. PMCID: Pmc2730951.
abnormal cytology. Int J Cancer, 126(3), 684–​691. Oderda G. 1999. Transmission of Helicobacter pylori infection. Can J
McColl KE, Watabe H, and Derakhshan MH. 2008. Role of gastric atrophy Gastroenterol, 13(7), 595–​597.
in mediating negative association between Helicobacter pylori infection Okuda K, Nakashima T, Sakamoto K, et  al. 1982. Hepatocellular carcinoma
and reflux oesophagitis, Barrett’s oesophagus and oesophageal adenocar- arising in noncirrhotic and highly cirrhotic livers:  a comparative study
cinoma. Gut, 57(6), 721–​723. of histopathology and frequency of hepatitis B markers. Cancer, 49(3),
McGowan CE, Edwards TP, Luong MU, and Hayashi PH. 2015. Suboptimal 450–​455.
surveillance for and knowledge of hepatocellular carcinoma among pri- Old LJ, Boyse EA, Oettgen HF, et al. 1966. Precipitating antibody in human
mary care providers. Clin Gastroenterol Hepatol, 13(4), 799–​804. serum to an antigen present in cultured burkitt’s lymphoma cells. Proc
McKhann CF. 1969. Primary malignancy in patients undergoing immuno- Natl Acad Sci U S A, 56(6), 1699–​1704. PMCID: PMC220158.
suppression for renal transplantation. Transplantation, 8(2), 209–​212. Pai SI, and Westra WH. 2009. Molecular pathology of head and neck can-
PMID: 4896335. cer:  implications for diagnosis, prognosis, and treatment. Annu Rev
McQuillan GM, Coleman PJ, Kruszon-​Moran D, et  al. 1999. Prevalence of Pathol, 4, 49–​70.
hepatitis B virus infection in the United States: the National Health and Palmer E, Newcombe RG, Green AC, et al. 2014. Human papillomavirus infec-
Nutrition Examination Surveys, 1976 through 1994. Am J Public Health, tion is rare in nonmalignant tonsil tissue in the UK: implications for tonsil
89(1), 14–​18. cancer precursor lesions. Int J Cancer, 135(10), 2437–​2443.
Megraud F, and Lehours P. 2007. Helicobacter pylori detection and antimicro- Papatheodoridis GV, Lampertico P, Manolakopoulos S, and Lok A. 2010.
bial susceptibility testing. Clin Microbiol Rev, 20(2), 280–​322. Incidence of hepatocellular carcinoma in chronic hepatitis B patients receiv-
Mehanna H, Beech T, Nicholson T, et  al. 2013. Prevalence of human papil- ing nucleos(t)ide therapy: a systematic review. J Hepatol, 53(2), 348–​356.
lomavirus in oropharyngeal and nonoropharyngeal head and neck can- Park JY, Forman D, Greenberg ER, and Herrero R. 2013. Helicobacter pylori
cer:  systematic review and meta-​analysis of trends by time and region. eradication in the prevention of gastric cancer:  are more trials needed?
Head Neck, 35(5), 747–​755. Curr Oncol Rep, 15(6), 517–​525.
Mentzel T, Knuutila S, and Lamovec J. 2013. Kaposi sarcoma. In:  Fletcher Parsonnet J. 2014. Are there benefits of Helicobacter pylori infection?
CDM, Bridge JA, Hogendoorn PCW, and Mertens F (Eds.), WHO clas- In:  Helicobacter pylori eradication as a strategy for preventing gastric
sification of tumours of soft tissue and bone (4th ed., pp. 151–​153). Lyon, cancer (pp. 72–​79). Lyon, France: International Agency for Research on
France: IARC. Cancer (IARC Working Group Reports No. 8).
Mesri EA, Cesarman E, and Boshoff C. 2010. Kaposi’s sarcoma and its associ- Peek RM, Jr., and Blaser MJ. 2002. Helicobacter pylori and gastrointestinal
ated herpesvirus. Nat Rev Cancer, 10(10), 707–​719. tract adenocarcinomas. Nat Rev Cancer, 2(1), 28–​37.
 459

Infectious Agents 459
Peleteiro B, Bastos A, Ferro A, and Lunet N. 2014. Prevalence of Helicobacter Schiffman M, and Wentzensen N. 2013. Human papillomavirus infection
pylori infection worldwide: a systematic review of studies with national and the multistage carcinogenesis of cervical cancer. Cancer Epidemiol
coverage. Dig Dis Sci, 59(8), 1698–​1709. Biomarkers Prev, 22(4), 553–​560.
Peleteiro B, Lunet N, Barros R, La Vecchia C, and Barros H. 2010. Factors Segura-​Lopez FK, Aviles-​Jimenez F, Guitron-​Cantu A, et  al. 2015. Infection
contributing to the underestimation of Helicobacter pylori-​associated gas- with Helicobacter bilis but not Helicobacter hepaticus was associated
tric cancer risk in a high-​prevalence population. Cancer Causes Control, with extrahepatic cholangiocarcinoma. Helicobacter, 20(3), 223–​ 230.
21(8), 1257–​1264. PMID: 25582431.
Penn I, Hammond W, Brettschneider L, and Starzl TE. 1969. Malignant lym- Serraino D, Corona RM, Giuliani M, et  al. 2003. Infection with human her-
phomas in transplantation patients. Transplant Proc, 1(1), 106–​ 112. pesvirus type 8 and Kaposi’s sarcoma in a central Italian area formerly
PMCID: PMC2845467. endemic for malaria. Infection, 31(1), 47–​50. PMID: 12590333.
Persson C, Canedo P, Machado JC, El-​ Omar EM, and Forman D. 2011. Shi J, Zhu L, Liu S, and Xie WF. 2005. A meta-​analysis of case-​control studies
Polymorphisms in inflammatory response genes and their association on the combined effect of hepatitis B and C virus infections in causing
with gastric cancer: a HuGE systematic review and meta-​analyses. Am J hepatocellular carcinoma in China. Br J Cancer, 92(3), 607–​612.
Epidemiol, 173(3), 259–​270. Shi Y, Wu YH, Wu W, et al. 2012. Association between occult hepatitis B infec-
Peto J. 1979. Dose-​response relationships for asbestos-​related disease: impli- tion and the risk of hepatocellular carcinoma: a meta-​analysis. Liver Int,
cations for hygiene standards. Part II. Mortality. Ann N Y Acad Sci, 330, 32(2), 231–​240.
195–​203. Smith BD, Beckett GA, Yartel A, et  al. 2014. Previous exposure to HCV
Peto J, Gilham C, Deacon J, et al. 2004. Cervical HPV infection and neoplasia among persons born during 1945–​ 1965:  prevalence and predictors,
in a large population-​based prospective study: the Manchester cohort. Br United States, 1999–​ 2008. Am J Public Health, 104(3), 474–​ 481.
J Cancer, 91, 942–​953. PMID: 24432883.
Plummer M, de Martel C, Vignat J, Ferlay J, Bray F, and Franceschi S. 2016. Snijders PJ, van den Brule AJ, and Meijer CJ. 2003. The clinical relevance of
Global burden of cancers attributable to infections in 2012: a synthetic human papillomavirus testing: relationship between analytical and clini-
analysis. Lancet Glob Health, 4(9), e609–​e616. cal sensitivity. J Pathol, 201(1), 1–​6.
Plummer M, Franceschi S, Vignat J, Forman D, and de Martel C. 2015. Global Sokal EM, Hoppenbrouwers K, Vandermeulen C, et  al. 2007. Recombinant
burden of gastric cancer attributable to Helicobacter pylori. Int J Cancer, gp350 vaccine for infectious mononucleosis:  a phase 2, randomized,
136(2), 487–​490. double-​blind, placebo-​controlled trial to evaluate the safety, immuno-
Plummer M, Peto J, and Franceschi S. 2012. Time since first sexual intercourse genicity, and efficacy of an Epstein-​Barr virus vaccine in healthy young
and the risk of cervical cancer. Int J Cancer, 130(11), 2638–​2644. adults. J Infect Dis, 196(12), 1749–​1753. PMID: 18190254.
Plummer M, van Doorn LJ, Franceschi S, et  al. 2007. Helicobacter pylori Sonnenberg A. 2013. Review article: historic changes of Helicobacter pylori-​
cytotoxin-​associated genotype and gastric precancerous lesions. J Natl associated diseases. Aliment Pharmacol Ther, 38(4), 329–​342.
Cancer Inst, 99(17), 1328–​1334. Spurgeon ME, Chung SH, and Lambert PF. 2014. Recurrence of cervical can-
Poiesz BJ, Ruscetti FW, Gazdar AF, et al. 1980. Detection and isolation of type cer in mice after selective estrogen receptor modulator therapy. Am J
C retrovirus particles from fresh and cultured lymphocytes of a patient Pathol, 184(2), 530–​540.
with cutaneous T-​cell lymphoma. Proc Natl Acad Sci U S A, 77(12), Steenbergen RD, Snijders PJ, Heideman DA, and Meijer CJ. 2014. Clinical
7415–​7419. PMCID: PMC350514. implications of (epi)genetic changes in HPV-​induced cervical precancer-
Polesel J, Franceschi S, Talamini R, et  al. 2011. Tobacco smoking, alco- ous lesions. Nat Rev Cancer, 14(6), 395–​405.
hol drinking, and the risk of different histological types of nasopha- Stein L, Urban MI, O’Connell D, et al. 2008. The spectrum of human immu-
ryngeal cancer in a low-​risk population. Oral Oncol, 47(6), 541–​545. nodeficiency virus-​associated cancers in a South African black popula-
PMID: 21478046. tion: results from a case-​control study, 1995–​2004. Int J Cancer, 122(10),
Poljak M, Cuzick J, Kocjan BJ, et al. 2012. Nucleic acid tests for the detection 2260–​2265.
of alpha human papillomaviruses. Vaccine, 30 Suppl 5, F100–​F106. Suerbaum S, and Josenhans C. 2007. Helicobacter pylori evolution and phe-
Quint W, Jenkins D, Molijn A, et  al. 2012. One virus, one lesion:  individ- notypic diversification in a changing host. Nat Rev Microbiol, 5(6),
ual components of CIN lesions contain a specific HPV type. J Pathol, 441–​452.
227(1), 62–​71. Tabor E. 1994. Tumor suppressor genes, growth factor genes, and oncogenes in
Raimondi S, Bruno S, Mondelli MU, and Maisonneuve P. 2009. Hepatitis C hepatitis B virus-​associated hepatocellular carcinoma. J Med Virol, 42(4),
virus genotype 1b as a risk factor for hepatocellular carcinoma develop- 357–​365.
ment: a meta-​analysis. J Hepatol, 50(6), 1142–​1154. Tahara T, Yamamoto E, Suzuki H, et al. 2014. Fusobacterium in colonic flora
Rietbergen MM, Braakhuis BJ, Moukhtari N, et  al. 2014. No evidence for and molecular features of colorectal carcinoma. Cancer Res, 74(5), 1311–​
active human papillomavirus (HPV) in fields surrounding HPV-​positive 1318. PMCID: PMC4396185.
oropharyngeal tumors. J Oral Pathol Med, 43(2), 137–​142. Take S, Mizuno M, Ishiki K, et al. 2011. The long-​term risk of gastric cancer
Rinaldi S, Plummer M, Biessy C, et al. 2011. Endogenous sex steroids and risk after the successful eradication of Helicobacter pylori. J Gastroenterol,
of cervical carcinoma:  results from the EPIC study. Cancer Epidemiol 46(3), 318–​324.
Biomarkers Prev, 20(12), 2532–​2540. Tanaka Y, Kurbanov F, Mano S, et  al. 2006. Molecular tracing of the global
Robbins HA, Li Y, Porras C, et al. 2014a. Glutathione S-​transferase L1 mul- hepatitis C virus epidemic predicts regional patterns of hepatocellular car-
tiplex serology as a measure of cumulative infection with human papil- cinoma mortality. Gastroenterology, 130(3), 703–​714.
lomavirus. BMC Infect Dis, 14, 120. Terrault NA. 2002. Sexual activity as a risk factor for hepatitis C. Hepatology,
Robbins HA, Pfeiffer RM, Shiels MS, et al. 2015. Excess cancers among HIV-​ 36(5 Suppl 1), S99–​105.
infected people in the United States. J Natl Cancer Inst, 107(4). The Cancer Genome Atlas Research Network. 2014. Comprehensive molecular
Robbins HA, Shiels MS, Pfeiffer RM, and Engels EA. 2014b. Epidemiologic characterization of gastric adenocarcinoma. Nature, 513(7517), 202–​209.
contributions to recent cancer trends among HIV-​infected people in the PMCID: Pmc4170219.
United States. AIDS, 28(6), 881–​890. Thomas DL, Astemborski J, Rai RM, et al. 2000. The natural history of hepa-
Roberts EA, and Yeung L. 2002. Maternal-​infant transmission of hepatitis C titis C virus infection:  host, viral, and environmental factors. JAMA,
virus infection. Hepatology, 36(5 Suppl 1), S106–​S113. 284(4), 450–​456.
Ronco G, Dillner J, Elfstrom KM, et al. 2014. Efficacy of HPV-​based screening Thorley-​Lawson DA, and Allday MJ. 2008. The curious case of the tumour
for prevention of invasive cervical cancer:  follow-​up of four European virus: 50 years of Burkitt’s lymphoma. Nat Rev Microbiol, 6(12), 913–​
randomised controlled trials. Lancet, 383(9916), 524–​532. 924. PMID: 19008891.
Rositch AF, Koshiol J, Hudgens MG, et al. 2013. Patterns of persistent genital Tomb JF, White O, Kerlavage AR, et al. 1997. The complete genome sequence
human papillomavirus infection among women worldwide:  a literature of the gastric pathogen Helicobacter pylori. Nature, 388(6642), 539–​547.
review and meta-​analysis. Int J Cancer, 133(6), 1271–​1285. Tommasino M. 2014. The human papillomavirus family and its role in carcino-
Rubenstein JH, and Chen JW. 2014. Epidemiology of gastroesophageal reflux genesis. Semin Cancer Biol, 26, 13–​21. PMID: 24316445.
disease. Gastroenterol Clin North Am, 43(1), 1–​14. Trikudanathan G, Philip A, Dasanu CA, and Baker WL. 2011. Association
Safaeian M, Porras C, Schiffman M, et al. 2010. Epidemiological study of anti-​ between Helicobacter pylori infection and pancreatic cancer: a cumula-
HPV16/​18 seropositivity and subsequent risk of HPV16 and -​18 infec- tive meta-​analysis. JOP, 12(1), 26–​31.
tions. J Natl Cancer Inst, 102(21), 1653–​1662. Uemura N, Okamoto S, Yamamoto S, et al. 2001. Helicobacter pylori infection
Saraswat V, Norris S, de Knegt RJ, et  al. 2015. Historical epidemiology of and the development of gastric cancer. N Engl J Med, 345(11), 784–​789.
hepatitis C virus (HCV) in select countries: volume 2. J Viral Hepat, 22 Umar M, Bushra HT, Ahmad M, et al. 2010. Hepatitis C in Pakistan: a review of
(Suppl 1), 6–​25. PMID: 25560839. available data. Hepat Mon, 10(3), 205–​214. PMCID: Pmc3269085.
460

460 PART III:  THE CAUSES OF CANCER


United Nations Development Programme. 2013. Human Development Report Xie T, Cui X, Zheng H, et al. 2013. Meta-​analysis: eradication of Helicobacter
2013. The rise of the South: human progress in a diverse world. New York: pylori infection is associated with the development of endoscopic gas-
United Nations Development Programme. troesophageal reflux disease. Eur J Gastroenterol Hepatol, 25(10),
Vaccarella S, Franceschi S, Snijders PJ, et al. 2010. Concurrent infection with 1195–​1205.
multiple human papillomavirus types: pooled analysis of the IARC HPV Yaghoobi M, Farrokhyar F, Yuan Y, and Hunt RH. 2010. Is there an increased
Prevalence Surveys. Cancer Epidemiol Biomarkers Prev, 19(2), 503–​510. risk of GERD after Helicobacter pylori eradication? a meta-​analysis. Am
Vaccarella S, Lortet-​Tieulent J, Plummer M, Franceschi S, and Bray F. 2013. J Gastroenterol, 105(5), 1007–​1013.
Worldwide trends in cervical cancer incidence:  Impact of screening Yanaoka K, Oka M, Ohata H, et al. 2009. Eradication of Helicobacter pylori
against changes in disease risk factors. Eur J Cancer, 49(15), 3262–​3273. prevents cancer development in subjects with mild gastric atrophy identi-
Verdonck K, Gonzalez E, Van Dooren S et al. 2007. Human T-​lymphotropic fied by serum pepsinogen levels. Int J Cancer, 125(11), 2697–​2703.
virus 1: recent knowledge about an ancient infection. Lancet Infect Dis, Yang HI, Lu SN, Liaw YF, et al. 2002. Hepatitis B e antigen and the risk of
7(4), 266–​281. PMID: 17376384. hepatocellular carcinoma. N Engl J Med, 347(3), 168–​174.
Wabinga HR, Nambooze S, Amulen PM, et al. 2014. Trends in the incidence of Yang HI, Yeh SH, Chen PJ, et al. 2008. Associations between hepatitis B virus
cancer in Kampala, Uganda 1991–​2010. Int J Cancer, 135(2), 432–​439. genotype and mutants and the risk of hepatocellular carcinoma. J Natl
Walboomers JM, Jacobs MV, Manos MM, et  al. 1999. Human papillomavi- Cancer Inst, 100(16), 1134–​1143.
rus is a necessary cause of invasive cervical cancer worldwide. J Pathol, Yang Y, Luo C, Feng R, and Bi S. 2011. The TNF-​alpha, IL-​1B and IL-​10
189(1), 12–​19. polymorphisms and risk for hepatocellular carcinoma:  a meta-​analysis.
Warren JR, and Marshall BJ. 1983. Unidentified curved bacilli on gastric epi- J Cancer Res Clin Oncol, 137(6), 947–​952.
thelium in active chronic gastritis. Lancet, 1(8336), 1273–​1275. Yeh SH, and Chen PJ. 2010. Gender disparity of hepatocellular carcinoma: the
Whitby D, Luppi M, Sabin C, et al. 2000. Detection of antibodies to human roles of sex hormones. Oncology, 78 Suppl 1, 172–​179.
herpesvirus 8 in Italian children: evidence for horizontal transmission. Br Yu MW, Yang YC, Yang SY, et al. 2002. Androgen receptor exon 1 CAG repeat
J Cancer, 82(3), 702–​704. PMCID: Pmc2363308. length and risk of hepatocellular carcinoma in women. Hepatology,
Whitby D, Marshall VA, Bagni RK et  al. 2007. Reactivation of Kaposi’s 36(1), 156–​163.
sarcoma-​ associated herpesvirus by natural products from Kaposi’s Yuan JM, Ross RK, Stanczyk FZ et al. 1995. A cohort study of serum testos-
sarcoma endemic regions. Int J Cancer, 120(2), 321–​ 328. terone and hepatocellular carcinoma in Shanghai, China. Int J Cancer,
PMCID: Pmc2857915. 63(4), 491–​493.
WHO. 2014. World Malaria Report 2014 (p. 228). Geneva, Switzerland: Zeng M, Mao XH, Li JX, et al. 2015. Efficacy, safety, and immunogenicity of
WHO. an oral recombinant Helicobacter pylori vaccine in children in China: a
WHO. 2015. Guidelines for the prevention, care and treatment of persons with randomised, double-​ blind, placebo-​ controlled, phase 3 trial. Lancet,
chronic hepatitis B infection (p. 136). Geneva, Switzerland: WHO. 386(10002), 1457–​1464.
Wong VW, and Janssen HL. 2015. Can we use HCC risk scores to individualize Zhao FH, Lewkowitz AK, Hu SY, et al. 2012. Prevalence of human papilloma-
surveillance in chronic hepatitis B infection? J Hepatol, 63(3), 722–​732. virus and cervical intraepithelial neoplasia in China: a pooled analysis of
PMID: 26026875. 17 population-​based studies. Int J Cancer, 131(12), 2929–​2938.
Wroblewski LE, Peek RM, Jr., and Wilson KT. 2010. Helicobacter pylori and Ziegler J, Newton R, Bourboulia D, et al. 2003. Risk factors for Kaposi’s sar-
gastric cancer:  factors that modulate disease risk. Clin Microbiol Rev, coma: a case-​control study of HIV-​seronegative people in Uganda. Int J
23(4), 713–​739. Cancer, 103(2), 233–​240. PMID: 12455038.
Wu CY, Lin JT, Ho HJ, et  al. 2014. Association of nucleos(t)ide analogue Ziegler JL. 1993. Endemic Kaposi’s sarcoma in Africa and local volcanic soils.
therapy with reduced risk of hepatocellular carcinoma in patients with Lancet, 342(8883), 1348–​1351. PMID: 7901641.
chronic hepatitis B: a nationwide cohort study. Gastroenterology, 147(1), Ziegler JL, Newton R, Katongole-​Mbidde E, et  al. 1997. Risk factors for
143–​151. Kaposi’s sarcoma in HIV-​positive subjects in Uganda. AIDS, 11(13),
Wundisch T, Dieckhoff P, Greene B, et al. 2012. Second cancers and residual 1619–​1626. PMID: 9365767.
disease in patients treated for gastric mucosa-​associated lymphoid tissue Zumkeller N, Brenner H, Zwahlen M, and Rothenbacher D. 2006. Helicobacter
lymphoma by Helicobacter pylori eradication and followed for 10 years. pylori infection and colorectal cancer risk: a meta-​analysis. Helicobacter,
Gastroenterology, 143(4), 936–​942. 11(2), 75–​80.
 461

25 Immunologic Factors

ERIC A. ENGELS AND ALLAN HILDESHEIM

OVERVIEW individual will be exposed, the adaptive immune response has evolved
to allow for real-​time generation of a genetically diverse immune rep-
The immune response is a highly complex system that has developed ertoire through a process of genetic recombination and somatic hyper-
to protect individuals from morbidity and mortality induced by exog- mutation of germline DNA. Cells generated through this process are
enous exposures, including infections. As summarized in this chapter, able to specifically recognize and target infecting pathogen and altered
alterations in the immune response, whether due to immunosuppres- host cells.
sive or immune stimulatory effects, have important consequences
with respect to cancer risk. The demonstration that individuals with
inherited immunological defects, acquired immunological deficien- Cells Types Involved in Immune Response
cies, chronic unresolved infections, and autoimmune conditions are at The two arms of the immune response function through a variety of
considerably increased risk for multiple cancers suggests an important cell types. The primary cells involved in the innate immune response
role for the immune response in the development of cancer at various are natural killer (NK) cells, neutrophils, macrophages, and dendritic
anatomical sites. Studies that have directly evaluated immunogenetic cells. NK cells are lymphocytes involved in recognizing and kill-
and immunological factors and cancer risk are beginning to identify ing infected or otherwise altered cells that have down-​regulated the
specific immunological risk factors associated with individual can- expression of self-​molecules on their surface. Neutrophils and mac-
cers. Furthermore, technological advances have made it increasingly rophages are phagocytes that engulf and eliminate microbes and
feasible to evaluate specific immunological factors and their relation- apoptotic/​necrotic cells. Importantly, macrophages and their derived
ship to cancer risk, suggesting that additional insights are likely in the specialized dendritic cells are involved in cross-​talk between the innate
coming years. and adaptive responses and are responsible for processing and present-
ing foreign antigens to effector cells of the adaptive immune response
OVERVIEW OF THE IMMUNE SYSTEM via cell surface molecules known as HLA.
The primary cells involved in the adaptive immune response are
The immune system is a highly complex network that has evolved to lymphocytes and their derived cells. Various types of lymphocytes
recognize and eliminate pathogenic microorganisms such as viruses exist. Cytotoxic (also called CD8 +) T-​lymphocytes are the primary
and bacteria (non-​self) and altered host cells (altered self), while effector cells responsible for specifically recognizing and killing
simultaneously recognizing self to avoid the destruction of healthy infected or otherwise altered cells that express non-​self antigens on
cells (tolerance) (Paul, 2013). Pathogens are highly variable microor- their surface. Helper (also called CD4 +) T-​cells assist in this process
ganisms that can quickly cause morbidity and mortality in their host. and also contribute to the process of antibody production described
To deal with these aspects of pathogenic infections, the immune sys- next. B-​lymphocytes mature into plasma cells that produce antibodies
tem has evolved to allow for (1)  rapid recognition of and response that specifically recognize and bind foreign pathogens. T-​regulatory
to highly conserved motifs present on the surface of pathogens, (2) (T-​reg) cells are produced to modulate/​downregulate responses so as
de novo generation of responses targeting specific pathogens to which to avoid exacerbated, uncontrolled responses that might be detrimental
it is exposed, and (3) long-​lasting memory to protect against reinfec- to the host.
tion with pathogens encountered in the past.
Compartments of the Immune Response
Innate Versus Adaptive Immune Responses The cells described in the preceding derive from and go through
The immune system can broadly be classified into two arms: innate and the process of differentiation, maturation, selection, expansion, and
adaptive. The innate response is designed to recognize key molecules development of long-​lived memory cells in distinct compartments
present on microbial surfaces (danger signals) that do not otherwise of the immune system. The primary lymphoid organs comprise the
exist on host cells. These responses are non-​specific in that they tar- bone marrow and the thymus. The bone marrow is the source of
get evolutionarily conserved pathogen-​associated molecular patterns self-​renewing hematopoietic stem cells. It is the site where B-​cells
(e.g., lipopolysaccharides in bacteria and double-​stranded ribonucleic undergo their initial differentiation from hematopoietic stem cells
acids in viruses) and do not require prior exposure for recognition to into naïve B-​cells, and ultimately where the mature B-​cells return
occur. As pathogen-​infected and otherwise altered host cells often stop after having encountered antigens in the secondary lymphoid organs
expressing self-​molecules known as human leukocyte antigens (HLA) so that they can serve as a reservoir for fully differentiated plasma
on their surface (as a mechanism for evading immune recognition cells in the periphery. Hematopoietic stem cells also leave the bone
by the adaptive response described in the following), innate immune marrow and seed the thymus, where they undergo differentiation
cells have also evolved to target and eliminate such cells. The innate into naïve T-​cells. It is in the thymus that T-​cells undergo the initial
response is designed to be rapid, to serve as a first line of defense process of selection, thus ensuring that those cells most adept at tar-
to limit infections, and to signal and activate the adaptive immune geting pathogens to which the host has been exposed (high-​affinity
response. cells) are selected for, while low-​affinity cells and cells that recog-
In contrast to the innate response, the adaptive response is generated nize self antigens are selected against. The lymph nodes, spleen, ton-
de novo after initial exposure (called priming) by a specific pathogen. sils, and mucosal associated lymphoid tissues (MALT, including
Since there is no a priori knowledge of the pathogens to which an the Peyer’s patches of the gastrointestinal tract) are considered

461
462

462 PART III:  THE CAUSES OF CANCER


secondary lymphoid organs. At these sites, antigen presenting cells DETERMINANTS OF THE IMMUNE RESPONSE
(APCs) returning from the periphery interact with naïve B-​and
T-​cells, enabling the process of maturation, selection, and expansion Age
of those cells most adept at targeting the pathogen at hand (i.e., high-​ It is known that the immune response varies considerably by age. The
affinity B-​and T-​cells), and the development of long-​lived memory immune response of newborns is distinct from that of adults (Dowling
cells. In addition to the selection and expansion of high-​ affinity and Levy, 2014). Given the limited exposure to antigens in utero,
B-​and T-​cells, the secondary lymphoid tissues also serve as sites where B-​and T-​cells in early life are largely naïve and there is a relative
B-​cells that recognize self are selected against and where peripheral deficit of memory cells. Infants are therefore highly reliant on the
tolerance is induced for self-​antigen targeting T-​cells that might have innate immune response (and passively transferred immunoglobulins
escaped the negative selection process in the thymus. The spleen also from the mother) for protection against pathogen exposure. Innate
serves as the site where damaged and dead cells and pathogens from and adaptive immune response in early life also differs from that of
circulation are cleared. adults with respect to the magnitude and types of responses observed.
Responses tend to be skewed toward a more anti-​inflammatory state
(Th2 response), and the reactivity of immune cells tends to be more
Effectors of the Immune Response modest than those in adults. This attenuated response in early life is
In addition to direct phagocytosis and killing of infected/​altered cells, understandable, as it avoids rejection of maternal and self antigens
cells of both the innate and adaptive immune system regulate and during fetal development/​early life and permits colonization of the
modulate the immune response through the production of soluble fac- skin and gut by commensal micro-​organisms. Nonetheless, these same
tors. These include cytokines (which can further be subdivided into characteristics make young children more susceptible to pathogenic
pro-​inflammatory Th1 cytokines and anti-​inflammatory Th2 cyto- infections and lead to an immune response among infected young chil-
kines) and chemokines that are produced by T-​cells and other immune dren that is less robust, thereby increasing the chances of the infection
and non-​immune cells. These secreted factors act to both attract becoming chronic (hepatitis B virus/​HBV, cytomegalovirus/​CMV) or
immune cells to the site of infection or tissue damage and to regulate progressing to clinical disease (human immunodeficiency virus/​HIV)
their own responses (autocrine response) or that of neighboring (para- (Prendergast et al., 2012).
crine response) or in some instances more distant cells. Plasma cells These differences in response in childhood versus adult life have
derived from B-​cells produce soluble factors called antibodies, which potential implications for cancer risk, since exposures occurring early
are the effectors of the humoral immune response through binding of in life lead to long-​term immunological responses that are distinct from
foreign pathogens to prevent reinfection and to facilitate phagocytosis what is observed when the same exposure first occurs in adulthood.
by APCs such as macrophages. One example of this is the higher predisposition to chronic infection
An important feature of the immune response is the ability of den- and liver disease in newborns infected with HBV (Prendergast et al.,
dritic cells and other APCs to process and present pieces of proteins 2012). Another is the higher risk of Hodgkin’s lymphoma (HL) follow-
(peptides) of an invading pathogen or altered cell to T-​cells so that ing adolescent infection with the Epstein-​Barr virus (EBV) (Hjalgrim
they are activated, and for activated T-​cells to recognize and eliminate et al., 2000). This latter association is thought to be explained by the
infected cells that present peptides from invading pathogens on their strong lymphoproliferative state (often leading to clinical infectious
surface. This process is mediated through Class I and II HLA mole- mononucleosis) induced by first infection with EBV in adolescence,
cules that are specialized in presenting peptides to the immune system once the immune system has matured.
on the surface of cells. Peptides are presented on the surface of APCs In adolescence, the well-​known phenomenon of thymus involution
by both classes of HLA and on the surface of infected non-​immune occurs (Sauce and Appay, 2011). This is the earliest manifestation of
cells by Class  I  HLA molecules. Genes that encode for HLA mol- the process of immunosenescence that occurs with aging. With this
ecules are highly polymorphic, resulting in diversity across individu- involution, production of new populations of naïve T-​cells is drasti-
als in a population with respect to the HLA molecules they produce. cally reduced, and in the ensuing years the pool of naïve T-​cells dimin-
Consequently, this results in varying efficiencies with which different ishes and is increasingly replaced by memory T-​cells. The smaller pool
individuals are able to present and respond to individual pathogens or of naïve cells leads to a diminished ability to adequately respond to
altered cells. This has implications for cancer risk, as will be discussed new pathogens at older ages.
later in this chapter. Additional changes in both the innate and adaptive immune response
occur later in life, including reduced NK cell cytotoxicity, reduced
number and function of APCs, and decreased naïve and memory
ABNORMALITIES OF THE IMMUNE RESPONSE B-​cells (Fulop and Montgomery, 2014; Kogut et al., 2012; Nikolich-​
Zugich, 2014; Shaw et al., 2013). Decreases in the class switch recom-
As will be described further in this chapter, alterations in this complex bination process of B-​cells that generates protective antibodies and
system disturb the careful balance of the immune response and can memory B-​cells is also observed. At the same time, the redirection of
lead to disease states. On the one hand, immunosuppression, whether responses from adaptive to innate immune responses leads to a dys-
inherited or acquired, can lead to increased susceptibility to infections regulated state characterized by increased levels of inflammatory mol-
that can cause cancer and reduced ability to target transformed cancer ecules. These changes increase susceptibility to new infections, reduce
cells for destruction. On the other, states that lead to chronic immune the ability to control existing chronic infections, and result in reduced
stimulation, such as autoimmunity due to an inability to distinguish responses to vaccination. These changes have also been hypothesized
self from non-​self, or hyper-​reactivity to external stimulants (as in to affect cancer risk through an inability to efficiently detect and target
allergic states), typically result in chronic inflammation and may result transformed cells (decreased immunosurveillance).
in increased risk of cancer development. More subtly, among individ- Like those seen in early life, characteristics of the immune response
uals with an otherwise healthy immune system, naturally occurring in late adult life likely have important implications for cancer develop-
variability in one’s ability to recognize and eliminate specific patho- ment. However, it has been difficult to tease apart effects on cancer risk
gens might predispose to specific cancers caused by infectious agents. of immune aging from those of other age-​related phenomenon.
In the remainder of this chapter, we summarize the primary deter-
minants of the immune response that should be considered when eval-
uating the link between immunity and cancer, discuss inherited and Sex
acquired immune variation associated with cancer development, and
briefly touch on the role of the immune response in determining prog- The effect of sex on immune response is more modest and less well
nosis after a cancer diagnosis and the promise of new immune-​based understood. The female excess for the majority of autoimmune condi-
therapies for some cancers. tions is suggestive of sex differences in the underlying immunological
 463

Immunologic Factors 463


conditions that predispose to these diseases (Whitacre, 2001). Also, intensity and destructiveness of disease activity, and in transplanta-
in many instances women have been found to develop a more robust tion, where they are used to prevent or treat graft rejection (in solid
immune response following vaccination compared to men, again organ transplantation) or graft-​versus-​host disease (in hematopoietic
suggesting sex differences in immune response (Klein et  al., 2010). stem cell transplantation). The adaptive immune system, specifically
These differences are supported by hormonal differences across the T-​cell immunity, is the primary target of these therapies, and while
sexes. In fact, hormonal receptors have been shown to be expressed the clinical intent is to achieve a narrow effect (e.g., abrogation of a
on immune cell subsets of the innate and adaptive immune response specific autoimmune disease process), the effects are generally broad
(Gabriel and Arck, 2014)  and estrogens, progesterone, and tes- and non-​specific. Immunosuppressive medications are often admin-
tosterone have distinct effects on cells expressing their receptors istered in combinations for prolonged periods (typically for months
(Nalbandian and Kovats, 2005; Tan et  al., 2015; Trigunaite et  al., or years), although the intensity of immunosuppression typically var-
2015). This may explain, in part, why females typically have elevated ies over time in concert with dosing to achieve the desired clinical
levels of T-​helper cells, and higher plasma levels of various cytokines effects.
and immunoglobulins compared to men. It is interesting to note that Several classes of medications used in this setting have broad
men with Klinefelter’s syndrome, a condition in which men are born effects on adaptive immunity, especially when used in combination,
with an extra X chromosome, have elevated serum levels of immu- including calcineurin inhibitors (e.g., cyclosporine and tacrolimus),
noglobulins and various cytokines, and androgen treatment results in antimetabolites (e.g., azathioprine, methotrexate, and mycophenolate
reductions in levels of these same markers, suggesting that the lack mofetil), and inhibitors of the mammalian target of rapamycin (mTOR
of testosterone in patients with Klinefelter’s syndrome enhances cel- inhibitors, e.g., sirolimus) (Halloran, 2004). Intravenous administra-
lular and humoral immunity and that androgen replacement treatment tion of antibodies against T-​cells or B-​cells (e.g., rituximab targeted
may reverse this effect (Kocar et  al., 2000). Finally, immunological at B-​cells) induces pronounced short-​term deficits in these arms of the
alterations observed in pregnancy, during the course of the menstrual immune system.
cycle, and among women using exogenous hormones further support Additional classes of medications have varying effects on the
the role of hormonal profile as a mediator of the immune response immune system. Fludarabine, used to treat chronic lymphocytic leu-
(Klein et al., 2010). kemia and other low-​grade lymphomas, causes a marked depletion of
T-​cells (Cheson, 1995). Antibodies against tumor necrosis factor alpha
(e.g., infliximab and etanercept) are used in the treatment of several
Host Genetics autoimmune diseases, including rheumatoid arthritis and Crohn’s dis-
Another intrinsic factor that influences the immune response is ease. Corticosteroids are widely used in the short-​term treatment of
genetics. The link between genetic mutations and primary immuno- common ailments such as asthma, or in combination with other medi-
deficiency disorders is proof-​of-​principle of the important role that cations for long-​term immunosuppression in transplantation or treat-
genetics plays in determining the immune response (see further dis- ment of autoimmune diseases.
cussion later in this chapter). In contrast to the single-​gene, high-​pen- Many of these immunosuppressive medications have somewhat
etrance model observed for these rare conditions, the effect of genetic non-​specific and overlapping effects on the adaptive immune system,
factors on immune response more broadly is understood to be com- and because they are often administered in combination, it is diffi-
plex and polygenic, making them more difficult to study. Nonetheless, cult to determine whether they differ with respect to their effects on
progress has been made in recent years in elucidating the underlying cancer risk. Also, it can be challenging to separate the effects of the
genetic underpinnings of the immune response. For example, stud- medications from the underlying disease processes for which they are
ies that have evaluated common genetic polymorphisms and autoim- the treatment. Indeed, cancer risk may be increased due to the effects
mune disease risk, particularly in the genome-​wide association study of chronic inflammation and immune activation seen in autoimmune
(GWAS) era, have uncovered various gene polymorphisms associated diseases, graft rejection, or graft-​versus-​host-​disease.
with modest risk for specific autoimmune conditions (Hu and Daly, In addition, it is apparent that several immunosuppressive medi-
2012). In some instances, initial findings from GWAS have led to fol- cations have indirect effects other than immunosuppression that
low-​up efforts that have identified functional variants associated with modulate cancer risk. For instance, calcineurin inhibitors and anti-
specific autoimmune conditions and/​or patterns in the genetic associa- metabolites cause DNA damage or impair DNA repair (IARC,
tions seen across autoimmune diseases suggestive of common or dis- 2012a, 2012b). Also, calcineurin inhibitors upregulate expression of
tinct biological causal pathways. GWAS conducted within carefully transforming growth factor beta-​1 (TGF-​beta1) and vascular endo-
controlled vaccination studies have provided another fruitful approach thelial growth factor (VEGF), which may promote the development
to identify genetic determinants of immune response (Kennedy et al., of cancer (Basu et al., 2008; Hojo et al., 1999). In contrast, mTOR
2012, 2014). While more difficult to study due to the absence of an inibitors may be relatively protective, by blocking the mTOR path-
outcome with a strong immunological phenotype (as seen for auto- way (a central pathway in cellular metabolism and proliferation)
immune conditions) and the lack of tight experimental control (as and downregulating the VEGF pathway (Gentzler et al., 2012; Guba
seen in vaccination studies), evaluation of healthy individuals have in et al., 2002).
some instances pointed to specific genes associated with variability
in the levels of specific immunological factors, including antibodies
and cytokines (Dehghan et al., 2011; Liao et al., 2012; Rubicz et al., HIV Infection and AIDS
2013). Taken together, studies to date confirm the role of genetics as a Human immunodeficiency virus type 1 (HIV) is a retrovirus that
determinant of immune response in health and disease states and not causes lifelong chronic infection. HIV is acquired through exposures
unexpectedly suggest that effects are highly polygenic. Despite this to blood or other bodily fluids, and most people become infected
progress, the fact that the effect of single genes on immune response is through sexual contact or the use of injection drugs. HIV infection
typically small and the association between individual immune param- typically results in progressive immunosuppression, which, when
eters and cancer are likely modest poses a challenge for studies aimed it becomes severe enough, is classified as acquired immunodefi-
to uncover the effect of genes that regulate the immune response on ciency syndrome (AIDS). HIV infects and destroys CD4-​positive
risk of cancer. T-​cells, and the major manifestation of AIDS is a profound deficit
in T-​cell immunity (Castro et al., 1992). The concentration of CD4-​
positive T-​cells in peripheral blood (i.e., CD4 count) is a commonly
Medications That Cause Immunosuppression used clinical measure of immune function in HIV-​infected people.
Immunosuppressive medications are utilized in the therapy of auto- Chronic polyclonal B-​cell activation is also present, along with defi-
immune or allergic diseases, where they are used to decrease the cits in adaptive B-​cell function (Moir and Fauci, 2009). Some of the
46

464 PART III:  THE CAUSES OF CANCER


immune abnormalities may result from chronic antigenic stimulation IMMUNE VARIATION AND CANCER
in response to the virus, or from increased permeability of the gas-
trointestinal tract and bacterial translocation (Brenchley et al., 2006). Inherited Conditions
Since 1996, combinations of medications targeted at HIV (i.e., highly Inherited Immunodeficiency Disorders and Cancer
active antiretroviral therapy [HAART]) have increasingly allowed
for the suppression of HIV replication and substantial reconstitution Primary immunodeficiency disorders (PID) comprise a diverse set of
of the immune system (Althoff et al., 2012; Palella et al., 1998). In conditions characterized by inherited defects that affect the immune
many cases, however, immune reconstitution is incomplete, and some system. Over 200 forms of PID have been identified to date (Al-​Herz
degree of chronic immune deficit and inflammation appears to persist et al., 2014; Ochs and Smith, 2014). Most PIDs are exceedingly rare.
(Lederman et al., 2013). Approximately half have been reported in ≤ 10 unrelated individu-
als, making their study and characterization of cancer risk difficult.
Causal attribution, when trying to understand increased cancer risk
Autoimmune and Allergic Conditions observed for these diseases, is also complicated by the fact that the
underlying molecular defects involved in PID affect critical cellular
Autoimmune diseases are characterized by a chronic immuno- processes that have pleiotropic effects, including immune deficien-
logic response directed at self antigens, leading to progressive tis- cies, autoimmunity, inflammation, DNA repair, and chromosomal
sue destruction (Mackay and Rose, 2006). Many such diseases are stability. Nonetheless, these single-​ gene diseases provide impor-
described, each defined by a constellation of specific clinical findings tant clues to the biological mechanisms linking immune response
and immunological markers. T-​cell infiltrates are commonly present in to cancer.
the targeted tissues, and polyclonal B-​cell activation may also occur. PIDs are broadly categorized based on the component of the
Patients with these diseases are treated chronically with a variety of immune system that is primarily involved (Al-​Herz et  al., 2014).
immunosuppressive medications, which vary according to the condi- Specifically, PIDs are currently classified based on the following
tion, although the targeted level of immunosuppression is usually less groupings: (1) combined immunodeficiency disorders (CIDs) (with or
intense than in transplantation. without syndromic features) in which both B-​and T-​cell functions are
Common allergic conditions include asthma, hay fever, and eczema, altered, (2) diseases with predominant antibody deficiencies, (3) dis-
and they are sometimes grouped together as manifestations of “atopy” eases of immune dysregulation, (4) defects of phagocyte number and/​
(Nelson and Blauvelt, 2012). These conditions are characterized by or function, (5)  defects in innate immunity, (6)  autoinflammatory
hypersensitivity to environmental exposures (i.e., allergens) involv- disorders, and (7)  complement deficiencies. It should be noted that
ing the production of IgE, degranulation of mast cells, and elevated because of the complex nature of these conditions, some PIDs fit the
levels of eosinophils in affected tissues and the circulation. Onset is criteria for more than one category.
frequently during childhood. The incidence of allergic conditions is Approximately two dozen PIDs have an elevated risk of malignancy
rising over time, perhaps due to improved hygiene and reduced expo- as one of their associated features (Al-​Herz et al., 2014), and 14–​16
sure to microbes in early life (Wills-​Karp et al., 2001). Immune-​modu- of these PIDs (depending on how conditions are grouped) have been
lating medications used to treat these conditions include systemic and reported in > 10 unrelated individuals (Table 25–1). Because immu-
inhaled corticosteroids (for asthma and hay fever), topical corticoste- nological deficiencies characterized by these PIDs invariably lead
roids (eczema), and antihistamines (hay fever). Because of the low to increased susceptibility to infections, much of the cancer excess
doses, short treatment courses, or localized administration of these observed occurs among infection-​associated cancers, primarily EBV-​
medications, patients with allergic conditions can be considered to be associated lymphomas. Furthermore, for several PIDs associated with
only minimally immunocompromised. cancer development (e.g., ataxia telangiectasia, Nijmegen breakage

Table 25–1.  Primary Immunodeficiency Diseases Associated with Cancer

International Union of Immunological


Condition Societies Classification* Associated Cancers

Ataxia telangiectasia (AT) Combined Immunodeficiency Disorders with Lymphoma, leukemia, breast, stomach,
Syndromic Features liver, thyroid
Wiskott-​Aldrich syndrome (WAS) Combined Immunodeficiency Disorders with Lymphoma
Syndromic Features
Nijmegen breakage syndrome (NBS) Combined Immunodeficiency Disorders with Lymphoma
Syndromic Features
Bloom syndrome (BS) Combined Immunodeficiency Disorders with Lymphoma, breast, colon, skin
Syndromic Features
Cartilage hair hypoplasia Combined Immunodeficiency Disorders with Lymphoma
Syndromic Features
Immunodeficiency with centromeric instability and Combined Immunodeficiency Disorders with Lymphoma
facial anomalies (ICF) Syndromic Features
X-​linked lymphoproliferative disease (XLP) Combined Immunodeficiency Disorders Lymphoma
Class-​switch recombination (CSR)/​CD40 ligand Combined Immunodeficiency Disorders Lymphoma, biliary tract, liver, pancreas
deficiency
Hyper-​IgE syndrome (HIES)/​DOCK8 deficiency Combined Immunodeficiency Disorders Lymphoma, HPV-​associated cancers
MST1 deficiency Combined Immunodeficiency Disorders Lymphoma
Common variable immunodeficiency disorders (CVID) Predominantly Antibody Deficiencies Lymphoma, breast, melanoma, stomach
Autoimmune lymphoproliferative sydrome (ALPS) Diseases of Immune Dysregulation Lymphoma
EVER1/​2 deficiencies Defects of Innate Immunity Non-​melanoma skin
Severe congenital neutropenia (SCN)—​ELA2/​HAX1 Congenital Defects of Phagocyte Number Leukemia
deficiencies and/​or Function

* Conditions can be classified into > 1 IUIS classification group. Herein each condition is classified into a single group hierarchically based on the order in which they are listed in
the table.
 465

Immunologic Factors 465


syndrome, and Bloom syndrome), the underlying genetic defect leads childhood. NK cell function is also altered in WAS. Autoimmune dis-
to immunological deficiency via DNA repair defects that affect the eases are commonly observed in WAS patients. WAS is a rare condi-
normal process of recombination and hypermutation observed in B tion diagnosed in 1 to 10 per million men. Life expectancy is typically
and T cells as part of their mechanism to induce immunological diver- 15–​20 years (Shin et al., 2012).
sity. The observed increase in cancers for these conditions might there- A predisposition to malignancies is seen in WAS, particularly lym-
fore also be related to these defects in DNA repair mechanism. phomas. In one series of 154 patients, cancer was observed in 13%
While EBV-​associated lymphomas of B-​cell origin (primarily NHL, of cases (average age: 9.5 years), over 70% of which were lympho-
but also HL) account for the majority of cancer excess observed in mas (Sullivan et al., 1994). In a more recent series of 50 patients, can-
PID, excesses in T-​ cell lymphomas (for Nijmegen breakage syn- cers were observed in a comparable proportion of cases (10%), were
drome), non-​EBV lymphomas (for X-​linked lymphoproliferative dis- restricted to lymphomas, and occurred preferentially among patients
ease), and leukemias (for ataxia telangiectasia, Nijmegen breakage with no WAS protein expression (Imai et al., 2004).
syndrome, and severe congenital neutropenia) have been reported. In In addition to AT, other PIDs caused by mutations in genes that lead
addition, solid tumors have been noted in excess of expectation for to chromosomal instability have been reported to be associated with
several PIDs. Examples include breast cancer in ataxia telangiectasia excess cancer risk. These include most notably NBS (an autosomal
and common variable immunodeficiency; gastric cancer in common recessive condition caused by NBS1 gene mutations) and Bloom syn-
variable immunodeficiency; liver, pancreas, and gallbladder cancers drome (an autosomal recessive condition caused by BLS gene muta-
in CD40 ligand deficiencies; HPV-​associated non-​melanoma skin can- tions), although other rarer conditions have also been suggested to be
cers in epidermodysplasia verucciformis; and various solid tumors in associated with increased risk of malignancies (Wegner et al., 2014). It
Bloom syndrome. has been reported that over 50% of individuals with NBS and Bloom
To follow, we describe individual PIDs that are reported to be asso- syndrome develop cancer, typically before age 20–​25. Greater than
ciated with increased risk of cancer, and summarize the breadth and 90% of malignancies seen in NBS are lymphomas of B-​and T-​cell
magnitude of the associations observed. It should be stressed that, in types (Gladkowska-​ Dura et  al., 2008). It has been estimated that
many instances, the absolute and relative risk estimates presented are NBS patients have a 50-​fold increased risk of early onset cancer and
based on modest size studies, given the rarity of the conditions being > 1000-​fold increased risk of lymphomas (Wegner et al., 2014). Bloom
considered, and that these estimates are therefore imprecise. syndrome patients have also been shown to have an excess risk of lym-
phomas, but in contrast to NBS, a large fraction of cancer diagnosed
Combined Immunodeficiencies Associated with in Bloom syndrome (particularly those diagnosed in adulthood) are
Syndromic Features. Ataxia telangiectasia (AT) is an autoso- carcinomas. Among carcinomas, colon, skin, and breast cancers pre-
mal recessive multisystem disease associated with progressive neu- dominate (German, 1997).
rodegeneration (ataxia), oculocutaneous telangiectasia, and variable Other immunodeficiencies with syndromic features that are associ-
degrees of immune deficiency. AT is caused by mutations in the ATM ated with an excess risk of cancer include cartilage hair hypoplasia
gene, which is known to be involved in numerous cellular functions, (an autosomal recessive condition caused by mutations in the DRMRP
including DNA repair. AT patients are predisposed to infections, par- gene), a disease reported to be associated with a 7-​fold increased risk
ticularly upper and lower respiratory infections. Immunodeficiency of cancer overall and a 90-​fold increased risk of NHL (Makitie, 2014);
observed in AT patients is highly variable, but involves both antibody and immunodeficiency with centromeric instability and facial anoma-
production (hypogammaglobulinemia) believed to be due to a defect lies (ICF; an autosomal recessive condition caused by mutations in
in B-​cell differentiation and cellular immunity resultant from faulty DNMT3B-​ICF1 or ZBTB24-​ICF2 genes) (Hansen et al., 2014).
development of the thymus. AT is estimated to occur at a frequency
ranging from 1:40,000 to 1:80,000 live births and occurs with approx- Other Combined Immunodeficiencies. X-​linked lym-
imately equal frequency in males and females (Yel et al., 2014). AT phoproliferative disease (XLP) is a condition that is characterized by
patients typically have a life span of about two decades. an exquisite susceptibility to EBV infection. Approximately 80% of
Predisposition to malignancies is a hallmark of AT. Approximately cases are attributable to mutations in the gene SH2D1A, whose pro-
a quarter of AT patients develop malignancies, typically in the first tein affects multiple intracellular signaling pathways that are involved
decade of life. While lymphomas predominate, solid tumors are also in the interaction of various immune effector cells after activation,
observed, and an excess of breast cancer among women has been including T-​cells and NK cells. XLP caused by this gene are referred
reported. In a study of 279 patients with AT from a French national reg- to as XLP-​1 to distinguish it from other XLP that are not caused by
istry (Suarez et al., 2015), 69 (24.7%) developed cancer. Cumulative SH2D1A. XLP-​1 is estimated to occur with an incidence of 1–​3 per
incidence of cancer was 10.4% at 10 years and increased to 38% at million males; prognosis is poor, with approximately 70% of cases
40 years. The majority of cancers were lymphomas, accounting for dying before the age of 10 years, many from fulminant EBV infection
55% of tumors. These non-​Hodgkin’s lymphomas (NHL) were pre- (Schuster and Latour, 2014).
dominantly of B-​cell origin (26 of 38), half of which were EBV-​asso- Approximately 30% of affected males develop lymphomas, with
ciated. At ages 0–​14, the observed incidence of NHL, HL, and acute an average age at diagnosis of 4–​6 years (Schuster and Kreth, 2000;
leukemia (AL) was 5667, 540, and 49 times higher than expectation, Schuster and Latour, 2014). The vast majority (90%) of lymphomas
respectively. Solid tumors were observed among nine AT patients, and are of B-​cell origin, and most occur at extranodal sites. The risk of
included breast, gastric, liver, and thyroid cancers, as well as a glioma lymphoma in XLP has been estimated to be about 200 times higher
diagnosed after treatment for HL. An excess of breast cancer mortal- than general population rates. Interestingly, the likelihood of develop-
ity was also noted in a separate study (Reiman et al., 2011), where a ing lymphoma in XLP is similar in EBV-​infected and EBV-​uninfected
standardized mortality ratio of 31 was reported among 141 women individuals, suggesting that lymphoma risk in XLP is not EBV specific
with AT. (Sumegi et al., 2000).
Wiskott-​Aldrich syndrome (WAS) is an X-​linked condition char- Class-​switch recombination (CSR) deficiencies (previously called
acterized by low platelet count (thrombocytopenia), reduced platelet X-​linked hyper-​IgM syndrome) include CD40 ligand deficiencies that
size, bloody diarrhea, and eczema. WAS is caused by mutations in the are X-​linked conditions characterized by low levels of serum IgG,
WAS gene, which is known to play an important role in cytoskeletal IgA, and IgE and normal or elevated levels of IgM (Notarangelo et al.,
remodeling following T-​cell receptor (TCR) binding and immune syn- 2006; Notarangelo et al., 2014). Patients with this condition typically
apse formation. WAS patients are predisposed to infections, including present with recurrent upper and lower respiratory tract infection.
respiratory and ear infections. Immunodeficiency observed in WAS Neutropenia is a common finding, contributing to the predisposition
patients involves both B-​cells and T-​cells, but varies, typically depend- to bacterial infections. As noted by its name, this condition is caused
ing on the WAS mutation observed and its effect on protein expression. in CD40 ligand mutations that impair the ability of B-​cells to undergo
While B-​and T-​cell circulating levels in infancy might be normal, their T-​cell-​mediated class switching from IgM-​producing cells to those
function is altered and levels tend to decline with increasing age during that produce other immunoglobulin classes. This condition is rare,
46

466 PART III:  THE CAUSES OF CANCER


usually detected in infancy, and estimated to occur in about 1:500,000 Early reports suggested that a third of cases developed squamous
male births per year (Winkelstein et  al., 2003). Affected individuals cell carcinomas of the skin over a period of decades, particularly in
typically survive into their forties. An interesting feature of this condi- sun-​exposed areas, but more recent studies with long follow-​up time
tion, in addition to its predisposition of lymphomas (Filipovich et al., suggest that virtually all cases are prone to the development of these
1994), is the reported elevation in incidence of liver, pancreas, and cancers (Majewski et al., 1997).
biliary tract cancers (Hayward et al., 1997). Severe congenital neutropenia (SCN) is a condition (usually
Other combined immunodeficiency conditions that are associated detected in infancy) characterized by maturation arrest of myelopoi-
with an excess risk of cancer include hyper-​IgE syndrome (HIES; an esis, resulting in reduced neutrophil counts, predisposition to recurrent
autosomal recessive condition caused by mutations in the DOCK8 bacterial and fungal infections, and decreased mineral bone density.
gene), a disease reported to cause cancer in close to 20% of affected The disease can be inherited in either an autosomal dominant or
individuals (half of whom develop hematological malignancies) recessive pattern, depending on the specific gene mutation involved.
(Aydin et  al., 2015); and MST1 (caused by mutations in the SKT4 Mutations in ELA2 and HAX1, both involved in the process of apopto-
gene), a disease thought to involve increased lymphoma risk but for sis, have been shown to be causal for the dominant and recessive forms
whom too few cases have been evaluated to permit definitive state- of this disease, respectively. Follow-​up of 374 cases with SCN treated
ments regarding cancer risk (Al-​Herz et al., 2014). with granulocyte colony stimulating factor (G-​CSF) therapy from an
international registry demonstrated a cumulative risk of myelodys-
Other Primary Immunodeficiency Disorders (PIDS).  plastic syndrome (MDS) or acute myeloid leukemia (AML) of 36%
Combined variable immunodeficiency (CVID) is a heterogeneous by 12 years after initiation of treatment (which occurred on average
condition that is characterized by marked reductions in serum immu- at age 3 years) (Rosenberg et  al., 2006). Of note, dose of G-​CSF
noglobulin levels (hypogammaglobulinemia), impaired antibody treatment was positively associated with risk of MDS/​AML, with a
responses, and recurrent bacterial infections (Hammarstrom, 2014). 1.2-​fold increase in risk per doubling of treatment dose. This suggests
A substantial proportion of cases develop inflammatory and autoim- the possible existence of a group of individuals at risk of cancer that
mune conditions. While abnormalities in B-​cell responses are the can be defined by non-​responsiveness to standard therapy.
primary immunological features of CVID, evidence has accumu-
lated to suggest the presence of T-​cell abnormalities as well (Ramesh Common Genetic Polymorphisms
et  al., 2015). The underlying molecular basis for CVID is unknown In addition to the rare and highly penetrant mutations that give rise to
for the vast majority of cases, and most cases are sporadic in nature. PIDs, more common polymorphisms in immune response genes with
Nonetheless, some specific autosomal recessive gene mutations have low penetrance are likely to contribute to cancer risk. As mentioned
been linked to CVID (including ICOS, CD19, CD20, CD21, CD81, earlier, these effects are likely to be polygenic and of modest effect
LRBA, BAFF receptor, CXCR4, NFKB2, and PIK3CD), and a recent size, making them more difficult to study than the link between single
GWAS revealed a potentially important role for copy number varia- gene mutations involved in PIDs and cancer. Despite these theoretical
tions (CNVs) in this condition (Orange et al., 2011). It is estimated that difficulties, past candidate-​based studies have shown consistent evi-
between 1:25,000 and 1:50,000 individuals develop CVID, making it dence for association between some immune response genes and vari-
one of the more common PIDs (Resnick et al., 2012). CVID occurs ous cancers, particularly cancers caused by viral infections and those
with approximately equal frequency in males and females. Diagnosis of hematopoietic origin.
occurs, on average, around age 25–​30 years; about 25%–​30% are diag- In particular, consistent associations with cancer have been
nosed before the age of 20 years. reported for HLA genes. These associations are biologically plausible
Registry-​based studies of several hundred cases each have reported since, as described earlier in this chapter, these are genes involved in
cancer in up to 15% of cases in the most recent report (Resnick et al., the presentation of foreign antigens to the adaptive arm of the immune
2012). A  large fraction of cancers are lymphomas, primarily B-​cell response and in innate responses mediated through HLA binding.
NHL. It has been estimated that individuals with CVID have a 6-​to Studies have shown specific associations between HLA alleles and
18-​fold increase in risk of NHL compared to the general population haplotypes for several cancers, including cervical cancer (caused by
(Mellemkjaer et al., 2002; Quinti et al., 2007). Elevations in other can- HPV) (Chen and Gyllensten, 2015; Hildesheim and Wang, 2002),
cers have also been reported, including gastric, breast, and melanoma nasopharyngeal carcinoma (caused by EBV) (Su et  al., 2013; Tang
(Resnick et  al., 2012). With respect to gastric cancer, earlier reports et al., 2012), hepatocellular carcinoma (frequently caused by HBV or
suggested a 50-​fold increase in risk, but the observed increase in risk HCV) (Jiang et al., 2013; Kumar et al., 2011; Li et al., 2012; Lin et al.,
is more modest in the latest report, suggesting changes with increased 2010; Lopez-​Vazquez et  al., 2004; Xin et  al., 2011), HL (approxi-
follow-​up or in more recent years (Mellemkjaer et  al., 2002; Quinti mately one-​third EBV-​associated) (Cozen et al., 2014; Hjalgrim et al.,
et al., 2007; Resnick et al., 2012). 2010; Huang et al., 2012; Urayama et al., 2012), and NHL (Skibola
Autoimmune lymphoproliferative syndrome (ALPS) is a condition et  al., 2012; Smedby et  al., 2011; Vijai et  al., 2015; Wang et  al.,
characterized by chronic lymphadenopathy and splenomegaly of early 2010a) among others. Findings from candidate gene studies of HLA
onset, autoimmune phenomenon, and expanded populations of T-​cells genes are now beginning to be confirmed by large, agnostic GWAS,
expressing specific T-​cell receptors α/​β and CD4/​CD8 double-​negative where polymorphisms in the MHC region of chromosome 6 (where
T-​cells (Fleisher et al., 2014). ALPS is typically inherited in an auto- HLA resides) have often provided the strongest evidence for asso-
somal dominant fashion, although autosomal recessive and recessive ciation with individual cancers. Furthermore, when HLA alleles are
forms of the disease have been reported less frequently. The majority inferred or directly measured in these GWAS, the SNP associations
of the conditions diagnosed to date are caused by mutations in the FAS are often in strong LD with known HLA alleles previously reported
gene (ALPS-​FAS). As a result, cases have an inability to respond to to be associated with the respective cancer being investigated. As
cell death signals. ALPS occurs in both males and females. The condi- expected, the specific HLA genes and alleles associated with different
tion is rare and no incidence figures are available. It has been estimated cancers vary, since presentation of peptides to the immune system
that risk of NHL is elevated 14-​fold in ALPS-​FAS and that of HL 51-​ by HLA molecules is antigen-​specific. Figure 25–1 summarizes key
fold compared to general population rates of similar age (Straus et al., findings from GWAS for hematopoietic and/​or infection-​associated
2001). cancers with strong evidence for associations within the MHC region
Epidermodysplasia verruciformis is a very rare autosomal reces- (Su et al., 2013).
sive condition, usually detected in early life, that is characterized by While the specific HLA alleles associated with different cancers
an exquisite and specific predisposition to cutaneous HPV infections. vary, some patterns are beginning to emerge that provide useful clues
This condition is caused by underlying mutations in EVER1/​2 genes to the underlying mechanisms of observed associations. First, HLA
(also known as TMC6/​8), which are involved in maintenance of zinc associations have been most consistent for cancers of hematopoietic
balance within epithelial and T-​cells, which when altered predispose origin and/​or those caused by viral infections. Of note, associations
to uncontrolled cutaneous HPV infections (Lazarczyk et  al., 2009). with HLA have not been noted for gastric cancer, a cancer linked to
 467

Immunologic Factors 467


70
Non-HLA HLA Class I Non-HLA Class I/II HLA Class II Non-HLA

Nasopharyngeal carcinoma
EBV-positive Hodgkin’s lymphoma
60
Hodgkin’s lymphoma
EBV-negative Hodgkin’s lymphoma
Nodular sclerosis Hodgkin’s lymphoma
Follicular lymphoma
Chronic lymphocytic leukemia
50 Lymphoma
Hepatocellular carcinoma
HCV-related hepatocellular carcinoma
HBV-related hepatocellular carcinoma
Cervical cancer
–Log10 (P value)

40 Lung cancer
Lung adenocarcinoma
Testicular germ cell tumor
Prostate cancer
Multiple cancers

30

20

10

0
29M 30M 31M 32M 33M 34M
Position in MHC Region (MB)
Figure 25–1.  Plot of strongest GWAS associations within the major histocompatibility complex (MHC) across anatomic sites. Source: Adapted from Su
et al., Front Oncol (2013).

H. pylori infection. This makes sense since H. pylori is an extracel- EBV-​associated cancers, while HLA Class II genes are most impor-
lular bacterial pathogen and the immune response to such pathogens tant for other viral-​associated cancers. Thus, the HLA Class I asso-
is driven by B-​rather than T-​cell responses, where HLA presenta- ciations are stronger than Class  II associations for EBV(+) HL
tion is not required. Second, while the strong linkage disequilibrium and for NPC, while HLA Class  II associations are stronger than
patterns in the MHC region make it difficult to disentangle specific Class  I  associations for cervical cancers and HCC (whether HBV
HLA alleles/​haplotypes that drive observed association, there is some or HCV related). It remains to be seen whether the close to 10%
indication that the strongest effects observed for lymphomas are with of gastric cancers associated with EBV infection have HLA associa-
HLA Class II rather than Class I genes. Again, this makes sense since tions and whether they are stronger for HLA Class I or II genes. The
Class II HLA molecules are only expressed in cells from the immune Class I predominance for EBV-​associated cancers is interesting bio-
lineage. One exception to this general pattern is observed for HL, logically since EBV is the only oncogenic virus that establishes long-​
where EBV(-​) HL appears to have a stronger Class II effect, while term latency in B-​cells. Taken together with findings from GWAS for
EBV(+) HL has a stronger Class  I  effect (Figure 25–2) (Urayama HIV, another virus that infects lymphocytes and for which strong
et al., 2012). Third, for cancers caused by viruses, evidence is accu- HLA Class I associations have been reported for progression to AIDS
mulating to suggest that HLA Class I genes are most important for (Martin and Carrington, 2013), this observation suggests that viral

Extended Class I Class I Class III Class II


30
EBV-positive HL
25 EBV-negative HL
–Log10 (P value)

20

15

10

5
Figure  25–2. Hodgkin’s lymphoma GWAS findings
0 within the major histocompatibility complex (MHC) by
27 28 29 30 31 32 33 tumor EBV status. Source:  Adapted from Urayama K,
Position in MHC Region (MB) et al., JNCI (2011).
468

468 PART III:  THE CAUSES OF CANCER


tropisms are important determinants of the type of HLA association HIV infection is associated with a specific spectrum of cancers
observed with the cancers that they cause. Finally, HLA associations (Dal Maso et al., 2003; Engels et al., 2006b, 2008; Frisch et al., 2001;
have been reported for other cancers, including non-​viral-​associated Grulich et al., 2002, 2007; Newnham et al., 2005; Shiels et al., 2009).
solid tumors and leukemias, but those associations have tended to Because of strongly elevated risks in association with advanced HIV
be less consistent than those observed for virus-​associated cancers infection, three cancers are considered to mark the onset of AIDS
and lymphomas (Bonamigo et al., 2012; Urayama et al., 2013; Wu when they occur in HIV-​infected people: KS, NHL, and cervical can-
et al., 2014). cer (Castro et al., 1992). There is less information on cancer risk in
Other than for HLA, results from candidate immune gene studies HIV-​infected people in sub-​Saharan Africa (Mbulaiteye et al., 2006;
and cancer risk have been largely conflicting and typically modest in Newton et  al., 2001; Tanon et  al., 2012), due to a paucity of relia-
size, making them difficult to interpret (Cerhan et  al., 2007; Engels ble data sources, even though the prevalence of HIV is highest in this
et al., 2007; Hildesheim and Wang, 2002; Hildesheim and Wang, 2012; region of the world. Nonetheless, there are very strong excesses of the
Schoof et  al., 2011; Shiels et  al., 2012a; Wang et  al., 2010b; Wang three AIDS-​defining cancers (KS, NHL, and cervical cancer) in HIV-​
SS et al., 2011). In recent years, more agnostic approaches have been infected people in Africa compared with the general population.
applied in large-​scale studies to query the entire genome for associa- The etiologic contribution of HIV-​induced immunosuppression per
tions with cancer. While these studies have the limitation of identi- se is variable across cancer types, but the importance of immunosup-
fying markers that might not be causal for disease but rather are in pression is indicated by several lines of evidence. First, many of the
linkage disequilibrium with the causal gene, they do identify chromo- same cancers are elevated in both HIV-​infected people and solid organ
somal regions linked to cancer development and provide an indication transplant recipients (Tables 25–3 and 25–4, and see further discus-
for genes in those regions that are potentially causal. A  query of the sion in this chapter) (Grulich et al., 2007). Second, risk for some can-
NHGRI catalog of published GWAS (EMBL-​EBI: performed on May cers in HIV-​infected people increases as the CD4 count declines and
16, 2015 @ www.genome.gov; restricting to etiology studies of cancers is higher after the onset of AIDS (Biggar et al., 2007; Engels et al.,
and lymphomas with greater than 500 cases and 500 controls) revealed 2008; Frisch et al., 2001; Guiguet et al., 2009). Third, the incidence
a total of 584 potential genes linked to one or more of 16 cancers stud- of some cancers is lower in HIV-​infected people receiving HAART
ied within 122 published reports. Review of these genes in six path- and has declined over calendar time after the introduction and with the
way databases (KEGG, PID_​NCI, REACTOME, PID_​BIOCARTA, increasing utilization of HAART (Cancer, 2000; Engels et al., 2006b,
NETPATH, and INOH) revealed 66 genes (11.3% of total) involved 2008; Ledergerber et  al., 1999; Robbins et  al., 2014). Nonetheless,
in the immune response (Table 25–2). Among others, these genes are the incidence of KS and NHL was declining steeply even before the
involved in inflammatory responses (e.g., CRP, IL13, IL4 genes) and introduction of HAART (Engels et al., 2006b), and whether this fall
modulation of responses to soluble immune factors (e.g., IL1RAP, was due to improvements in HIV therapy before 1996 or other factors
IRF4, TNFBR2 genes). As expected, the magnitude of associations is unknown.
observed for individual gene polymorphisms are modest, suggesting KS is one of the most common malignancies in HIV-​infected peo-
that, at best, they each account for a small proportion of these cancers. ple, and the relative risk associated with HIV infection is profound.
In the calendar period before the availability of HAART (i.e., before
1996), KS risk was elevated more than 50,000-​fold among people with
Acquired Conditions AIDS in the United States (Engels et  al., 2006b). KS is caused by
Kaposi sarcoma–​associated herpesvirus (KSHV, also known as human
Cancers Associated with HIV Infection and AIDS herpesvirus 8). The onset of the HIV epidemic led to an explosive
The effect of immunosuppression on cancer risk has been best stud- increase in the incidence of KS in the United States (Shiels MS et al.,
ied in association with HIV/​AIDS and solid organ transplantation, 2011). KS is also one of the most common cancers in the general pop-
because these conditions are relatively common, readily diagnosed ulation in sub-​Saharan Africa (Mbulaiteye et al., 2006; Tanon et al.,
by clinicians, and reportable to state or national registries. The idea 2012; Wabinga et al., 2000), due to the high burden of both HIV and
that these immunosuppressed groups would have an elevated risk of KSHV infection in this region. Among HIV-​infected people, KS risk
cancer is related to the “immune surveillance hypothesis,” which was increases with the degree of immunosuppression (i.e., at lower CD4
first framed by Burnet in 1957 (Burnet, 1957)  and which has been counts) (Biggar et al., 2007; Guiguet et al., 2009). KS incidence has
further developed and updated more recently (Dunn et al., 2002). This declined substantially over time in the United States (Engels et  al.,
hypothesis states that an intact immune system can recognize pre-​neo- 2006b, 2008; Robbins et al., 2014), presumably due, at least in part,
plastic or neoplastic cells as foreign due to the expression by those to improvements in HIV therapy, but risk remains substantially higher
cells of novel proteins (i.e., neoantigens), and that the immune system than in the general population.
can contain or destroy these cells, thereby preventing the development NHL is also common in HIV-​ infected people (Dal Maso et  al.,
of cancer. A corollary of this hypothesis is that people with a weakened 2003; Engels et  al., 2006b, 2008; Frisch et  al., 2001; Grulich
immune system would have increased cancer risk. et  al., 2002, 2007; Newnham et  al., 2005; Shiels et  al., 2009).
The broadest form of the immune surveillance hypothesis appears NHL actually comprises a constellation of distinct entities defined
to be false, because as described in the following, HIV-​infected people by histologic features and clinical characteristics. Among the
and transplant recipients do not have an elevated risk for cancer of all NHL subtypes that arise in HIV-​ infected people, diffuse large
types. Rather, the cancers that are most strongly increased are caused B-​cell lymphoma (DLBCL) is the most common, and risks of DLBCL,
by viruses. Indeed, the greatly elevated risks for Kaposi sarcoma (KS) Burkitt lymphoma, and central nervous system (CNS) NHL are strongly
and Merkel cell carcinoma among people with AIDS (Beral et  al., elevated. Among people with AIDS in the pre-​HAART era, risks were
1990; Engels et  al., 2002)  were important clues that led to the dis- elevated 60-​to 100-​fold for DLBCL, 50-​to 60-​fold for Burkitt lym-
covery of the viral causes of these cancers. Thus, the immune system phoma, and 5000-​fold for CNS NHL (Engels et al., 2006b). The inci-
appears to recognize and contain or clear cells that express viral pro- dence rates of both DLBCL and CNS NHL are lower at higher CD4
teins more readily than for other precancerous or cancerous cells. counts and in people who have not yet developed AIDS (Biggar et al.,
Much of the available data on cancer risk in HIV-​infected peo- 2007; Engels et al., 2008), and both NHL subtypes have declined over
ple come from linkage of large population-​based HIV and cancer time with improvements in HIV therapy (Engels et al., 2006b, 2008). In
registries, particularly from the United States, Europe, and Australia contrast, associations with Burkitt lymphoma are less clear-​cut (Biggar
(Dal Maso et  al., 2003; Engels et  al., 2006b, 2008; Frisch et  al., et  al., 2007; Engels et  al., 2006b, 2008). For unknown reasons, asso-
2001; Grulich et  al., 2002, 2007; Newnham et  al., 2005; Shiels ciations of HIV infection with NHL appear somewhat more modest
et  al., 2009). Results are typically described in terms of standard- in Africa than in developed countries, with relative risks in the range
ized incidence ratios (SIRs) comparing cancer incidence in HIV-​ of 4–​7 (Mbulaiteye et al., 2006; Sitas et al., 2000; Tanon et al., 2012).
infected people to that expected based on general population rates Several less common NHL subtypes, including marginal zone, lympho-
(Table 25–3). plasmacytic, and T-​cell NHLs, are also increased in HIV-​infected people
 469

Table 25–2.  Genes Involved in Immune Response That Have Been Implicated in Cancer from Genome-​Wide Association Studies

Cancers for Which Associations


GENE Symbol Gene Name* Reported References

ADH1C Alcohol dehydrogenase 1C Upper aerodigestive tract cancers McKay et al., 2011
ANK2 Ankyrin 2 Prostate cancer Tao et al., 2012
AR Androgen receptor Prostate cancer Kote-​Jarai et al., 2011
ATF1 Activating transport factor 1 Colorectal cancer Houlston et al., 2010
BMP2 Bone morphogenetic protein 2 Colorectal cancer Peters et al., 2013
CCND1 Cyclin D1 Breast cancer Ahsan et al., 2014; Michailidou et al., 2013;
Turnbull et al., 2010a
CCND2 Cyclin D2 Colorectal cancer Jia et al., 2013; Peters et al., 2013;
Zhang B et al., 2014a
CCNE1 Cyclin E1 Bladder cancer Figueroa et al., 2014b; Rothman et al., 2010
CD180 CD 180 molecule Prostate cancer Tao et al., 2012
CDH1 Cadherin 1 Colorectal cancer Study et al., 2008
CLDN11 Claudin 11 Prostate cancer Kote-​Jarai et al., 2011
CRP C-​reactive protein Lung cancer Amos et al., 2008
CSNK1A1 Casein kinase 1 Esophageal cancer Wu et al., 2011
CYCS Cytochrome C Colorectal cancer Jiao et al., 2012
DAB2 Dab, mitogen-​responsive phosphoprotein, Pancreatic cancer Wu et al., 2012b
homolog 2
DUSP4 Dual specificity phosphatase 4 Colorectal cancer Fernandez-​Rozadilla et al., 2013
EOMES Eomesodermin Hodgkin lymphoma Frampton et al., 2013
ERAP1 Endoplasmic reticulum aminopeptidase 1 Hodgkin lymphoma Urayama et al., 2012
ESR1 Estrogen receptor 1 Breast cancer Couch et al., 2013; Garcia-​Closas et al., 2013;
Long et al., 2012; Siddiq et al., 2012;
Zheng et al., 2009
FGF10 Fibroblast growth factor 10 Prostate cancer Kote-​Jarai et al., 2011
FGFR2 Fibroblast growth factor receptor 2 Breast cancer Ahsan et al., 2014; Elgazzar et al., 2012;
Fletcher et al., 2011; Gaudet et al., 2010;
Hunter et al., 2007; Li et al., 2011; Low
et al., 2013; Michailidou et al., 2013;
Thomas et al., 2009; Turnbull et al., 2010a
GATA3 GATA binding protein 3 Colorectal cancer and Hodgkin lymphoma Cozen et al., 2014; Figueiredo et al., 2014
GNG2 Guanine nucleotide binding protein Lung and non-​melanoma skin cancers Zhang et al., 2013; Zhang R et al., 2014
(G protein) gamma 2
GRHL1 Grainyhead-​Like 1 Prostate cancer Eeles et al., 2013
HLA-​DQB2 Human leukocyte antigen DQ beta 2 Lymphoma Vijai et al., 2013
HLA-​F Human leukocyte antigen F Prostate cancer Tao et al., 2012
IL13 Interleukin 13 Hodgkin’s lymphoma Cozen et al., 2014; Urayama et al., 2012
IL1RAP Interleukin 1 receptor accessory protein Lung cancer Amos et al., 2008
IL4 Interleukin 4 Hodgkin’s lymphoma Cozen et al., 2014
INHBA Inhibin beta A Prostate cancer Tao et al., 2012
IRF4 Interferon regulatory factor 4 Non-​melanoma skin cancer Zhang et al., 2013
ITGA6 Integrin alpha 6 Breast and prostate cancers Eeles et al., 2009; Michailidou et al., 2013
ITPR1 Inositol 1,4,5-​trisphosphate receptor type 1 Breast cancer Michailidou et al., 2013
JUP Junction plakoglobin Esophageal cancer (squamous cell) Wu et al., 2012a
KITLG KIT ligand Testicular germ cell cancer Turnbull et al., 2010b
KLC3 Kinesin light chain 3 Lung cancer Wang et al., 2013
MAP3K1 Mitogen-​activated protein kinase kinase Breast cancer Ahsan et al., 2014; Michailidou et al., 2013;
kinase 1, E3 ubiquitin protein ligase Thomas et al., 2009; Turnbull et al., 2010a
MUC1 Mucin 1, cell surface associated Esophageal and gastric cancers Abnet et al., 2010
MYB V-​Myb avian myeloblastosis viral oncogene Hodgkin lymphoma Frampton et al., 2013
homolog
NLRP1 NLR family pyrin domain containing 1 Non-​small cell lung cancer Yoon et al., 2010
NRG1 Neuregulin 1 Thyroid cancer Gudmundsson et al., 2012
PAG1 Phosphoprotein membrane anchor with Bladder cancer Figueroa et al., 2014b
glycosphingolipid microdomains 1
PARK2 Parkin RBR E3 ubiquitin protein ligase Pancreatic cancer Low et al., 2010
POLR1D Polymerase (RNA) I polypeptide D, 16kDa Large B-​cell lymphoma Kumar et al., 2011b
PRKAA1 Protein kinase, AMP-​activated alpha 1 Gastric cancer Shi et al., 2011
catalytic subunit
PRKACB Protein kinase CAMP-​dependent catalytic Breast cancer (male) Orr et al., 2012
beta
PRKAR2B Protein kinase CAMP-​dependent regulatory Bladder cancer Figueroa et al., 2014a
type II beta
PTPN2 Protein tyrosine phosphatase non-​receptor Esophageal cancer (squamous cell) Wu et al., 2012a
type 2
RUNX1 Runt-​related transcription factor 1 Esophageal cancer Wu et al., 2011
(continued)
470

470 PART III:  THE CAUSES OF CANCER


Table 25–2. Continued

Cancers for Which Associations


GENE Symbol Gene Name* Reported References

SIAH2 Siah E3 ubiquitin protein ligase 2 Breast cancer Elgazzar et al., 2012
SKAP1 Src kinase–​associated phosphoprotein 1 Ovarian cancer Goode et al., 2010; Pharoah et al., 2013
SKIL SKI-​like proto-​oncogene Prostate cancer Kote-​Jarai et al., 2011
SLC22A1 Solute carrier family 22 (organic cation Prostate cancer Eeles et al., 2009; Schumacher et al., 2011
transporter) member 1
SMAD3 SMAD family member 3 Mothers Against Lung cancer Zhang R et al., 2014
Decapentaplegic Homolog 3
SMAD7 SMAD family member 7 Mothers Against Colorectal cancer Broderick et al., 2007; Peters et al., 2012;
Decapentaplegic Homolog 7 Peters et al., 2013; Tenesa et al., 2008;
Tomlinson et al., 2008; Zhang B et al.,
2014a; Zhang B et al., 2014b
SYK Spleen tyrosine kinase Prostate cancer Tao et al., 2012
SYNJ2 Synaptojanin 2 Colorectal cancer Jiao et al., 2012
TCF3 Transcription factor 3 Hodgkin’s lymphoma Cozen et al., 2014
TCF7L2 Transcription factor 7-​like 2 (T-​cell specific Breast and colorectal cancers Couch et al., 2013; Michailidou et al., 2013;
HMG-​box) Zhang B et al., 2014a
TERT Telomerase reverse transcriptase Breast, lung, prostate, and testicular germ Garcia-​Closas et al., 2013; Hu et al., 2011;
cell cancers Kote-​Jarai et al., 2011; Lan et al., 2012;
Michailidou et al., 2013; Purrington et al.,
2014; Turnbull et al., 2010b
TET2 Tet methylcytosine dioxygenase 2 Breast cancer Michailidou et al., 2013
TFF2 Trefoil factor 2 Pancreatic cancer Wu et al., 2012b
TGFBR2 Transforming growth factor beta receptor II Breast cancer Michailidou et al., 2013
(70/​80kDa)
TNFRSF19 Tumor necrosis factor receptor superfamily Lung cancer Hu et al., 2011
member 19
TNRC6B Trinucleotide repeat containing 6B Prostate cancer Amin Al et al., 2013; Sun et al., 2009;
Tao et al., 2012
TUBA1C Tubulin alpha 1C Prostate cancer Kote-​Jarai et al., 2011

* From GeneCards Human Gene Database.

(Gibson et  al., 2014b), and the risk of multiple myeloma is elevated et  al., 2009). HAART use is associated with decreased persistence
(Clifford et al., 2005; Dal Maso et al., 2003; Engels et al., 2006b, 2008; of cervical HPV infection and decreased progression to preneo-
Frisch et al., 2001; Grulich et al., 2002, 2007; Newnham et al., 2005; plastic lesions (Minkoff et  al., 2010), and the incidence of cervical
Shiels et al., 2009). cancer has declined over time among HIV-​infected women in the
Epstein-​Barr virus (EBV) plays an important role in the etiology of United States (Robbins et  al., 2014). For anal cancer, associations
DLBCL, Burkitt lymphoma, and CNS NHL. EBV genomic material with depressed CD4 counts are apparent over prolonged intervals,
and viral proteins can be detected in a sizable fraction of DLBCL and suggesting that HIV-​related immunosuppression affects early stages
Burkitt lymphoma tumors, and in virtually all CNS NHLs, that arise in on the etiologic pathway from HPV infection to cancer (Bertisch
HIV-​infected people (International Agency for Cancer on Research, 1997). et al., 2013). Paradoxically, some recent data indicate a rising trend for
Chronic B-​cell activation also may contribute to the etiology of NHL anal cancer among HIV-​infected people in the United States (Robbins
among HIV-​infected people (Landgren et al., 2010; Vendrame et al., 2014). et al., 2014). HPV is involved in the etiology of a subset of oropha-
HIV-​infected people have an elevated risk of anogenital cancers ryngeal cancers, but risk for oropharyngeal cancer is only modestly
(Chaturvedi et  al., 2009; Dal Maso et  al., 2003; Engels et  al., 2006b, elevated among HIV-​infected people (Chaturvedi et al., 2009).
2008; Frisch et al., 2001; Grulich et al., 2002, 2007; Newnham et al., HIV infection is also associated with elevated risk for other less
2005; Shiels et al., 2009). Among these cancers, the most common are common malignancies in which viruses are implicated. HL, espe-
cervical and anal cancers, but risk is also increased for vulvar, vag- cially the mixed cellularity subtype, occurs in substantial excess,
inal, and penile cancers (Chaturvedi et  al., 2009). Anogenital cancers and most tumors manifest evidence of EBV infection (Clifford
are largely caused by oncogenic subtypes of human papillomavirus et al., 2005; Dal Maso et al., 2003; Engels et al., 2006b, 2008; Frisch
(HPV). HIV-​infected women manifest an elevated prevalence of cervi- et  al., 2001; Grulich et  al., 2002, 2007; Newnham et  al., 2005;
cal HPV infection, which appears at least partly related to decreased Shiels et al., 2009). Children with AIDS have a markedly elevated
HPV clearance, and there is an increased incidence of cervical atypia risk of leiomyosarcoma causally associated with EBV (Simard
and in situ cervical cancer (De Vuyst et al., 2008). Cervical cancer is et  al., 2012). HIV-​infected people also manifest an increased risk
the most common cancer among HIV-​infected women in sub-​Saharan for liver cancer (specifically hepatocellular carcinoma, caused by
Africa (Mbulaiteye et al., 2006; Newton et al., 2001; Tanon et al., 2012). hepatitis B and C viruses), although the contribution of immuno-
High-​risk sexual behaviors, as well as inadequate screening for cervi- suppression is uncertain (Clifford et  al., 2005; Dal Maso et  al.,
cal cancer and incomplete follow-​up for treatment of precursor lesions, 2003; Engels et al., 2006b, 2008; Frisch et al., 2001; Grulich et al.,
likely explain some of the excess risk for cervical cancer among HIV-​ 2002, 2007; Newnham et al., 2005; Shiels et al., 2009). Liver can-
infected women in both developed and developing countries. In the cer risk is highest in subgroups of the HIV population with the
United States, risk of anal cancer is highest among HIV-​infected homo- greatest blood-​borne exposure to hepatitis C virus (i.e., injection
sexual men (reflecting sexual acquisition of anal HPV infection), but all drug users, hemophiliacs) (Sahasrabuddhe et  al., 2012). The inci-
subgroups of HIV-​infected people manifest an elevated risk compared dence of liver cancer is increasing among HIV-​infected people in
with the general population (Chaturvedi et al., 2009). the United States (Robbins et al., 2014; Sahasrabuddhe et al., 2012),
Among HIV-​ infected people, risk of cervical and anal cancer perhaps due to an increase in the duration of hepatitis B and C
increases with immunosuppression (Bertisch et  al., 2013; Guiguet virus infection with prolonged survival. Risk is elevated 13-​fold
 471

Immunologic Factors 471


Table 25–3.  Risk for Selected Cancers in Registry-​Based Cohort Studies of HIV-​Infected People

Frisch et al., Grulich et al., Dal Maso et al., Newnham et al., Engels et al., Engels et al.,
Study 2001 2002 2003 2005 2006b* 2008*

STUDY CHARACTERISTICS
HIV/​AIDS status AIDS HIV or AIDS AIDS HIV or AIDS AIDS HIV without
AIDS
Country US Australia Italy England US US
Calendar years 1980–​1996 1985–​1999 1985–​1998 1985–​2001 1996–​2002 1991–​2002
Number of people 302,834 13,067 12,104 33,190 107,417 57,350
STANDARDIZED INCIDENCE RATIO FOR CANCER (95% CI)
Infection-​related cancers
KS 178 (173–​182) —​ 1750 (1600–​1900) —​ 3640 (3330–​3980) 800 (650–​970)
NHL 73 (70–​75) —​ 350 (320–​390) 43 (39–​46) 23 (21–​25) 5.6 (4.7–​6.6)
Cervix 5.2 (3.8–​6.9) —​ 22 (13–​35) 1.0 (0.2–​2.9) 5.3 (3.6–​7.6) 2.6 (1.6–​3.9)
Anus 23 (17–​30) 37 (18–​68) 34 (12–​74) 23 (14–​37) 20 (14–​26) 8.1 (4.4–​14)
Liver 3.1 (2.0–​4.6) 2.7 (0.6–​8.0) 1.9 (0.4–​5.6) 5.6 (3.0–​9.6) 3.3 (2.0–​5.1) 2.7 (1.4–​4.7)
Hodgkin lymphoma 6.7 (5.3–​8.3) 7.9 (4.4–​13) 16 (12–​22) 5.6 (4.0–​7.7) 14 (11–​17) 4.5 (2.9–​6.6)
Non-​infection-​related cancers
Lung 2.8 (2.4–​3.1) 1.4 (0.8–​2.3) 2.4 (1.5–​3.7) 2.2 (1.6–​3.1) 2.6 (2.1–​3.1) 2.3 (1.8–​2.8)
Kidney 1.2 (0.7–​1.8) 0.8 (0.2–​2.3) 1.1 (0.2–​3.2) 1.1 (0.4–​2.5) 1.8 (1.1–​2.8) —​
Melanoma 1.0 (0.7–​1.5) 1.3 (0.9–​1.9) 0.8 (0.2–​2.4) 0.2 (0–​0.6) 1.0 (0.5–​1.8) —​
Colorectum 0.6 (0.4–​0.9) 0.5 (0.2–​0.9) 1.4 (0.6–​2.6) 0.9 (0.5–​1.5) 1.0 (0.7–​1.4) —​
Prostate 0.5 (0.4–​0.7) 1.1 (0.5–​1.9) 1.2 (0.1–​4.3) 0.9 (0.3–​2.0) 0.5 (0.4–​0.7) —​
Breast 0.5 (0.3–​0.8) 1.1 (0.2–​2.3) 0.7 (0.1–​2.0) 0.8 (0.4–​1.4) 0.8 (0.5–​1.2) —​

Abbreviations: CI = confidence interval; KS = Kaposi sarcoma; NHL = non-​Hodgkin’s lymphoma.


Some estimates were calculated using observed and expected counts calculated from data presented in the cited reports.
*Engels et al. (2006b) also contains additional data for calendar years that overlap with Frisch et al. (2001). Engels et al. (2008) contains additional data on people with AIDS that
overlap with Engels et al. (2006b). The overlapping data are not presented in the table.

for Merkel cell carcinoma (Engels et al., 2002), a rare skin cancer Shiels et al., 2009). The deficit in prostate cancer may partly reflect
caused by the Merkel cell polyomavirus. A strongly elevated risk lower levels of screening or may instead be due to unknown biological
for conjunctival carcinoma has been noted, especially among HIV-​ processes (Marcus et al., 2014; Shiels et al., 2010b).
infected people in Africa (Newton et  al., 2001), although a viral In the United States (and presumably other countries), the HIV
etiology is not established. epidemic has had a measurable impact on the general population
Lung cancer is one of the most common malignancies in HIV-​ trends for KS, NHL, and anal cancer, due to the very high relative
infected people, and risk is elevated approximately 1.5-​to 3-​fold risks associated with HIV infection (Shiels et al., 2012b; Shiels MS
compared with the general population (Dal Maso et al., 2003; Engels et al., 2011). HIV led to a marked increase in the incidence of KS
et  al., 2006b, 2008; Frisch et  al., 2001; Grulich et  al., 2002, 2007; in sub-​Saharan Africa (Wabinga et  al., 2000). HIV infection does
Newnham et al., 2005; Shiels et al., 2009). All subtypes of lung can- not increase risk for common cancers typically associated with aging
cer arise in excess (Chaturvedi et al., 2007). To some extent, this ele- (e.g., prostate, breast, and colorectal cancers), and HIV infection is
vation reflects the very high frequency of current or former smoking not associated with an earlier age at diagnosis for most cancers (Shiels
among HIV-​infected people (estimated to be > 80% in many US stud- et al., 2010c). Nonetheless, with improvements in HIV therapy and
ies) (Kirk et al., 2011), and almost all cases of lung cancer occur in greater longevity, the HIV population is aging. Consequently, there
smokers (Engels et al., 2006a). Importantly, however, the elevation in has been a shift over time in the cancer burden from AIDS-​defin-
lung cancer risk appears to be greater than explained by smoking alone ing cancers to other cancers, including those that are common in the
(Chaturvedi et al., 2007; Engels et al., 2006a; Shiels et al., 2010a; Sigel general population at older ages (Robbins et al., 2015b; Shiels MS
et al., 2012). Although lung cancer can occur in HIV-​infected people et al., 2011).
in the absence of immunosuppression, risk increases with declining Finally, it is of interest to consider whether antiretroviral medica-
CD4 counts (Guiguet et al., 2009), and there has been a decline in lung tions used to treat HIV infection may affect cancer risk, especially
cancer incidence in recent years that may be due to improved HIV since the medications must be used lifelong to suppress viral rep-
therapy (Robbins et al., 2014). One hypothesis is that lung cancers in lication. It is difficult to discern any adverse effects, since the ben-
HIV-​infected people occur due to the synergistic effects of tobacco efits associated with improved immune function are substantial.
and the chronic inflammation that arises from repeated lung infections Initial attention regarding carcinogenicity focused of one class of
(Engels, 2009). HIV medications, nucleoside reverse transcriptase inhibitors, which
There is a slight increase in the incidence of melanoma demon- can cause DNA damage in vitro (Olivero, 2007). Perhaps the best
strated in some studies of HIV-​infected people (Table 25–3) (Dal data on the effects of these drugs come from assessment of HIV-​unin-
Maso et  al., 2003; Engels et  al., 2006b, 2008; Frisch et  al., 2001; fected children who were exposed to zidovudine in utero; these stud-
Grulich et al., 2002, 2007; Lanoy et al., 2009; Newnham et al., 2005; ies, although limited in the number of individuals evaluated and the
Shiels et al., 2009). Based on limited data, there also appears to be a length of follow-​up, have not identified a clear increase in cancer risk
2-​to 3-​fold elevated risk of basal cell and squamous cell skin can- (Benhammou et al., 2008; Hanson et al., 1999). More recently, cohort
cers, with risk for squamous cell skin cancer increasing at lower CD4 studies have compared cancer risk associated with the use of two
counts (Silverberg et al., 2013). broad classes of antiretroviral medications:  protease inhibitors and
HIV-​infected people do not have an elevated risk for other common non-​nucleoside reverse transcriptase inhibitors (Bruyand et al., 2015;
cancers. Indeed, many studies demonstrate a reduced risk for breast Chao et al., 2012). An increased risk of anal cancer has been observed
and prostate cancers, and colorectal cancer risk is similar to that in the for HIV-​infected patients treated with protease inhibitors, but this
general population (Dal Maso et al., 2003; Engels et al., 2006b, 2008; association does not have a ready biological explanation (Bruyand
Frisch et al., 2001; Grulich et al., 2002, 2007; Newnham et al., 2005; et al., 2015; Chao et al., 2012).
472

472 PART III:  THE CAUSES OF CANCER

Cancers Associated with Solid Organ Transplantation considered part of a spectrum of “post-​transplant lymphoprolifera-
Solid organ transplantation is a life-​saving procedure for patients tive disorder” (PTLD) (Swerdlow et al., 2008). PTLD ranges from
with end-​stage organ disease, most commonly of the kidney, liver, benign hyperplasia of lymphoid tissue resembling infectious mon-
heart, or lung. Recipients of these organs must receive lifelong onucleosis, to aggressive polyclonal proliferation of lymphocytes,
immunosuppressive therapy to prevent immune rejection of their to frank malignancy (including NHL, HL, and multiple myeloma).
donor organ. Immunosuppression is maintained with combinations PTLD is largely caused by EBV, which can efficiently drive lym-
of medications, which commonly include a calcineurin inhibitor. phocyte proliferation in the absence of effective immune control
Immunosuppression is most intense in the 1–​2  years immediately (Swerdlow et al., 2008). People who develop primary EBV infection
after transplantation, due to the use of induction therapy with T-​cell after transplantation (primarily pediatric transplant recipients) are at
depleting or modulating antibodies, as well as high doses of oral greatest risk of PTLD, due to the lack of preexisting immunity to
immunosuppressive agents. In the absence of organ rejection, medi- EBV (Opelz et al., 2009).
cations are subsequently tapered to simplify the medication regimen NHL exhibits a biphasic onset after transplantation (Clarke et al.,
and lower medication doses over a period of several years. Rejection 2013; Engels et  al., 2011; van Leeuwen et  al., 2009). Risk is very
of the donor organ is treated with increases in immunosuppressive high in the first year after transplant and is associated with primary
therapy. EBV infection and use of intensive induction immunosuppression
Because transplant immunosuppression mainly targets T-​cell func- (mainly with T-​cell-​depleting antibodies) (Gibson et  al., 2014a; van
tion, the spectrum of cancer in transplant recipients resembles that Leeuwen et  al., 2009). Risk subsequently falls and then rises again
seen in HIV-​infected patients (Tables 25–3 and 25–4) (Adami et al., approximately 3–​5 years after transplantation (Engels et al., 2011; van
2003; Collett et  al., 2010; Engels et  al., 2011; Grulich et  al., 2007; Leeuwen et al., 2009). The majority of post-​transplant NHLs manifest
Vajdic et  al., 2006; Villeneuve et  al., 2007). However, solid organ evidence for EBV infection in tumor cells, although a greater fraction
transplant recipients are at increased risk for several cancers not asso- of late-​onset NHLs are EBV negative compared to early-​onset cases
ciated with HIV infection, which may partly reflect the effects of (Swerdlow et al., 2008). DLBCL is the most common NHL subtype
end-​stage organ disease, other comorbid medical conditions, or direct in solid organ transplant recipients, and risk is elevated 14-​fold com-
carcinogenic effects of immunosuppressive medications. A  simple pared with the general population (Clarke et al., 2013). Risk is strongly
marker of immune status, such as the CD4 count measured in periph- elevated for Burkitt lymphoma and increased for a number of other
eral blood, is not available for transplant recipients. As a result, the less common NHL subtypes as well (Clarke et al., 2013). Transplant
argument that immunosuppression itself is responsible for an increase recipients also have an elevated risk of HL and plasma cell neoplasms
in cancer risk is mainly based on the similarity in the cancer risks (including multiple myeloma), and a large fraction of these cases are
between HIV-​infected people and transplant recipients, the known EBV-​positive (Clarke et al., 2013; Engels et al., 2013).
etiologic contribution of viruses, and the absence of other obvious NHL risk is highest in recipients of a heart, lung, intestine, or pan-
explanations. creas (Clarke et al., 2013; Engels et al., 2011; Opelz and Dohler, 2004),
Other than skin cancers (discussed later in the chapter), NHL is which may reflect the variable intensity of immunosuppression across
the most common malignancy that arises in solid organ transplant transplants of different organs, young age of recipients (for intestinal
recipients (Adami et  al., 2003; Collett et  al., 2010; Engels et  al., transplantation), or the exposure of recipients to donor lymphocytes or
2011; Grulich et al., 2007; Madeleine et al., 2013; Vajdic et al., 2006; EBV delivered with the organ graft (for lung or intestinal transplanta-
Villeneuve et al., 2007). An increased risk for NHL was first described tion). Solid organ transplant recipients have a markedly elevated risk
in kidney transplant recipients in 1973 (Hoover and Fraumeni, 1973), for developing NHL in the transplanted organ (Gibson et  al., 2014a),
which was one of the first clues that this malignancy is caused by which could be caused by chronic immune stimulation against donor
immunosuppression. In the setting of transplantation, NHL is now HLA antigens. Nonetheless, the majority of NHLs that arise in transplant

Table 25–4.  Risk for Selected Cancers in Registry-​Based Cohort Studies of Solid Organ Transplant Recipients

Study Adami et al., 2003 Vajdic et al., 2006 Villeneuve et al., 2007 Collett et al., 2010 Engels et al., 2011

STUDY CHARACTERISTICS
Transplanted organs All Kidney Kidney Kidney* All
Country Sweden Australia Canada UK US
Calendar year of transplants 1970–​1997 Pre-​1986–​2003 1981–​1998 1980–​2007 1987–​2008
Number of recipients 5931 10,180 11,155 25,104 175,732
STANDARDIZED INCIDENCE RATIO FOR CANCER (95% CI)
Infection-​related cancers
KS —​ 208 (114–​349) —​ 17 (8.9–​30) 61 (51–​73)
NHL 6.0 (4.4–​8.0) 9.9 (8.4–​12) 8.8 (7.4–​11) 13 (11–​14) 7.5 (7.2–​7.9)
Cervix 2.0 (0.7–​4.7) 2.5 (1.3–​4.3) 1.6 (0.6–​3.4) 2.3 (1.4–​3.5) 1.0 (0.8–​1.4)
Anus 10 (2.8–​26) 2.8 (1.5–​4.6) —​ 10 (6.6–​15) 5.8 (4.7–​7.2)

Liver 1.1 (0.3–​2.8) 3.2 (1.5–​5.9) 1.8 (0.6–​4.3) 2.4 (1.5–​3.8) 1.0 (0.8–​1.3)
Hodgkin’s lymphoma 2.2 (0.3–​8.1) 3.8 (1.5–​7.7) 3.6 (1.7–​6.9) 7.4 (5.3–​10) 3.6 (2.9–​4.4)
Non-​infection-​related cancers
Lung 1.7 (1.1–​2.5) 2.5 (2.0–​3.0) 2.1 (1.7–​2.5) 1.4 (1.2–​1.6) 2.0 (1.9–​2.1)
Kidney 4.9 (3.3–​7.1) 7.3 (5.7–​9.2) 7.3 (5.7–​9.2) 7.9 (6.7–​9.3) 4.7 (4.3–​5.0)
Melanoma 1.8 (1.0–​3.0) 2.5 (2.1–​3.1) 1.9 (1.2–​3.0) 2.6 (2.0–​3.3) 2.4 (2.1–​2.6)
Colorectum 2.0 (1.4–​2.7) 1.7 (1.4–​2.1) 1.4 (1.0–​1.8) 1.8 (1.6–​2.1) 1.2 (1.2–​1.3)
Prostate 1.1 (0.7–​1.7) 1.0 (0.7–​1.3) 0.9 (0.6-​1.3) 1.1 (0.9–​1.4) 0.9 (0.9–​1.0)
Breast 1.0 (0.6–​1.5) 1.0 (0.8–​1.3) 1.3 (1.0–​1.7) 1.0 (0.8–​1.2) 0.9 (0.8–​0.9)

Abbreviations: CI = confidence interval; KS = Kaposi sarcoma; NHL = non-​Hodgkin’s lymphoma.


Some estimates were calculated using observed and expected counts calculated from data presented in the cited reports.
*
Collett et al. (2010) also presents cancer risk estimates separately for 12,504 recipients of other organs.

The estimate for liver cancer reported in Engels et al. (2011) included prevalent liver cancers in liver recipients. The estimate in the table is instead derived from Koshiol et al. (2014),
which evaluated the same transplant population.
 473

Immunologic Factors 473


recipients are derived from recipient rather than donor lymphocytes RCC (Hurst et al., 2010; Levine et al., 1991). Most often, a patient’s
(Kinch et al., 2014). HLA mismatch between the organ donor and recip- native kidneys are left in place at the time of kidney transplantation,
ient is a risk factor for post-​transplant NHL (Hussain et al., 2015), as and following transplantation, most RCCs develop in the chronically
is chronic immune activation (Engels et al., 2012; Haque et al., 2011). damaged native kidneys rather than the donor kidney. Nonetheless,
As seen in HIV-​infected people, solid organ transplant recipients risk of RCC is also elevated in recipients of organs other than the kid-
also have an elevated risk for KS and HPV-​related anogenital cancers ney (Collett et al., 2010; Engels et al., 2011). The occurrence of RCC
(Adami et al., 2003; Collett et al., 2010; Engels et al., 2011; Grulich in transplant recipients may partly reflect non-​immune side effects
et  al., 2007; Madeleine et  al., 2013; Vajdic et  al., 2006; Villeneuve of immunosuppressive medications, since some are nephrotoxic, and
et al., 2007). The elevation in KS risk, while marked, is less than seen RCC risk is not elevated among HIV-​infected people (Table 25–3).
in HIV (Table 25–4), which may reflect a lower prevalence of KSHV Furthermore, kidney recipients are at increased risk of cancers of
infection. Of interest, some KS tumors are derived from donor cells, the renal pelvis and bladder (Adami et al., 2003; Collett et al., 2010;
suggesting that KS progenitor cells may be conveyed with transplanta- Engels et al., 2011; Grulich et al., 2007; Madeleine et al., 2013; Vajdic
tion (Barozzi et al., 2003). Furthermore, case reports document regres- et al., 2006; Villeneuve et al., 2007), which may be caused by toxic
sion of transplant-​associated KS when patients are switched from a exposures (e.g., analgesics, herbal remedies) that can lead to renal
calcineurin inhibitor-​based regimen to sirolimus (Stallone et al., 2005), failure (Gokmen et al., 2012c, 2013).
consistent with a strong therapeutic effect arising from blocking the Solid organ transplant recipients have an elevated risk of lung can-
mTOR pathway in this tumor. Cervical cancer risk is only slightly cer (Table 25–4), although the increase is less than that seen in HIV-​
increased or (in some transplant populations) not increased (Adami infected people, and a contribution due to immunosuppression is not
et  al., 2003; Collett et  al., 2010; Engels et  al., 2011; Grulich et  al., established. Tobacco use is certainly a major factor (Dickson et al.,
2007; Vajdic et al., 2006; Villeneuve et al., 2007); this pattern likely 2006; Minai et al., 2008). Lung cancer risk is especially high in lung
reflects prevention through effective screening, since risk for in  situ recipients (Collett et al., 2010; Engels et al., 2011); many lung recipi-
cervical cancer is elevated (Madeleine et al., 2013). ents receive only a single donor lung, and lung cancers typically arise
The contribution of immunosuppression to the development of hepa- in the end-​stage native lung (Dickson et al., 2006; Minai et al., 2008).
tobiliary cancers in solid organ transplant recipients is uncertain. Liver Solid organ transplant recipients have modestly increased risk for
recipients have an elevated risk for developing HCC. Of note, HCC colorectal cancer (Table 25–4) (Adami et  al., 2003; Collett et  al.,
is a common indication for liver transplantation, and misdiagnosis of 2010; Engels et  al., 2011; Grulich et  al., 2007; Vajdic et  al., 2006;
some prevalent cancers as incident cases can inflate the risk estimate Villeneuve et al., 2007). Among transplant recipients, risk is most ele-
(Engels et al., 2011). Nonetheless, the elevated risk is not entirely arti- vated for proximal colon cancer, not increased for distal colon can-
factual, and HCC incidence in liver recipients is associated with HBV cer, and actually decreased for rectal cancer (Safaeian et  al., 2015).
and HCV infections as well as diabetes mellitus (Koshiol et al., 2014). Although this pattern is unexplained, the absence of a similar eleva-
HCC risk is not elevated in other recipients of other types of organs. tion in colon cancer risk among HIV-​infected people argues against
Cholangiocarcinoma also arises in excess among transplant recipients a major contribution from immunosuppression. Indeed, much of the
and is associated with the presence of primary sclerosing cholangitis risk may be related to strong increases in colon cancer among liver
as an indication for liver transplant and with the use of azathioprine as recipients with underlying primary sclerosing cholangitis (a condition
maintenance immunosuppressive therapy (Koshiol et al., 2014). associated with inflammatory bowel disease) and lung recipients with
Solid organ transplant recipients have a remarkably elevated risk of cystic fibrosis (associated with chronic gastrointestinal malabsorption)
non-​melanoma skin cancers (approximately 20-​to 100-​fold elevation) (Safaeian et al., 2015). Colorectal cancer risk is also associated with
(Bannon et al., 2014; Jensen et al., 1999; Lindelof et al., 2000; Moloney use of cyclosporine and azathioprine (Safaeian et al., 2015).
et al., 2006). Although risk is elevated for basal cell carcinoma, risk is Transplant recipients also have a reduced risk of prostate and
most increased for squamous cell carcinoma, so that there is a reversal breast cancers (Adami et al., 2003; Collett et al., 2010; Engels et al.,
of the usual preponderance of basal cell carcinomas over squamous cell 2011; Grulich et  al., 2007; Vajdic et  al., 2006; Villeneuve et  al.,
carcinomas. Risk is much higher than that observed in HIV-​infected 2007) (Table 25–4), as seen in HIV-​infected people. Again, the rea-
people (Silverberg et  al., 2013), suggesting the presence of distinct son for these deficits is unknown, and there may be a biological
immune-​related pathways or important contributions from non-​immu- explanation. Alternatively, the pattern could reflect the screening of
nologic factors. One possibility is that there are direct DNA damaging transplant candidates and the removal of prevalent cancers prior to
effects from immunosuppressive medications, especially azathioprine, transplantation.
which appears to act synergistically with ultraviolet radiation to dam- In rare instances, a donor cancer is transmitted along with the trans-
age the skin (Ingvar et  al., 2010; O’Donovan et  al., 2005). Similar planted organ (Desai et al., 2012; Nalesnik et al., 2011; Penn, 1997;
mechanisms may explain the elevated risks for melanoma (Robbins Strauss and Thomas, 2010). Such cancers can be due to the presence
et  al., 2015a), lip cancer (standardized incidence ratios of 17–​ 53) of small tumors of the donor organ that are undetected at the time of
(Adami et al., 2003; Collett et al., 2010; Engels et al., 2011; Grulich transplantation (e.g., a small renal cell carcinoma in the donor kid-
et al., 2007; Vajdic et al., 2006; Villeneuve et al., 2007), and Merkel cell ney) or due to microscopic metastases of other cancers (e.g., mela-
carcinoma (standardized incidence ratio of 24) (Clarke et al., 2015). Of noma) that are not known to the transplant team. Organ donors are
interest, randomized trials have demonstrated that maintenance immu- screened for a remote history or the current presence of an undocu-
nosuppression with sirolimus (an mTOR pathway inhibitor) reduces mented cancer, and most such individuals are prevented from donating
the incidence of non-​melanoma skin cancers in solid organ transplant (Nalesnik et  al., 2011). Transmitted cancers frequently present at a
recipients, compared with other (mainly calcineurin inhibitor-​based) disseminated stage and behave aggressively, in part due to the presence
regimens (Euvrard et al., 2012; Yanik et al., 2015) of immunosuppression.
Kidney cancer arises in excess among transplant recipients (Adami
et al., 2003; Collett et al., 2010; Engels et al., 2011; Grulich et al., Cancers Associated with Hematopoietic
2007; Madeleine et  al., 2013; Vajdic et  al., 2006; Villeneuve et  al., Stem Cell Transplantation
2007), which may be due to a complex assortment of factors. Risk of Hematopoietic stem cell transplantation (HSCT) involves the
renal cell carcinoma (RCC) is most increased among kidney recipi- transplantation of multipotent hematopoietic stem cells from bone
ents (Collett et al., 2010; Engels et al., 2011), and a large fraction of marrow, peripheral blood, or umbilical cord blood. HSCT is most
RCC cases have a papillary histology, as opposed to the predomi- commonly used as treatment for hematologic malignancies, but is
nance of clear cell RCCs observed in the general population (Klatte also sometimes used to treat some non-​malignant hematologic con-
et al., 2010). The excess of RCCs in kidney recipients is partly due ditions or solid tumors. As part of the procedure, it may be necessary
to the effects of end-​stage renal disease. Patients with end-​stage renal to ablate the recipient’s own bone marrow, typically with cytotoxic
disease treated with long-​term dialysis can develop acquired poly- chemotherapy and/​or high-​dose irradiation used as a conditioning
cystic kidney disease, which is associated with the development of regimen. The transplant procedure can be autologous (when the
47

474 PART III:  THE CAUSES OF CANCER


patient’s own stem cells are used) or allogeneic (when the stem cells The medications used to treat autoimmune conditions likely play
come from a donor). a role in the development of some cancers, including NHL. Patients
A major complication of allogeneic HSCT is graft-​versus-​host dis- treated with methotrexate can develop EBV-​ positive lymphomas
ease (GVHD), in which donor-​derived T-​cells attack the recipient’s resembling cases of PTLD, and the tumors can regress following dis-
tissues, most notably the skin, gastrointestinal tract, and liver. Acute continuation of this drug (Kamel et al., 1993), supporting a direct con-
GVHD presents in the first few months after HSCT and is a risk fac- tribution of immunosuppressive therapy. Use of azathioprine or the
tor for chronic GVHD, which can persist for years. Because severe related drug 6-​mercaptopurine in patients with inflammatory bowel
GVHD is a major source of morbidity and mortality in HSCT recipi- disease is associated with an elevated risk of skin cancer (Ariyaratnam
ents, allogeneic HSCT recipients are administered GVHD prophylaxis and Subramanian, 2014), either as a result of immunosuppression or
with immunosuppressive medications in the months immediately fol- sensitization to ultraviolet radiation. Although there has been concern
lowing transplantation. Occurrence of GVHD prompts intensification that medications that inhibit the action of tumor necrosis alpha might
of these medications and the instigation of prolonged immunosuppres- increase the risk of NHL and other cancers (Brown et al., 2002), most
sive therapy. The immunosuppressive medication regimens resemble studies have failed to confirm an association (Lopez-​Olivo et al., 2012;
those used in solid organ transplant recipients (e.g., prophylaxis with Nyboe Andersen et al., 2014).
a calcineurin inhibitor, and treatment of acute GVHD with pulsed ste- A few autoimmune diseases are associated with an elevated risk of
roids). One notable difference is that, in the absence of GVHD, HSCT carcinomas arising in the affected organ, again likely due to chronic
recipients can often be weaned from immunosuppressive medications immune-​related damage and inflammation. Inflammatory bowel dis-
over a period of several years, whereas cessation of immunosuppres- ease (including ulcerative colitis and Crohn’s disease) is accompanied
sive therapy is not possible for solid organ recipients. Autologous by an elevated risk of colorectal cancer, and the magnitude of risk is
HSCT is not associated with GVHD and so does not require the related to the severity, extent, and duration of colonic inflammation
administration of long-​term immunosuppressive therapy. (Beaugerie and Itzkowitz, 2015). Crohn’s disease, which frequently
As in solid organ transplantation, PTLD is an important compli- involves the small intestine, is associated with more than 20-​fold
cation of allogeneic HSCT. PTLD typically develops in the first few increased risk of small intestine cancer (Beaugerie and Itzkowitz,
years post-​ transplantation (Landgren et  al., 2009). EBV infection 2015; Jess et  al., 2005). Patients with primary sclerosing cholangi-
plays an important role in driving lymphocyte proliferation (Zutter tis (which is itself associated with ulcerative colitis) have strongly
et  al., 1988). In contrast to what is observed following solid organ increased risk of cholangiocarcinoma (Beaugerie and Itzkowitz, 2015;
transplantation, the majority of PTLDs in HSCT recipients are derived Claessen et al., 2009).
from donor lymphocytes (Zutter et al., 1988). While some PTLD cases Finally, some autoimmune diseases are associated with cancer
are polyclonal in nature, many cases can be classified as monomorphic because of reverse causation (i.e., the cancer or its immediate precur-
NHL or HL (Landgren et al., 2009; Rowlings et al., 1999; Swerdlow sor predisposes to the development of the autoimmune condition).
et al., 2008). Broad depletion of T-​cells utilized to prevent GVHD is a Examples of such associations include the increased prevalence of der-
strong risk factor for PTLD (Landgren et al., 2009). matomyositis in patients with many different solid tumors (Kurzrock
HSCT recipients are also at elevated risk of developing solid tumors and Cohen, 1995)  and of autoimmune hemolytic anemia and idio-
(Rizzo et al., 2009). Most notably, allogeneic recipients have strongly pathic thrombocytopenic purpura in patients with some subtypes of
increased risk for squamous cell carcinomas of the oral cavity, upper NHL (Anderson et al., 2009; Visco et al., 2014).
gastrointestinal tract, and skin (Rizzo et  al., 2009). Chronic GVHD
commonly affects these sites, and chronic GVHD is a risk factor for Allergic Conditions and Cancer
these malignancies (Curtis et  al., 2005), suggesting that sustained It has been challenging to definitively determine whether allergic
inflammation resulting from GVHD contributes to carcinogenesis. disorders affect the risk of cancer. Although numerous studies have
However, as in solid organ transplantation, it is difficult to disentangle evaluated the relationship between allergic conditions and a number
whether some increase in risk for these cancers results from directly of different cancers, many of the associations are modest in magni-
carcinogenic effects of the immunosuppressive medications. Radiation tude and vary across studies, making a definitive conclusion difficult
therapy given as part of the therapy for the primary cancers that are (Turner et al., 2006).
the indication for HSCT, and the HSCT conditioning regimen itself, Two opposing mechanisms have been hypothesized to explain
are also risk factors for the development of some solid cancers (Rizzo the variable associations observed with allergic conditions. The
et al., 2009). immune surveillance hypothesis was mentioned earlier in the
context of HIV infection and transplantation, where it was used
Autoimmune Diseases and Cancer to explain why the risk of some cancers is increased in people
Patients with certain autoimmune diseases have an increased risk of with immunosuppressive conditions. In the context of atopy, this
NHL, and some of these associations appear to be specific accord- hypothesis is used to explain a decrease in cancer risk. Specifically,
ing to NHL subtype (Morton et al., 2014). Notably, risk of DLBCL the immune surveillance hypothesis posits that an allergic immune
is elevated in patients with systemic lupus erythematosus, Sjögren response may serve to direct T-​cells to tissues where there is pre-
syndrome, and perhaps rheumatoid arthritis (Anderson et  al., 2009; cancerous damage, which would thereby facilitate the immune
Ekstrom Smedby et al., 2008; Morton et al., 2014), likely reflecting destruction of these lesions. Alternatively, allergic responses may
the effects of chronic immune activation. Given the parallel with the lead to chronic tissue damage, which would perhaps predict that
immunodeficiency conditions described earlier, it is likely that the atopy would promote the development of cancer. It is unclear
associations are due to etiological links with these conditions or their which of these competing hypotheses has a more compelling evi-
treatment. Very strong associations are also described for rarer NHL dence base in general, and indeed it is possible that allergy may
subtypes that arise at sites affected by specific autoimmune conditions, have differing effects for different cancers.
presumably because of chronic localized immune stimulation. Patients A methodologic issue in epidemiologic studies of allergic conditions
with Sjögren syndrome have 1000-​fold increased risk for developing and cancer is that many such studies rely on participants’ self-​reports
marginal zone lymphoma in the salivary glands, the site of chronic to ascertain the presence of allergic conditions. Because symptoms of
immune damage in this disorder (Ekstrom Smedby et al., 2008). Celiac allergic conditions are often mild, and the conditions may be confused
disease is associated with a strongly increased risk of T-​cell lympho- with one another or with non-​allergic conditions, the studies may be
mas, including a 250-​fold elevation in risk of enteropathy-​associated prone to misclassification. Also, subjects may inaccurately recall the
T-​cell lymphoma of the small intestine (Ekstrom Smedby et al., 2008). presence of allergic conditions from which they may have suffered in
T-​cell lymphoma risk is also increased in patients with psoriasis or childhood. When cited in the following, measures of association are
other cutaneous autoimmune diseases (Anderson et al., 2009; Ekstrom from large well-​designed studies or meta-​analyses.
Smedby et  al., 2008), but this perhaps reflects diagnostic confusion Several large cohort studies have evaluated cancer risk in association
since many T-​cell lymphomas affect the skin. with asthma or hay fever. Estimates of overall cancer risk vary across
 475

Immunologic Factors 475


studies of people with asthma, from modest protection to slightly surveillance, when risk of these cancers is not strongly increased in
increased risk (i.e., relative risks in the range of 0.7 to 1.1) (Gonzalez-​ immunosuppressed HIV-​infected people or transplant recipients.
Perez et al., 2006; Kallen et al., 1993; Mills et al., 1992; Turner et al.,
2005; Vesterinen et al., 1993). For specific cancer sites, the most con- Inflammation and Cancer
sistent association with asthma is the finding of an elevated risk of Acute inflammation resultant from tissue insults is beneficial and con-
lung cancer (relative risks ranging from 1.1 to 1.7) (Gonzalez-​Perez tributes to the elimination of the agent causing the insult and to tissue
et al., 2006; Kallen et al., 1993; Mills et al., 1992; Turner et al., 2005; repair. However, if the response leads to a chronic inflammatory state,
Vesterinen et al., 1993). Overall cancer risk appears similar in people it can predispose these same tissues to damage that is hypothesized to
with hay fever to people without this condition (Mills et  al., 1992; be a risk factor for tumor development through several mechanisms,
Turner et al., 2005). including increased cellular proliferation and the generation of free
The association between asthma and lung cancer risk has been radicals that can lead to DNA damage (Balkwill and Mantovani, 2001;
evaluated in numerous case-​control studies in addition to the cohort Coussens and Werb, 2002; Grivennikov et al., 2010). Several lines of
studies mentioned in the preceding (Rosenberger et al., 2012; Santillan evidence point to a role for inflammation in the development of cancer
et al., 2003). An association between asthma and lung cancer risk has at various anatomic sites. Immune infiltrates (e.g., neutrophils, mac-
been observed in studies that adjust for tobacco use (Rosenberger rophages, and tumor-​infiltrating lymphocytes) are commonly present
et al., 2012; Santillan et al., 2003). Proposed biological mechanisms at tumor sites and likely play an important part in shaping the tumor
for an increase in lung cancer among individuals with asthma include microenvironment. There is also evidence showing that some agents
damage to the lung from chronic inflammation, reduced clearance of considered carcinogenic induce chronic inflammation, some inflam-
inhaled toxins, or increased pulmonary levels of damaging free radi- matory medical conditions predispose to cancer, genes associated
cals. Nonetheless, one concern has been that the prevalence of smok- with cancer development code for proteins involved in inflammation
ing may be higher in people who report a history of asthma than in responses, and use of anti-​inflammatory agents reduce cancer risk.
people without this history, either because smoking may increase Most infections classified as Class  I  carcinogens by the IARC
the risk of asthma or due to confusion in the reports from study par- induce strong and prolonged inflammatory responses (see Chapter 24).
ticipants between asthma and chronic obstructive pulmonary disease. Helicobacter pylori (causes gastric cancer), hepatitis B and C viruses
Indeed, the association between asthma and lung cancer risk is weaker (cause liver cancer and NHL), human immunodeficiency virus (predis-
among never-​smokers than in ever-​smokers (Rosenberger et al., 2012), poses to multiple cancers, as described earlier), Schistosoma haemato-
suggesting that some estimates may suffer from residual confound- bium (causes bladder cancer), and Opisthorchis viverrini (liver flukes;
ing by smoking. Furthermore, there is substantial heterogeneity across causes biliary track cancers) all induce chronic infections that lead to
studies in the estimated magnitude of the associations of asthma with sustained inflammation at the infection site, which is hypothesized
lung cancer. to partially account for the carcinogenic effects of these infections.
Associations between allergic conditions and risk of brain tumors Similarly, several chemicals and particulates classified as Class I car-
have been extensively investigated. Many case-​control studies have cinogens by IARC, including tobacco, asbestos, diesel exhaust, and
demonstrated protective associations with risk of glioma, for allergic formaldehyde (see also Chapters  11 and 16)  induce inflammation,
conditions overall as well as specifically for asthma (odds ratios [OR] which may be partly responsible for the chronic tissue effects that lead
0.6–​0.8), eczema (OR 0.5–​0.9), and hay fever (OR 0.7–​1.0) (Chen to cancer.
et al., 2011; Zhao et al., 2014). In contrast, associations of serum IgE Various medical conditions that predispose to cancer development
levels with subsequent risk of glioma have been borderline and incon- are also known to induce strong chronic inflammation responses (see
sistent (Calboli et  al., 2011; Schlehofer et  al., 2011; Schwartzbaum discussion earlier in this chapter). These include liver cirrhosis (asso-
et al., 2012). Use of antihistamine medications, frequently utilized by ciated with liver cancer) (El-​Serag and Rudolph, 2007), inflammatory
patients in the management of hay fever, are inversely associated with bowel disease (associated with colorectal and small bowel cancers)
glioma (McCarthy et al., 2011). Use of antihistamine medications may (Beaugerie and Itzkowitz, 2015), gallstones and primary sclerosing
simply be a marker for the presence of allergy. Indeed, in one study that cholangitis (associated with biliary track cancers) (Claessen et  al.,
stratified subjects based on the presence of allergic conditions, antihis- 2009; Maurer et al., 2009), chronic atrophic gastritis (associated with
tamine use was not associated with glioma risk overall, and long-​term gastric cancer) (Sipponen and Maaroos, 2015), chronic lung diseases
use was actually associated with increased risk of high-​grade tumors including chronic obstructive pulmonary disease (associated with lung
(Scheurer et al., 2011). For meningioma, there is some evidence for a cancer) (Brenner et  al., 2011), and diabetes and obesity (associated
decreased risk in association with eczema (Wang M et al., 2011). with numerous cancers) (Giovannucci et al., 2010) (see Chapter 20).
Risk of pancreatic cancer appears reduced in people with allergies, In some instances, these conditions are a near necessary precondition
primarily due to associations with respiratory allergies and hay fever for the development of the cancer with which they are associated, add-
in particular (OR 0.6–​0.9) (Gandini et  al., 2005; Olson et  al., 2013; ing to the biological plausibility and likely importance of inflamma-
Turner et  al., 2005). In contrast, asthma is not associated with pan- tion in the development of these cancers. For example, 70%–​90% of
creatic cancer risk (Gandini et  al., 2005; Olson et  al., 2013; Turner liver cancers are associated with liver cirrhosis (El-​Serag and Rudolph,
et al., 2005). 2007), upward of 80% of gallbladder cancers are preceded by gall-
Hay fever is associated with a modestly reduced risk of develop- stones (Hsing et al., 2007), and Helicobacter pylori–​induced chronic
ing NHL (OR 0.8), and the association does not appear to vary across atrophic gastritis and intestinal metaplasia are considered precursors to
NHL subtypes (Morton et al., 2014). A strong positive association is gastric cancer (Correa, 1988).
observed between eczema and risk of developing mycosis fungoi- As summarized earlier in this chapter, studies of genetic varia-
des/​Sézary syndrome (Morton et al., 2014), but as mentioned earlier tion and cancer also point to a role for genes involved in inflamma-
regarding psoriasis and other autoimmune diseases that affect the skin, tion responses and cancer at various sites (Table 25–2). It should be
this association may represent diagnostic confusion between cutane- noted, however, that the magnitude of the effect for genetic variation
ous diseases. Data on the association of atopy with childhood acute in inflammation genes and individual cancers is typically small and
lymphoblastic leukemia are conflicting (Chang et al., 2012; Linabery therefore not likely to account for a large proportion of disease.
et al., 2010). Finally, evidence has accumulated over the past decade suggest-
Despite much epidemiologic research, it is not straightforward ing that regular, long-​term use of aspirin and possibly other non-​
to reach an etiologic conclusion. The effects, even for some of the ­steroidal anti-​inflammatory drugs (NSAIDs) reduce the risk of some
most promising protective associations (glioma, pancreatic cancer, cancers, most notably colorectal cancer (Thun et al., 2012). NSAIDs
and NHL), are somewhat weak, and the adverse association between act through various mechanisms, including inhibition of enzymes
asthma and lung cancer could be due to confounding. Also, it is unclear (called cyclo-​oxygenases or COX enzymes) involved in the forma-
why certain allergic conditions would decrease the risk of glioma tion of inflammation-​modulating prostaglandins. While low doses
and pancreatic cancer, presumably as a result of enhanced immune of NSAIDs (e.g., 75–​100 mg of aspirin daily) recommended for
476

476 PART III:  THE CAUSES OF CANCER


cardioprotection are required to induce their analgesic and antipyretic efforts to understand the temporal relationship between chronic
effects, considerably higher doses are required to reduce inflamma- inflammation and cancer. Furthermore, the inflammatory response
tion (Thun et al., 2012). It was therefore initially uncertain whether is highly complex and not yet fully understood. Many cell types
low-​dose aspirin use that has been shown to protect against cardio- and effector molecules with pleiotropic effects are involved in the
vascular disease would also translate to risk reduction for cancers. In inflammatory response. Fully defining the pattern of inflammatory
a pooled analysis of six randomized trials that evaluated daily low-​ response and its relationship with cancer has therefore been diffi-
dose aspirin, there was evidence that regular use for 3+ years was cult. Until recently, this difficulty has been exacerbated by the ina-
associated with a 24% reduction in risk of all cancers (OR = 0.76; bility to measure the large number of known inflammation response
95% CI = 0.66, 0.88) (Rothwell et al., 2012). When individual can- molecules using small volumes of available specimens in existing
cers were evaluated in a meta-​analysis of these six randomized trials, studies. This latter problem has been ameliorated in recent years,
evidence for a protective effect was strongest for colorectal cancer, as highly multiplexed, reproducible assays requiring small amounts
where daily aspirin use for 5+ years was associated with a 45% reduc- of specimen have come online (Chaturvedi et  al., 2011). Finally,
tion (OR  =  0.55; 95% CI 0.41–​0.76) (Algra and Rothwell, 2012). even when significant associations between inflammation markers
In this same analysis, estimates of risk reduction using data from and a particular cancer have been observed, it has been difficult to
observational studies yielded comparable and statistically significant ascertain whether those associations mediate previously established
results (risk reduction ranging from 32%–​49%). This justified the risk factors (i.e., causal mediator) for the cancer under study or are
use of both randomized and observational study data to evaluate the explained by them (i.e., confounding).
risk of individual cancers other than colorectal cancer, for which Despite the difficulties listed previously, some progress has been
the randomized trials alone were often underpowered. Evidence for made in evaluating the association between molecular markers of
risk reduction with maximum reported aspirin use that was statisti- inflammation and various cancers. The most extensively studied
cally significant in meta-​analyses of both observational case-​control inflammation biomarker studied to date has been C-​reactive protein
studies and randomized trials was observed for biliary tract (43%–​ (CRP), an acute-​phase inflammation marker in blood induced by the
65% reduction reported) and esophageal (42%–​ 53% reduction pro-​inflammatory cytokine IL-​6 and produced by the liver. CRP is
reported) cancers (Algra and Rothwell, 2012). Statistically signifi- readily measured and has been convincingly linked to risk of cardi-
cant evidence for protection in observational studies alone (with sim- ovascular disease (CVD), so it was considered a good candidate to
ilarly trending but non-​significant effects observed for randomized evaluate in studies of inflammation and cancer. In contrast to CVD,
trials) was also observed for gastric (39% reduction reported) and however, evidence linking CRP levels to cancer risk have been con-
breast (15%–​19% reduction reported) cancers. Effects were weaker flicting. Nonetheless, suggestive evidence for a positive association
or not evident for other cancers. between circulating, pre-​diagnostic CRP levels and cancer risk has
In general, comparable but more limited risk reductions have been been reported for numerous cancers, including colorectal cancer
observed when NSAIDs other than aspirin were evaluated (Rostom (Aleksandrova et al., 2010, 2014b; Ananthakrishnan et al., 2014; Chan
et al., 2007). Evidence for a protective effect has also been stronger et al., 2011; Guo et al., 2013; Song et al., 2013; Toriola et al., 2013;
when higher-​dose use or higher-​risk populations (e.g., individuals Tsilidis et al., 2008), lung cancer (Guo et al., 2013; Shiels et al., 2014,
with a history of colorectal adenomas) were evaluated (Dube et  al., 2015b; Zhou et al., 2012), hepatocellular carcinoma (Aleksandrova
2007). At the time of last review in 2007, the US Preventive Services et al., 2014a; Chen et al., 2015), gallbladder cancer (Aleksandrova
Task Force (USPSTF) recommended against the routine use of aspi- et al., 2014a), gastric cancer (Sasazuki et al., 2010), esophageal carci-
rin and NSAIDs to prevent colorectal cancer in individuals at aver- noma (Hardikar et al., 2014a), ovarian cancer (Ose et al., 2015; Poole
age risk for colorectal cancer, in large part due to the competing et  al., 2013; Trabert et  al., 2014) and endometrial cancer (Wang T
risks associated with the risk increases observed for gastrointestinal et al., 2011). In published meta-​analyses, significant evidence for an
bleeding and hemorrhagic stroke (Dube et  al., 2007; Rostom et  al., association with CRP were noted for all cancers combined, lung can-
2007). Recommendations are currently undergoing re-​review by the cer, colon cancer, and ovarian cancer. Increases in risk of disease
USPSTF, and a recommendation is being contemplated for low-​dose for the individual cancers evaluated were on the order of 10%–​30%
aspirin use for the primary prevention of colorectal cancer in adults per unit change in natural logarithm CRP levels (Guo et al., 2013;
ages 50–​59 years who have a 10% or greater 10-​year cardiovascular Poole et al., 2013; Tsilidis et al., 2008; Zhou et al., 2012). Whether
disease risk, are not at increased risk for bleeding, have a life expec- these associations reflect a causal link and, if so, whether they medi-
tancy of at least 10  years, and are willing to take low-​dose aspiring ate previously known cancer risk factor associations remain to be
daily for at least 10 years (http://​www.uspreventiveservicestaskforce. determined.
org/​BrowseRec/​Search?s=aspirin). Additional markers of inflammation have been directly evaluated
The multiple lines of evidence linking inflammation to cancer for their association with cancer, most notably IL-​6, IL-​8, sTNFRI,
development have motivated studies to directly assess the asso- and sTNFRII (Aleksandrova et al., 2014a; Brenner et al., 2014; Chan
ciation between specific markers of inflammation and cancer risk et al., 2011; Dossus et al., 2013; Edlefsen et al., 2014; Epplein et al.,
(Brenner et al., 2014; Carrick et al., 2015). These studies have not 2013; Hardikar et al., 2014a; Heikkila et al., 2008, 2009; Ho et al.,
been straightforward and have been difficult to interpret, however, 2014; Il’yasova et al., 2005; Kakourou et al., 2015; Ose et al., 2015;
for several reasons. First, tumors induce inflammation themselves, Poole et al., 2013; Shiels et al., 2014; Song et al., 2013; Trabert et al.,
making epidemiologic studies of inflammation and cancer particu- 2014; Vendrame et al., 2014; Wang T et al., 2011; Zhou et al., 2012).
larly prone to reverse causality. As a result, prevalent case-​control As for CRP, while evidence for an association between these markers
studies, while useful for initial screening of relevant inflammation and various cancers have been reported, results have not been entirely
markers, are difficult to interpret, and cohort studies or nested case-​ consistent. Finally, some recent studies have utilized highly multi-
control studies with pre-​diagnostic specimens are needed to care- plexed assays to assess the role of the inflammation pathway in can-
fully account for issues of reverse causality. Second, biologically cer development (Bassig et al., 2015; De Roos et al., 2012; Purdue
relevant inflammation occurs at the tissue level, and measurement of et  al., 2013, 2015; Shiels et  al., 2013, 2014, 2015b; Trabert et  al.,
tissue-​specific inflammation preceding cancer is difficult in the con- 2014). These studies are noteworthy because of their attempt to more
text of epidemiologic studies (Basavaraju et al., 2015; Joshu et al., comprehensively evaluate the link between inflammation and can-
2014; Koshiol and Lin, 2012; Vignozzi and Maggi, 2014). Most cer and to identify the subset of markers with consistent, reproduc-
epidemiological studies of inflammation and cancer have opted to ible associations. Of note are results from discovery and replication
measure circulating levels of inflammation markers, which may or efforts to evaluate inflammation markers associated with NHL and
may not be a good surrogate for inflammation at the tissue level. lung cancer. Studies of NHL have demonstrated robust and consist-
Third, levels of inflammatory markers can vary over time (Hardikar ent associations with B-​cell activation and pro-​inflammatory markers,
et al., 2014b) and cohort studies with available pre-​diagnostic speci- including three soluble receptors (sCD23, sCD27, and sCD30) and a
mens typically have limited serial sampling, again complicating pro-​inflammatory marker receptor (sTNFRII). Odds ratio estimates
 47

Immunologic Factors 477


comparing upper to lower tertiles for these markers were in the range system (Schumacher and Schreiber, 2015). One potential advantage of
of 2–​3. Studies of lung cancer have identified a set of acute-​phase immunotherapy is that the exquisite specificity of the immune system
markers (CRP and SAA), a pro-​inflammatory marker receptor (sTN- might allow targeting of only tumor cells, thereby minimizing treat-
FRII), and a chemokine (CXCL9/​MIG) that are reproducibly asso- ment toxicity.
ciated with future lung cancer risk. Odds ratio estimates comparing Of note, the cancers that are the most frequent focus in the devel-
upper to lower quartiles were in the range of 1.5–​2.0. For both of opment of immunotherapies are not the same as the cancers that are
these cancers, the associations were evident many years before can- increased in immunosuppressed populations (e.g., virus-​related can-
cer diagnosis for many of the markers, suggestive of a true etiological cers). The reasons for this disjunction are not entirely clear. A major
association rather than reverse causality. focus of immunotherapy has been directed against melanoma, which
is increased, but not strongly, in HIV-​infected people and transplant
recipients. Nonetheless, melanoma has been considered an attractive
CANCER OUTCOMES target based on the large number of neoantigens present in this malig-
nancy (Schumacher and Schreiber, 2015), the prognostic relevance
Immunity, Cancer Stage, and Outcomes of tumor-​infiltrating lymphocytes (Clemente et  al., 1996), and the
observation that, in rare cases, melanoma can regress spontaneously
In comparison to investigations relating immunity with the risk of
(presumably due to the development of an effective immune response)
developing cancer, less is known about how immune function affects
(Nathanson, 1976).
the behavior of cancers or the prognosis of cancer patients follow-
Several approaches to immunotherapy have been utilized across a
ing diagnosis. As noted earlier, inflammatory cell infiltrates contribute
range of cancers. Administration of interferon α-​2b has clinical effi-
prominently to the tumor microenvironment. These inflammatory cells
cacy in stimulating an immune response in the treatment of patients
are thought to reflect both the complex processes leading to the cancer
with metastatic melanoma (Kirkwood et al., 1996). Sipuleucel-​T is a
and the host’s immune response to the tumor (Coussens and Werb,
cellular vaccine used in the treatment of advanced stage prostate can-
2002). Inflammatory cells within tumors may play a role in promoting
cer (Karan et al., 2012); treatment consists of isolation of a sample of
metastases (DeNardo et al., 2008), and some studies have described
the patient’s circulating antigen presenting cells, expansion of these
associations of specific features of these infiltrates with the risk of
cells in the presence of a fusion protein based on prostatic acid phos-
cancer recurrence. For example, the intensity of lymphocyte infiltra-
phatase, then reinfusion of the antigen presenting cells back into the
tion has prognostic relevance for melanomas that have begun to grow
patient. Instillation of the bacterium Bacillus Calmette-​Guérrin into
vertically, as indicated by increased tumor thickness (Clemente et al.,
the bladder stimulates a brisk local immune response, which is used as
1996). Likewise, the presence of CD8 positive T-​cells in colorectal
therapy for bladder cancer (Fuge et al., 2015). Rituximab, a monoclo-
and ovarian cancers is associated with improved survival (Gooden
nal antibody against the B-​cell surface protein CD20, is an important
et al., 2011).
component of therapy of some NHLs and probably acts, at least in
Other limited research has related the presence of immune-​related
part, by instigating an immune response against tumor cells (Perez-​
conditions in cancer patients to stage at cancer diagnosis, which itself
Callejo et al., 2015).
has prognostic significance. However, cancer stage is somewhat com-
A major effort in experimental immunotherapy has been directed
plex to assess, because it reflects not only the aggressiveness of a
toward the treatment of cancer through the administration of tumor-​
malignancy but also the timing of diagnosis, which can be affected
directed T-​cells (Rosenberg and Restifo, 2015). This therapy has
by screening or delays in access to medical care. HIV-​infected people
been most actively pursued for metastatic melanoma. Treatment
have an increased risk of being diagnosed at an advanced stage com-
typically involves surgical excision of a tumor lesion, isolation
pared to HIV-​uninfected people for many cancers (especially for lung
and expansion of specific populations of tumor-​infiltrating T-​cells,
and prostate cancers) (Brock et  al., 2006; Shiels et  al., 2015a), but
and reinfusion of these cells into the patient. While progress has
this may simply be due to delayed diagnosis. Along the same lines,
been made along these lines, the complexity and expense of this
transplant recipients tend to present at earlier stages for many cancers,
approach has limited wider evaluation. Similarly, infusion of EBV-​
which probably reflects increased surveillance (Shiels et al., 2015a).
directed T-​cells has been utilized in the treatment of PTLD (Bollard
Both HIV-​infected patients and transplant recipients are at increased
et al., 2012).
risk for presenting with advanced stage tumors for melanoma and
Recently, substantial success has been reported for immuno-
bladder cancer (Shiels et  al., 2015a), which may be most consistent
therapies targeting checkpoint proteins including the programmed
with an increased aggressiveness related to immunosuppression for
death 1 protein (PD-1) and cytotoxic T-lymphocyte-associated
these two cancers.
antigen 4 (CTLA-4). These molecules, which are expressed on the
There are also limited data relating immune conditions to out-
surface of lymphocytes during the inflammatory process, bind to
comes after cancer diagnosis. HIV-​ infected patients have an
their cognate ligands on other cells, down-regulating the immune
increased risk of dying from their cancer compared to HIV-​unin-
response and thus serving to limit tissue destruction. Some cancer
fected patients, for a range of malignancies including cancers of
cells also express the ligands, thereby inhibiting T-cell response
the colorectum, lung, prostate, breast, and pancreas, and melanoma
against tumor neoantigens. Among patients with metastatic mela-
(Coghill et al., 2015; Marcus et al., 2015). Also, transplant recipi-
noma, lung cancer, and other malignancies, infusion of monoclonal
ents with melanoma are more likely to die from their melanoma
antibodies directed against checkpoint molecules has proven effec-
than melanoma patients in the general population (Robbins et al.,
tive in stimulating anti-tumor immunity and prolonging disease-
2015a). It is possible that some of these differences arise from dif-
free survival (Brahmer et  al., 2012; Hodi et  al., 2010; Topalian
ferences in cancer treatment or from inaccuracies in assigning the
et al., 2012).
cause of death in patients with multiple medical conditions. Among
Finally, for HIV-​infected patients, one approach to treating KS is
patients with glioma, elevated serum levels of IgE (indicating the
simply to administer HAART if the patient is not already receiving
presence of atopy) are associated with improved survival (Wrensch
it, which leads to an improvement in immune status (Bower et  al.,
et al., 2006).
2009). Likewise, reduction in the intensity of immunosuppression (by
reducing the doses or eliminating some of the immunosuppressant
medications) is also used in the treatment of some forms of PTLD
Immunotherapy for Cancer
in transplant recipients (Preiksaitis and Keay, 2001). In addition, the
Substantial laboratory and clinical effort has been devoted to devel- infusion of T-​cells directed against EBV can be used in the treatment
oping effective immunotherapy for the treatment of cancer, espe- of PTLD (Khanna et  al., 1999; Rooney et  al., 1995). Nonetheless,
cially for metastatic cancers (Kirkwood et  al., 2012). This approach these approaches are not uniformly successful by themselves, and
is predicated on the idea that tumor cells frequently express neoan- other forms of directed treatment (e.g., chemotherapy) are often
tigens that could potentially be recognized by the adaptive immune required.
478

478 PART III:  THE CAUSES OF CANCER

FUTURE DIRECTIONS Althoff KN, Buchacz K, Hall HI, et al. 2012. U.S. trends in antiretroviral therapy
use, HIV RNA plasma viral loads, and CD4 T-​lymphocyte cell counts among
HIV-​infected persons, 2000 to 2008. Ann Intern Med, 157(5), 325–​335.
As summarized in this chapter, immune factors play a role in the devel-
Amin Al OA, Kote-​Jarai Z, Schumacher FR, et  al. 2013. A meta-​analysis of
opment of cancers at many, if not most, anatomical sites. However, genome-​wide association studies to identify prostate cancer susceptibil-
the immune response is a highly complex and multifaceted system ity loci associated with aggressive and non-​aggressive disease. Hum Mol
that varies over time, making it difficult to study. As a result, much Genet, 22(2), 408–​415.
remains to be learned regarding the specific immunological param- Amos CI, Wu X, Broderick P, et al. 2008. Genome-​wide association scan of
eters involved in cancer development and the underlying mechanisms tag SNPs identifies a susceptibility locus for lung cancer at 15q25.1. Nat
of observed associations. As patterns of immune response become bet- Genet, 40(5), 616–​622. PMCID: PMC2713680.
ter understood and novel technologies for querying the immune sys- Ananthakrishnan AN, Cheng SC, Cai T, et  al. 2014. Serum inflammatory
tem become more widely available, it is likely that our understanding markers and risk of colorectal cancer in patients with inflammatory
bowel diseases. Clin Gastroenterol Hepatol, 12(8), 1342–​ 1348 e1341.
of immunological factors linked to cancer development will increase.
PMCID: PMC4085150.
Promising areas for future discoveries include the delineation of spe- Anderson LA, Gadalla S, Morton LM, et al. 2009. Population-​based study of
cific components of the immune response linked to cancer (made pos- autoimmune conditions and the risk of specific lymphoid malignancies.
sible by “-​omic” technologies), the discovery of new infections linked Int J Cancer, 125(2), 398–​405. PMCID: PMC2692814.
to cancer (made possible by the continued study of immunosuppressed Ariyaratnam J, and Subramanian V. 2014. Association between thiopurine use and
populations), the elucidation of the role of early life immunological nonmelanoma skin cancers in patients with inflammatory bowel disease: a
imprinting on cancer risk later in life (made possible by studies that meta-​analysis. Am J Gastroenterol, 109(2), 163–​169. PMID: 24419479.
measure biologically relevant exposures and their immunological Aydin SE, Kilic SS, Aytekin C et  al. 2015. DOCK8 deficiency:  clinical and
implications in early life), increased understanding of the role of tissue immunological phenotype and treatment options—​ a review of 136
patients. J Clin Immunol, 35(2), 189–​198. PMID: 25627830.
microenviroment on immunological responses that modulate cancer
Balkwill F, and Mantovani A. 2001. Inflammation and cancer: back to Virchow?
risk and prognosis (made possible by systematic tissue banking studies Lancet, 357(9255), 539–​545. PMID: 11229684.
and novel technologies that can be applied to these collections), and Bannon FJ, McCaughan JA, Traynor C, et  al. 2014. Surveillance of non-
better delineation of the patterns of immune response over time that melanoma skin cancer incidence rates in kidney transplant recipients in
are associated with protection and risk for cancer (made possible by Ireland. Transplantation, 98(6), 646–​652. PMID: 24798309.
cohort studies with repeated and appropriate biological sampling). As Barozzi P, Luppi M, Facchetti F, et al. 2003. Post-​transplant Kaposi sarcoma
we learn more about the immunological components that drive cancer originates from the seeding of donor-​derived progenitors. Nat Med, 9(5),
development and its progression, new avenues and opportunities for 554–​561. PMID: 12692543.
improved primary and secondary prevention and treatment of cancer Basavaraju U, Shebl FM, Palmer AJ, et  al. 2015. Cytokine gene polymor-
phisms, cytokine levels and the risk of colorectal neoplasia in a screened
by targeting and harnessing the immune response are possible.
population of Northeast Scotland. Eur J Cancer Prev, 24(4), 296–​304.
PMCID: PMC4411199.
Bassig BA, Shu XO, Koh WP, et al. 2015. Soluble levels of CD27 and CD30
ACKNOWLEDGMENTS are associated with risk of non-​Hodgkin lymphoma in three Chinese pro-
spective cohorts. Int J Cancer, 137(11), 2688–​2695. PMID: 26095604.
The authors would like to acknowledge Qi Yang from the Center for Basu A, Contreras AG, Datta D, et al. 2008. Overexpression of vascular endo-
Genomics Research at the NCI-​LEIDOS in Frederick, Maryland, for thelial growth factor and the development of post-​transplantation cancer.
her support with the pathway-​based analysis required for Table 25–2, Cancer Res, 68(14), 5689–​5698. PMCID: PMC2630878.
David Check for his assistance with the figures, and Sandra Brown for Beaugerie L, and Itzkowitz SH. 2015. Cancers complicating inflammatory
her assistance with text formatting and referencing. bowel disease. N Engl J Med, 372(15), 1441–​1452.
Benhammou V, Warszawski J, Bellec S, et  al. 2008. Incidence of cancer in
References children perinatally exposed to nucleoside reverse transcriptase inhibi-
tors. AIDS, 22(16), 2165–​2177. PMID: 18832880.
Abnet CC, Freedman ND, Hu N, et al. 2010. A shared susceptibility locus in Beral V, Peterman TA, Berkelman RL, and Jaffe HW. 1990. Kaposi’s sarcoma
PLCE1 at 10q23 for gastric adenocarcinoma and esophageal squamous among persons with AIDS:  a sexually transmitted infection? Lancet,
cell carcinoma. Nat Genet, 42(9), 764-​U751. PMCID: PMC2947317. 335(8682), 123–​128.
Adami J, Gabel H, Lindelof B, et al. 2003. Cancer risk following organ trans- Bertisch B, Franceschi S, Lise M, et al. 2013. Risk factors for anal cancer in
plantation:  a nationwide cohort study in Sweden. Br J Cancer, 89(7), persons infected with HIV: a nested case-​control study in the Swiss HIV
1221–​1227. PMCID: PMC2394311. Cohort Study. Am J Epidemiol, 178(6), 877–​884. PMID: 23900553.
Ahsan H, Halpern J, Kibriya MG, et  al. 2014. A genome-​wide association Biggar RJ, Chaturvedi AK, Goedert JJ, Engels EA, and HIV AIDS Cancer
study of early-​onset breast cancer identifies PFKM as a novel breast Match Study. 2007. AIDS-​related cancer and severity of immunosup-
cancer gene and supports a common genetic spectrum for breast can- pression in persons with AIDS. J Natl Cancer Inst, 99(12), 962–​972.
cer at any age. Cancer Epidemiol Biomark Prev, 23(4), 658–​ 669. PMID: 17565153.
PMCID: PMC3990360. Bollard CM, Rooney CM, and Heslop HE. 2012. T-​cell therapy in the treatment
Al-​Herz W, Bousfiha A, Casanova JL, et al. 2014. Primary immunodeficiency of post-​transplant lymphoproliferative disease. Nat Rev Clin Oncol, 9(9),
diseases:  an update on the classification from the international union 510–​519. PMCID: PMC3743122.
of immunological societies expert committee for primary immunodefi- Bonamigo RR, Carvalho AV, Sebastiani VR, Silva CM, and Pinto AC. 2012.
ciency. Front Immunol, 5, 162. PMCID: PMC4001072. HLA and skin cancer. An Bras Dermatol, 87(1), 9–​16; quiz 17–​18.
Aleksandrova K, Boeing H, Nothlings U, et al. 2014a. Inflammatory and meta- PMID: 22481646.
bolic biomarkers and risk of liver and biliary tract cancer. Hepatology, Bower M, Weir J, Francis N, et al. 2009. The effect of HAART in 254 consecu-
60(3), 858–​871. PMCID: PMC4231978. tive patients with AIDS-​related Kaposi’s sarcoma. AIDS, 23(13), 1701–​
Aleksandrova K, Jenab M, Boeing H, et al. 2010. Circulating C-​reactive protein 1706. PMID: 19550283.
concentrations and risks of colon and rectal cancer:  a nested case-​con- Brahmer JR, Tykodi SS, Chow LQ, et al. 2012. Safety and activity of anti-​PD-​
trol study within the European Prospective Investigation into Cancer and L1 antibody in patients with advanced cancer. N Engl J Med, 366(26),
Nutrition. Am J Epidemiol, 172(4), 407–​418. PMID: 20634278. 2455–​2465. PMCID: PMC3563263.
Aleksandrova K, Jenab M, Bueno-​de-​Mesquita HB, et  al. 2014b. Biomarker Brenchley JM, Price DA, Schacker TW, et al. 2006. Microbial translocation is a
patterns of inflammatory and metabolic pathways are associated with risk cause of systemic immune activation in chronic HIV infection. Nat Med,
of colorectal cancer: results from the European Prospective Investigation 12(12), 1365–​1371.
into Cancer and Nutrition (EPIC). Eur J Epidemiol, 29(4), 261–​275. Brenner DR, McLaughlin JR, and Hung RJ. 2011. Previous lung diseases and
PMID: 24791703. lung cancer risk: a systematic review and meta-​analysis. PLoS One, 6(3),
Algra AM, and Rothwell PM. 2012. Effects of regular aspirin on long-​term e17479. PMCID: PMC3069026.
cancer incidence and metastasis:  a systematic comparison of evidence Brenner DR, Scherer D, Muir K, et  al. 2014. A review of the application
from observational studies versus randomised trials. Lancet Oncol, 13(5), of inflammatory biomarkers in epidemiologic cancer research. Cancer
518–​527. PMID: 22440112. Epidemiol Biomarkers Prev, 23(9), 1729–​1751. PMCID: PMC4155060.
 479

Immunologic Factors 479


Brock MV, Hooker CM, Engels EA, et al. 2006. Delayed diagnosis and ele- Clemente CG, Mihm MC, Jr., Bufalino R, et  al. 1996. Prognostic value of
vated mortality in an urban population with HIV and lung cancer: impli- tumor infiltrating lymphocytes in the vertical growth phase of primary
cations for patient care. J Acquir Immune Defic Syndr, 43(1), 47–​55. cutaneous melanoma. Cancer, 77(7), 1303–​1310. PMID: 8608507.
PMID: 16936558. Clifford GM, Polesel J, Rickenbach M, et al. 2005. Cancer risk in the Swiss
Broderick P, Carvajal-​Carmona L, Pittman AM et  al. 2007. A genome-​ HIV Cohort Study:  associations with immunodeficiency, smoking, and
wide association study shows that common alleles of SMAD7 highly active antiretroviral therapy. J Natl Cancer Inst, 97(6), 425–​432.
influence colorectal cancer risk. Nat Genet, 39(11), 1315–​ 1317. PMID: 15770006.
PMID: 17934461. Coghill AE, Shiels MS, Suneja G, and Engels EA. 2015. Elevated cancer-​spe-
Brown SL, Greene MH, Gershon SK, Edwards ET, and Braun MM. 2002. cific mortality among HIV-​infected persons in the U.S. J Clin.Oncol., 33
Tumor necrosis factor antagonist therapy and lymphoma develop- (21), 2376-​2383. PMCID: PMC4500831.
ment:  twenty-​six cases reported to the Food and Drug Administration. Collett D, Mumford L, Banner NR, Neuberger J, and Watson C. 2010.
Arthritis Rheum, 46(12), 3151–​3158. PMID: 12483718. Comparison of the incidence of malignancy in recipients of different
Bruyand M, Ryom L, Shepherd L, et al. 2015. Cancer risk and use of protease types of organ: a UK Registry audit. Am J Transplant, 10(8), 1889–​1896.
inhibitor or nonnucleoside reverse transcriptase inhibitor-​based combina- PMID: 20659094.
tion antiretroviral therapy:  the D:  A:  D study. J Acquir Immune Defic Correa P. 1988. A human model of gastric carcinogenesis. Cancer Res, 48(13),
Syndr, 68(5), 568–​577. PMID: 25763785. 3554–​3560. PMID: 3288329.
Burnet M. 1957. Cancer:  a biological approach. III. Viruses associated with Couch FJ, Wang X, McGuffog L, et  al. 2013. Genome-​wide association
neoplastic conditions. IV. Practical applications. Br Med J, 1(5023), 841–​ study in BRCA1 mutation carriers identifies novel loci associated
847. PMCID: PMC1973618. with breast and ovarian cancer risk. PLoS Genet, 9(3), e1003212.
Calboli FC, Cox DG, Buring JE, et al. 2011. Prediagnostic plasma IgE levels PMCID: PMC3609646.
and risk of adult glioma in four prospective cohort studies. J Natl Cancer Coussens LM, and Werb Z. 2002. Inflammation and cancer. Nature, 420(6917),
Inst, 103(21), 1588–​1595. PMCID: PMC3206038. 860–​867. PMCID: PMC2803035.
International Agency for Cancer on Research. 1997. Epstein-​Barr Virus and Cozen W, Timofeeva MN, Li D, et  al. 2014. A meta-​analysis of Hodgkin
Kaposi’s Sarcoma Herpesvirus/​ Human Herpesvirus 8. Lyon, France, lymphoma reveals 19p13.3 TCF3 as a novel susceptibility locus. Nat
375–494. Commun, 5, 3856. PMCID: PMC4055950.
International Collboration on HIV and Cancer. 2000. Highly active antiretro- Curtis RE, Metayer C, Rizzo JD, et al. 2005. Impact of chronic GVHD therapy
viral therapy and incidence of cancer in human immunodeficiency virus-​ on the development of squamous-​cell cancers after hematopoietic stem-​
infected adults. J Natl Cancer Inst, 92(22), 1823–​1830. cell transplantation: an international case-​control study. Blood, 105(10),
Carrick DM, Chaturvedi AK, Shiels MS, et  al. 2015. Using immune marker 3802–​3811. PMCID: PMC1895092.
panels to evaluate the role of inflammation in cancer:  summary of an Dal Maso L, Franceschi S, Polesel J, et  al. 2003. Risk of cancer in per-
NCI-​sponsored workshop. Cancer Epidemiol Biomarkers Prev, 24(9), sons with AIDS in Italy, 1985–​ 1998. Br J Cancer, 89(1), 94–​ 100.
1427–​1433. PMCID: PMC4690808. PMCID: PMC2394201.
Castro K, Ward J, Slutsker L, et al. 1992. 1993 revised classification system for De Roos AJ, Mirick DK, Edlefsen KL, et al. 2012. Markers of B-​cell activa-
HIV infection and expanded surveillance case definition for AIDS among tion in relation to risk of non-​Hodgkin lymphoma. Cancer Res, 72(18),
adolescents and adults. MMWR, 41(RR-​17), 1–​19. 4733–​4743. PMCID: PMC3445724.
Cerhan JR, Ansell SM, Fredericksen ZS, et al. 2007. Genetic variation in 1253 De Vuyst H, Lillo F, Broutet N, and Smith JS. 2008. HIV, human papillomavi-
immune and inflammation genes and risk of non-​Hodgkin lymphoma. rus, and cervical neoplasia and cancer in the era of highly active antiret-
Blood, 110(13), 4455–​4463. PMCID: PMC2234796. roviral therapy. Eur J Cancer Prev, 17(6), 545–​554. PMID: 18941376.
Chan AT, Ogino S, Giovannucci EL, and Fuchs CS. 2011. Inflammatory mark- Dehghan A, Dupuis J, Barbalic M, et al. 2011. Meta-​analysis of genome-​wide
ers are associated with risk of colorectal cancer and chemopreventive association studies in >80 000 subjects identifies multiple loci for C-​reac-
response to anti-​inflammatory drugs. Gastroenterology, 140(3), 799–​808, tive protein levels. Circulation, 123(7), 731–​738. PMCID: PMC3147232.
quiz e711. PMCID: PMC3049858. DeNardo DG, Johansson M, and Coussens LM. 2008. Immune cells as media-
Chang JS, Tsai YW, Tsai CR, and Wiemels JL. 2012. Allergy and risk of child- tors of solid tumor metastasis. Cancer Metastasis Rev, 27(1), 11–​18.
hood acute lymphoblastic leukemia: a population-​based and record-​based PMID: 18066650.
study. Am J Epidemiol, 176(11), 970–​978. PMID: 23171876. Desai R, Collett D, Watson CJ, et  al. 2012. Cancer transmission from organ
Chao C, Leyden WA, Xu L, et  al. 2012. Exposure to antiretroviral therapy donors-​unavoidable but low risk. Transplantation, 94(12), 1200–​1207.
and risk of cancer in HIV-​infected persons. AIDS, 26(17), 2223–​2231. PMID: 23269448.
PMCID: PMC4562666. Dickson RP, Davis RD, Rea JB, and Palmer SM. 2006. High frequency of
Chaturvedi AK, Kemp TJ, Pfeiffer RM, et  al. 2011. Evaluation of multi- bronchogenic carcinoma after single-​lung transplantation. J Heart Lung
plexed cytokine and inflammation marker measurements:  a methodo- Transplant, 25(11), 1297–​1301. PMCID: PMC3693444.
logic study. Cancer Epidemiol Biomarkers Prev, 20(9), 1902–​ 1911. Dossus L, Lukanova A, Rinaldi S, et al. 2013. Hormonal, metabolic, and inflam-
PMCID: PMC3400264. matory profiles and endometrial cancer risk within the EPIC cohort: a fac-
Chaturvedi AK, Madeleine MM, Biggar RJ, and Engels EA. 2009. Risk of tor analysis. Am J Epidemiol, 177(8), 787–​799. PMID: 23492765.
human papillomavirus-​associated cancers among persons with AIDS. Dowling DJ, and Levy O. 2014. Ontogeny of early life immunity. Trends
J Natl Cancer Inst, 101(16), 1120–​1130. PMCID: PMC2728745. Immunol, 35(7), 299–​310. PMCID: PMC4109609.
Chaturvedi AK, Pfeiffer RM, Chang L, et al. 2007. Elevated risk of lung cancer Dube C, Rostom A, Lewin G, et al. 2007. The use of aspirin for primary pre-
among people with AIDS. AIDS, 21(2), 207–​213. PMID: 17197812. vention of colorectal cancer:  a systematic review prepared for the U.S.
Chen C, Xu T, Chen J, et al. 2011. Allergy and risk of glioma: a meta-​analysis. Preventive Services Task Force. Ann Intern Med, 146(5), 365–​375.
Eur J Neurol, 18(3), 387–​395. PMID: 20722711. Dunn GP, Bruce AT, Ikeda H, Old LJ, and Schreiber RD. 2002. Cancer immu-
Chen D, and Gyllensten U. 2015. Lessons and implications from association noediting:  from immunosurveillance to tumor escape. Nat Immunol,
studies and post-​GWAS analyses of cervical cancer. Trends Genet, 31(1), 3(11), 991–​998. PMID: 12407406.
41–​54. PMID: 25467628. Edlefsen KL, Martinez-​ Maza O, Madeleine MM, et  al. 2014. Cytokines
Chen W, Wang JB, Abnet CC, et  al. 2015. Association between C-​reactive in serum in relation to future non-​Hodgkin lymphoma risk:  evidence
protein, incident liver cancer, and chronic liver disease mortality in the for associations by histologic subtype. Int J Cancer, 135(4), 913–​922.
Linxian Nutrition Intervention Trials: a nested case-​control study. Cancer PMID: 24488825.
Epidemiol Biomarkers Prev, 24(2), 386–​392. PMCID: PMC4323937. Eeles RA, Kote-​Jarai Z, Al Olama AA, et al. 2009. Identification of seven new
Cheson BD. 1995. Infectious and immunosuppressive complications of purine prostate cancer susceptibility loci through a genome-​wide association
analog therapy. J Clin Oncol, 13(9), 2431–​2448. PMID: 7666104. study. Nat Genet, 41(10), 1116–​1121. PMCID: PMC2846760.
Claessen MM, Vleggaar FP, Tytgat KM, Siersema PD, and van Buuren HR. Eeles RA, Olama AA, Benlloch S, et al. 2013. Identification of 23 new prostate
2009. High lifetime risk of cancer in primary sclerosing cholangitis. cancer susceptibility loci using the iCOGS custom genotyping array. Nat
J Hepatol, 50(1), 158–​164. PMID: 19012991. Genet, 45(4), 385–​391, 391e381–​382. PMCID: PMC3832790.
Clarke CA, Morton LM, Lynch C, et  al. 2013. Risk of lymphoma subtypes Ekstrom Smedby K, Vajdic CM, Falster M, et al. 2008. Autoimmune disorders
after solid organ transplantation in the United States. Br J Cancer, 109(1), and risk of non-​Hodgkin lymphoma subtypes: a pooled analysis within
280–​288. PMCID: PMC3708563. the InterLymph Consortium. Blood, 111(8), 4029–​4038.
Clarke CA, Robbins HA, Tatalovich Z, et  al. 2015. Risk of merkel cell car- El-​Serag HB, and Rudolph KL. 2007. Hepatocellular carcinoma:  epidemiol-
cinoma after solid organ transplantation. J Natl Cancer Inst, 107(2). ogy and molecular carcinogenesis. Gastroenterology, 132(7), 2557–​2576.
PMCID: PMC4311175. PMID: 17570226.
480

480 PART III:  THE CAUSES OF CANCER


Elgazzar S, Zembutsu H, Takahashi A, et al. 2012. A genome-​wide association Garcia-​Closas M, Couch FJ, Lindstrom S, et al. 2013. Genome-​wide associa-
study identifies a genetic variant in the SIAH2 locus associated with hor- tion studies identify four ER negative-​specific breast cancer risk loci. Nat
monal receptor-​positive breast cancer in Japanese. J Hum Genet, 57(12), Genet, 45(4), 392–​398, 398e391–​392. PMCID: PMC3771695.
766–​771. PMID: 22951594. Gaudet MM, Kirchhoff T, Green T, et al. 2010. Common genetic variants and
Engels EA. 2009. Non-​ AIDS-​ defining malignancies in HIV-​ infected per- modification of penetrance of BRCA2-​associated breast cancer. PLoS
sons:  etiologic puzzles, epidemiologic perils, prevention opportunities. Genet, 6(10), e1001183. PMCID: PMC2965747.
AIDS, 23(8), 875–​885. PMCID: PMC2677638. Gentzler RD, Altman JK, and Platanias LC. 2012. An overview of the mTOR
Engels EA, Biggar RJ, Hall HI, et al. 2008. Cancer risk in people infected with pathway as a target in cancer therapy. Expert Opin Ther Targets, 16(5),
human immunodeficiency virus in the United States. Int J Cancer, 123(1), 481–​489. PMID: 22494490.
187–​194. PMID: 18435450. German J. 1997. Bloom’s syndrome. XX: the first 100 cancers. Cancer Genet
Engels EA, Brock MV, Chen J, et al. 2006a. Elevated incidence of lung can- Cytogenet, 93(1), 100–​106. PMID: 9062585.
cer among HIV-​infected individuals. J Clin Oncol, 24(9), 1383–​1388. Gibson TM, Engels EA, Clarke CA, et al. 2014a. Risk of diffuse large B-​cell
PMID: 16549832. lymphoma after solid organ transplantation in the United States. Am J
Engels EA, Clarke CA, Pfeiffer RM, et  al. 2013. Plasma cell neoplasms in Hematol, 89(7), 714–​720. PMCID: PMC4069221.
US solid organ transplant recipients. Am J Transplant, 13(6), 1523–​1532. Gibson TM, Morton LM, Shiels MS, Clarke CA, and Engels EA. 2014b. Risk
PMCID: PMC3676887. of non-​ Hodgkin lymphoma subtypes in HIV-​ infected people during
Engels EA, Frisch M, Goedert JJ, Biggar RJ, and Miller RW. 2002. Merkel the HAART era:  a population-​based study. AIDS, 28(15), 2313–​2318.
cell carcinoma and HIV infection. Lancet, 359(9305), 497–​ 498. PMCID: PMC4260326.
PMID: 11853800. Giovannucci E, Harlan DM, Archer MC, et al. 2010. Diabetes and cancer: a
Engels EA, Pfeiffer RM, Fraumeni JF, Jr, et al. 2011. Spectrum of cancer risk consensus report. Diabetes Care, 33(7), 1674–​1685.
among US solid organ transplant recipients. JAMA, 306(17), 1891–​1901. Gladkowska-​Dura M, Dzierzanowska-​Fangrat K, Dura WT, et al. 2008. Unique
PMCID: PMC3310893. morphological spectrum of lymphomas in Nijmegen breakage syndrome
Engels EA, Pfeiffer RM, Goedert JJ, et al. 2006b. Trends in cancer risk among (NBS) patients with high frequency of consecutive lymphoma formation.
people with AIDS in the United States 1980–​2002. AIDS, 20(12), 1645–​ J Pathol, 216(3), 337–​344. PMID: 18788073.
1654. PMID: 16868446. Gokmen MR, Cosyns JP, Arlt VM, et al. 2013. The epidemiology, diagnosis,
Engels EA, Preiksaitis J, Zingone A, and Landgren O. 2012. Circulating anti- and management of aristolochic acid nephropathy:  a narrative review.
body free light chains and risk of posttransplant lymphoproliferative dis- Ann Intern Med, 158(6), 469–​477. PMID: 23552405.
order. Am J Transplant, 12(5), 1268–​1274. PMCID: PMC3338891. Gonzalez-​Perez A, Fernandez-​Vidaurre C, Rueda A, Rivero E, and Garcia
Engels EA, Wu X, Gu J, et al. 2007. Systematic evaluation of genetic variants Rodriguez LA. 2006. Cancer incidence in a general population of
in the inflammation pathway and risk of lung cancer. Cancer Res, 67(13), asthma patients. Pharmacoepidemiol Drug Saf, 15(2), 131–​ 138.
6520–​6527. PMID: 17596594. PMID: 16287212.
Epplein M, Xiang YB, Cai Q, et  al. 2013. Circulating cytokines and gas- Goode EL, Chenevix-​Trench G, Song H, et  al. 2010. A genome-​wide asso-
tric cancer risk. Cancer Causes Control, 24(12), 2245–​ 2250. ciation study identifies susceptibility loci for ovarian cancer at 2q31 and
PMCID: PMC3828746. 8q24. Nat Genet, 42(10), 874–​879. PMCID: PMC3020231.
Euvrard S, Morelon E, Rostaing L, et al. 2012. Sirolimus and secondary skin-​ Gooden MJ, de Bock GH, Leffers N, Daemen T, and Nijman HW. 2011. The
cancer prevention in kidney transplantation. N Engl J Med, 367(4), 329–​ prognostic influence of tumour-​ infiltrating lymphocytes in cancer:  a
339. PMID: 22830463. systematic review with meta-​analysis. Br J Cancer, 105(1), 93–​103.
Fernandez-​Rozadilla C, Cazier JB, Tomlinson IP, et  al. 2013. A colorectal PMCID: PMC3137407.
cancer genome-​wide association study in a Spanish cohort identifies two Grivennikov SI, Greten FR, and Karin M. 2010. Immunity, inflammation, and
variants associated with colorectal cancer risk at 1p33 and 8p12. BMC cancer. Cell, 140(6), 883–​899. PMCID: PMC2866629.
Genomics, 14, 55. PMCID: PMC3616862. Grulich AE, Li YM, McDonald A, et  al. 2002. Rates of non-​AIDS-​defining
Figueiredo JC, Hsu L, Hutter CM, et al. 2014. Genome-​wide diet-​gene interac- infection before and cancers in people with HIV after AIDS diagnosis.
tion analyses for risk of colorectal cancer. PLoS Genet, 10(4), e1004228. AIDS, 16(8), 1155–​1161. PMID: 12004274
PMCID: PMC3990510. Grulich AE, van Leeuwen MT, Falster MO, and Vajdic CM. 2007. Incidence
Figueroa JD, Han SS, Garcia-​Closas M, et al. 2014a. Genome-​wide interaction of cancers in people with HIV/​ AIDS compared with immunosup-
study of smoking and bladder cancer risk. Carcinogenesis, 35(8), 1737–​ pressed transplant recipients: a meta-​analysis. Lancet, 370(9581), 59–​67.
1744. PMCID: PMC4123644. PMID: 17617273.
Figueroa JD, Ye Y, Siddiq A, et al. 2014b. Genome-​wide association study iden- Guba M, von Breitenbuch P, Steinbauer M, et  al. 2002. Rapamycin inhibits
tifies multiple loci associated with bladder cancer risk. Hum Mol Genet, primary and metastatic tumor growth by antiangiogenesis:  involve-
23(5), 1387–​1398. PMCID: PMC3919005. ment of vascular endothelial growth factor. Nat Med, 8(2), 128–​135.
Filipovich AH, Mathur A, Kamat D, Kersey JH, and Shapiro RS. 1994. PMID: 11821896.
Lymphoproliferative disorders and other tumors complicating immuno- Gudmundsson J, Sulem P, Gudbjartsson DF et  al. 2012. Discovery of com-
deficiencies. Immunodeficiency, 5(2), 91–​112. PMID: 8032367. mon variants associated with low TSH levels and thyroid cancer risk. Nat
Fleisher TA, Rieux-​ Laucat F, and Puck JM. 2014. Autoimmune Genet, 44(3), 319–​322. PMCID: PMC3655412.
Lymphoproliferative Syndrome Primary Immunodeficiency Diseases (pp. Guiguet M, Boue F, Cadranel J et al. 2009. Effect of immunodeficiency, HIV
368–​386). Oxford: Oxford University Press. viral load, and antiretroviral therapy on the risk of individual malignan-
Fletcher O, Johnson N, Orr N, et  al. 2011. Novel breast cancer susceptibil- cies (FHDH-​ANRS CO4):  a prospective cohort study. Lancet Oncol,
ity locus at 9q31.2:  results of a genome-​wide association study. J Natl 10(12), 1152–​1159. PMID: 19818686.
Cancer Inst, 103(5), 425–​435. PMID: 21263130. Guo YZ, Pan L, Du CJ, Ren DQ, and Xie XM. 2013. Association between C-​
Frampton M, da Silva Filho MI, Broderick P, et al. 2013. Variation at 3p24.1 reactive protein and risk of cancer: a meta-​analysis of prospective cohort
and 6q23.3 influences the risk of Hodgkin’s lymphoma. Nat Commun, 4, studies. Asian Pacific J Cancer Prev, 14(1), 243–​248. PMID: 23534731
2549. PMID: 24149102. Halloran PF. 2004. Immunosuppressive drugs for kidney transplantation. N
Frisch M, Biggar RJ, Engels EA, Goedert JJ, and GroupAIDS Cancer Engl J Med, 351(26), 2715–​2729. PMID: 15616206.
Match Registry Study Group. 2001. Association of cancer with AIDS-​ Hammarstrom L. 2014. Genetic Approach to Common Variable
related immunosuppression in adults. JAMA, 285(13), 1736–​1745. Immunodeficiency and IgA Deficiency Primary Immunodeficiency
PMID: 11277828. Diseases (pp. 356–​365). Oxford: Oxford University Press.
Fuge O, Vasdev N, Allchorne P, and Green JS. 2015. Immunotherapy for blad- Hansen R, Weemaes CM, and van der Maarel S. 2014. Immunodeficiency
der cancer. Res Rep Urol, 7, 65–​79. PMCID: PMC4427258. with Centromere Instability and Facial Anomalies (ICF Syndrome)
Fulop T, and Montgomery RR. 2014. Editorial overview:  Immune senes- Primary Immunodeficiency Diseases (pp. 662–​ 671). Oxford:  Oxford
cence: known knowns and unknown unknowns. Curr Opin Immunol, 29, University Press.
vii–​ix. PMCID: PMC4389771. Hanson IC, Antonelli TA, Sperling RS, et al. 1999. Lack of tumors in infants
Gabriel G, and Arck PC. 2014. Sex, immunity and influenza. J Infect Dis, 209 with perinatal HIV-​1 exposure and fetal/​neonatal exposure to zidovu-
Suppl 3, S93–​99. PMID: 24966196. dine. J Acquir Immune Defic Syndr Hum Retrovirol, 20(5), 463–​467.
Gandini S, Lowenfels AB, Jaffee EM, Armstrong TD, and Maisonneuve P. PMID: 10225228.
2005. Allergies and the risk of pancreatic cancer:  a meta-​analysis with Haque T, Chaggar T, Schafers J, et al. 2011. Soluble CD30: a serum marker for
review of epidemiology and biological mechanisms. Cancer Epidemiol Epstein-​Barr virus-​associated lymphoproliferative diseases. J Med Virol,
Biomarkers Prev, 14(8), 1908–​1916. PMID: 16103436. 83(2), 311–​316. PMID: 21181928.
 481

Immunologic Factors 481


Hardikar S, Onstad L, Song X, et al. 2014a. Inflammation and oxidative stress cohort. Cancer Epidemiol Biomarkers Prev, 14(10), 2413–​ 2418.
markers and esophageal adenocarcinoma incidence in a Barrett’s esopha- PMID: 16214925.
gus cohort. Cancer Epidemiol Biomarkers Prev, 23(11), 2393–​ 2403. Imai K, Morio T, Zhu Y, et al. 2004. Clinical course of patients with WASP gene
PMCID: PMC4221307. mutations. Blood, 103(2), 456–​464. PMID: 12969986.
Hardikar S, Song X, Kratz M, et al. 2014b. Intraindividual variability over time Ingvar A, Smedby KE, Lindelof B, et al. 2010. Immunosuppressive treatment
in plasma biomarkers of inflammation and effects of long-​term storage. after solid organ transplantation and risk of post-​transplant cutaneous
Cancer Causes Control, 25(8), 969–​976. PMCID: PMC4169209. squamous cell carcinoma. Nephrol Dial Transplant, 25(8), 2764–​2771.
Hayward AR, Levy J, Facchetti F, et al. 1997. Cholangiopathy and tumors of PMID: 19729465.
the pancreas, liver, and biliary tree in boys with X-​linked immunodefi- Jensen P, Hansen S, Moller B, et  al. 1999. Skin cancer in kidney and heart
ciency with hyper-​IgM. J Immunol, 158(2), 977–​983. PMID: 8993019. transplant recipients and different long-​term immunosuppressive therapy
Heikkila K, Ebrahim S, and Lawlor DA. 2008. Systematic review of the asso- regimens. J Am Acad Dermatol, 40(2 Pt 1), 177–​186. PMID: 10025742.
ciation between circulating interleukin-​6 (IL-​6) and cancer. Eur J Cancer, Jess T, Gamborg M, Matzen P, Munkholm P, and Sorensen TI. 2005. Increased
44(7), 937–​945. PMID: 18387296. risk of intestinal cancer in Crohn’s disease:  a meta-​analysis of popula-
Heikkila K, Harris R, Lowe G, et al. 2009. Associations of circulating C-​reac- tion-​
based cohort studies. Am J Gastroenterol, 100(12), 2724–​ 2729.
tive protein and interleukin-​6 with cancer risk: findings from two prospec- PMID: 16393226.
tive cohorts and a meta-​analysis. Cancer Causes Control, 20(1), 15–​26. Jia WH, Zhang B, Matsuo K, et al. 2013. Genome-​wide association analyses
PMID: 18704713. in East Asians identify new susceptibility loci for colorectal cancer. Nat
Hildesheim A, and Wang CP. 2012. Genetic predisposition factors and naso- Genet, 45(2), 191–​196. PMCID: PMC3679924.
pharyngeal carcinoma risk:  a review of epidemiological association Jiang DK, Sun J, Cao G, et al. 2013. Genetic variants in STAT4 and HLA-​DQ
studies, 2000–​2011:  Rosetta Stone for NPC:  genetics, viral infection, genes confer risk of hepatitis B virus-​related hepatocellular carcinoma.
and other environmental factors. Semin Cancer Biol, 22(2), 107–​116. Nat Genet, 45(1), 72–​75. PMCID: PMC4105840.
PMCID: PMC3296903. Jiao S, Hsu L, Berndt S, et  al. 2012. Genome-​ wide search for gene-​
Hildesheim A, and Wang SS. 2002. Host and viral genetics and risk of cervical gene interactions in colorectal cancer. PLoS One, 7(12), e52535.
cancer: a review. Virus Res, 89(2), 229–​240. PMID: 12445662. PMCID: PMC3530500.
Hjalgrim H, Askling J, Sorensen P, et al. 2000. Risk of Hodgkin’s disease and Joshu CE, Tsilidis KK, Peskoe SB, et al. 2014. The association between cir-
other cancers after infectious mononucleosis. J Natl Cancer Inst, 92(18), culating high-​sensitivity C-​reactive protein concentration and pathologic
1522–​1528. PMID: 10995808. measures of colonic inflammation. Cancer Causes Control, 25(4), 409–​
Hjalgrim H, Rostgaard K, Johnson PC, et al. 2010. HLA-​A alleles and infec- 418. PMCID: PMC3951758.
tious mononucleosis suggest a critical role for cytotoxic T-​cell response Kakourou A, Koutsioumpa C, Lopez DS, et  al. 2015. Interleukin-​6 and risk
in EBV-​related Hodgkin lymphoma. Proc Natl Acad Sci U S A, 107(14), of colorectal cancer: results from the CLUE II cohort and a meta-​analy-
6400–​6405. PMCID: PMC2851961. sis of prospective studies. Cancer Causes Control, 26(10), 1449–​1460.
Ho GY, Wang T, Zheng SL, et al. 2014. Circulating soluble cytokine receptors PMCID: PMC4763881.
and colorectal cancer risk. Cancer Epidemiol Biomarkers Prev, 23(1), Kallen B, Gunnarskog J, and Conradson TB. 1993. Cancer risk in asthmatic
179–​188. PMCID: PMC3947182. subjects selected from hospital discharge registry. Eur Respir J, 6(5),
Hodi FS, O’Day SJ, McDermott DF, et al. 2010. Improved survival with ipi- 694–​697. PMID: 8519380.
limumab in patients with metastatic melanoma. N Engl J Med, 363(8), Kamel OW, van de Rijn M, Weiss LM, et al. 1993. Brief report: reversible lym-
711–​723. PMCID: PMC3549297. phomas associated with Epstein-​Barr virus occurring during methotrex-
Hojo M, Morimoto T, Maluccio M, et al. 1999. Cyclosporine induces cancer ate therapy for rheumatoid arthritis and dermatomyositis. N Engl J Med,
progression by a cell-​autonomous mechanism. Nature, 397(6719), 530–​ 328(18), 1317–​1321. PMID: 8385742.
534. PMID: 10028970. Karan D, Holzbeierlein JM, Van Veldhuizen P, and Thrasher JB. 2012. Cancer
Hoover R, and Fraumeni JF, Jr. 1973. Risk of cancer in renal-​transplant recipi- immunotherapy: a paradigm shift for prostate cancer treatment. Nat Rev
ents. Lancet, 2(7820), 55–​57. PMID: 4123616. Urol, 9(7), 376–​385. PMID: 22641164.
Houlston RS, Cheadle J, Dobbins SE, et  al. 2010. Meta-​analysis of three Kennedy RB, Ovsyannikova IG, Haralambieva IH, et al. 2014. Genome-​wide
genome-​wide association studies identifies susceptibility loci for colorec- SNP associations with rubella-​specific cytokine responses in measles-​
tal cancer at 1q41, 3q26.2, 12q13.13 and 20q13.33. Nat Genet, 42(11), mumps-​rubella vaccine recipients. Immunogenetics, 66(7–​8), 493–​499.
973–​977. PMID: 20972440. PMCID: PMC4096048.
Hsing AW, Gao YT, Han TQ, et  al. 2007. Gallstones and the risk of biliary Kennedy RB, Ovsyannikova IG, Pankratz VS, et  al. 2012. Genome-​ wide
tract cancer:  a population-​based study in China. Br J Cancer, 97(11), genetic associations with IFNgamma response to smallpox vaccine. Hum
1577–​1582. Genet, 131(9), 1433–​1451. PMCID: PMC4170655.
Hu X, and Daly M. 2012. What have we learned from six years of GWAS in auto- Khanna R, Bell S, Sherritt M, et al. 1999. Activation and adoptive transfer of
immune diseases, and what is next? Curr Opin Immunol, 24(5), 571–​575. Epstein-​Barr virus-​specific cytotoxic T cells in solid organ transplant
Hu Z, Wu C, Shi Y, et  al. 2011. A genome-​wide association study identifies patients with posttransplant lymphoproliferative disease. Proc Natl Acad
two new lung cancer susceptibility loci at 13q12.12 and 22q12.2 in Han Sci U S A, 96(18), 10391–​10396. PMCID: PMC17898.
Chinese. Nat Genet, 43(8), 792–​796. PMID: 21725308. Kinch A, Cavelier L, Bengtsson M, et al. 2014. Donor or recipient origin of
Huang X, Kushekhar K, Nolte I, et  al. 2012. HLA associations in classical posttransplant lymphoproliferative disorders following solid organ trans-
Hodgkin lymphoma:  EBV status matters. PLoS One, 7(7), e39986. plantation. Am J Transplant, 14(12), 2838–​2845. PMID: 25307322.
PMCID: PMC3393726. Kirk GD, Merlo CA, and Lung HIV Study. 2011. HIV infection in the etiol-
Hunter DJ, Kraft P, Jacobs KB, et al. 2007. A genome-​wide association study ogy of lung cancer: confounding, causality, and consequences. Proc Am
identifies alleles in FGFR2 associated with risk of sporadic postmeno- Thorac Soc, 8(3), 326–​332. PMCID: PMC3132793.
pausal breast cancer. Nat Genet, 39(7), 870–​874. PMCID: PMC3493132. Kirkwood JM, Butterfield LH, Tarhini AA, et al. 2012. Immunotherapy of can-
Hurst FP, Jindal RM, Graham LJ, et al. 2010. Incidence, predictors, costs, and cer in 2012. CA Cancer J Clin, 62(5), 309–​335. PMCID: PMC3445708.
outcome of renal cell carcinoma after kidney transplantation:  USRDS Kirkwood JM, Strawderman MH, Ernstoff MS, et al. 1996. Interferon alfa-​2b
experience. Transplantation, 90(8), 898–​904. PMID: 21248500. adjuvant therapy of high-​risk resected cutaneous melanoma: the Eastern
Hussain SK, Makgoeng SB, Everly MJ, et al. 2015. HLA and risk of diffuse Cooperative Oncology Group Trial EST 1684. J Clin Oncol, 14(1), 7–​17.
large B-​cell lymphoma after solid organ transplantation. Transplantation. PMID: 8558223.
PMID: 26636741. Klatte T, Seitz C, Waldert M, et  al. 2010. Features and outcomes of renal
IARC. 1997 International Agency for Research on Cancer. Epstein-Barr virus cell carcinoma of native kidneys in renal transplant recipients. BJU Int,
and Kaposi’s sarcoma herpesvirus/human herpesvirus 8. Lyon, France: 105(9), 1260–​1265. PMID: 19818071.
1997:375-494. Klein SL, Jedlicka A, and Pekosz A. 2010. The Xs and Y of immune responses
IARC. 2012a. Azathioprine Pharmaceuticals (volume 100A, pp. 319–​331). to viral vaccines. Lancet Infect Dis, 10(5), 338–​349. PMID: 20417416.
Lyon: IARC. Kocar IH, Yesilova Z, Ozata M, et al. 2000. The effect of testosterone replace-
IARC. 2012b. Ciclosporin Pharmaceuticals (volume 100A, pp. 337–​345). ment treatment on immunological features of patients with Klinefelter’s
Lyon: IARC. syndrome. Clin Exp Immunol, 121(3), 448–​452. PMCID: PMC1905740.
IARC. 2012c. Phenacetin Pharmaceuticals (volume 100A, pp. 377–​ 398). Kogut I, Scholz JL, Cancro MP, and Cambier JC. 2012. B cell maintenance and
Lyon: IARC. function in aging. Semin Immunol, 24(5), 342–​349. PMID: 22560930.
Il’yasova D, Colbert LH, Harris TB, et al. 2005. Circulating levels of inflam- Koshiol J, and Lin SW. 2012. Can tissue-​based immune markers be used for
matory markers and cancer risk in the health aging and body composition studying the natural history of cancer? Ann Epidemiol, 22(7), 520–​530.
482

482 PART III:  THE CAUSES OF CANCER


Koshiol J, Pawlish K, Goodman MT, McGlynn KA, and Engels EA. Madeleine MM, Finch JL, Lynch CF, Goodman MT, and Engels EA. 2013.
2014. Risk of hepatobiliary cancer after solid organ transplant in the HPV-​related cancers after solid organ transplantation in the United States.
United States. Clin Gastroenterol Hepatol, 12(9), 1541–​1549 e1543. Am J Transplant, 13(12), 3202–​3209. PMCID: PMC4049182.
PMCID: PMC4064001. Majewski S, Jablonska S, and Orth G. 1997. Epidermodysplasia verruciformis.
Kote-​Jarai Z, Olama AA, Giles GG, et al. 2011. Seven prostate cancer suscepti- Immunological and nonimmunological surveillance mechanisms: role in
bility loci identified by a multi-​stage genome-​wide association study. Nat tumor progression. Clin Dermatol, 15(3), 321–​334. PMID: 9255439.
Genet, 43(8), 785–​791. PMCID: PMC3396006. Makitie O. 2014. Cartilage-​ Hair Hypoplasia Primary Immunodeficiency
Kumar V, Kato N, Urabe Y, et al. 2011a. Genome-​wide association study identi- Diseases. (3rd ed., pp. 484–​488). Oxford: Oxford University Press.
fies a susceptibility locus for HCV-​induced hepatocellular carcinoma. Nat Marcus JL, Chao C, Leyden WA, et  al. 2015. Survival among HIV-​infected
Genet, 43(5), 455–​458. PMID: 21499248. and HIV-​ uninfected individuals with common non-​ AIDS-​defining can-
Kumar V, Matsuo K, Takahashi A, et al. 2011b. Common variants on 14q32 and cers. Cancer Epidemiol Biomarkers Prev, 24(8), 1167–​ 1173. PMCID:
13q12 are associated with DLBCL susceptibility. J Hum Genet, 56(6), PMC4526437.
436–​439. PMID: 21471979. Marcus JL, Chao CR, Leyden WA, et al. 2014. Prostate cancer incidence and
Kurzrock R, and Cohen PR. 1995. Cutaneous paraneoplastic syndromes in prostate-​specific antigen testing among HIV-​positive and HIV-​negative
solid tumors. Am J Med, 99(6), 662–​671. PMID: 7503090. men. J Acquir Immun Defic Syndr, 66(5), 495–​502. PMID: 24820107.
Lan Q, Hsiung CA, Matsuo K, et al. 2012. Genome-​wide association analysis Martin MP, and Carrington M. 2013. Immunogenetics of HIV disease. Immunol
identifies new lung cancer susceptibility loci in never-​smoking women in Rev, 254(1), 245–​264. PMCID: PMC3703621.
Asia. Nat Genet, 44(12), 1330–​1335. PMCID: PMC4169232. Maurer KJ, Carey MC, and Fox JG. 2009. Roles of infection, inflammation, and
Landgren O, Gilbert ES, Rizzo JD, et al. 2009. Risk factors for lymphoprolifer- the immune system in cholesterol gallstone formation. Gastroenterology,
ative disorders after allogeneic hematopoietic cell transplantation. Blood, 136(2), 425-​440. PMCID: PMC2774219.
113(20), 4992–​5001. PMCID: PMC2686146. Mbulaiteye SM, Katabira ET, Wabinga H, et  al. 2006. Spectrum of cancers
Landgren O, Goedert JJ, Rabkin CS, et al. 2010. Circulating serum free light among HIV-​ infected persons in Africa:  the Uganda AIDS-​ Cancer
chains as predictive markers of AIDS-​related lymphoma. J Clin Oncol, Registry Match Study. Int J Cancer, 118(4), 985-​990. PMID: 16106415.
28(5), 773–​779. PMCID: PMC2834393. McCarthy BJ, Rankin K, Il’yasova D, et al. 2011. Assessment of type of allergy
Lanoy E, Dores GM, Madeleine MM, et al. 2009. Epidemiology of nonkera- and antihistamine use in the development of glioma. Cancer Epidemiol
tinocytic skin cancers among persons with AIDS in the United States. Biomarkers Prev, 20(2), 370-​378. PMCID: PMC3077928.
AIDS, 23(3), 385–​393. PMCID: PMC2728602. McKay JD, Truong T, Gaborieau V, et  al. 2011. A genome-​wide association
Lazarczyk M, Cassonnet P, Pons C, Jacob Y, and Favre M. 2009. The EVER study of upper aerodigestive tract cancers conducted within the INHANCE
proteins as a natural barrier against papillomaviruses: a new insight into consortium. PLoS Genet, 7(3), e1001333. PMCID: PMC3060072.
the pathogenesis of human papillomavirus infections. Microbiol Mol Biol Mellemkjaer L, Hammarstrom L, Andersen V, et al. 2002. Cancer risk among
Rev, 73(2), 348–​370. PMCID: PMC2698414. patients with IgA deficiency or common variable immunodeficiency
Ledergerber B, Egger M, Erard V, et al. 1999. AIDS-​related opportunistic ill- and their relatives:  a combined Danish and Swedish study. Clin Exper
nesses occurring after initiation of potent antiretroviral therapy: the Swiss Immunol, 130(3), 495–​500. PMID: 12452841
HIV Cohort Study. JAMA, 282(23), 2220–​2226. PMID: 10605973. Michailidou K, Hall P, Gonzalez-​Neira A, et al. 2013. Large-​scale genotyping
Lederman MM, Funderburg NT, Sekaly RP, Klatt NR, and Hunt PW. 2013. identifies 41 new loci associated with breast cancer risk. Nat Genet, 45(4),
Residual immune dysregulation syndrome in treated HIV infection. Adv 353–​361. PMCID: PMC3771688.
Immunol, 119, 51–​83. PMCID: PMC4126613. Mills PK, Beeson WL, Fraser GE, and Phillips RL. 1992. Allergy and can-
Levine E, Slusher SL, Grantham JJ, and Wetzel LH. 1991. Natural history of cer:  organ site-​specific results from the Adventist Health Study. Am J
acquired renal cystic disease in dialysis patients: a prospective longitudi- Epidemiol, 136(3), 287–​295. PMID: 1415150.
nal CT study. AJR Am J Roentgenol, 156(3), 501–​506. PMID: 1899744. Minai OA, Shah S, Mazzone P, et al. 2008. Bronchogenic carcinoma after lung
Li J, Humphreys K, Heikkinen T, et al. 2011. A combined analysis of genome-​ transplantation:  characteristics and outcomes. J Thorac Oncol, 3(12),
wide association studies in breast cancer. Breast Cancer Res Treat, 1404–​1409. PMID: 19057264.
126(3), 717–​727. PMID: 20872241. Minkoff H, Zhong Y, Burk RD, et al. 2010. Influence of adherent and effective
Li S, Qian J, Yang Y, et al. 2012. GWAS identifies novel susceptibility loci on antiretroviral therapy use on human papillomavirus infection and squa-
6p21.32 and 21q21.3 for hepatocellular carcinoma in chronic hepatitis B mous intraepithelial lesions in human immunodeficiency virus-​positive
virus carriers. PLoS Genet, 8(7), e1002791. PMCID: PMC3395595. women. J Infect Dis, 201(5), 681–​690. PMCID: PMC2818607.
Liao M, Ye F, Zhang B, et  al. 2012. Genome-​wide association study iden- Moir S, and Fauci AS. 2009. B cells in HIV infection and disease. Nat Rev
tifies common variants at TNFRSF13B associated with IgG level in Immunol, 9(4), 235–​245. PMCID: PMC2779527.
a healthy Chinese male population. Genes Immun, 13(6), 509–​ 513. Moloney FJ, Comber H, O’Lorcain P, et al. 2006. A population-​based study of
PMID: 22673310. skin cancer incidence and prevalence in renal transplant recipients. Br J
Lin ZH, Xin YN, Dong QJ et al. 2010. Association between HLA-​DRB1 alleles Dermatol, 154(3), 498–​504. PMID: 16445782.
polymorphism and hepatocellular carcinoma:  a meta-​ analysis. BMC Morton LM, Slager SL, Cerhan JR, et al. 2014. Etiologic heterogeneity among non-​
Gastroenterol, 10, 145. PMCID: PMC3016400. Hodgkin lymphoma subtypes:  the InterLymph Non-​Hodgkin Lymphoma
Linabery AM, Jurek AM, Duval S, and Ross JA. 2010. The association Subtypes Project. J Natl Cancer Inst Monogr, 2014(48), 130–​144.
between atopy and childhood/​adolescent leukemia: a meta-​analysis. Am J Nalbandian G, and Kovats S. 2005. Understanding sex biases in immu-
Epidemiol, 171(7), 749–​764. PMCID: PMC2877483. nity:  effects of estrogen on the differentiation and function of antigen-​
Lindelof B, Sigurgeirsson B, Gabel H, and Stern RS. 2000. Incidence of skin presenting cells. Immunol Res, 31(2), 91–​106. PMID: 15778508.
cancer in 5356 patients following organ transplantation. Br J Dermatol, Nalesnik MA, Woodle ES, Dimaio JM, et al. 2011. Donor-​transmitted malig-
143(3), 513–​519. PMID: 10971322. nancies in organ transplantation:  assessment of clinical risk. Am J
Long J, Cai Q, Sung H et  al. 2012. Genome-​wide association study in east Transplant, 11(6), 1140–​1147. PMID: 21645251.
Asians identifies novel susceptibility loci for breast cancer. PLoS Genet, Nathanson. 1976. Spontaneous regression of malignant melanoma: a review of
8(2), e1002532. PMCID: PMC3285588. the literature on incidence, clinical features, and possible mechanisms.
Lopez-​Olivo MA, Tayar JH, Martinez-​Lopez JA, et al. 2012. Risk of malignan- Natl Cancer Inst Monogr, 44, 67–​76. PMID: 799762.
cies in patients with rheumatoid arthritis treated with biologic therapy: a Nelson F, and Blauvelt C. 2012. Allergy. In: Holgate ST, Broide DH, Church
meta-​analysis. JAMA, 308(9), 898–​908. PMID: 22948700. MK, and Martinez FD (Eds.), 4th ed. Edinburgh: Elsevier.
Lopez-​Vazquez A, Rodrigo L, Mina-​Blanco A, et  al. 2004. Extended human Newnham A, Harris J, Evans HS, Evans BG, and Moller H. 2005. The risk of
leukocyte antigen haplotype EH18.1 influences progression to hepatocel- cancer in HIV-​infected people in southeast England: a cohort study. Br J
lular carcinoma in patients with hepatitis C virus infection. J Infect Dis, Cancer, 92(1), 194–​200. PMCID: PMC2361741.
189(6), 957–​963. PMID: 14999597. Newton R, Ziegler J, Beral V, et al. 2001. A case-​control study of human immu-
Low SK, Kuchiba A, Zembutsu H, et al. 2010. Genome-​wide association study nodeficiency virus infection and cancer in adults and children residing
of pancreatic cancer in Japanese population. PLoS One, 5(7), e11824. in Kampala, Uganda. Int J Cancer, 92(5), 622–​627. PMID: 11340563.
PMCID: PMC2912284. Nikolich-​
Zugich J. 2014. Aging of the T cell compartment in mice and
Low SK, Takahashi A, Ashikawa K, et  al. 2013. Genome-​wide association humans:  from no naive expectations to foggy memories. J Immunol,
study of breast cancer in the Japanese population. PLoS One, 8(10), 193(6), 2622–​2629. PMCID: PMC4157314.
e76463. PMCID: PMC3797071. Notarangelo LD, Giliani S, and Plebani A. 2014. CD40 and CD40 Ligand
Mackay IR, and Rose RR (Eds.). 2006. The Autoimmune Diseases. 4th ed. Deficiencies Primary Immunodeficiency Diseases (3rd ed., pp. 324–​342).
St. Louis, MO: Academic Press. Oxford: Oxford University Press.
 483

Immunologic Factors 483


Notarangelo LD, Lanzi G, Peron S, and Durandy A. 2006. Defects of class-​switch Reiman A, Srinivasan V, Barone G, et al. 2011. Lymphoid tumours and breast
recombination. J Allergy Clin Immunol, 117(4), 855–​864. PMID: 16630945. cancer in ataxia telangiectasia; substantial protective effect of residual
Nyboe Andersen N, Pasternak B, Basit S, et  al. 2014. Association between ATM kinase activity against childhood tumours. Br J Cancer, 105(4),
tumor necrosis factor-​alpha antagonists and risk of cancer in patients with 586–​591. PMCID: PMC3170966.
inflammatory bowel disease. JAMA, 311(23), 2406–​2413. Resnick ES, Moshier EL, Godbold JH, and Cunningham-​Rundles C. 2012.
O’Donovan P, Perrett CM, Zhang X, et al. 2005. Azathioprine and UVA light Morbidity and mortality in common variable immune deficiency over 4
generate mutagenic oxidative DNA damage. Science, 309(5742), 1871–​ decades. Blood, 119(7), 1650–​1657. PMCID: PMC3286343.
1874. PMCID: PMC2426755. Rizzo JD, Curtis RE, Socie G, et  al. 2009. Solid cancers after alloge-
Ochs HD, Smith C, and Puck JM. 2014. Primary Immunodeficiency Diseases. neic hematopoietic cell transplantation. Blood, 113(5), 1175–​ 1183.
3rd ed. Oxford: Oxford University Press. PMCID: PMC2635083.
Olivero OA. 2007. Mechanisms of genotoxicity of nucleoside reverse Robbins HA, Clarke CA, Arron ST, et  al. 2015a. Melanoma risk and survival
transcriptase inhibitors. Environ Mol Mutagen, 48(3–​4), 215–​223. among organ transplant recipients. J Invest Dermatol, 135(11), 2657–​2665.
PMID: 16395695. PMCID: PMC4640996.
Olson SH, Hsu M, Satagopan JM, et  al. 2013. Allergies and risk of pancre- Robbins HA, Pfeiffer RM, Shiels MS, et  al. 2015b. Excess cancers among
atic cancer: a pooled analysis from the Pancreatic Cancer Case-​Control HIV-​infected people in the United States. J Natl Cancer Inst, 107(4).
Consortium. Am J Epidemiol, 178(5), 691–​700. PMCID: PMC3755648. PMCID: PMC4334816.
Opelz G, Daniel V, Naujokat C, and Dohler B. 2009. Epidemiology of pretrans- Robbins HA, Shiels MS, Pfeiffer RM, and Engels EA. 2014. Epidemiologic
plant EBV and CMV serostatus in relation to posttransplant non-​Hodgkin contributions to recent cancer trends among HIV-​infected people in the
lymphoma. Transplantation, 88(8), 962–​967. PMID: 19855238. United States. AIDS, 28(6), 881–​890. PMID: 24300545.
Opelz G, and Dohler B. 2004. Lymphomas after solid organ transplantation: a Rooney CM, Smith CA, Ng CYC, et al. 1995. Use of gene-​modified virus-​spe-
collaborative transplant study report. Am J Transplant, 4(2), 222–​230. cific T-​lymphocytes to control Epstein-​Barr-​virus-​related lymphoprolif-
PMID: 14974943. eration. Lancet, 345(8941), 9–​13. PMID: 7799740
Orange JS, Glessner JT, Resnick E, et al. 2011. Genome-​wide association iden- Rosenberg PS, Alter BP, Bolyard AA, et al. 2006. The incidence of leukemia
tifies diverse causes of common variable immunodeficiency. J Allergy and mortality from sepsis in patients with severe congenital neutrope-
Clin Immunol, 127(6), 1360–​1367 e1366. PMCID: PMC3646656. nia receiving long-​ term G-​ CSF therapy. Blood, 107(12), 4628–​ 4635.
Orr N, Lemnrau A, Cooke R et al. 2012. Genome-​wide association study iden- PMCID: PMC1895804.
tifies a common variant in RAD51B associated with male breast cancer Rosenberg SA, and Restifo NP. 2015. Adoptive cell transfer as personal-
risk. Nat Genet, 44(11), 1182–​1184. PMCID: PMC3722904. ized immunotherapy for human cancer. Science, 348(6230), 62–​ 68.
Ose J, Schock H, Tjonneland A, et al. 2015. Inflammatory markers and risk of PMID: 25838374.
epithelial ovarian cancer by tumor subtypes: The EPIC Cohort. Cancer Rosenberger A, Bickeboller H, McCormack V, et al. 2012. Asthma and lung
Epidemiol Biomarkers Prev, 24(6), 951–​961. PMCID: PMC4454588. cancer risk: a systematic investigation by the International Lung Cancer
Palella FJ, Jr, Delaney KM, Moorman AC, et al. 1998. Declining morbidity and mor- Consortium. Carcinogenesis, 33(3), 587–​597. PMCID: PMC3291861.
tality among patients with advanced human immunodeficiency virus infec- Rostom A, Dube C, Lewin G, et al. 2007. Nonsteroidal anti-​inflammatory drugs
tion. HIV Outpatient Study Investigators. N Engl J Med, 338(13), 853–​860. and cyclooxygenase-​2 inhibitors for primary prevention of colorectal can-
Paul W. 2013. Fundamental Immunology. 7th ed. Philadelphia:  Lipincott cer: a systematic review prepared for the U.S. Preventive Services Task
Williams and Wilkins. Force. Ann Intern Med, 146(5), 376–​389. PMID: 17339623.
Penn I. 1997. Transmission of cancer from organ donors. Ann Transplant, 2(4), Rothman N, Garcia-​Closas M, Chatterjee N et al. 2010. A multi-​stage genome-​
7–​12. PMID: 9869872. wide association study of bladder cancer identifies multiple susceptibility
Perez-​Callejo D, Gonzalez-​Rincon J, Sanchez A, Provencio M, and Sanchez-​ loci. Nat Genet, 42(11), 978–​984. PMCID: PMC3049891.
Beato M. 2015. Action and resistance of monoclonal CD20 antibodies Rothwell PM, Price JF, Fowkes FG, et  al. 2012. Short-​term effects of daily
therapy in B-​cell non-​Hodgkin lymphomas. Cancer Treat Rev, 41(8), aspirin on cancer incidence, mortality, and non-​vascular death: analysis
680–​689. PMID: 26045227. of the time course of risks and benefits in 51 randomised controlled trials.
Peters U, Hutter CM, Hsu L, et al. 2012. Meta-​analysis of new genome-​wide Lancet, 379(9826), 1602–​1612. PMID: 22440946.
association studies of colorectal cancer risk. Hum Genet, 131(2), 217–​ Rowlings PA, Curtis RE, Passweg JR, et  al. 1999. Increased incidence of
234. PMCID: PMC3257356. Hodgkin’s disease after allogeneic bone marrow transplantation. J Clin
Peters U, Jiao S, Schumacher FR, et  al. 2013. Identification of genetic sus- Oncol, 17(10), 3122–​3127. PMID: 10506608.
ceptibility loci for colorectal tumors in a genome-​wide meta-​analysis. Rubicz R, Yolken R, Drigalenko E, et  al. 2013. A genome-​wide integrative
Gastroenterology, 144(4), 799–​807, e724. PMCID: PMC3636812. genomic study localizes genetic factors influencing antibodies against
Pharoah PD, Tsai YY, Ramus SJ, et al. 2013. GWAS meta-​analysis and replica- Epstein-​ Barr virus nuclear antigen 1 (EBNA-​ 1). PLoS Genet, 9(1),
tion identifies three new susceptibility loci for ovarian cancer. Nat Genet, e1003147. PMCID: PMC3542101.
45(4), 362–​370, 370e361–​362. PMCID: PMC3693183. Safaeian M, Robbins HA, Berndt SI, et al. 2015. Risk of colorectal cancer after
Poole EM, Lee IM, Ridker PM, et al. 2013. A prospective study of circulating solid organ transplantation in the United States. Am J Transplant, 16(3),
C-​reactive protein, interleukin-​6, and tumor necrosis factor alpha receptor 960-​967. PMID: 26731613
2 levels and risk of ovarian cancer. Am J Epidemiol, 178(8), 1256–​1264. Sahasrabuddhe VV, Shiels MS, McGlynn KA, and Engels EA. 2012. The risk
PMCID: PMC3792725. of hepatocellular carcinoma among individuals with acquired immuno-
Preiksaitis JK, and Keay S. 2001. Diagnosis and management of posttransplant deficiency syndrome in the United States. Cancer, 118(24), 6226–​6233.
lymphoproliferative disorder in solid-​organ transplant recipients. Clin PMCID: PMC3965349.
Infect Dis, 33 Suppl 1, S38–​46. PMID: 11389521. Santillan AA, Camargo CA, Jr., and Colditz GA. 2003. A meta-​analysis of
Prendergast AJ, Klenerman P, and Goulder PJ. 2012. The impact of differential asthma and risk of lung cancer (United States). Cancer Causes Control,
antiviral immunity in children and adults. Nat Rev Immunol, 12(9), 636–​ 14(4), 327–​334. PMID: 12846363.
648. PMID: 22918466. Sasazuki S, Inoue M, Sawada N, et  al. 2010. Plasma levels of C-​ reac-
Purdue MP, Hofmann JN, Kemp TJ, et  al. 2013. A prospective study of 67 tive protein and serum amyloid A  and gastric cancer in a nested case-​
serum immune and inflammation markers and risk of non-​Hodgkin lym- control study:  Japan Public Health Center-​ based prospective study.
phoma. Blood, 122(6), 951–​957. PMCID: PMC3739038. Carcinogenesis, 31(4), 712–​718. PMID: 20083644.
Purdue MP, Lan Q, Kemp TJ, et al. 2015. Elevated serum sCD23 and sCD30 up Sauce D, and Appay V. 2011. Altered thymic activity in early life: how does it affect
to two decades prior to diagnosis associated with increased risk of non-​ the immune system in young adults? Curr Opin Immunol, 23(4), 543–​548.
Hodgkin lymphoma. Leukemia, 29(6), 1429–​1431. PMID: 25567136. Scheurer ME, Amirian ES, Davlin SL, et al. 2011. Effects of antihistamine and
Purrington KS, Slager S, Eccles D et al. 2014. Genome-​wide association study anti-​inflammatory medication use on risk of specific glioma histologies.
identifies 25 known breast cancer susceptibility loci as risk factors for tri- Int J Cancer, 129(9), 2290–​2296. PMCID: PMC3125483.
ple-​negative breast cancer. Carcinogenesis, 35(5), 1012–​1019. PMCID: Schlehofer B, Siegmund B, Linseisen J, et  al. 2011. Primary brain tumours
PMC4004200. and specific serum immunoglobulin E: a case-​control study nested in the
Quinti I, Soresina A, Spadaro G, et al. 2007. Long-​term follow-​up and outcome European Prospective Investigation into Cancer and Nutrition cohort.
of a large cohort of patients with common variable immunodeficiency. Allergy, 66(11), 1434–​1441. PMID: 21726235.
J Clin Immunol, 27(3), 308–​316. PMID: 17510807. Schoof N, Iles MM, Bishop DT, et  al. 2011. Pathway-​ based analysis
Ramesh M, Hamm D, Simchoni N, and Cunningham-​Rundles C. 2015. Clonal of a melanoma genome-​ wide association study:  analysis of genes
and constricted T cell repertoire in common variable immune deficiency. related to tumour-​ immunosuppression. PLoS One, 6(12), e29451.
Clin Immunol, S1521-​6616(15)00004-​2. PMCID: PMC4501913. PMCID: PMC3246481.
48

484 PART III:  THE CAUSES OF CANCER


Schumacher FR, Berndt SI, Siddiq A, et  al. 2011. Genome-​wide association Skibola CF, Conde L, Foo JN, et  al. 2012. A meta-​analysis of genome-​wide
study identifies new prostate cancer susceptibility loci. Hum Mol Genet, association studies of follicular lymphoma. BMC Genomics, 13, 516.
20(19), 3867–​3875. PMCID: PMC3168287. PMCID: PMC3534234.
Schumacher TN, and Schreiber RD. 2015. Neoantigens in cancer immunother- Smedby KE, Foo JN, Skibola CF, et al. 2011. GWAS of follicular lymphoma
apy. Science, 348(6230), 69–​74. PMID: 25838375. reveals allelic heterogeneity at 6p21.32 and suggests shared genetic
Schuster V, and Kreth HW. 2000. X-​linked lymphoproliferative disease is susceptibility with diffuse large B-​cell lymphoma. PLoS Genet, 7(4),
caused by deficiency of a novel SH2 domain-​containing signal transduc- e1001378. PMCID: PMC3080853.
tion adaptor protein. Immunol Rev, 178, 21–​28. PMID: 11213803 Song M, Wu K, Ogino S, et al. 2013. A prospective study of plasma inflamma-
Schuster V, and Latour S. 2014. X-​Linked Lymphoproliferative Diseases Primary tory markers and risk of colorectal cancer in men. Br J Cancer, 108(9),
Immunodeficiency Diseases (pp. 557–​579). Oxford: Oxford University Press. 1891–​1898. PMCID: PMC3658520.
Schwartzbaum J, Ding B, Johannesen TB, et  al. 2012. Association between Stallone G, Schena A, Infante B, et al. 2005. Sirolimus for Kaposi’s sarcoma
prediagnostic IgE levels and risk of glioma. J Natl Cancer Inst, 104(16), in renal-​transplant recipients. N Engl J Med, 352(13), 1317–​ 1323.
1251–​1259. PMCID: PMC3424222. PMID: 15800227.
Shaw AC, Goldstein DR, and Montgomery RR. 2013. Age-​dependent dys- Straus SE, Jaffe ES, Puck JM, et al. 2001. The development of lymphomas in
regulation of innate immunity. Nat Rev Immunol, 13(12), 875–​ 887. families with autoimmune lymphoproliferative syndrome with germline
PMCID: PMC4096436. Fas mutations and defective lymphocyte apoptosis. Blood, 98(1), 194–​
Shi YY, Hu ZB, Wu C, et al. 2011. A genome-​wide association study identi- 200. PMID: 11418480.
fies new susceptibility loci for non-​cardia gastric cancer at 3q13.31 and Strauss DC, and Thomas JM. 2010. Transmission of donor melanoma by organ
5p13.1. Nat Genet, 43(12), 1215–​U1266. transplantation. Lancet Oncol, 11(8), 790–​796. PMID: 20451456.
Shiels MS, Cole SR, Kirk GD, and Poole C. 2009. A meta-​analysis of the inci- Study C, Houlston RS, Webb E, et  al. 2008. Meta-​analysis of genome-​wide
dence of non-​AIDS cancers in HIV-​infected individuals. J Acquir Immune association data identifies four new susceptibility loci for colorectal can-
Defic Syndr, 52(5), 611–​622. PMCID: PMC2790038. cer. Nat Genet, 40(12), 1426–​1435. PMCID: PMC2836775.
Shiels MS, Cole SR, Mehta SH, and Kirk GD. 2010a. Lung cancer inci- Su WH, Hildesheim A, and Chang YS. 2013. Human leukocyte antigens and
dence and mortality among HIV-​infected and HIV-​uninfected injec- epstein-​barr virus-​associated nasopharyngeal carcinoma: old associations
tion drug users. J Acquir Immune Defic Syndr, 55(4), 510–​ 515. offer new clues into the role of immunity in infection-​associated cancers.
PMCID: PMC2974802. Front Oncol, 3, 299.
Shiels MS, Copeland G, Goodman MT, et al. 2015a. Cancer stage at diagnosis Suarez F, Mahlaoui N, Canioni D et al. 2015. Incidence, presentation, and prog-
in patients infected with the human immunodeficiency virus and trans- nosis of malignancies in ataxia-​telangiectasia: a report from the French
plant recipients. Cancer, 121(12), 2063–​2071. PMCID: PMC4470321. national registry of primary immune deficiencies. J Clin Oncol, 33(2),
Shiels MS, Engels EA, Shi J, et al. 2012a. Genetic variation in innate immu- 202–​208. PMID: 25488969.
nity and inflammation pathways associated with lung cancer risk. Cancer, Sullivan KE, Mullen CA, Blaese RM, and Winkelstein JA. 1994. A multiinsti-
118(22), 5630–​5636. PMCID: PMC3485420. tutional survey of the Wiskott-​Aldrich syndrome. J Pediatr, 125(6 Pt 1),
Shiels MS, Goedert JJ, Moore RD, Platz EA, and Engels EA. 2010b. Reduced 876–​885. PMID: 7996359.
risk of prostate cancer in U.S.  men with AIDS. Cancer Epidemiol Sumegi J, Huang D, Lanyi A, et  al. 2000. Correlation of mutations of the
Biomarkers Prev, 19(11), 2910–​2915. PMCID: PMC2976800. SH2D1A gene and Epstein-​Barr virus infection with clinical phenotype
Shiels MS, Katki HA, Hildesheim A, et  al. 2014. Circulating inflammation and outcome in X-​ linked lymphoproliferative disease. Blood, 96(9),
markers, risk of lung cancer, and utility for risk stratification. JNCI, 3118–​3125. PMID: 11049992.
107(10), djv199 Sun J, Zheng SL, Wiklund F, et  al. 2009. Sequence variants at 22q13 are
Shiels MS, Pfeiffer RM, Chaturvedi AK, Kreimer AR, and Engels EA. associated with prostate cancer risk. Cancer Res, 69(1), 10–​ 15.
2012b. Impact of the HIV epidemic on the incidence rates of anal can- PMCID: PMC2705898.
cer in the United States. J Natl Cancer Inst, 104(20), 1591–​ 1598. Swerdlow SH, Webber SA, Chadburn A, and Ferry JA. 2008. WHO classifica-
PMCID: PMC3611819. tion of tumours of hematopoietic and lymphoid tissues. In: Swerdlow SH,
Shiels MS, Pfeiffer RM, and Engels EA. 2010c. Age at cancer diagnosis among Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J, and Vardiman
persons with AIDS in the United States. Ann Intern Med, 153(7), 452–​ JW (Eds.), (pp. 343–​349). Lyon: IARC Press.
460. PMCID: PMC3071616. Tan IJ, Peeva E, and Zandman-​Goddard G. 2015. Hormonal modulation of the
Shiels MS, Pfeiffer RM, Gail MH, et  al. 2011. Cancer burden in the HIV-​ immune system: a spotlight on the role of progestogens. Autoimmun Rev,
infected population in the United States. J Natl Cancer Inst, 103(9), 753–​ 14(6), 536–​542. PMID: 25697984.
762. PMCID: PMC3086877. Tang M, Lautenberger JA, Gao X, et  al. 2012. The principal genetic deter-
Shiels MS, Pfeiffer RM, Hall HI, et al. 2011. Proportions of Kaposi sarcoma, minants for nasopharyngeal carcinoma in China involve the HLA
selected non-​Hodgkin lymphomas, and cervical cancer in the United class  I  antigen recognition groove. PLoS Genet, 8(11), e1003103.
States occurring in persons with AIDS, 1980–​2007. JAMA, 305(14), PMCID: PMC3510037.
1450–​1459. Tanon A, Jaquet A, Ekouevi DK, et al. 2012. The spectrum of cancers in West
Shiels MS, Pfeiffer RM, Hildesheim A, et al. 2013. Circulating inflammation Africa:  associations with human immunodeficiency virus. PLoS One,
markers and prospective risk for lung cancer. J Natl Cancer Inst, 105(24), 7(10), e48108. PMCID: PMC3483170.
1871–​1880. PMCID: PMC3888091. Tao S, Wang Z, Feng J, et  al. 2012. A genome-​wide search for loci inter-
Shiels MS, Shu X, Chaturvedi A, et al. (2015b). A prospective study of immune acting with known prostate cancer risk-​ associated genetic variants.
and inflammation markers and risk of lung cancer among never smoking Carcinogenesis, 33(3), 598–​603. PMCID: PMC3291863.
women in Shanghai. Tenesa A, Farrington SM, Prendergast JG, et  al. 2008. Genome-​wide asso-
Shin CR, Kim MO, Li D et al. 2012. Outcomes following hematopoietic cell ciation scan identifies a colorectal cancer susceptibility locus on 11q23
transplantation for Wiskott-​Aldrich syndrome. Bone Marrow Transplant, and replicates risk loci at 8q24 and 18q21. Nat Genet, 40(5), 631–​637.
47(11), 1428–​1435. PMID: 22426750. PMCID: PMC2778004.
Siddiq A, Couch FJ, Chen GK et al. 2012. A meta-​analysis of genome-​wide Thomas G, Jacobs KB, Kraft P, et  al. 2009. A multistage genome-​ wide
association studies of breast cancer identifies two novel suscepti- association study in breast cancer identifies two new risk alleles
bility loci at 6q14 and 20q11. Hum Mol Genet, 21(24), 5373–​5384. at 1p11.2 and 14q24.1 (RAD51L1). Nat Genet, 41(5), 579–​ 584.
PMCID: PMC3510753. PMCID: PMC2928646.
Sigel K, Wisnivesky J, Gordon K, et al. 2012. HIV as an independent risk factor Thun MJ, Jacobs EJ, and Patrono C. 2012. The role of aspirin in cancer preven-
for incident lung cancer. AIDS, 26(8), 1017–​1025. PMCID: PMC3580210. tion. Nat Rev Clin Oncol, 9(5), 259–​267. PMID: 22473097.
Silverberg MJ, Leyden W, Warton EM, et al. 2013. HIV infection status, immu- Tomlinson IP, Webb E, Carvajal-​Carmona L, et  al. 2008. A genome-​wide
nodeficiency, and the incidence of non-​melanoma skin cancer. J Natl association study identifies colorectal cancer susceptibility loci
Cancer Inst, 105(5), 350–​360. PMCID: PMC3589255. on chromosomes 10p14 and 8q23.3. Nat Genet, 40(5), 623–​ 630.
Simard EP, Shiels MS, Bhatia K, and Engels EA. 2012. Long-​term cancer risk PMID: 18372905.
among people diagnosed with AIDS during childhood. Cancer Epidemiol Topalian SL, Hodi FS, Brahmer JR, et al. 2012. Safety, activity, and immune
Biomarkers Prev, 21(1), 148–​154. PMCID: PMC3312034. correlates of anti-​PD-​1 antibody in cancer. N Engl J Med, 366(26), 2443–​
Sipponen P, and Maaroos HI. 2015. Chronic gastritis. Scand J Gastroenterol, 2454. PMCID: PMC3544539.
50(6), 657–​667. PMCID: PMC4673514. Toriola AT, Cheng TY, Neuhouser ML, et al. 2013. Biomarkers of inflammation
Sitas F, Pacella-​Norman R, Carrara H, et al. 2000. The spectrum of HIV-​1 related are associated with colorectal cancer risk in women but are not suitable as
cancers in South Africa. Int J Cancer, 88(3), 489–​492. PMID: 11054682. early detection markers. Int J Cancer, 132(11), 2648–​2658.
 485

Immunologic Factors 485


Trabert B, Pinto L, Hartge P, et al. 2014. Pre-​diagnostic serum levels of inflam- Wang SS, Menashe I, Cerhan JR, et  al. 2011. Variations in chromosomes 9
mation markers and risk of ovarian cancer in the prostate, lung, colorectal and 6p21.3 with risk of non-​Hodgkin lymphoma. Cancer Epidemiol
and ovarian cancer (PLCO) screening trial. Gynecol Oncol, 135(2), 297–​ Biomarkers Prev, 20(1), 42–​49. PMCID: PMC3817834.
304. PMCID: PMC4254357. Wang T, Rohan TE, Gunter MJ et  al. 2011. A prospective study of inflam-
Trigunaite A, Dimo J, and Jorgensen TN. 2015. Suppressive effects of mation markers and endometrial cancer risk in postmenopausal hor-
androgens on the immune system. Cell Immunol, 294(2), 87–​ 94. mone nonusers. Cancer Epidemiol Biomarkers Prev, 20(5), 971–​977.
PMID: 25708485. PMCID: PMC3096873.
Tsilidis KK, Branchini C, Guallar E, et al. 2008. C-​reactive protein and colorec- Wegner R, German J, Chrzanowska KH, Digweed M, and Stumm M. 2014.
tal cancer risk: a systematic review of prospective studies. Int J Cancer, Chromosomal Instability Syndromes Other Than Ataxia-​Telangiectasia
123(5), 1133–​1140. PMID: 18528865. Primary Immunodeficiency Diseases (pp. 632–​ 661). Oxford:  Oxford
Turnbull C, Ahmed S, Morrison J, et al. 2010a. Genome-​wide association study University Press.
identifies five new breast cancer susceptibility loci. Nat Genet, 42(6), Whitacre CC. 2001. Sex differences in autoimmune disease. Nat Immunol,
504–​507. PMCID: PMC3632836. 2(9), 777–​780. PMID: 11526384.
Turnbull C, Rapley EA, Seal S, et al. 2010b. Variants near DMRT1, TERT and Wills-​Karp M, Santeliz J, and Karp CL. 2001. The germless theory of allergic
ATF7IP are associated with testicular germ cell cancer. Nat Genet, 42(7), disease: revisiting the hygiene hypothesis. Nat Rev Immunol, 1(1), 69–​75.
604–​607. PMCID: PMC3773909. PMID: 11905816.
Turner MC, Chen Y, Krewski D, and Ghadirian P. 2006. An overview of the Winkelstein JA, Marino MC, Ochs H, et  al. 2003. The X-​linked hyper-​IgM
association between allergy and cancer. Int J Cancer, 118(12), 3124–​ syndrome:  clinical and immunologic features of 79 patients. Medicine
3132. PMID: 16395696. (Baltimore), 82(6), 373–​384. PMID: 14663287.
Turner MC, Chen Y, Krewski D, et al. 2005. Cancer mortality among US men Wrensch M, Wiencke JK, Wiemels J, et al. 2006. Serum IgE, tumor epidermal
and women with asthma and hay fever. Am J Epidemiol, 162(3), 212–​221. growth factor receptor expression, and inherited polymorphisms associated
PMID: 15987724. with glioma survival. Cancer Res, 66(8), 4531–​4541. PMID: 16618782.
Urayama KY, Jarrett RF, Hjalgrim H, et  al. 2012. Genome-​wide associa- Wu C, Hu Z, He Z, et  al. 2011. Genome-​wide association study identifies
tion study of classical Hodgkin lymphoma and Epstein-​Barr virus sta- three new susceptibility loci for esophageal squamous-​cell carcinoma in
tus-​defined subgroups. J Natl Cancer Inst, 104(3), 240–​253. PMCID: Chinese populations. Nat Genet, 43(7), 679–​684. PMID: 21642993.
PMC3274508. Wu C, Kraft P, Zhai K, et  al. 2012a. Genome-​wide association analyses of
Urayama KY, Thompson PD, Taylor M, Trachtenberg EA, and Chokkalingam esophageal squamous cell carcinoma in Chinese identify multiple sus-
AP. 2013. Genetic variation in the extended major histocompatibility ceptibility loci and gene-​environment interactions. Nat Genet, 44(10),
complex and susceptibility to childhood acute lymphoblastic leukemia: a 1090–​1097. PMCID: PMC3513832.
review of the evidence. Front Oncol, 3, 300. PMCID: PMC3859964. Wu C, Miao X, Huang L, et al. 2012b. Genome-​wide association study identi-
Vajdic CM, McDonald SP, McCredie MR, et  al. 2006. Cancer incidence fies five loci associated with susceptibility to pancreatic cancer in Chinese
before and after kidney transplantation. JAMA, 296(23), 2823–​ 2831. populations. Nat Genet, 44(1), 62–​66. PMID: 22158540.
PMID: 17179459. Wu C, Wang Z, Song X, et al. 2014. Joint analysis of three genome-​wide asso-
van Leeuwen MT, Grulich AE, Webster AC, et al. 2009. Immunosuppression ciation studies of esophageal squamous cell carcinoma in Chinese popu-
and other risk factors for early and late non-​Hodgkin lymphoma after lations. Nat Genet, 46(9), 1001–​1006. PMCID: PMC4212832.
kidney transplantation. Blood, 114(3), 630–​637. PMID: 19443660. Xin YN, Lin ZH, Jiang XJ, et al. 2011. Specific HLA-​DQB1 alleles associated
Vendrame E, Hussain SK, Breen EC, et al. 2014. Serum levels of cytokines and with risk for development of hepatocellular carcinoma: a meta-​analysis.
biomarkers for inflammation and immune activation, and HIV-​associated World J Gastroenterol, 17(17), 2248–​2254. PMCID: PMC3092879.
non-​Hodgkin B-​cell lymphoma risk. Cancer Epidemiol Biomarkers Prev, Yanik EL, Siddiqui K, and Engels EA. 2015. Sirolimus effects on cancer inci-
23(2), 343–​349. PMCID: PMC3948172. dence after kidney transplantation:  a meta-​analysis. Cancer Med, 4(9),
Vesterinen E, Pukkala E, Timonen T, and Aromaa A. 1993. Cancer incidence 1448–​1459. PMCID: PMC4567030.
among 78,000 asthmatic-​ patients. Int J Epidemiol, 22(6), 976–​ 982. Yel L, Lavin MF, and Shiloh Y. 2014. Ataxia-​Telangiectasia Primary
PMID: 8144310. Immunodeficiency Diseases (pp. 602–​631). Oxford: Oxford University Press.
Vignozzi L, and Maggi M. 2014. Prostate cancer: intriguing data on inflamma- Yoon KA, Park JH, Han J, et al. 2010. A genome-​wide association study reveals
tion and prostate cancer. Nat Rev Urol, 11(7), 369–​370. PMID: 24980193. susceptibility variants for non-​small cell lung cancer in the Korean popu-
Vijai J, Kirchhoff T, Schrader KA, et  al. 2013. Susceptibility loci associated lation. Hum Mol Genet, 19(24), 4948–​4954. PMID: 20876614.
with specific and shared subtypes of lymphoid malignancies. PLoS Genet, Zhang B, Jia WH, Matsuda K, et al. 2014a. Large-​scale genetic study in East
9(1), e1003220. PMCID: PMC3547842. Asians identifies six new loci associated with colorectal cancer risk. Nat
Vijai J, Wang Z, Berndt SI, et al. 2015. A genome-​wide association study of Genet, 46(6), 533–​542. PMCID: PMC4068797.
marginal zone lymphoma shows association to the HLA region. Nat Zhang B, Jia WH, Matsuo K, et al. 2014b. Genome-​wide association study iden-
Commun, 6, 5751. PMCID: PMC4287989. tifies a new SMAD7 risk variant associated with colorectal cancer risk in
Villeneuve PJ, Schaubel DE, Fenton SS, et al. 2007. Cancer incidence among East Asians. Int J Cancer, 135(4), 948–​955. PMCID: PMC4051826.
Canadian kidney transplant recipients. Am J Transplant, 7(4), 941–​948. Zhang M, Song F, Liang L, et al. 2013. Genome-​wide association studies iden-
PMID: 17331115. tify several new loci associated with pigmentation traits and skin can-
Visco C, Barcellini W, Maura F, et al. 2014. Autoimmune cytopenias in chronic cer risk in European Americans. Hum Mol Genet, 22(14), 2948–​2959.
lymphocytic leukemia. Am J Hematol, 89(11), 1055–​1062. PMCID: PMC3690971.
Wabinga HR, Parkin DM, Wabwire-​Mangen F, and Nambooze S. 2000. Trends Zhang R, Chu M, Zhao Y, et  al. 2014. A genome-​wide gene-​environment
in cancer incidence in Kyadondo County, Uganda, 1960–​1997. Br J interaction analysis for tobacco smoke and lung cancer susceptibility.
Cancer, 82(9), 1585–​1592. PMCID: PMC2363394. Carcinogenesis, 35(7), 1528–​1535. PMCID: PMC4076813.
Wang LE, Gorlova OY, Ying J, et  al. 2013. Genome-​wide association study Zhao H, Cai W, Su S, et al. 2014. Allergic conditions reduce the risk of gli-
reveals novel genetic determinants of DNA repair capacity in lung cancer. oma:  a meta-​analysis based on 128,936 subjects. Tumour Biol, 35(4),
Cancer Res, 73(1), 256–​264. PMCID: PMC3537906. 3875–​3880. PMID: 24347487.
Wang M, Chen C, Qu J, et al. 2011. Inverse association between eczema and Zheng W, Long J, Gao YT, et al. 2009. Genome-​wide association study identi-
meningioma:  a meta-​analysis. Cancer Causes Control, 22(10), 1355–​ fies a new breast cancer susceptibility locus at 6q25.1. Nat Genet, 41(3),
1363. PMID: 21710191. 324–​328. PMCID: PMC2754845.
Wang SS, Abdou AM, Morton LM, et  al. 2010a. Human leukocyte antigen Zhou B, Liu J, Wang ZM, and Xi T. 2012. C-​ reactive protein, interleu-
class I and II alleles in non-​Hodgkin lymphoma etiology. Blood, 115(23), kin 6 and lung cancer risk:  a meta-​analysis. PLoS One, 7(8), e43075.
4820–​4823. PMCID: PMC2890176. PMCID: PMC3422305.
Wang SS, Gonzalez P, Yu K et al. 2010b. Common genetic variants and risk Zutter MM, Martin PJ, Sale GE, et  al. 1988. Epstein-​Barr virus lymphopro-
for HPV persistence and progression to cervical cancer. PLoS One, 5(1), liferation after bone marrow transplantation. Blood, 72(2), 520–​ 529.
e8667. PMCID: PMC2801608. PMID: 2840986.
486
 487

IV
CANCERS BY TISSUE OF ORIGIN
 489

26 Nasopharyngeal Cancer

ELLEN T. CHANG AND ALLAN HILDESHEIM

OVERVIEW TUMOR CLASSIFICATION

The main epithelial malignancy arising in the nasopharynx is NPC is identified using ICD-​O-​3 site codes C110–​C119. Three main
nasopharyngeal carcinoma (NPC). Although rare throughout most histologic subtypes are currently recognized by the World Health
of the world, NPC has a unique geographic distribution, with high-​ Organization (WHO):  non-​keratinizing carcinoma (differentiated or
risk endemic areas in southern China and parts of Southeast Asia, undifferentiated); keratinizing squamous cell carcinoma; and basaloid
intermediate incidence rates elsewhere in Southeast Asia, North squamous cell carcinoma. Non-​keratinizing carcinoma is designated
Africa, the Arctic, the Middle East, and among Asian and Pacific by the ICD-​O-​3 histology code 8072/​3; keratinizing squamous cell
Islander migrants, and very low risk in other areas. The great carcinoma as 8071/​3; and basaloid carcinoma as code 8083/​3. The lat-
majority of NPC cases worldwide are non-​keratinizing tumors that ter morphologically resembles squamous cell carcinomas that occur
predominate in endemic, high-​incidence areas and comprise up to commonly elsewhere in the head and neck region (Chan et al., 2005).
half of tumors in low-​incidence populations. The other histologic Other rare malignant epithelial tumors that arise in the nasopharynx,
subtype of NPC, keratinizing squamous cell carcinoma, accounts including adenocarcinoma and salivary-​gland-​type carcinoma, are not
for 40%–​ 50% of cases in low-​ incidence areas. Infection with considered in this chapter.
Epstein-​Barr virus (EBV) is a necessary but not sufficient cause Histologic classification of NPC has changed relatively little since
of non-​keratinizing NPC, but it is more weakly associated with 1978, when WHO characterized the three histologic subtypes of NPC
keratinizing squamous cell tumors. Tobacco smoking increases as squamous cell carcinoma (WHO type 1), non-​keratinizing carci-
the risk of both subtypes. Other established cofactors for non-​ noma (WHO type 2), and undifferentiated carcinoma (WHO type
keratinizing NPC include dietary consumption of salt-​preserved 3)  (Shanmugaratnam and Sobin, 1978). A  1991 reclassification by
fish and other preserved foods, and germline polymorphisms in WHO retained the category of squamous cell carcinoma and com-
the human leukocyte antigen (HLA) genes HLA-​A*02:07-​B*46:01 bined the other two subtypes into the single category of non-​keratin-
and HLA-​A*11:01, which affect proteins that present antigens to izing carcinoma; the latter was then subclassified as differentiated or
immune cells. Exposures in childhood are thought to be particu- undifferentiated (Shanmugaratnam, 1991). The most recent WHO
larly important in the etiology of non-​keratinizing NPC. In high-​ classification in 2005 added basaloid squamous cell carcinoma as
incidence areas, EBV infection is typically acquired in infancy or an additional, rare (< 0.2%) histologic subtype (Chan et  al., 2005).
early childhood, inherited polymorphisms in HLA may affect the Updated classification schemes that consider the molecular profile of
immune response to the virus, and in southern China, infants were tumors are currently being considered; these may provide more accu-
traditionally weaned on salt-​preserved fish with a high content rate predictions of prognosis (Wang et al., 2011).
of nitrosamines. Less is known about the etiology of keratinizing As noted earlier, the non-​keratinizing carcinoma subtype predom-
squamous cell NPC, or any histologic subtype of NPC in areas inates in high-​incidence areas. Previous infection with EBV can be
with low and intermediate incidence rates. demonstrated in essentially 100% of tumors by in situ hybridization
for EBV-​encoded early RNA tumors (Chan et al., 2005). The samples
that have been characterized histologically and tested for EBV are not
DISEASE BURDEN population-​based, yet they have been assumed to be representative
because of the distinctive geographic clustering. In contrast, keratin-
NPC is a relatively rare cancer in most areas of the world. While NPC izing squamous cell carcinoma can comprise up to 40%–​50% of NPC
is not the only malignancy that arises in the nasopharynx, it comprises tumors in low-​incidence areas such as the United States (Vaughan
the great majority of cases. Thus, data on incidence and mortality rates et al., 1996; Wang Y et al., 2013). The reported frequencies of NPC
for cancers located within the nasopharynx are generally considered histologic subtypes vary widely within low-​risk geographic regions.
to represent NPC. WHO has attributed this heterogeneity to a combination of unclear
Worldwide, approximately 86,700 newly diagnosed cases and boundaries between the histopathologic categories, sampling error due
50,800 deaths from NPC occur each year, making it the 24th most to the small size of biopsies, and suboptimal intra-​and inter-​observer
common cancer and the 23rd leading cause of cancer death in the reproducibility of classification (Chan et al., 2005).
world (Ferlay et al., 2013). The United States contributes minimally
to this international disease burden, with approximately 2,000 NPC
cases and 800 deaths identified annually. In the United States, NPC PRECANCEROUS OR PRECURSOR LESIONS
ranks 26th among cancers in terms of incidence and 27th in terms of
deaths. In situ NPC in the absence of invasive carcinoma is very rare (Chan
Over 80% of NPC cases and deaths that occur annually (70,100 et al., 2005). The scarcity of in situ cancers suggests either that invasive
and 41,400, respectively) are in Asia, while 10% of cases and 13% NPC does not typically develop from in situ cancer, or that progression
of deaths (8300 and 5500, respectively) occur in Africa (Ferlay et al., to invasive disease is so rapid that the in situ phase generally is not
2013). Even when averaged across all of Asia, NPC ranks only 22nd detected. Three small case series and one case report have described
in cancer incidence and 20th in cancer mortality. It ranks higher in patients with NPC in situ. These included 11 (Pathmanathan et  al.,
Southeast Asia, however, where NPC is the 10th most common cancer 1995), two (Cheung et  al., 1998), one (Pak et  al., 2005), and three
and cause of cancer death, accounting for approximately 25,600 new patients (Pak et al., 2002), all of whom were positive for EBV, indi-
diagnoses and 14,200 deaths each year. cating that EBV infection of the nasopharyngeal epithelium precedes

489
490

490 PART IV:  Cancers by Tissue of Origin


tumor invasiveness. Studies of tumor progression from in situ to inva- people and twice as high in Cantonese speakers as in the Hakka,
sive cancer have all been small. Two of three patients with untreated Hokkien, and Chiu Chau linguistic/​ethnic groups (Li et al., 1985).
NPC in situ and neither of two patients with radiotherapy-​treated NPC Across Southeast Asia, groups with a higher degree of racial and
in situ were reported as subsequently having developed invasive NPC social admixture with southern Chinese, and especially with the
(Cheung et al., 1998; Pak et al., 2005). ancestors of the Tanka called the Bai-​Yue (also called “proto-​Tai-​
Kadai,” “proto-​Austronesian,” or “proto-​Zhuang”), generally have
higher NPC risk (Wee et  al., 2010). This pattern is observed in
SURVIVAL Singapore, for example, where NPC incidence is highest among
Chinese, intermediate among Malays who have historically inter-
Tumor node metastasis (TNM) stage is the strongest established prog- married with Chinese, and lowest among Indians, who have rela-
nostic factor for NPC (Edge et al., 2010; Sobin et al., 2009). Survival tively little admixture with Chinese (Figure 26–2) (Forman et  al.,
rates vary considerably across centers. For example, at large oncology 2013; Ho, 1976). In the United States, NPC incidence is highest
centers in Hong Kong and China, recent 5-​year disease-​specific sur- among Chinese, followed by Filipinos (who have historical ties to
vival rates were 80%–​85% for all stages, 92%–​100% for stage I, 87%–​ southern Chinese), and substantially lower among Koreans, non-​
96% for stage II, 79%–​83% for stage III, and 65%–​70% for stage IV Hispanic whites, Japanese (who, like Koreans, have had traditional
(Lee et al., 2005; Sun et al., 2013). Other centers in Asia have reported ties mainly with northern China), and Hispanic whites (Figure 26–2)
5-​year overall survival rates of 60%–​90% for stage I, 57%–​79% for (Forman et al., 2013).
stage II, 44%–​77% for stage III, and 17%–​43% for stage IV (Dou No clear incidence patterns by county-​level socioeconomic status
et al., 2014; Leung et al., 2005; Phua et al., 2011, 2013). Some overlap are evident for NPC in the United States (SEER, 2015).
in the relative survival for stages I and IIA and a large gap between
stages IIA and IIB (Leung et al., 2005) suggest that stage IIA could
potentially be considered along with stage I as early-​stage NPC. In GEOGRAPHIC VARIATION
general, the stage distribution of NPC at diagnosis is skewed toward
more advanced stages due to the non-​specific symptoms and corre- The distinctive international pattern of incidence and mortality rates
spondingly late diagnosis of the disease. of NPC has been described earlier. The rates are highest in southern
Circulating plasma/​serum EBV DNA levels have been shown to China and parts of Southeast Asia, intermediate elsewhere in China
predict NPC survival (Chan et  al., 2002; Lin et  al., 2004; Lo et  al., and Southeast Asia, North Africa, the Arctic, and the Middle East,
2000; Wang WY et al., 2013). In particular, higher pre-​treatment and and lowest (below 1 per 100,000 person-​years) throughout the rest of
especially post-​treatment circulating EBV DNA levels are signifi- the world. Globally, NPC incidence rates vary by more than 200-​fold
cantly associated with poorer overall, disease-​specific, progression-​ between the highest-​and lowest-​incidence regions.
free, and relapse-​free survival. Population-​based cancer registry data show that in 2003–​2007, the
highest NPC incidence rates worldwide were recorded in Zhongshan
City, China, where the age-​standardized incidence rate (per 100,000)
DESCRIPTIVE EPIDEMIOLOGY was 26.8 in males (based on 799 cases) and 10.7 in females (based
on 326 cases) (Figures 26–2 and 26–3) (Forman et  al., 2013). The
Demographic Patterns next highest rates (per 100,000) were reported in Macau, China (15.7
In high-​incidence areas, the age-​specific incidence rates of NPC based on 246 cases among males; 6.7 based on 104 cases among
increase monotonically up to around age 45–​59 years, followed by a females), and Hong Kong, China (14.4 based on 3245 cases among
plateau or modest decline (Figures 26–1a and 26–1b) (Forman et al., males; 4.9 based on 1247 cases among females) (Figures 26–2 and
2013). This age-​related pattern is consistent with the role of early-​ 26–3). Intermediate rates (per 100,000) also prevail in both native
life exposure to EBV and other risk factors in the development of and migrant Asian populations and in the Arctic, North Africa, and
NPC in high-​incidence areas. In low-​incidence areas, the age-​specific the Middle East, generally ranging from 2–​6 among males and 1–​3
NPC incidence rates show a minor peak at around age 15–​19 years, among females (Figure 26–2).
especially in males, followed by a slight decline and then a mono-
tonic rise from around age 30–​34 to 65–​79 years (Figures 26–1c and
26–1d) (Bray et al., 2008; Forman et al., 2013). The early peak has TEMPORAL TRENDS
been documented in Australia, northern and western Europe, North
America, and low-​risk Asian countries (India and Japan), with some Reports from high-​risk areas, including Singapore, Taiwan, Hong
geographic variation in the timing of the peak and the duration of the Kong, and Guangzhou, have documented declines in NPC incidence
subsequent decline (Bray et  al., 2008). The bimodal pattern could and/​or mortality in recent years, although the rates in some areas
be consistent with exposures occurring at different times of life. The of southern China appear to have changed little over time (Deng
early peak may reflect inherited or other early-​life risk factors (per- et  al., 2014; Wei et  al., 2010). Specifically, among Chinese males
haps including EBV infection), whereas the later peak may reflect in Singapore, the annual age-​standardized incidence rate of NPC
long-​term adult environmental exposures. It has been hypothesized (per 100,000) fell from 18.7 in 1988–​1992 to 10.3 in 2003–​2007
that the bimodal age-​ incidence pattern occurs also in high-​ inci- (National Registry of Diseases Office, 2009, 2013). Between the late
dence regions, but that the early peak is obscured by genetic and 1970s and 2008, the age-​standardized incidence rate of NPC in Hong
epigenetic factors that are associated with increased NPC risk (Bray Kong decreased by 66.1% for males and by 73.7% for females (Lau
et al., 2008). et al., 2013), while in urban Guangzhou, China, the average annual
Across all geographic areas, regardless of background NPC rates, percentage change in the NPC incidence rate between 2000 and 2011
the age-​adjusted incidence rate of NPC is consistently around two to was –​3.26% for males and –​5.74% for females (Li et al., 2014). The
three times higher in males than in females, with even higher ratios decline in Taiwan has been smaller, with age-​standardized NPC inci-
in some areas (Figure 26–2) (Forman et  al., 2013). The male pre- dence rates (per 100,000) decreasing from 9.50 in males and 4.12
dominance of NPC is largely unexplained by established risk factors in females in 1980–​1984 to 8.59 in males and 2.88 in females in
(Edgren et al., 2012). 2000–​2006 (Chiang et  al., 2010). An analysis of national cancer
The distribution of NPC varies substantially by geographic mortality data in China found comparable declines in NPC mortality
region and race/​ethnicity, even within countries. For example, in in rural and urban areas between 1987–​1991 and 2007–​2009 (Guo
China a clear gradient runs from north to south, with the highest et al., 2012).
rates in southern China (Figures 26–2 and 26–3) (Forman et  al., In contrast, age-​standardized NPC incidence rates (per 100,000)
2013). Historically, in the high-​incidence southern Chinese prov- held steady at approximately 28 among males and 11 among females
ince of Guangdong, NPC occurrence was highest in the Tanka boat in Zhongshan, China, between 1970–​ 1974 and 2005–​ 2007 (Wei
 491

Nasopharyngeal Cancer 491
(a) 120 (b) 60
China, Hong Kong China, Hong Kong
China, Zhongshan City China, Zhongshan City

Incidence rate (per 100,000 person-years)


Incidence rate (per 100,000 person-years) Singapore Chinese Singapore Chinese
100 50

80 40

60 30

40 20

20 10

0 0
4
9

15 4
20 9
25 4
30 9
35 4
40 9
45 4
50 9
55 4
60 9
65 4
70 9
75 4
80 9
4
+

4
9

15 4
20 9
25 4
30 9
35 4
40 9
45 4
50 9
55 4
60 9
65 4
70 9
75 4
80 9
4
+
0–
5–

–1
–1
–2
–2
–3
–3
–4
–4
–5
–5
–6
–6
–7
–7
–8
85

0–
5–

–1
–1
–2
–2
–3
–3
–4
–4
–5
–5
–6
–6
–7
–7
–8
85
10

10
Age group (years) Age group (years)

(c) 2.5 (d) 1


USA whites USA whites
Scandinavia 0.9 Scandinavia
Incidence rate (per 100,000 person-years)

Japan

Incidence rate (per 100,000 person-years)


Japan
2 0.8

0.7
1.5 0.6

0.5
1
0.4

0.3
0.5
0.2

0.1
0
0
15 4
20 9
25 4
30 9
35 4
40 9
45 4
50 9
55 4
60 9
65 4
70 9
75 4
80 9
4
4
9
–1
–1
–2
–2
–3
–3
–4
–4
–5
–5
–6
–6
–7
–7
–8
0–
5–

4
9

15 4
20 9
25 4
30 9
35 4
40 9
45 4
50 9
55 4
60 9
65 4
70 9
75 4
80 9
4
10

0–
5–

–1
–1
–2
–2
–3
–3
–4
–4
–5
–5
–6
–6
–7
–7
–8
10

Age group (years) Age group (years)

Figure 26–1.  Age-specific incidence rates of nasopharyngeal carcinoma in high- and low-incidence areas, 2003–2007. (a) Incidence among males in
high-​incidence areas (Hong Kong, China; Zhongshan City, China; and Singapore Chinese).  (b) Incidence among females in high-​incidence areas.  (c)
Incidence among males in low-​incidence areas (United States: National Program of Cancer Registries, 42 states, whites; Scandinavia: Denmark,
Finland, Iceland, Norway, and Sweden; and Japan: Aichi Prefecture, Fukui Prefecture, Hiroshima, Miyagi Prefecture, Nagasaki Prefecture, Niigata
Prefecture, Osaka Prefecture, and Saga Prefecture).  (d) Incidence among females in low-​incidence areas.  Data from Cancer Incidence in Five
Continents, Vol. X (Forman et al., 2013).

et al., 2010), while reported NPC mortality rates (per 100,000) did Trends in incidence rates in intermediate-​ risk countries have
not change substantially between 1971–​1973 and 2004–​2005 among been mixed. In Algeria, the age-​standardized incidence rate (per
males (7.00 to 7.24, standardized to the 1982 China age structure) or 100,000) among males increased from 4.5 in 1986–​1990 to 7.3 in
females (2.96 to 2.54) in Guangxi, China (Deng et al., 2014). 1996–​2000 and declined thereafter to 4.8 in 2006–​2010. In contrast,
The observed decreases in NPC incidence in several high-​ risk the incidence rate among females was relatively stable at about 2
regions may be due in part to declining dietary consumption of salt-​ per 100,000 from 1986 through 2010 (Hamdi Cherif et  al., 2014).
preserved fish and increasing consumption of fresh fruits and vegeta- In intermediate-​risk Shanghai, the NPC incidence rate was stable at
bles, although reductions in the prevalence of tobacco smoking also roughly 3 per 100,000 between 1973 and 2005 among adolescent
may have contributed. The prevalence of salt-​preserved fish consump- and young-​adult males, but declined by 2% per year among females
tion in southern China and elsewhere has decreased in Southeast Asia, of the same age (from 2.1 to 1.1 per 100,000 between 1973–​1975
based on limited information derived from epidemiologic studies of and 2003–​2005) (Wu et al., 2012). Reports from low-​risk areas also
NPC. Up to 48% of adults and 47% of children reportedly consumed have been mixed, with the incidence in some countries reported to
salt-preserved fish in studies conducted in the 1980s and early 1990s. be stable (Lau et al., 2013; Xie et al., 2012) and others (including the
This has decreased to 4% among adults and 2% among children in US) declining (Lau et  al., 2013; SEER, 2015). Some authors have
studies conducted in the late 1990s and 2000s (IARC, 2012d). The reported a decrease in keratinizing but not non-​keratinizing NPC,
once-​common traditional practice of feeding salt-​preserved fish during possibly due to reductions in smoking (Arnold et  al., 2013; Sun
weaning and early childhood has become rare (IARC, 2012d). (The et al., 2005). For instance, in studies of Chinese males living in the
epidemiologic literature on salt-preserved fish consumption in relation Los Angeles and San Francisco/​Oakland regions of California, the
to NPC risk is discussed later.) Changes in NPC screening, diagnosis, incidence of keratinizing squamous cell NPC decreased significantly
or classification are unlikely to have had an appreciable effect on tem- by 71% (95% confidence interval [CI] = –​85, –​45%) between 1992
poral incidence and mortality trends. and 2002, whereas the incidence of non-​keratinizing NPC increased
492

30
Males
Females

25

Incidence rate (per 100,000 person-years)


20

15

10

Figure 26–2.  Age-​standardized (to world


standard population) incidence rates of
0
nasopharyngeal carcinoma in selected ao

se

an

An les no

an

te

ite

an se

er ian

i
An n-H s: K ck
ity

jin y
ity

or alay
ng

a, isia tif

ud
geographic regions, 2003‒2007. Regions

it

es

ad ort
hi


la
C

iC

ne

er ane
ac

di

pi

re
Ko

Sa
n

,N
ge : B
, L ing e: M

In

In
o
n

l
,M

hi

hi
are selected to illustrate the gradient ha

Fi

a,
ha

Am Jap
, L s: H nic

h:
C

C
e:
on

i
, A Ang ani
ng

s:
na

ei
gs

or
e:

ic
os les

le

le
ge
in incidence from northern to southern
,H

lg
,B
ha

un
:
hi

sp

p
ap

ap
n

or

iy
es
e

A
s
ho

An , L An
C

, L ge

R
ng
,S

na
na

T
ap

i
ng

el
China, the variation in incidence among
,Z

SA An

A
hi
hi

ng
na

os SA os
Si

bi
C
C

a:
o
na

ge os

SA ele
Si
hi

L
os

ra
sk
racial/​ethnic groups in Singapore and the
hi

s
,

iA
g
SA

s:

la
C

le

ud
SA
United States, and intermediate rates in

U
U

os
U

Sa
SA
U

,L

U
the Arctic, North Africa, and the Middle

U
SA
,L
East. Data from Cancer Incidence in Five

U
SA
Continents, Vol. X (Forman et al., 2013).

Harbin City: 1.3

Beijing City: 1.2

Cixian County: 1.0

Yangcheng County: 0.4

Jiaxing City: 3.9


Yanting County: 1.6
Wuhan City: 3.6 Shanghai City: 3.7
Jiashan County: 4.0 Haining County: 4.3

Qidong County: 1.9

Hong Kong: 14.4


Macau: 15.7
Zhongshan City: 26.8
Figure 26–3.  Age-​standardized (to world
standard population) incidence rates per
100,000 of nasopharyngeal carcinoma
among males in China, 2003–​2007. Map
shows the north-​to-​south gradient in inci-
dence within China. Data from Cancer
Incidence in Five Continents, Vol. X
(Forman et al., 2013).
 493

Nasopharyngeal Cancer 493
nonsignificantly (+ 56%, 95% CI = –​36, 277%) and the incidence of tumors establishes the presence of EBV in the initial NPC progen-
undifferentiated NPC was fairly stable (+12%, 95% CI = –​43, 120%) itor cell (Niedobitek, 2000; Raab-​Traub and Flynn, 1986). Clonal
(Sun et al., 2005). Overall, given the sharper declines in high-​inci- EBV is also detected in rare cases of NPC in situ in the absence of
dence areas, these global patterns have resulted in a narrowing of the invasive NPC, as described earlier (Cheung et al., 1998; Pak et al.,
international variation in NPC incidence and mortality. 2002, 2005; Pathmanathan et  al., 1995). The genomic diversity of
EBV in NPC biopsies has only recently begun to be appreciated,
and no specific viral strains or sequence variants have yet been con-
MIGRANT STUDIES sistently associated with NPC risk or clinical characteristics. Whole-​
genome sequencing of nine EBV genomes derived from NPC tumor
Migrant studies suggest that a combination of environmental, cultural, biopsy specimens from Hong Kong, compared with a reference
and heritable genetic factors contribute to NPC risk. Early studies EBV genome, revealed more than 1,700 sequence variations, repre-
showed that individuals with low-​risk North American or European senting greater sequence diversity than previously captured (Kwok
parents who were born in high-​risk regions had a significantly higher et al., 2012, 2014). Further sequencing in large studies will be nec-
risk of NPC than would be expected from their parentage. For exam- essary to determine whether any such variations play a role in NPC
ple, between 1955–​1964, five of 273 white male decedents with NPC development.
in California had been born in China or the Philippines, more than Retrospective studies in the 1970s established that NPC patients
40 times the 0.11 expected based on controls with cancer of the oro- have higher levels of several antibodies against EBV antigens,
pharynx, hypopharynx, or pharynx, not otherwise specified (Buell, including immunoglobulin G (IgG) and IgA against the viral capsid
1973). Similarly, the age-​standardized annual incidence rate of NPC antigen (VCA), early antigen (EA), and latent viral nuclear antigens
among males of French origin born in the Maghreb (Northwest Africa) 1 and 2 (EBNA1 and EBNA2), and neutralizing antibodies against
was 1.7 per 100,000, compared with 0.3 per 100,000 for French males EBV-​specific DNase, compared with controls (de-​The et al., 1978;
born in France (Jeannel et al., 1993). Henderson et  al., 1977; Lin et  al., 1977). Subsequent prospective
The increased risk of NPC among migrants from high- to lower- studies demonstrated increases in these antibody titers several years
risk host countries remains higher in first-​generation migrants. In before NPC development (Zeng et al., 1982, 1983, 1985). A land-
Sweden, compared with native-​born Swedes, standardized incidence mark prospective cohort study by Chien et al. (2001), with 131,981
ratios (SIRs) for NPC were significantly elevated among first-​gen- person-​years of follow-​up for 9699 men in Taiwan, revealed that the
eration immigrants born in Southeast Asia (SIR = 35.60 for males, cumulative incidence of NPC per 100,000 person-​years was 11.2 for
24.63 for females), North Africa (SIR = 12.45 for males, 34.73 for subjects with negative serological test results for anti-​VCA IgA and
females), and Asian Arab countries (SIR = 4.92 for males, 10.95 for anti-​DNase neutralizing antibodies, 45.0 for those who were posi-
females) (Mousavi and Hemminki, 2015). In Israel, the odds ratio tive for one marker, and 371.0 for those who were positive for both
(OR) of NPC comparing individuals under 30 years of age born in markers. The relative risk (RR) was 32.8 (95% CI = 7.3–​147.2) for
Africa to African parents versus those born in Israel to Israel-​born two versus no markers, and increased with longer duration of fol-
parents was 3.51 (95% CI = 1.21–​10.1), and that for Israel-​born indi- low-​up. Consequently, anti-​VCA IgA, anti-​EA IgA, and anti-​EBV
viduals with a mother born in Africa was 4.44 (95% CI = 1.67–​11.8) DNase antibodies form the basis of long-​ standing immunofluo-
(Parkin and Iscovich, 1997). Age-​standardized NPC incidence rates rescence-​based screening tests for NPC in high-​risk populations.
among Chinese migrants were shown to decline with increasing dis- Circulating cell-​free EBV DNA is a valuable biomarker for NPC
tance from China. Progressively lower rates were observed among detection and prognosis. Its value for NPC screening in unaffected
Chinese in Singapore, Hawaii, and California (Yu and Hussain, 2009). persons, however, is probably limited (Chan et  al., 2013; Ji et  al.,
Nevertheless, in the United States from 2008–​2012, the age-​adjusted 2014).
incidence rates of NPC among all Asians and Pacific Islanders (3.58 A recent study in Taiwan demonstrates the value of screening for
and 1.23 per 100,000 among males and females, respectively) were NPC in high-​incidence groups using enzyme-​linked immunosorbent
five to six times higher than rates among non-​Hispanic whites (SEER, assay (ELISA) kits for IgA against VCA and EBNA1 (Coghill et al.,
2015). When interpreting these results, it should be noted that Asians 2014). The study measured pre-​diagnostic serum levels of ELISA-​
and Pacific Islanders in the United States include migrants from high-​, based IgA antibodies against several EBV peptides in 21 NPC cases
intermediate-​, and low-​incidence areas in Asia, as well as US-​born and 84 controls from high-​risk multiplex NPC families. The detec-
Asian and Pacific Islander Americans. tion of anti-​EBNA1 IgA had 80% sensitivity to identify future cases,
although specificity was only 60%; thus, a more specific screening
test is still needed. The enrollment phase of a cluster randomized
ETIOLOGIC FACTORS controlled trial in two cities in southern China demonstrated the
feasibility of mass screening for NPC using ELISA-​based sero-
Epstein-​Barr Virus logic testing for anti-​VCA and EBNA1 IgA, followed by fiberoptic
EBV is a ubiquitous herpesvirus that infects nearly all humans, usually endoscopy and/​or biopsy for individuals with a positive screening
during childhood, and persists latently for life (Klein et al., 2010). In test result (Liu et al., 2013). Of 28,688 participants, 862 (3.0%) were
areas with high NPC incidence, EBV infection typically occurs during classified as having a high-​risk serologic screening result. Thirty-​
infancy or early childhood, and over 90% of the population is infected eight subjects in this group (4.4% of all 862 high-​risk subjects;
by age 8 years (Kangro et al., 1994; Xiong et al., 2014). In low-​inci- 5.8% of those who underwent fiberoptic endoscopy and 43.7% of
dence areas, primary EBV infection usually occurs later and is less those who underwent biopsy) were found to have NPC, including 29
pervasive. For example, between 2003 and 2010, only 50% of the US (76.3%) with no symptoms at the time of screening and 27 (71.1%)
population was seropositive for EBV at ages 6–​8  years and 69% at with clinical stage I or II disease. These initial results suggest that
ages 15–​17 years (Balfour et al., 2013). EBV-​based screening for NPC in high-​incidence areas could reduce
EBV was first classified by the International Agency for Research mortality through early detection and treatment. Longer follow-​up
on Cancer (IARC) as a human carcinogen in 1997, based largely on of the randomized communities is needed to establish screening effi-
the evidence regarding NPC (IARC, 1997). At the time, evidence of cacy (Hildesheim, 2013).
the etiologic involvement of EBV in NPC had been accumulating for A few studies have investigated interactions between EBV and other
more than three decades (Old et al., 1966), and it continues to expand NPC risk factors. Positive interactions between cigarette smoking and
(IARC, 2012b). However, the incidence of NPC is much lower than anti-​EBV antibody titers were found in some (Hsu et  al., 2009; Lin
the prevalence of EBV infection in all countries, indicating the impor- et al., 1979; Xu et al., 2012) but not all (Chang et al., 2012) studies.
tance of cofactors, especially for non-​keratinized tumors. Cigarette smoke extract has been shown to promote EBV activation
The detection of clonally identical EBV DNA, RNA, and/​or gene in vitro (Xu et al., 2012), suggesting that tobacco smoke may increase
products in the malignant cells of virtually all non-​keratinizing NPC NPC risk by inducing EBV reactivation.
49

494 PART IV:  Cancers by Tissue of Origin


In addition to the probable role of earlier age of primary EBV infec- Dietary Consumption of Salt-​Preserved
tion, host cofactors are likely to modulate the association between Fish and Other Foods
EBV and NPC. Genetic susceptibility to both EBV infection and
NPC is suggested by greater EBV IgA seroreactivity in unaffected Dietary consumption of Chinese-​style salt-preserved fish is classified
first-​degree relatives of NPC cases than in general community mem- by the IARC as carcinogenic to humans based on sufficient evidence
bers. Two-​to six-​fold differences in the seroprevalence or serore- regarding NPC (IARC, 1993, 2012d). Chinese-​style salt-preserved
activity of anti-​VCA and/​or anti-​EBNA IgA have been reported in fish is prepared by salting, brining (soaking in salted water), dry-​salt-
Taiwan (Pickard et al., 2004), southern China (Qin et al., 2011a), and ing (mixing with dry salt and allowing the resulting brine to drain
Indonesia (Hutajulu et  al., 2012). Shared environmental risk factors away), pickle curing (mixing with salt and storing in the resulting
probably also contribute to these findings, as spouses of multiplex brine), or a combination of these methods. Salting may result in partial
NPC cases in Taiwan and of sporadic (but not multiplex) NPC cases decomposition of the fish. After a few days of salting, fish are dried in
in southern China had a significantly higher seroprevalence of anti-​ the sun for 1–​7 days and then are stored for 4–​5 months before being
EBV IgA than community controls (Pickard et al., 2004; Qin et al., consumed.
2011a). Further insight into host susceptibility to EBV infection and Evidence that consumption of Chinese-​ style salt-preserved fish
NPC development comes from evidence of a functional deficit in the increases NPC risk emerged in the 1960s and 1970s, when Ho noted
CD8+ T-​cell immune response to EBNA1 in NPC cases (Fogg et al., that the Tanka boat people were at increased risk of NPC and hypoth-
2009). Specifically, EBNA1-​ specific CD8+ T-​ cells were detected esized that salt-​preserved fish, a staple of the Tanka diet, might account
in 10 of 14 healthy donors, with confirmation of HLA class  I  pres- for the excess risk, especially when coupled with a deficiency of fresh
entation of EBNA1 epitopes to CD8+ T-​cells in six of six donors. fruits and vegetables (Ho, 1967; Ho et al., 1978). Chinese-​style salt-
By contrast, only three of 22 patients with EBV-​associated NPC had preserved fish has traditionally been a popular dietary staple through-
detectable EBNA1-​specific CD8+ T-​cells (P = 0.0003 versus healthy out high-​risk regions in southern China and Southeast Asia (IARC,
donors), and a functional deficit in the EBNA1-​specific response was 1993, 2012d). Consumption of salt-​preserved foods in the traditional
evidenced by rescue of that response via short-​term peptide and cyto- diet in other geographic regions, such as North Africa and the Arctic,
kine stimulation in eight of 14 patients. Meanwhile, the frequency of also may contribute to the NPC incidence rates in these areas. The
the LMP2-​specific CD8+ T-​cell response did not differ significantly mechanism by which consumption of Chinese-​style salt-preserved fish
between NPC patients and controls, indicating a specific deficit in the causes NPC is not established, but may involve exposure to carcino-
immune response to EBNA1. genic N-​nitroso compounds and/​or reactivation of latent EBV infec-
tion (IARC, 1993, 2012d).
Nearly all case-​control studies of salt-​preserved fish in relation to
NPC risk have been conducted in Chinese populations. No cohort
Infections Other Than EBV studies of the association have yet been published. Among the nearly
High-​risk human papillomavirus (HPV) DNA has been detected in a 20 case-​control studies of Chinese populations, the OR for NPC asso-
minority of NPC tumors, especially in low-​incidence regions. HPV ciated with weekly versus no consumption or rare consumption of
appears not to play an important etiologic role in high-​incidence areas salt-​preserved fish has generally ranged from 1.1 to 4, while that for
(Dogan et al., 2014; Giannoudis et al., 1995; Hording et al., 1994; Lin daily consumption ranges from 1.8 to 20, with positive trends by fre-
Z et  al., 2014). HPV-​positive tumors in the nasopharynx may actu- quency of consumption (Guo et al., 2009; Henderson and Louie, 1978;
ally be oropharyngeal carcinomas that have been misclassified, since IARC, 2012d; Jia et al., 2010; Xu et al., 2012; Yu et al., 1986, 1988,
tumors of the oropharynx have a strong etiologic link with HPV (see 1989; Yuan et al., 2000b; Zheng X et al., 1994). A small case-​control
Chapter 29) and can extend into the nasopharynx. In a histopathologic study of Alaska natives (based on 13 NPC cases and matched controls
study of 45 non-​keratinizing NPC tumor specimens in the United with questionnaire data) also found a positive association with salt-
States, four (9%) were positive for HPV. Records on three of these preserved fish consumption in childhood (Lanier et al., 1980).
cases included information on stage at diagnosis. All three involved Findings have not been consistent regarding whether consump-
tumor extension into the oropharynx (Singhi et al., 2012). In contrast, tion of salt-​ preserved fish during weaning and childhood confers
only four (12%) of 34 HPV-​negative NPCs with information on stage greater risk than exposure in adulthood. Some studies support this
had oropharyngeal involvement. hypothesis (Ning et al., 1990; Yu et al., 1986), whereas others do not
Registry studies suggest that NPC risk may be doubled among indi- (Armstrong et al., 1998; Yu et al., 1988, 1989; Zheng X et al., 1994).
viduals with AIDS, but excess risk is not observed among patients who Consumption of salt-​preserved fish during weaning might be expected
receive immunosuppressive therapy following solid organ transplant. to increase NPC risk as a result of food regurgitation into the nasophar-
The SIR for AIDS patients in the United States was identical (2.4) for ynx. Differences in risk by age at first exposure are highly correlated
both keratinizing and non-​keratinizing tumors (Shebl et al., 2010). No with dietary patterns throughout life. A case-​control study of NPC in
increase in risk was observed among recipients of solid organ trans- Taiwan estimated dietary intake of nitrosamines and their precursors
plants in the United States (SIR = 0.96, 95% CI = 0.42–​1.90) (Engels from a variety of foods, and found positive associations with consump-
et al., 2011). Since the incidence rate of NPC did not increase appreci- tion of nitrite and nitrosamines from foods other than soybean products
ably after the 1980s as a consequence of the AIDS epidemic, it seems during childhood and weaning, but not adulthood (Ward et al., 2000).
unlikely that alterations in immune control of EBV infection in adult- ORs for the highest versus lowest quartile of consumption of non-​soy-
hood play a major role in the development of NPC. bean nitrosamines, as reported by subjects’ mothers, were 3.9 (95%
Multiple studies of prior chronic ear, nose, throat, and lower respi- CI = 1.4–​10.4) for intake during weaning, 2.6 (95% CI = 1.0–​7.0) at
ratory tract conditions, including allergic rhinitis (Chung et al., 2014; age 3 years, and 2.2 (95% CI = 0.8–​5.6) at age 10 years. In light of the
Lin KT et  al., 2015), rhinitis or rhinosinusitis (Hung et  al., 2013; generally decreasing population-​level consumption of salt-preserved
Yu et al., 1990; Yuan et al., 2000a), otitis media (Huang WY et al., fish, especially in early life (IARC, 2012d), future studies may lack
2012; Yu et al., 1990; Yuan et al., 2000a), and nasal polyps (Yu et al., sufficient exposure heterogeneity to resolve the question of whether
1990; Yuan et al., 2000a), have reported positive associations on the risk varies by age at first exposure.
order of a two-​fold or greater increase in NPC risk. Repeated benign Some evidence suggests that the strength of the association between
inflammation and infection of the respiratory tract may increase the NPC and consumption of salt-​preserved fish has declined over time
susceptibility of the nasopharyngeal mucosa to NPC development. (IARC, 2012d). This pattern could reflect a shift toward commercially
Some nasopharyngeal flora also may contribute to the local forma- produced products and away from home preparation (Ward et  al.,
tion of carcinogenic N-​nitroso compounds through the reduction of 2000) or a decrease in the amount of fish consumed. Increasing eco-
nitrates (Charriere et  al., 1991). Recall bias and prodromal symp- nomic development, growing awareness of the potential carcinogenic-
toms have been difficult to rule out as potential explanations for these ity of salt-​preserved fish, and a societal shift toward a more Western
findings. dietary pattern may have resulted in secular changes in the association
 495

Nasopharyngeal Cancer 495
between salt-​preserved fish consumption and NPC risk (Zhai et  al., significantly heterogeneous, with a higher point estimate in low-​inci-
2014). However, detailed temporal patterns in salt-preserved fish pro- dence populations (meta-​RR = 1.76, 95% CI = 1.47–​2.10) than high-​
duction and consumption have not been well documented. Any meta-​ incidence populations (meta-​RR  =  1.29, 95% CI  =  1.05–​1.58). The
analysis or pooled analysis of salt-​preserved fish consumption would association also was stronger for squamous cell NPC (meta-​RR = 2.20;
be challenged by the between-​study heterogeneity resulting from such 95% CI = 1.63–​1.98) than for undifferentiated NPC (RR = 1.27, 95%
time trends, as well as regional variation in fish-​preparation methods CI = 0.98–​1.66). Exposure to passive smoking during childhood was
and consequent nitrosamine levels. not significantly associated with NPC risk (meta-​OR based on five
Consumption of other preserved food items that are high in nitro- studies = 1.29, 95% CI = 0.80–​2.09). Three studies detected signifi-
samines also has been shown to be associated with increased risk of cant positive associations (Armstrong et al., 2000; Nesic et al., 2010;
NPC in diverse geographic regions, although with less strength and Yuan et al., 2000a), whereas two did not (Cheng et al., 1999; Yu et al.,
consistency (Armstrong et  al., 1998; Fachiroh et  al., 2012; Farrow 1990). The relatively modest association with active tobacco smoking
et  al., 1998; Feng et  al., 2007; Jeannel et  al., 1990; Jia et  al., 2010; translates to an attributable fraction for NPC that is roughly one-​tenth
Ning et al., 1990; Xu et al., 2012; Yu et al., 1988; Yuan et al., 2000b). that for lung cancer and other respiratory tract malignancies, and an
For example, in a US study, total preserved meat intake was calculated even smaller absolute contribution (IARC, 2004b, 2012d).
based on intake of bacon, ham, sausage, liverwurst, and hot dogs; this Studies of the association between exposure to domestic wood fire
variable was found to be associated with increased risk of non-​keratin- and NPC risk have yielded inconsistent results. Case-​control studies
izing NPC (OR for highest vs. lowest quartile = 4.59, 95% CI = 0.78–​ in Malaysia and southern China reported OR estimates between 2.1
27.01, Ptrend = 0.04), but not squamous cell NPC (Ptrend = 0.58) (Farrow and 5.4 for exposure to domestic wood fire (Armstrong et al., 1978;
et al., 1998). In North Africa, preserved foods include quaddid (dried Guo et  al., 2009; Zheng YM et  al., 1994), whereas other studies in
mutton stored in oil), harissa (a spicy condiment containing red pep- Hong Kong (Yu et al., 1986), southern China (Yu et al., 1990, 1988),
per, olive oil, garlic, caraway, and salt), toklia (a basic stewing prepa- and North Africa (Feng et  al., 2009) found no significant associa-
ration containing red and black pepper, garlic, oil, caraway, salt, and tion. The associations reported with burning incense or anti-​mosquito
coriander), khelii (salted, spicy, dried meat cooked and preserved in coils also have been mixed, with some positive (Geser et  al., 1978;
a mixture of oil and melted bovine greases), rancid butter, and rancid Shanmugaratnam and Sobin, 1978; West et al., 1993) and some null
sheep fat. Some of these, particularly quaddid, rancid butter, and ran- (Yu et al., 1990, 1986, 1988). One study in North Africa found a pos-
cid sheep fat, have been found to be associated with increased risk of itive association of NPC risk with cannabis smoking (OR for ≥ 2,000
NPC, with ORs on the order of 2–​3 for frequent consumption (Feng exposures versus none = 3.25, 95% CI = 1.24–​8.50) and with hav-
et al., 2007; Jeannel et al., 1990). A meta-​analysis of six case-​control ing had a non-​ventilated kitchen during childhood (OR = 1.64, 95%
studies in China, Southeast Asia, and North Africa reported a two-​fold CI = 1.09–​2.48), but not with use of a water pipe (OR = 0.49, 95% CI
increase (meta-​OR = 2.04, 95% CI = 1.43–​2.92) in NPC risk associated = 0.20–​1.23) (Feng et al., 2009). Ever use of snuff in this study was
with high versus low consumption of preserved vegetables, despite associated with increased risk of differentiated NPC (OR = 30.2, 95%
difference in food preservation and consumption levels across studies CI = 1.67–​546) but not undifferentiated NPC (OR = 0.97, 95% CI =
(Gallicchio et  al., 2006). In contrast, frequent consumption of fresh 0.59–​1.58).
fruits and/​or vegetables has been associated with a lower risk of NPC Chewing of betel quid—​a mixture of areca nut, betel leaf, and other
in several studies from high-​, intermediate-​, and low-​incidence regions ingredients, with or without tobacco—​is common in India and many
(Armstrong et al., 1998; Jia et al., 2010; Liu et al., 2012; Polesel et al., parts of Asia, and is classified by the IARC as a cause of cancers of the
2013; Ward et  al., 2000; Yu et  al., 1989; Yuan et  al., 2000b). In the oral cavity and esophagus in humans (IARC, 2004a, 2012d). However,
meta-​analysis by Gallicchio et  al. described earlier, high versus low whether use of betel quid causes NPC is unclear, due to potential con-
dietary intake of all non-​preserved vegetables was associated with founding by tobacco smoking. In a community-​based cohort of 19,719
36% lower risk of NPC (meta-​OR  =  0.64, 95% CI  =  0.48–​0.85), men in Taiwan, NPC mortality was higher among those who smoked
based on five contributing case-​control studies. Stronger inverse asso- and chewed betel quid (RR = 4.2, 95% CI = 1.5–​11.4; 8 deaths) than
ciations were observed with consumption of green leafy vegetables those who only smoked (RR  =  1.3, 95% CI  =  0.6–​3.0; 18 deaths),
(OR  =  0.55, 95% CI  =  0.32–​0.96) and non-​starchy roots and tubers compared with those who did neither (9 deaths) (Wen et  al., 2005).
(OR = 0.55, 95% CI = 0.34–​0.88). Hypothesized mechanisms for the A case-​control study of NPC high-​risk families in Taiwan found that
apparent protective effect of fresh fruits and vegetables against NPC ≥ 20 years of betel nut use was associated with increased NPC risk in
include prevention of oxidation and nitrosamine formation. families with late-​onset NPC at or after age 40 years (OR versus never
use = 2.44, 95% CI = 1.16–​5.13), but not early-​onset NPC (OR = 0.71,
95% CI  =  0.27–​1.84) (Yang et  al., 2005). By contrast, use of betel
Tobacco and Other Smoke nut or betel quid was not associated with NPC risk after multivari-
ate adjustment in case-​control studies in the Philippines (OR = 0.56,
The IARC classifies the evidence as sufficient that tobacco smoking 95% CI = 0.18–​1.8) (West et al., 1993) and Thailand (OR = 1.34, 95%
causes cancer of the nasopharynx in humans (IARC, 2004b, 2012d). CI = 0.64–​2.83) (Fachiroh et al., 2012)
In recent case-​control studies, the OR for NPC among current or ever
versus never cigarette smokers has ranged from approximately 1.2 to
3.0, with generally positive (albeit not always monotonic) exposure–​
Alcohol
response trends for intensity, duration, and pack-​years of smoking
(Fachiroh et al., 2012; Feng et al., 2009; Ji et al., 2011; Polesel et al., The results of studies of alcohol consumption and risk of NPC have
2011; Xu et al., 2012). A meta-​analysis of 28 case-​control studies and been inconsistent; both positive (Armstrong et al., 1998; Ruan et al.,
four prospective cohort studies published between 1979 and 2011 2010; Vaughan et  al., 1996) and null findings (Chen et  al., 1990;
reported a meta-​RR of 1.60 (95% CI = 1.38–​1.87) for ever versus never Cheng et al., 1999; Fachiroh et al., 2012; Ji et al., 2011; Sriamporn
smoking (Xue et al., 2013). Higher risk was observed for those with et  al., 1992; Xu et  al., 2012; Zheng X et  al., 1994) have been
greater cumulative smoking, whether measured as pack-​years (meta-​ reported from case-​control studies. A meta-​analysis of 11 case-​con-
RR for ≥ 30 = 2.93, 95% CI = 1.87–​4.61), cigarettes per day (meta-​RR trol studies of variable quality reported a meta-​OR of 1.33 (95%
for higher exposure, with variable cut-​points by study  =  1.98, 95% CI = 1.09–​1.62) for the highest versus lowest category of alcohol
CI = 1.31–​2.98), or years of smoking (meta-​RR for longer duration, intake, based on variable definitions of high and low intake among
with variable cut-​points by study = 2.26, 95% CI = 1.32–​3.84). Risk studies (Chen et  al., 2009). The association was only marginally
was not associated with earlier age at initiation (meta-​RR for earliest statistically non-significant in studies that controlled for smoking
group, with variable cutpoints by study = 1.17, 95% CI = 0.78–​1.75). (meta-​OR = 1.26, 95% CI = 0.99–​1.62). Among the six studies that
Colinearity of the various measures of smoking makes it difficult considered at least three categories of alcohol intake frequency, a J-​
to measure their independent associations with risk. Results were shaped trend was evident, with NPC risk decreasing up to 15 drinks
496

496 PART IV:  Cancers by Tissue of Origin


per week and increasing thereafter. Data were insufficient to stratify (Hildesheim et al., 2001), a pattern that lends biological plausibility to
analyses by NPC histologic type, which may contribute to the het- the association.
erogeneity of results. A two-​fold or greater increased risk of NPC among woodwork-
ers and other individuals potentially exposed to wood dust has been
reported in several studies conducted in both high-​and low-​incidence
Tea and Traditional Herbal Medicine regions (Armstrong et  al., 2000; Demers et  al., 1995; Hildesheim
An inverse association between tea consumption and NPC risk has et al., 2001; Sriamporn et al., 1992; Vaughan and Davis, 1991). Other
been reported in several case-​control studies in China and Taiwan (Hsu studies did not find an association (Siew et al., 2012; Stellman et al.,
et al., 2012a; Ruan et al., 2010; Xu et al., 2012), with OR estimates 1998; Vaughan et al., 2000). Two cohort studies of woodworkers have
ranging from 0.4 to 0.6 for any consumption, and significant inverse reported results for NPC. One was a pooled study of 28,704 work-
relationships with frequency of consumption. No prospective studies ers from five studies in the United Kingdom and United States that
have yet replicated these findings. Chinese herbal tea and Cantonese detected an SMR of 2.4 (95% CI = 1.1–​4.5) based on nine NPC deaths
slow-​cooked soup, which are forms of traditional Chinese medicine, (7 among furniture workers [SMR = 2.9, 95% CI = 1.2–​5.9] and two
were both inversely associated with NPC risk in two case-​control stud- among plywood workers [SMR = 4.6, 95% CI = 0.6–​16.4]) (Demers
ies (although exposure–​response trends were not evident for herbal et al., 1995). In this study, an excess of NPC was observed among those
tea) (Jia et al., 2010; Xu et al., 2012), but a positive association with with either definite or possible exposure to wood dust, first employ-
herbal tea was detected in another study (Zheng YM et al., 1994). In ment prior to 1950, and 30 or more years since first employment. An
general, traditional herbal medicines are inconsistently associated with analysis based on 45,399 men (12.5% of the American Cancer Society
NPC risk, conferring a two-​to four-​fold increase in risk in some stud- Cancer Prevention Study II) found no excess of NPC mortality among
ies (Hildesheim et al., 1992; Shanmugaratnam et al., 1978; West et al., men who self-​ reported employment in a wood-​ related occupation
1993; Zheng X et al., 1994), but not others (Jia et al., 2010; Yu et al., (RR = 1.44, 95% CI = 0.19–​10.9) or occupational exposure to wood
1986, 1990). dust (RR = 0.44, 95% CI = 0.06–​3.29) (Stellman et al., 1998). In each
case the result was based on only one NPC death.
The NPC case-​ control study with the most detailed informa-
tion about wood dust exposure detected positive exposure–​response
Occupational Exposures trends with increasing duration of exposure (OR for > 10 years versus
The two occupational exposures classified by the IARC as causally none = 2.2, 95% CI = 0.93–​5.3, Ptrend = 0.03 excluding the 10 years
related to NPC in humans are formaldehyde (IARC, 2006, 2012c) and before diagnosis or interview) and cumulative exposure (OR for ≥
wood dust (IARC, 1995, 2012a). The association between occupa- 25 intensity-​years versus none = 2.4, 95% CI = 1.2–​5.1, Ptrend = 0.02)
tional exposure to formaldehyde and risk of NPC has been investigated (Hildesheim et al., 2001). In this study, which was based in Taiwan,
mainly in low-​incidence regions. The main evidence comes from a associations with wood dust exposure (as with formaldehyde) were
retrospective cohort mortality study of 25,619 workers at 10 US form- stronger among individuals seropositive for EBV antibodies associ-
aldehyde production or usage plants, where an excess of NPC mortal- ated with NPC risk.
ity was detected based on seven NPC deaths among all workers (six Several studies in China found that NPC risk was increased by
among formaldehyde-​exposed workers) from pre-​1966 through 1979. about three-​fold among textile workers, with some evidence of posi-
The standardized mortality ratio [SMR] comparing all workers to US tive exposure–​response trends, potentially due to cotton dust exposure
rates was (3.18 [p < 0.05] [Blair et al., 1986]). In further follow-​up, (Li et al., 2006; Ng, 1986; Zheng et al., 1992). However, some con-
eight NPC deaths were identified among the exposed workers by 1994 tradictory results have been reported (Yu et al., 1990). Other occupa-
(SMR = 2.10, 95% CI = 1.05–​4.21) (Hauptmann et  al., 2004), and tional exposures, including industrial heat and combustion products
nine through 2004. One case identified in the longest follow-​up was (Armstrong et al., 2000; Yu et al., 1990), solvents (Guo et al., 2009;
shown by secondary sources to be oropharyngeal cancer. Exclusion Hildesheim et al., 2001), and pesticides (Li et al., 2006; West et al.,
of this case reduced the SMR through 2004 to 1.84 (95% CI = 0.84–​ 1993; Zhu et al., 2002; Zou et al., 2000), have not consistently been
3.49) (Beane Freeman et  al., 2013). Statistically significant positive associated with NPC risk.
exposure–​response trends by peak exposure and cumulative exposure
were detected in follow-​up through 1994 (Hauptmann et  al., 2004),
but these were no longer statistically significant in the analysis through HOST FACTORS
2004 (Beane Freeman et al., 2013). Five NPC deaths, all in the highest
Immune Suppression
category of peak exposure, occurred at a single plant. At the remaining
nine plants, the SMR for NPC was below 1.0 (McLaughlin and Tarone, Some evidence suggests that immune suppression in adulthood is not
2014) and exposure–​response trends were in a positive direction but likely to play a major role in the development of NPC. The absence
not significant (Beane Freeman et al., 2013). of a large increase in NPC risk among AIDS patients (Shebl et  al.,
No excess of NPC was observed in recent cohort or nested case-​con- 2010) and solid organ transplant recipients (Engels et al., 2011), men-
trol studies of formaldehyde-​exposed workers in Finland, England, and tioned earlier, supports this hypothesis, as do null studies of cancer
Wales, or in studies from the United States published after the 2006 in patients with primary immunodeficiency disorders (Rezaei et  al.,
IARC monograph on formaldehyde (Coggon et al., 2014; Hauptmann 2011; Salavoura et al., 2008), although the statistical power of the lat-
et al., 2009; Meyers et al., 2013; Siew et al., 2012). A meta-​analysis of ter was low. The strong association between NPC and genetic variants
six case-​control studies and seven cohort studies found a marginally in the HLA region suggests that certain aspects of immune function do
non-significant excess risk of NPC in case-​control studies (overall meta-​ affect NPC risk. Except for the null studies of cancer in patients with
OR = 1.22, 95% CI = 1.00–​1.50). Adjustment for smoking reduced the primary immunodeficiency disorders (Rezaei et al., 2011; Salavoura
meta-​OR estimate to 1.10 (95% CI = 0.80–​1.51). Conflicting results et  al., 2008), these data suggest that defects in immune function in
were observed among cohort studies that excluded (meta-​SMR = 0.72, childhood, when EBV infection first occurs, may be more important
95% CI = 0.40–​1.28) or included (meta-​SMR = 1.60 [95% CI not than immunodeficiency in adulthood.
reported]) the anomalous US plant (Bachand et al., 2010).
Evidence regarding the association between formaldehyde and Familial Aggregation
risk of NPC in high-​incidence areas (i.e., predominantly non-​keratin- Epidemiologic studies have demonstrated a 4-​to 20-​fold increased risk
izing NPC) is sparse. However, one study conducted in Taiwan that of NPC associated with a first-​degree family history of the disease,
collected extensive occupational history information found evidence thereby establishing strong familial susceptibility (Friborg et al., 2005;
for an association between formaldehyde exposure and NPC risk that Hsu et al., 2011; Ji et al., 2011; Jia et al., 2004; Zeng and Jia, 2002).
was stronger among individuals seropositive for any of various anti-​ A cohort study of the entire population born in Greenland (73,222 per-
EBV antibodies associated with NPC (96% of cases, 29% of controls) sons), followed from 1973 to 2002, used population-​based registry
 497

Nasopharyngeal Cancer 497
linkages to identify relatives and their cancer diagnoses (Friborg et al., specific causal variants (Su et al., 2013). Relatively consistent findings
2005). This study reported an RR of 8.0 (95% CI = 4.1–​14.0) for NPC are positive associations with HLA-​A*02:07 (in linkage disequilibrium
among first-​degree relatives, with no significant differences in risk by with HLA-​B*46:01) and HLA-​B*58:01 (in linkage disequilibrium with
age and sex of the relative and index case or type of relation. In low-​ HLA-​A*33:03) and an inverse association with HLA-​A*11:01 (in link-
incidence regions, the number of NPC cases with a family history of age disequilibrium with HLA-​B*13) (Bei et al., 2012; Goldsmith et al.,
NPC is small, precluding robust analyses of the risk of familial NPC in 2002; Hildesheim and Wang, 2012). Other HLA class I genes (HLA-​
such areas (Vaughan et al., 1996). Nevertheless, familial aggregation of C, -​E, -​G, and others) and class II genes (HLA-​DR, -​DP, and -​DQ) are
NPC has been documented in high-​, intermediate-​, and low-​incidence less well studied and are not consistently associated with NPC risk.
populations (Bei et al., 2012; Zeng and Jia, 2002), especially in south-
ern China, where one family of 109 relatives across four generations
has had 15 members affected by NPC (Zhang and Zhang, 1999). Metabolism and Biotransformation Genes
The risk of malignancies other than NPC does not generally appear Compelling evidence that diet and tobacco smoking affect NPC risk
to be elevated in NPC families (Jia et al., 2004; Yu et al., 2009), nor makes it plausible that genetic variation in phase I and II biotransfor-
have studies observed an excess of NPC in any of the known inherited mation genes, including those in the glutathione-​S-​transferase (GST),
cancer syndromes (Garber and Offit, 2005). These observations argue cytochrome P450 (CYP), and N-​ acetyltransferase (NAT) families,
against a familial syndrome that includes NPC as well as other cancers. could contribute to NPC risk by influencing the metabolism of nitro-
However, some studies, including the Greenland cohort study, suggest samines and other environmental carcinogens. Several meta-​analy-
that risk of other virus-​associated malignancies, such as cancers of ses of up to 15 case-​control studies of the GSTM1 and GSTT1 null
the salivary glands and uterine cervix, is increased in NPC families polymorphisms, which result in reduced detoxifying enzyme levels,
(Albeck et al., 1993; Friborg et al., 2005). A complex segregation anal- have consistently demonstrated an increased risk of NPC in associa-
ysis of 1903 Cantonese pedigrees in southern China concluded that tion with the GSTM1 null genotype (meta-​ORs = 1.42–​1.54), and most
the observed pattern of inheritance was better explained by multiple also showed a positive association with the GSTT1 null genotype
genetic and environmental risk factors than by a major susceptibility (meta-​ORs  =  1.12–​2.27) (Jin et  al., 2014; Murthy et  al., 2013; Sun
gene (Jia et al., 2005). et al., 2012; Wei et al., 2013; Zhuo X et al., 2009). In the largest of
Four familial linkage studies of NPC have been conducted in these meta-​analyses (Wei et  al., 2013), 12 of 14 included studies of
affected siblings or extended pedigrees to map shared regions of the GSTM1 and 9 of 10 studies of GSTT1 were conducted in Asians, such
genome that may confer disease susceptibility (Feng et al., 2002; Hu that results in Asians (OR  =  1.53, 95% CI  =  1.24–​1.88 for GSTM1
et al., 2008; Lu et al., 1990; Xiong et al., 2004). These studies found null; OR  =  2.28, 95% CI  =  1.48–​3.50 for GSTT1 null) were nearly
disparate results that included (a)  a putative susceptibility locus at identical to the overall findings (OR = 1.54, 95% CI = 1.28–​1.86 and
6p22 based on 30 affected siblings from southern China and Southeast OR = 2.25, 95% CI = 1.50–​3.36, respectively).
Asia (Lu et al., 1990); (b) a locus at 4p12–​p15 that further narrowed Likewise, genetic polymorphisms in CYP2E1 that may result in
to the promoter region of a novel gene, LOC344967, based on 20 mul- increased metabolic activation of xenobiotic carcinogens have been
tiplex families from southern China (Feng et  al., 2002; Jiang et  al., associated with at least a doubling of NPC risk for homozygous vari-
2006); (c)  a locus at 3p21.31–​21.2 based on 18 high-​risk southern ants in several studies (Hildesheim et al., 1995, 1997; Jia et al., 2009;
Chinese families (Xiong et al., 2004); and (d) a locus at 5p13.1 based Kongruttanachok et  al., 2001; Yang et  al., 2005). Results are variable
on 15 multiplex southern Chinese families (Hu et al., 2008). Further with respect to potential interactions with smoking. A Taiwan case-​con-
sequencing in combination with functional characterization, along trol study with 364 NPC cases and 320 controls found that the increased
with replication studies, are needed to better understand these findings risk associated with the homozygous CYP2E1 RsaI c2 allele (rs2031920)
and to resolve whether the observed differences are due to chance or was restricted to non-​smokers (RR = 9.3, 95% CI = 2.7–​32) and was
variation in methods, populations, or disease subtypes. not detected among smokers (RR = 1.1 for all smokers; RR = 0.96 for
smokers of > 30 years’ duration) (Hildesheim et al., 1997). However, a
Human Leukocyte Antigen Genes case-​control study in Guangdong, China, with 755 cases and 755 con-
Studies of HLA associations in the 1970s formed the basis of the long-​ trols found that significant positive associations with seven CYP2E1 sin-
standing hypothesis that HLA-​mediated variation in the ability to pres- gle-​nucleotide polymorphisms (SNPs) (not including rs2031920) and
ent EBV antigens to immune cells may influence NPC risk (Simons two CYP2E1 haplotypes were detectable only among smokers aged <
et al., 1975). In particular, a two-​to three-​fold increase in NPC risk 46 years (ORs for SNPs = 1.88–​2.99, P = 0.0140–​0.0001; ORs for hap-
was associated with HLA-​A2-​B46 and B17 in Asians (Simons et al., lotypes = 1.65 and 2.58, P = 0.026 and 0.007, respectively) (Jia et al.,
1974, 1978), whereas a 30%–​50% decrease in risk of NPC was asso- 2009). Studies of other CYP and NAT genes are limited in number, with-
ciated with HLA-​A11, B13, and A2, in Asians and whites (Burt et al., out consistent results (Bendjemana et al., 2006; Cheng et al., 2003; Jiang
1994, 1996; Chan et al., 1983; Simons et al., 1978), The former poly- et al., 2004; Lee et al., 1998; Tiwawech et al., 2006).
morphisms were postulated to confer decreased ability to present EBV-​
antigen-​presenting capability to cytotoxic T-​cells, whereas the latter
were hypothesized to increase antigen-​presenting ability. The appar-
DNA Repair and Tumor Suppressor Genes
ent inconsistency of the positive association with HLA-​A2 in Asian
populations and the inverse association in Caucasian populations was Genetic variants in DNA repair genes might modify any protective
reconciled once four-​digit HLA typing by PCR was introduced; the effects against DNA damage caused by environmental carcinogens
HLA-​A2 association in Asians is driven by HLA-​A*02:07, whereas or EBV. A meta-​analysis of up to five case-​control studies of coding
that in Caucasian populations is driven by HLA-​A*02:01 (Hildesheim SNPs in the X-​ray repair cross-​complementing group 1 gene (XRCC1),
et al., 2002). Of note, HLA-​A*02:07, which is almost exclusively pres- which is involved in the repair of DNA single-​strand breaks, found
ent in East Asians, also has been shown to be associated with increased a positive association with the Arg99Gln genotype (OR = 1.30, 95%
risk of EBV-​positive classical Hodgkin lymphoma, another EBV-​asso- CI = 1.01–​1.69 for Gln/​Gln versus Arg/​Arg, based on 1,049 cases and
ciated malignancy, in Chinese subjects (Huang X et  al., 2012). The 1,066 controls), but no association with Arg194Trp (OR = 1.79, 95%
role of HLA-​A*02:07 in the development of EBV-​related tumors may CI = 0.50–​6.40 for Trp/​Trp versus Arg/​Arg) or Arg280His (OR = 0.98,
be related to the less effective cytotoxic T-​cell response to EBV LMP2 95% CI = 0.50–​1.94 for His/​His versus Arg/​Arg) (Huang et al., 2011).
in individuals carrying HLA-​A*02:07 than in those carrying other Other reports of significant associations with genetic variation in
HLA-​A*02 alleles (Lee et al., 1997). nibrin (NBS1), a protein involved in double-​strand break repair (Zheng
Although more than 100 candidate-​gene association studies have et al., 2011), and with a member of the RAD51 protein family, which
reported associations between HLA alleles or haplotypes and NPC also is involved in repairing DNA double-​strand breaks (Qin et  al.,
risk, the high density of genes (> 150 genes) and strong linkage dis- 2011b), require independent confirmation. The latter study found no
equilibrium in the HLA region have made it challenging to identify significant association with most haplotypes of 88 DNA repair genes
498

498 PART IV:  Cancers by Tissue of Origin


(including XRCC1) in a case-​control study of 755 NPC cases and 755 findings were found to be driven largely by HLA-​A*11:01, and less so
controls, perhaps indicating that any influence of DNA repair gene by HLA-​A*02:07.
polymorphisms is moderate at most. A meta-​analysis of four NPC GWAS confirmed many of the asso-
In two meta-​analyses of a genetic variant in codon 72 of the TP53 ciations summarized in the preceding and identified a novel associa-
tumor-​suppressor gene (rs1042522, Arg72Pro), which is involved in tion between genetic variation in the CLPTM1L/​TERT region and NPC
cell cycle arrest in response to DNA damage, those with the homozy- risk, suggesting that factors involved in telomere length maintenance
gous Pro/​Pro genotype were found to have a two-​fold increased risk could be involved in NPC pathogenesis (Bei et al., 2016).
of NPC compared with Arg/​Arg carriers (Cai et  al., 2014; Zhuo XL
et  al., 2009). The larger meta-​analysis (Cai et  al., 2014), based on a
total of seven studies with 1133 cases and 1678 controls, detected this OPPORTUNITIES FOR PREVENTION
association in both Asians (OR = 1.79, 95% CI = 1.40–​2.30) and whites
(OR = 2.72, 95% CI = 1.44–​5.12), although it was stronger in the latter. The recently declining incidence rates of NPC worldwide, particu-
larly in high-​risk areas, demonstrate an important role of modifiable
Genome-​Wide Association Studies environmental or behavioral risk factors for NPC that could be targets
Five genome-​wide association studies (GWAS) with information on for prevention. Tobacco control policies and dietary modifications to
400,000–​600,000 SNPs in hundreds of NPC cases and controls have decrease intake of salt-​preserved foods and increase consumption of
been conducted to identify susceptibility loci for NPC. Collectively, fresh fruits and vegetables are important for the prevention of NPC
these studies have provided strong confirmation of previous HLA find- as well as other cancers. However, they may not prevent a large pro-
ings, but do not confirm other genes previously found in candidate-​ portion of future cases, given the fairly modest risk associations and
SNP studies to be associated with NPC risk. indications that salt-preserved fish consumption has already declined
The first NPC GWAS, which included an initial sample set of in high-​risk regions (Lau et  al., 2013). Development of a vaccine
111 Malaysian Chinese NPC cases and 260 controls, along with a against EBV could provide a basis for more widespread prevention of
validation set of 168 cases and 252 controls, identified a significant NPC and other EBV-​associated malignancies that sum to as many as
association with rs2212020 in the integrin α-​9 gene at 3p21 (OR = 200,000 new cancer cases each year, but substantial barriers stand in
2.24, Pcombined = 8.27 × 10–​7) (Ng et al., 2009). the way of such an advance (Cohen et al., 2011, 2013).
A Taiwan-​based GWAS with a discovery set of 277 NPC cases Secondary NPC prevention through EBV-​based screening, leading
and 285 controls and two validation sets with a combined 635 cases to early disease detection and curative treatment, also could reduce
and 1640 controls identified two significant susceptibility loci in the NPC mortality in high-​risk populations. Although promising early
HLA-​A region at 6p21.3 (rs2517713, OR = 1.88, Pcombined = 3.9 × 10–​20; results using ELISA to test for anti-​EBV IgA antibodies have emerged
rs2975042, OR = 1.86, Pcombined = 1.6 × 10–​19), and an independent asso- from the Taiwan multiplex family study (Coghill et al., 2014) and the
ciation with rs29232 in the gamma aminobutyric b receptor (GABBR1) preliminary phase of the southern Chinese screening trial described
gene at the same locus (OR = 1.67, Pcombined = 8.97 × 10–​17, Presidual < 5 × earlier (Liu et al., 2013), a beneficial effect has yet to be demonstrated
10–​4 after adjustment for the HLA-​A SNPs) (Tse et al., 2009). A fol- in a randomized, controlled prospective screening trial.
low-​up study using PCR-​based genotyping for the HLA-​A gene and
for 12 significant GWAS-​identified SNPs in 6p21 found that most of
FUTURE RESEARCH DIRECTIONS
the associations detected in the GWAS were due to previously identi-
fied HLA-​A associations, with the possible exception of the association
Although EBV contributes to virtually all non-​keratinizing NPC cases,
with rs29232 (GABBR1) (Hsu et al., 2012b).
it is unclear why this ubiquitous virus causes NPC in a small fraction
A subsequent GWAS with 1583 southern Chinese NPC cases and
of those infected. Early age at primary EBV infection appears to have
1894 controls in the discovery phase, plus 3507 cases and 3063 cases in
an important etiologic influence. Although several cofactors for NPC
the validation phase, identified three new susceptibility loci: rs9510787
have been established (Table 26–1), at present the majority of cases are
in the tumor necrosis factor receptor superfamily, member 19, on 13q12
(OR = 1.20, Pcombined = 1.53 × 10–​9); rs6774494 in the myelodysplasia 1
and ecotropic viral insertion site 1 fusion proteins on 3q26 (OR = 0.84, Table 26–1.  Known and Suspected Risk Factors for Nasopharyngeal
Pcombined = 1.34 × 10–​8); and rs1412829 in the cyclin-​dependent kinase Carcinoma (See Text for Details).
inhibitors 2A and 2B clusters on 9p21 (OR = 0.78, Pcombined = 4.84 ×
10–​7) (Bei et al., 2010). This study also confirmed the importance of the Risk Factor Level of Evidence
HLA region in NPC by identifying significant loci in HLA-​A, HLA-​B/​C,
and HLA-​DQ/​DR (rs2860580, OR  =  0.58, Pcombined  =  4.88 × 10–​67; Epstein-​Barr virus infection Strong
rs2894207, OR = 0.61, 95% CI = 0.57–​0.66, Pcombined = 3.42 × 10–​33; Chinese-​style salt-preserved fish Strong
rs28421666, OR = 0.67, Pcombined = 2.49 × 10–​18). Family history Strong
The etiologic relevance of the HLA class I region was once again Certain HLA-​A and HLA-​B alleles Strong
highlighted by the results of a GWAS of 567 southern Chinese NPC Tobacco smoke Strong to moderate
cases and 476 controls for discovery and 346 cases and 629 controls Formaldehyde Moderate
for replication, in which significant associations were identified with Preserved foods other than salt-preserved Moderate
glutamine at HLA-​A amino acid site 62 (Pcombined = 7.38 × 10–​29), leu- fish
cine at HLA-​B amino acid site 116 (Pcombined = 6.51 × 10–​19), and tryp- Chronic respiratory tract conditions Moderate
tophan at HLA-​C amino acid site 156 (Pcombined  =  6.84 × 10–​8) (Tang Wood dust Moderate
et  al., 2012). High-​resolution HLA molecular typing in a combined HIV/​AIDS Moderate
1405 cases and 2650 controls revealed independent associations with Fresh fruits and vegetables (inverse) Moderate
HLA-​A*11:01 (P = 1.7 × 10–​19), HLA-​B*38:02 (P = 7.0 × 10–​11), HLA-​ CYP and GST polymorphisms Moderate
B*55:02 (P = 1.6 × 10–​10), and HLA-​C*12:02 (P = 4.3 × 10–​5), and with Other types of smoke Weak
HLA haplotypes including combinations of these alleles. Alcohol Weak
A fifth GWAS conducted with 184 Malaysian Chinese NPC cases Herbal medicines Weak
and 236 controls in the discovery phase and 260 cases and 245 con- Tea (inverse) Weak
trols in the replication phase also confirmed the involvement of HLA-​ Other HLA class I and class II alleles Weak
A in NPC risk, based on a significant association with rs3869062 DNA repair gene polymorphisms Weak
(Pcombined = 1.73 × 10–​9) (Chin et al., 2015). Fine mapping of the HLA-​A CYP = cytochrome P-​450; GST = glutathione-​S-​transferase; HIV/​AIDS = human
region revealed associations with SNPs in the HLA-​A antigen peptide immunodeficiency virus/​acquired immunodeficiency syndrome; HLA = human leukocyte
binding groove, the T-​cell receptor binding site, and other loci; these antigen.
 49

Nasopharyngeal Cancer 499

Box 26–1  EVIDENCE SUPPORTING THE INVOLVEMENT and newer studies are due to differences in research methods, inherent
OF EARLY-LIFE EXPOSURES IN NASOPHARYNGEAL exposure characteristics, human behavioral patterns that affect expo-
CARCINOMA DEVELOPMENT sure received, the prevalence of interacting exposures, histologic sub-
type distributions, or a combination of these or other factors.
1. Early peak and later plateau/​decline in age-​incidence curve
2. Etiologic role of Epstein-​Barr virus infection, which usually References
occurs in early life Albeck H, Bentzen J, Ockelmann HH, et al. 1993. Familial clusters of nasopha-
3. Possibly stronger association with consumption of Chinese-​style ryngeal carcinoma and salivary gland carcinomas in Greenland natives.
salt-preserved fish in infancy or childhood than in adulthood Cancer, 72(1), 196–​200. PMID: 8508406.
4. Modest or null association with immune suppression in adult- Armstrong RW, Imrey PB, Lye MS, et al. 1998. Nasopharyngeal carcinoma in
hood (e.g., HIV/​AIDS, solid organ transplantation), but strong Malaysian Chinese: salted fish and other dietary exposures. Int J Cancer,
77(2), 228–​235. PMID: 9650558.
and consistent association with genetic determinants of immune Armstrong RW, Imrey PB, Lye MS, et  al. 2000. Nasopharyngeal carcinoma
function (i.e., HLA alleles) in Malaysian Chinese: occupational exposures to particles, formaldehyde
5. Positive exposure–​response trend with longer duration of smok- and heat. Int J Epidemiol, 29(6), 991–​998. PMID: 11101539.
ing, implying earlier initiation Armstrong RW, Kutty MK, and Armstrong MJ. 1978. Self-​specific environ-
ments associated with nasopharyngeal carcinoma in Selangor, Malaysia.
HIV/​AIDS = human immunodeficiency virus/​acquired immunodeficiency Soc Sci Med, 12(3D–​4D), 149–​156. PMID: 734454.
syndrome; HLA = human leukocyte antigen. Arnold M, Wildeman MA, Visser O, et al. 2013. Lower mortality from nasopha-
ryngeal cancer in The Netherlands since 1970 with differential incidence
trends in histopathology. Oral Oncol, 49(3), 237–​243. PMID: 23089459.
Bachand AM, Mundt KA, Mundt DJ, and Montgomery RR. 2010.
Epidemiological studies of formaldehyde exposure and risk of leukemia
not explained by known risk factors such as family history, certain HLA and nasopharyngeal cancer: a meta-​analysis. Crit Rev Toxicol, 40(2), 85–​
alleles, Chinese-​ style salt-preserved fish consumption, and tobacco 100. PMID: 20085478.
smoking. It remains to be determined whether most NPC is due to as-​ Balfour HH, Jr., Odumade OA, Schmeling DO, et al. 2013. Behavioral, viro-
yet unidentified risk factors, heterogeneity in exposure, or a difference logic, and immunologic factors associated with acquisition and severity
in host response to a known cause such as EBV. Chance events contrib- of primary Epstein-​Barr virus infection in university students. J Infect
ute to risk, but would not be expected to affect international variation. Dis, 207(1), 80–​88. PMCID: PMC3523797.
Beane Freeman LE, Blair A, Lubin JH, et al. 2013. Mortality from solid tumors
A distinctive aspect of NPC that bears further research is the impact of
among workers in formaldehyde industries: an update of the NCI cohort.
early-​life exposures on risk. As summarized in Box 26–1, several lines of Am J Ind Med, 56(9), 1015–​1026. PMID: 23788167.
epidemiologic evidence point to the involvement of childhood exposures in Bei JX, Jia WH, and Zeng YX. 2012. Familial and large-​scale case-​control
NPC development. These include the relatively young average age at onset studies identify genes associated with nasopharyngeal carcinoma. Semin
in high-​incidence areas, along with the early incidence peak among ado- Cancer Biol, 22(2), 96–​106. PMID: 22313875.
lescents and young adults, at least in low-​incidence areas; the fundamental Bei JX, Li Y, Jia WH, et al. 2010. A genome-​wide association study of naso-
involvement of EBV infection, which first occurs during early childhood pharyngeal carcinoma identifies three new susceptibility loci. Nat Genet,
in the vast majority of people in high-​incidence areas; the stronger RRs 42(7), 599–​603. PMID: 20512145.
in association with consumption of salt-​preserved fish during infancy and Bei JX, Su WH, Ng CC, et al. 2016. A GWAS meta-​analysis and replication
study identifies a novel locus within CLPTM1L/​TERT associated with
childhood than adulthood in some studies; the positive association with
nasopharyngeal carcinoma in individuals of Chinese ancestry. Cancer
cigarette smoking, which generally begins in adolescence (although results Epidemiol Biomarkers Prev, 25(1), 188–​192. PMCID: PMC4713263.
for passive smoking during childhood are equivocal); and the weak or null Bendjemana K, Abdennebi M, Gara S, et al. 2006. [Genetic polymorphism of
associations with immune deficiency in adulthood, countered by strong gluthation-​S transferases and N-​acetyl transferases 2 and nasopharyn-
associations with genetically determined immune function. Given this col- geal carcinoma:  the Tunisia experience]. Bull Cancer, 93(3), 297–​302.
lective evidence, the search for novel risk factors for NPC may be most PMID: 16567317.
productive if focused on other early-​life (including heritable) exposures. Blair A, Stewart P, O’Berg M, et al. 1986. Mortality among industrial work-
Other potentially fruitful avenues for future epidemiologic research ers exposed to formaldehyde. J Natl Cancer Inst, 76(6), 1071–​1084.
on NPC include investigating how EBV sequence variation influ- PMID: 3458945.
Bray F, Haugen M, Moger TA, et  al. 2008. Age-​incidence curves of naso-
ences NPC risk, delving into the molecular pathways underlying the
pharyngeal carcinoma worldwide:  bimodality in low-​risk populations
involvement of HLA genes in NPC development, and exploring how and aetiologic implications. Cancer Epidemiol Biomarkers Prev, 17(9),
the local nasopharyngeal microbiome might affect the risk of malig- 2356–​2365. PMID: 18768504.
nancy. Measuring the levels of nitrosamines in various foods across Buell P. 1973. Race and place in the etiology of nasopharyngeal cancer: a study
different geographic regions or identifying a valid and accessible based on California death certificates. Int J Cancer, 11(2), 268–​272.
biomarker for nitrosamine consumption would improve the assess- PMID: 4790695.
ment of exposure to key dietary risk factors for NPC. Interactions Burt RD, Vaughan TL, McKnight B, et al. 1996. Associations between human
among environmental, viral, and genetic risk factors in NPC await leukocyte antigen type and nasopharyngeal carcinoma in Caucasians in
elucidation in large, probably pooled analyses. Shared and distinct the United States. Cancer Epidemiol Biomarkers Prev, 5(11), 879–​887.
Burt RD, Vaughan TL, Nisperos B, Swanson M, and Berwick M. 1994. A pro-
risk factors for keratinizing and non-​keratinizing NPC have not yet
tective association between the HLA-​A2 antigen and nasopharyngeal car-
been delineated clearly, and molecular differences between histologi- cinoma in US Caucasians. Int J Cancer, 56(4), 465–​467.
cal subtypes also are not well established. Few epidemiologic stud- Cai K, Wang Y, Zhao X, and Bao X. 2014. Association between the P53 codon
ies have been conducted of NPC in intermediate-​incidence regions. 72 polymorphism and nasopharyngeal cancer risk. Tumour Biol, 35(3),
In addition, host and environmental prognostic factors for NPC are 1891–​1897. PMID: 24114013.
largely unstudied. To reduce the global health burden due to NPC and Chan AT, Lo YM, Zee B, et  al. 2002. Plasma Epstein-​Barr virus DNA and
other EBV-​related malignancies, progress is needed in the develop- residual disease after radiotherapy for undifferentiated nasopharyngeal
ment of an EBV vaccine and/​or effective, most likely EBV-​based, carcinoma. J Natl Cancer Inst, 94(21), 1614–​1619. PMID: 12419787.
cancer screening strategies. Chan JKC, Bray F, McCarron P, et  al. 2005. Nasopharyngeal carcinoma.
In: Barnes L, Eveson J W, Reichart P, and Sidransky D (Eds.), World Health
As southern China and other areas with historically high NPC inci-
Organization Classification of Tumours:  Pathology & Genetics:  Head
dence rates undergo rapid economic development, the ensuing environ- and Neck Tumours (pp. 85–​97). Lyon: International Agency for Research
mental and lifestyle transitions among people living in these regions on Cancer (IARC).
will almost certainly lead to changes in the prevalence of risk factors Chan KC, Hung EC, Woo JK, et al. 2013. Early detection of nasopharyngeal
and their relative importance in the etiology of NPC. A challenge will carcinoma by plasma Epstein-​Barr virus DNA analysis in a surveillance
be to determine whether any discrepancies in results between older program. Cancer, 119(10), 1838–​1844. PMID: 23436393.
50

500 PART IV:  Cancers by Tissue of Origin


Chan SH, Day NE, Kunaratnam N, Chia KB, and Simons MJ. 1983. HLA Farrow DC, Vaughan TL, Berwick M, et  al. 1998. Diet and nasopharyn-
and nasopharyngeal carcinoma in Chinese: a further study. Int J Cancer, geal cancer in a low-​risk population. Int J Cancer, 78(6), 675–​679.
32(2), 171–​176. PMID: 9833758.
Chang CM, Yu KJ, Hsu WL, et  al. 2012. Correlates of anti-​EBV EBNA1 Feng BJ, Huang W, Shugart YY, et al. 2002. Genome-​wide scan for familial
IgA positivity among unaffected relatives from nasopharyn- nasopharyngeal carcinoma reveals evidence of linkage to chromosome 4.
geal carcinoma multiplex families. Br J Cancer, 106(1), 206–​ 209. Nat Genet, 31(4), 395–​399. PMID: 12118254.
PMCID: PMC3251852. Feng BJ, Jalbout M, Ayoub WB, et al. 2007. Dietary risk factors for nasopha-
Charriere M, Poirier S, Calmels S, et al. 1991. Microflora of the nasopharynx ryngeal carcinoma in Maghrebian countries. Int J Cancer, 121(7), 1550–​
in Caucasian and Maghrebian subjects with and without nasopharyngeal 1555. PMID: 17582611.
carcinoma. IARC Sci Publ, 105, 158–​161. PMID: 1855841. Feng BJ, Khyatti M, Ben-​Ayoub W, et al. 2009. Cannabis, tobacco and domes-
Chen CJ, Liang KY, Chang YS, et al. 1990. Multiple risk factors of nasopharyn- tic fumes intake are associated with nasopharyngeal carcinoma in North
geal carcinoma: Epstein-​Barr virus, malarial infection, cigarette smoking Africa. Br J Cancer, 101(7), 1207–​1212. PMCID: PMC2768108.
and familial tendency. Anticancer Res, 10(2B), 547–​553. PMID: 2161639. Ferlay J, Soerjomataram I, Ervik M, et  al. 2013. GLOBOCAN 2012 v1.0,
Chen L, Gallicchio L, Boyd-​Lindsley K, et al. 2009. Alcohol consumption and Cancer Incidence and Mortality Worldwide:  IARC CancerBase No. 11
the risk of nasopharyngeal carcinoma: a systematic review. Nutr Cancer, [Internet]. Lyon:  International Agency for Research on Cancer; 2013.
61(1), 1–​15. PMCID: PMC3072894. Available from: http://​globocan.iarc.fr. Accessed January 2015.
Cheng YJ, Chien YC, Hildesheim A, et  al. 2003. No association between Fogg MH, Wirth LJ, Posner M, and Wang F. 2009. Decreased EBNA-​1-​spe-
genetic polymorphisms of CYP1A1, GSTM1, GSTT1, GSTP1, NAT2, cific CD8+ T cells in patients with Epstein-​Barr virus-​associated naso-
and nasopharyngeal carcinoma in Taiwan. Cancer Epidemiol Biomarkers pharyngeal carcinoma. Proc Natl Acad Sci U S A, 106(9), 3318–​3323.
Prev, 12(2), 179–​180. PMID: 12582034. PMCID: PMC2651339.
Cheng YJ, Hildesheim A, Hsu MM et  al. 1999. Cigarette smoking, alcohol Forman D, Bray F, Brewster DH, et al. (Eds.). 2013. Cancer Incidence in Five
consumption and risk of nasopharyngeal carcinoma in Taiwan. Cancer Continents, Vol. X (electronic version) Lyon:  IARC. http://​ci5.iarc.fr.
Causes Control, 10(3), 201–​207. PMID: 10454065. Accessed January 2015.
Cheung F, Pang SW, Hioe F, et al. 1998. Nasopharyngeal carcinoma in situ: two Friborg J, Wohlfahrt J, Koch A, et al. 2005. Cancer susceptibility in nasopha-
cases of an emerging diagnostic entity. Cancer, 83(6), 1069–​ 1073. ryngeal carcinoma families: a population-​based cohort study. Cancer Res,
PMID: 9740069. 65(18), 8567–​8572. PMID: 16166338.
Chiang CJ, Chen YC, Chen CJ, et al. 2010. Cancer trends in Taiwan. Jpn J Clin Gallicchio L, Matanoski G, Tao XG, et  al. 2006. Adulthood consumption
Oncol, 40(10), 897–​904. PMID: 20495192. of preserved and nonpreserved vegetables and the risk of nasopharyn-
Chien YC, Chen JY, Liu MY, et al. 2001. Serologic markers of Epstein-​Barr geal carcinoma: a systematic review. Int J Cancer, 119(5), 1125–​1135.
virus infection and nasopharyngeal carcinoma in Taiwanese men. N Engl PMID: 16570274.
J Med, 345(26), 1877–​1882. PMID: 11756578. Garber JE, and Offit K. 2005. Hereditary cancer predisposition syndromes.
Chin YM, Mushiroda T, Takahashi A, et  al. 2015. HLA-​A SNPs and amino J Clin Oncol, 23(2), 276–​292. PMID: 15637391.
acid variants are associated with nasopharyngeal carcinoma in Malaysian Geser A, Charnay N, Day NE, de-​The G, and Ho HC. 1978. Environmental fac-
Chinese. Int J Cancer, 136(3), 678–​687. PMID: 24947555. tors in the etiology of nasopharyngeal carcinoma: report on a case-​control
Chung SD, Wu CS, Lin HC, and Hung SH. 2014. Association between aller- study in Hong Kong. IARC Sci Publ, 20, 213–​229. PMID: 730191.
gic rhinitis and nasopharyngeal carcinoma:  a population-​based study. Giannoudis A, Ergazaki M, Segas J, et  al. 1995. Detection of Epstein-​Barr
Laryngoscope, 124(8), 1744–​1749. PMID: 24272528. virus and human papillomavirus in nasopharyngeal carcinoma by the
Coggon D, Ntani G, Harris EC, and Palmer KT. 2014. Upper airway cancer, polymerase chain reaction technique. Cancer Lett, 89(2), 177–​ 181.
myeloid leukemia, and other cancers in a cohort of British chemical PMID: 7889526.
workers exposed to formaldehyde. Am J Epidemiol, 179(11), 1301–​1311. Goldsmith DB, West TM, and Morton R. 2002. HLA associations with naso-
PMID: 24714728. pharyngeal carcinoma in Southern Chinese:  a meta-​ analysis. Clin
Coghill AE, Hsu WL, Pfeiffer RM, et al. 2014. Epstein-​Barr virus serology as Otolaryngol Allied Sci, 27(1), 61–​67. PMID: 11903375.
a potential screening marker for nasopharyngeal carcinoma among high-​ Guo P, Huang ZL, Yu P, and Li K. 2012. Trends in cancer mortality in China: an
risk individuals from multiplex families in Taiwan. Cancer Epidemiol update. Ann Oncol, 23(10), 2755–​2762. PMID: 22492700.
Biomarkers Prev, 23(7), 1213–​1219. PMCID: PMC4082438. Guo X, Johnson RC, Deng H, et al. 2009. Evaluation of nonviral risk factors for
Cohen JI, Fauci AS, Varmus H, and Nabel GJ. 2011. Epstein-​Barr virus:  an nasopharyngeal carcinoma in a high-​risk population of Southern China.
important vaccine target for cancer prevention. Sci Transl Med, 3(107), Int J Cancer, 124(12), 2942–​2947. PMID: 19296536.
107fs107. PMCID: PMC3501269. Hamdi Cherif M, Serraino D, Mahnane A, et  al. 2014. Time trends of can-
Cohen JI, Mocarski ES, Raab-​Traub N, Corey L, and Nabel GJ. 2013. The cer incidence in Setif, Algeria, 1986–​2010: an observational study. BMC
need and challenges for development of an Epstein-​Barr virus vaccine. Cancer, 14, 637. PMCID: PMC4164767.
Vaccine, 31(Suppl 2), B194–​196. PMCID: PMC3636506. Hauptmann M, Lubin JH, Stewart PA, Hayes RB, and Blair A. 2004. Mortality
de-​The G, Lavoue MF, and Muenz L. 1978. Differences in EBV antibody from solid cancers among workers in formaldehyde industries. Am J
titres of patients with nasopharyngeal carcinoma originating from high, Epidemiol, 159(12), 1117–​1130. PMID: 15191929.
intermediate and low incidence areas. IARC Sci Publ, 20, 471–​ 481. Hauptmann M, Stewart PA, Lubin JH, et  al. 2009. Mortality from lympho-
PMID: 215529. hematopoietic malignancies and brain cancer among embalmers
Demers PA, Boffetta P, Kogevinas M, et  al. 1995. Pooled reanalysis of can- exposed to formaldehyde. J Natl Cancer Inst, 101(24), 1696–​ 1708.
cer mortality among five cohorts of workers in wood-​related industries. PMCID: PMC2794303.
Scand J Work Environ Health, 21(3), 179–​190. PMID: 7481605. Henderson BE, and Louie E. 1978. Discussion of risk factors for nasopharyn-
Deng W, Long L, Li JL, et al. 2014. Mortality of major cancers in Guangxi, geal carcinoma. IARC Sci Publ, 20, 251–​260. PMID: 730193.
China: sex, age and geographical differences from 1971 and 2005. Asian Henderson BE, Louie EW, Jing JS, and Alena B. 1977. Epstein-​Barr virus
Pac J Cancer Prev, 15(4), 1567–​1574. PMID: 24641369. and nasopharyngeal carcinoma: is there an etiologic relationship? J Natl
Dogan S, Hedberg ML, Ferris RL, et  al. 2014. Human papillomavirus and Cancer Inst, 59(5), 1393–​1395. PMID: 198562.
Epstein-​Barr virus in nasopharyngeal carcinoma in a low-​incidence popu- Hildesheim A. 2013. Invited commentary: Epstein-​Barr virus-​based screening
lation. Head Neck, 36(4), 511–​516. PMID: 23780921. for the early detection of nasopharyngeal carcinoma: a new frontier. Am J
Dou H, Hu D, Lam C, et al. 2014. Retrospective analysis of results of treatment Epidemiol, 177(3), 251–​253. PMCID: PMC3626052.
for nasopharyngeal carcinoma in Macao. Chin J Cancer Res, 26(2), 148–​ Hildesheim A, Anderson LM, Chen CJ, et al. 1997. CYP2E1 genetic polymor-
158. PMCID: PMC4000909. phisms and risk of nasopharyngeal carcinoma in Taiwan. J Natl Cancer
Edge S, Byrd DR, Compton CC, et  al (Eds.). 2010. AJCC Cancer Staging Inst, 89(16), 1207–​1212. PMID: 9274915.
Manual, 7th ed. New York: Springer. Hildesheim A, Apple RJ, Chen CJ, et al. 2002. Association of HLA class I and
Edgren G, Liang L, Adami HO, and Chang ET. 2012. Enigmatic sex disparities II alleles and extended haplotypes with nasopharyngeal carcinoma in
in cancer incidence. Eur J Epidemiol, 27(3), 187–​196. PMID: 22212865. Taiwan. J Natl Cancer Inst, 94(23), 1780–​1789.
Engels EA, Pfeiffer RM, Fraumeni JF, Jr, et al. 2011. Spectrum of cancer risk Hildesheim A, Chen CJ, Caporaso NE, et  al. 1995. Cytochrome P4502E1
among US solid organ transplant recipients. JAMA, 306(17), 1891–​1901. genetic polymorphisms and risk of nasopharyngeal carcinoma:  results
PMCID: PMC3310893. from a case-​ control study conducted in Taiwan. Cancer Epidemiol
Fachiroh J, Sangrajrang S, Johansson M, et al. 2012. Tobacco consumption and Biomarkers Prev, 4(6), 607–​610. PMID: 8547826.
genetic susceptibility to nasopharyngeal carcinoma (NPC) in Thailand. Hildesheim A, Dosemeci M, Chan CC, et  al. 2001. Occupational expo-
Cancer Causes Control, 23(12), 1995–​2002. PMID: 23085811. sure to wood, formaldehyde, and solvents and risk of nasopharyngeal
 501

Nasopharyngeal Cancer 501
carcinoma. Cancer Epidemiol Biomarkers Prev, 10(11), 1145–​ 1153. IARC. 2012d. IARC Monographs on the Evaluation of Carcinogenic Risks to
PMID: 11700262. Humans. Volume 100: A Review of Human Carcinogens. Part E: Personal
Hildesheim A, and Wang CP. 2012. Genetic predisposition factors and naso- Habits and Indoor Combustions. Lyon: International Agency for Research
pharyngeal carcinoma risk:  a review of epidemiological association on Cancer (IARC).
studies, 2000–​2011:  Rosetta Stone for NPC:  genetics, viral infection, Jeannel D, Ghnassia M, Hubert A, et al. 1993. Increased risk of nasopharyngeal
and other environmental factors. Semin Cancer Biol, 22(2), 107–​116. carcinoma among males of French origin born in Maghreb (north Africa).
PMCID: PMC3296903. Int J Cancer, 54(4), 536–​539. PMID: 8514445.
Hildesheim A, West S, DeVeyra E, et al. 1992. Herbal medicine use, Epstein-​ Jeannel D, Hubert A, de Vathaire F, et al. 1990. Diet, living conditions and naso-
Barr virus, and risk of nasopharyngeal carcinoma. Cancer Res, 52(11), pharyngeal carcinoma in Tunisia: a case-​control study. Int J Cancer, 46(3),
3048–​3051. 421–​425.
Ho HC. 1976. Epidemiology of nasopharyngeal carcinoma. In:  Hirayama T Ji MF, Huang QH, Yu X, et al. 2014. Evaluation of plasma Epstein-​Barr virus
(Ed.), Cancer Asia (pp. 49–​61). Baltimore, MD: University Park Press. DNA load to distinguish nasopharyngeal carcinoma patients from healthy
Ho JHC. 1967. Nasopharyngeal carcinoma in Hong Kong. In: Muir CS, and high-​risk populations in Southern China. Cancer, 120(9), 1353–​1360.
Shanmugaratnam K (Eds.), Cancer of the Nasopharynx (pp. 58–​63). PMID: 24477877.
Copenhagen: Munksgaard. Ji X, Zhang W, Xie C, et al. 2011. Nasopharyngeal carcinoma risk by histologic
Ho JHC, Huang DP, and Fong YY. 1978. Salted fish and nasopharyngeal carci- type in central China: impact of smoking, alcohol and family history. Int J
noma in southern Chinese. Lancet, 2(8090), 626. PMID: 80545. Cancer, 129(3), 724–​732. PMID: 20878958.
Hording U, Nielsen HW, Daugaard S, and Albeck H. 1994. Human papillo- Jia WH, Collins A, Zeng YX, et  al. 2005. Complex segregation analysis of
mavirus types 11 and 16 detected in nasopharyngeal carcinomas by nasopharyngeal carcinoma in Guangdong, China:  evidence for a multi-
the polymerase chain reaction. Laryngoscope, 104(1 Pt 1), 99–​ 102. factorial mode of inheritance (complex segregation analysis of NPC in
PMID: 8295466. China). Eur J Hum Genet, 13(2), 248–​252. PMID: 15483644.
Hsu WL, Chen JY, Chien YC, et al. 2009. Independent effect of EBV and cig- Jia WH, Feng BJ, Xu ZL, et  al. 2004. Familial risk and clustering of naso-
arette smoking on nasopharyngeal carcinoma: a 20-​year follow-​up study pharyngeal carcinoma in Guangdong, China. Cancer, 101(2), 363–​369.
on 9,622 males without family history in Taiwan. Cancer Epidemiol PMID: 15241835.
Biomarkers Prev, 18(4), 1218–​1226. PMID: 19336547. Jia WH, Luo XY, Feng BJ, et al. 2010. Traditional Cantonese diet and nasopha-
Hsu WL, Pan WH, Chien YC, et al. 2012a. Lowered risk of nasopharyngeal car- ryngeal carcinoma risk: a large-​scale case-​control study in Guangdong,
cinoma and intake of plant vitamin, fresh fish, green tea and coffee: a case-​ China. BMC Cancer, 10, 446. PMCID: PMC2931495.
control study in Taiwan. PLoS One, 7(7), e41779. PMCID: PMC3407060. Jia WH, Pan QH, Qin HD, et al. 2009. A case-​control and a family-​based asso-
Hsu WL, Tse KP, Liang S et al. 2012b. Evaluation of human leukocyte antigen-​A ciation study revealing an association between CYP2E1 polymorphisms
(HLA-​A), other non-​HLA markers on chromosome 6p21 and risk of naso- and nasopharyngeal carcinoma risk in Cantonese. Carcinogenesis,
pharyngeal carcinoma. PLoS One, 7(8), e42767. PMCID: PMC3413673. 30(12), 2031–​2036. PMCID: PMC2792314.
Hsu WL, Yu KJ, Chien YC, et  al. 2011. Familial tendency and risk of naso- Jiang JH, Jia WH, Chen HK, et al. 2004. Genetic polymorphisms of CYP2A13
pharyngeal carcinoma in Taiwan:  effects of covariates on risk. Am J and its relationship to nasopharyngeal carcinoma in the Cantonese popu-
Epidemiol, 173(3), 292–​299. PMCID: PMC3105265. lation. J Transl Med, 2(1), 24. PMCID: PMC455696.
Hu LF, Qiu QH, Fu SM et al. 2008. A genome-​wide scan suggests a suscepti- Jiang RC, Qin HD, Zeng MS, et al. 2006. A functional variant in the transcrip-
bility locus on 5p 13 for nasopharyngeal carcinoma. Eur J Hum Genet, tional regulatory region of gene LOC344967 cosegregates with disease
16(3), 343–​349. PMID: 18197201. phenotype in familial nasopharyngeal carcinoma. Cancer Res, 66(2),
Huang GL, Guo HQ, Yu CY, et al. 2011. XRCC1 polymorphisms and risk of 693–​700. PMID: 16423998.
nasopharyngeal carcinoma:  a meta-​analysis. Asian Pac J Cancer Prev, Jin B, Dong P, Li K, Shen B, and Xie J. 2014. Meta-​analysis of the association
12(9), 2329–​2333. PMID: 22296379. between GSTT1 null genotype and risk of nasopharyngeal carcinoma in
Huang WY, Lin CC, Jen YM, et  al. 2012. Association between adult otitis Chinese. Tumour Biol, 35(1), 345–​349. PMID: 23949879.
media and nasopharyngeal cancer: a nationwide population-​based cohort Kangro HO, Osman HK, Lau YL, et  al. 1994. Seroprevalence of antibodies
study. Radiother Oncol, 104(3), 338–​342. PMID: 22981611. to human herpesviruses in England and Hong Kong. J Med Virol, 43(1),
Huang X, Hepkema B, Nolte I, et  al. 2012. HLA-​ A*02:07 is a protec- 91–​96. PMID: 8083655.
tive allele for EBV negative and a susceptibility allele for EBV posi- Klein G, Klein E, and Kashuba E. 2010. Interaction of Epstein-​Barr virus
tive classical Hodgkin lymphoma in China. PLoS One, 7(2), e31865. (EBV) with human B-​ lymphocytes. Biochem Biophys Res Commun,
PMCID: PMC3280205. 396(1), 67–​73. PMID: 20494113.
Hung SH, Chen PY, Lin HC, Ting J, and Chung SD. 2013. Association of rhi- Kongruttanachok N, Sukdikul S, Setavarin S, et al. 2001. Cytochrome P450
nosinusitis with nasopharyngeal carcinoma:  a population-​based study. 2E1 polymorphism and nasopharyngeal carcinoma development in
Laryngoscope. PMID: 24114992. Thailand: a correlative study. BMC Cancer, 1(1), 4. PMCID: PMC32196.
Hutajulu SH, Ng N, Jati BR, et al. 2012. Seroreactivity against Epstein-​Barr Kwok H, Tong AH, Lin CH, et  al. 2012. Genomic sequencing and com-
virus (EBV) among first-​degree relatives of sporadic EBV-​associated parative analysis of Epstein-​ Barr virus genome isolated from pri-
nasopharyngeal carcinoma in Indonesia. J Med Virol, 84(5), 768–​776. mary nasopharyngeal carcinoma biopsy. PLoS One, 7(5), e36939.
PMID: 22431025. PMCID: PMC3349645.
IARC. 1993. Some naturally occurring substances:  food Items and constitu- Kwok H, Wu CW, Palser AL, et al. 2014. Genomic diversity of Epstein-​Barr
ents, heterocyclic aromatic amines and mycotoxins. IARC Monogr Eval virus genomes isolated from primary nasopharyngeal carcinoma biopsy
Carcinog Risks Hum, 56, 1–​599. samples. J Virol, 88(18), 10662–​10672. PMCID: PMC4178900.
IARC. 1995. Wood dust and formaldehyde. IARC Monogr Eval Carcinog Risks Lanier A, Bender T, Talbot M, et al. 1980. Nasopharyngeal carcinoma in Alaskan
Hum, 62, 1–​405. Eskimos Indians, and Aleuts: a review of cases and study of Epstein-​Barr
IARC. 1997. Epstein-​Barr virus and Kaposi’s herpesvirus/​human herpesvirus virus, HLA, and environmental risk factors. Cancer, 46(9), 2100–​2106.
8. IARC Monogr Eval Carcinog Risks Hum, 70, 1–​524. PMID: 6253051.
IARC. 2004a. Betel-​quid and areca-​nut chewing and some areca-​nut-​derived Lau HY, Leung CM, Chan YH, et al. 2013. Secular trends of salted fish con-
nitrosamines. IARC Monogr Eval Carcinog Risks Hum, 85, 1–​334. sumption and nasopharyngeal carcinoma:  a multi-​ jurisdiction eco-
IARC. 2004b. Tobacco smoke and involuntary smoking. IARC Monogr Eval logical study in 8 regions from 3 continents. BMC Cancer, 13, 298.
Carcinog Risks Hum, 83, 1–​1452. PMCID: PMC3729410.
IARC. 2006. Formaldehyde, 2-​butoxyethanol and 1-​tert-​butoxypropan-​2-​ol. Lee AW, Sze WM, Au JS, et  al. 2005. Treatment results for nasopharyngeal
IARC Monogr Eval Carcinog Risks Hum, 88, 1–​478. carcinoma in the modern era:  the Hong Kong experience. Int J Radiat
IARC. 2012a. Arsenic, metals, fibres, and dusts. Volume 100 C: A review of Oncol Biol Phys, 61(4), 1107–​1116. PMID: 15752890.
human carcinogens. IARC Monogr Eval Carcinog Risks Hum, 100(Pt Lee E, Huang Y, Zhao B, et  al. 1998. Genetic polymorphism of conjugating
C), 1–​501. enzymes and cancer risk: GSTM1, GSTT1, NAT1 and NAT2. J Toxicol
IARC. 2012b. Biological agents. Volume 100 B: A review of human carcino- Sci, 23(Suppl 2), 140–​142. PMID: 9760451.
gens. IARC Monogr Eval Carcinog Risks Hum, 100(Pt B), 1–​475. Lee SP, Tierney RJ, Thomas WA, Brooks JM, and Rickinson AB. 1997.
IARC. 2012c. Chemical agents and related occupations. Volume 100 Conserved CTL epitopes within EBV latent membrane protein 2: a poten-
F: A review of human carcinogens. IARC Monogr Eval Carcinog Risks tial target for CTL-​based tumor therapy. J Immunol, 158(7), 3325–​3334.
Hum, 100(Pt F), 1–​599. PMID: 9120290.
502

502 PART IV:  Cancers by Tissue of Origin


Leung TW, Tung SY, Sze WK, et al. 2005. Treatment results of 1070 patients Pak MW, To KF, Lo YM, et  al. 2002. Nasopharyngeal carcinoma in
with nasopharyngeal carcinoma: an analysis of survival and failure pat- situ (NPCIS):  pathologic and clinical perspectives. Head Neck, 24(11),
terns. Head Neck, 27(7), 555–​565. PMID: 15880410. 989–​995. PMID: 12410533.
Li CC, Yu MC, and Henderson BE. 1985. Some epidemiologic observations of Parkin DM, and Iscovich J. 1997. Risk of cancer in migrants and their descen-
nasopharyngeal carcinoma in Guangdong, People’s Republic of China. dants in Israel:  II. Carcinomas and germ-​cell tumours. Int J Cancer,
Natl Cancer Inst Monogr, 69, 49–​52. PMID: 3834344. 70(6), 654–​660. PMID: 9096645.
Li K, Lin GZ, Shen JC, and Zhou Q. 2014. Time trends of nasopharyngeal carci- Pathmanathan R, Prasad U, Sadler R, Flynn K, and Raab-​Traub N. 1995.
noma in urban Guangzhou over a 12-​year period (2000–​2011): declines in Clonal proliferations of cells infected with Epstein-​Barr virus in preinva-
both incidence and mortality. Asian Pac J Cancer Prev, 15(22), 9899–​9903. sive lesions related to nasopharyngeal carcinoma. N Engl J Med, 333(11),
PMID: 25520125. 693–​698. PMID: 7637746.
Li W, Ray RM, Gao DL, et al. 2006. Occupational risk factors for nasopha- Phua CE, Tan BS, Yong TK, and Govindasamy M. 2011. Retrospective analysis
ryngeal cancer among female textile workers in Shanghai, China. Occup of results of treatment for nasopharyngeal carcinoma in Penang General
Environ Med, 63(1), 39–​44. PMCID: PMC2078032. Hospital from 2001–​2005. Asian Pac J Cancer Prev, 12(12), 3197–​3200.
Lin JC, Wang WY, Chen KY, et al. 2004. Quantification of plasma Epstein-​Barr PMID: 22471453.
virus DNA in patients with advanced nasopharyngeal carcinoma. N Engl Phua VCE, Loo WH, Yusof MM, et al. 2013. Treatment outcome for nasopha-
J Med, 350(24), 2461–​2470. PMID: 15190138. ryngeal carcinoma in University Malaya Medical Centre from 2004–​2008.
Lin KT, Huang WY, Lin CC et  al. 2015. Subsequent risk of nasopharyngeal Asian Pac J Cancer Prev, 14(8), 4567–​4570. PMID: 24083703.
carcinoma among patients with allergic rhinitis: a nationwide population-​ Pickard A, Chen CJ, Diehl SR, et  al. 2004. Epstein-​Barr virus seroreactivity
based cohort study. Head Neck, 37(3), 413–​417. PMID: 24435940. among unaffected individuals within high-​risk nasopharyngeal carcinoma
Lin TM, Yang CS, Chiou JF et al. 1977. Antibodies to Epstein-​Barr virus capsid families in Taiwan. Int J Cancer, 111(1), 117–​123. PMID: 15185352.
antigen and early antigen in nasopharyngeal carcinoma and comparison Polesel J, Franceschi S, Talamini R, et al. 2011. Tobacco smoking, alcohol drink-
groups. Am J Epidemiol, 106(4), 336–​339. PMID: 199059. ing, and the risk of different histological types of nasopharyngeal cancer
Lin TM, Yang CS, Tu SM, et al. 1979. Interaction of factors associated with in a low-​risk population. Oral Oncol, 47(6), 541–​545. PMID: 21478046.
cancer of the nasopharynx. Cancer, 44(4), 1419–​1423. PMID: 227565. Polesel J, Serraino D, Negri E, et al. 2013. Consumption of fruit, vegetables,
Lin Z, Khong B, Kwok S, et al. 2014. Human papillomavirus 16 detected in and other food groups and the risk of nasopharyngeal carcinoma. Cancer
nasopharyngeal carcinomas in white Americans but not in endemic Causes Control, 24(6), 1157–​1165. PMID: 23535867.
Southern Chinese patients. Head Neck, 36(5), 709–​714. PMID: 23616441. Qin HD, Jia WH, Zhang LL, et  al. 2011a. Elevated Epstein-​Barr virus sero-
Liu YT, Dai JJ, Xu CH et al. 2012. Greater intake of fruit and vegetables is associ- reactivity among unaffected members of families with nasopharyngeal
ated with lower risk of nasopharyngeal carcinoma in Chinese adults: a case-​ carcinoma. J Med Virol, 83(10), 1792–​1798. PMID: 21837797.
control study. Cancer Causes Control, 23(4), 589–​599. PMID: 22392078. Qin HD, Shugart YY, Bei JX, et al. 2011b. Comprehensive pathway-​based asso-
Liu Z, Ji MF, Huang QH, et al. 2013. Two Epstein-​Barr virus-​related serologic ciation study of DNA repair gene variants and the risk of nasopharyngeal
antibody tests in nasopharyngeal carcinoma screening:  results from the carcinoma. Cancer Res, 71(8), 3000–​3008. PMCID: PMC3383064.
initial phase of a cluster randomized controlled trial in Southern China. Raab-​Traub N, and Flynn K. 1986. The structure of the termini of the Epstein-​
Am J Epidemiol, 177(3), 242–​250. PMID: 23255783. Barr virus as a marker of clonal cellular proliferation. Cell, 47(6),
Lo YM, Chan AT, Chan LY, et  al. 2000. Molecular prognostication of naso- 883–​889. PMID: 3022942.
pharyngeal carcinoma by quantitative analysis of circulating Epstein-​Barr Rezaei N, Hedayat M, Aghamohammadi A, and Nichols KE. 2011. Primary
virus DNA. Cancer Res, 60(24), 6878–​6881. PMID: 11156384. immunodeficiency diseases associated with increased susceptibility
Lu SJ, Day NE, Degos L, et al. 1990. Linkage of a nasopharyngeal carcinoma to viral infections and malignancies. J Allergy Clin Immunol, 127(6),
susceptibility locus to the HLA region. Nature, 346(6283), 470–​471. 1329–​1341 e1322; quiz 1342–​1323. PMID: 21514636.
PMID: 2377207. Ruan HL, Xu FH, Liu WS, et al. 2010. Alcohol and tea consumption in relation
McLaughlin JK, and Tarone RE. 2014. Mortality from solid tumors in the to the risk of nasopharyngeal carcinoma in Guangdong, China. Front Med
updated NCI formaldehyde cohort. Am J Ind Med, 57(4), 486–​ 487. China, 4(4), 448–​456. PMID: 21110141.
PMID: 24501017. Salavoura K, Kolialexi A, Tsangaris G, and Mavrou A. 2008. Development
Meyers AR, Pinkerton LE, and Hein MJ. 2013. Cohort mortality study of of cancer in patients with primary immunodeficiencies. Anticancer Res,
garment industry workers exposed to formaldehyde: update and inter- 28(2B), 1263–​1269. PMID: 18505064.
nal comparisons. Am J Ind Med, 56(9), 1027–​1039. PMID: 23788124. SEER. 2015. Surveillance, Epidemiology, and End Results (SEER) Program (www.
Mousavi SM, and Hemminki K. 2015. Cancer incidence, trends, and survival seer.cancer.gov/​data/​seerstat/​nov2014/​) SEER*Stat Database:  Incidence—​
among immigrants to Sweden: a population-​based study. Eur J Cancer SEER 18 Regs Research Data + Hurricane Katrina Impacted Louisiana Cases,
Prev, 24(Suppl 1), S1–​S63. PMID: 25599121. Nov 2014 Sub (2000–​2012) <Katrina/​Rita Population Adjustment>—​Linked
Murthy AK, Kumar V, and Suresh KP. 2013. Meta-​analysis of GSTM1 and to County Attributes—​Total U.S., 1969–​2013 Counties, National Cancer
GSTT1 polymorphisms and risk of nasopharyngeal cancer. Asian Pac J Institute, DCCPS, Surveillance Research Program, Surveillance Systems
Cancer Prev, 14(3), 1697–​1701. PMID: 23679259. Branch, released April 2015, based on the November 2014 submission.
National Registry of Diseases Office. 2009. Trends in Cancer Incidence in Shanmugaratnam K. 1991. Histological Typing of Tumours of the Upper
Singapore 1968–​2007. Singapore: Singapore Cancer Registry. Respiratory Tract and Ear, 2nd ed. Berlin: Springer-​Verlag.
National Registry of Diseases Office. 2013. Singapore Cancer Registry Interim Shanmugaratnam K, and Sobin LH. 1978. Histological Typing of Upper
Annual Registry Report: Trends in Cancer Incidence in Singapore 2008–​ Respiratory Tract Tumours. Geneva: World Health Organization.
2012. Singapore: Singapore Cancer Registry. Shanmugaratnam K, Tye CY, Goh EH, and Chia KB. 1978. Etiological factors
Nesic V, Sipetic S, Vlajinac H, Stosic-​Divjak S, and Jesic S. 2010. Risk factors in nasopharyngeal carcinoma: a hospital-​based, retrospective, case-​con-
for the occurrence of undifferentiated carcinoma of nasopharyngeal type: a trol, questionnaire study. IARC Sci Publ, 20, 199–​212. PMID: 730190.
case-​control study. Srp Arh Celok Lek, 138(1–​2), 6–​10. PMID: 20422906. Shebl FM, Bhatia K, and Engels EA. 2010. Salivary gland and nasopha-
Ng TP. 1986. A case-​referent study of cancer of the nasal cavity and sinuses in ryngeal cancers in individuals with acquired immunodeficiency
Hong Kong. Int J Epidemiol, 15(2), 171–​175. PMID: 3721678. syndrome in United States. Int J Cancer, 126(10), 2503–​ 2508.
Ng WT, Choi CW, Lee MC, et al. 2009. Familial nasopharyngeal carcinoma PMCID: PMC2847048.
in Hong Kong: epidemiology and implication in screening. Fam Cancer, Siew SS, Kauppinen T, Kyyronen P, Heikkila P, and Pukkala E. 2012.
8(2), 103–​108. PMID: 18726711. Occupational exposure to wood dust and formaldehyde and risk of nasal,
Niedobitek G. 2000. Epstein-​Barr virus infection in the pathogenesis of nasopha- nasopharyngeal, and lung cancer among Finnish men. Cancer Manag
ryngeal carcinoma. Mol Pathol, 53(5), 248–​254. PMCID: PMC1186977. Res, 4, 223–​232. PMCID: PMC3421471.
Ning JP, Yu MC, Wang QS, and Henderson BE. 1990. Consumption of salted Simons MJ, Chan SH, Wee GB et  al. 1978. Nasopharyngeal carcinoma and
fish and other risk factors for nasopharyngeal carcinoma (NPC) in histocompatibility antigens. IARC Sci Publ, 20, 271–​282. PMID: 730194.
Tianjin, a low-​risk region for NPC in the People’s Republic of China. Simons MJ, Wee GB, Chan SH, et al. 1975. Immunogenetic aspects of naso-
J Natl Cancer Inst, 82(4), 291–​296. PMID: 2299678. pharyngeal carcinoma (NPC) III. HL-​a type as a genetic marker of NPC
Old LJ, Boyse EA, Oettgen HF, et al. 1966. Precipitating antibody in human predisposition to test the hypothesis that Epstein-​Barr virus is an etiologi-
serum to an antigen present in cultured Burkitt’s lymphoma cells. Proc cal factor in NPC. IARC Sci Publ, 11(Pt 2), 249–​258. PMID: 191368.
Natl Acad Sci U S A, 56(6), 1699–​1704. PMCID: PMC220158. Simons MJ, Wee GB, Day NE, et  al. 1974. Immunogenetic aspects of
Pak MW, To KF, Lee JC, Liang EY, and van Hasselt CA. 2005. In vivo real-​time nasopharyngeal carcinoma:  I.  Differences in HL-​ A antigen profiles
diagnosis of nasopharyngeal carcinoma in situ by contact rhinoscopy. between patients and control groups. Int J Cancer, 13(1), 122–​ 134.
Head Neck, 27(11), 1008–​1013. PMID: 16134185. PMID: 4131857.
 503

Nasopharyngeal Cancer 503
Singhi AD, Califano J, and Westra WH. 2012. High-​risk human papillomavirus in Xie SH, Gong J, Yang NN, et al. 2012. Time trends and age-​period-​cohort
nasopharyngeal carcinoma. Head Neck, 34(2), 213–​218. PMID: 21484924. analyses on incidence rates of nasopharyngeal carcinoma during
Sobin LH, Gospodarowicz MK, and Wittekind C (Eds.). 2009. TNM 1993–​2007 in Wuhan, China. Cancer Epidemiol, 36(1), 8–​10. PMID:
Classification of Malignant Tumours, 7th ed. New York: Wiley-​Blackwell. 21846597.
Sriamporn S, Vatanasapt V, Pisani P, Yongchaiyudha S, and Rungpitarangsri V. Xiong G, Zhang B, Huang MY, et al. 2014. Epstein-​Barr virus (EBV) infec-
1992. Environmental risk factors for nasopharyngeal carcinoma: a case-​ tion in Chinese children: a retrospective study of age-​specific prevalence.
control study in northeastern Thailand. Cancer Epidemiol Biomarkers PLoS One, 9(6), e99857. PMCID: PMC4051769.
Prev, 1(5), 345–​348. PMID: 1305465. Xiong W, Zeng ZY, Xia JH, et al. 2004. A susceptibility locus at chromosome
Stellman SD, Demers PA, Colin D, and Boffetta P. 1998. Cancer mortality and 3p21 linked to familial nasopharyngeal carcinoma. Cancer Res, 64(6),
wood dust exposure among participants in the American Cancer Society 1972–​1974. PMID: 15026332.
Cancer Prevention Study-​II (CPS-​II). Am J Ind Med, 34(3), 229–​237. Xu FH, Xiong D, Xu YF, et al. 2012. An epidemiological and molecular study
PMID: 9698991. of the relationship between smoking, risk of nasopharyngeal carcinoma,
Su WH, Hildesheim A, and Chang YS. 2013. Human leukocyte antigens and and Epstein-​Barr virus activation. J Natl Cancer Inst, 104(18), 1396–​
epstein-​barr virus-​associated nasopharyngeal carcinoma: old associations 1410. PMID: 22972969.
offer new clues into the role of immunity in infection-​associated cancers. Xue WQ, Qin HD, Ruan HL, Shugart YY, and Jia WH. 2013. Quantitative asso-
Front Oncol, 3, 299. PMCID: PMC3856645. ciation of tobacco smoking with the risk of nasopharyngeal carcinoma: a
Sun LM, Epplein M, Li CI, Vaughan TL, and Weiss NS. 2005. Trends in the comprehensive meta-​analysis of studies conducted between 1979 and
incidence rates of nasopharyngeal carcinoma among Chinese Americans 2011. Am J Epidemiol, 178(3), 325–​338. PMCID: PMC3727336.
living in Los Angeles County and the San Francisco metropolitan area, Yang XR, Diehl S, Pfeiffer R, et al. 2005. Evaluation of risk factors for naso-
1992–​2002. Am J Epidemiol, 162(12), 1174–​1178. PMID: 16282240. pharyngeal carcinoma in high-​ risk nasopharyngeal carcinoma fami-
Sun X, Su S, Chen C, et  al. 2013. Long-​term outcomes of intensity-​modu- lies in Taiwan. Cancer Epidemiol Biomarkers Prev, 14(4), 900–​905.
lated radiotherapy for 868 patients with nasopharyngeal carcinoma: An PMID: 15826929.
analysis of survival and treatment toxicities. Radiother Oncol. PMID: Yu KJ, Hsu WL, Chiang CJ, et al. 2009. Cancer patterns in nasopharyngeal car-
24231245. cinoma multiplex families in Taiwan. Int J Cancer, 124(7), 1622–​1625.
Sun ZF, Zhang J, Xu HM, Wang GL, and Dong P. 2012. Association between PMCID: PMC2796215.
GSTM1 polymorphism and nasopharyngeal cancer susceptibility: a meta-​ Yu MC, Garabrant DH, Huang TB, and Henderson BE. 1990. Occupational
analysis. Asian Pac J Cancer Prev, 13(11), 5817–​5821. PMID: 23317262. and other non-​ dietary risk factors for nasopharyngeal carcinoma in
Tang M, Lautenberger JA, Gao X, et  al. 2012. The principal genetic deter- Guangzhou, China. Int J Cancer, 45(6), 1033–​1039. PMID: 2351484.
minants for nasopharyngeal carcinoma in China involve the HLA Yu MC, Ho JH, Lai SH, and Henderson BE. 1986. Cantonese-​style salted fish
class  I  antigen recognition groove. PLoS Genet, 8(11), e1003103. as a cause of nasopharyngeal carcinoma: report of a case-​control study in
PMCID: PMC3510037. Hong Kong. Cancer Res, 46(2), 956–​961. PMID: 3940655.
Tiwawech D, Srivatanakul P, Karalak A, and Ishida T. 2006. Cytochrome P450 Yu MC, Huang TB, and Henderson BE. 1989. Diet and nasopharyngeal car-
2A6 polymorphism in nasopharyngeal carcinoma. Cancer Lett, 241(1), cinoma: a case-​control study in Guangzhou, China. Int J Cancer, 43(6),
135–​141. PMID: 16377082. 1077–​1082. PMID: 2732001.
Tse KP, Su WH, Chang KP, et  al. 2009. Genome-​wide association study Yu MC, Mo CC, Chong WX, Yeh FS, and Henderson BE. 1988. Preserved
reveals multiple nasopharyngeal carcinoma-​associated loci within the foods and nasopharyngeal carcinoma:  a case-​control study in Guangxi,
HLA region at chromosome 6p21.3. Am J Hum Genet, 85(2), 194–​203. China. Cancer Res, 48(7), 1954–​1959. PMID: 3349469.
PMCID: PMC2725267. Yu WM, and Hussain SS. 2009. Incidence of nasopharyngeal carcinoma
Vaughan TL, and Davis S. 1991. Wood dust exposure and squamous cell can- in Chinese immigrants, compared with Chinese in China and South
cers of the upper respiratory tract. Am J Epidemiol, 133(6), 560–​564. East Asia: review. J Laryngol Otol, 123(10), 1067–​1074. PMID:
PMID: 2006642. 19486543.
Vaughan TL, Shapiro JA, Burt RD, et  al. 1996. Nasopharyngeal cancer in a Yuan JM, Wang XL, Xiang YB, et al. 2000a. Non-​dietary risk factors for naso-
low-​risk population:  defining risk factors by histological type. Cancer pharyngeal carcinoma in Shanghai, China. Int J Cancer, 85(3), 364–​369.
Epidemiol Biomarkers Prev, 5(8), 587–​593. PMID: 8824359. PMID: 10652428.
Vaughan TL, Stewart PA, Teschke K, et  al. 2000. Occupational exposure to Yuan JM, Wang XL, Xiang YB, et al. 2000b. Preserved foods in relation to risk
formaldehyde and wood dust and nasopharyngeal carcinoma. Occup of nasopharyngeal carcinoma in Shanghai, China. Int J Cancer, 85(3),
Environ Med, 57(6), 376–​384. PMID: 10810126. 358–​363. PMID: 10652427.
Wang HY, Sun BY, Zhu ZH, et al. 2011. Eight-​signature classifier for prediction Zeng Y, Zhang LG, Li HY, et al. 1982. Serological mass survey for early detec-
of nasopharyngeal carcinoma survival. J Clin Oncol, 29(34), 4516–​4525. tion of nasopharyngeal carcinoma in Wuzhou City, China. Int J Cancer,
PMID: 22025164. 29(2), 139–​141. PMID: 6895884.
Wang WY, Twu CW, Chen HH, et  al. 2013. Long-​term survival analysis of Zeng Y, Zhang LG, Wu YC, et  al. 1985. Prospective studies on nasopha-
nasopharyngeal carcinoma by plasma Epstein-​Barr virus DNA levels. ryngeal carcinoma in Epstein-​ Barr virus IgA/​ VCA antibody-​ posi-
Cancer, 119(5), 963–​970. PMID: 23065693. tive persons in Wuzhou City, China. Int J Cancer, 36(5), 545–​547.
Wang Y, Zhang Y, and Ma S. 2013. Racial differences in nasopharyngeal PMID: 4055129.
carcinoma in the United States. Cancer Epidemiol, 37(6), 793–​ 802. Zeng Y, Zhong JM, Li LY, et  al. 1983. Follow-​up studies on Epstein-​Barr
PMCID: PMC3851929. virus IgA/​VCA antibody-​positive persons in Zangwu County, China.
Ward MH, Pan WH, Cheng YJ, et  al. 2000. Dietary exposure to nitrite and Intervirology, 20(4), 190–​194. PMID: 6317603.
nitrosamines and risk of nasopharyngeal carcinoma in Taiwan. Int J Zeng YX, and Jia WH. 2002. Familial nasopharyngeal carcinoma. Semin
Cancer, 86(5), 603–​609. PMID: 10797279. Cancer Biol, 12(6), 443–​450. PMID: 12450730.
Wee JT, Ha TC, Loong SL, and Qian CN. 2010. Is nasopharyngeal cancer Zhai FY, Du SF, Wang ZH, et  al. 2014. Dynamics of the Chinese diet and
really a “Cantonese cancer”? Chin J Cancer, 29(5), 517–​526. PMID: the role of urbanicity, 1991–​ 2011. Obes Rev, 15(Suppl 1), 16–​ 26.
20426903. PMCID: PMC3868998.
Wei K, Xu Y, Liu J, Zhang W, and Liang Z. 2010. No incidence trends and Zhang F, and Zhang J. 1999. Clinical hereditary characteristics in nasopharyn-
no change in pathological proportions of nasopharyngeal carcinoma in geal carcinoma through Ye-​Liang’s family cluster. Chin Med J (Engl),
Zhongshan in 1970–​2007. Asian Pac J Cancer Prev, 11(6), 1595–​1599. 112(2), 185–​187. PMID: 11593591.
PMID: 21338202. Zheng J, Zhang C, Jiang L, et  al. 2011. Functional NBS1 polymorphism is
Wei Y, Zhou T, Lin H, et al. 2013. Significant associations between GSTM1/​ associated with occurrence and advanced disease status of nasopharyn-
GSTT1 polymorphisms and nasopharyngeal cancer risk. Tumour Biol, geal carcinoma. Mol Carcinog, 50(9), 689–​696. PMID: 21656575.
34(2), 887–​894. PMID: 23275251. Zheng W, McLaughlin JK, Gao YT, Gao RN, and Blot WJ. 1992. Occupational
Wen CP, Tsai SP, Cheng TY, et al. 2005. Uncovering the relation between betel risks for nasopharyngeal cancer in Shanghai. J Occup Med, 34(10), 1004–​
quid chewing and cigarette smoking in Taiwan. Tob Control, 14(Suppl 1), 1007. PMID: 1403187.
i16–​22. PMCID: PMC1766184. Zheng X, Yan L, Nilsson B, Eklund G, and Drettner B. 1994. Epstein-​Barr virus
West S, Hildesheim A, and Dosemeci M. 1993. Non-​viral risk factors for naso- infection, salted fish and nasopharyngeal carcinoma: a case-​control study
pharyngeal carcinoma in the Philippines:  results from a case-​control in southern China. Acta Oncol, 33(8), 867–​872. PMID: 7818917.
study. Int J Cancer, 55(5), 722–​727. PMID: 7503957. Zheng YM, Tuppin P, Hubert A, et  al. 1994. Environmental and dietary risk
Wu QJ, Vogtmann E, Zhang W, et al. 2012. Cancer incidence among adoles- factors for nasopharyngeal carcinoma:  a case-​control study in Zangwu
cents and young adults in urban Shanghai, 1973–​2005. PLoS One, 7(8), County, Guangxi, China. Br J Cancer, 69(3), 508–​ 514. PMCID:
e42607. PMCID: PMC3411830. PMC1968852.
504

504 PART IV:  Cancers by Tissue of Origin


Zhu K, Levine RS, Brann EA, et  al. 2002. Case-​control study evaluating Zhuo XL, Cai L, Xiang ZL, et al. 2009. TP53 codon 72 polymorphism contrib-
the homogeneity and heterogeneity of risk factors between sinonasal utes to nasopharyngeal cancer susceptibility: a meta-​analysis. Arch Med
and nasopharyngeal cancers. Int J Cancer, 99(1), 119–​123. PMID: Res, 40(4), 299–​305. PMID: 19608020.
11948502. Zou J, Sun Q, Akiba S, et  al. 2000. A case-​control study of nasopharyngeal
Zhuo X, Cai L, Xiang Z, Li Q, and Zhang X. 2009. GSTM1 and GSTT1 poly- carcinoma in the high background radiation areas of Yangjiang, China.
morphisms and nasopharyngeal cancer risk:  an evidence-​based meta-​ J Radiat Res, 41(Suppl), 53–​62. PMID: 11142212.
analysis. J Exp Clin Cancer Res, 28, 46. PMCID: PMC2669055.
 50

27 Cancer of the Larynx

ANDREW F. OLSHAN AND MIA HASHIBE

OVERVIEW not change appreciably between 1973 and 2012 in data compiled
by the NCI Surveillance, Epidemiology, and End Results (SEER)
Cancer of the larynx includes malignant tumors of the glottis, supra- Program (SEER, 2015). Approximately half (51.0%) of laryngeal can-
glottis, subglottis, and laryngeal cartilage. Older treatments for cers in the United States are glottic, 35.3% supraglottic, 1.9% subglot-
laryngeal cancer caused significant disfigurement, difficulty with swal- tic, 0.7% involve laryngeal cartilage, and 2.5% overlap the boundaries
lowing and speech, and poor quality of life. Newer treatment methods of two or more subsites so their point of origin cannot be determined.
seek to preserve laryngeal function. Worldwide, an estimated 157,000 For 8.6% of tumors, the anatomic location is not otherwise speci-
new cases and 83,000 deaths from laryngeal cancer occurred in 2012, fied (NOS). Males have more glottic tumors than females (55.6% vs.
accounting for 1.1% of all new cancer cases and 1.0% of all cancer 31.8%) and fewer supraglottic tumors (30.7% vs. 54.1%). Among both
deaths. The risk of cancer of the larynx is nearly five times higher in sexes, whites have more glottic tumors (51.8%) than blacks (44.2%).
men than women in the United States. Incidence and mortality rates Blacks have a greater proportion of supraglottic tumors (40.6%) than
of these cancers in males are decreasing in most high-​income coun- whites (34.8%).
tries, and the rates have decreased in all racial and ethnic groups in the
United States.
Active cigarette smoking is the strongest risk factor and explains STAGE AT DIAGNOSIS
the greatest proportion of cases. The association with smoking has
strengthened in both sexes over time, but decreases in smoking prev- Over half (56.2%) of cases reported to SEER registries from 2008 to
alence have decreased the number of cases in men and women. All 2012 with information on stage were classified as localized, 24.5%
tobacco products are strongly associated with increased risk, especially as regional, and 18.3% as distant (SEER, 2015). Glottic cancer has
when combined with alcohol consumption. Alcohol consumption in the highest percentage of localized disease (81.5%). The correspond-
people who have never smoked and several occupational exposures ing percentages for other subtypes were 48.1% for tumors of laryn-
are more weakly associated than smoking. Dietary patterns and indi- geal cartilage, 47.1% for subglottic, 29.7% for supraglottic, 29.7%
vidual food groups and nutrients have been associated with lower risk. for overlapping lesions, and 25.2% for NOS. Of all tumors, 57.7%
Any potential causal role for high-​risk types (e.g., HPV-​16, HPV-​18) among males and 50.2% among females were localized at the time of
of the human papillomavirus (HPV) is uncertain and is likely to be diagnosis.
small compared to the role of smoking or the association of HPV with
oropharyneal cancer. Potential genetic susceptibility factors have been
identified from candidate gene and genome-​wide association studies TUMOR PROGRESSION MODELS
(GWAS). Evidence for molecularly defined subgroups of laryngeal
cancer is rapidly emerging. Molecular profiling indicates many shared In terms of histologic appearance, laryngeal cancers develop through
features between cancers of the larynx and other smoking-​related a series of epithelial mucosal changes. The earliest visible precursor
squamous cell carcinomas. lesions are keratoses that may appear bilaterally on the vocal cords.
It has been estimated that the annual incidence of laryngeal keratoses
in the United States is 10.2 lesions per 100,000 for males and 2.1 per
CLASSIFICATION 100,000 for females (Bouquot and Gnepp, 1991). More than half of
keratotic lesions also contain dysplasia. However, even lesions without
Subtypes of laryngeal cancer are classified according to their topogra- dysplasia have the potential for malignant transformation (Isenberg
phy. The International Classification of Diseases for Oncology (ICD-​ et al., 2008). A meta-​analysis of 940 laryngeal lesions with dysplasia
O-​3) designates codes for cancers of the glottis (C32.0), supraglottis reported that the cumulative incidence of malignant transformation
(C32.1), subglottis (32.2), laryngeal cartilage (C32.3), overlapping was 14% (95% CI = 8%, 22%) (Weller et al., 2010). In Weller’s series,
lesion of larynx (C32.8), and larynx NOS (C32.9) (Fritz A, 2000). malignant transformation occurred in 10.6% (CI  =  5.1%, 20.7%) of
Glottic tumors arise from the true vocal cords, including the anterior lesions with mild/​moderate dysplasia and 30.4% (CI = 16.1%, 49.9%)
and posterior commissures. Supraglottic tumors occur proximal to the of lesions with severe dysplasia. The mean time to transformation
true vocal cords—​specifically the false vocal cords, arytenoids, epi- was 5.8 years (range = 1.8–​14.4 years). A study of disease progres-
glottis, and aryepiglottic folds. Subglottic tumors form below the apex sion among 107 cases at a single institution in relation to sex and age
of the ventricle and extend to the lower border of the cricoid cartilage. at diagnosis of dysplasia found no significant trend during the time
Hypopharyngeal cancers are rare and are often combined with cancers period 1993–​2012 (Karatayli-​ozgursoy et al., 2015). Genomic analy-
of the larynx in epidemiologic studies because of their similar clinical ses of laryngeal lesions with keratosis and dysplasia have included
management (Hashibe et  al., 2009). The epidemiologic evidence on very few cases (Bartlett et al., 2012).
risk factors for cancers of the hypopharynx is limited but is described Large molecular studies have generally grouped all head and
later in this chapter under etiologic factors. neck cancers together, rather than reporting results specifically for
Approximately 13,430 new cases of laryngeal cancer were pro- laryngeal cancers. However, Makitie and Monni reviewed studies
jected to occur in the United States in 2016 (American Cancer Society, of genetic alterations in laryngeal cancer based on microsatellite
2016). These cases represent about 1% of all incident cancers in the markers, mutation detection, immunohistochemistry, and compara-
United States and 22% of all head and neck cancers. Most laryn- tive genomic hybridization (Makitie and Monni, 2009). They identi-
geal cancers (95%) are squamous cell carcinomas (Surveillance, fied early progression events including allele loss at 3p, 9p21, and
Epidemiology, and End Results [SEER], 2015). This proportion did 17p13—​loci that contain tumor suppressor genes including FHIT

505
506

506 PART IV:  Cancers by Tissue of Origin


(3p), CDKN2A (9p21), and TP53 (17p13). Common amplifications Demographic Patterns
were mapped to 3q, 5p, 8q, 11q13, and 18p; losses were located at
3p, 5q, 8p, 9p, 11q23–​24, 13q, and 18q (Makitie and Monni, 2009). Incidence and Mortality Rates (United States)
These results were generally consistent with a genetic progression Table 27–1 presents US incidence and mortality rates based on the
model for head and neck cancer, based on cytogenetic characteristics NCI SEER registries from 2008 to 2012 (Howlander et  al., 2015).
of cell lines and tumors (Califano et al., 1996). This model proposed Overall, the age-​adjusted invasive laryngeal cancer incidence rates
that multiple genetic alterations accumulate at key loci, affect- per 100,000 are almost five times higher for males (5.8) than females
ing specific events in progression. Loss of heterozygosity (LOH) (1.2); among males, the rates are highest in black men (8.8) and lowest
at 9p21 was thought to be linked to the transition from normal to in Asian/​Pacific Islanders (2.4). For both sexes combined, lower rates
hyperplasia; LOH at 3p21 and 17p13 to progression from hyperpla- (per 100,000) are seen for Hispanics (2.3), American Indians/​Alaskan
sia to dysplasia; LOH at 11q13, 13q21, and 14q32 to the transition Natives (2.3), and Asian/​Pacific Islanders (1.2). In all races combined,
from dysplasia to carcinoma in situ (CIS); and LOH at 6p, 8, 4q27, the incidence rate in US SEER areas was below 1.0 per 100,000 from
10q23 LOH to progression from CIS to carcinoma. Another general age 25 years to 44 years. The incidence rate per 100,000 rose progres-
model of molecular pathogenesis for head and neck cancer included sively with age from 2.3 at ages 45–​49 to 7.7 at ages 55–​59, and to
loss of multiple tumor suppressor genes that affect initiation (TP53), 16.4 at ages 70–​74 years. Mortality rates are considerably lower than
progression (Rb/​INK4/​ARF), and maintenance (Notch) (Rothenberg incidence rates, reflecting the availability of effective treatment. The
and Ellisen, 2012). age-​standardized death rate (per 100,000) is nearly five times higher
One important area of molecular cancer research involves gene-​ in men (1.9) than in women (0.4) in the United States (Table 27–1).
expression profiling of tumors to identify tumor heterogeneity and
subgroups with distinctive prognosis and possibly etiology. A  series Incidence and Mortality Rates (International)
of studies have identified gene expression patterns that differentiate International incidence and mortality data are available through the
normal epithelium from squamous cell carcinoma of the head and registry system of the International Agency for Research on Cancer
neck (Makitie and Monni, 2009). Based on 60 head and neck can- (IARC) and the GLOBOCAN 2012 estimates (Ferlay et  al., 2015).
cers that included 24 laryngeal cancers, Chung identified four sub- Based on 156,877 cases worldwide, the age-​adjusted incidence rate
groups with different prognostic outcomes (Chung et al., 2004). In an was 2.1 per 100,000 in both sexes combined. The incidence rate (per
analysis that did not stratify by tumor site, these subgroups included 100,000) was nearly eight times higher in men (3.9) than women (0.5),
(1) high expression of TGF-​α; (2) mesenchymal signature; (3) normal a larger sex ratio than for any other cancer site. The incidence rate (per
tonsillar epithelial samples positive for cytokeratin-​15 expression; and 100,000) was somewhat higher in economically more developed (2.7)
(4) patients with tobacco exposure. Another gene expression study of than less developed (1.9) regions. The male/​female (M/​F) sex ratio
head and neck cancers by Walter also identified four different gene was slightly higher for less developed (3.5/​0.4 = 8.7) than more devel-
expression profiles, but designated these as basal, mesenchymal, atypi- oped (5.1/​0.6  =  8.5) countries. The highest incidence rates for men
cal, and classical (Walter et al., 2013). The Walter et al. study included (per 100,000) were observed in the Caribbean (7.9) and in Central and
30 laryngeal cases in a total of 138 head and neck cancers. Among Eastern Europe (7.8), Southern Europe (7.2), and Western Asia (6.5).
the laryngeal cancers, 11 (37%) were considered classical, 10 (33%) Lower rates were seen for Middle Africa (1.4) and Western Africa
basal, 5 (17%) atypical, and 4 (13%) mesenchymal. Tumors with (1.4). Globally, the mortality rate (per 100,000) was 10 times higher
the classical signature were obtained from patients with the heaviest in men (2.0) than women (0.2). The M/​F sex ratio for mortality was
smoking history and had elevated expression of genes associated with higher for more developed countries (2.2/​0.2 = 11) than less developed
tobacco smoke exposure. Basal cancers had a similar gene expression countries (2.0/​0.3 = 6.6). As was the case for incidence rates, mortal-
signature as basal cells from normal human airway epithelium, with ity rates (per 100,000) for males were highest in the Caribbean (4.0),
high expression of genes such as COL17A1, TGFA, EGFR, and TP63. Central and Eastern Europe (4.9), and South America (3.0).
Tumors with the atypical signature had a strong HPV-​positive expres-
sion pattern, whereas those with a mesenchymal signature showed Trends in Incidence and Mortality in the United States
elevated expression of genes involved in the epithelial to mesenchy- The incidence and death rates from laryngeal cancer are decreasing
mal transition. Despite the limited sample size, Walter showed a strong among men and women in the United States. The average annual per-
correlation between the head and neck cancer subgroups and subtypes cent change (AAPC) in SEER from 2003 to 2012 showed statistically
of lung squamous cell carcinoma reported elsewhere (Wilkerson et al.,
2010). However, Walter was not able to confirm the association with
survival reported by Chung. Table 27–​1. Incidence and Mortality Rates from Cancer of the Larynx: 
The most comprehensive analysis of the genome of head and US SEER Program (2008–​2012)
neck cancer to date has been reported by The Cancer Genome Atlas
(2015). Their analysis of 279 head and neck tumors identified four Incidencea Mortalityb
gene expression subtypes as reported by Chung et  al. (2004) and
Walter et al. (2013). The proportionate contributions of the smoking-​ Race/​Ethnicity Total Males Females Total Males Females
related “classical” tumors and non-​HPV related “atypical” cancers
were higher for laryngeal cancer than for other head and neck cancers. All Races 3.2 5.8 1.2 1.1 1.9 0.4
Here too the classical subtype was strongly associated with a history White 3.3 5.8 1.2 1.0 1.8 0.4
of heavy smoking and gene alterations of TP53, CDKN2A, 3q, and White Hispanic 2.4 4.7 0.5 0.9 1.7 0.2
oxidative stress genes. White 3.5 6.0 1.4 1.0 1.8 0.4
Non-​Hispanic
Black 4.7 8.8 1.7 1.8 3.6 0.6
Asian/​Pacific 1.2 2.4 0.3 0.4 0.8 0.1
DESCRIPTIVE EPIDEMIOLOGY Islander
American Indian 2.3 4.3 —​ 0.7 1.4 0.2
Disease Burden Alaska Native
Worldwide, an estimated 157,000 new cases and 83,000 deaths from Hispanic 2.3 4.5 0.5 0.8 1.7 0.2
laryngeal cancer occurred in 2012 (Ferlay et al., 2015). These com-
prise 1.1% of all new cases of cancer and 1.0% of all cancer deaths a
Rates per 100,000, age adjusted to the 2000 US population, SEER (2008–​2012), SEER
estimated for that year. The American Cancer Society projects that 18 areas.
b
Rates per 100,000, age adjusted to the 2000 US population, SEER (2008–​2012), US
13,430 new cases and 3620 deaths from laryngeal cancer will occur in Mortality Files.
the United States in 2016 (American Cancer Society, 2016). —​Statistic not shown. Rate based on less than 16 cases.
 507

Cancer of the Larynx 507


significant decreases in incidence (–​ 2.6%) and mortality (–​ 2.2%) but lower survival for regional stage (43.2% vs. 48.2%). Whites had
(Figure 27–1) in both sexes combined. This pattern held for every race/​ better survival for all stages, with the largest difference seen for distant
ethnicity group (Table 27–2), except for a small, non-​significant increase stage (36.3% vs. 30.5% for blacks).
for American Indians/​Native Alaskans. The largest AAPC decrease in
incidence (–​3.3%) was observed among blacks, followed by Asian/​
ETIOLOGIC FACTORS
Pacific Islanders (–​3.0%) and non-​Hispanic whites (–​2.3%). Similarly,
the largest AAPC decrease in mortality was among blacks (–​3.3%). In
general, larger decreases in incidence were found for females compared Tobacco
to males (data not shown). For all races, females had a –​3.1% decrease The causal relationship between tobacco smoking and laryngeal can-
in incidence compared with males (–​1.9%). For mortality, males had a cer is well established (Hashibe et al., 2009; IARC, 2004). Smoking
slightly larger decrease (–​2.5%) than females for all races combined. cigarettes, cigars, or pipes increases risk. The association with ciga-
rette smoking is stronger than for any other cancer except lung cancer
International Trends in Incidence and Mortality (see Chapter 11 in this volume).
Based on cancer incidence data from Cancer Incidence in Five Multiple international cohort and case control studies have shown
Continents, the incidence rates for laryngeal cancer have generally dose–​ response relationships between increasing risk of laryngeal
been declining steadily in men since 1980, as shown for Europe (Figure cancer and daily consumption and duration of cigarette smoking.
27–2), Asia and Oceania (Figure 27–3), and Africa and the Americas A pooled analysis of large cohort studies in the United States shows
(Figure 27–4) (Australian Cancer Database, 2015; Engholm, 2015; that the mortality rate from laryngeal cancer among cigarette smok-
Ferlay, 2014a, 2014b; Hamdi, 2013; United Kingdom, 2015). Among ers and the strength of the association between current smoking and
women, the incidence rates of laryngeal cancer have also been declin- death from laryngeal cancer have increased over time, especially
ing (Figures 27–5, 27–6, and 27–7, although more modestly than the among women (Carter et  al., 2015). In contrast, tumor registries in
rates among men in most locations. Substantial declines have been many countries show that the incidence rates from laryngeal cancer in
observed in India, Thailand, and the United States (whites and blacks). the general population (including current, former, and never-​smokers)
have decreased among men (Figures 27–2, 27–3, and 27–4) but not
women (Figures 27–5, 27–6, and 27–7), due to the earlier decline in
Survival (United States) the prevalence of smoking among men than women.
Table 27–3 presents the 5-​year relative survival percentages from Pipe and cigar smoking are also associated with increased incidence
the SEER Program, 2005–​2011. Overall, relative survival was better and death rates from laryngeal cancer among men. In an analysis of
for males (61.2%) than for females (56.3%), and for whites (61.2%) the American Cancer Society (ACS) cohort Cancer Prevention Study
than for blacks (52.9%). Using the SEER Summary Stage 2000 data II (CPS-​II), Henley et al. (2004) reported a strong association between
(table 2), males had slightly better survival than females for localized exclusive pipe smoking by men and mortality from laryngeal can-
stage tumors (76.8% vs. 71.5%) and distant stage (35.5% vs. 34.3%), cer for both current smokers (HR  =  13.1; 95% CI  =  5.2, 33.1) and

Incidence Mortality
Rate per 100,000 Rate per 100,000
10 10
White
9 Black 9

8 APIb 8
AI/ANc
7 7
Hispanicd
6 6

5 5

4 4

3 3

2 2

1 1

0 0
1975 1980 1985 1990 1995 2000 2005 2012 1975 1980 1985 1990 1995 2000 2005 2012
Year of Diagnosis Year of Death

Figure 27–1.  SEER Incidence and Death Rates: Cancer of the Larynx, Both Sexes.a Source: Incidence data for whites and blacks are from the SEER 9
areas (San Francisco, Connecticut, Detroit, Hawaii, Iowa, New Mexico, Seattle, Utah, Atlanta). Incidence data for Asian/Pacific Islanders, American Indians/
Alaska Natives and Hispanics are from the SEER 13 areas (SEER 9 areas, San Jose-Monterey, Los Angeles, Alaska Native Registry and Rural Georgia).
Mortality data are from US Mortality Files, National Center for Health Statistics, CDC.
a
Rates are age-adjusted to the 2000 US Std Population (19 age groups - Census P25-1103).
Regression lines are calculated using the Joinpoint Regression Program Version 4.2.0. April 2015, National Cancer Institute. Joinpoint analyses for Whites
and Blacks during the 1975-2012 period allow a maximum of 5 joinpoints. Analyses for other ethnic groups during the period 1992-2012 allow a maximum
of 3 joinpoints.
b
API= Asian/Pacific Islander.
c
AI/AN = American Indian/Alaska Native. Rates for American Indian/Alaska native are based on the CHSDA/Contract Health Service Delivery Area)
counties.
d
Hispanic is not mutually exclusive from whites, blacks, Asian/Pacific Islanders, and American Indians/ Alaska Natives. Incidence data for Hispanics
are based on NHIA and exclude cases from the Alaska Native Registry. Mortality data for Hispanics exclude cases from New Hampshire and Oklahoma.
508

508 PART IV:  Cancers by Tissue of Origin


Table 27–​2. Trends in Incidence and Mortality: US SEER Program 14
(2003–​2012)

Incidence Mortality
Race/​Ethnicity AAPCa (%) AAPCb (%)
12
All Races –​2.6 –​2.2
White –​2.4 –​1.9
White Hispanic –​1.8 –​2.5
White Non-​Hispanic –​2.3 –​1.8
Black –​3.3 –​3.3 10
Asian/​Pacific Islander –​3.0 –​1.9
Amer Ind/​Alaska Nat 0.6 –​2.6
Hispanic –​1.8 –​2.5

Rates per 100,000


a
Average annual percent change (AAPC) based on a joinpoint analysis with up to 3 8
joinpoints over the diagnosis years 1992–​2012.
b
The 2003–​2012 mortality AAPCs are based on a Joinpoint analysis using years of death
1992–​2012.

14
4

12
2

10
0
1970 1975 1980 1985 1990 1995 2000 2005 2010

*China Israel *Japan *India


Singapore *Philippines Australia *Thailand
Rates per 100,000

8
*Regional data

Figure 27–​3.  Trends in age standardized incidence rates (ASR) from laryn-


geal cancer (C32) in selected regions of Asia and Oceania (men). Rates
6
have been smoothed using 5-​year average. Source: C15 other database.

former smokers (HR = 5.6; 95% CI = 1.5, 21.2). The association with


4
pipe smoking was stronger for laryngeal cancer than for lung cancer.
By comparison, the HR for lung cancer among current versus never-​
smokers was 5.0 (95% CI = 4.16, 6.01) and for former smokers was
1.70 (95% CI = 1.23, 2.36).
Similar findings were reported for men who exclusively smoked
cigars in the ACS CPS-​II cohort (Shapiro et  al., 2000). The relative
2
risk (RR) estimate for current cigar smokers was (RR = 10.3, 95% CI
= 2.6, 41.0) (Shapiro et al., 2000). A pooled analysis of case control
studies in Europe reported an odds ratio (OR) of 5.60 (95% CI = 1.64,
19.13) for exclusive cigar use, compared to 11.11 for exclusive ciga-
rette use (Hashibe et al., 2007a, 2007b). There were no exclusive pipe
0
1970 1975 1980 1985 1990 1995 2000 2005 2010 smokers in the study population.
International Head and Neck Cancer Consortium (INHANCE)
Denmark Finland *France *Italy investigators performed a pooled analysis of 1026 hypopharyngeal
Norway Slovakia UK: England Sweden cancer cases in relation to cigarette, cigar, and pipe smoking (Wyss
et  al., 2013). Among never-​cigarette-​smokers they found OR esti-
*Regional data mates of 2.51 (95% CI  =  0.90,  –​6.97) comparing current to never-​
cigar-​smokers and 3.90 (95% CI = 1.39, 10.92) for pipe smokers. The
Figure  27–​
2. Trends in age standardized incidence rates (ASR) from OR estimates for larynx cancer were 6.31 (95% CI = 3.09, 12.92) for
laryngeal cancer (C32) in selected regions of Europe (men). Rates have cigar smoking and 3.53 (CI  =  1.48, 8.37) for pipe smoking among
been smoothed using 5-​year average. Source: C15 other database. never-​cigarette-​smokers.
 509

Cancer of the Larynx 509


14 4

3.5
12

3
10

2.5
Rates per 100,000

Rates per 100,000


8

1.5

4
1

2
0.5

0 0
1970 1975 1980 1985 1990 1995 2000 2005 2010 1970 1975 1980 1985 1990 1995 2000 2005 2010

*USA: Black *USA: White Denmark Finland *France *Italy


Norway Slovakia UK: England Sweden
*Regional data
*Regional data
Figure 27–​4.  Trends in age standardized incidence rates (ASR) from laryn-
geal cancer (C32) in the United States (men). Rates have been smoothed Figure  27–​
5. Trends in age standardized incidence rates (ASR) from
using 5-​year average. Source: C15 other database. laryngeal cancer (C32) in selected regions of Europe (women). Rates have
been smoothed using 5-​year average. Source: C15 other database.

Some variation in the strength of the association between smoking tobacco) and non-​ filtered cigarettes compared to filter tip, low-​
and laryngeal cancer has been reported by anatomic subsite (Olshan, ventilation cigarettes.
2010). The INHANCE group, the largest pooling study of laryngeal The association between smoking and laryngeal cancer is greater
cancer (22 international studies), conducted an analysis of tobacco use for women than men in terms of relative risk, but lower in terms of
using the largest sample of hypopharynx cases (1026 cases) assem- absolute risk. Carter et al. (2015) reported that the hazard ratio for cur-
bled to date (Wyss et  al., 2013). They reported an adjusted OR of rent versus never smoking and death from laryngeal cancer was 107.4
6.48 (CI = 4.94, 8.51) for cancer of the hypopharynx among people (95% CI = 25.1, 459.4) in women and 15.5 (95% CI = 9.3, 25.8) in
who had ever smoked compared to those who had never smoked. The men in a pooled analysis of large cohort studies in the United States,
adjusted OR for larynx cancer was 8.33 (CI = 7.07, 9.81). All other even though the absolute death rate from laryngeal cancer among
head and neck cancer sites had smaller ORs than hypopharynx. A case smokers was three times higher in men than women. Freedman et al.
control study in Central and Eastern Europe (384 cases) compared the (2007) reported that the hazard ratio associated with smoking was sig-
OR estimates for current versus never smokers for both supraglottic nificantly larger in women (HR 12.96; 95% CI = 7.81, 21.52) than in
tumors (OR = 3.23; CI = 1.72, 6.42) and glottic tumors (OR = 2.61; men (5.45; 95% CI = 4.22, 7.05; P for interaction: < .001) for all head
CI  =  1.67, 4.08) (Hashibe et  al., 2007a). Although the overall point and neck cancers combined in the AARP cohort. A European cohort
estimates did not differ, the association with duration of smoking and study (EPIC), found larger hazard ratios (HR) for female (HR 20.37;
pack-​years of smoking was consistently greater for supraglottic than 95% CI  =  4.86, 85.5) than male (HR 13.24; 95% CI  =  6.00, 29.20)
glottic tumors. The authors estimated that 87% of laryngeal cancers in current versus never smokers (Agudo et al., 2012). The higher HR in
Central Europe are due to tobacco use. women than men signifies that the proportion of laryngeal cancers
Some variation in the strength of the association has also been caused by smoking is higher, even though the absolute risk is lower.
reported by type of tobacco (Olshan, 2010). For example, some stud- An analysis of pooled data from international case control studies
ies have reported stronger associations with supraglottic tumors from showed that a history of ever having smoked cigarettes was associ-
smoking black (air-​cured) tobacco compared to blonde (flue-​cured ated with a smaller OR in younger adults (≤ 45 years) than in older
510

510 PART IV:  Cancers by Tissue of Origin


4 4

3.5 3.5

3 3

2.5 2.5

Rates per 100,000


Rates per 100,000

2 2

1.5 1.5

1 1

0.5 0.5

0 0
1970 1975 1980 1985 1990 1995 2000 2005 2010 1970 1975 1980 1985 1990 1995 2000 2005 2010
Australia Israel *India Singapore *USA: Black *USA: White
*China *Philippines *Japan *Thailand
*Regional data
*Regional data
Figure  27–​
7. Trends in age standardized incidence rates (ASR) from
Figure 27–​6.  Trends in age standardized incidence rates (ASR) from laryn- laryngeal cancer (C32) in the United States (women). Rates have been
geal cancer (C32) in selected regions of Asia and Oceania (women). Rates smoothed using 5-​year average. Source: C15 other database.
have been smoothed using 5-​year average. Source: C15 other database.

(> 45 years) adults. However, the odds ratios for those under age 45 self-​reported light versus deep inhalation (OR = 0.22; CI = 0.09, 0.55).
were larger across pack-​year categories compared to older adults, However, clear dose–​response relationships were seen with the dura-
although the estimates for the younger group tended to be impre- tion and intensity of smoking, even in those who reported only puffing
cise (Toporcov et  al., 2015). Ramroth et  al. (2011) assessed risk in lightly on cigarettes.
relation to self-​reported depth of inhalation in a German case control Several recent international studies have examined the association
study (257 cases) and found significantly lower risk in smokers who with larynx cancer in relation to time since cessation of smoking, and

Table 27–​3.  Five-​Year Relative Survival: US SEER Program (2005–​2011) by Race, Sex, and Stage

5-​Year Relative
Survival % Total Males Females Total Males Females Total Males Females

2011a 60.2 61.2 56.3 61.2 62.2 57.0 52.9 53.7 49.5
All Stagesb 60.6 61.5 56.9 61.6 62.6 57.6 53.3 53.8 51.2
Localized 75.9 76.8 71.5 75.7 76.7 70.9 74.2 74.4 72.9
Regional 44.5 43.2 48.2 45.4 44.0 49.3 40.8 39.8 43.3
Distant 35.2 35.5 34.3 36.3 37.7 34.6 30.5 30.1 31.6
Unstaged 55.7 57.3 49.1 56.8 58.9 47.3 50.7 49.4 —​

a
SEER 18 areas. Period survival estimate for 2011 using conditional survival estimates joined from several 3-​year calendar block cohorts.
b
Stage at diagnosis classified using SEER summary stage 2000.
—​Statistic could not be calculated due to fewer than 25 cases during the time period.
 51

Cancer of the Larynx 511


age at cessation. An Italian case control study reported that 10–​19 cavity and pharynx. At light and moderate levels of alcohol consump-
years after cessation the OR for laryngeal cancer was 0.33 (CI = 0.23, tion, males and females had similar pooled RR estimates; for heavy
0.47) compared to those who continued smoking. The greatest reduc- drinkers, the RR was higher for men (RR = 2.77; CI = 2.15, 3.57) than
tion was seen in smokers who quit by age ≤ 35 years (OR = 0.11; CI = women (RR = 1.55; CI = 0.45, 5.34), although only one study reported
0.06, 0.23) (Bosetti et al., 2008). female-​specific results. Results stratified by geographic region showed
Among those who do not drink alcohol, tobacco smoking is more similar RR estimates for European and North American studies; RRs
strongly associated with laryngeal than with oral cavity cancer. The were elevated, but lower for Asian studies, though only four Asian
International Head and Neck Cancer Consortium (INHANCE) evalu- studies were included.
ated the separate and joint effects of tobacco and alcohol use, based The INHANCE pooling project also examined the association
on analyses of 2959 cases. The OR for people who reported smok- between alcohol consumption and laryngeal cancer among indi-
ing but not drinking was 6.76 (95% CI = 4.58, 9.96) (Hashibe et al., viduals who had never used tobacco (121 cases). Among these, risk
2009). This association was stronger than for cancers of the oral cav- was associated with the amount but not the duration of drinking.
ity (1.74; CI = 1.10, 2.76) or pharynx (1.91; CI = 1.39, 2.62). A higher Consumption of more than 5 drinks per day was associated with
OR [12.83 (95% CI  =  7.95, 20.71)] was observed for people who an OR of 2.98 (95% CI  =  1.72, 5.17); consumption for more than
reported smoking more than 20 cigarettes per day without alcohol 40  years was associated with an OR of 1.0 (95% CI  =  0.58, 1.73)
consumption. The comparable odds ratios for oral cavity and phar- (Hashibe et  al., 2007b). The INHANCE group also examined the
ynx were lower; 3.13 (95% CI = 1.14,8.59) and 2.83 (95% CI = 1.66, association with different types of alcohol (Purdue et al., 2009). The
4.82), respectively. OR for participants who reported drinking more than 15 drinks per
An INHANCE analysis of passive smoking exposure based on 71 week of only beer was 2.7 (95% CI = 1.7, 4.4); 1.9 (95% CI = 0.9, 3.9)
laryngeal cancers among people who have never smoked reported for those reporting only liquor consumption; and 3.9 (95% CI = 1.2,
an elevated but statistically borderline OR for those who were invol- 13.0) for those who consumed only wine. These associations were
untarily exposed to tobacco smoke at home or work (OR  =  1.71; weaker for laryngeal cancer than for oral cavity or pharyngeal can-
CI = 0.98, 3.00) (Lee et al., 2008). Exposure at home for more than cer. The separate and joint associations with alcohol and tobacco use
15 years was associated with an OR of 2.58 (95% CI = 1.20, 5.57). were also examined by INHANCE (Hashibe et al., 2009). The analy-
A Taiwanese cohort study of 176 cases of laryngeal cancer reported sis included 2959 cases and found a weakly elevated OR for a history
increased risk associated with betel-​quid chewing (Lee et al., 2011). of alcohol use but no smoking (OR = 1.21; 95% CI = 0.77, 1.92). The
Individuals who chewed 20 or more quids daily had an adjusted hazard OR for 3 or more drinks per week among individuals who reported
ratio of 1.7 (95% CI = 1.2, 2.6) compared to non-​users. In an Indian never smoking was 3.16 (95% CI  =  1.23, 8.16). The joint effects
case control study with 511 cases, Sapkota reported strong associa- of tobacco and alcohol were greater than additive. The authors esti-
tions between bidi smoking and cancers of the glottis (OR = 6.80; 95% mated that the population-​attributable risk for tobacco and alcohol
CI = 4.64, 9.97) and supraglottis (OR = 7.54; 95% CI = 3.84, 14.74) consumption was 89% for laryngeal cancer, compared to 64% for
(Sapkota et al., 2007). Gutkha was the only chewing tobacco product oral cavity and 72% for pharyngeal cancer.
associated with an elevated OR in the Sapkota study among individuals A large analysis (4818 cases) from the INHANCE international
with no history of active smoking (OR = 2.55; 95% CI = 0.62, 10.44). pooling project reported a slightly stronger association for ≥ 5 drinks
Other smaller Indian studies have also reported an elevated risk for per day for persons 45 years of age or older (OR = 2.82; CI = 2.40,
bidi smoking and use of smokeless tobacco (Jayalekshmi et al., 2013; 3.25) than persons less than or equal to age 45  years (OR  =  1.81;
Pednekar et al., 2011). Kapil found an OR of 2.37 (95% CI = 1.12, CI  =  1.13, 2.90) (Toporcov et  al., 2015). The significance of this is
5.06) for those who chewed betel leaf with tobacco compared to non-​ unclear.
users (Kapil et al., 2005).
In an analysis based on pooled international studies, Berthiller
reported that having ever smoked marijuana was not associated with Diet
increased risk (OR = 0.98; CI = 0.69, 1.39) (Berthiller et al., 2009). Studies consistently report an inverse association between greater
Neither frequency nor length nor time using marijuana was associated consumption of fruits and vegetables and risk of laryngeal cancer
with laryngeal cancer, except among those with greater than 20 years (Lucenteforte et al., 2010). Some studies also suggest increased risk
of use (OR = 1.42; CI = 0.84, 2.38). associated with consumption of meat and eggs. Higher levels of retinol,
total calories, protein, and fat have been associated with an increased
risk in some but not all studies; dairy products have also been reported
Alcohol to be inversely associated with risk (Lucenteforte et al., 2010).
Epidemiologic studies have shown that alcohol consumption in the The World Cancer Research Fund and the American Institute for
absence of tobacco smoking has a weak independent association with Cancer Research (WCRF/​ AICR) have designated the evidence as
laryngeal cancer (Hashibe et al., 2009). Limited evidence suggests that “probable” that non-​starchy vegetables, fruits, and foods containing
supraglottic tumors may be more strongly associated with alcohol than carotenoids decrease the risk of cancers of the mouth, pharynx, and
glottic tumors (Hashibe et al., 2007a). Other features of the association larynx combined (WCRF/​AICR, 2007).
with alcohol, such as differences between males and females and the Recent case control studies including a case control study in
strength of the association for smoking combined with heavy alcohol Uruguay (275 cases) have examined dietary patterns in relation to
consumption, are less well studied. laryngeal cancer (De Stefani et al., 2013). Adherence to a dietary pat-
A recent meta-​ analysis of 41 case control and cohort studies tern classified as “prudent” was associated with reduced risk: (OR =
compared 7059 cases in patients who reported a history of alcohol 0.54; 95% CI = 0.36, 0.80), whereas diets described as “starchy plants”
consumption with 2575 cases who reported no history of alcohol con- (OR = 1.62; 1.08, 2.41), “Western diet” (OR = 1.84; 1.14, 2.98), or
sumption (Bagnardi et al., 2014). The pooled RR estimates for light “drinker” (OR = 2.86; 1.80, 4.56) were associated with increased risk.
drinkers (< 12.5 grams of alcohol per day), moderate drinkers (≤ 50 An earlier analysis from the same study reported that a high-​fat die-
g/​day), and heavy drinkers (> 50 g/​day) were 0.87 (95% CI = 0.68, tary pattern had an OR of 1.47 (95% CI = 0.77, 2.82; and a “healthy”
1.11), 1.44 (95% CI = 1.25, 1.66), and 2.65 (95% CI = 2.19, 3.19), pattern had a decreased odds (OR = 0.64; 95% CI = 0.24, 1.18) (De
respectively. Although study-​ specific smoking-​adjusted RRs were Stefani et al., 2007). A US case control study of 419 cases used factor
used in the meta-​analysis, residual confounding by tobacco use cannot analysis to identify dietary patterns and found decreased risk associ-
be ruled out. Compared to the dose–​response relationship with cancers ated with higher consumption of fruits, vegetables, and lean protein
of the oral cavity and pharynx, the relationship between the amount (highest consumption quartile OR = 0.73; CI = 0.48, 1.10) and an ele-
of alcohol consumed and laryngeal cancer risk was less steep and flat- vated OR for the dietary pattern characterized by fried foods, high-​fat
tened out at consumption above 100g/​day at an RR of about 4.0. This and processed meats, and sweets (highest consumption quartile OR =
contrasts with a maximum RR of about 8.0 for cancers of the oral 2.12; CI = 1.21, 3.72) (Bradshaw et al., 2012).
512

512 PART IV:  Cancers by Tissue of Origin


The INHANCE pooling project (4073 cases) evaluated different A Dutch case-​control case-​cohort study (216 cases) found weak to
food groups and reported inverse associations between risk and fruit moderately elevated hazard ratios (HR) for asbestos exposure based
intake (highest quartile OR = 0.59; 95% CI = 0.45, 0.79) and white on estimates from a job-​exposure matrix (Offermans et  al., 2014).
meat (OR = 0.84; 95% CI = 0.70, 1.00) and increased risk associated The HR for the highest cumulative exposure times–​exposure inten-
with red meat (OR = 1.50; 95% CI = 0.96, 2.36) and processed meat sity category was 1.51 (95% CI = 0.92, 2.49) and that for the lon-
(OR = 1.36; 95% CI = 1.06, 1.73) (Chuang et al., 2012). Higher dietary gest duration of high exposure was 6.36 (95% CI  =  2.18, 18.53).
scores based on high fruit and vegetable and low red meat intake were There was a higher HR for individuals who had ever been exposed
associated with a decreased OR (one score unit OR  =  0.89; 95% to asbestos for supraglottic cancer (HR = 2.14; 95% CI = 1.13, 4.04)
CI = 0.82, 0.96). but not glottic cancer (HR  =  0.94; 95% CI  =  0.61, 1.44); analyses
Studies have also evaluated micronutrients and other dietary using other exposure metrics did not show a clear difference by
components. The INHANCE pooling study of 1545 cases examined laryngeal subsite and had wide confidence intervals. An elevated
vitamin C intake and found that the highest quintile of self-​reported SMR was found for Chinese male workers in an asbestos factory
vitamin C intake was associated with reduced odds of laryngeal using chrysotile (SMR = 4.26; 95% CI = 1.17, 15.52) (Wang, Lin,
cancer (OR = 0.52; 95% CI = 0.40, 0.68) overall, as well as among et  al., 2013). A  case control study in Central and Eastern Europe
current tobacco users (OR = 0.55; 95% CI = 0.4, 0.72) and heavy (316 cases; Shangina et al., 2006) found no overall association with
drinkers (OR = 0.38; 95% CI = 0.25, 0.59) (Edefonti et al., 2015). asbestos exposure (OR = 0.86; 95% CI = 0.51, 1.45). A cohort study
Another INHANCE analysis reported decreased odds ratios for any of Swedish male construction workers (227 cases) found elevated
vitamin supplementation (OR = 0.68; 95% CI = 0.37, 1.25), vita- rate ratios for those exposed to asbestos (RR = 1.9; 95% CI = 1.2,
min A supplement use (OR = 0.56; 95% CI = 0.22, 1.24), vitamin 3.1) and mineral wool (RR = 1.6; CI = 1.03, 2.4), but no association
C (OR = 0.70; 95% CI = 0.46, 1.08), and vitamin E (OR = 0.50; with cement dust, asphalt, or stone dust (Purdue et al., 2006). A small
95% CI = 0.10, 2.46) (Li et al., 2013). A recent INHANCE analysis Boston area case control study (118 cases) reported no association
found that total carotenoid intake was associated with reduced risk with asbestos exposure (OR =1.04; 95% CI = 0.64, 1.67) (Langevin
(highest quintile OR = 0.61; 95% CI = 0.50, 0.76) (Leoncini et al., et al., 2013).
2016). Intake of beta-​carotene, beta-​cryptoxanthin, and lutein plus A meta-​analysis of cohort and case control studies that examined
zeaxanthin also had reduced odds ratios. The inverse association incidence and mortality from laryngeal cancer in relation to silica dust
with lycopene intake was statistically borderline (highest quintile exposure reported moderately increased risk (e.g., SIR  =  1.50; 95%
OR  =  0.83; 95% CI  =  0.68, 1.02). Another INHANCE analysis CI  =  0.59, 2.42 in cohort studies; and in case control studies, 1.39;
showed no association with allium vegetables—​specifically, garlic 95% CI = 1.17, 1.67) (Chen and Tse, 2012). In another study, a weak
or onion consumption (Galeone et  al., 2015). A  large Italian and but statistically insignificant increase in risk was found for uranium
Swiss case control study (851 cases) reported elevated odds ratios miners exposed to radon and their short-​lived progeny (OR  =  1.13;
for red meat intake (≥ 90 grams a day: OR = 2.34; 95% CI = 1.82, 95% CI = 0.75, 1.70) (Mohner 2008).
3.00) (Di Maso et  al., 2013). The same study noted a decreased A series of analyses from a German case control study (257 cases)
OR for consumption of cruciferous vegetables (>1 portion a reported increased odds ratios associated with several occupations
week: OR = 0.84; 95% CI = 0.67, 1.05) (Bosetti et al., 2012) and (road construction worker, carpenter, metal production and process-
citrus fruit or citrus juice (> 4 portions per week:  OR  =  0.55; ing worker, farmer) and exposures (cement dust, and wood dust)
95% CI  =  0.37,0. 83 (Foschi et  al., 2010). An analysis of dietary (Dietz et al., 2004; Becher, 2005; Ramroth and Dietz, 2008). Shangina
fiber showed about 40%–​60% decreased odds for total soluble, reported no association with wood dust exposure (OR  =  0.81; 95%
total insoluble fiber consumption, vegetable fiber, and fruit fiber CI = 0.48, 1.37) and non-​significantly increased risk associated with
(Pelucchi et al., 2003). For total fiber intake, the decrease was simi- exposure to soot (OR  =  1.30; 95% CI  =  0.68, 2.48), formaldehyde
lar for supraglottic and glottic tumors. (OR  =  1.68; 95% CI  =  0.85, 3.31), and coal dust (OR  =  1.81; 95%
CI = 0.94, 3.47) (Shangina et al., 2006).
Occupation
Human Papillomavirus
Cohort and case control studies of occupational populations have
reported several exposures associated with increased risk of laryngeal Human papillomavirus virus (HPV), especially the “high-​risk” HPV
cancer (Olshan, 2010). Occupational exposure to asbestos has been subtypes 16 and 18, are established risk factors for cancers of the
the best studied (Institute of Medicine, 2006), but other researchers oropharynx (see Chapters  24 and 29 in this volume). The associa-
have shown, albeit inconsistently, an elevated risk with man-​made tion between HPV and laryngeal cancer is weaker and the fraction of
vitreous fibers, mustard gas, sulfuric acid, solvents, formaldehyde, cases likely to be smaller for cancers of the larynx (Chaturvedi and
wood dust, and nickel refining. A variety of occupations and indus- Gillison, 2010). A  recent examination of the prevalence of HPV in
tries have also shown to be associated with cancer of the larynx. archival samples from laryngeal tumors in selected US states detected
These include metal-​related occupations, paint and print workers, HPV in 20.9% of laryngeal tumor specimens (n = 148) versus 70.1%
truck drivers, railway workers, butchers, textile workers, plastic and of oropharyngeal cancers (Saraiya et al., 2015). Among a series of 91
rubber manufacturing, agriculture, and construction work. The occu- laryngeal tumors, three cases (3.2%) were found to be HPV positive
pational studies are limited by imprecise data on specific exposures by in situ hybridization for “high-​risk” HPV types (16, 18, 31, 33, 35,
and exposure levels, and in some cases by inability to adjust for 39, 45, 51, 52, 56, 58, 66) and P16 immunohistochemistry, whereas six
tobacco and alcohol use. cases (6.6%) were positive for HPV by p16 immunochemistry (Upile
Recent meta-​analyses and several studies of administrative data sets et  al., 2014). A  German study reported that of 102 laryngeal tumor
have reported associations between laryngeal cancer and other expo- samples, three (2.9%) were “HPV-​driven,” that is, they had a pattern of
sures, occupations, and industries. A  meta-​analysis of cohort stud- HPV DNA-​and RNA-​positivity and p16 high expression and low pRB
ies of workers employed in crop-​protection chemical and pesticide expression (Halec et al., 2013). The analysis of archival tissue samples
manufacturing reported an overall summary standardized mortality from the NCI’s SEER residual tissue repository (148 cases) found that
ratio (SMR) of 1.58 (95% CI = 1.09, 2.31) and an SMR of 1.51 (95% 21% were HPV positive, and of these, 6% were HPV-​16 positive. They
CI  =  1.01, 2.78) for phenoxy herbicide manufacturing (Jones et  al., also found that more women than men were HPV positive (Hernandez
2009). A US death certificate study reported a statistically insignificant et al., 2014).
association with employment as a butcher (OR = 1.19; 95% CI = 0.92, Human papillomavirus (HPV) can disrupt the cell cycle through
1.54) (Besson et  al., 2006). An analysis of a large Swedish census inactivation of the Rb pathway via the HPV E7 protein and inactivation
and cancer data set reported elevated standardized incidence ratios of the TP53 pathway through the HPV E6 protein. A large European
(SIR) for waiters (SIR = 2.70; 95% CI = 1.39, 4.44) and hairdressers case control study (529 hypopharynx/​laryngeal cases) analyzed HPV
(SIR = 1.78; 95% CI = 1.10, 2.62) (Ji and Hemminki, 2005). 16 L1, E6, and E7 seropositivity (Anantharaman et  al., 2013). The
 513

Cancer of the Larynx 513


strongest association was reported for combined HPV 16 E6 and E7 HOST FACTORS
positivity (OR 30.84; 95% CI  =  3.11, 306.0). A  statistically signifi-
cant association was observed for HPV 16 E6 alone (OR = 4.18; 95% Predisposing Medical Conditions
CI = 1.54, 11.32), but not for HPV 16 E7 (OR 1.53; 95% CI = 0.93,
2.51) or HPV 16 L1 (OR = 1.54; 95% CI = 0.81, 2.94). By contrast, for Most of the recent epidemiologic literature on medical history has
oropharynx cancer, the comparable odds ratios were 8.60, 132.0, and focused on the association between diabetes and the risk of laryngeal
897.66. An association between HPV 6 E7 and laryngeal cancer was cancer. The results of these studies have been inconsistent. A  study
also found (OR = 3.25; 95% CI = 1.46, 7.24). of discharge data from US Veterans Affairs Hospitals based on 1413
A meta-​analysis of 55 studies (2559 cases) found an overall HPV men with diabetes and laryngeal cancer (Atchison et al., 2011) found
prevalence of 28%. High-​risk HPV types were present in 26.6% of decreased risk in veterans with diabetes (RR = 0.76; 95% CI = 0.71,
samples and with HPV 16 in 19.8% of samples (LiGao et al., 2013). 0.80). The inverse association persisted in analyses examining race
Using data from 12 studies with normal laryngeal mucosa compari- and time since the diagnosis of diabetes. However, an analysis of
son (control) samples, the authors reported a summary OR of 5.39 the Clinical Practice Research Datalink in the United Kingdom (680
(CI = 3.32, 8.73), with larger estimates for high-​risk HPV types (HPV cases) found no association with prescriptions for Metformin (Becker
16, 18, 33, 35, 39, 45, 51, 52, 56, 58, 59, and 68; OR = 5.74; CI = 3.05, et al., 2014). No statistically significant association between diabetes
10.80), including the most common, HPV 16 (OR = 6.07; CI = 3.44, and risk of cancer of the larynx was found in a large Italian/​Swiss
10.70). case control study of 782 cases (Bosetti et al., 2012), where the OR
associated with a history of diabetes was slightly but not statistically
significantly elevated (OR = 1.23; 95% CI = 0.86, 1.74).
Other Etiologic Factors Several other medical conditions and treatments have also been
Various other possible risk factors for laryngeal cancer have been studied in relation to laryngeal cancer. A  Swedish cohort study of
studied. Body size has been examined in several case control and patients who underwent gastrectomy for peptic ulcer disease reported
cohort studies. Gaudet reported an inverse association between an elevated SIR (2.0; 95% CI  =  1.5, 2.6) (Lagergren and Lindam,
body mass index (BMI) and the incidence of laryngeal cancer in the 2012). A  South Korean case control study (408 cases) using health
INHANCE pooled case control study of 17 studies and 2837 cases insurance data reported an elevated OR for inhaled corticosteroid use
(Gaudet et  al., 2010). A  BMI < 18.5  kg/​m3 was associated with of any type (OR = 1.60; 95% CI = 0.68, 1.18 (Lee et al., 2013), but not
increased risk (OR  =  1.63; 95% CI  =  1.23, 2.17) whereas a BMI for specific types or cumulative dose (largest dose: OR = 0.84; 95%
> 30 was associated with reduced risk (OR = 0.54; 95% CI = 0.45, CI = 0.45, 1.55).
0.64). The inverse association with obesity was observed only among Previous studies have inconsistently suggested an association with
smokers/​drinkers, whereas the increased risk with low BMI persisted gastrointestinal reflux disease (GERD). A  small case control study
in analyses stratified on tobacco and alcohol use. Lubin also exam- (96 cases) reported an adjusted OR of 2.11 (95% CI  =  1.16, 3.85)
ined INHANCE data and found a similar association with BMI by for a history of GERD obtained from the medical records (Vaezi
sex (Lubin et al., 2010). Another INHANCE analysis showed a small et al., 2006). A European case control study of 183 hypopharyngeal
decrease in the OR associated with increasing height (Leoncini cancer cases reported that a history of regurgitation or medication
et al., 2014). were not associated with risk of hypopharyngeal cancer (regur-
An analysis of the NIH-​AARP cohort study (211 cases; Etemadi gitation OR  =  0.62; 95% CI  =  0.37, 1.00); medication for regurgi-
et  al., 2014)  found an increased but highly imprecise association tation OR  =  0.62; 95% CI  =  0.24, 1.59) (Macfarlane et  al., 2012).
between lower BMI (18.5 to < 25.0) and risk of laryngeal cancer Heartburn was associated with a decreased hypopharyngeal cancer
(OR = 2.18; CI = 0.68, 6.98). No association was observed with BMI risk (OR = 0.64; 95% CI = 0.44, 0.93). No association with larynx
≥ 30 (OR = 1.04; 95% CI = 0.70, 1.55). The lowest categories of waist cancer was reported for these factors.
and hip circumference each had odds ratios below 1.0. Height and
waist-​to-​hip ratio showed no association. A  recent pooling of data Inherited Susceptibility
from 20 cohort studies (142 laryngeal cases) found increased risk asso-
Epidemiologic research has shown moderately increased risk of laryn-
ciated with higher BMI (HR = 1.42; CI = 1.19, 1.70, per 5 kg/​m2), but
geal cancer to be associated with a family history of head and neck
no associations with height, waist circumference, hip circumference,
or other cancers (Wang et  al., 2010). An INHANCE analysis (2572
or waist-​to-​hip ratio (Gaudet et al., 2015). The differences between the
cases) of laryngeal cancer found an OR of 2.07 (95% CI = 1.57, 2.73)
case control and cohort study findings have been attributed to selection
for a family history of head and neck cancer among first-​degree rela-
bias in hospital-​based studies, effects on nutritional status caused by
tives, in analyses adjusted for tobacco and alcohol use (Negri et al.,
the cancer itself, and residual confounding by smoking and alcohol
2009). A  family history of other head and neck cancers related to
use (Gaudet et al., 2015). Recreational physical activity was examined
smoking (lung, nasopharynx, nasal cavity, nasal cavity, paranasal
in the INHANCE data (612 cases) (Nicolotti et al., 2011). Moderate
sinuses, esophagus, stomach, pancreas, liver, kidney, urinary bladder,
levels of activity were associated with decreased risk (OR = 0.81; 95%
uterine cervix, myeloid leukemia) in first-​degree relatives was sig-
CI  =  0.60, 1.11) compared to low or no physical activity. However,
nificantly associated with laryngeal cancer (OR = 1.27; 95 CI = 1.09,
high physical activity was associated with increased risk (OR = 1.73,
1.47), whereas a family history of other cancers was not (OR = 1.10;
95% CI = 1.04, 2.88).
95% CI = 0.95, 1.22). An Italian/​Swiss case control study (852 cases)
A multi-​center study (240 cases) of markers of oral health reported
reported associations with family history of cancer among first-​
an elevated OR for poor oral hygiene (European OR  =  1.95; 95%
degree relatives, overall (OR = 2.8; 95% CI = 1.5, 5.1), by age of the
CI = 1.02, 3.71; Latin America OR = 1.46; 95% CI = 1.05, 2.04) (Guha
index’s cancer diagnosis (< 60 years: OR = 3.5; 95% CI = 1.4, 8.7; ≥
et al., 2007). The INHANCE analysis of education and income (6485
60 years: OR = 2.2; CI = 1.0, 5.1), sex (male: OR = 3.2; CI = 1.7, 6.0;
cases) showed that low educational status (no education or completed
females not estimable), and for the cancer site of a specific relative
first stage of basic education, or at most primary education) compared
(colorectal: OR = 1.5; CI = 1.0, 2.3) (Turati et al., 2013).
to high educational status (vocational or university degree) had an OR
Epidemiologic studies continue to report candidate gene/​ SNPs
of 1.69 (95% CI = 1.24, 2.32) (Conway et al., 2014).
(single nucleotide polymorphisms) associated with laryngeal cancer,
Analysis of sexual behavior (including the practice of oral sex, num-
largely including genes previously examined (Wang, Chu, et al., 2013).
ber of lifetime sexual partners, age at sexual debut, history of same-​
A meta-​analysis found no association with the XRCC1 Arg399Gln
sex contact, history of oral-​anal contact) and head and neck cancer
SNP (Chen et  al., 2013). An Eastern European multicenter study
risk using INHANCE pooled data did not show any associations with
(326 cases) examined 37 SNPs in several DNA repair and cell cycle
laryngeal (940 cases) or hypopharyngeal cancer (251 cases) (Heck
gene SNPs (XPA, OGG1, APEX, MGMT, CHEK2) and reported asso-
et al., 2010). Cancers of the oropharynx showed elevated odds ratios
ciations with MGMT and CHEK2 (Hall et al., 2007). A Chinese case
for some sexual behavior history factors.
514

514 PART IV:  Cancers by Tissue of Origin


control study (271 cases) reported elevated odds ratios for ERCC1 and Chemoprevention
ERCC2 SNPs (Sun, 2015). Another Chinese study reported associa- Because of the association between intake of antioxidant vitamins and
tions with genes in the nucleotide excision repair pathway: ERCC1, reduced incidence of head and neck cancer in epidemiologic studies,
XPD, XPB, XPF, XPG, and XPA SNPs (Lu et al., 2014). Haplotypes prevention studies have assessed whether retinoids can reduce the first
of the double-​strand break repair gene, NSB1, were associated with occurrence of head and neck cancer and/​or second primary tumors.
decreased odds of laryngeal cancer in a combined US and Chinese Early studies of premalignant oral lesions and head and neck cancers
study (Park et al., 2010). indicated a possible reduction in risk from 13-​cis-​retinoic acid, but
A Polish study evaluated smoking-​related loci and found no associa- concern about toxicity and efficacy (overall and long term) limited
tions with laryngeal cancer (Jaworowska et al., 2011). Polymorphisms the use of this agent (William, 2010). More recent studies, including
in phase 1 and phase 2 metabolic genes combined (COX2, UGT1A1, larger randomized trials, have not shown a consistent benefit from
UGT1A6, UGT1A7) were associated with risk in a Dutch study (Lacko α-​tocopherol or β-​carotene. The Alpha-​Tocopherol Beta-​Carotene
et al., 2009). The same study group also reported an elevated OR for Cancer Prevention (ATBC) study (Wright et  al., 2007)  was a pri-
SNP combinations predicted to result in varying epoxide hydrolase, mary prevention randomized trial of daily doses of α-​tocopherol or β-​
UGT1A6, UGT1A7, and COX2 activity levels (Lacko et  al., 2013). carotene given to male smokers. No evidence was observed that either
A meta-​analysis including 20 studies (2124 cases) found a summary agent reduced the overall incidence of upper aerodigestive tract cancer.
OR of 1.40 (95% CI = 0.90, 2.16) for the GSTT1 null genotype (Ying However, an exploratory analysis found reduced incidence of laryngeal
et  al., 2012). A  meta-​analysis including Asian and Caucasian popu- cancer in subjects treated with β-​carotene (RR = 0.28; 95% CI = 0.10,
lations (2809 cases) (Xiao et  al., 2013)  reported a summary OR for 0.75). A second randomized trial that administered α-​tocopherol and
the GSTM1 deletion genotype of 1.44 (95% CI = 1.19, 1.73), with a β-​carotene supplements to head and neck cancer patients (225 laryn-
larger effect among Asians (OR = 1.89; 95% CI = 1.28, 2.77). Another geal cases in each trial arm; 82% of all patients in the treatment arm
meta-​analysis of GSTM1 (2562 cases) (Zhang et al., 2014) reported a and 84% of patients in the placebo arm) reported a higher incidence
summary OR of 1.22 (95% CI = 1.10, 1.36) and also a strong effect for rate of second tumors in the β-​carotene arm during supplementation
Asians (OR = 1.71; 95% CI = 1.34, 2.19; Caucasians: OR = 1.13; 95% (HR = 2.88; 95% CI = 1.56, 5.31), but a lower rate after the discon-
CI = 1.00, 1.27). A meta-​analysis of NAT2 genotypes (7 studies, 980 tinuation of treatment (HR = 0.41; 95% CI = 0.16, 1.03) (Bairati et al.,
cases (Ying et al., 2011) found an elevated OR for the NAT slow acety- 2005). A similar pattern was seen following supplementation with α-​
lator genotype among Asian studies (OR = 1.99; 95% CI = 1.10, 3.63). tocopherol. After 8 years, there was no difference between treatment
A meta-​analysis of 24 studies (1072 cases) reported no association arms in the rate of second tumors. Earlier studies had not reported a
between CYP2E1 SNPs and laryngeal cancer (Lu et al., 2011). A meta-​ reduction in the incidence of second primary tumors (Mayne et  al.,
analysis (1022 cases) reported weak associations for two CYP1A1 2001; Toma et al., 2003).
SNPs (Zhuo et  al., 2009). An Indian case control study (750 cases) Studies have not yet consistently identified a successful approach
noted elevated odds ratios for CYP1A1, CYP1B1, and CYP2E1 SNPs for chemoprevention of primary head and neck cancers or second
(Maurya et al., 2015). Elevated odds ratios were reported for CYP1B1 primary cancers. Most have focused on oral cancer and have not
(rs1056827) and CYP2E1 (rs3813867) SNPs in a Chinese case control been designed to examine laryngeal cancer separately. More recent
study (552 cases) (Jin et al., 2014). Another Chinses study (300 cases) chemoprevention studies of head and neck cancer have included
reported elevated odds ratios for CYP2C19 (Feng et al., 2011). molecular risk stratification, as in the Erlotinib Prevention of Oral
Genes involved in alcohol metabolism have been studied in relation Cancer (EPOC) study (William and El-​Naggar, 2015). With the rap-
to laryngeal cancer in several studies. Hashibe reported decreased odds idly evolving molecular characterization of head and neck tumors,
ratios of laryngeal or hypopharynx cancer (1659 cases) associated with chemoprevention may offer new opportunities to reduce the risk of
two alcohol dehydrogenase gene (ADH) SNPs, ADH1B (rs1229984; specific subtypes.
OR = 0.71; 95% CI = 0.57, 0.88) and ADH7 (rs1573496; OR = 0.78;
95% CI = 0.65, 0.93) (Hashibe et al., 2008). A case control study from
North Carolina (442 cases) evaluated SNPs of ADH1B, ADH1C, ADH4, Screening and Early Detection
ADH7, ALDH2, and CYP2E1 and found an elevated OR for ADH1B
The use of biomarkers such as saliva or blood to detect genetic altera-
(rs17028834; OR = 1.5; CI = 1.1, 2.0) (Hakenwerth et al., 2011).
tions involved in head and neck cancer, preferably detection at an
A European case control study (326 cases) examined genes in the
early stage, has been explored for at least two decades (Boyle et al.,
inflammatory pathway and found a decreased OR associated with
1994). The expanding list of genomic alterations in head and neck
a polymorphism of IL8 (rs4073; OR  =  0.70; 95% CI  =  0.50, 0.98)
cancers now offers additional opportunities for biomarker develop-
(Campa et al., 2007). A Brazilian study (237 cases) reported an inverse
ment. A recent proof-​of-​principle study (Wang et al., 2015) evaluated
association with an SNP involved in folic acid metabolism (SHMT1)
whether somatic mutations and HPV could be detected in the saliva
(OR = 0.48; 95% CI = 0.27, 0.86) (Succi et al., 2013). An analysis of
and plasma of patients with head and neck cancers. A  96% detec-
cases (n = 721) and controls from the M. D. Anderson Cancer Center
tion rate was achieved when both saliva and plasma were used. The
found an association with an SHTM1 haplotype (OR  =  1.64; 95%
results were best for oral cancers. For laryngeal cancers, mutations
CI = 1.12, 2.40) (Zhang et al., 2005).
were detected in 70% of patients in saliva samples, 86% in plasma,
There have been few genome-​wide association studies (GWAS) spe-
and 100% when both saliva and plasma were tested. All except two
cifically of laryngeal cancer. A  GWAS of pooled case control studies
of the laryngeal cancer patients had later stage disease. The value of
(2787 cases) reported associations for laryngeal cancer with ADH1B
these biomarkers for detecting early-​stage cancers of the larynx is
(rs1229984; OR  =  0.75; 95% CI  =  0.65, 0.86), ADH7 (rs1573496;
uncertain.
OR  =  0.79; 95% CI  =  0.71, 0.88), ADH1C (rs698; OR  =  1.06; 95%
CI = 0.99, 1.13), ALDH2 (rs4767364; OR = 1.07; 95% CI = 1.00, 1.15),
and a 4q21 variant (rs1494961; OR = 1.08; 95% CI = 1.02, 1.16) (McKay FUTURE RESEARCH
et  al., 2011). A  Chinese study identified three loci, including FADS1
(rs174549), AIF1 (rs2857595), and TBX5 (rs10492336), with significant Cigarette smoking remains by far the most important cause of laryn-
p-​values (x 10–​14 or greater) that could be replicated (Wei et al., 2014). geal cancer. The decreasing incidence of laryngeal cancer among
men in high-​income countries is encouraging. The incidence rate
PREVENTION among women is expected to decrease soon, consistent with the
delayed reduction in smoking among women. Additional efforts are
Primary Prevention needed to prevent the uptake of cigarette smoking and/​or other forms
of tobacco use and to promote cessation. Reductions in other risk
Alcohol control and tobacco control are discussed in Chapters 12 and factors are also important. These include heavy alcohol consump-
62.1, respectively, in this volume. tion, unhealthy dietary patterns, and certain occupational exposures.
 51

Cancer of the Larynx 515


Large epidemiologic studies are needed to further clarify whether Boyle JO, Mao L, Brennan JA, et al. 1994. Gene mutations in saliva as molec-
other factors also affect risk. These include HPV, GERD, diabetes, ular markers for head and neck squamous cell carcinomas. Am J Surg,
and obesity. Ideally, epidemiologic studies can be designed to assess 168(5), 429–​432. PMID: 7977967.
etiologic heterogeneity between these exposures and the molecular Bradshaw PT, Siega-​Riz AM, Campbell M, et al. 2012. Associations between
dietary patterns and head and neck cancer:  the Carolina head and
and topographic subsites of laryngeal cancer. It would be important neck cancer epidemiology study. Am J Epidemiol, 175, 1225–​ 1233.
to know whether the histological distribution of laryngeal cancer has PMID: 22575416.
changed, given the rise in smoking-​related adenocarcinomas and the Califano J, van der Riet P, Westra W, et al. 1996. Genetic progression model for
decrease in squamous cell carcinomas observed for lung and laryn- head and neck cancer: implications for field cancerization. Cancer Res,
geal cancers. 56(11), 2488–​2492. PMID: 8653682.
Genomic studies of both germline and somatic tumor tissue have Campa D, Hashibe M, Zaridze D, et  al. 2007. Association of common poly-
provided intriguing clues in recent years. Further replication and morphisms in inflammatory genes with risk of developing cancers of
refinement of this work will require adequately sized studies that allow the upper aerodigestive tract. Cancer Causes Control, 18, 449–​ 455.
laryngeal cancers to be separated from oral cavity and oropharyngeal PMID: 17356794.
Cancer Genome Atlas Network. 2015. Comprehensive genomic characteriza-
cancers. Large studies are also a requirement to characterize molecu- tion of head and neck squamous cell carcinomas. Nature, 517(7536),
lar subtypes. Recent research with biomarkers has shown promise for 576–​582. PMCID: PMC25631445.
the detection of head and neck cancers. Further studies are needed Carter BD, Freedman ND, and Jacobs EJ. 2015. Smoking and mortality: beyond
to confirm whether genomic markers can be used for early detection, established causes. N Engl J Med, 372(22), 2170. PMID: 26017836.
markers of prognosis, or therapeutic targets. Chaturvedi A, and Gillison M. 2010. Human papillomavirus and head and neck
Although trials of chemopreventive agents have not as yet been suc- cancer In: Olshan AF (Ed.), Epidemiology, pathogenesis, and prevention
cessful in preventing the transformation of pre-​malignant lesions into of head and neck cancer (pp. 87–​116). New York: Springer.
laryngeal cancer or in reducing the incidence of second tumors, this Chen M, and Tse LA. 2012. Laryngeal cancer and silica dust exposure:  a
may change as new agents are identified and molecular markers are systematic review and meta-​analysis. Am J Ind Med, 55(8), 669–​676.
PMID: 22457229.
further characterized. Chen W, Wang ZY, Xu FL, et al. 2013. Association of XRCC1 genetic poly-
In summary, cigarette smoking and the use of other combustible morphism (Arg399Gln) with laryngeal cancer: a meta-​analysis based on
tobacco products are the main cause of laryngeal cancer, accounting 4,031 subjects. Tumor Biology, 35, 1637–​1640. PMID: 24194393.
for the majority of cases. As such, research and implementation of Chuang S-​C, Jenab M, Heck JE, et al. 2012. Diet and the risk of head and neck
measures to reduce exposure to tobacco products is of utmost impor- cancer: a pooled analysis in the INHANCE consortium. Cancer Causes
tance to accelerating the decline of this cancer worldwide. Control, 23, 69–​88. PMID: 22037906.
Chung CH, Parker JS, Karaca G, et al. 2004. Molecular classification of head
and neck squamous cell carcinomas using patterns of gene expression.
References Cancer Cell, 5(5), 489–​500. PMID: 15144956.
Agudo A, Bonet C, Travier N, et al. 2012. Impact of cigarette smoking on can- Conway DI, Brenner DR, Mcmahon AD, et  al. 2014. Estimating and
cer risk in the European prospective investigation into cancer and nutri- explaining the effect of education and income on head and neck
tion study. J Clin Oncol, 30, 4550–​4557. PMID: 23169508. cancer risk:  INHANCE consortium pooled analysis of 31 case-​
American Cancer Society. 2016. Cancer facts & ­figures 2016. Atlanta: American control studies from 27 countries. Int J Cancer, 44, 1125–​1139.
Cancer Society. PMID: 24996155.
Anantharaman D, Gheit T, Waterboer T, et  al. 2013. Human papillomavirus De Stefani E, Boffetta P, Correa P, et  al. 2013. Dietary patterns and risk of
infections and upper aero-​digestive tract cancers: the ARCAGE study. J cancers of the upper aerodigestive tract:  a factor analysis in Uruguay.
Natl Cancer Inst, 105, 536–​545. PMID: 23503618. Nutrition Cancer, 65, 384–​389. PMID: 23530637.
Australian Cancer Database. 2015. Australian Institute of Health and Welfare. De Stefani E, Boffetta P, Ronco AL, et al. 2007. Dietary patterns and risk of
Available from: http://​www.aihw.gov.au/​. laryngeal cancer: an exploratory factor analysis in Uruguayan men. Int J
Atchison EA, Gridley G, Carreon JD, Leitzmann MF, and Mcglynn KA. 2011. Cancer, 121, 1086–​1091. PMID: 17437274.
Risk of cancer in a large cohort of U.S.  veterans with diabetes. Int J Di Maso M, Talamini R, Bosetti C, et al. 2013. Red meat and cancer risk in a
Cancer, 128, 635–​643. PMCID: PMC2962873. network of case-​control studies focusing on cooking practices. Annals
Bagnardi V, Rota M, Botteri E, et  al. 2014. Alcohol consumption and site-​ Oncology, 24, 3107–​3112. PMID: 24121119.
specific cancer risk: a comprehensive dose–​response meta-​analysis. Br J Dietz A, Ramroth H, Urban T, Ahrens W, and Becher H. 2004. Exposure to
Cancer, 112, 580–​593. PMCID: PMC4453639. cement dust, related occupational groups and laryngeal cancer risk: results
Bairati I, Meyer F, Gelinas M, et al. 2005. A randomized trial of antioxidant of a population based case-​control study. Int J Cancer, 108, 907–​911.
vitamins to prevent second primary cancers in head and neck cancer PMID: 14712496.
patients. J Natl Cancer Inst, 97(7), 481–​488. PMID: 15812073. Edefonti V, Hashibe M, Parpinel M, et al. 2015. Natural vitamin C intake and
Bartlett RS, Heckman WW, Isenberg J, Thibeault SL, and Dailey SH. 2012. the risk of head and neck cancer: a pooled analysis in the International
Genetic characterization of vocal fold lesions:  leukoplakia and carci- Head and Neck Cancer Epidemiology Consortium. Int J Cancer, 137(2),
noma. Laryngoscope, 122, 336–​342. PMCID: PMC3389785. 448–​462. PMCID: PMC4428957.
Becker C, Jick SS, Meier CR, and Bodmer M. 2014. Metformin and the risk of Engholm G. 2015. NORDCAN:  Cancer incidence, mortality, prevalence
head and neck cancer: a case-​control analysis. Diabetes Obes Metab, 16, and survival in the Nordic Countries, Version 7.1 (07.2015). Available
1148–​1154. PMID: 25041125. from: http://​www.ancr.nu. Accessed October 2015.
Berthiller J, Lee YCA, Boffetta P, Wei Q, et al. 2009. Marijuana smoking and the Etemadi A, O’Doherty MG, Freedman ND, et al. 2014. A prospective cohort
risk of head and neck cancer: pooled analysis in the INHANCE Consortium. study of body size and risk of head and neck cancers in the NIH-​AARP
Cancer Epidemiol Bio Prev, 18, 1544–​1551. PMID: 20519118. Diet and Health Study. Cancer Epidemiol Biomarkers Prev, 23, 2422–​
Besson H, Banks R, and Boffetta P. 2006. Cancer mortality among butch- 2429. PMID: 25172872.
ers: a 24-​state death certificate study. J Occup Envir Med, 48, 289–​293. Feng J, Li L, Zhao YS, et al. 2011. Interaction between CYP 2C19*3 polymor-
PMID: 16531833. phism and smoking in relation to laryngeal carcinoma in the Chinese Han
Bosetti C, Filomeno M, Riso P, et al. 2012. Cruciferous vegetables and can- population. Genet Molec Res, 10, 3331–​3337. PMID: 22183943.
cer risk in a network of case-​control studies. Ann Oncol, 23, 2198–​2203. Ferlay J. 2014a. Cancer incidence in five continents, CI5plus. IARC CancerBase
PMID: 22328735. No. 9. Available from: http://​ci5.iarc.fr.
Bosetti C, Gallus S, Peto R, et al. 2008. Tobacco smoking, smoking cessation, Ferlay J, Bray F, Steliarova-​Foucher E, and Forman D. 2014b. Cancer incidence
and cumulative risk of upper aerodigestive tract cancers. Am J Epidemiol, in five continents, CI5plus. IARC CancerBase No. 9. Lyon: International
167, 468–​473. PMID: 18056925. Agency for Research on Cancer.
Bosetti C, Rosato V, Polesel J, et al. 2012. Diabetes mellitus and cancer risk Ferlay J, Soerjomataram I, Dikshit R, et al. 2015. Cancer incidence and mor-
in a network of case-​control studies. Nutrition Cancer, 64, 643–​651. tality worldwide: sources, methods and major patterns in GLOBOCAN
PMID: 22519904. 2012. Int J Cancer, 136(5), E359–​386. PMID: 25220842.
Bouquot JE, and Gnepp DR. 1991. Laryngeal precancer: a review of the lit- Foschi R, Pelucchi C, Dal Maso L, et al. 2010. Citrus fruit and cancer risk in
erature, commentary, and comparison with oral leukoplakia. Head Neck, a network of case-​control studies. Cancer Causes Control, 21, 237–​242.
13(6), 488–​497. PMID: 1791144. PMID: 19856118.
516

516 PART IV:  Cancers by Tissue of Origin


Freedman ND, Abnet CC, Leitzmann MF, Hollenbeck AR, and Schatzkin Jin J, Lin F, Liao S, Bao Q, and Ni L. 2014. Effects of SNPs (CYP1B1*2
A. 2007. Prospective investigation of the cigarette smoking-​ head G355T, CYP1B1*3 C4326G, and CYP2E1*5 G-​1293C), smoking, and
and neck cancer association by sex. Cancer, 110, 1593–​ 1601. drinking on susceptibility to laryngeal cancer among Han Chinese. PLoS
PMID: 17724671. ONE, 9, e106580. PMCID: PMC4191961.
Fritz A Perry C, Jack A, et al., eds. 2013. International classification of diseases Jones DR, Sutton AJ, Abrams KR, et al. 2009. Systematic review and meta-​
for oncology. 3rd ed. 1st Revision Geneva: World Health Organization. analysis of mortality in crop protection product manufacturing workers. J
Galeone C, Turati F, and Zhang Z. 2015. Relation of allium vegetables intake Occup Environ Med, 66, 7–​15. PMID: 19095707.
with head and neck cnancers: evidence from the INHANCE consortium. Kapil U, Singh P, Bahadur S, et al. 2005. Assessment of risk factors in laryn-
Molec Nutrit Food Res, 59, 1641–​50. PMCID: PMC4579039. geal cancer in India: a case-​control study. Asian Pac J Cancer Prev, 6(2),
Gaudet MM, Kitahara CM, Newton CC, et al. 2015. Anthropometry and head 202–​207. PMID: 16101334.
and neck cancer:a pooled analysis of cohort data. Int J Epidemiol, 44, Karatayli-​ozgursoy S, Pacheco-​lopez P, Hillel AT, et al. 2015. Laryngeal dys-
673–​681. PMCID: PMC4481608. plasia, demographics, and treatment a single-​institution, 20-​year review.
Gaudet MM, Olshan AF, Chuang S-​C, et al. 2010. Body mass index and risk JAMA Otolaryngol Head Neck Surg, 141, 313–​8. PMID: 25654369.
of head and neck cancer in a pooled analysis of case-​control studies in Lacko M, Roelofs HM, te Morsche RH, et al. 2009. Genetic polymorphisms
the International Head and Neck Cancer Epidemiology (INHANCE) in the tobacco smoke carcinogens detoxifying enzyme UGT1A7
Consortium. Int J Epidemiol, 39, 1091–​1102. PMID: 20123951. and the risk of head and neck cancer. Head Neck, 31, 1274–​1281.
Guha N, Boffetta P, Wünsch Filho V, et al. 2007. Oral health and risk of squa- PMID: 19602651.
mous cell carcinoma of the head and neck and esophagus: results of two Lacko M, Voogd AC, Roelofs HM, et  al. 2013. Combined effect of genetic
multicentric case-​ control studies. Am J Epidemiol, 166, 1159–​ 1173. polymorphisms in phase I and II biotransformation enzymes on head and
PMID: 17761691. neck cancer risk. Head Neck, 35, 858–​867. PMID: 19602651.
Hakenwerth AM, Millikan RC, Rusyn I, et  al. 2011. Joint effects of alcohol Lagergren J, and Lindam A. 2012. Increased risk of laryngeal and pharyngeal
consumption and polymorphisms in alcohol and oxidative stress metabo- cancer after gastrectomy for ulcer disease in a population-​based cohort
lism genes on risk of head and neck cancer. Cancer Epidemiol Biomarkers study. Br J Cancer, 106, 1342–​1345. PMID: 22453126.
Prev, 20, 2438–​2449. PMCID: PMC3210881. Langevin SM, Sullivan MHO, Valerio JL, et  al. 2013. Occupational asbes-
Halec G, Holzinger D, Schmitt M, et al. 2013. Biological evidence for a causal tos exposure is associated with pharyngeal squamous cell carcinoma in
role of HPV16 in a small fraction of laryngeal squamous cell carcinoma. men from the greater Boston area. J Occup Environ Med, 70, 858–​863.
Br J Cancer, 109, 172–​183. PMID: 23778529. PMCID: PMC4227396.
Hall J, Hashibe M, Boffetta P, et al. 2007. The association of sequence variants Lee CH, Hyun MK, Jang EJ, et al. 2013. Inhaled corticosteroid use and risks
in DNA repair and cell cycle genes with cancers of the upper aerodiges- of lung cancer and laryngeal cancer. Respir Med, 107, 1222–​ 1233.
tive tract. Carcinogenesis, 28, 665–​671. PMID: 17040931. PMID: 23768737.
Hamdi A. 2013. Incidences, trends, survival. Available from:  http://​www. Lee CH, Lee KW, Fang FM, et al. 2011. The use of tobacco-​free betel-​quid in
registre-​cancer-​setif.com. conjunction with alcohol/​tobacco impacts early-​onset age and carcinoma
Hashibe M, Boffetta P, Zaridze D, et al. 2007a. Contribution of tobacco and distribution for upper aerodigestive tract cancer. J Oral Pathol Med, 40,
alcohol to the high rates of squamous cell carcinoma of the supraglot- 684–​692. PMID: 21385213.
tis and glottis in Central Europe. Am J Epidemiol, 165, 814–​ 820. Lee YCA, Boffetta P, Sturgis EM, et al. 2008. Involuntary smoking and head
PMID: 17244634. and neck cancer risk. Cancer Epidemiol Biomarkers Prev, 17, 1974–​
Hashibe M, Brennan P, Benhamou S, et al. 2007b. Alcohol drinking in never 1981. PMID: 18708387.
users of tobacco, cigarette smoking in never drinkers, and the risk of Leoncini E, Edefonti V, Hashibe M, and Parpinel M. 2016. Carotenoid intake
head and neck cancer:  pooled analysis in the international head and and head and neck cancer:  a pooled analysis in the International Head
neck cancer epidemiology consortium. J Natl Cancer Inst, 99, 777–​789. and Neck Cancer Epidemiology Consortium. Eur J Epidemiology,31(4),
PMID: 17505073. 369–​383. PMID: 18708387.
Hashibe M, Brennan P, Chuang SC, et al. 2009. Interaction between tobacco and Leoncini E, Ricciardi W, Cadoni G, Arzani D, et al. Adult height and head and
alcohol use and the risk of head and neck cancer: pooled analysis in the neck cancer: a pooled analysis within the. Eur J Epidemiol, 29, 35–​48.
International Head and Neck Cancer Epidemiology Consortium. Cancer PMID: 24271556.
Epidemiol Biomarkers Prev, 18(2), 541–​550. PMCID: PMC3051410. Li Q, Chuang SC, Neto JE, et al. 2013. Vitamin or mineral supplement intake
Hashibe M, McKay JD, Curado MP, et al. 2008. Multiple ADH genes are asso- and the risk of head and neck cancer: pooled analysis in the INHANCE
ciated with upper aerodigestive cancers. Nature Genetics, 40, 707–​709. consortium. Int J Cancer, 131, 1686–​1699. PMCID: PMC3376697.
PMID: 18500343. Li X, Gao L, Li H, et  al. 2013. Human papillomavirus infection and laryn-
Heck JE, Berthiller J, Vaccarella S, et al. 2010. Sexual behaviours and the risk geal cancer risk: a systematic review and meta-​analysis. J Infect Dis, 207,
of head and neck cancers: a pooled analysis in the International Head and 479–​88. PMID: 23175764.
Neck Cancer Epidemiology (INHANCE) consortium. Int J Epidemiol, Lu B, Li J, Gao Q, et  al. 2014. Laryngeal cancer risk and common single
39, 166–​181. PMID: 20022926. nucleotide polymorphisms in nucleotide excision repair pathway genes
Henley SJ, Thun MJ, Chao A, and Calle EE. 2004. Association between exclu- ERCC1, ERCC2, ERCC3, ERCC4, ERCC5 and XPA. Gene, 542, 64–​68.
sive pipe smoking and mortality from cancer and other diseases. J Natl PMID: 24582975.
Cancer Inst, 96, 853–​861. PMID: 15173269. Lu D, Yu X, and Du Y. 2011. Meta-​analyses of the effect of cytochrome P450
Hernandez BY, Goodman MT, Lynch CF, et al. 2014. Human papillomavirus 2E1 gene polymorphism on the risk of head and neck cancer. Molec Biol
prevalence in invasive laryngeal cancer in the United States. PLoS ONE, Reports, 38, 2409–​2416. PMID: 21076874.
9, e115931. PMCID: PMC4278830. Lubin JH, Gaudet MM, Olshan AF, et  al. 2010. Body mass index, cigarette
Howlander N, Noone AM, Krapcho M, et  al. 2015. SEER Cancer Statistics smoking, and alcohol consumption and cancers of the oral cavity, phar-
Review, 1975–​2012, National Cancer Institute. Bethesda, MD, http://​seer. ynx, and larynx: Modeling odds ratios in pooled case-​control data. Am J
cancer.gov/​csr/​1975_​2012/​, based on November 2014 SEER data submis- Epidemiol, 171, 1250–​1261. PMID: 20494999.
sion, posted to the SEER website April 2015. Lucenteforte E, Garavello W, Bosetti C, and La Vecchia C. 2010. Dietary fac-
IARC. 2004. Tobacco smoke and involuntary smoking. IARC Monogr Eval tors. In: Olshan AF (Ed.), Epidemiology, pathogenesis, and prevention of
Carcinog Risks Hum, 83, 1182. PMID: 15285078. head and neck cancer (pp. 117–​136). New York: Springer.
Institute of Medicine. 2006. Asbestos: selected cancers. Washington, DC: IOM. Macfarlane TV, Macfarlane GJ, Thakker NS, et al. 2012. Role of medical his-
PMID: 20669440. tory and medication use in the aetiology of upper aerodigestive tract can-
Isenberg JS, Crozier DL, and Dailey SH. 2008. Institutional and comprehen- cers in Europe: The ARCAGE study. Annals Oncology, 23, 1053–​1060.
sive review of laryngeal leukoplakia. Ann Otol Rhinol Laryngol, 117(1), PMID: 21828376.
74–​79. PMID: 18254375. Makitie AA, and Monni O. 2009. Molecular profiling of laryngeal cancer.
Jaworowska E, Trubicka J, Lener MR, et  al. 2011. Smoking related cancers Expert Rev Anticancer Ther, 9(9), 1251–​1260. PMID: 19761429.
and loci at chromosomes 15q25, 5p15, 6p22.1 and 6p21.33 in the Polish Maurya SS, Tridiv K, Dhawan A, et al. 2015. Gene-​environment interactions
population. PLoS ONE, 6, 3–​8. PMID: 21966413. indetermining differences in genetic susceptibility to cancer in subsites of
Jayalekshmi PA, Nandakumar A, Akiba S, Gangadharan P, and Koriyama C. the head and neck. Envir Molec Mutagen, 56, 313–​321. PMID: 19844953.
2013. Associations of tobacco use and alcohol drinking with laryngeal Mayne ST, Cartmel B, Baum M, et al. 2001. Randomized trial of supplemental
and hypopharyngeal cancer risks among men in Karunagappally, Kerala, beta-​carotene to prevent second head and neck cancer. Cancer Res, 61(4),
India:Karunagappally Cohort Study. PLoS ONE, 8, 1–​8. PMID: 24015309. 1457–​1463. PMID: 11245451.
Ji J, and Hemminki K. 2005. Occupation and upper aerodigestive tract can- McKay JD, Truong T, Gaborieau V, et  al. 2011. A genome-​wide associa-
cers: a follow-​up study in Sweden. J Occup Environ Med, 47, 785–​795. tion study of upper aerodigestive tract cancers conducted within the
PMID: 16093928. INHANCE consortium. PLoS Genetics, 7. PMID: 21437268.
 517

Cancer of the Larynx 517


Negri E, Boffetta P, Berthiller J, et al. 2009. Family history of cancer: pooled Upile NS, Shaw RJ, Jones TM, et al. 2014. Squamous cell carcinoma of the
analysis in the International Head and Neck Cancer Epidemiology head and neck outside the oropharynx is rarely human papillomavirus
Consortium. Int J Cancer, 124, 394–​401. PMID: 18814262. related. Laryngoscope, 2739–​2744. PMID: 25042603.
Nicolotti N, Chuang S-​C, Cadoni G, et al. 2011. Recreational physical activity Vaezi MF, Qadeer Ma, Lopez R, and Colabianchi N. 2006. Laryngeal cancer
and risk of head and neck cancer: a pooled analysis within the interna- and gastroesophageal reflux disease: a case-​control study. Am J Med, 119,
tional head and neck cancer epidemiology (INHANCE) Consortium. Eur 768–​776. PMID: 16945612.
J Epidemiol, 26, 619–​628. PMID: 21842237. Walter V, Yin X, Wilkerson MD, et  al. 2013. Molecular subtypes in
Offermans NSM, Vermeulen R, Burdorf A, et al. 2014. Occupational asbestos head and neck cancer exhibit distinct patterns of chromosomal
exposure and risk of pleural mesothelioma, lung cancer, and laryngeal gain and loss of canonical cancer genes. PLoS ONE, 8(2), e56823.
cancer in the prospective Netherlands cohort study. J Occup Environ Med, PMCID: PMC3579892.
56, 6–​19. PMID: 24351898. Wang LE, Surgis E, and Wei Q. 2010. Host susceptibility and molecular epi-
Olshan AF. 2010. Occupation and other risk factors for head and neck cancer. demiology. In: Olshan AF (Ed.), Epidemiology, pathogenesis, and pre-
In: Olshan AF (Ed.), Epidemiology, pathogenesis, and prevention of head vention of head and neck cancer (pp. 155–​185). New York: Springer.
and neck cancer (pp. 137–​154). New York: Springer. Wang X, Lin S, Yu I, et  al. 2013. Cause-​specific mortality in a Chinese
Park SL, Bastani D, Goldstein BY, et al. 2010. Associations between NBS1 poly- chrysotile textile worker cohort. Cancer Science, 104, 245–​ 249.
morphisms, haplotypes and smoking-​related cancers. Carcinogenesis, 31, PMID: 23121131.
1264–​1271. PMID: 20478923. Wang Y, Chu X, Meng X, and Zou F. 2013. Association of X-​ray repair cross
Pednekar MS, Gupta PC, and Hebert JR. 2011. Association of tobacco hab- complementing group 1 Arg399Gln polymorphisms with the risk of squa-
its, including bidi smoking, with overall and site-​specific cancer inci- mous cell carcinoma of the head and neck:  evidence from an updated
dence:  results from the Mumbai cohort study. Cancer Causes Control, meta-​analysis. PloS ONE, 8, e77898. PMID: 24205020.
22, 859–​868. PMCID: PMC3756904. Wang Y, Springer S, Mulvey CL, et  al. 2015. Detection of somatic muta-
Pelucchi C, Talamini R, Levi F, et al. 2003. Fibre intake and laryngeal cancer tions and HPV in the saliva and plasma of patients with head and
risk. Annals Oncology, 14, 162–​167. PMID: 12488309. neck squamous cell carcinomas. Sci Transl Med, 7(293), 293ra104.
Purdue MP, Hashibe M, Berthiller J, et al. 2009. Type of alcoholic beverage PMID: 26109104.
and risk of head and neck cancer: a pooled analysis within the INHANCE WCRF/​AICR. 2007. World Cancer Research Fund /​American Institute for
Consortium. Am J Epidemiol, 169, 132–​142. PMID: 19064644. Cancer Research. Food, nutrition, and the prevention of cancer: a global
Purdue MP, Järvholm B, Bergdahl Ia, Hayes RB, and Baris D. 2006. Occupational perspective. Washington, DC: WCRF/​AICR.
exposures and head and neck cancers among Swedish construction work- Wei Q, Yu D, Liu M, et  al. 2014. Genome-​wide association study identi-
ers. Scand J Work Environ Health, 32, 270–​275. PMID: 16932824. fies three susceptibility loci for laryngeal squamous cell carci-
Ramroth H, and Dietz A. 2008. Occupational wood dust exposure and the risk noma in the Chinese population. Nature Genetics, 46, 1110–​1114.
of laryngeal cancer: a population based case-​control study in Germany. PMID: 25194280.
Am J Indust Med, 51, 648–​655. PMID: 18626911. Weller MD, Nankivell PC, McConkey C, Paleri V, and Mehanna HM. 2010.
Ramroth H, Dietz A, and Becher H. 2011. Intensity and inhalation of smoking The risk and interval to malignancy of patients with laryngeal dysplasia: a
in the aetiology of laryngeal cancer. Int J Environ Res Public Health, 8, systematic review of case series and meta-​analysis. Clin Otolaryngol,
976–​984. PMID: 21695025. 35(5), 364–​372. PMID: 21108746.
Rothenberg SM, and Ellisen LW. 2012. The molecular pathogenesis of head Wilkerson MD, Yin X, Hoadley KA, et  al. 2010. Lung squamous cell carci-
and neck squamous cell carcinoma. J Clin Invest, 122(6), 1951–​1957. noma mRNA expression subtypes are reproducible, clinically important,
PMCID: PMC3589176. and correspond to normal cell types. Clin Cancer Res, 16(19), 4864–​
Sapkota A, Gajalakshmi V, Jetly DH et al. 2007. Smokeless tobacco and increased 4875. PMCID: PMC2953768.
risk of hypopharyngeal and laryngeal cancers: a multicentric case-​control William WN, Jr. 2010. Chemoprevention of head and neck cancers. In: Olshan
study from India. Int J Cancer, 121, 1793–​1798. PMID: 17583577. AF (Ed.), Epidemiology, pathogenesis, and prevention of head and neck
Saraiya M, Unger ER, Thompson TD, et al. 2015. US assessment of HPV types cancer (pp. 187–​202). New York: Springer.
in cancers:  implications for current and 9-​valent HPV vaccines. J Natl William WN, Jr, and El-​Naggar AK. 2015. A novel target for oral cancer che-
Cancer Inst, 107. PMID: 25925419. moprevention? Notch quite, yet. Cancer Prev Res (Phila), 8(4), 262–​265.
SEER. Surveillance, Epidemiology, and End Results Program. 2015. (www.seer. PMCID: PMC4383667.
cancer.gov) SEER*Stat Database: Incidence—​SEER 9 Regs Research Data, Wright ME, Virtamo J, Hartman AM, et  al. 2007. Effects of alpha-​tocopherol
Nov 2014 Sub (1973–​2012) Katrina/​Rita Population Adjustment—​Linked and beta-​ carotene supplementation on upper aerodigestive tract can-
to County Attributes—​Total US, 1969–​2013 Counties, National Cancer cers in a large, randomized controlled trial. Cancer, 109, 891–​ 898.
Institute, DCCPS, Surveillance Research Program, Surveillance Systems PMID: 17265529.
Branch, released April 2015, based on the November 2014 submission. Wyss A, Hashibe M, Chuang SC, et al. 2013. Cigarette, cigar, and pipe smoking
Shangina O, Brennan P, Szeszenia-​Dabrowska N, et  al. 2006. Occupational and the risk of head and neck cancers: pooled analysis in the International
exposure and laryngeal and hypopharyngeal cancer risk in Central and Head and Neck Cancer Epidemiology Consortium. Am J Epidemiol,
Eastern Europe. Am J Epidemiol, 164, 367–​375. PMID: 16801374. 178(5), 679–​690. PMCID: PMC3755640.
Shapiro JA, Jacobs EJ, and Thun MJ. 2000. Cigar smoking in men and risk of Xiao H, Li M, Tian L, et al. 2013. Quantitative assessment of the association
death from tobacco-​related cancers. J Natl Cancer Inst, 92(4), 333–​337. between GSTM1 null genotype and laryngeal cancer risk. Eur Arch Oto-​
PMID: 10675383. Rhino-​Laryngol, 270, 615–​622. PMID: 22972496.
Succi M, de Castro TB, Galbiatti ALS, et  al. 2013. DNMT3B C46359T and Ying XJ, Dong P, Shen B, et al. 2011. Possible association of NAT2 polymor-
SHMT1 C1420T polymorphisms in the folate pathway in carcinogenesis of phism with laryngeal cancer risk:  an evidence-​based meta-​analysis. J
head and neck. Molecular Biology Reports, 41, 581–​589. PMID: 24362509. Cancer Res Clin Oncol, 137, 1661–​1667. PMID: 21877196.
Sun Y, Tan L, Li H, Qin X, and Liu J. 2015. Association of NER pathway gene Ying XJ, Xu CZ, Xu HM, et al. 2012. Lack of association between glutathione
polymorphisms with susceptibility to laryngeal cancer in a Chinese popu- S-​transferase T1 gene polymorphism and laryngeal cancer susceptibil-
lation. Int J Clin Exp Pathol, 8, 11615–​11621. PMCID: PMC4637715. ity: a meta-​analysis based on 2,124 cases and 2,059 controls. Gene, 494,
Toma S, Bonelli L, Sartoris A, et al. 2003. Beta-​carotene supplementation in 11–​16. PMID: 22207034.
patients radically treated for stage I-​II head and neck cancer: results of a Zhang Y, Chen W, Ji J-​F, et al. 2014. GSTM1 null polymorphisms is associated
randomized trial. Oncol Rep, 10(6), 1895–​1901. PMID: 14534715. with laryngeal cancer risk:  a meta-​analysis. Tumor Biology, 35, 6303–​
Toporcov TN, Znaor a, Zhang Z-​F, et al. 2015. Risk factors for head and neck 6309. PMID: 24643684.
cancer in young adults: a pooled analysis in the INHANCE consortium. Zhang Z, Shi Q, Sturgis EM, Spitz MR, and Wei Q. 2005. Polymorphisms and
Int J Epidemiol, 1–​17. PMCID: PMC4339764. haplotypes of serine hydroxymethyltransferase and risk of squamous cell
Turati F, Edefonti V, Bosetti C, et al. 2013. Family history of cancer and the carcinoma of the head and neck: a case-​control analysis. Pharmacogenet
risk of cancer: a network of case-​control studies. Annals Oncology, 24, Genomics, 15, 557–​564. PMID: 16006999.
2651–​2656. PMID: 23884440. Zhuo WL, Wang Y, Zhuo XL, et  al. 2009. Polymorphisms of CYP1A1 and
United Kingdom, England, Office for National Statistics. Available from: http://​ GSTM1 and laryngeal cancer risk:  evidence-​ based meta-​ analyses. J
www.ons.gov.uk/​ons/​. Cancer Res Clin Oncol, 135, 1081–​1090. PMID: 19252926.
518
 519

28 Lung Cancer

MICHAEL J. THUN, S. JANE HENLEY, AND WILLIAM D. TRAVIS

OVERVIEW (ICD-​10 codes C33–​C34:  ICD-​O-​2/​3 codes C33.9–​C34.9). Our dis-


cussion is limited to carcinomas, which comprise the great majority of
Lung cancer is the most common cancer worldwide, ranking first lung cancers. Other primary malignancies that occur in or adjacent to
in men and third in women for new cases and first in both sexes for the lung include carcinoid tumors, accounting for less than 1% of lung
deaths. Dynamic global patterns in lung cancer incidence predomi- cancers (Chapter 35), pleural mesothelioma (Chapter 16), lymphomas
nantly reflect past and current patterns of cigarette smoking. Lung (Chapters 39, 40), and Kaposi sarcoma (Chapters 24, 25).
cancer incidence rates in most high-​income countries have decreased We first discuss lung cancer as a global public health problem, con-
substantially among men, but continue to increase at older ages in sidering how the occurrence of all forms of lung cancer combined
women. More than half of all cases occur in low-​and middle-​income varies by gender, birth cohort, and level of economic development.
countries where cigarette smoking remains common, especially among We then describe the three main histologic subtypes of lung cancer
men. In high-​income countries, strong birth cohort patterns dominate with respect to their clinical and epidemiologic features and molecu-
temporal trends in lung cancer incidence and mortality; these paral- lar pathogenesis. We consider how the changing histologic and topo-
lel birth cohort patterns in the uptake in cigarette smoking 50 years graphic distribution of these subtypes relates to changes in tumor
earlier. In the United States and the United Kingdom, the average classification as well as in cigarette design and composition. We then
relative risk (RR) for lung cancer mortality associated with continued discuss the known and suspected causes of lung cancer, staging, sur-
cigarette smoking increased across birth cohorts first among men and vival, prevention strategies including screening, and future research
then women. Now, in contemporary cohorts, the relative risk for cur- directions.
rent smokers is about 25 in both sexes. The histologic and topographic
distribution of lung cancer also has changed, corresponding to changes
in cigarette design and composition. Unlike smoking cessation, which DISEASE BURDEN
dramatically reduces risk, design changes in cigarettes that dilute the
smoke provide no health benefit. In the United States, active cigarette Lung cancer is the most common cancer worldwide, accounting for an
smoking accounts for an estimated 95% of lung cancer cases among estimated 1.8 million new cases and 1.6 million deaths in 2012 (Ferlay
smokers and 80% to 85% in the general population; secondhand et al., 2013). Globally, it ranks first in men and third in women in terms
smoke causes an estimated 7,300 lung cancer deaths among never of incident cases and first in both sexes for cancer deaths. In high-​income
smokers. The International Agency for Research on Cancer (IARC) countries, where incidence and mortality rates are decreasing in men of all
has designated at least 7 occupations and 18 exposures other than ages and in younger women (Peto et al., 2015), it remains the third lead-
active smoking, such as secondhand smoke, radon gas, diesel exhaust, ing cause of death from any cause, after heart disease and stroke (Lozano
and particulate air pollution as carcinogenic to humans for lung cancer. et  al., 2012). The number of deaths from lung cancer in high-​income
If lung cancer in never smokers were classified as a separate entry, it countries exceeds the combined number of deaths from cancers of the
would rank among the 10 most common cancers in the United States. colorectum, breast, and prostate (Torre et  al., 2015). Historically, lung
Targeted therapies have improved survival for a subset of lung can- cancer became the leading cause of cancer deaths in the United States in
cer patients with adenocarcinoma and mutations in EGRF or fusions the mid-​1950s among men and in 1987 in women (Siegel et al., 2015).
involving ALK or ROS, although most patients develop resistance to In LMICs, the lung cancer burden is increasing rapidly in men but
these agents over time. not women. This increase is caused by the high prevalence of cigarette
smoking in males, low smoking cessation rates, and population aging.
Massive numbers of young adult males in LMICs are smokers and are
INTRODUCTION now surviving into middle and older ages when lung cancer becomes
common (see Chapter 11).
Lung cancer occurrence has changed more in the last century than
any other cancer. In high-​income countries, the large increase in lung
cancer mortality rates, first observed among men after World War DESCRIPTIVE EPIDEMIOLOGY
I and women after World War II, has dominated cancer death rates for
decades. Similarly, the downturn in lung cancer incidence and mortal- In most countries, lung cancer occurrence predominantly reflects cur-
ity rates among men in most economically developed countries during rent and lifetime patterns of cigarette smoking. Variations in lung can-
the last 10–​25 years has exceeded the decrease in any other cancer. cer incidence and mortality are used as sentinel indicators of the impact
In contrast, the lung cancer burden in low-​and middle-​income of current and cumulative tobacco smoking in economically developed
countries (LMICs) continues to increase rapidly in terms of the num- countries (Peto et  al., 1992, 2017; Thun et  al., 2012). This approach
ber of cases and deaths, and now accounts for more than half of all is not applicable to all populations, however. For example, women in
lung cancer cases worldwide. Tobacco smoking remains highly preva- China have much higher lung cancer rates than would be expected from
lent among men in LMICs; exposure to other causes of lung cancer in their low lifetime history of active smoking (Jha and Peto, 2014).
occupational and community settings are also much higher in LMICs
than in economically developed countries. Among women in China,
Geographic Variation
for example, lung cancer incidence is high, despite a low prevalence
of active smoking. Figure 28–​1 shows the variation in lung cancer incidence and mor-
This chapter discusses the epidemiology of primary cancers of the tality rates by gender and WHO region, according to Globocan esti-
trachea, bronchus, and lung, commonly referred to as “lung” cancers mates for 2012 (Ferlay et al., 2013). The geographic patterns were

519
520

520 PART IV:  Cancers by Tissue of Origin


Male Female
North America
Micronesia
Eastern Asia
More developed regions
Western Europe
Central and Eastern Europe
Southern Europe
Northern Europe
Australia/New Zealand
Polynesia
World
Western Asia
Less developed regions
Southeast Asia
Caribbean
Southern Africa
South America
Melanesia
Northern Africa
South-Central Asia
Central America
Eastern Africa
Middle Africa
Western Africa
60 40 20 0 20 40 60
Incidence Mortality

Figure 28–​1.  Age-​standardized lung cancer incidence and mortality rates (per 100,000) by gender and WHO geographic region, as estimated for 2012.
Source: Ferlay et al. (2013).

similar for incidence and mortality rates. Lung cancer incidence et al., 2003). Lung cancer mortality rates in this age group and trends
rates varied by 25-​fold in men and 34-​fold in women across WHO in the rates over a 10-​year period were strongly and inversely corre-
regions. In men, the highest incidence rates (per 100,000) were in lated with the index of state tobacco control activities.
Central and Eastern Europe (53.5) and Eastern Asia (50.4), whereas
in women the rates were highest in Northern America (33.8) and
Northern Europe (23.7). Notably low incidence rates (per 100,000) Temporal Variation
were observed in Middle and Western Africa for both men (2.0) The increase in lung cancer occurrence during the first half of the
and women (1.7). More than half (58%) of cases newly diagnosed twentieth century was captured largely by mortality statistics, since
in 2012 occurred in LMICs; about one-​third occurred in China very few population-​based cancer incidence registries were then in
(Tobacco Atlas, 2015). existence. The temporal patterns are similar for incidence and mortal-
Gender-​specific maps of the age-​standardized lung cancer inci- ity data however. Figure 28–​3 shows the trends in gender-​specific inci-
dence rates (per 100,000) are shown by country in 2012 (Figure 28–​2) dence rates in selected high-​and middle-​income countries from 1954
(Ferlay et  al., 2013). (It should be noted that the scale used to dis- to 2005, based on Cancer Incidence in Five Continents (Parkin et al.,
play the rates differs for men and women.) In men, the incidence rates 2014). We first discuss the trends in age-​standardized incidence rates
in 2012 were highest in Central and Eastern Europe and in Eastern in these countries, and then illustrate how age-​specific rates and birth
Asia; among women, the rates were highest in Denmark, the United cohort trends provide an earlier indication of changes in smoking-​
Kingdom, Canada, and the United States. The geographic patterns of related lung cancer than age-​standardized rates.
lung cancer incidence shown here correspond closely to the patterns
of daily cigarette smoking among men and women in 2012, shown in
Chapter 11 (Figure 11–​2). Age-​Standardized Incidence Rates
Lung cancer incidence and mortality rates also vary substantially
within many countries. For example, in the United States, incidence Much of the long-​term increase in lung cancer occurrence among men
and mortality rates vary by more than 3-​fold among states (American preceded the advent of population-​based cancer incidence registries.
Cancer Society, 2016). Researchers have used the regional variation to An exception to this is the National Cancer Registry in Denmark,
assess the impact of tobacco control activities at the state level. Jemal which captures several decades of the protracted increase in lung can-
and colleagues correlated an index of state tobacco control activities cer incidence in men as well as women (Figure 28–​3). In all of the
with lung cancer death rates in young adults (ages 30–​39  years) to high-​income countries for which data are shown, the dominant feature
assess the early effects of state tobacco control efforts on risk (Jemal among men is the large decrease in the age-​standardized lung cancer
 521
Men Women

0 20 40 60 80 0 10 20 30 40

Figure 28–​2.  Map of age-​standardized lung cancer incidence rates (per 100,000) in men and women* by country of residence, 2012. *Note that scale differs for men and women.
Source: Ferlay et al. (2013).
52

522 PART IV:  Cancers by Tissue of Origin


(a) Males (b) Females
140 45

40
120
US Black
US Black
35

100
30

UK, Scotland

Rate per 100,000


Rate per 100,000

80
25

UK, Scotland
Italy
US White 20 US White
60

Denmark Australia 15
Australia
40
Italy
10

Denmark
20
5
India India

0 0
1954 1964 1974 1984 1994 2004 1955 1965 1975 1985 1995 2005
Year Year

Figure 28–​3.  Trends in lung cancer incidence in selected countries among males and females. Source: Ferlay et al. (2013).

incidence rate, beginning in the mid-​1980s. This contrasts with the ages than are age-​standardized rates. Birth cohort patterns represent
continuing increase in incidence among women in Italy, Denmark, the age-​specific incidence rates among people born in the same 5-​or
and Australia, and the absence of a major change in lung cancer inci- 10-​year calendar period as they advance together through age and
dence in either sex in India. In the United Kingdom (Scotland) and calendar time. Birth cohort patterns are highly informative about the
among US blacks, the incidence rates in women reached a plateau dur- evolution of tobacco smoking in the United States, as discussed in
ing the 1990s. Only among whites in the United States did the age-​ Chapter 11. They reflect shared patterns of tobacco use that people in
standardized lung cancer incidence rate begin to decrease in women as the same birth cohort acquire as they pass through critical periods of
well as men during the time period shown. life. The birth cohort patterns of smoking are mirrored by similar pat-
Not shown in Figure 28–​3 are the decreasing trends in the age-​ terns in lung cancer risk, 20–​50 years later.
standardized lung cancer incidence rates among men in many other Figure 28–​4 shows birth cohort patterns in lung cancer mortality
high-​and middle-​income countries, including Hungary, Slovakia, and rates by sex in the US general population from 1930 to 2009. In men,
others in Eastern Europe (Ferlay J et al., 2013). It is not yet possible to the age-​specific death rates increase from the birth cohort 1870–​1874
assess the long-​term impact of smoking on lung cancer among men in to 1910–​1914, and then decrease in later birth cohorts. In women, the
China (Zhao et al., 2010) or Japan (Funatogawa et al., 2013). Access age-​specific death rates continue to increase until at least the birth
to cigarettes in these countries was limited during and after World War cohort 1930–​1934. In both sexes, the slope of the initial increase in
II, and widespread cigarette smoking began considerably later than in lung cancer before age 40 years appears to flatten in birth cohorts after
the West. In general, middle-​and low-​income countries have limited 1910–​1914 in men and after 1930–​1934 in women. This is consist-
historical data on cigarette smoking, and even past trends in lung can- ent with a reduction in smoking-​related lung cancer at younger ages,
cer are difficult to assess. The age-​standardized death rate from lung following reductions in smoking initiation and, more recently, in
cancer is reportedly increasing among men in Uganda, as represented consumption.
by the population-​based cancer registry (Ferlay et al., 2013). Birth cohort patterns and age-​specific rates at younger ages are
informative in several ways. First, they show the same wave-​like
progression in lung cancer risk across successive birth cohorts of
Age-​Specific and Birth Cohort Trends men and women that characterizes the trends in smoking prevalence
As noted, age-​specific rates and birth cohort trends are more sensi- (Holford et  al., 2014)  (Chapter  11). Second, they provide an earlier
tive to changes in smoking behavior and lung cancer risk at younger indication of the changing impact of smoking than age-​standardized
 523

Lung Cancer 523


(a) Males (b) Females
600 600

500 500

400 400
Deaths per 100,000

Deaths per 100,000


300 300

200 200

100 100

1880
1870 1880
1870
40
00

20

30

50
90

10

60

60
50
40
19

19 0

30
20
19

19

19

19
18

19

19

0
1

189900

19
19

19
19

1 19

0 0
1930– 1940– 1950– 1960– 1970– 1980– 1990– 2000– 1930– 1940– 1950– 1960– 1970– 1980– 1990– 2000–
1934 1944 1954 1964 1974 1984 1994 2004 1934 1944 1954 1964 1974 1984 1994 2004

Calendar Period of Death Calendar Period of Death

Figure 28–​4.  Birth cohort patterns in lung cancer death rates by sex, United States, 1930–​2009. Source: National Center for Health Statistics (2013a).

rates or age-​specific rates at older ages. Third, they help to inform the 1983; Holford et al., 2014). These statistical models provide a some-
interpretation of trends in age-​standardized cancer rates in the general what more formal approach for partitioning age, cohort, and period
population. effects, based on a log-​linear model of temporal trends in cancer
Figure 28–​5 shows the age-​standardized lung cancer death rates for rates. The

You might also like