You are on page 1of 37

Critical Reviews in Oncology/Hematology 96 (2015) 220256

The role of ABC transporters in ovarian cancer progression and


chemoresistance
M.P. Ween a , M.A. Armstrong b , M.K. Oehler c,d , C. Ricciardelli d,
a

Lung Research, Hanson Institute and Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide
b Data Management and Analysis Centre, University of Adelaide, Australia
c Gynaecological Oncology Department, Royal Adelaide Hospital, Australia
School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, Australia
Accepted 18 May 2015

Contents
1.
2.

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 221
ABC transporters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 222
2.1. The ABCA family . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 222
2.1.1. ABCA1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 222
2.1.2. ABCA2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 222
2.1.3. ABCA3 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 225
2.1.4. ABCA4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
2.1.5. ABCA5 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
2.1.6. ABCA6 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227
2.1.7. ABCA7 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227
2.1.8. ABCA8 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227
2.1.9. ABCA9 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227
2.1.10. ABCA10 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227
2.1.11. ABCA12 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227
2.2. The ABCB family . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227
2.2.1. ABCB1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227
2.2.2. ABCB2 and ABCB3 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 230
2.2.3. ABCB4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 230
2.2.4. ABCB5 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 231
2.2.5. ABCB6 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 231
2.2.6. ABCB7 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 232
2.2.7. ABCB8 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 232
2.2.8. ABCB9 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 232
2.2.9. ABCB10 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 232
2.2.10. ABCB11 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 232
2.3. The ABCC family . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 232
2.3.1. ABCC1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 232
2.3.2. ABCC2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 234
2.3.3. ABCC3 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 234
2.3.4. ABCC4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 234
2.3.5. ABCC5 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 234

Corresponding author at: The University of Adelaide, School of Paediatrics and Reproductive Health, Medical School North, University of Adelaide, Frome
Rd., Adelaide 5005, Australia. Tel.: +61 08 8313 8255; fax: +61 08 8313 6387.
E-mail address: carmela.ricciardelli@adelaide.edu.au (C. Ricciardelli).

http://dx.doi.org/10.1016/j.critrevonc.2015.05.012
1040-8428/ 2015 Elsevier Ireland Ltd. All rights reserved.

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

3.
4.
5.
6.

221

2.3.6. ABCC6 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 235


2.3.7. ABCC7 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 235
2.3.8. ABCC8 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 236
2.3.9. ABCC9 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 236
2.3.10. ABCC10 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 236
2.3.11. ABCC11 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 236
2.3.12. ABCC12 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 236
2.4. The ABCD family . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 236
2.4.1. ABCD1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 236
2.4.2. ABCD2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 236
2.4.3. ABCD3 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 236
2.4.4. ABCD4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
2.5. The ABCE family . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
2.5.1. ABCE1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
2.6. The ABCF family . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
2.6.1. ABCF1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
2.6.2. ABCF2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
2.6.3. ABCF3 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
2.7. The ABCG family . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
2.7.1. ABCG1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
2.7.2. ABCG2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
2.7.3. ABCG4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 238
2.7.4. ABCG5 and ABCG8 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 238
Cancer stem cells and ABC transporters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 238
Clinical trials using inhibitors of ABC transporters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 238
Alternative strategies to overcome ovarian cancer MDR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 240
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 241
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 242
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 242
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 242
Biography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 255

Abstract
Over 80% of ovarian cancer patients develop chemoresistance which results in a lethal course of the disease. A well-established cause
of chemoresistance involves the family of ATP-binding cassette transporters, or ABC transporters that transport a wide range of substrates
including metabolic products, nutrients, lipids, and drugs across extra- and intra-cellular membranes. Expressions of various ABC transporters,
shown to reduce the intracellular accumulation of chemotherapy drugs, are increased following chemotherapy and impact on ovarian cancer
survival. Although clinical trials to date using ABC transporter inhibitors have been disappointing, ABC transporter inhibition remains an
attractive potential adjuvant to chemotherapy. A greater understanding of their physiological functions and role in ovarian cancer chemoresistance will be important for the development of more effective targeted therapies. This article will review the role of the ABC transporter
family in ovarian cancer progression and chemoresistance as well as the clinical attempts used to date to reverse chemoresistance.
2015 Elsevier Ireland Ltd. All rights reserved.

Keywords: Ovarian cancer; ABC transporter; Chemoresistance; Multi-drug resistance; Prognosis; Clinical trials

1. Introduction
Ovarian cancer is the most lethal gynaecological cancer
and ranks as the sixth most common cause of cancer-related
death in women in the Western world [1]. Unfortunately,
ovarian cancer often goes undetected due to vague symptoms and is rarely diagnosed at an early stage which has
a substantially higher survival rate than advanced disease
[24]. Despite a high initial response to chemotherapy, the
majority of patients with advanced ovarian cancer relapse and

subsequently develop chemoresistance which results in treatment failure and death of the patient [5]. Chemoresistance,
either intrinsic or acquired, is the main factor contributing to ovarian cancer death. Cancer cells often develop
resistance to many functionally and structurally unrelated
anti-cancer drugs, including those not yet given to patients,
a phenomenon known as multidrug resistance (MDR). It is
believed that chemoresistance develops for two main reasons. Tumours may be populated by a minority of resistance
cells including tumour cells with stem cell like properties

222

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

that have inherent resistance to drugs and are capable of


repopulating the tumour after treatment [6]. Alternatively,
tumour cells acquire resistance as a result of chemotherapy treatment induced changes. Multiple mechanisms have
been characterized which include changes in cellular repair
mechanisms, alterations in apoptotic pathways, increased
or decreased expression of metabolic enzymes, mutations,
alterations in the tumour microenvironment and increased
expression/activity of drug efflux pumps and altered uptake
pathways which result in reduced cellular accumulation of
chemotherapy drugs [79]. One of the most important MDR
mechanisms is an increased efflux rate of anti-cancer drugs
from cancer cells by members of the ATP-binding cassette
(ABC) transporters family [10]. This review will discuss the
role of the ABC transporter family in ovarian cancer progression and chemoresistance.

2. ABC transporters
ABC transporters transport a wide range of substrates,
including metabolic products, nutrients, lipids, and drugs,
across extracellular and intracellular membranes, and have
been shown to play an important role in many human diseases
[1122]. ABC transporters differ from classical selective
transporters by way of their promiscuity for structurally
and chemically diverse substrates [23]. Phylogenetic analysis
places the 48 known human ABC transporters into 7 distinct
subfamilies (ABCA-ABCG, Table 1) [24]. ABC transporters
are complex translocation systems that couple the hydrolysis
of ATP to the unidirectional movement of a wide range of distinct compounds across the phospholipid bilayer of cellular
membranes. All ABC transporters consist of transmembrane
domains (TMDs) and nucleotide-binding domains (NBDs).
In humans, they either contain one, two, or three TMDs paired
with an NBD (Fig. 1) The NBDs act as motor domains
by hydrolysing ATP molecules to drive the transport cycle
(Fig. 2) [2527]. Whilst ABC transporters have many physiological functions, they are best known for their involvement
in drug efflux. In fact, a single ABC transporter can be capable
of transporting a wide range of structurally and chemically
diverse substrates, both physiological as well as chemotherapeutic. A thorough review of the literature has found that
currently, at least 20 ABC transporters are capable of exporting anti-cancer drugs (Table 1). It is even thought that ABC
transporters may be capable of transporting multiple substrates at once [28]. However, it remains unclear why ABC
transporters have such a vast range of substrates, but it is
believed that they contain many different substrate recognition sites. A greater understanding this phenomenon will
allow for a greater ability to inhibit the chemotherapeutic
efflux capacity of ABC transporters [29].
Only a handful of ABC transporter crystal structures
are currently available. After the retraction of the incorrect
structures of MsbA [30] there was only one high resolution structure of an ABC exporter, Staphylococcus aureus

Sav1866 protein available [31,32]. However, recent publications have elucidated TM287-TM288 (TM287/288) from
Thermotoga maritima [33], mouse ABCB1 [34], Pyrococcus abyssi ABCE1 [35,36], Caenorhabditis elegans ABCB1
[37], and also human ABCB10 [38] (Fig. 3). A very recent
paper shows ABCB1 interacting with several designed ligands and provides useful insight in understanding how ABC
transporters can recognise a wide range of structurally diverse
ligands [39].
2.1. The ABCA family
2.1.1. ABCA1
ABCA1 mediates the apolipoprotein-dependent formation of a high-density lipoproteincholesterol complex [40].
Very little is known about its role in cancer, but a recent
study found ABCA1 was elevated in radiation surviving
medulloblastoma cells and broad ABC exporter inhibitors
verapamil and reserpine could resensitise cells to radiotherapy [41]. ABCA1 was also found to be up-regulated in
pancreatic ductal adenocarcinomas [42] and was part of a
gene signature predicting higher risk of relapse in colon
cancer patients [43]. Increased ABCA1 expression was also
shown to be associated with poor treatment response in breast
cancer patients [44]. Two recent studies suggest it also plays
a role in ovarian cancer. The first showed that ABCA1 was
significantly associated with reduced overall survival (OS)
in serous ovarian cancer patients in two independent datasets
and the suppression of ABCA1 expression lead to reduced
ovarian cancer cell growth and migration [45]. The second
found higher ABCA1 methylation levels in ovarian cancer
tissues compared to normal ovaries and was associated with
significantly shorter progression-free survival (PFS) [46].
Lentiviral knockdown of ABCA1 in cisplatin resistant ovarian cancer cells resulted in their re-sensitisation, representing
the first evidence that ABCA1 may play a role in drug resistance [47]. These findings are supported by Oiso et al. that
found ABCA1 was up-regulated in cisplatin resistant human
epidermoid carcinoma cells compared to the parental cells
[48].
2.1.2. ABCA2
ABCA2 has been suggested to be involved in intracellular
sterol regulation [49]. ABCB2 expression has been reported
to be up-regulated in melanoma [50], hepatocellular carcinoma [51], breast cancer [52], colon adenocarcinoma [53]
and leukaemia [54,55]. A single study in ovarian cancer found
up-regulated ABCA2 expression in an estramustine resistant
strain of SKOV-3 compared with the parent cell line [56].
Antisense targeting of ABCA2 resulted in downregulation of
ABCA2 and re-sensitisation to estramustine [56]. Increased
survival in the presence of cytotoxic agents is thought to be
due to the capacity of ABCA2 to sequester drugs into lysosomal organelles. Human Protein Atlas (HPA) shows medium
or high ABCA2 expression in ovarian tumour cells in all 12 of

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

223

Table 1
Human ABC transporters and their known substrates and inhibitors.
Names

Cancer drug substrates

Inhibitors

ABCA1
ABC1
HDLDT1
TGD
CERP
ABCA2
ABC2
KIAA1062
ABCA3
ABC3
ABCC

Cisplatin [47]

Glibenclamide/Glyburide [383], JNJ-26854165 [384]

Mitoxantrone [385], Estramustine [385], Methotrexate [54]

None identified

ABCA4
ABCR
ABCA5
ABCA6
EST155051
ABCA7
ABCX
ABCA-SSN
ABCA8
KIAA082
ABCA9
EST640918
ABCA10
EST698739
ABCA12
ABCA13
ABCB1
MDR1
Pgp
GP170
PGY1
MDR/TAP ABC20
CD243

ABCB2
TAP1
ABCB3
TAP2
ABCB4
PGP3
MDR3
PGY3
MDR3 PGP1111
GBD1
PFIC-3
ABC21
ABCB5
ABCB6
MTABC3
ABCB7
ABC7

Cisplatin [59], Dasatinib [386], Daunorubicin [387], Doxorubicin Genistein [388], Indomethacin [386], PK11195 [389]
[55], Etoposide [387], Imatinib [386], Methotrexate [54],
Mitoxantrone [387], Nilotinib [386], Paclitaxel [59], Vincristine
[387]
None identified
None identified
None identified
None identified

U0126 [390]
IGF-1 [391]

None identified

None identified

Digoxin [392]
None identified

MS-209 [393], MK-571[393]1, Ochratoxin A [393],


Verapamil [393], Probenecid [393]
None identified

None identified

None identified

None identified
None identified
5-Fluorouracil [394], Actinomycin D [395], Bisantrene [396],
Dasatinib [397], Daunorubicin [78], Digoxin [398], Docetaxel
[399], Doxorubicin [400], Epirubicin [401], Etoposide/VP-16
[402], Homoharringtonine [403], Irinotecan [404], Mitoxantrone
[317], Paclitaxel [405], Teniposide [406], Topotecan [407],
Vinblastine [408], Vincristine [408], Vindesine [408], Vinorelbine
[408]

None identified

None identified
None identified
Agosterol A [409], Annamycin [410], Anthranilamide
[411], Antimalarials [412,413], Apatinib [414], BBA [415],
CBT-1 [416], Curcumin [353], Cyclosporin A [417],
Disulfiram [418], Dofequidar [419],
Elacridar/GG918/GF120918 [420], Epigallocatechin
gallates [348], FG020326 [421], Flavinoids [422],
Ginsenosides [423], HG-829 [424], Ko143 [425], Lapatinib
[426], Latilagascenes [427], Mitotane/NSC-38721 [428],
MK571 [425], Monoterpenoids [429], MS-209 [430],
Neratinib [431], OC-144-093/ONT-093 [432], Piperine
[433], PK11195 [434], Pluronic L61 [420], Polyphenols
[348], Quinoline [435], R101933 [436], Reversan [320],
S9788 [437], Saracatinib [330], Tariquidar/XR-9576 [308],
U0126 [390], V-104 [420], Valspodar/PSC 833 [438],
Verapamil [439], VX-710/Biricodar [440], WK-x-34 [441],
Zosquidar/LY-335979 [442]
None identified

None identified

None identified

Daunorubicin [443], Digoxin [443], Paclitaxel [443], Vinblastine


[443]

Cyclosporin A [443], Valspodar/PSC833 [443], Verapamil


[443]

5-Fluorouracil [394], Camptothecin [394], Doxorubicin [286],


Irinotecan [394], Mitozantrone [394], Topotecan [394]
None identified

None identified

None identified

None Identified

None identified

224

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

Table 1 (Continued)
Names

Cancer drug substrates

Inhibitors

ABCB8
MABC1
ABCB9
ABCB10
MTABC2
ABCB11
SPGP
BSEP
ABCC1
MRP-1
CFTR/MRP
GS-X
ABC29

Doxorubicin [165]

None identified

None identified
None identified

None identified
None identified

Paclitaxel [175]

Cyclosporin A [175], Valspodar/PSC833 [175], Verapamil


[175]

ABCC2
MRP-2
cMOAT

ABCC3
MRP-3
cMOAT-2
ABCC4
MRP-4
MOAT-B
ABCC5
MRP-5
MOAT-C
ABCC6
MRP-6
ABCC7
CFTR
ABCC8
SUR1
ABCC9
SUR2
ABCC10
MRP-7

ABCC11
MRP-8
ABCC12
MRP-9
ABCD1
ALD
ABCD2
ALD1
ALDR
ABCD3
PXMP1
PMP70
ABCD4
PMP69
P70R

Chlorambucil [444], Daunorubicin [445], Doxorubicin [445],


Epirubicin [446], Etoposide/VP-16 [447], Glucuronide [448],
Imatinib [447], Irinotecan [447], Melphalan [444], Methotrexate
[447], Mitoxantrone [447], Paclitaxel [182], saquinivir [447],
SN-38 [447], Vinblastine [449], Vincristine [408], Vinorelbine
[450]

Agosterol A [409], Annamycin [451], Antimalarials [452],


Biricodar/VX-710 [453], CBT-1 [416], Clotrimazole [454],
Curcumin [353], Cyclosporin A [453], Disulfiram [418],
Dofequidar [419], Elacridar/GG918/GF120918 [453],
Flavinoids [422], Flavopiridol [455], Glibenclamide [456],
Glutathione ethacrynic acid [23], Indomethacin [457],
LY465803 [458], LY475776 [459], MK571 [460], MS-209
[453], Non-nucleoside RT inhibitors [460], Nucleoside RT
inhibitors [460], Piperine [433], PK11195 [434],
Polyphenols [349], Pyrrolopyrimidines [461], Reversan
[320], Sulindac [327], Tariquidar/XR-9576, V-104 [420],
Verapamil [453], VX-710/Biricodar [453]
Cyclosporin A [467], MK571 [467], Non-nucleoside RT
Carboplatin [462], Cisplatin [176], Doxorubicin [225],
Epirubicin [463], Etoposide/VP-16 [225], Irinotecan [464],
inhibitors [460], Nucleoside RT inhibitors [460], PAK-104P
Irinotecan/CPT-11 [225], Methotreaxate [448], Mitoxantrone [465], [467], Triptanthin [468], Valspodar/PSC833 [467]
Paclitaxel [225], saquinivir [463], SN-38 [225], Sulfinpyrazone
[466], Topotecan [463], Vinblastine [192], Vincristine [225]
Etoposide/VP-16 [469], Methotrexate [448], Tenoposide [469],
MK571 [460], Non-nucleoside RT inhibitors [460],
Vincristine [394]
Nucleoside RT inhibitors [460], U0126 [390]
5-Fluorouracil [470], 6-mercaptopurine [471], 6-thioguanine [471],
Bisantrene [394], Irinotecan/CPT11 [225], Methotrexate [471],
Nucleoside monophosphates [472], Topotecan [225], Vinblastine
[443]
5-Fluorouracil [471], 6-mercaptopurine [471], 6-thioguanine [471],
Bisantrene [394], Doxorubicin [286], Gemcitabine [471],
Methotrexate [474], Mitozantrone [394], Nucleoside
monophosphates
Cisplatin [475], Daunorubicin [475], Doxorubicin [475],
Etoposide/VP-16 [475], Tenopiside [475]
None identified

Antimalarials [452], MK571 [471], PCS833 [471],


Polyphenols [349], Sildenafil [473], Trequinsin [473],
Zaprinast [473]

None identified

None identified

None identified

None identified

MK571 [471], Polyphenols [349], Sildenafil [473],


Trequinsin [473], Zaprinas [473]

None identified
None identified

Docetaxel [225], Doxorubicin [225], Paclitaxel [225], Vinblastine Cyclosporin A [225], Erlotinib [477], Glycolithocholate-3[225], Vincristine [225], Vinorelbine [476]
sulfate [225], Imatinib [478], Lapatinib [477], LTC4 [225],
MK571 [225], Nilotonib [478], Sulfinpyrazone [476],
Tandutinib/MLN518 [479], Tariquidar/XR-9576 [480]
5-Fluorouracil [225], 6-mercaptopurine [481], ddC [225],
None identified
Methotrexate [225], Pemetrexed [482], PMEA [225]
None identified
None identified
None identified
None identified

None identified
None identified

None identified

None identified

None identified

None identified

None identified

None identified

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

225

Table 1 (Continued)
Names

Cancer drug substrates

Inhibitors

ABCE1
OABP
RNS41
ABCF1
ABC50
ABCF2
ABCF3
ABCG1
ABC8
White
ABCG2
BCRP
MXR1
ABCP
CD338
EST157481
ABC15
BMDP
MRX
ABCG4
White2
ABCG5
White3
STSL
Sterolin-1
ABCG8
GBD4
STSL
Sterolin-2

5-Fluorouracil [483], Fasudil [484]

None identified

None identified

None identified

None identified
None identified
Doxorubicin [394]

None identified
None identified
None identified

Bisantrene [485], Dasatinib [397], Doxorubicin [486],


Daunorubicin [486], Epirubicin[487], Flavopiridol [256],
Irinotecan/CPT-11 [264], Methotrexate [448], Mitoxantrone [486],
Nilotinib [397], SN-38 [264], Topotecan [488], Tyrosine kinase
inhibitors [321]

Apatinib [414], Curcumin [353], Curcumin [352],


Dofequidar [419], Elacridar/GG918/GF120918 [425],
Flavinoids [350], Fumitremorgin C [489], Gefitinib [490],
Ginsenosides [491], Imatinib Cyclosporin A [417], Ko143
[425], lapatinib [426], MK571 [425], Novobiocin [492],
Piperine [433], PK11195 [434], Polyphenols [351],
Tariquidar/XR-9576 [493], UR-COP269 [494],
VX-710/Biricodar [440], WK-X-34 [441]

None identified

None identified

None identified

None identified

None identified

None identified

Each ABC transporter is listed along with any known synonyms. Any chemotherapeutic agents shown to be transported via knockdown or labelling studies are
listed. Chemotherapeutic agents commonly used in the treatment of ovarian cancer are in bold. Additionally, substances which have been shown to inhibit the
ABC transporters are listed.

their tissue samples [57]. These findings indicate that the role
of ABCA2 in ovarian cancer should be further investigated.
2.1.3. ABCA3
ABCA3 is known for its role in production of pulmonary
surfactant in type 2 pneumocytes. ABCA3 has been shown

to be up-regulated in some leukemias and breast cancer


[52,54,58]. Silencing of ABCA3 in non-small-cell lung cancer cells resensitised the cells to cisplatin and paclitaxel
[59]. Additionally, Oiso et al. recently found ABCA3 was upregulated in cisplatin resistant human epidermoid carcinoma
cells compared to parental cells [48]. Furthermore, ABCA3

Fig. 1. The basic structure of ABC transporters. A typical ABC transporter consists of two transmembrane domains and two nucleotide binding domains.
However, variations exist on this basic structure. Some ABCC family members have three transmembrane domains and two nucleotide binding domains. Just
over a third of all ABC transporters are transcribed as half transporters, with a single transmembrane domain and a single nucleotide binding domain. It is
believed that the majority of these form homo- or hetero-dimers in order to form a complex capable of transport. ABCE1 has no TMDs.

226

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

Fig. 2. A typical transport process of an ABC transporter. ABC transporters are believed to exist in an inward facing conformation when not actively transporting,
i.e., with their TMD opening facing the interior of the cell. When a complex, such as a chemotherapeutic agent, is recognised by the TMDs, the NBDs dimerise,
powered by ATP hydrolysis, and trigger a change in the conformation of the transporter to the outward facing configuration, releasing the complex outside the
cell.

Fig. 3. Cartoon representations of known ABC exporter structures. Structures were downloaded from the RSCB Protein Databank and oriented and exported
using PYMOL [3133,35,3739].

was found to be up-regulated in cisplatin, paclitaxel, topotecan, and vincristine resistant ovarian cancer cells [60]. HPA
found medium to high ABCA3 expression in ovarian cancer cells in 7 of their 11 tissue samples [57]. These studies
suggest that ABCA3 may play a role in ovarian cancer drug
resistance mechanisms.

2.1.4. ABCA4
ABCA4 is well known for its role in Stargardt disease, an
inherited form of juvenile macular degeneration [12]. However, only a handful of studies so far have been undertaken
to determine its role in cancer. ABCA4 was found to be upregulated significantly in an induced MDR breast cancer cell
line [61]. Additionally, ABCA4 was found to be up-regulated
almost 2.5 fold in stage I epithelial ovarian cancer compared
with borderline ovarian tumours [62], which could make it
a useful marker for distinguishing between borderline and
early stage ovarian cancer. A recent study has found that

high ABCA4 expression was associated with chemotherapy


response in ovarian cancer patients [63].

2.1.5. ABCA5
Little is known about the physiological function of
ABCA5 since its discovery in 2003. ABCA5 was significantly
down-regulated in breast tumours compared with control
tissues [52] and was also recently identified as a putative
biomarker of osteosarcoma tumour stem cells [64]. It has also
been linked to the development of oesophageal cancer [65].
HPA found medium staining for ABCA5 in ovarian cancer
cells in all 11 of their tissue samples [57] and ABCA5 mRNA
was detected in undifferentiated ovarian carcinomas [53].
Furthermore, Hedditch et al. found that low expression levels
of ABCA5 was associated with shorter OS in serous ovarian cancer patients [45]. However, ABCA5 is up-regulated
in doxorubicin, paclitaxel, and vincristine resistant ovarian
cancer cells providing a possible link to drug resistance [60].

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

Further studies will determine whether ABCA5 is involved


in mediating drug resistance in ovarian cancer.
2.1.6. ABCA6
Very little is known about the physiological function
ABCA6. It has been proposed that may be involved in
macrophage lipid homeostasis [66] but there have been
limited studies in cancer. Whilst a study by Ohtsuki et al.
found no ABCA6 mRNA in a range of cancer cell lines
and tissues including ovarian cancer [53], other researchers
described increased ABCA6 expression in lymphoma cell
lines that exhibited a weak response to drug treatment
[67]. ABCA6 expression was significantly down-regulated
in breast tumours and ovarian serous papillary carcinomas compared with normal tissues [52,68]. Furthermore,
ABCA6 expression was found to be up-regulated in serous
ovarian tumours and associated with reduced OS [45], indicating that further investigation into this ABC transporter is
needed.
2.1.7. ABCA7
ABCA7 is most well-known for its link with Alzheimers
disease [65]. It was also reported to be up-regulated in pancreatic ductal adenocarcinoma and breast cancer [42,52].
Colorectal cancer patients with low ABCA7 transcript levels
had significantly shorter PFS [69]. ABCA7 mRNA expression
was shown to increase in head and neck squamous cell and
human epidermoid carcinoma cell lines in response to cisplatin and paclitaxel [48,70]. HPA found higher staining for
ABCA7 in all of their ovarian cancer tissues [57] but to date
no ovarian cancer studies investigating ABCA7 expression
have been published.
2.1.8. ABCA8
The function of ABCA8 is currently unknown, although
it has been postulated that it may play a role in myelin formation [71]. ABCA8 expression was reduced in ovarian cancers
compared to normal ovarian tissues [52,57]. ABCA8 was
also significantly down-regulated in a methotrexate, cisplatin,
doxorubicin, vincristine, topotecan, and paclitaxel-resistant
variant of the W1 ovarian cancer cell line [60]. In contrast,
ABCA8 expression was found to be positively associated with
gemcitabine resistance in ovarian cancer cell lines [72] and
significantly associated with reduced OS in serous ovarian
cancer patients [45]. This conflicting data indicates that further work is necessary to assess the exact role of ABCA8 in
ovarian cancer.
2.1.9. ABCA9
The function of ABCA9 remains unconfirmed; however it
may play a role in monocyte differentiation and lipid homeostasis [73]. Its role in cancer is poorly understood as only a
few studies have been published [52,74]. High expression
of ABCA9 was associated with significantly reduced survival in serous ovarian cancer patients [45]. ABCA9 was also
found to be hypermethylated before and after treatment with

227

DNA hypomethylating agent (decitabine) in ovarian cancer


patients with a PFS 6 months, but not in patients with <6
months PFS [75], indicating a possible role for ABCA9 in
ovarian cancer progression.
2.1.10. ABCA10
ABCA10 is perhaps the least investigated of the ABCA
family since its discovery in 2003. Its function and substrate are unknown, although its expression has been shown
to be regulated by cholesterol, indicating a possible role in
lipid transport similar to other members of the ABCA family
[76]. A single study showed that ABCA10 was significantly
down-regulated in breast tumours [52]. Normal ovarian tissue showed no reactivity for ABCA10 and only 5 out of 12
ovarian cancer tissue samples showed even low staining, suggesting ABCA10 may be down-regulated in ovarian cancer
[52]. There have been no other reports to date for ovarian
cancer.
2.1.11. ABCA12
Mutations of ABCA12 are the underlying cause for the
skin condition harlequin ichthyosis, the most severe variant
of autosomal recessive congenital ichthyosis [14]. ABCA12s
physiological function is thought to maintain the epidermal lipid permeability barrier by facilitating formation of
ceramide linoleic esters [77]. However, despite the vast number of studies on the role of ABCA12 in ichthyosis, there is
only a handful investigating its role in cancer. ABCA12 was
significantly up-regulated in breast and colorectal tumour tissues [52,69] and high expression was associated with poor
treatment response in breast cancer patients [44,52]. HPA
found moderate to high staining for ABCA12 in 8 of their 11
ovarian cancer tissue samples [57]. There have been no other
reports to date for ovarian cancer.
2.2. The ABCB family
2.2.1. ABCB1
ABCB1 (MDR1), also known as p-glycoprotein is the
most studied ABC transporter and first to be identified [78].
It is a 170 kDa protein localized at the apical surface of
many cells and is thought to play an important role in cell
detoxification [79,80]. ABCB1 transports large hydrophobic, uncharged, or slightly positively charged compounds
and plays a critical role in drug efflux and chemoresistance
in many malignancies [8183]. It transports chemotherapy
drugs including paclitaxel, doxorubicin, topotecan, docetaxel
but not platinums or cyclophosphamide [84] (Table 1).
ABCB1 positivity ranges from 7% to 92.8% across multiple ovarian cancer studies [8398] (Table 2). The largest
of these studies included 1036 primary ovarian cancers
and 2137 metastatic cancers which reported low ABCB1
expression (10% and 12% in primary and metastatic cancer, respectively) [92]. However, this study did not specify
the ovarian cancer subtypes examined or relate ABCB1
expression to survival outcome. Many studies have found

228

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

Table 2
Summary of ABCB1 studies in ovarian cancer tissue cohorts.
Cohort details

Type of analysis

Major findings

Ref.

Pre-treatment advanced stage (n = 5)


Subtype not known
Prior to treatment (n = 33)
Post chemotherapy (n = 24)

Immunohistochemistry
Antibody not known (only abstract available)
Immunohistochemistry (paraffin sections, two step
immunoperoxidase)
Monoclonal antibodies C219 and JSB-1
Immunohistochemistry (frozen sections)
C219 monoclonal antibody
5% cut point
Immunohistochemistry (paraffin, sections)
C219 monoclonal antibody
Semi-quantitative RT-PCR

ABCB1 expressed in 40% of cases

[85]

Pre-treatment (n = 21)
Post chemotherapy (n = 13)
Subtypes not specified
Pre-treatment advanced stage (n = 20)
Subtypes not specified
Pre-treatment (n = 46)
Post chemotherapy (n = 14)
Subtypes not specified
Pre-treatment (n = 52)
Serous (23)
Mucinous (7)
Endometrioid (9)
Clear cell (4)
undifferentiated (1)
Other (5)
Metastatic (3)
Pre-treatment (n = 57)
Serous (40)
Mucinous (5)
Endometrioid (4)
Undifferentiated (8)
Pre-treatment advanced (n = 89)
Serous (54)
Mucinous (9)
Endometrioid (7)
Clear cell (10)
Undifferentiated (9)
Pre-treatment (n = 58)
Serous (24)
Mucinous (12)
Endometrioid (13)
Clear cell (6)
Undifferentiated (3)
Pre-treatment (n = 53)
Serous (34)
Mucinous (1)
Endometrioid (7)
Clear cell (3)
Mixed (7)

Quantitative RT-PCR

Immunohistochemistry (frozen sections


Mouse monoclonal (MRK-16 &
JSB-1)
10% cutpoint

ABCB1 expression observed in 7% of cases and [86]


only in 2 patients that were clinically drug
resistant
ABCB1 expression in 71.4% of untreated cases [87]
and 61.5% after treatment with
platinum/cyclophosphamide
[84]
ABCB1 expression observed in 35% of cases
ABCB1 expressed in 65.2% untreated samples [111]
and 100% of tumours after treatment with
ABCB1 substrates
Highest ABCB1 expression was observed in
[107]
serous and mucinous subtypes
No relationship with chemotherapy response or
survival

[88]
ABCB1 expressed in 16% of cases.
No relationship with chemotherapy response or
survival

Immunohistochemistry (paraffin, microwave retrieval) ABCB1 expressed in 13% cases. ABCB1


Mouse Monoclonal JSB-1
expression not related to any clinical or
pathological parameters or patient outcome

[89]

Immunohistochemistry (paraffin)
Rabbit polyclonal (Oncogene Research products)
Cut point 30%

ABCB1 expressed in 27.6% of cases. No


relationship with survival

[90]

Quantitative RT-PCR

[99]
ABCB1 expressed in 35.6% of cases
ABCB1 expression associated with reduced PFS

Pre-treatment (n = 115)
Mucinous/Clear cell (89)
Other (69)
Pre-treatment advanced primary (n = 1036)
Metastatic (n = 2137)
Subtypes not specified
Pre-treatment (n = 24)
Subtypes not known as only abstract
available

Immunohistochemistry (frozen) JSB-1 monoclonal


antibody
Cut point 10%
Immunohistochemistry (paraffin)
JSB1 mouse monoclonal

Pre-treatment (n = 30)
Serous
Pre-treatment advanced (n = 27)
Subtypes not specified
Pre-treatment (n = 131)
Serous (90)
Clear cell (41)

Quantitative RT-PCR

ABCB1 expressed in 17.4% cases


No relationship with chemotherapy response or
survival
ABCB1 expressed in 12% primary cancers and
10% of metastatic lesions.
Survival analysis not performed
ABCB1 expressed in 25% of cases.
No relationship with chemotherapy response.
ABCB1 was increased in tumour tissues after
chemotherapy
ABCB1 undetectable in any tumour

Immunohistochemistry
Antibody details not known (only abstract available)

Quantitative RT-PCR

[91]

[92]

[93]

[190]

ABCB1 expressed in 96% cases


[100]
ABCB1 expression increased in non-responders
[94]
Immunohistochemistry (paraffin, microwave retrieval) ABCB1 expressed in 46.7% of serous and
Mouse monoclonal C494 (DAKO, Glostrup,
41.5% clear cell cancers.
Denmark)
No relationship with chemotherapy response in
Cut point 10%
either serous or clear carcinoma

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

229

Table 2 (Continued)
Cohort details

Type of analysis

Major findings

Ref.

Pre-treatment (n = 93)
Serous (57)
Mucinous (7)
Endometrioid (13)
Clear cell (16)
Pre-treatment and paired samples at
recurrence (n = 32)
Serous (26)
Clear cell (2)
Endometrioid (2)
Mixed (2)
Pre-treatment (n = 59)
Serous (18)
Mucinous (19)
Endometrioid (9)
Clear cell (8)
Other (5)
Pre-treatment (n = 50)
Serous (40)
Endometrioid (4)
Mucinous (1)
Others (5)
Pretreatment (n = 52)
Stage III serous

Immunohistochemistry (paraffin, microwave


retrieval)
C-19 goat polyclonal
Cut point 10%

ABCB1 expressed in 12.9% cases


ABCB1 more frequent in clear cell carcinoma
No relationship with survival

[95]

Immunohistochemistry (paraffin, microwave


retrieval)
Mouse monoclonal c494 and c219 antibodies
0, 1+ vs 2+, 3+ staining

[96]
c494 antibody: ABCB1 expressed in 50% of
untreated samples and 75% of recurrent samples.
c219 antibody: ABCB1 expressed in 46.9% of
untreated and 21.9% recurrent samples.
ABCB1 expression could predict response to
paclitaxel and survival in recurrent samples.
ABCB1 expressed in 30.5%
[97]
No relationship with survival

Pre-treatment serous (n = 60)

Pre-treatment (n = 43) and post


chemotherapy (n = 30)
Serous (37)
Endometrioid (3)
Other (3)
Pre-treatment (n = 211)
Serous (86)
Endometrioid (42)
Clear cell (16)
Mucinous (14)
Undifferentiated (38)
Other (15)
Pre-treatment (n = 120)
Serous (70)
Mucinous (23)
Undifferentiated (11)
Endometrioid (8)
Clear cell (8)
Pre-treatment (n = 80)
Subtypes not indicated in abstract
Pre-treatment serous (n = 143)
All patients received 1st line paclitaxel
and carboplatin
Pre-treatment (n = 111)
Serous (83)
Endometrioid (14)
Mucinous (5)
Clear cell (6)
Undifferentiated (3)

Immunohistochemistry (paraffin, microwave


retrieval)
JSB-1

Quantitative RT-PCR

ABCB1 expression in 98% of cases. High


ABCB1 associated with reduced PFS

[495]

Immunohistochemistry (paraffin, microwave


retrieval)
Clone MDR 88 (Biogenix)
Positive if membrane staining present
Immunohistochemistry (paraffin, microwave
retrieval)
JSB-1 & C494
Immunohistochemistry (paraffin, microwave
retrieval)
C219
Cut point: Histoscore >03

ABCB1 expressed in 92.8% of cases


High ABCB1 associated with reduced OS

[101]

ABCB1 expressed in 20% of cases using both


JSB-1 and C494. Increased ABCB1 expression
independent predictor of reduced OS.
High ABCB1 expressed in 53.5% of untreated
cases and 30% of cases after chemotherapy.
High ABCB1 significantly associated with
reduced PFS in untreated cases.

[83]

Quantitative RT-PCR

ABCB1 expressed in 85.9% cases


High ABCB1 significantly associated with
reduced PFS but OS

[102]

Immunohistochemistry (paraffin, microwave


retrieval, immunofluorescence)
Rabbit anti human sc-1517-R (Santa Cruz)
Cut point 25%

ABCB1 expressed in 80% of cases. High


ABCB1 significantly associated with tumour
stage, grade and disease relapse

[103]

Immunohistochemistry
Antibody not known (only abstract available)

ABCB1 expressed in 57.5% of cases. High


[104]
ABCB1 expression significantly associated with
OS
High ABCB1 significantly associated with
[105]
shorter PFS

Quantitative RT-PCR

Immunohistochemistry
(paraffin, microwave retrieval)
MDR88 (BioGenex)
Cut point 85%

ABCB1 expression associated with PFS


(P = 0.053)

References in bold indicate studies that found relationships with chemotherapy response or survival outcome.

[98]

[106]

230

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

a relationship between high ABCB1 mRNA or ABCB1 protein levels and poor ovarian cancer outcome [83,96,98106]
whilst several others have not [8891,93,94,97,107]. The
reason for this inconsistency is not clear but possible reasons may be due to small cohort size and the inclusion of
different ovarian cancer subtypes and stages in the analysis. Three studies that investigated expression in the serous
subtype only did report a significant relationship with high
ABCB1 protein or ABCB1 mRNA levels and poor patient
outcome [83,101,105]. However, clinically validated assays
for ABCB1 detection in tissues have not been established and
the variability may also be due to the use of different antibodies and methods used for assessment. Many antibodies
are not specific for ABCB1 and cross react with pyruvate
carboxylase (JSB-1) and other ABC transporter proteins
[108].
Increased ABCB1 mRNA and ABCB1 protein has been
seen in many drug resistant ovarian cancer cell lines
[60,82,84,109,110]. However, several studies found no difference in ABCB1 protein or ABCB1 mRNA levels in tissues
from treated versus untreated ovarian cancers [86,87,96]
whilst others researchers reported marked differences following treatment [93,96,111]. Several studies have found
that ABCB1 is expressed in ovarian tumours which have
been exposed to its substrates, such as paclitaxel, but not
in chemonaive tumours or tumours exposed to non ABCB1
substrates including cisplatin [111,112]. Our own data concurs with these findings as we found no ABCB1 expression
in ovarian cancer cells treated with the platinum, carboplatin
[113]. Our recent immunohistochemistry studies detected
minimal ABCB1 protein in ovarian cancer tissues collected
at surgery prior to chemotherapy treatment (Fig. 4A and B)
but found increased ABCB1 levels in tumour tissues from
patients that received neoadjuvant carboplatin and paclitaxel treatment (Fig. 4D). Minimal ABCB1 expression was
observed in a patient that received neoadjuvant single carboplatin treatment (Fig. 4C). However, high ABCB1 levels were
present in recurrent ovarian cancers from patients that had
received both carboplatin and paclitaxel treatment (Fig. 4E
and F).
Whilst some earlier studies reported that ABCB1 polymorphisms were associated with ovarian cancer outcome
and toxicity to taxane and platinum based chemotherapy
[114119], a recent systemic review found no consistent association with any ABCB1 SNPs and clinical outcome [120].
A large comprehensive study looking at 21 different ABCB1
SNPS in 4025 patients did not find any association between
ABCB1 SNPs and PFS or OS [105]. These findings were also
confirmed in an independent study of over 10,000 ovarian
cancer cases [121]. Several ABCB1 SNPs have been reported
to alter paclitaxel clearance [116,122], substrate specificity
[123] and neutrophil toxicity [124] but other studies found
no relationship with toxicity and neutropenia [125,126]. The
relationship of ABCB1 SNPs with toxicity needs to be also
assessed in the large well defined clinical cohorts like survival
outcome.

2.2.2. ABCB2 and ABCB3


ABCB2 and ABCB3, also known as TAP-1 and TAP2, respectively, form a heterodimer (Fig. 1) to form TAP
(transporter associated with antigen processing) which delivers cytosolic peptides into the endoplasmic reticulum (ER),
where they bind to nascent MHC class I molecules. TAP is
part of the peptide-loading complex (PLC) along with 2
microglobulin and other molecules which acts to keep hold
of MHC molecules until they have been fully loaded with
peptides ready for presentation to cytotoxic T lymphocytes
[98,104,238,263].
Many studies have shown that low ABCB2 and ABCB3
levels in various cancer types are linked to poor outcome,
suggesting that their down-regulation may aid cancer cells to
evade immune detection [44,127136]. ABCB2 is also downregulated in metastatic cancers compared to primary cancers
[137139]. In ovarian cancer high ABCB2 and ABCB3
expression was associated with low grade cancers, negative
lymph node status and increased OS [140142]. However,
another study found no relationship between ABCB2 and
survival outcome [143].
There is also some evidence that ABCB2 and ABCB3 are
linked with chemotherapy resistance. ABCB2 is increased
in acute myeloid leukaemia cells (AML) compared with
healthy bone marrow [55] and both ABCB2 and ABCB3 were
up-regulated in breast cancers following treatment with neoadjuvant chemotherapy [52]. Another study by Xu et al. has
suggested that ABCB2 may be involved in the resistance
of pancreatic cancer cells to gemcitabine [144]. Furthermore, ABCB3 is increased in recurrent ovarian cancers [145].
Our own preliminary analysis showed variable ABCB3 protein levels in ovarian cancer tissues. Tissues from patients
after neoadjuvent carboplatin treatment (Fig. 4C) and after
adjuvant carboplatin + paclitaxel chemotherapy at recurrence
(Fig. 4E) had higher ABCB3 levels compared with tissues
from untreated ovarian cancers (Fig. 4A and B). Further work
exploring the role of the TAP heterodimer in ovarian cancer
cells is required.
2.2.3. ABCB4
ABCB4 (also known as MDR3) is normally involved
in the transport of phospholipids from the liver hepatocytes into bile [146]. It is often considered a homologue of
ABCB1; however, very little is known about its role in cancer.
ABCB4 protein is up-regulated in soft tissue sarcomas [147]
and increased levels are associated with poor outcome in
patients with acute leukaemia [148]. ABCB4 is up-regulated
in pancreatic ductal adenocarcinoma and is thought to contribute its poor treatment response to chemotherapy treatment
[42] but high expression in lung cancer cells suppressed
their growth [149]. ABCB4 was found to be up-regulated
16-fold in paclitaxel resistant ovarian cancer cell lines compared with the parental cell line and siRNA knockdown of
ABCB4 increased the sensitivity to paclitaxel [82]. Furthermore, ABCB4 was up-regulated 176-fold in doxorubicin and
147-fold in vincristine resistant sublines of ovarian cancer

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

231

Fig. 4. ABC transporter expression in ovarian cancer tissues. Formalin fixed paraffin sections (5 m) were immunostained with antibodies to ABCB1 (mouse
monoclonal, clone F4, 1/1200, P7965, Sigma Aldrich), ABCB3 (rabbit polyclonal 1/750, Ab130414, Abcam), ACBC2 (mouse monoclonal clone M2 I-4, 1/50,
Abcam) and ABCG2 (mouse monoclonal clone BXP-21, 1/100, ab3380, Abcam) using citrate buffer microwave retrieval as previously described [496]. (A)
Untreated stage IIIC serous ovarian carcinoma. (B) Untreated stage IIIC serous ovarian carcinoma. (C) Stage IV serous carcinoma treated with neoadjuvent
carboplatin alone. (D) Stage III peritoneal carcinoma treated with neoadjuvent carboplatin + paclitaxel. (E) Recurrent mucinous ovarian carcinoma treated
with carboplatin + paclitaxel. (F) Recurrent endometrioid ovarian carcinoma treated with carboplatin + paclitaxel. Bar = 100 m. All images are at the same
magnification.

cells [60], providing evidence that ABCB4 may play a similar


role in MDR like ABCB1.
2.2.4. ABCB5
The exact function of ABCB5 remains unknown, although
it has been found to be increased in melanoma [150] and
placental tumours [151]. High ABCB5 expression is an independent prognostic factor for predicting oral squamous cell
carcinoma recurrence [152]. It has been linked to chemoresistance in melanoma [153], liver cancer [154], breast cancer

[155], and colon cancer [156]. c-MYC was shown to bind to


the ABCB5 promoter region and a c-MYC inhibitor increased
colon cancer cell sensitivity to fluorouracil [157]. ABCB5 has
been shown to transport doxorubicin and topotecan in non
ovarian cancer cells (Table 1) but its role has not yet been
confirmed in ovarian cancer cells.
2.2.5. ABCB6
ABCB6 is involved in hematopoesis and is a mitochondrial transporter [158]. It is highly expressed in most normal

232

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

tissues. ABCB6 is up-regulated in colorectal cancer [69],


glioma [159], hepatocellular carcinoma [51], and melanoma
cells [136]. Furthermore, ABCB6 was significantly increased
in residual breast cancers after chemotherapy [44]. ABCB6
was found to be up-regulated 15-fold in paclitaxel resistant
ovarian cancer cells compared with the parental cell line
[160]. Additionally, ABCB6 was found to be up-regulated
in cisplatin, doxorubicin, methotrexate, paclitaxel, topotecan, and vincristine resistant sublines of ovarian cancer cells
[60], indicating a role for ABCB6 in ovarian cancer drug
resistance.
2.2.6. ABCB7
A mutation of the ABCB7 gene is the underlying cause
of X-linked sideroblastic anaemia with ataxia [13]. Only a
handful of studies have reported a possible role of ABCB7 in
cancer. ABCB7 expression was found to be high in retinoblastoma cells [161]. A thesis covering a wide range of ABC
transporters found that ABCB7 mRNA levels were increased
in a platinum-resistant ovarian carcinoma cell line [162],
however further work exploring the role of ABCB7 in ovarian
cancer is required.
2.2.7. ABCB8
Whilst the precise function of ABCB8 is unknown, it has
recently been suggested to be involved in mitochondrial iron
export essential for baseline cardiac function [163]. Similar
to ABCB7, there has been very little research into the role of
ABCB8 in cancer. ABCB8 has been shown to be up-regulated
in several drug resistant cell lines [164]. It is also known
that ABCB8 transports doxorubicin in melanoma, breast, and
ovarian cancer cells [165]. To date no ovarian cancer studies
investigating ABCB8 expression have been published.
2.2.8. ABCB9
ABCB9 translocates a broad spectrum of peptides from
the cytosol into the lumen of lysosomes [166]. It is often
called TAP-like protein, due to its antigen presenting role.
Whilst ABCB9 has not been studied as thoroughly as ABCB2
and ABCB3 which make up the TAP complex, it is highly
expressed in most normal tissues but to a lesser extent in most
cancer tissues, including ovarian cancer [57]. A very recent
study showed that ABCB9 confers resistance to cisplatin in
lung cancer cells via the micro RNA miR-31 [167] which
has also been shown to confer resistance to chemotherapyinduced apoptosis in prostate and colon cancer [168,169].
However, ABCB9 has not been studied in ovarian cancer to
date.
2.2.9. ABCB10
ABCB10 one of the three ABC transporters found in the
inner membrane of mitochondria, is essential for erythropoiesis [170] and is thought to act as a homodimer (Fig. 1).
Whilst little is known about the role of ABCB10 in cancer,
ABCB10 expression has been associated with cisplatin resistance in a range of cancer cell lines [171,172]. Gillet et al.

also found that ABCB10 mRNA levels in peritoneal effusions could predict shorter PFS in ovarian cancer patients
[173]. Further studies are required to understand the role of
ABCB10 in ovarian cancer.
2.2.10. ABCB11
ABCB11 is the bile salt exporter protein (BSEP). Thus,
ABCB11 research has so far been focused on liver disease.
BSEP disease is caused by a wide range of mutations in
ABCB11 which result in ABCB11 deficiency and a higher
risk of liver cancer especially in children [174]. ABCB11 is
up-regulated in pancreatic ductal adenocarcinoma [42] but
down-regulated in breast tumours [52]. ABCB11 is often
called the sister to Pgp/ABCB1 and has been shown to confer
some level of resistance to paclitaxel [175] but has not been
studied in ovarian cancer.
2.3. The ABCC family
The largest sub-family of ABC transporters is the ABCC
family (also known as multidrug resistance protein/MRP
family ABCC1-12). This ABC transporter family transports hydrophobic anionic conjugates and exports uncharged
hydrophic compounds.
2.3.1. ABCC1
In contrast to ABCB1, which extrudes xenobiotics into
bile, intestine, and blood for terminal elimination from the
body, ABCC1 (MRP-1) is a basolateral transporter that
allows movement of compounds away from luminal surfaces and into tissues that lie beneath the basement membrane
[176]. Its structure is similar to ABCB1 but it has an additional
5 transmembrane helices at the N-terminus of the protein,
creating a third TMD, MSD0 (Fig. 1). The MSD0 domain
of ABCC1 is grossly dispensable for its transport function
however the cytoplasmic loop connecting the third TMD is
required for its activity [177].
Unlike ABCB1, ABCC1 does not seem to have a critical
role in the bodys normal elimination of toxins, which may
make it a more suitable therapeutic target. Its physiological
function is thought to protect cells from chemical toxicity
and oxidative stress and to mediate inflammatory responses
[178]. ABCC1 transports vinca alkaloids, anthracyclines,
methotrexate, leukotriene C4, and substrates conjugated with
glucaronide, sulphate, or glutathione [179184].
ABCC1 has been found to be increased in a range of
cancers including hepatocellular carcinoma [51], paediatric
acute lymphoblastic leukaemia [185], colorectal cancers
[69], nasopharyngeal carcinoma [186], pancreatic ductal adenocarcinoma [42], and breast cancer [52]. ABCC1 positivity
ranges from 22.2% to 68.0% across multiple ovarian cancer studies [88,90,91,114,187,188] (Table 3). However, there
is a discrepancy between studies indicating ABCC1 mRNA
or ABCC1 protein levels can predict poor chemotherapy
response and reduced survival [90,106,145,187190] which
was not reported by other groups [88,91,94,99,100,114]. The

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

233

Table 3
Summary of ABCC1 studies in ovarian cancer tissue cohorts.
Cohort details

Type of analysis

Major findings

Ref.

Pre-treatment (n = 57)
Serous (40)
Mucinous (5)
Endometrioid (4)
Undifferentiated (8)
Pre-treatment (n = 53)
Serous (34)
Mucinous (1)
Endometrioid (7)
Other (10)
Pre-treatment (n = 32)
Serous (23)
Muncinous (9)
Pre-treatment (n = 115)
Mucinous/Clear cell (89)
Other (69)
Pre-treatment (n = 58)
Serous (24)
Mucinous (12)
Endometrioid (13)
Other (9)
Pre-treatment (n = 30)
Serous

Immunohistochemistry (frozen sections


Mouse monoclonal MrPr1

ABCC1 expressed in 68% of cases.


No relationship with chemotherapy response or
survival

[88]

Quantitative RT-PCR

ABCC1 expressed in 100% of cases. ABCC1


expression was not associated PFS

[99]

Quantitative RT-PCR

ABCC1 expressed in 97% of cases. ABCC1 levels


were 2-fold higher in non-responders compared to
responders
ABCC1 expressed in 44% cases
No relationship with chemotherapy response or PFS

[189]

ABCC1 expressed in 22.4% of cases. No


relationship with survival

[90]

High ABCC1 expressed in 50% of serous carcinoma


and significantly associated an unfavourable
outcome and reduced PFS
ABCC1 expressed in 100% cases
No relationship with chemotherapy response

[190]

Immunohistochemistry (paraffin, microwave


retrieval)
Rabbit polyclonal C-10 (Santa Cruz)
Cut point 10%
Immunohistochemistry (paraffin)
Rabbit polyclonal C-10 (Santa Cruz)
Cut point 5%
Quantitative RT-PCR

ABCC1 expressed in 27.8% of serous and 19.5%


clear cell cancers.
No relationship with chemotherapy response in
either cancer type
ABCC1 expressed in 38.3% of cases.
No relationship with chemotherapy response or PFS

[94]

ABCC1 expressed in 84.8% of cases

[187]

Immunohistochemistry (paraffin, microwave


retrieval)
Rat monoclonal MRPr1
Histoscore 6
Quantitative RT-PCR

High ABCC1 expressed in 55% of cases. High


ABCC1 expression associated with high tumour
grade, higher stage and reduced OS.
ABCC1 expression was increased in recurrent
cancers compared to primary cancers and benign
tumours

[145]

Immunohistochemistry (paraffin, heat retrieval)


Mouse monoclonal clone QCRL-2

ABCC1 expression not associated with survival


outcome

[197]

Immunohistochemistry (paraffin, microwave


retrieval)
Rat monoclonal MRPr1
Cut point 10%

ABCC1 expressed in 30% of cases. ABCC1


expression significantly associated with tumour
grade

[188]

Pre-treatment advanced
(n = 27)
Serous (25)
Clear cell (1)
Endometrioid (1)
Pre-treatment (n = 131)
Serous (90)
Clear cell (41)
Pre-treatment (n = 60)
Serous (50)
Endometrioid (10)
Pre-treatment (n = 129)
Serous (78)
Non-serous (37)
Undifferentiated (14)

Pre-treatment (n = 50)
Serous (40)
Endometroid (7)
Mucinous (1)
Others (2)
Recurrent disease (n = 50)
Serous (44)
Endometroid (2)
Mucinous (1)
Others (3)
Pre-treatment (n = 47)
Serous (34)
Non-serous (13)
Pretreatment (n = 127)
Serous (83)
Undifferentiated (12)
Clear cell (12)
Endometrioid (12)
Mucinous (6)
Others (2)

Immunohistochemistry (frozen)
MRPm6 monoclonal antibody
Cut point 10%
Immunohistochemistry (paraffin)
MRPm6 monoclonal antibody
Cut point 30%

Quantitative RT-PCR

Quantitative RT-PCR

[91]

[100]

[114]

234

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

Table 3 (Continued)
Cohort details

Type of analysis

Major findings

Ref.

Pre-treatment (n = 111)
Serous (83)
Endometrioid (14)
Mucinous (5)
Clear cell (6)
Undifferentiated (3)

Immunohistochemistry (paraffin, microwave


retrieval)
33A6 (Novacastra)
Cut point 85%

High ABCC1 expression associated reduced PFS


and OS

[106]

References in bold indicate studies that found relationships with chemotherapy response or survival outcome.

reason for this inconsistency is not clear but a possible reason may be due to the inclusion of different ovarian cancer
subtypes and stages in the analysis. In addition the use of
different antibodies and methods may also contribute to the
variance. Like ABCB1, ABCC1 SNPs were not associated
with ovarian cancer outcome [125].
2.3.2. ABCC2
ABCC2 (MRP-2), the canalicular multispecific organic
anion transporter (cMOAT), is 190 kDa and has 49% AA
identity with ABCC1 [191]. It is thought to have a physiological role as an organic anion pump in the liver and play a role
in detoxification of alkylating agents in the apical epithelium of the liver and kidney [192,193]. ABCC2 positivity
varies from 16% to 100% in ovarian cancer tissue cohorts
[91,114,194,195] (Table 4). The largest cohort examined to
date included 115 ovarian cancer tumours [91]. There are
studies which described ABCC2 protein or ABCC2 mRNA
levels to be associated with outcome [145,196,197] while
others did not [91,100,190,195]. The reason for this inconsistency is not clear but possible reasons may due to small cohort
size and the inclusion of different ovarian cancer subtypes in
the analysis. Another possible explanation for this inconsistency may lie in the localisation of ABCC2 within the cell.
Surowiak et al. found that higher nuclear ABCC2 levels both
before and after chemotherapy resulted in cisplatin resistance
and shorter survival time. However, total and cytoplasmic
ABCC2 levels were not associated with outcome [196].
Our data concurs with these findings as we found increased
ABCC2 expression in ovarian cancer cells treated with carboplatin [113]. Furthermore, minimal ABCC2 protein was
detected by immunohistochemistry in ovarian cancer tissues
collected at surgery prior to chemotherapy treatment (Fig. 4A
and B) but increased nuclear ABCC2 levels were observed in
tumour tissues from patients that received either neoadjuvant
single carboplatin (Fig. 4C) or carboplatin + paclitaxel treatment (Fig. 4D). High nuclear ABCC2 levels were also present
in a recurrent ovarian cancer from a patient that had received
adjuvant carboplatin + paclitaxel treatment (Fig. 4E). As for
ABCB1 and ABCC1, ABCC2 SNPs appear not be associated
with ovarian cancer outcome [125,198].
2.3.3. ABCC3
ABCC3 (MRP-3) has 58% AA identity with ABCC1 [191]
and transports organic conjugates and nucleosides [180].
ABCC3 expression is increased in lung cancer [199], breast

cancer [200], cervical cancer [201], hepatocellular carcinoma


[51], and pancreatic cancer [42]. Less is known about the role
of ABCC3 in ovarian cancer compared with ABCC1 and
ABCC2. ABCC3 expression correlates with ABCC1 [190]
and found to be higher in clear cell than serous papillary carcinomas [190]. ABCC3 was localized in the plasma membrane
and cytoplasm and ABCC3 mRNA levels were significantly
lower in ovarian cancer patients with a PFS period 2 years
[190]. ABCC3 was significantly elevated in recurrent ovarian
cancers [145] as well as in late stage ovarian cancers compared with early stage lesions [202]. Whilst studies have not
yet been undertaken to determine whether ABCC3 transports
ovarian cancer chemotherapy drugs, our own recent studies
show that treatment of ovarian cancer cells with carboplatin
increases the expression of the extracellular matrix component, hyaluronan, which in turn increased the expression of
ABCC3 in cells expressing the hyaluronan receptor, CD44
[113].
2.3.4. ABCC4
ABCC4 has the ability to transport molecules involved in
cellular signalling including cyclic nucleotides, eicosanoids,
urate, and conjugated steroids [203]. ABCC4 is increased
in a range of cancer including hepatocellular carcinoma
[51], breast cancer [52], melanoma [136] and lung cancer
[204]. ABCC4 protein is increased in oesophageal [205] and
prostate cancer [206]. ABCC4 has recently been linked with
increased cancer cell proliferation [204,205,207,208]. Little
is known about the role of ABCC4 in ovarian cancer but Bagnoli et al. showed that high ABCC4 expression was associated
with PFS [188], and Beretta et al. suggested that ABCC4 glycosylation may play a role in ovarian cancer chemoresistance
[209].
2.3.5. ABCC5
ABCC5 (MRP-5) is known to transport organic conjugates and nucleosides [180]. Increased ABCC5 expression
has been associated with hepatocellular carcinoma [51],
nasopharyngeal carcinoma [186], pancreatic ductal adenocarcinoma [42], and breast cancer [44]. Only one study
currently has shown a potential role for ABCC5 in ovarian
cancer. Whilst ABCC5 expression was not associated with
histo-pathological parameters or PFS [210], it was found to
be elevated in late stage ovarian cancer patients compared
with early stage ovarian cancer [202].

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

235

Table 4
Summary of ABCC2 studies in ovarian cancer tissue cohorts.
Cohort details

Type of analysis

Major findings

Ref.

Pre treatment (n = 115)


Mucinous/Clear cell (89)
Other (69)
Pre treatment (n = 17)
Subtypes not specified

Immunohistochemistry (frozen)
Mouse monoclonal cloneM2 III-6
Cut point 10%
Immunohistochemistry (paraffin, heat
retrieval)
Rabbit polyclonal EAG5 &
Mouse monoclonal clone M2 III-6
Quantitative RT-PCR

16% positive
No relationship with 1st line chemotherapy
response or PFS
94.1% positive with EAG5 antibody
100% positive with M2-III-6 antibody

[91]

ABCC2 expressed in 100% cases


No relationship with chemotherapy response
ABCC2 expression was not associated with
chemotherapy response or PFS.
ABCC2 expressed in 48.3% of cases.
No relationship with chemotherapy response
or PFS
ABCC2 expressed in 56% of platinum
sensitive tumours and 235 of platinum
resistant tumours. ABCC2 expression not
correlated with platinum response

[100]

Immunohistochemistry (paraffin, heat


retrieval)
Mouse monoclonal clone M2 I-4
Cut point not indicated

Increased nuclear ABCC2 expression was


associated with reduced PFS and OS.

[196]

Quantitative RT-PCR

ABCC2 expression was increased in


recurrent cancers compared to primary
cancers and benign tumours

[145]

Immunohistochemistry (paraffin, heat


retrieval)
Mouse monoclonal clone M2 III-6
Cut point 10%

High ABCC2 expression associated with


reduced PFS

[197]

Advanced pre treatment (n = 27)


Subtypes not specified
Pre-treatment (n = 30)
Serous
Pretreatment (n = 60)
Serous (50)
Endometrioid (10)
Pretreatment (n = 22)
Platinum sensitive (9)
Serous (9)
Platinum resistant (13)
Serous (9)
Clear cell (1)
Mucinous (1)
Endometrioid (1)
Pretreatment (n = 41)
Serous (35)
Endometrioid (3)
Others (3)
Pre-treatment (n = 50)
Serous (40)
Endometroid (7)
Mucinous (1)
Others (2)
Recurrent disease (n = 50)
Serous (44)
Endometroid (2)
Mucinous (1)
Others (3)
Pre-treatment (n = 47)
Serous (34)
Non-serous (13)

Quantitative RT-PCR
Immunohistochemistry (paraffin)
Rabbit polyclonal H-17 (Santa Cruz)
Cut point 5%
Immunohistochemistry (paraffin, heat
retrieval)
Mouse monoclonal clone M2 III-6

[194]

[190]
[114]

[195]

References in bold indicate studies that found relationships with chemotherapy response or survival outcome.

2.3.6. ABCC6
ABCC6 is best known for its mutations causing the softtissue mineralisation in pseudoxanthoma elasticum due to a
decrease in the release of ATP in the liver by ABCC6 leading
to reduced production of the mineralization inhibitor inorganic pyrophosphate [22]. Whilst it has been shown that
ABCC6 can transport a range of cancer drugs (Table 1),
there is little known about the role of ABCC6 in cancer
chemoresistance. There have been suggestions that ABCC6
is coamplified with ABCC1 because of its close location on
the same chromosome rather than having a specific regulatory control [211]. ABCC6 has, however, been shown to be
up-regulated in breast cancer [52] and it is part of a methylation profiling test 11-gene set which can predict bladder
cancer with 87% accuracy [212]. ABCC6 was found to be upregulated in mucinous ovarian adenocarcinomas compared

with benign mucinous cystadenomas [213]. ABCC6 has also


been shown to be up-regulated in paclitaxel and topotecan
resistant sublines of ovarian cancer cells [60].
2.3.7. ABCC7
ABCC7 is a chloride channel most commonly known
as cystic fibrosis transmembrane conductance regulator
(CFTR). ABCC7 mutations are responsible for creating
excess mucus found in the lungs in cystic fibrosis patients
[11]. Whilst no drug transport role for ABCC7 has been
reported, ABCC7 expression is increased in cervical cancer
[214] and ABCC7 mutations have been linked with pancreatic cancer [215]. However, low expression was associated
with advanced stage, lymph node metastasis and poor survival in non-small cell lung cancer patients [216] ABCC7
mRNA and protein expression has also been associated with

236

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

breast cancer treatment response [44,217] and tumour growth


suppression in prostate cancer [218]. ABCC7 expression has
been associated with increased oestrogen production and is
reduced in patients with polycystic ovary syndrome [16]. A
single study demonstrating that high ABCC7 expression was
associated with FIGO stage, poor histopathological grade and
serum CA125 links ABCC7 to ovarian cancer [219].

2.3.12. ABCC12
As one of the more recently discovered ABC transporters,
ABCC12 is poorly understood and its function is unknown.
ABCC12 expression is up-regulated in hepatocellular carcinoma [51] and breast cancer [52,226], but no studies
involving ovarian cancer have been reported to date.
2.4. The ABCD family

2.3.8. ABCC8
ABCC8 (sulfonylurea receptor 1/SUR1) is best known as
a sensor of intracellular levels of ATP and ADP and facilitates
the open or closing its associated KATP channel [220]. It is the
target of the sulfonylurea class of antidiabetic drugs which
promote insulin release from pancreatic beta cells. Whilst it
has not been reported to be involved in chemoresistance, it
has been associated with more aggressive cervical cancers
[221], high grade breast cancers [52], and brain metastases
[222]. No studies to date, have reported a role for ABCC8 in
ovarian cancer.
2.3.9. ABCC9
Splice variants arising from a single ABCC9 gene give
rise to SUR2A and SUR2B which like ABCC8 interacts
with the KATP channel in beta cells of the pancreas. Most
recently, ABCC9 mutations have been shown to result in
Cant syndrome, a rare genetic disease that results in bone
abnormalities, increased body hair, and an enlarged heart
[18]. ABCC9 variants have also been linked to the duration of
sleep required by individuals to feel rested [223]. Currently,
no studies link ABCC9 to any cancer.
2.3.10. ABCC10
ABCC10 is a 170 kDa protein that is localized on the
basolateral cell membrane which is well known as a broad
xenobiotic transporter (Table 1). ABCC10 has been associated with myeloid leukaemia [55], hepatocellular carcinoma
[51], pancreatic ductal adenocarcinoma [42], breast cancer
[52], as well as low grade colorectal tumours [69]. The
ABCC10 gene is hypermethylated in ovarian cancer patients
with a longer PFS [75]. Furthermore, ABCC10 expression
levels are increased in ovarian cancer xenograft tumours following treatment with doxetaxel [224]. Given the ability of
ABCC10 to efflux ovarian cancer therapeutics, further studies
to examine its role in chemoresistance are warranted.
2.3.11. ABCC11
A SNP in the ABCC11 gene is responsible for determination of human earwax type and presence of underarm odour
[225]. Despite having been found to transport a variety of
chemotherapeutic agents (Table 1), its role in cancer remains
relatively unexplored. ABCC11 expression is increased in
hepatocellular carcinoma [51], and breast cancer [44,52], but
no studies involving ovarian cancer have been reported.

The ABCD family (ABCD1-4) are peroxisomal halftransporters [227] (Fig. 1) whose mutations lead to impaired
peroxisomal beta-oxidation and accumulation of saturated
very long-chain fatty acids in tissues and body fluids associated with X-adrenoleukodystrophy, an inherited disorder
of peroxisomal metabolism, biochemically characterised by
accumulation of saturated very long chain fatty acids [15].
2.4.1. ABCD1
As so many of the ABC half transporters are shown to
dimerise to function, it was assumed that the ABCD transporters also formed dimers. Cell FRET analysis showed that
ABCD1 forms a homodimer, and can form a heterodimer
with ABCD3 [228]. Whilst little is known about the role of
ABCD1 in cancer, ABCD1 has been reported to be downregulated in renal cancer [229] and to be up-regulated in
melanoma [74] and breast cancer [52]. To date, there have
been no studies investigating the role of ABCD1 in ovarian
cancer.
2.4.2. ABCD2
ABCD2 has a functional redundancy with ABCD1. However, studies have also suggested that ABCD2 has a specific
role in lipid metabolism [230]. ABCD2 has only recently
become a subject of investigation in cancer and its role in
cancer remains undefined. ABCD2 expression is significantly
down-regulated in breast cancer and patients with higher
ABCD2 mRNA levels were more likely to respond to treatment [52]. ABCD2 has been shown to be up-regulated in
a topotecan resistant ovarian cancer subline [60] and high
ABCD2 expression correlated with poor ovarian cancer OS
[173].
2.4.3. ABCD3
ABCD3 is one of the most abundant peroxisomal
membrane proteins. Cell FRET analysis has shown that
ABCD3 forms a homodimer as well as a heterodimer with
ABCD1.[228] The ABCD3 expression is significantly upregulated in breast cancer [52] and associated with prostate
cancer aggressiveness [231,232]. ABCD3 is part of a 9-gene
panel along with ABCC3 that can predict non-small cell lung
cancer diagnosis with 99.75% predictive accuracy [233]. Furthermore, a recent conference abstract suggests that silencing
ABCD3 in a prostate cancer cell line resulted in increased
sensitivity to paclitaxel [232]. As paclitaxel is a key ovarian cancer drug these findings suggest that further studies

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

examining its role in ovarian cancer chemoresistance are


warranted.
2.4.4. ABCD4
ABCD4 has recently been shown not to be a peroxisomal
membrane protein but to reside in the ER. It has a 2527%
AA identity with the other three ABCD proteins but lacks
an N-terminal domain [234]. ABCD4 mutations have also
recently been shown to be involved in a lysosomal vitamin
B12 disorder [235]. There is a scarcity of studies investigating
the role of ABCD4 in cancer. A recent gene expression study
showed that the ABCD4 gene is significantly down-regulated
in breast tumours [52]. No studies to date, have reported a
role for ABCD4 in ovarian cancer.
2.5. The ABCE family
2.5.1. ABCE1
The only existing ABCE transporter, ABCE1/RLI, is
highly expressed in human lung adenocarcinoma tissues and
metastatic lymph nodes [236]. ABCE1 is a unique ABC
transporter as it has no TMDs and is therefore more akin to
an ABC-type ATPase than an ABC transporter. The structure of non-human ABCE1 has been solved [35] and it
likely plays a role in mRNA translation [237]. ABCE1 is
up-regulated in colorectal cancer [69], breast cancer [52],
melanoma [136], and hepatocellular carcinoma [51]. It has
been associated with proliferation, motility, and invasiveness
of oesophageal cancer cells [238]. Certain mutations are associated with prostate cancer [239], and ABCE1 siRNA knock
down inhibited the proliferation, invasiveness of small cell
lung cancer cells [240]. There have been no ovarian cancer
studies to date investigating ABCE1 expression.
2.6. The ABCF family
The ABCF transporters also do not have transmembrane
domains. They are similarly thought to play a role in mRNA
translation [237].
2.6.1. ABCF1
The ABCF1 gene is up-regulated in childhood T-cell acute
lymphoblastic leukaemia [54], and colorectal cancer [241],
as well as liver cancer where it was associated with poor outcome [242]. ABCF1 is up-regulated in breast cancer [52]
and is part of a six gene signature which can be used to
predict breast cancer patient outcome [243]. A single study
investigating the role of ABCF1 in ovarian cancer found a 3fold increase in ABCF1 in high grade stage I serous ovarian
carcinoma compared with normal tissue [62].
2.6.2. ABCF2
ABCF2 expression is significantly increased in melanoma
[136]. In breast cancer increased expression is associated with
treatment response and negatively associated with metastasis
[44,69,244]. Low ABCF2 expression was also found to be

237

an independent prognostic factor for survival in cervical cancer [245]. In ovarian cancer, it has been shown that ABCF2
is up-regulated after chemotherapy in high grade epithelial
ovarian cancers when compared with matching tissue before
treatment [246]. Interestingly, Tsuda et al. showed that levels
of ABCF2 mRNA and protein were increased in clear cell
compared with serous ovarian carcinomas [247] suggesting
a possible reason why clear cell carcinomas have a poorer
prognosis and are often more chemoresistant [94,248,249].
However, a later study showed that whilst ABCF2 expression was detected in the majority of clear cell ovarian cancer
patients it was not associated with clinical stage, response
to chemotherapy, or outcome [250]. Furthermore, a study
by Auner et al. did not find ABCF2 to be elevated in recurrent ovarian cancer [145]. These conflicting reports indicate
that further work is necessary to elucidate whether or not
ABCF2 plays any role in ovarian cancer progression and
chemoresistance.
2.6.3. ABCF3
ABCF3 is the least studied member of the ABCF family.
ABCF3 has been reported to be significantly up-regulated
in breast cancer [52], melanoma [136], and cervical cancer
[251]. No studies have linked ABCF3 to ovarian cancer.
2.7. The ABCG family
The ABCG family are half transporters thought to homoor hetero-dimerise in order to function as lipid transporters
(Fig. 1).
2.7.1. ABCG1
ABCG1 is the human homologue of the Drosophila white
gene, and is involved in the transport of cholesterol and phospholipids. It is up-regulated in breast cancer [52], pancreatic
ductal adenocarcinoma [42] and prostate cancer [252]. The
ABCG1 gene is hypermethylated before and after decitabine
treatment in ovarian cancer patients with longer survival [75].
Further studies are required to understand its role in ovarian
cancer.
2.7.2. ABCG2
ABCG2 is also known as the breast cancer resistance
protein (BCRP) or the mitoxantrone transporter (MXR1).
ABCG2 plays a protective role in the mammary glands
and can concentrate toxins released into breast milk [253].
ABCG2 is also localized in the placenta where it effluxes
xenobiotics and toxic endogenous compounds to protect
the developing foetus [254]. It has also been shown to
efflux urate and ABCG2 mutations have been linked to gout
[255]. ABCG2 is up-regulated in a wide range of cancers,
including nasopharyngeal carcinoma [186], pancreatic ductal adenocarcinoma [42], breast cancer [256], adenoid cystic
carcinoma [257], esophageal cancer [258], and tongue cancer
[259].

238

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

ABCG2 is a marker of ovarian cancer stem cells and


highly expressed in side population (SP) cells and ovarian
cancer spheroids which have higher chemoresistance to cisplatin or paclitaxel [260,261] (see Cancer stem cells and
ABC transporters section below). Like ABCB1, there has
been some controversy whether ABCG2 SNPs are associated
with ovarian cancer outcome. One study found no relationship [125] whilst another study found an ABCG2 variant to
be associated with longer PFS [198]. Interestingly an ABCG2
SNP was associated with increased oral bioavailability of the
ABCG2 substrate, topotecan [262]. Two polymorphic loci in
the ABCG2 promoter were associated with lower ABCG2
levels and higher plasma concentrations of erlotinib in a
prospective clinical study of 80 patients with non-small-cell
lung cancer, head and neck cancer, and ovarian cancer [263].
ABCG2 expression is increased in topotecan resistant
ovarian cancer cells [264,265]. ABCG2 is induced by physiological concentrations of oestrogen in the PA-1 ovarian
cancer cell line transfected to overexpress oestrogen receptor suggesting the possibility that estrogens can directly
stimulate the production of this multidrug resistance protein [266]. Our own recent immunohistochemistry studies
detected minimal ABCG2 protein in ovarian cancer tissues
prior to chemotherapy (Fig. 4A and B) but found increased
membranous ABCG2 levels in tumour tissues from patients
with recurrent ovarian cancers that had received combined
carboplatin + paclitaxel treatment (Fig. 4E and F).
2.7.3. ABCG4
The function of ABCG4 in humans is unknown, but mouse
experiments suggest it transports oxysterols in the brain
[267]. Very little is known about any possible function in
cancer, but it has been shown to be highly expressed in nonsmall-cell lung cancer and correlate with grade and stage
[268]. ABCG4 was found to be hypermethylated before and
after decitabine treatment in ovarian cancer patients with
longer survival [75]. Further studies are required to understand it role in ovarian cancer.
2.7.4. ABCG5 and ABCG8
The ABCG5 and ABCG8 proteins unite to form the
functional heterodimeric transporter [269] and are involved
in the secretion of plant sterols and cholesterol into the bile in
the liver. Mutations in either ABCG5 or ABCG8 cause sitosterolemia, a rare autosomal recessive disorder characterised
by accumulation of both plant-derived sterols and animalderived sterol (cholesterol) in plasma and tissues, leading
to the development of xanthomas [270]. Whilst most of the
attention has been directed at liver and gall bladder diseases,
ABCG5 and ABCG8 were also found to be significantly
down-regulated in breast cancer [52] and overexpressed in
gastric cancer [271]. Additionally, ABCG5 was associated
with poor outcome in node-negative colorectal cancer [272]
and linked with chemoresistance of head and neck cancers
[273]. No studies to date have investigated whether altered
sterol/cholesterol transport by ABCG5/8 plays a role in

ovarian cancer. This heterodimer warrants further study, as


early studies show that cholesterol availability in ovarian
cancer cells may play a role in their chemoresistance [274].

3. Cancer stem cells and ABC transporters


Ovarian tumour initiating cancer cells and SP cells with
stem cell-like properties have been shown to overexpress several ABC drug transporters including ABCB1 and ABCG2
[275277]. Chemoresistance in SP cells is mediated by both
ABCB1 and ABCG2 [260,275,278280]. When injected i.p.
into nude mice, SP cells were more tumorigenic than nonSP cells [275]. Ovarian cancer stem cells characterised by
CD44+ CD117+ isolated from SKOV3 cells when grown in
3D culture where reported to be more resistant to anti-cancer
drugs and had increased ABCG2 and ABCB1 expression
[281]. A study by Bourguignon and colleagues showed that
activation of the stem cell marker Nanog was mediated by
ABCB1 via hyaluronan-CD44 interactions [282]. A more
recent study demonstrated that chemoresistance in ovarian
tumour-initiating cells is mediated by c-Kit and involves the
activation of ABCG2 [283]. Other ABC transporters shown
to be up-regulated in cancer stem cells are ABCA3, ABCA5
and ABCB5. ABCA3 is up-regulated in AML SP and SP
cells isolated from neuroblastomas [284,285] and ABCA5
has been identified in osteosarcoma stem cells [64]. ABCB5
is increased in cancer stem cells identified in melanoma [286]
and squamous cell carcinomas [152].

4. Clinical trials using inhibitors of ABC transporters


Although ABC transporter inhibitors have been very
effective in preclinical studies [287289] (Table 1), clinical trials involving a range of ABC transport inhibitors have
proven disappointing [29,289291]. Historically, research on
blocking ABC transporter mediated MDR has focused on
inhibiting the activity of the transporters. In the 1980s Verapamil was discovered, the first drug to inhibit ABCB1s role
in decreasing sensitivity to vincristine and vinblastine [292].
Then came the discovery that cyclosporine A (CsA) could
reverse primary resistance of leukaemia and ascites carcinoma cells to vincristine and daunorubicin [293,294]. Both
of these drugs work by interacting with the substrate binding
pocket of ABCB1 [295,296] but failed in drug trials because
of immunosuppressive and nephrotoxic effects [297].
First generation ABCB1 inhibitors nicardipine and
nifedipine which also inhibit ABCB1 efflux were similarly
found to be quite toxic when given at high doses required to
inhibit ABCB1. The second generation inhibitors included
an analogue of cyclosporine A (CsA), PSC 833 or valspodar,
which was 10 fold more potent than CsA, but did not elicit
the toxicity or immunosuppressive response of CsA [298]. A
phase I/II trial where valspodar was given to ovarian cancer
patients who had failed platinum- and anthracycline-based

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

chemotherapy showed some promise of acceptable toxicity


and efficacy [299]. Among the 33 patients treated at the
maximal tolerated dose for doxorubicin, one (3%) had a
complete response, four (12%) had a partial response and an
additional seven patients (21%) experienced a stabilization
of their previously progressive disease. Another phase I
study treating patients with advanced breast and ovarian
cancer with pegylated liposomal doxorubicin and valsopdar
also did not report increased toxicity [300]. Some clinical
benefit was observed in 5 out of 10 patients with advanced
ovarian cancer receiving a combination of paclitaxel and
valspodar [301]. However a phase II trial with patients with
refractory ovarian cancer found that valspodar was ineffective at reversing paclitaxel resistance [302]. Valspodar failed
to enhance efficacy of paclitaxel and carboplatin treatment
and was more toxic in a phase III trial with advanced
ovarian cancer patients [303]. Similarly, dexverapamil, a
dextrorotatory isomer of verapamil never progressed beyond
phase II clinical trials for a range of cancers in the 90s (but
not ovarian cancer) despite data suggesting tolerable toxicity
and some level of efficacy [304,305]. Similarly, another
second generation inhibitor, biricodar, known to inhibit
efflux activity of ABCB1 as well as ABCC1 and ABCG2,
showed some efficacy in a phase II trial in ovarian cancer
patients [306] but did not have sufficient efficacy to progress
to phase III for ovarian cancer or other malignancies [307].
A wide range of third generation ABCB1 inhibitors have
also been evaluated in clinical trials. Tariquidar which inhibits
ABCB1 by binding at a site distinct from the paclitaxel and
vinblastine recognition site [308] provided enhanced sensitivity to doxorubicin, paclitaxel, etoposide, and vincristine
in phase III trials with lung cancer patients. However, trials had to be terminated due to its increased toxicity and
interaction with the chemotherapeutics. A trial investigating
the efficacy of tariquidar in addition to docetaxel in ovarian cancer patients showed a partial response in a single
patient after 5 failed lines of chemotherapy [309]. The results
were similar for other third generation ABCB1 inhibitors
such as zosuquidar, elacradir, and dofequidar. Zosuquidar
(LY335979), a difluorocyclopropyl quinoline, which binds
with high affinity to ABCB1 did not result in pharmacokinetic interactions with co-administered doxorubicin [310].
In a phase I trial of 36 patients with resistant cancers treated
with zosuquidar and docetaxel, 14 patients had disease stabilization and one patient with ovarian carcinoma, previously
treated with paclitaxel before the study had a partial response
with the combined treatment [311]. However, zosuquidar
did not provide any additional survival benefit for breast
cancer patients with metastatic or locally recurrent breast
cancer who received prior chemotherapy [312]. Elacridar
(GF-120918) which inhibits the activity of both ABCB1 and
ABCG2 was shown to increase the oral bioavailability of
topotecan but was not further investigated in phase II/III clinical trials [313,314]. Dofequidar (MS209) can be given in
combination with docetaxel, with limited effect on docetaxel
toxicity or pharmacokinetics [315]. Although dofequidar plus

239

cyclophosphamide, doxorubicin, and fluorouracil (CAF)


therapy in comparison with CAF alone was well tolerated
in patients with advanced or recurrent breast cancer it only
had limited efficacy in patients who had not received prior
therapy [316].
Recently, two new ABCB1 inhibitors have been identified
by screening small molecule compounds together with paclitaxel against resistant ovarian cancer cells lines. NSC23925
which was synthesized in the 1940s as an anti-malarial drug,
inhibits ABCB1 and increases accumulation of paclitaxel
but does not inhibit ABCC1 or ABCG2-mediated MDR.
Its inhibitory effect was not altered by co-incubation with
the ABCB1 inhibitor verapamil, suggesting that NSC23925
itself is not a substrate of ABCB1 [317]. In follow up
studies, NSC23925 completely reversed paclitaxel resistance in SKOV-3TR ovarian MDR mouse xenograft models,
without increasing the level of paclitaxel toxicity [318].
Similarly, NSC77037 reversed ABCB1 MDR without reversing ABCC1 or ABCG2-mediated MDR [319]. The ABCC1
inhibitor reversan has recently being identified by screening
small molecule libraries [320]. Reversan which can inhibit
the function of both ABCC1 and ABCB1 was found to be
non-toxic in murine models of neuroblastoma [320] but has
yet to be tested in patients.
Tryosine kinase inhibitors erotinib, getitinib, and imatinib increase the accumulation and cytotoxicity of ABCG2
substrates, mitoxantrone and methotrexate [321] and also
inhibit expression of ABCG2 via the P13K-AKT pathway
[322]. Lapatinib, a small pan-erbB inhibitor which can block
ABCB1 and ABCG2 efflux of toptecan, failed to show sufficient efficacy in refractory ovarian cancer patients to progress
to phase III trials [323]. Athough imatinib treatment was well
tolerated it did not produce clinical responses in patients
with recurrent ovarian cancer either alone [324,325] or in
combination with docetaxel [326].
Other agents including the non-steroidal antiinflammatory drug sulindac and the src inhibitor saracatinib
(AZD0530) shown to inhibit ABC transporter activity have
also been evaluated in clinical trials. A Phase I clinical
trial found that the combination of epirubicin with sulindac
was well tolerated in patients with advanced cancer [327].
Recent studies have shown that sulindac can inhibit ovarian
cancer cell proliferation and reduce tumour growth in mice
[328]. Sulindac is being studied in clinical trials for colon
cancer [329], however, no studies have been reported to
date in patients with refractory ovarian cancer. Saracatinib
which was shown to inhibit ABCB1 function and enhance
the effect of paclitaxel in ABCB1 overexpressing cancer
cells [330] has just completed a Phase II trial in patients
with relapsed platinum-resistant ovarian cancer [331]. The
phosohodiesterase-5 inhibitor sildenafil, has also been
shown to inhibit both ABCB1 and ABCG2 at clinical
achievable concentrations and efflux several chemotherapy
drugs including paclitaxel [332]. In animal models sildenafil
improved the efficacy of doxorubicin against prostate cancer
[333]. However no clinical trials have been reported to date

240

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

examining the effect of phosohodiesterase-5 inhibitors on


ABC transporters. The anti-viral drug and protease inhibitor
ritonavir has also been shown to be a potent ABCB1
inhibitor [334]. In combination with docetaxel, ritonavir was
well tolerated and increased the bioavailability of docetaxel
[335].
A major reason for the failure of three generations of
ABC inhibitors has been the unexpected alterations in the
pharmacokinetics of chemotherapy drugs and increased toxicity when combined with ABC transporters inhibitors. This
is likely due to the fact that the physiological function of
many ABC transporters is to provide protection against toxins. Thus inhibiting ABC transporters is likely also to inhibit
their ability to reduce the toxicity of chemotherapy drugs.
Furthermore ABC transporters are involved in the absorption,
excretion and distribution of chemotherapy agents. Many
inhibitors may yet be proven to be effective treatments if
they can be delivered with greater specificity to the cancer cells and increase the bioavailability of the drug only
in the area of the tumour. In addition to these issues, many
clinical trials were initiated without adequate preclinical evaluation. Increased accumulation of chemotherapy agents in
cell lines expressing a single ABC transporter fail to take
into consideration the physiological functions of the transporters in a whole organism, and the possible co-expression
of many transporters within tumours and neighbouring tissues. Analysis of the structures of ABCB1 in the presence
of several substrates including paclitaxel and doxorubicin
have shown a large degree of flexibility in the transporter
cavity when it comes to substrate interaction, highlighting
the difficulty of designing competitive inhibitors [336338].
Additionally, many ABC transporter inhibitors exhibit crossinhibition with other necessary proteins, leading to increased
side effects. Thus all new ABC transporter inhibitors need
to be tested in preclinical animal models with established
tumours. There is also much speculation as to the best animal models to use for ovarian cancer as they do not model all
subtypes and are not truly reflective of human disease [29]. In
addition to this, most clinical trials did not specifically target
patients with particular cancer type, with many assessing the
effects on tumours from a range of origins. Another potential
reason for the failure of the ABC transporter inhibitor trials
is that most patients in the clinical studies were enrolled after
multiple lines of therapy, and often differing therapies, making it difficult to assess their current chemoresistant states.
Furthermore, none of the trials selected patients based on the
expression levels of the ABC transporters, thereby allowing
effects seen in patients with high ABC transporter expression to be potentially masked by patients not expressing the
inhibitors intended target at all. And probably most importantly, most clinical trials to date have only targeted ABCB1
and more recently ABCC1 and ABCG2. This is a major flaw,
as Table 1 shows, many ABC transporters are capable of
effluxing chemotherapy agents than most would generally be
aware and this number is increasing each year. Single ABC
transporter inhibitors fail to take this into account, and any

effects of the inhibitor may well be masked by other ABC


transporters still left in play. Future clinical trials should take
these many factors into account to ensure the best outcome.

5. Alternative strategies to overcome ovarian cancer


MDR
The use of monoclonal antibodies, siRNAs, antisense
oligonucleotides and transcription factor regulators are alternative approaches to target ABC transporters [289,291].
Double stranded antisense oligonucleotides were effective
at inhibiting ABCB1 expression in A2780/Adr ovarian cancer cell lines [339]. Several studies have shown that RNAi
methods are effective at reversing chemotherapy resistance in
ovarian cancer cells in vitro and in vivo studies [82,340,341].
ABCC2 siRNA were also shown to be effective at inhibiting
ABCC2 expression and reversing resistance against cisplatin and paclitaxel of the human ovarian carcinoma cell
line A2780RCIS [342]. Treatment with DNA G-quadruplex
oligonucleotides decreased expression of ABCB1 in SKOV3 cells and reversed their resistance to paclitaxel [343].
Although promising, none these approaches have made it into
clinical trials yet. A major challenge of using RNAi mediated
methods is to specifically deliver them to resistant tumour
cells and avoid systemic side effects.
Another area under current investigation is the use of ABC
transporter inhibitors to block the bodys natural ability to
efflux and remove drugs administered orally, which show
reduced efficacy in comparison to intravenous administration. In particular, the ABCB1 inhibitor, HM30181 could
increase the bioavailability of paclitaxel in rats and inhibited
the growth of colon tumour xenografts with greater efficacy
when given orally instead of intravenously [344]. Follow up
phase I trials have shown that HM30181 is well tolerated
by patients [345,346]. Phase Ib trials are now recruiting to
test OraxolTM , a new oral formulation consisting of paclitaxel and HM30181 in a range of cancer patients including
ovarian cancer [347].
Many natural products have also shown some promise
in reversing ABC transporter mediated chemoresistance in
ovarian cancer cells. Several compounds including curcumin
and plant polyphenols demonstrated broad spectrum effects
on more than one ABC drug transporter [348355]. The
major components of green tea, epigallocatechin gallate, and
cruciferous vegetables, sulforaphane respectively were effective at overcoming paclitaxel resistance in SKOV3TR-ip2
xenografts by inducing apoptosis [356]. However a recent
phase trial II found that epigallocatechin gallate enriched
green tea did not improve recurrence rates in women with
advanced ovarian cancer after completing 68 cycles of first
line chemotherapy [357]. A recent study found that Brucea
javanica fruit oil emulsion in conjunction with cisplatin treatment led to smaller tumour in SKOV-3 xenograft mice and
downregulation of ABCC1 [358]. Natural products have great
potential for treating refractory disease in cancer patients;

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

however unpredictable pharmacokinetic drug interactions


with chemotherapeutics and toxicity will need to be carefully
evaluated.
Alternative drug delivery approaches using liposomes and
nanoparticles have also been tested in vitro and in vivo
ovarian cancer models. Liposomes containing tariquidar and
paclitaxel resulted in greater toxicity to the drug resistant ovarian cancer SKOV-3TR [359,360]. Biodegradable
polymeric nanoparticle formulations combining ABCB1
siRNA and paclitaxel could increase cytotoxicity in MDR
SKOV3(TR) cells by increasing cellular levels of paclitaxel
[361]. Chitosan shRNA ABCB1 plasmid nanoparticles were
also effective at reversing paclitaxel resistance of A2780/TS
ovarian cancer cells [362]. Nanoscale coordination polymers
containing cisplatin and ABCB1 siRNA reduced the growth
of cisplatin resistant SKOV-3 xenografts in mice [363].
CD44 targeted HA-PEI/HA-PEG/ABCB1 siRNA nanoparticles caused decreased tumour growth after treatment with
paclitaxel in taxane resistant SKOV-3 and OVCAR-8 cells
[364,365]. Nanoparticle technology holds great promise for
targeted delivery of specific ABC inhibitors in combination
with chemotherapy drugs and deserves further attention.
Studies have shown that as cancer cells become more
resistant to one drug, they can simultaneously become more
susceptible to another, an event termed collateral sensitivity [366368]. In particular, common ovarian cancer drugs,
taxanes and platinum based agents, showed an inverse
relationship in resistance [369]. This concept has been extensively reviewed in [370]. A greater understanding of how
this occurs may lead to future therapies that can manipulate
collateral sensitivity to established drugs.
Another approach is to utilize compounds which lead
to a reduction in ABC transporter expression. One such
compound is NSC73306 which causes a loss of ABCB1
expression in vitro as well as a reduced MDR phenotype
[371,372]. NSC73306 exhibited selective toxicity to ABCB1
expressing MDR cancer cells by reducing expression of
ABCB1 rather than suppressing ABCB1 function [371]. An
additional 15 ABCB1 selective inhibitors were identified
by screening the National Cancer Institutes Developmental
Therapeutics Program drug repository [373]. These compounds along with NSC73306 and TSC Dp44mT [374] which
exploit ABCB1 expression rather ABCB1 function to induce
cytotoxicity have great potential to treat ABCB1 mediated
MDR. However to date there have been limited pre clinical
studies in mice and none of these have been tested in clinical
trials to date.
Antibodies are another exciting avenue being explored to
reverse ABC transporter mediated MDR. Antibodies against
ABCB1 have been shown to promote both lymphocyte- and
monocyte-mediated tumour cell killing [375]. Immunisation
against the extracellular loops of the mouse homologue of
ABCB1 doubled the survival time of xenograft mice [376].
Mouse monoclonal ABCB1 antibody UIC2 was shown to
inhibit the in vitro efflux of drug substrates but required
a substrate-bound form of ABCB1 in order to bind almost

241

100% of surface ABCB1 [377,378]. It was recently shown


that treatment with the UIC2 antibody as well as doxorubicin
dramatically reduced the size of epidermoid carcinomas in
SCID mice [379]. HPMA copolymers containing chemotherapy agents have used UIC2 as a way to target ABCB1
expressing ovarian cancer cells and showed a drastically
reduced IC50 for both chemotherapy agents tested [380]. An
ABCB1 antibody has also been conjugated to lentivirus as
a way to target gene therapy delivery [381]. These study
shows that there may be a way to specifically target cells
over-expressing ABC transporters, overcoming the problems
of toxicity via inhibition of physiological function in normal
tissues.

6. Conclusion
It has become increasingly apparent that many ABC transporters and not just ABCB1 play important roles in MDR
with 20 ABC transporters now known to efflux chemotherapy
drugs. ABC transporters contribute to tumour biology independently of their ability to efflux cytotoxic drugs and are also
involved in lipid export and lipid homeostasis and mediate the
release of bioactive lipids that activate signalling cascades
involved in cell proliferation, migration and tumorigenesis
[290]. Furthermore, ABC transporters also have important
roles in drug distribution and drug access across blood brain
barrier and intestine [382]. Unfortunately clinical trials evaluating three generations of ABC inhibitors have failed. A lot
has been learned from the experience and many reasons for
the failure have been realised. A major reason has been the
unexpected alterations in the pharmacokinetics of chemotherapy drugs and increased toxicity when combined with ABC
transporters inhibitors. One reason for these outcomes may
be the method of inhibition. As many of the ABC transporters
commonly targeted serve important physiological functions,
blocking this function results in the side effects outweighing any effect on chemoresistance. One way around this
issue in the future would be to have inhibitors designed to
block the recognition sites of the chemotherapy drugs, whilst
allowing the ABC transporter to perform its physiological
function. One of the best ways to achieve this would be to
structurally design inhibitors for the drug recognition sites.
However, this is not as easily done as said, as only a handful of ABC transporter structures have been determined to
date [3137], and most recently, human ABCB10 [38] and
mouse ABCB1 in the presence of designed substrates [39].
This is due to the difficulties in expressing, purifying, and
crystallizing the proteins in their native conformation in cellular membranes. It is our belief, that once structures become
more readily accessible, including structures of the transporters interacting with chemotherapeutic and physiological
substrates, that ABC inhibitors will prove a valid and effective way to reduce chemoresistance. Furthermore, improved
models are needed to improve pre-clinical selection if we
want to see more success in future clinical trials.

242

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

This review has revealed that there is more to the MDR


phenomenon than the most commonly studied ABC transporters, including ABCB1, ABCC1, and ABCG2. Indeed,
a thorough review of the literature has identified at least
20 ABC transporters that are capable of effluxing a range
of chemotherapy drugs (Table 1). As many transporters and
drugs have never been tested, it is extremely likely that this
number will continue to rise. It has become apparent that a
large number of ABC transporters could well be contributing to the MDR but have been neglected in favour of their
more popular cousins. The advancements in technology in
the omics fields provide researchers with the opportunity to
investigate the changes in cancer cells of not one or a few,
but all the ABC transporters in response to chemotherapy
drugs and new inhibitors. It is likely that multiple agents
will need to be combined to inhibit a wider range of ABC
transporters to prove effective at combating chemoresistance.
ABC transporter inhibition still remains an attractive potential adjuvant to chemotherapy but the discovery of novel
nontoxic inhibitors and the development of more effective
targeted therapies will be required to impact clinical outcome.
The most effective therapies will be those that find a way to
target not only multiple transporters at once, but also only
target cancer cells, avoiding toxicity effects from inhibiting
the physiological functions of the ABC transporters.

Conict of interest
The authors have no conflict of interest to declare.

Acknowledgements
This research has been funded by the Ovarian Cancer
Research Foundation (OCRF), Australia, Royal Adelaide
Hospital Clinical Research Fund, Cancer Council of South
Australia & South Australian Health and Medical Research
Institute.

References
[1] Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global
cancer statistics. CA Cancer J Clin 2011;61:6990.
[2] Ahmed FY, Wiltshaw E, AHern RP, Nicol B, Shepherd J, Blake P,
et al. Natural history and prognosis of untreated stage I epithelial
ovarian carcinoma. J Clin Oncol 1996;14:296875.
[3] Cannistra SA. Cancer of the ovary. N Engl J Med 2004;351:251929.
[4] Bast Jr RC, Hennessy B, Mills GB. The biology of ovarian cancer:
new opportunities for translation. Nat Rev Cancer 2009;9:41528.
[5] Agarwal R, Kaye SB. Ovarian cancer: strategies for overcoming resistance to chemotherapy. Nat Rev Cancer 2003;3:50216.
[6] Luqmani YA. Mechanisms of drug resistance in cancer chemotherapy.
Medical principles and practice: international journal of the Kuwait
University. Health Sci Cent 2005;14(Suppl. 1):3548.
[7] Sharom FJ. ABC multidrug transporters: structure, function and role
in chemoresistance. Pharmacogenomics 2008;9:10527.

[8] Galluzzi L, Senovilla L, Vitale I, Michels J, Martins I, Kepp


O, et al. Molecular mechanisms of cisplatin resistance. Oncogene
2012;31:186983.
[9] Thibault B, Castells M, Delord JP, Couderc B. Ovarian cancer microenvironment: implications for cancer dissemination and
chemoresistance acquisition. Cancer Metastasis Rev 2013.
[10] Dassa E, Schneider E. The rise of a protein family: ATP-binding
cassette systems. Res Microbiol 2001;152:203.
[11] Riordan JR, Rommens JM, Kerem B, Alon N, Rozmahel R, Grzelczak
Z, et al. Identification of the cystic fibrosis gene: cloning and characterization of complementary DNA. Science 1989;245:106673.
[12] Allikmets R, Shroyer NF, Singh N, Seddon JM, Lewis RA, Bernstein
PS, et al. Mutation of the Stargardt disease gene (ABCR) in age-related
macular degeneration. Science 1997;277:18057.
[13] Bekri S, Kispal G, Lange H, Fitzsimons E, Tolmie J, Lill R, et al.
Human ABC7 transporter: gene structure and mutation causing Xlinked sideroblastic anemia with ataxia with disruption of cytosolic
iron-sulfur protein maturation. Blood 2000;96:325664.
[14] Akiyama M, Sugiyama-Nakagiri Y, Sakai K, McMillan JR, Goto M,
Arita K, et al. Mutations in lipid transporter ABCA12 in harlequin
ichthyosis and functional recovery by corrective gene transfer. J Clin
Invest 2005;115:177784.
[15] Matsukawa T, Asheuer M, Takahashi Y, Goto J, Suzuki Y, Shimozawa
N, et al. Identification of novel SNPs of ABCD1, ABCD2, ABCD3,
and ABCD4 genes in patients with X-linked adrenoleukodystrophy
(ALD) based on comprehensive resequencing and association studies
with ALD phenotypes. Neurogenetics 2011;12:4150.
[16] Chen H, Guo JH, Lu YC, Ding GL, Yu MK, Tsang LL, et al.
Impaired CFTR-dependent amplification of FSH-stimulated estrogen production in cystic fibrosis and PCOS. J Clin Endocrinol Metab
2012;97:92332.
[17] Allen M, Zou F, Chai HS, Younkin CS, Crook J, Pankratz VS, et al.
Novel late-onset Alzheimer disease loci variants associate with brain
gene expression. Neurology 2012;79:2218.
[18] Harakalova M, van Harssel JJ, Terhal PA, van Lieshout S, Duran K,
Renkens I, et al. Dominant missense mutations in ABCC9 cause Cantu
syndrome. Nat Genet 2012;44:7936.
[19] Pahnke J, Frohlich C, Krohn M, Schumacher T, Paarmann K. Impaired
mitochondrial energy production and ABC transporter function-A
crucial interconnection in dementing proteopathies of the brain. Mech
Ageing Dev 2013;134:50615.
[20] Chen P, Dai Y, Wu X, Wang Y, Sun S, Xiao J, et al. Mutations in the
ABCA3 gene are associated with cataract-microcornea syndrome.
Invest Ophthalmol Vis Sci 2014;55:803143.
[21] Westerterp M, Bochem AE, Yvan-Charvet L, Murphy AJ, Wang
N, Tall AR. ATP-binding cassette transporters, atherosclerosis, and
inflammation. Circ Res 2014;114:15770.
[22] Jansen RS, Duijst S, Mahakena S, Sommer D, Szeri F, Varadi A,
et al. ABCC6-mediated ATP secretion by the liver is the main source
of the mineralization inhibitor inorganic pyrophosphate in the systemic circulation-brief report. Arterioscler Thromb Vascular Biol
2014;34:19859.
[23] Zaman GJ, Lankelma J, van Tellingen O, Beijnen J, Dekker H,
Paulusma C, et al. Role of glutathione in the export of compounds
from cells by the multidrug-resistance-associated protein. Proc Natl
Acad Sci U S A 1995;92:76904.
[24] Dean M, Allikmets R. Complete characterization of the human ABC
gene family. J Bioenerg Biomembr 2001;33:4759.
[25] Kerr ID. Structure and association of ATP-binding cassette
transporter nucleotide-binding domains. Biochim Biophys Acta
2002;1561:4764.
[26] Moody JE, Thomas PJ. Nucleotide binding domain interactions during the mechanochemical reaction cycle of ATP-binding cassette
transporters. J Bioenerg Biomembr 2005;37:4759.
[27] Davidson AL, Dassa E, Orelle C, Chen J. Structure, function, and
evolution of bacterial ATP-binding cassette systems. Microbiol Mol
Biol Rev 2008;72:31764.

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256


[28] Borst P, Zelcer N, van de Wetering K, Poolman B. On the putative
co-transport of drugs by multidrug resistance proteins. FEBS Lett
2006;580:108593.
[29] Tamaki A, Ierano C, Szakacs G, Robey RW, Bates SE. The controversial role of ABC transporters in clinical oncology. Essays Biochem
2011;50:20932.
[30] Chang G, Roth CB, Reyes CL, Pornillos O, Chen YJ, Chen AP.
Retraction. Science 2006;314:1875.
[31] Dawson RJP, Locher KP. Structure of a bacterial multidrug ABC
transporter. Nature 2006;443:1805.
[32] Dawson RJP, Locher KP. Structure of the multidrug ABC transporter
Sav1866 from Staphylococcus aureus in complex with AMP-PNP.
FEBS Lett 2007;581:9358.
[33] Hohl M, Briand C, Grutter MG, Seeger MA. Crystal structure of a
heterodimeric ABC transporter in its inward-facing conformation. Nat
Struct Mol Biol 2012;19:395402.
[34] Li J, Jaimes KF, Aller SG. Refined structures of mouse P-glycoprotein.
Protein Sci 2013.
[35] Karcher A, Schele A, Hopfner KP. X-ray structure of the complete ABC enzyme ABCE1 from Pyrococcus abyssi. J Biol Chem
2008;283:796271.
[36] Becker T, Franckenberg S, Wickles S, Shoemaker CJ, Anger AM,
Armache JP, et al. Structural basis of highly conserved ribosome
recycling in eukaryotes and archaea. Nature 2012;482:5016.
[37] Jin MS, Oldham ML, Zhang Q, Chen J. Crystal structure of the
multidrug transporter P-glycoprotein from Caenorhabditis elegans.
Nature 2012;490:5669.
[38] Shintre CA, Pike AC, Li Q, Kim JI, Barr AJ, Goubin S, et al. Structures of ABCB10, a human ATP-binding cassette transporter in apoand nucleotide-bound states. Proc Natl Acad Sci U S A 2013;110:
97105.
[39] Szewczyk P, Tao H, McGrath AP, Villaluz M, Rees SD, Lee SC,
et al. Snapshots of ligand entry, malleable binding and induced helical
movement in P-glycoprotein. Acta Crystallogr D Biol Crystallogr
2015;71:73241.
[40] Fitzgerald ML, Morris AL, Chroni A, Mendez AJ, Zannis VI,
Freeman MW. ABCA1 and amphipathic apolipoproteins form highaffinity molecular complexes required for cholesterol efflux. J Lipid
Res 2004;45:28794.
[41] Ingram WJ, Crowther LM, Little EB, Freeman R, Harliwong I, Veleva
D, et al. ABC transporter activity linked to radiation resistance and
molecular subtype in pediatric medulloblastoma. Exp Hematol Oncol
2013;2:26.
[42] Mohelnikova-Duchonova B, Brynychova V, Oliverius M, Honsova E,
Kala Z, Muckova K, et al. Differences in transcript levels of ABC
transporters between pancreatic adenocarcinoma and nonneoplastic
tissues. Pancreas 2013;42:70716.
[43] Vargas T, Moreno-Rubio J, Herranz J, Cejas P, Molina S,
Gonzalez-Vallinas M, et al. ColoLipidGene: signature of lipid
metabolism-related genes to predict prognosis in stage-II colon cancer
patients. Oncotarget 2015.
[44] Park S, Shimizu C, Shimoyama T, Takeda M, Ando M, Kohno T, et al.
Gene expression profiling of ATP-binding cassette (ABC) transporters
as a predictor of the pathologic response to neoadjuvant chemotherapy
in breast cancer patients. Breast Cancer Res Treat 2006;99:917.
[45] Hedditch EL, Gao B, Russell AJ, Lu Y, Emmanuel C, Beesley J, et al.
ABCA transporter gene expression and poor outcome in epithelial
ovarian cancer. J Natl Cancer Inst 2014:106.
[46] Chou JL, Huang RL, Shay J, Chen LY, Lin SJ, Yan PS, et al. Hypermethylation of the TGF-beta target, ABCA1 is associated with poor
prognosis in ovarian cancer patients. Clin Epigenet 2015;7:1.
[47] Chou J, Chen Y, Su H, Lin S, Huang Y, Deatherage DE, et al.
Hypomethylation of TGF-beta target gene, ABCA1 in ovarian cancer and cancer initialing cell and is associated with poor prognosis
in cancer patients. In: Proceedings of the 102nd annual meeting
of the American Association for Cancer Research (AACR). 2011.
p. 83.

243

[48] Oiso S, Takayama Y, Nakazaki R, Matsunaga N, Motooka C, Yamamura A, et al. Factors involved in the cisplatin resistance of KCP4
human epidermoid carcinoma cells. Oncol Rep 2013.
[49] Davis Jr W, Boyd JT, Ile KE, Tew KD. Human ATP-binding cassette
transporter-2 (ABCA2) positively regulates low-density lipoprotein
receptor expression and negatively regulates cholesterol esterification in Chinese hamster ovary cells. Biochim Biophys Acta
2004;1683:89100.
[50] Alla V, Kowtharapu BS, Engelmann D, Emmrich S, Schmitz U, Steder
M, et al. E2F1 confers anticancer drug resistance by targeting ABC
transporter family members and Bcl-2 via the p73/DNp73-miR-205
circuitry. Cell Cycle 2012;11:306778.
[51] Borel F, Han R, Visser A, Petry H, van Deventer SJ, Jansen PL,
et al. Adenosine triphosphate-binding cassette transporter genes upregulation in untreated hepatocellular carcinoma is mediated by
cellular microRNAs. Hepatology 2012;55:82132.
[52] Hlavac V, Brynychova V, Vaclavikova R, Ehrlichova M, Vrana
D, Pecha V, et al. The expression profile of ATP-binding cassette transporter genes in breast carcinoma. Pharmacogenomics
2013;14:51529.
[53] Ohtsuki S, Kamoi M, Watanabe Y, Suzuki H, Hori S, Terasaki T. Correlation of induction of ATP binding cassette transporter A5 (ABCA5)
and ABCB1 mRNAs with differentiation state of human colon tumor.
Biol Pharm Bull 2007;30:11446.
[54] Efferth T, Gillet JP, Sauerbrey A, Zintl F, Bertholet V, de Longueville
F, et al. Expression profiling of ATP-binding cassette transporters
in childhood T-cell acute lymphoblastic leukemia. Mol Cancer Ther
2006;5:198694.
[55] Steinbach D, Gillet JP, Sauerbrey A, Gruhn B, Dawczynski K,
Bertholet V, et al. ABCA3 as a possible cause of drug resistance
in childhood acute myeloid leukemia. Clin Cancer Res 2006;12:
435763.
[56] Laing NM, Belinsky MG, Kruh GD, Bell DW, Boyd JT, Barone L,
et al. Amplification of the ATP-binding cassette 2 transporter gene is
functionally linked with enhanced efflux of estramustine in ovarian
carcinoma cells. Cancer Res 1998;58:13327.
[57] Uhlen M, Oksvold P, Fagerberg L, Lundberg E, Jonasson K, Forsberg M, et al. Towards a knowledge-based Human Protein Atlas. Nat
Biotechnol 2010;28:124850.
[58] Wulf GG, Modlich S, Inagaki N, Reinhardt D, Schroers R, Griesinger
F, et al. ABC transporter ABCA3 is expressed in acute myeloid
leukemia blast cells and participates in vesicular transport. Haematologica 2004;89:13957.
[59] Overbeck TR, Hupfeld T, Krause D, Waldmann-Beushausen R, Chapuy B, Guldenzoph B, et al. Intracellular ATP-binding cassette
transporter A3 is expressed in lung cancer cells and modulates susceptibility to cisplatin and paclitaxel. Oncology 2013;84:
36270.
[60] Januchowski R, Zawierucha P, Andrzejewska M, Rucinski M, Zabel
M. Microarray-based detection and expression analysis of ABC and
SLC transporters in drug-resistant ovarian cancer cell lines. Biomed
Pharmacother 2013;67:2405.
[61] Liu Y, Peng H, Zhang JT. Expression profiling of ABC transporters in a
drug-resistant breast cancer cell line using AmpArray. Mol Pharmacol
2005;68:4308.
[62] Chien J, Fan JB, Bell DA, April C, Klotzle B, Ota T, et al. Analysis of
gene expression in stage I serous tumors identifies critical pathways
altered in ovarian cancer. Gynecol Oncol 2009;114:311.
[63] Nymoen DA, Holth A, Hetland Falkenthal TE, Trope CG, Davidson
B. CIAPIN1 and ABCA13 are markers of poor survival in metastatic
ovarian serous carcinoma. Mol Cancer 2015;14:317.
[64] Saini V, Hose CD, Monks A, Nagashima K, Han B, Newton DL, et al.
Identification of CBX3 and ABCA5 as putative biomarkers for tumor
stem cells in osteosarcoma. PLoS ONE 2012;7:e41401.
[65] Huang D, Gao Q, Guo L, Zhang C, Jiang W, Li H, et al. Isolation and
identification of cancer stem-like cells in esophageal carcinoma cell
lines. Stem Cells Dev 2009;18:46573.

244

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

[66] Kaminski WE, Wenzel JJ, Piehler A, Langmann T, Schmitz G.


ABCA6, a novel a subclass ABC transporter. Biochem Biophys Res
Commun 2001;285:1295301.
[67] Islam TC, Asplund AC, Lindvall JM, Nygren L, Liden J, Kimby E,
et al. High level of cannabinoid receptor 1, absence of regulator of
G protein signalling 13 and differential expression of Cyclin D1 in
mantle cell lymphoma. Leukemia 2003;17:188090.
[68] Santin AD, Zhan F, Bellone S, Palmieri M, Cane S, Bignotti E, et al.
Gene expression profiles in primary ovarian serous papillary tumors
and normal ovarian epithelium: identification of candidate molecular markers for ovarian cancer diagnosis and therapy. Int J Cancer
2004;112:1425.
[69] Hlavata I, Mohelnikova-Duchonova B, Vaclavikova R, Liska V, Pitule P, Novak P, et al. The role of ABC transporters in progression
and clinical outcome of colorectal cancer. Mutagenesis 2012;27:
18796.
[70] Theile D, Ketabi-Kiyanvash N, Herold-Mende C, Dyckhoff G, Efferth
T, Bertholet V, et al. Evaluation of drug transporters significance for
multidrug resistance in head and neck squamous cell carcinoma. Head
Neck 2011;33:95968.
[71] Kim WS, Hsiao JH, Bhatia S, Glaros EN, Don AS, Tsuruoka S, et al.
ABCA8 stimulates sphingomyelin production in oligodendrocytes.
Biochem J 2013;452:40110.
[72] Bou Zgheib N, Xiong Y, Marchion DC, Bicaku E, Chon HS, Stickles
XB, et al. The O-glycan pathway is associated with in vitro sensitivity
to gemcitabine and overall survival from ovarian cancer. Int J Oncol
2012;41:17988.
[73] Piehler A, Kaminski WE, Wenzel JJ, Langmann T, Schmitz
G. Molecular structure of a novel cholesterol-responsive A subclass ABC transporter, ABCA9. Biochem Biophys Res Commun
2002;295:40816.
[74] Chen KG, Valencia JC, Gillet JP, Hearing VJ, Gottesman MM.
Involvement of ABC transporters in melanogenesis and the development of multidrug resistance of melanoma. Pigment Cell Melanoma
Res 2009;22:7409.
[75] Matei D, Fang F, Shen C, Schilder J, Arnold A, Zeng Y, et al. Epigenetic resensitization to platinum in ovarian cancer. Cancer Res
2012;72:2197205.
[76] Wenzel JJ, Kaminski WE, Piehler A, Heimerl S, Langmann T, Schmitz
G. ABCA10, a novel cholesterol-regulated ABCA6-like ABC transporter. Biochem Biophys Res Commun 2003;306:108998.
[77] Zuo Y, Zhuang DZ, Han R, Isaac G, Tobin JJ, McKee M, et al.
ABCA12 maintains the epidermal lipid permeability barrier by
facilitating formation of ceramide linoleic esters. J Biol Chem
2008;283:3662435.
[78] Kartner N, Shales M, Riordan JR, Ling V. Daunorubicin-resistant
Chinese hamster ovary cells expressing multidrug resistance and a
cell-surface P-glycoprotein. Cancer Res 1983;43:44139.
[79] Higgins CF. ABC transporters: from microorganisms to man. Annu
Rev Cell Biol 1992;8:67113.
[80] Gottesman MM, Pastan I, Ambudkar SV. P-glycoprotein and multidrug resistance. Curr Opin Genet Dev 1996;6:6107.
[81] Fojo AT, Ueda K, Slamon DJ, Poplack DG, Gottesman MM, Pastan
I. Expression of a multidrug-resistance gene in human tumors and
tissues. Proc Natl Acad Sci U S A 1987;84:2659.
[82] Duan Z, Brakora KA, Seiden MV. Inhibition of ABCB1 (MDR1) and
ABCB4 (MDR3) expression by small interfering RNA and reversal
of paclitaxel resistance in human ovarian cancer cells. Mol Cancer
Ther 2004;3:8338.
[83] Yakirevich E, Sabo E, Naroditsky I, Sova Y, Lavie O, Resnick
MB. Multidrug resistance-related phenotype and apoptosis-related
protein expression in ovarian serous carcinomas. Gynecol Oncol
2006;100:1529.
[84] Veneroni S, Zaffaroni N, Daidone MG, Benini E, Villa R, Silvestrini
R. Expression of P-glycoprotein and in vitro or in vivo resistance to
doxorubicin and cisplatin in breast and ovarian cancers. Eur J Cancer
1994;30:10027.

[85] Bell DR, Gerlach JH, Kartner N, Buick RN, Ling V. Detection of
P-glycoprotein in ovarian cancer: a molecular marker associated with
multidrug resistance. J Clin Oncol 1985;3:3115.
[86] Rubin SC, Finstad CL, Hoskins WJ, Saigo PE, Provencher DM, Federici MG, et al. Expression of P-glycoprotein in epithelial ovarian
cancer: evaluation as a marker of multidrug resistance. Am J Obstet
Gynecol 1990;163:6973.
[87] van der Zee AG, Hollema H, de Jong S, Boonstra H, Gouw A,
Willemse PH, et al. P-glycoprotein expression and DNA topoisomerase I and II activity in benign tumors of the ovary and in malignant
tumors of the ovary, before and after platinum/cyclophosphamide
chemotherapy. Cancer Res 1991;51:591520.
[88] Izquierdo MA, van der Zee AG, Vermorken JB, van der Valk P, Belien
JA, Giaccone G, et al. Drug resistance-associated marker Lrp for
prediction of response to chemotherapy and prognoses in advanced
ovarian carcinoma. J Natl Cancer Inst 1995;87:12307.
[89] van der Zee AG, Hollema H, Suurmeijer AJ, Krans M, Sluiter WJ,
Willemse PH, et al. Value of P-glycoprotein, glutathione S-transferase
pi, c-erbB-2, and p53 as prognostic factors in ovarian carcinomas. J
Clin Oncol 1995;13:708.
[90] Yokoyama Y, Sato S, Fukushi Y, Sakamoto T, Futagami M, Saito Y.
Significance of multi-drug-resistant proteins in predicting chemotherapy response and prognosis in epithelial ovarian cancer. J Obstet
Gynaecol Res 1999;25:38794.
[91] Arts HJ, Katsaros D, de Vries EG, Massobrio M, Genta F, Danese S,
et al. Drug resistance-associated markers P-glycoprotein, multidrug
resistance-associated protein 1, multidrug resistance-associated protein 2, and lung resistance protein as prognostic factors in ovarian
carcinoma. Clin Cancer Res 1999;5:2798805.
[92] Tewari KS, Kyshtoobayeva AS, Mehta RS, Yu IR, Burger RA, DiSaia
PJ, et al. Biomarker conservation in primary and metastatic epithelial
ovarian cancer. Gynecol Oncol 2000;78:1306.
[93] Ozalp SS, Yalcin OT, Tanir M, Kabukcuoglu S, Etiz E. Multidrug
resistance gene-1 (Pgp) expression in epithelial ovarian malignancies.
Eur J Gynaecol Oncol 2002;23:33740.
[94] Itamochi H, Kigawa J, Sugiyama T, Kikuchi Y, Suzuki M, Terakawa
N. Low proliferation activity may be associated with chemoresistance in clear cell carcinoma of the ovary. Obstet Gynecol 2002;100:
2817.
[95] Ikeda K, Sakai K, Yamamoto R, Hareyama H, Tsumura N, Watari H,
et al. Multivariate analysis for prognostic significance of histologic
subtype, GST-pi, MDR-1, and p53 in stages II-IV ovarian cancer. Int
J Gynecol Cancer 2003;13:77684.
[96] Penson RT, Oliva E, Skates SJ, Glyptis T, Fuller Jr AF, Goodman A,
et al. Expression of multidrug resistance-1 protein inversely correlates
with paclitaxel response and survival in ovarian cancer patients: a
study in serial samples. Gynecol Oncol 2004;93:98106.
[97] Huang X, Ushijima K, Komai K, Takemoto Y, Motoshima S, Kamura
T, et al. Co-expression of Y box-binding protein-1 and P-glycoprotein
as a prognostic marker for survival in epithelial ovarian cancer.
Gynecol Oncol 2004;93:28791.
[98] Surowiak P, Materna V, Denkert C, Kaplenko I, Spaczynski M,
Dietel M, et al. Significance of cyclooxygenase 2 and MDR1/Pglycoprotein coexpression in ovarian cancers. Cancer Lett 2006;235:
27280.
[99] Kavallaris M, Leary JA, Barrett JA, Friedlander ML. MDR1 and
multidrug resistance-associated protein (MRP) gene expression in
epithelial ovarian tumors. Cancer Lett 1996;102:716.
[100] Kamazawa S, Kigawa J, Kanamori Y, Itamochi H, Sato S, Iba T,
et al. Multidrug resistance gene-1 is a useful predictor of Paclitaxelbased chemotherapy for patients with ovarian cancer. Gynecol Oncol
2002;86:1716.
[101] Raspollini MR, Amunni G, Villanucci A, Boddi V, Taddei
GL. Increased cyclooxygenase-2 (COX-2) and P-glycoprotein-170
(MDR1) expression is associated with chemotherapy resistance and
poor prognosis. Analysis in ovarian carcinoma patients with low and
high survival. Int J Gynecol Cancer 2005;15:25560.

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256


[102] Lu L, Katsaros D, Wiley A, Rigault de la Longrais IA, Puopolo M, Yu
H. Expression of MDR1 in epithelial ovarian cancer and its association
with disease progression. Oncol Res 2007;16:395403.
[103] Chen H, Hao J, Wang L, Li Y. Coexpression of invasive markers
(uPA, CD44) and multiple drug-resistance proteins (MDR1, MRP2)
is correlated with epithelial ovarian cancer progression. Br J Cancer
2009;101:43240.
[104] Lu D, Shi HC, Wang ZX, Gu XW, Zeng YJ. Multidrug
resistance-associated biomarkers PGP, GST-pi, Topo-II and LRP as
prognostic factors in primary ovarian carcinoma. Br J Biomed Sci
2011;68:6974.
[105] Johnatty SE, Beesley J, Gao B, Chen X, Lu Y, Law MH, et al.
ABCB1 (MDR1) polymorphisms and ovarian cancer progression and
survival: a comprehensive analysis from the Ovarian Cancer Association Consortium and The Cancer Genome Atlas. Gynecol Oncol
2013;131:814.
[106] Sedlakova I, Laco J, Caltova K, Cervinka M, Tosner J, Rezac A,
et al. Clinical significance of the resistance proteins LRP, Pgp, MRP1,
MRP3, and MRP5 in epithelial ovarian cancer. Int J Gynecol Cancer
2015;25:23643.
[107] Arao S, Suwa H, Mandai M, Tashiro H, Miyazaki K, Okamura
H, et al. Expression of multidrug resistance gene and localization
of P-glycoprotein in human primary ovarian cancer. Cancer Res
1994;54:13559.
[108] Rao VV, Anthony DC, Piwnica-Worms D. Multidrug resistance Pglycoprotein monoclonal antibody JSB-1 crossreacts with pyruvate
carboxylase. J Histochem Cytochem 1995;43:118792.
[109] Kiguchi K, Iwamori Y, Suzuki N, Kobayashi Y, Ishizuka B, Ishiwata
I, et al. Characteristic expression of globotriaosyl ceramide in human
ovarian carcinoma-derived cells with anticancer drug resistance. Cancer Sci 2006;97:13216.
[110] Ho EA, Soo PL, Allen C, Piquette-Miller M. Impact of intraperitoneal,
sustained delivery of paclitaxel on the expression of P-glycoprotein
in ovarian tumors. J Control Release 2007;117:207.
[111] Holzmayer TA, Hilsenbeck S, Von Hoff DD, Roninson IB. Clinical
correlates of MDR1 (P-glycoprotein) gene expression in ovarian and
small-cell lung carcinomas. J Natl Cancer Inst 1992;84:148691.
[112] Bourhis J, Goldstein LJ, Riou G, Pastan I, Gottesman MM, Benard J.
Expression of a human multidrug resistance gene in ovarian carcinomas. Cancer Res 1989;49:50625.
[113] Ricciardelli C, Ween MP, Lokman NA, Tan IA, Pyragius CE,
Oehler MK. Chemotherapy-induced hyaluronan production: a novel
chemoresistance mechanism in ovarian cancer. BMC Cancer
2013;13:476.
[114] Obata H, Yahata T, Quan J, Sekine M, Tanaka K. Association between
single nucleotide polymorphisms of drug resistance-associated genes
and response to chemotherapy in advanced ovarian cancer. Anticancer
Res 2006;26:222732.
[115] Kim HS, Kim MK, Chung HH, Kim JW, Park NH, Song YS,
et al. Genetic polymorphisms affecting clinical outcomes in epithelial
ovarian cancer patients treated with taxanes and platinum compounds: a Korean population-based study. Gynecol Oncol 2009;113:
2649.
[116] Green H, Soderkvist P, Rosenberg P, Mirghani RA, Rymark P,
Lundqvist EA, et al. Pharmacogenetic studies of Paclitaxel in
the treatment of ovarian cancer. Basic Clin Pharmacol Toxicol
2009;104:1307.
[117] Johnatty SE, Beesley J, Paul J, Fereday S, Spurdle AB, Webb PM,
et al. ABCB1 (MDR 1) polymorphisms and progression-free survival
among women with ovarian cancer following paclitaxel/carboplatin
chemotherapy. Clin Cancer Res 2008;14:5594601.
[118] Peethambaram P, Fridley BL, Vierkant RA, Larson MC, Kalli
KR, Elliott EA, et al. Polymorphisms in ABCB1 and ERCC2
associated with ovarian cancer outcome. Int J Mol Epidemiol Genet
2011;2:18595.
[119] Hamidovic A, Hahn K, Kolesar J. Clinical significance of ABCB1
genotyping in oncology. J Oncol Pharm Pract 2010;16:3944.

245

[120] Diaz-Padilla I, Amir E, Marsh S, Liu G, Mackay H. Genetic polymorphisms as predictive and prognostic biomarkers in gynecological
cancers: a systematic review. Gynecol Oncol 2012;124:35465.
[121] White KL, Vierkant RA, Fogarty ZC, Charbonneau B, Block
MS, Pharoah PD, et al. Analysis of over 10,000 Cases finds no
association between previously reported candidate polymorphisms
and ovarian cancer outcome. Cancer Epidemiol Biomarkers Prev
2013;22:98792.
[122] Yamaguchi H, Hishinuma T, Endo N, Tsukamoto H, Kishikawa Y,
Sato M, et al. Genetic variation in ABCB1 influences paclitaxel pharmacokinetics in Japanese patients with ovarian cancer. Int J Gynecol
Cancer 2006;16:97985.
[123] Fung KL, Gottesman MM. A synonymous polymorphism in a common MDR1 (ABCB1) haplotype shapes protein function. Biochim
Biophys Acta 2009;1794:86071.
[124] Bergmann TK, Brasch-Andersen C, Green H, Mirza MR, Skougaard
K, Wihl J, et al. Impact of ABCB1 Variants on neutrophil depression:
a pharmacogenomic study of paclitaxel in 92 women with ovarian
cancer. Basic Clin Pharmacol Toxicol 2011.
[125] Marsh S, Paul J, King CR, Gifford G, McLeod HL, Brown R. Pharmacogenetic assessment of toxicity and outcome after platinum plus
taxane chemotherapy in ovarian cancer: the Scottish Randomised
Trial in Ovarian Cancer. J Clin Oncol 2007;25:452835.
[126] Bergmann TK, Green H, Brasch-Andersen C, Mirza MR, Herrstedt J,
Holund B, et al. Retrospective study of the impact of pharmacogenetic
variants on paclitaxel toxicity and survival in patients with ovarian
cancer. Eur J Clin Pharmacol 2011;67:693700.
[127] Muller M, Agaimy A, Zenk J, Ettl T, Iro H, Hartmann A, et al. The
prognostic impact of human leukocyte antigen (HLA) class I antigen
abnormalities in salivary gland cancer. A clinicopathological study of
288 cases. Histopathology 2013;62:84759.
[128] Zeng Q, Wang XL, Xu ZG, Lu N, Zheng S, Zhao QZ, et al. [Association of antigen processing machinery and HLA class I defects with
clinicopathological outcome in laryngeal squamous cell carcinoma].
Zhonghua Er Bi Yan Hou Tou Jing Wai Ke Za Zhi 2012;47:23440.
[129] Lopez-Albaitero A, Nayak JV, Ogino T, Machandia A, Gooding W,
DeLeo AB, et al. Role of antigen-processing machinery in the in vitro
resistance of squamous cell carcinoma of the head and neck cells to
recognition by CTL. J Immunol 2006;176:34029.
[130] Korkolopoulou P, Kaklamanis L, Pezzella F, Harris AL, Gatter KC.
Loss of antigen-presenting molecules (MHC class I and TAP-1) in
lung cancer. Br J Cancer 1996;73:14853.
[131] Cromme FV, van Bommel PF, Walboomers JM, Gallee MP, Stern
PL, Kenemans P, et al. Differences in MHC and TAP-1 expression in
cervical cancer lymph node metastases as compared with the primary
tumours. Br J Cancer 1994;69:117681.
[132] Seliger B, Stoehr R, Handke D, Mueller A, Ferrone S, Wullich B,
et al. Association of HLA class I antigen abnormalities with disease
progression and early recurrence in prostate cancer. Cancer Immunol
Immunother 2010;59:52940.
[133] Tanaka K, Tsuchikawa T, Miyamoto M, Maki T, Ichinokawa M, Kubota KC, et al. Down-regulation of Human Leukocyte Antigen class I
heavy chain in tumors is associated with a poor prognosis in advanced
esophageal cancer patients. Int J Oncol 2012;40:96574.
[134] Kasajima A, Sers C, Sasano H, Johrens K, Stenzinger A, Noske A,
et al. Down-regulation of the antigen processing machinery is linked
to a loss of inflammatory response in colorectal cancer. Hum Pathol
2010;41:175869.
[135] Meissner M, Reichert TE, Kunkel M, Gooding W, Whiteside TL,
Ferrone S, et al. Defects in the human leukocyte antigen class I
antigen processing machinery in head and neck squamous cell carcinoma: association with clinical outcome. Clin Cancer Res 2005;11:
255260.
[136] Heimerl S, Bosserhoff AK, Langmann T, Ecker J, Schmitz G.
Mapping ATP-binding cassette transporter gene expression profiles in melanocytes and melanoma cells. Melanoma Res 2007;17:
26573.

246

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

[137] Kaklamanis L, Leek R, Koukourakis M, Gatter KC, Harris AL. Loss of


transporter in antigen processing 1 transport protein and major histocompatibility complex class I molecules in metastatic versus primary
breast cancer. Cancer Res 1995;55:51914.
[138] Bandoh N, Ogino T, Katayama A, Takahara M, Katada A, Hayashi
T, et al. HLA class I antigen and transporter associated with antigen
processing downregulation in metastatic lesions of head and neck
squamous cell carcinoma as a marker of poor prognosis. Oncol Rep
2010;23:9339.
[139] Liu Y, Komohara Y, Domenick N, Ohno M, Ikeura M, Hamilton
RL, et al. Expression of antigen processing and presenting molecules
in brain metastasis of breast cancer. Cancer Immunol Immunother
2012;61:789801.
[140] Ge HL, Zhang XR, Wang Y, Ma AL, Zhang HZ, Wang SJ, et al.
[Study of HLA molecules and its associated genes expression in
human ovarian cancer cells]. Xi Bao Yu Fen Zi Mian Yi Xue Za
Zhi 2003;19:414.
[141] Han LY, Fletcher MS, Urbauer DL, Mueller P, Landen CN, Kamat
AA, et al. HLA class I antigen processing machinery component
expression and intratumoral T-Cell infiltrate as independent prognostic markers in ovarian carcinoma. Clin Cancer Res 2008;14:
33729.
[142] Leffers N, Gooden MJ, Mokhova AA, Kast WM, Boezen HM, Ten
Hoor KA, et al. Down-regulation of proteasomal subunit MB1 is
an independent predictor of improved survival in ovarian cancer.
Gynecol Oncol 2009;113:25663.
[143] Vitale M, Pelusi G, Taroni B, Gobbi G, Micheloni C, Rezzani R,
et al. HLA class I antigen down-regulation in primary ovary carcinoma lesions: association with disease stage. Clin Cancer Res
2005;11:6772.
[144] Xu M, Li L, Liu Z, Jiao Z, Xu P, Kong X, et al. ABCB2 (TAP1) as
the downstream target of SHH signaling enhances pancreatic ductal
adenocarcinoma drug resistance. Cancer Lett 2013;333:1528.
[145] Auner V, Sehouli J, Oskay-Oezcelik G, Horvat R, Speiser P, Zeillinger
R. ABC transporter gene expression in benign and malignant ovarian
tissue. Gynecol Oncol 2010;117:198201.
[146] Morita SY, Terada T. Molecular mechanisms for biliary phospholipid
and drug efflux mediated by ABCB4 and bile salts. BioMed Res Int
2014;2014:954781.
[147] Citti A, Boldrini R, Inserra A, Alisi A, Pessolano R, Mastronuzzi
A, et al. Expression of multidrug resistance-associated proteins in
paediatric soft tissue sarcomas before and after chemotherapy. Int J
Oncol 2012;41:11724.
[148] Arai Y, Masuda M, Sugawara I, Arai T, Motoji T, Tsuruo T, et al.
Expression of the MDR1 and MDR3 gene products in acute and
chronic leukemias. Leuk Res 1997;21:3139.
[149] Kiehl S, Herkt SC, Richter AM, Fuhrmann L, El-Nikhely N, Seeger W,
et al. ABCB4 is frequently epigenetically silenced in human cancers
and inhibits tumor growth. Sci Rep 2014;4:6899.
[150] Frank NY, Pendse SS, Lapchak PH, Margaryan A, Shlain D, Doeing C,
et al. Regulation of progenitor cell fusion by ABCB5 P-glycoprotein,
a novel human ATP-binding cassette transporter. J Biol Chem
2003;278:4715665.
[151] Volpicelli ER, Lezcano C, Zhan Q, Girouard SD, Kindelberger
DW, Frank MH, et al. The multidrug-resistance transporter ABCB5
is expressed in human placenta. Int J Gynecol Pathol 2014;33:
4551.
[152] Grimm M, Krimmel M, Polligkeit J, Alexander D, Munz A, Kluba
S, et al. ABCB5 expression and cancer stem cell hypothesis in oral
squamous cell carcinoma. Eur J Cancer 2012;48:318697.
[153] Chartrain M, Riond J, Stennevin A, Vandenberghe I, Gomes B,
Lamant L, et al. Melanoma chemotherapy leads to the selection of
ABCB5-expressing cells. PLoS ONE 2012;7:e36762.
[154] Cheung ST, Cheung PF, Cheng CK, Wong NC, Fan ST.
Granulin-epithelin precursor and ATP-dependent binding cassette
(ABC)B5 regulate liver cancer cell chemoresistance. Gastroenterology 2011;140:34455.

[155] Yang JY, Ha SA, Yang YS, Kim JW. p-Glycoprotein ABCB5 and YB1 expression plays a role in increased heterogeneity of breast cancer
cells: correlations with cell fusion and doxorubicin resistance. BMC
Cancer 2010;10:388.
[156] Wilson BJ, Schatton T, Zhan Q, Gasser M, Ma J, Saab KR, et al.
ABCB5 identifies a therapy-refractory tumor cell population in colorectal cancer patients. Cancer Res 2011;71:530716.
[157] Kugimiya N, Nishimoto A, Hosoyama T, Ueno K, Enoki T, Li TS,
et al. The c-MYC-ABCB5 axis plays a pivotal role in 5-fluorouracil
resistance in human colon cancer cells. J Cell Mol Med 2015.
[158] Krishnamurthy PC, Du G, Fukuda Y, Sun D, Sampath J, Mercer
KE, et al. Identification of a mammalian mitochondrial porphyrin
transporter. Nature 2006;443:5869.
[159] Zhao SG, Chen XF, Wang LG, Yang G, Han DY, Teng L, et al.
Increased expression of ABCB6 enhances protoporphyrin IX accumulation and photodynamic effect in human glioma. Ann Surg Oncol
2013;20:437988.
[160] Duan Z, Lamendola DE, Duan Y, Yusuf RZ, Seiden MV. Description
of paclitaxel resistance-associated genes in ovarian and breast cancer
cell lines. Cancer Chemother Pharmacol 2005;55:27785.
[161] Hendig D, Langmann T, Zarbock R, Schmitz G, Kleesiek K, Gotting C. Characterization of the ATP-binding cassette transporter
gene expression profile in Y79: a retinoblastoma cell line. Mol Cell
Biochem 2009;328:8592.
[162] Young LC [Thesis] The contribution of members of the ATP binding cassette superfamily to the multidrug resistance of lung cancer.
Queens University; 2000.
[163] Ichikawa Y, Bayeva M, Ghanefar M, Potini V, Sun L, Mutharasan
RK, et al. Disruption of ATP-binding cassette B8 in mice leads to
cardiomyopathy through a decrease in mitochondrial iron export. Proc
Natl Acad Sci U S A 2012;109:41527.
[164] Yasui K, Mihara S, Zhao C, Okamoto H, Saito-Ohara F, Tomida A,
et al. Alteration in copy numbers of genes as a mechanism for acquired
drug resistance. Cancer Res 2004;64:140310.
[165] Elliott AM, Al-Hajj MA. ABCB8 mediates doxorubicin resistance in
melanoma cells by protecting the mitochondrial genome. Mol Cancer
Res 2009;7:7987.
[166] Demirel O, Bangert I, Tampe R, Abele R. Tuning the cellular trafficking of the lysosomal peptide transporter TAPL by its N-terminal
domain. Traffic 2010;11:38393.
[167] Dong Z, Zhong Z, Yang L, Wang S, Gong Z. MicroRNA-31 inhibits
cisplatin-induced apoptosis in non-small cell lung cancer cells by regulating the drug transporter ABCB9. Cancer Lett 2014;343:24957.
[168] Bhatnagar N, Li X, Padi SK, Zhang Q, Tang MS, Guo B.
Downregulation of miR-205 and miR-31 confers resistance to
chemotherapy-induced apoptosis in prostate cancer cells. Cell Death
Dis 2010;1:e105.
[169] Wang CJ, Stratmann J, Zhou ZG, Sun XF. Suppression of microRNA31 increases sensitivity to 5-FU at an early stage, and affects cell
migration and invasion in HCT-116 colon cancer cells. BMC Cancer
2010;10:616.
[170] Yamamoto M, Arimura H, Fukushige T, Minami K, Nishizawa Y,
Tanimoto A, et al. Abcb10 role in heme biosynthesis in vivo: Abcb10
knockout in mice causes anemia with protoporphyrin IX and iron
accumulation. Mol Cell Biol 2014;34:107784.
[171] Konkimalla VB, Kaina B, Efferth T. Role of transporter genes in
cisplatin resistance. In Vivo 2008;22:27983.
[172] Oiso S, Takayama Y, Nakazaki R, Matsunaga N, Motooka C,
Yamamura A, et al. Factors involved in the cisplatin resistance
of KCP4 human epidermoid carcinoma cells. Oncol Rep 2014;31:
71926.
[173] Gillet JP, Wang J, Calcagno AM, Green LJ, Varma S, Bunkholt
Elstrand M, et al. Clinical relevance of multidrug resistance gene
expression in ovarian serous carcinoma effusions. Mol Pharm
2011;8:20808.
[174] Strautnieks SS, Byrne JA, Pawlikowska L, Cebecauerova D, Rayner
A, Dutton L, et al. Severe bile salt export pump deficiency: 82

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

[175]

[176]
[177]

[178]

[179]
[180]

[181]

[182]

[183]

[184]

[185]

[186]

[187]

[188]

[189]

[190]

[191]
[192]

[193]

[194]

different ABCB11 mutations in 109 families. Gastroenterology


2008;134:120314.
Childs S, Yeh RL, Hui D, Ling V. Taxol resistance mediated by transfection of the liver-specific sister gene of P-glycoprotein. Cancer Res
1998;58:41607.
Kruh GD, Belinsky MG. The MRP family of drug efflux pumps.
Oncogene 2003;22:753752.
Bakos E, Evers R, Szakacs G, Tusnady GE, Welker E, Szabo K,
et al. Functional multidrug resistance protein (MRP1) lacking the Nterminal transmembrane domain. J Biol Chem 1998;273:3216775.
Cole SP, Deeley RG. Multidrug resistance mediated by the
ATP-binding cassette transporter protein MRP. BioEssays
1998;20:93140.
Eckford PD, Sharom FJ. ABC efflux pump-based resistance to
chemotherapy drugs. Chem Rev 2009;109:29893011.
Borst P, Evers R, Kool M, Wijnholds J. A family of drug transporters: the multidrug resistance-associated proteins. J Natl Cancer
Inst 2000;92:1295302.
Cole SP, Bhardwaj G, Gerlach JH, Mackie JE, Grant CE, Almquist
KC, et al. Overexpression of a transporter gene in a multidrug-resistant
human lung cancer cell line. Science 1992;258:16504.
Cole SP, Sparks KE, Fraser K, Loe DW, Grant CE, Wilson GM,
et al. Pharmacological characterization of multidrug resistant MRPtransfected human tumor cells. Cancer Res 1994;54:590210.
Grant CE, Valdimarsson G, Hipfner DR, Almquist KC, Cole SP,
Deeley RG. Overexpression of multidrug resistance-associated protein (MRP) increases resistance to natural product drugs. Cancer Res
1994;54:35761.
Zaman GJ, Flens MJ, van Leusden MR, de Haas M, Mulder HS,
Lankelma J, et al. The human multidrug resistance-associated protein
MRP is a plasma membrane drug-efflux pump. Proc Natl Acad Sci U
S A 1994;91:88226.
Rahgozar S, Moafi A, Abedi M, Entezar EGM, Moshtaghian J, Ghaedi
K, et al. mRNA expression profile of multidrug resistant genes in acute
lymphoblastic leukemia of children, a prognostic value for ABCA3
and ABCA2. Cancer Biol Ther 2013:15.
Du SS, Yao KT. [Expression of ATP-binding cassette transporter
genes in nasopharyngeal carcinoma]. Nan Fang Yi Ke Da Xue Xue
Bao 2008;28:44952.
Faggad A, Darb-Esfahani S, Wirtz R, Sinn B, Sehouli J, Konsgen D,
et al. Expression of multidrug resistance-associated protein 1 in invasive ovarian carcinoma: implication for prognosis. Histopathology
2009;54:65766.
Bagnoli M, Beretta GL, Gatti L, Pilotti S, Alberti P, Tarantino E, et al.
Clinicopathological impact of ABCC1/MRP1 and ABCC4/MRP4 in
epithelial ovarian carcinoma. BioMed Res Int 2013;2013:143202.
Kigawa J, Minagawa Y, Cheng X, Terakawa N. Gammaglutamyl cysteine synthetase up-regulates glutathione and multidrug
resistance-associated protein in patients with chemoresistant epithelial ovarian cancer. Clin Cancer Res 1998;4:173741.
Ohishi Y, Oda Y, Uchiumi T, Kobayashi H, Hirakawa T, Miyamoto S,
et al. ATP-binding cassette superfamily transporter gene expression in
human primary ovarian carcinoma. Clin Cancer Res 2002;8:376775.
Borst P, Evers R, Kool M, Wijnholds J. The multidrug resistance
protein family. Biochim Biophys Acta 1999;1461:34757.
Borst P, Kool M, Evers R. Do cMOAT (MRP2), other MRP
homologues, and LRP play a role in MDR? Semin Cancer Biol
1997;8:20513.
Smitherman PK, Townsend AJ, Kute TE, Morrow CS. Role of multidrug resistance protein 2 (MRP2, ABCC2) in alkylating agent
detoxification: MRP2 potentiates glutathione S-transferase A1-1mediated resistance to chlorambucil cytotoxicity. J Pharmacol Exp
Ther 2004;308:2607.
Sandusky GE, Mintze KS, Pratt SE, Dantzig AH. Expression of
multidrug resistance-associated protein 2 (MRP2) in normal human
tissues and carcinomas using tissue microarrays. Histopathology
2002;41:6574.

247

[195] Guminski AD, Balleine RL, Chiew YE, Webster LR, Tapner M, Farrell GC, et al. MRP2 (ABCC2) and cisplatin sensitivity in hepatocytes
and human ovarian carcinoma. Gynecol Oncol 2006;100:23946.
[196] Surowiak P, Materna V, Kaplenko I, Spaczynski M, DolinskaKrajewska B, Gebarowska E, et al. ABCC2 (MRP2, cMOAT) can
be localized in the nuclear membrane of ovarian carcinomas and correlates with resistance to cisplatin and clinical outcome. Clin Cancer
Res 2006;12:714958.
[197] Kim H, Kim K, No JH, Jeon YT, Jeon HW, Kim YB. Prognostic value
of biomarkers related to drug resistance in patients with advanced
epithelial ovarian cancer. Anticancer Res 2012;32:58994.
[198] Tian C, Ambrosone CB, Darcy KM, Krivak TC, Armstrong
DK, Bookman MA, et al. Common variants in ABCB1, ABCC2
and ABCG2 genes and clinical outcomes among women with
advanced stage ovarian cancer treated with platinum and taxane-based
chemotherapy: a Gynecologic Oncology Group study. Gynecol Oncol
2012;124:57581.
[199] Zhao Y, Lu H, Yan A, Yang Y, Meng Q, Sun L, et al. ABCC3 as a
marker for multidrug resistance in non-small cell lung cancer. Sci Rep
2013;3:3120.
[200] Partanen L, Staaf J, Tanner M, Tuominen VJ, Borg A, Isola J. Amplification and overexpression of the ABCC3 (MRP3) gene in primary
breast cancer. Genes Chromosomes Cancer 2012;51:83240.
[201] Narayan G, Bourdon V, Chaganti S, Arias-Pulido H, Nandula SV,
Rao PH, et al. Gene dosage alterations revealed by cDNA microarray
analysis in cervical cancer: identification of candidate amplified and
overexpressed genes. Genes Chromosomes Cancer 2007;46:37384.
[202] Sedlakova I, Laco J, Tosner J, Caltova K, Cervinka M, Rezac A,
et al. [Proteins of resistance and drug resistance in ovarian carcinoma
patients]. Klin Onkol 2012;25:45763.
[203] Russel FG, Koenderink JB, Masereeuw R. Multidrug resistance protein 4 (MRP4/ABCC4): a versatile efflux transporter for drugs and
signalling molecules. Trends Pharmacol Sci 2008;29:2007.
[204] Zhao X, Guo Y, Yue W, Zhang L, Gu M, Wang Y. ABCC4 is required
for cell proliferation and tumorigenesis in non-small cell lung cancer.
Onco Targets Ther 2014;7:34351.
[205] Sun Y, Shi N, Lu H, Zhang J, Ma Y, Qiao Y, et al. ABCC4 copy number
variation is associated with susceptibility to esophageal squamous cell
carcinoma. Carcinogenesis 2014.
[206] Ho LL, Kench JG, Handelsman DJ, Scheffer GL, Stricker PD, Grygiel
JG, et al. Androgen regulation of multidrug resistance-associated protein 4 (MRP4/ABCC4) in prostate cancer. Prostate 2008;68:14219.
[207] Zhang Z, Wang J, Shen B, Peng C, Zheng M. The ABCC4 gene
is a promising target for pancreatic cancer therapy. Gene 2012;491:
1949.
[208] Chen L, Gu J, Xu L, Qu C, Zhang Y, Zhang W. RNAi-mediated
silencing of ATP-binding cassette C4 protein inhibits cell growth in
MGC80-3 gastric cancer cell lines. Cell Mol Biol 2014;60:15.
[209] Beretta GL, Benedetti V, Cossa G, Assaraf YG, Bram E, Gatti
L, et al. Increased levels and defective glycosylation of MRPs in
ovarian carcinoma cells resistant to oxaliplatin. Biochem Pharmacol
2010;79:110817.
[210] Sedlakova I, Laco J, Tosner J, Spacek J, Cermakova E. [Drug resistance proteins LRP, Pgp, MRP1, MRP3 and MRP5 in ovarian cancer
patients]. Ceska Gynekol 2013;78:54553.
[211] Kool M, van der Linden M, de Haas M, Baas F, Borst P. Expression of
human MRP6, a homologue of the multidrug resistance protein gene
MRP1, in tissues and cancer cells. Cancer Res 1999;59:17582.
[212] Yu J, Zhu T, Wang Z, Zhang H, Qian Z, Xu H, et al. A novel set of
DNA methylation markers in urine sediments for sensitive/specific
detection of bladder cancer. Clin Cancer Res 2007;13:7296304.
[213] Wamunyokoli FW, Bonome T, Lee JY, Feltmate CM, Welch WR,
Radonovich M, et al. Expression profiling of mucinous tumors of the
ovary identifies genes of clinicopathologic importance. Clin Cancer
Res 2006;12:690700.
[214] Wu Z, Peng X, Li J, Zhang Y, Hu L. Constitutive activation of
nuclear factor kappaB contributes to cystic fibrosis transmembrane

248

[215]

[216]

[217]

[218]

[219]

[220]
[221]

[222]

[223]

[224]

[225]
[226]

[227]
[228]

[229]

[230]

[231]

[232]

[233]
[234]

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256


conductance regulator expression and promotes human cervical
cancer progression and poor prognosis. Int J Gynecol Cancer
2013;23:90615.
McWilliams RR, Petersen GM, Rabe KG, Holtegaard LM, Lynch PJ,
Bishop MD, et al. Cystic fibrosis transmembrane conductance regulator (CFTR) gene mutations and risk for pancreatic adenocarcinoma.
Cancer 2010;116:2039.
Li J, Zhang JT, Jiang X, Shi X, Shen J, Feng F, et al. The cystic fibrosis
transmembrane conductance regulator as a biomarker in non-small
cell lung cancer. Int J Oncol 2015;46:210715.
Zhang JT, Jiang XH, Xie C, Cheng H, Da Dong J, Wang Y, et al.
Downregulation of CFTR promotes epithelial-to-mesenchymal transition and is associated with poor prognosis of breast cancer. Biochim
Biophys Acta 2013;1833:29619.
Xie C, Jiang XH, Zhang JT, Sun TT, Dong JD, Sanders AJ, et al.
CFTR suppresses tumor progression through miR-193b targeting
urokinase plasminogen activator (uPA) in prostate cancer. Oncogene
2013;32:e17, 2282-91, 91.
Xu J, Yong M, Li J, Dong X, Yu T, Fu X, et al. High level of CFTR
expression is associated with tumor aggression and knockdown of
CFTR suppresses proliferation of ovarian cancer in vitro and in vivo.
Oncol Rep 2015;33:222734.
Nichols CG. KATP channels as molecular sensors of cellular
metabolism. Nature 2006;440:4706.
Vazquez-Ortiz G, Garcia JA, Ciudad CJ, Noe V, Penuelas S, LopezRomero R, et al. Differentially expressed genes between high-risk
human papillomavirus types in human cervical cancer cells. Int J
Gynecol Cancer 2007;17:48491.
Thompson EM, Pishko GL, Muldoon LL, Neuwelt EA. Inhibition
of SUR1 decreases the vascular permeability of cerebral metastases.
Neoplasia 2013;15:53543.
Allebrandt KV, Amin N, Muller-Myhsok B, Esko T, Teder-Laving M,
Azevedo RV, et al. A K(ATP) channel gene effect on sleep duration:
from genome-wide association studies to function in Drosophila. Mol
Psychiatry 2013;18:12232.
De Souza R, Zahedi P, Badame RM, Allen C, Piquette-Miller M.
Chemotherapy dosing schedule influences drug resistance development in ovarian cancer. Mol Cancer Ther 2011;10:128999.
Kruh GD, Guo Y, Hopper-Borge E, Belinsky MG, Chen ZS. ABCC10,
ABCC11, and ABCC12. Pflug Arch 2007;453:67584.
Bera TK, Iavarone C, Kumar V, Lee S, Lee B, Pastan I. MRP9,
an unusual truncated member of the ABC transporter superfamily,
is highly expressed in breast cancer. Proc Natl Acad Sci U S A
2002;99:69977002.
Morita M, Imanaka T. Peroxisomal ABC transporters: structure, function and role in disease. Biochim Biophys Acta 2012;1822:138796.
Hillebrand M, Verrier SE, Ohlenbusch A, Schafer A, Soling HD,
Wouters FS, et al. Live cell FRET microscopy: homo- and heterodimerization of two human peroxisomal ABC transporters,
the adrenoleukodystrophy protein (ALDP, ABCD1) and PMP70
(ABCD3). J Biol Chem 2007;282:269977005.
Hour TC, Kuo YZ, Liu GY, Kang WY, Huang CY, Tsai YC, et al.
Downregulation of ABCD1 in human renal cell carcinoma. Int J Biol
Markers 2009;24:1718.
Fourcade S, Ruiz M, Guilera C, Hahnen E, Brichta L, Naudi A, et al.
Valproic acid induces antioxidant effects in X-linked adrenoleukodystrophy. Hum Mol Genet 2010;19:200514.
Reams RR, Kalari KR, Wang H, Odedina FT, Soliman KF, Yates
C. Detecting genegene interactions in prostate disease in African
American men. Infect Agents Cancer 2011;6(Suppl. 2):S1.
Reams RR, Jones Triche J, Hwang H, Soliman KF, Yates CC. ABCD3
gene important in prostate cancer APS experimental biology. Boston,
USA: FASEB; 2013.
Tran QN. A novel method for finding non-small cell lung cancer
diagnosis biomarkers. BMC Med Genomics 2013;6(Suppl. 1):S11.
Kashiwayama Y, Seki M, Yasui A, Murasaki Y, Morita M, Yamashita
Y, et al. 70-kDa peroxisomal membrane protein related protein

[235]

[236]
[237]

[238]

[239]
[240]

[241]

[242]

[243]

[244]

[245]

[246]

[247]

[248]

[249]

[250]

[251]

[252]

(P70R/ABCD4) localizes to endoplasmic reticulum not peroxisomes,


and NH2-terminal hydrophobic property determines the subcellular localization of ABC subfamily D proteins. Exp Cell Res
2009;315:190205.
Coelho D, Kim JC, Miousse IR, Fung S, du Moulin M, Buers I,
et al. Mutations in ABCD4 cause a new inborn error of vitamin B12
metabolism. Nat Genet 2012;44:11525.
Ren Y, Li Y, Tian D. Role of the ABCE1 gene in human lung adenocarcinoma. Oncol Rep 2012;27:96570.
Chen ZQ, Dong J, Ishimura A, Daar I, Hinnebusch AG, Dean M.
The essential vertebrate ABCE1 protein interacts with eukaryotic
initiation factors. J Biol Chem 2006;281:74527.
Li XR, Yang LZ, Huo XQ, Wang Y, Yang QH, Zhang QQ. Effects of
silencing the ATP-binding cassette protein E1 gene by electroporation
on the proliferation and migration of EC109 human esophageal cancer
cells. Mol Med Rep 2015.
Silverman RH. Implications for RNase L in prostate cancer biology.
Biochemistry 2003;42:180512.
Huang B, Gao Y, Tian D, Zheng M. A small interfering ABCE1targeting RNA inhibits the proliferation and invasiveness of small
cell lung cancer. Int J Mol Med 2010;25:68793.
Camps J, Nguyen QT, Padilla-Nash HM, Knutsen T, McNeil NE,
Wangsa D, et al. Integrative genomics reveals mechanisms of copy
number alterations responsible for transcriptional deregulation in
colorectal cancer. Genes Chromosomes Cancer 2009;48:100217.
Cheung ST, Cheung PF, Cheng CK, Fan ST. Genomics approach to
identify drug transporter ABCF1 associated with liver cancer recurrence and chemo-resistance. In: Proceedings of the 103rd annual
meeting of the American Association for Cancer Research (AACR).
2012.
He M, Mangiameli DP, Kachala S, Hunter K, Gillespie J, Bian X,
et al. Expression signature developed from a complex series of mouse
models accurately predicts human breast cancer survival. Clin Cancer
Res 2010;16:24959.
Ogawa Y, Tsuda H, Hai E, Tsuji N, Yamagata S, Tokunaga S, et al.
Clinical role of ABCF2 expression in breast cancer. Anticancer Res
2006;26:180914.
Nishimura S, Tsuda H, Miyagi Y, Hirasawa A, Suzuki A, Kataoka F,
et al. Can ABCF2 protein expression predict the prognosis of uterine
cancer? Br J Cancer 2008;99:16515.
LEsperance S, Popa I, Bachvarova M, Plante M, Patten N, Wu L,
et al. Gene expression profiling of paired ovarian tumors obtained
prior to and following adjuvant chemotherapy: molecular signatures
of chemoresistant tumors. Int J Oncol 2006;29:524.
Tsuda H, Ito YM, Ohashi Y, Wong KK, Hashiguchi Y, Welch WR,
et al. Identification of overexpression and amplification of ABCF2 in
clear cell ovarian adenocarcinomas by cDNA microarray analyses.
Clin Cancer Res 2005;11:68808.
Behbakht K, Randall TC, Benjamin I, Morgan MA, King S, Rubin SC.
Clinical characteristics of clear cell carcinoma of the ovary. Gynecol
Oncol 1998;70:2558.
Goff BA, Sainz de la Cuesta R, Muntz HG, Fleischhacker D, Ek
M, Rice LW, et al. Clear cell carcinoma of the ovary: a distinct
histologic type with poor prognosis and resistance to platinumbased chemotherapy in stage III disease. Gynecol Oncol 1996;60:
4127.
Tsuda H, Ito K, Yaegashi N, Hirasawa A, Sudo T, Kita T, et al. Relationship between ABCF2 expression and response to chemotherapy
or prognosis in clear cell adenocarcinoma of the ovary. Int J Gynecol
Cancer 2010;20:7947.
Choi YW, Bae SM, Kim YW, Lee HN, Park TC, Ro DY, et al. Gene
expression profiles in squamous cell cervical carcinoma using arraybased comparative genomic hybridization analysis. Int J Gynecol
Cancer 2007;17:68796.
Trasino SE, Kim YS, Wang TT. Ligand, receptor, and cell typedependent regulation of ABCA1 and ABCG1 mRNA in prostate
cancer epithelial cells. Mol Cancer Ther 2009;8:193445.

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256


[253] Jonker JW, Merino G, Musters S, van Herwaarden AE, Bolscher
E, Wagenaar E, et al. The breast cancer resistance protein BCRP
(ABCG2) concentrates drugs and carcinogenic xenotoxins into milk.
Nat Med 2005;11:1279.
[254] Young AM, Allen CE, Audus KL. Efflux transporters of the human
placenta. Adv Drug Deliv Rev 2003;55:12532.
[255] Woodward OM, Kottgen A, Coresh J, Boerwinkle E, Guggino WB,
Kottgen M. Identification of a urate transporter, ABCG2, with a common functional polymorphism causing gout. Proc Natl Acad Sci U S
A 2009;106:1033842.
[256] Robey RW, Medina-Perez WY, Nishiyama K, Lahusen T, Miyake K,
Litman T, et al. Overexpression of the ATP-binding cassette halftransporter, ABCG2 (Mxr/BCrp/ABCP1), in flavopiridol-resistant
human breast cancer cells. Clin Cancer Res 2001;7:14552.
[257] Li W, Tamamura R, Wang B, Liu Q, Liu H, Liu T, et al. Expressions of ABCG2, CD133, and podoplanin in salivary adenoid cystic
carcinoma. BioMed Res Int 2014;2014:132349.
[258] Liu L, Zuo LF, Guo JW. ABCG2 gene amplification and expression
in esophageal cancer cells with acquired adriamycin resistance. Mol
Med Rep 2014;9:1299304.
[259] Yanamoto S, Yamada S, Takahashi H, Naruse T, Matsushita Y, Ikeda
H, et al. Expression of the cancer stem cell markers CD44v6 and
ABCG2 in tongue cancer: effect of neoadjuvant chemotherapy on
local recurrence. Int J Oncol 2014;44:115362.
[260] Zhang S, Balch C, Chan MW, Lai HC, Matei D, Schilder JM, et al.
Identification and characterization of ovarian cancer-initiating cells
from primary human tumors. Cancer Res 2008;68:431120.
[261] He QZ, Luo XZ, Wang K, Zhou Q, Ao H, Yang Y, et al. Isolation and
characterization of cancer stem cells from high-grade serous ovarian
carcinomas. Cell Physiol Biochem 2014;33:17384.
[262] Sparreboom A, Loos WJ, Burger H, Sissung TM, Verweij J, Figg
WD, et al. Effect of ABCG2 genotype on the oral bioavailability of
topotecan. Cancer Biol Ther 2005;4:6508.
[263] Rudin CM, Liu W, Desai A, Karrison T, Jiang X, Janisch L, et al. Pharmacogenomic and pharmacokinetic determinants of erlotinib toxicity.
J Clin Oncol 2008;26:111927.
[264] Maliepaard M, van Gastelen MA, de Jong LA, Pluim D, van Waardenburg RC, Ruevekamp-Helmers MC, et al. Overexpression of the
BCRP/MXR/ABCP gene in a topotecan-selected ovarian tumor cell
line. Cancer Res 1999;59:455963.
[265] Jia P, Wu S, Li F, Xu Q, Wu M, Chen G, et al. Breast cancer resistance
protein-mediated topotecan resistance in ovarian cancer cells. Int J
Gynecol Cancer 2005;15:10428.
[266] Ee PL, Kamalakaran S, Tonetti D, He X, Ross DD, Beck WT. Identification of a novel estrogen response element in the breast cancer
resistance protein (ABCG2) gene. Cancer Res 2004;64:124751.
[267] Bojanic DD, Tarr PT, Gale GD, Smith DJ, Bok D, Chen B, et al.
Differential expression and function of ABCG1 and ABCG4 during
development and aging. J Lipid Res 2010;51:16981.
[268] Zhang Z, Li X, Wang X, Zhou Y, Xu H, Wang J, et al. [The expression
of ABCG4, V-ATPase and clinic significance of their correlation with
NSCLC.]. Zhongguo Fei Ai Za Zhi 2008;11:6915.
[269] Graf GA, Yu L, Li WP, Gerard R, Tuma PL, Cohen JC, et al. ABCG5
and ABCG8 are obligate heterodimers for protein trafficking and
biliary cholesterol excretion. J Biol Chem 2003;278:4827582.
[270] Berge KE, Tian H, Graf GA, Yu L, Grishin NV, Schultz J, et al. Accumulation of dietary cholesterol in sitosterolemia caused by mutations
in adjacent ABC transporters. Science 2000;290:17715.
[271] Aquea G, Bresky G, Lancellotti D, Madariaga JA, Zaffiri V, Urzua
U, et al. Increased expression of P2RY2, CD248 and EphB1 in
gastric cancers from Chilean patients. Asian Pac J Cancer Prev
2014;15:19316.
[272] Hostettler L, Zlobec I, Terracciano L, Lugli A. ABCG5-positivity
in tumor buds is an indicator of poor prognosis in node-negative
colorectal cancer patients. World J Gastroenterol 2010;16:7329.
[273] Yu CC, Chen YW, Chiou GY, Tsai LL, Huang PI, Chang CY, et al.
MicroRNA let-7a represses chemoresistance and tumourigenicity in

[274]

[275]

[276]

[277]

[278]

[279]

[280]

[281]

[282]

[283]

[284]

[285]

[286]

[287]

[288]

[289]

[290]

[291]

[292]

249

head and neck cancer via stem-like properties ablation. Oral Oncol
2011;47:20210.
Mazzoni A, Trave F. Cytoplasmic membrane cholesterol and doxorubicin cytotoxicity in drug-sensitive and multidrug-resistant human
ovarian cancer cells. Oncol Res 1993;5:7582.
Hu L, McArthur C, Jaffe RB. Ovarian cancer stem-like sidepopulation cells are tumourigenic and chemoresistant. Br J Cancer
2010;102:127683.
Rizzo S, Hersey JM, Mellor P, Dai W, Santos-Silva A, Liber D,
et al. Ovarian cancer stem cell-like side populations are enriched
following chemotherapy and overexpress EZH2. Mol Cancer Ther
2011;10:32535.
Zhang QH, Dou HT, Xu P, Zhuang SC, Liu PS. Tumor recurrence and
drug resistance properties of side population cells in high grade ovary
cancer. Drug Res 2015;65:1537.
Dou J, Jiang C, Wang J, Zhang X, Zhao F, Hu W, et al. Using ABCG2molecule-expressing side population cells to identify cancer stem-like
cells in a human ovarian cell line. Cell Biol Int 2011;35:22734.
Ricci F, Bernasconi S, Perego P, Ganzinelli M, Russo G, Bono F,
et al. Ovarian carcinoma tumor-initiating cells have a mesenchymal
phenotype. Cell Cycle 2012;11:196676.
Fong MY, Kakar SS. The role of cancer stem cells and the side
population in epithelial ovarian cancer. Histol Histopathol 2010;25:
11320.
Chen J, Wang J, Chen D, Yang J, Yang C, Zhang Y, et al. Evaluation
of characteristics of CD44 + CD117+ ovarian cancer stem cells in
three dimensional basement membrane extract scaffold versus two
dimensional monocultures. BMC Cell Biol 2013;14:7.
Bourguignon LYW, Peyrollier K, Xia W, Gilad E. Hyaluronan-CD44
interaction activates stem cell marker Nanog, Stat-3-mediated MDR1
gene expression, and ankyrin-regulated multidrug efflux in breast and
ovarian tumor cells. J Biol Chem 2008;283:1763551.
Chau WK, Ip CK, Mak AS, Lai HC, Wong AS. c-Kit mediates
chemoresistance and tumor-initiating capacity of ovarian cancer
cells through activation of Wnt/beta-catenin-ATP-binding cassette G2
signaling. Oncogene 2013;32:276781.
Norwood K, Wang RY, Hirschmann-Jax C, Andreeff M, Brenner
MK, Goodell MA, et al. An in vivo propagated human acute myeloid
leukemia expressing ABCA3. Leuk Res 2004;28:2959.
Hirschmann-Jax C, Foster AE, Wulf GG, Nuchtern JG, Jax TW,
Gobel U, et al. A distinct side population of cells with high drug
efflux capacity in human tumor cells. Proc Natl Acad Sci U S A
2004;101:1422833.
Frank NY, Margaryan A, Huang Y, Schatton T, Waaga-Gasser
AM, Gasser M, et al. ABCB5-mediated doxorubicin transport
and chemoresistance in human malignant melanoma. Cancer Res
2005;65:432033.
Amiri-Kordestani L, Basseville A, Kurdziel K, Fojo AT, Bates SE.
Targeting MDR in breast and lung cancer: discriminating its potential
importance from the failure of drug resistance reversal studies. Drug
Resist Updates 2012;15:5061.
Gatti L, Cossa G, Beretta GL, Zaffaroni N, Perego P. Novel insights
into targeting ATP-binding cassette transporters for antitumor therapy.
Curr Med Chem 2011;18:423749.
Shukla S, Ohnuma S, Ambudkar SV. Improving cancer chemotherapy with modulators of ABC drug transporters. Curr Drug Targets
2011;12:62130.
Fletcher JI, Haber M, Henderson MJ, Norris MD. ABC transporters in cancer: more than just drug efflux pumps. Nat Rev Cancer
2010;10:14756.
Yu M, Ocana A, Tannock IF. Reversal of ATP-binding cassette drug
transporter activity to modulate chemoresistance: why has it failed to
provide clinical benefit? Cancer Metastasis Rev 2013;32:21127.
Tsuruo T, Iida H, Tsukagoshi S, Sakurai Y. Overcoming of vincristine
resistance in P388 leukemia in vivo and in vitro through enhanced
cytotoxicity of vincristine and vinblastine by verapamil. Cancer Res
1981;41:196772.

250

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

[293] Slater LM, Sweet P, Stupecky M, Gupta S. Cyclosporin A reverses


vincristine and daunorubicin resistance in acute lymphatic leukemia
in vitro. J Clin Invest 1986;77:14058.
[294] Slater LM, Sweet P, Stupecky M, Wetzel MW, Gupta S. Cyclosporin
A corrects daunorubicin resistance in Ehrlich ascites carcinoma. Br J
Cancer 1986;54:2358.
[295] Cornwell MM, Pastan I, Gottesman MM. Certain calcium channel
blockers bind specifically to multidrug-resistant human KB carcinoma membrane vesicles and inhibit drug binding to P-glycoprotein.
J Biol Chem 1987;262:216670.
[296] Foxwell BM, Mackie A, Ling V, Ryffel B. Identification of the multidrug resistance-related P-glycoprotein as a cyclosporine binding
protein. Mol Pharmacol 1989;36:5436.
[297] Shukla S, Wu CP, Ambudkar SV. Development of inhibitors of ATPbinding cassette drug transporters: present status and challenges.
Expert Opin Drug Metab Toxicol 2008;4:20523.
[298] Twentyman PR, Bleehen NM. Resistance modification by PSC-833, a
novel non-immunosuppressive cyclosporin [corrected]. Eur J Cancer
1991;27:163942.
[299] Baekelandt M, Lehne G, Trope CG, Szanto I, Pfeiffer P, Gustavssson
B, et al. Phase I/II trial of the multidrug-resistance modulator valspodar combined with cisplatin and doxorubicin in refractory ovarian
cancer. J Clin Oncol 2001;19:298393.
[300] Fracasso PM, Blum KA, Ma MK, Tan BR, Wright LP, Goodner SA, et al. Phase I study of pegylated liposomal doxorubicin
and the multidrug-resistance modulator, valspodar. Br J Cancer
2005;93:4653.
[301] Chico I, Kang MH, Bergan R, Abraham J, Bakke S, Meadows B,
et al. Phase I study of infusional paclitaxel in combination with the
P-glycoprotein antagonist PSC 833. J Clin Oncol 2001;19:83242.
[302] Fracasso PM, Brady MF, Moore DH, Walker JL, Rose PG, Letvak L,
et al. Phase II study of paclitaxel and valspodar (PSC 833) in refractory
ovarian carcinoma: a gynecologic oncology group study. J Clin Oncol
2001;19:297582.
[303] Lhomme C, Joly F, Walker JL, Lissoni AA, Nicoletto MO, Manikhas
GM, et al. Phase III study of valspodar (PSC 833) combined with
paclitaxel and carboplatin compared with paclitaxel and carboplatin
alone in patients with stage IV or suboptimally debulked stage III
epithelial ovarian cancer or primary peritoneal cancer. J Clin Oncol
2008;26:267482.
[304] Motzer RJ, Lyn P, Fischer P, Lianes P, Ngo RL, Cordon-Cardo C,
et al. Phase I/II trial of dexverapamil plus vinblastine for patients
with advanced renal cell carcinoma. J Clin Oncol 1995;13:195865.
[305] Lehnert M, Mross K, Schueller J, Thuerlimann B, Kroeger N, Kupper H. Phase II trial of dexverapamil and epirubicin in patients
with non-responsive metastatic breast cancer. Br J Cancer 1998;77:
115563.
[306] Seiden MV, Swenerton KD, Matulonis U, Campos S, Rose P, Batist
G, et al. A phase II study of the MDR inhibitor biricodar (INCEL, VX710) and paclitaxel in women with advanced ovarian cancer refractory
to paclitaxel therapy. Gynecol Oncol 2002;86:30210.
[307] Gandhi L, Harding MW, Neubauer M, Langer CJ, Moore M, Ross
HJ, et al. A phase II study of the safety and efficacy of the multidrug resistance inhibitor VX-710 combined with doxorubicin and
vincristine in patients with recurrent small cell lung cancer. Cancer
2007;109:92432.
[308] Martin C, Berridge G, Mistry P, Higgins C, Charlton P, Callaghan R.
The molecular interaction of the high affinity reversal agent XR9576
with P-glycoprotein. Br J Pharmacol 1999;128:40311.
[309] Kelly RJ, Draper D, Chen CC, Robey RW, Figg WD, Piekarz
RL, et al. A pharmacodynamic study of docetaxel in combination
with the P-glycoprotein antagonist tariquidar (XR9576) in patients
with lung, ovarian, and cervical cancer. Clin Cancer Res 2011;17:
56980.
[310] Rubin EH, de Alwis DP, Pouliquen I, Green L, Marder P, Lin Y, et al. A
phase I trial of a potent P-glycoprotein inhibitor, Zosuquidar.3HCl trihydrochloride (LY335979), administered orally in combination with

[311]

[312]

[313]

[314]

[315]

[316]

[317]

[318]

[319]

[320]

[321]

[322]

[323]

[324]

[325]

[326]

doxorubicin in patients with advanced malignancies. Clin Cancer Res


2002;8:37107.
Fracasso PM, Goldstein LJ, de Alwis DP, Rader JS, Arquette MA,
Goodner SA, et al. Phase I study of docetaxel in combination with the
P-glycoprotein inhibitor, zosuquidar, in resistant malignancies. Clin
Cancer Res 2004;10:72208.
Ruff P, Vorobiof DA, Jordaan JP, Demetriou GS, Moodley SD, Nosworthy AL, et al. A randomized, placebo-controlled, double-blind
phase 2 study of docetaxel compared to docetaxel plus zosuquidar
(LY335979) in women with metastatic or locally recurrent breast
cancer who have received one prior chemotherapy regimen. Cancer
Chemother Pharmacol 2009;64:7638.
Malingre MM, Beijnen JH, Rosing H, Koopman FJ, Jewell RC, Paul
EM, et al. Co-administration of GF120918 significantly increases the
systemic exposure to oral paclitaxel in cancer patients. Br J Cancer
2001;84:427.
Kuppens IE, Witteveen EO, Jewell RC, Radema SA, Paul EM,
Mangum SG, et al. A phase I, randomized, open-label, parallel-cohort,
dose-finding study of elacridar (GF120918) and oral topotecan in
cancer patients. Clin Cancer Res 2007;13:327685.
Dieras V, Bonneterre J, Laurence V, Degardin M, Pierga JY, Bonneterre ME, et al. Phase I combining a P-glycoprotein inhibitor,
MS209, in combination with docetaxel in patients with advanced
malignancies. Clin Cancer Res 2005;11:625660.
Saeki T, Nomizu T, Toi M, Ito Y, Noguchi S, Kobayashi T, et al. Dofequidar fumarate (MS-209) in combination with cyclophosphamide,
doxorubicin, and fluorouracil for patients with advanced or recurrent
breast cancer. J Clin Oncol 2007;25:4117.
Duan Z, Choy E, Hornicek FJ. NSC23925, identified in a highthroughput cell-based screen, reverses multidrug resistance. PLoS
ONE 2009;4:e7415.
Duan Z, Li X, Huang H, Yuan W, Zheng SL, Liu X, et al.
Synthesis and evaluation of (2-(4-methoxyphenyl)-4-quinolinyl)(2piperidinyl)methanol (NSC23925) isomers to reverse multidrug
resistance in cancer. J Med Chem 2012;55:311321.
Susa M, Choy E, Yang C, Schwab J, Mankin H, Hornicek F, et al.
Multidrug resistance reversal agent, NSC77037, identified with a cellbased screening assay. J Biomol Screen 2010;15:28796.
Burkhart CA, Watt F, Murray J, Pajic M, Prokvolit A, Xue C, et al.
Small-molecule multidrug resistance-associated protein 1 inhibitor
reversan increases the therapeutic index of chemotherapy in mouse
models of neuroblastoma. Cancer Res 2009;69:657380.
Ozvegy-Laczka C, Hegedus T, Varady G, Ujhelly O, Schuetz
JD, Varadi A, et al. High-affinity interaction of tyrosine kinase
inhibitors with the ABCG2 multidrug transporter. Mol Pharmacol
2004;65:148595.
Nakanishi T, Shiozawa K, Hassel BA, Ross DD. Complex interaction of BCRP/ABCG2 and imatinib in BCR-ABL-expressing
cells: BCRP-mediated resistance to imatinib is attenuated by
imatinib-induced reduction of BCRP expression. Blood 2006;108:
67884.
Weroha SJ, Oberg AL, Ziegler KL, Dakhilm SR, Rowland KM, Hartmann LC, et al. Phase II trial of lapatinib and topotecan (LapTop)
in patients with platinum-refractory/resistant ovarian and primary
peritoneal carcinoma. Gynecol Oncol 2011;122:11620.
Coleman RL, Broaddus RR, Bodurka DC, Wolf JK, Burke TW,
Kavanagh JJ, et al. Phase II trial of imatinib mesylate in patients
with recurrent platinum- and taxane-resistant epithelial ovarian and
primary peritoneal cancers. Gynecol Oncol 2006;101:12631.
Noguera IR, Sun CC, Broaddus RR, Branham D, Levenback CF,
Ramirez PT, et al. Phase II trial of imatinib mesylate in patients
with recurrent platinum- and taxane-resistant low-grade serous carcinoma of the ovary, peritoneum, or fallopian tube. Gynecol Oncol
2012;125:6405.
Matei D, Emerson RE, Schilder J, Menning N, Baldridge LA, Johnson
CS, et al. Imatinib mesylate in combination with docetaxel for the
treatment of patients with advanced, platinum-resistant ovarian cancer

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

[327]

[328]

[329]

[330]

[331]

[332]

[333]

[334]

[335]

[336]

[337]

[338]

[339]

[340]

[341]

[342]

[343]

and primary peritoneal carcinomatosis: a Hoosier Oncology Group


trial. Cancer 2008;113:72332.
OConnor R, OLeary M, Ballot J, Collins CD, Kinsella P, Mager
DE, et al. A phase I clinical and pharmacokinetic study of the multidrug resistance protein-1 (MRP-1) inhibitor sulindac, in combination
with epirubicin in patients with advanced cancer. Cancer Chemother
Pharmacol 2007;59:7987.
Duncan K, Uwimpuhwe H, Czibere A, Sarkar D, Libermann TA,
Fisher PB, et al. NSAIDs induce apoptosis in nonproliferating ovarian cancer cells and inhibit tumor growth in vivo. IUBMB Life
2012;64:63643.
National Institutes of Health Clinical C, National Cancer I,
Charite University BG. Surgery plus sulindac or surgery alone
for advanced colorectal cancer. http://ClinicalTrials.gov/show/
NCT01856322.2019
Liu KJ, He JH, Su XD, Sim HM, Xie JD, Chen XG, et al.
Saracatinib (AZD0530) is a potent modulator of ABCB1-mediated
multidrug resistance in vitro and in vivo. Int J Cancer 2013;132:
22435.
University College L, AstraZeneca, Cancer Research UK.
Saracatinib and paclitaxel in platinum-resistant ovarian cancer;
2012.
http://clinicaltrials.gov/ct2/show/NCT01196741?term=
NCT01196741&rank=1
Shi Z, Tiwari AK, Shukla S, Robey RW, Singh S, Kim IW, et al.
Sildenafil reverses ABCB1- and ABCG2-mediated chemotherapeutic
drug resistance. Cancer Res 2011;71:302941.
Das A, Durrant D, Mitchell C, Mayton E, Hoke NN, Salloum FN,
et al. Sildenafil increases chemotherapeutic efficacy of doxorubicin in
prostate cancer and ameliorates cardiac dysfunction. Proc Natl Acad
Sci U S A 2010;107:182027.
Bierman WF, Scheffer GL, Schoonderwoerd A, Jansen G, van Agtmael MA, Danner SA, et al. Protease inhibitors atazanavir, lopinavir
and ritonavir are potent blockers, but poor substrates, of ABC transporters in a broad panel of ABC transporter-overexpressing cell lines.
J Antimicrobial Chemother 2010;65:167280.
Oostendorp RL, Huitema A, Rosing H, Jansen RS, Ter Heine R,
Keessen M, et al. Coadministration of ritonavir strongly enhances
the apparent oral bioavailability of docetaxel in patients with solid
tumors. Clin Cancer Res 2009;15:422833.
Martinez L, Arnaud O, Henin E, Tao H, Chaptal V, Doshi R,
et al. Understanding polyspecificity within the substrate-binding
cavity of the human multidrug resistance P-glycoprotein. FEBS J
2014;281:67382.
Zhang J, Sun T, Liang L, Wu T, Wang Q. Drug promiscuity of Pglycoprotein and its mechanism of interaction with paclitaxel and
doxorubicin. Soft Matter 2014;10:43845.
Chufan EE, Sim HM, Ambudkar SV. Molecular basis of the polyspecificity of P-glycoprotein (ABCB1): recent biochemical and structural
studies. Adv Cancer Res 2015;125:7196.
Jekerle V, Kassack MU, Reilly RM, Wiese M, Piquette-Miller M.
Functional comparison of single- and double-stranded mdr1 antisense
oligodeoxynucleotides in human ovarian cancer cell lines. J Pharm
Pharm Sci 2005;8:51627.
Lu S, Huang Q, Wang Z, Song Y, Wang L. Reversal of multi-drug resistance by vector-based-shRNA-mdr1 in vitro and in vivo. J Huazhong
Univ Sci Technol Med Sci 2009;29:6204.
Zhang H, Wang J, Cai K, Jiang L, Zhou D, Yang C, et al.
Downregulation of gene MDR1 by shRNA to reverse multidrugresistance of ovarian cancer A2780 cells. J Cancer Res Ther 2012;8:
22631.
Materna V, Stege A, Surowiak P, Priebsch A, Lage H. RNA
interference-triggered reversal of ABCC2-dependent cisplatin resistance in human cancer cells. Biochem Biophys Res Commun
2006;348:1537.
Wang B, Li S, Meng X, Shang H, Guan Y. Inhibition of mdr1 by
G-quadruplex oligonucleotides and reversal of paclitaxel resistance
in human ovarian cancer cells. Tumour Biol 2015.

251

[344] Kwak JO, Lee SH, Lee GS, Kim MS, Ahn YG, Lee JH, et al.
Selective inhibition of MDR1 (ABCB1) by HM30181 increases oral
bioavailability and therapeutic efficacy of paclitaxel. Eur J Pharmacol
2010;627:928.
[345] Kim TE, Gu N, Yoon SH, Cho JY, Park KM, Shin SG, et al. Tolerability
and pharmacokinetics of a new P-glycoprotein inhibitor, HM30181,
in healthy Korean male volunteers: single- and multiple-dose randomized, placebo-controlled studies. Clin Ther 2012;34:48294.
[346] Cha YJ, Lee H, Gu N, Kim TE, Lim KS, Yoon SH, et al. Sustained
Increase in the Oral Bioavailability of Loperamide after a Single
Oral Dose of HM30181, a P-glycoprotein Inhibitor, in Healthy Male
Participants. Basic Clin Pharmacol Toxicol 2013.
[347] Kinex Pharmaceuticals I, Atlantic Research G. A study to
evaluate safety, tolerability, pharmacokinetics and activity of oraxol in subjects with advanced malignancies; 2014.
http://ClinicalTrials.gov/show/NCT01967043
[348] Mei Y, Qian F, Wei D, Liu J. Reversal of cancer multidrug resistance
by green tea polyphenols. J Pharmacy Pharmacol 2004;56:130714.
[349] Wu CP, Calcagno AM, Hladky SB, Ambudkar SV, Barrand MA.
Modulatory effects of plant phenols on human multidrug-resistance
proteins 1, 4 and 5 (ABCC1, 4 and 5). FEBS J 2005;272:472540.
[350] Zhang S, Yang X, Morris ME. Flavonoids are inhibitors of breast cancer resistance protein (ABCG2)-mediated transport. Mol Pharmacol
2004;65:120816.
[351] Cooray HC, Janvilisri T, van Veen HW, Hladky SB, Barrand MA.
Interaction of the breast cancer resistance protein with plant polyphenols. Biochem Biophys Res Commun 2004;317:26975.
[352] Shukla S, Zaher H, Hartz A, Bauer B, Ware JA, Ambudkar
SV. Curcumin inhibits the activity of ABCG2/BCRP1, a multidrug resistance-linked ABC drug transporter in mice. Pharm Res
2009;26:4807.
[353] Limtrakul P, Chearwae W, Shukla S, Phisalphong C, Ambudkar
SV. Modulation of function of three ABC drug transporters, Pglycoprotein (ABCB1), mitoxantrone resistance protein (ABCG2)
and multidrug resistance protein 1 (ABCC1) by tetrahydrocurcumin,
a major metabolite of curcumin. Mol Cell Biochem 2007;296:8595.
[354] Singh AP, Singh RK, Kim KK, Satyan KS, Nussbaum R, Torres M,
et al. Cranberry proanthocyanidins are cytotoxic to human cancer cells
and sensitize platinum-resistant ovarian cancer cells to paraplatin.
Phytother Res 2009;23:106674.
[355] Shin SY, Jung H, Ahn S, Hwang D, Yoon H, Hyun J, et al. Polyphenols bearing cinnamaldehyde scaffold showing cell growth inhibitory
effects on the cisplatin-resistant A2780/Cis ovarian cancer cells.
Bioorg Med Chem 2014;22:180920.
[356] Chen H, Landen CN, Li Y, Alvarez RD, Tollefsbol TO. Epigallocatechin gallate and sulforaphane combination treatment induce apoptosis
in paclitaxel-resistant ovarian cancer cells through hTERT and Bcl-2
down-regulation. Exp Cell Res 2013;319:697706.
[357] Trudel D, Labbe DP, Araya-Farias M, Doyen A, Bazinet L, Duchesne
T, et al. A two-stage, single-arm, phase II study of EGCG-enriched
green tea drink as a maintenance therapy in women with advanced
stage ovarian cancer. Gynecol Oncol 2013;131:35761.
[358] Zhao N, Li YH, Wu XK, Wang GY, Cai DY, Han FJ. [Effect of Brucea
javanica fruit oil emulsion combined cisplatin on the growth inhibition
of transplanted tumor in human ovarian cancer SKOV3 nude mice: an
experimental study]. Zhongguo Zhong Xi Yi Jie He Za Zhi Zhongguo
Zhongxiyi Jiehe Zazhi 2015;35:5762.
[359] Patel NR, Rathi A, Mongayt D, Torchilin VP. Reversal of multidrug
resistance by co-delivery of tariquidar (XR9576) and paclitaxel using
long-circulating liposomes. Int J Pharm 2011;416:2969.
[360] Ganta S, Devalapally H, Amiji M. Curcumin enhances oral
bioavailability and anti-tumor therapeutic efficacy of paclitaxel
upon administration in nanoemulsion formulation. J Pharm Sci
2010;99:463041.
[361] Yadav S, van Vlerken LE, Little SR, Amiji MM. Evaluations of
combination MDR-1 gene silencing and paclitaxel administration
in biodegradable polymeric nanoparticle formulations to overcome

252

[362]

[363]

[364]

[365]

[366]

[367]

[368]

[369]

[370]

[371]

[372]

[373]

[374]

[375]

[376]

[377]

[378]

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256


multidrug resistance in cancer cells. Cancer Chemother Pharmacol
2009;63:71122.
Yang Y, Wang Z, Li M, Lu S. Chitosan/pshRNA plasmid nanoparticles targeting MDR1 gene reverse paclitaxel resistance in ovarian
cancer cells. J Huazhong Univ Sci Technol Med Sci 2009;29:
23942.
He C, Liu D, Lin W. Self-assembled nanoscale coordination polymers carrying siRNAs and cisplatin for effective treatment of resistant
ovarian cancer. Biomaterials 2015;36:12433.
Yang X, Iyer AK, Singh A, Milane L, Choy E, Hornicek FJ, et al. Cluster of differentiation 44 targeted hyaluronic acid based nanoparticles
for MDR1 siRNA delivery to overcome drug resistance in ovarian
cancer. Pharm Res 2014.
Yang X, Lyer AK, Singh A, Choy E, Hornicek FJ, Amiji MM, et al.
MDR1 siRNA loaded hyaluronic acid-based CD44 targeted nanoparticle systems circumvent paclitaxel resistance in ovarian cancer. Sci
Rep 2015;5:8509.
Jensen PB, Christensen IJ, Sehested M, Hansen HH, Vindelov L.
Differential cytotoxicity of 19 anticancer agents in wild type and
etoposide resistant small cell lung cancer cell lines. Br J Cancer
1993;67:31120.
Jensen PB, Holm B, Sorensen M, Christensen IJ, Sehested M. In vitro
cross-resistance and collateral sensitivity in seven resistant smallcell lung cancer cell lines: preclinical identification of suitable drug
partners to taxotere, taxol, topotecan and gemcitabin. Br J Cancer
1997;75:86977.
Gupta RS. Cross-resistance of vinblastine- and taxol-resistant mutants
of Chinese hamster ovary cells to other anticancer drugs. Cancer Treat
Rep 1985;69:51521.
Stordal B, Pavlakis N, Davey R. A systematic review of platinum and
taxane resistance from bench to clinic: an inverse relationship. Cancer
Treat Rev 2007;33:688703.
Szakacs G, Hall MD, Gottesman MM, Boumendjel A, Kachadourian
R, Day BJ, et al. Targeting the Achilles heel of multidrug-resistant
cancer by exploiting the fitness cost of resistance. Chem Rev
2014;114:575374.
Ludwig JA, Szakacs G, Martin SE, Chu BF, Cardarelli C, Sauna ZE,
et al. Selective toxicity of NSC73306 in MDR1-positive cells as a
new strategy to circumvent multidrug resistance in cancer. Cancer
Res 2006;66:480815.
Fung KL, Tepede AK, Pluchino KM, Pouliot LM, Pixley JN, Hall
MD, et al. Uptake of compounds that selectively kill multidrugresistant cells: the copper transporter SLC31A1 (CTR1) increases
cellular accumulation of the thiosemicarbazone NSC73306. Mol
Pharm 2014;11:2692702.
Turk D, Hall MD, Chu BF, Ludwig JA, Fales HM, Gottesman
MM, et al. Identification of compounds selectively killing multidrugresistant cancer cells. Cancer Res 2009;69:8293301.
Whitnall M, Howard J, Ponka P, Richardson DR. A class of iron
chelators with a wide spectrum of potent antitumor activity that overcomes resistance to chemotherapeutics. Proc Natl Acad Sci U S A
2006;103:149016.
Heike Y, Hamada H, Inamura N, Sone S, Ogura T, Tsuruo
T. Monoclonal anti-P-glycoprotein antibody-dependent killing of
multidrug-resistant tumor cells by human mononuclear cells. Jpn J
Cancer Res 1990;81:115561.
Pawlak-Roblin C, Tosi PF, Perrin L, Devy J, Venteo L, Albert
P, et al. Inhibition of multidrug resistance by immunisation with
synthetic P-glycoprotein-derived peptides. Eur J Cancer 2004;40:
60613.
Goda K, Fenyvesi F, Bacso Z, Nagy H, Marian T, Megyeri A, et al.
Complete inhibition of P-glycoprotein by simultaneous treatment
with a distinct class of modulators and the UIC2 monoclonal antibody.
J Pharmacol Exp Ther 2007;320:818.
Mechetner EB, Roninson IB. Efficient inhibition of P-glycoproteinmediated multidrug resistance with a monoclonal antibody. Proc Natl
Acad Sci U S A 1992;89:58248.

[379] Szaloki G, Krasznai ZT, Toth A, Vizkeleti L, Szollosi AG, Trencsenyi


G, et al. The strong in vivo anti-tumor effect of the UIC2 monoclonal antibody is the combined result of Pgp inhibition and antibody
dependent cell-mediated cytotoxicity. PLOS ONE 2014;9:e107875.
[380] Fowers KD, Kopecek J. Targeting of multidrug-resistant human ovarian carcinoma cells with anti-P-glycoprotein antibody conjugates.
Macromol Biosci 2012;12:50214.
[381] Morizono K, Xie Y, Ringpis GE, Johnson M, Nassanian H, Lee B,
et al. Lentiviral vector retargeting to P-glycoprotein on metastatic
melanoma through intravenous injection. Nat Med 2005;11:34652.
[382] Schinkel AH, Jonker JW. Mammalian drug efflux transporters of the
ATP binding cassette (ABC) family: an overview. Adv Drug Deliv
Rev 2003;55:329.
[383] Hastie C, Masters JR, Moss SE, Naaby-Hansen S. Interferongamma reduces cell surface expression of annexin 2 and suppresses
the invasive capacity of prostate cancer cells. J Biol Chem
2008;283:12595603.
[384] Jones RJ, Gu D, Bjorklund CC, Kuiatse I, Remaley AT, Bashir T, et al.
The novel anticancer agent JNJ-26854165 induces cell death through
inhibition of cholesterol transport and degradation of ABCA1. J
Pharmacol Exp Ther 2013;346:38192.
[385] Boonstra R, Timmer-Bosscha H, van Echten-Arends J, van der Kolk
DM, van den Berg A, de Jong B, et al. Mitoxantrone resistance in a
small cell lung cancer cell line is associated with ABCA2 upregulation. Br J Cancer 2004;90:24117.
[386] Hupfeld T, Chapuy B, Schrader V, Beutler M, Veltkamp C, Koch R,
et al. Tyrosinekinase inhibition facilitates cooperation of transcription
factor SALL4 and ABC transporter A3 towards intrinsic CML cell
drug resistance. Br J Haematol 2013;161:20413.
[387] Chapuy B, Koch R, Radunski U, Corsham S, Cheong N, Inagaki
N, et al. Intracellular ABC transporter A3 confers multidrug resistance in leukemia cells by lysosomal drug sequestration. Leukemia
2008;22:157686.
[388] Dai W, Wang F, He L, Lin C, Wu S, Chen P, et al. Genistein inhibits
hepatocellular carcinoma cell migration by reversing the epithelialmesenchymal transition: partial mediation by the transcription factor
NFAT1. Mol Carcinog 2015;54:30111.
[389] Mendonca-Torres MC, Roberts SS. The translocator protein (TSPO)
ligand PK11195 induces apoptosis and cell cycle arrest and sensitizes
to chemotherapy treatment in pre- and post-relapse neuroblastoma
cell lines. Cancer Biol Ther 2013;14:31926.
[390] Shukla A, Hillegass JM, MacPherson MB, Beuschel SL, Vacek
PM, Pass HI, et al. Blocking of ERK1 and ERK2 sensitizes human
mesothelioma cells to doxorubicin. Mol Cancer 2010;9:314.
[391] Gai J, Ji M, Shi C, Li W, Chen S, Wang Y, et al. FoxO regulates
expression of ABCA6, an intracellular ATP-binding-cassette transporter responsive to cholesterol. Int J Biochem Cell Biol 2013;45:
26519.
[392] Wakaumi M, Ishibashi K, Ando H, Kasanuki H, Tsuruoka S. Acute
digoxin loading reduces ABCA8A mRNA expression in the mouse
liver. Clin Exp Pharmacol Physiol 2005;32:103441.
[393] Tsuruoka S, Ishibashi K, Yamamoto H, Wakaumi M, Suzuki M,
Schwartz GJ, et al. Functional analysis of ABCA8, a new drug transporter. Biochem Biophys Res Commun 2002;298:415.
[394] Huang Y, Anderle P, Bussey KJ, Barbacioru C, Shankavaram U,
Dai Z, et al. Membrane transporters and channels: role of the transportome in cancer chemosensitivity and chemoresistance. Cancer Res
2004;64:4294301.
[395] Safa AR, Glover CJ, Sewell JL, Meyers MB, Biedler JL, Felsted RL.
Identification of the multidrug resistance-related membrane glycoprotein as an acceptor for calcium channel blockers. J Biol Chem
1987;262:78848.
[396] Zhang XP, Ritke MK, Yalowich JC, Slovak ML, Ho JP, Collins
KI, et al. P-glycoprotein mediates profound resistance to bisantrene.
Oncol Res 1994;6:291301.
[397] Hegedus C, Ozvegy-Laczka C, Apati A, Magocsi M, Nemet K, Orfi
L, et al. Interaction of nilotinib, dasatinib and bosutinib with ABCB1

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

[398]

[399]

[400]

[401]

[402]

[403]

[404]

[405]

[406]

[407]

[408]
[409]

[410]

[411]

[412]

[413]

[414]

[415]

and ABCG2: implications for altered anti-cancer effects and pharmacological properties. Br J Pharmacol 2009;158:115364.
Tanigawara Y, Okamura N, Hirai M, Yasuhara M, Ueda K, Kioka N,
et al. Transport of digoxin by human P-glycoprotein expressed in a
porcine kidney epithelial cell line (LLC-PK1). J Pharmacol Exp Ther
1992;263:8405.
Wils P, Phung-Ba V, Warnery A, Lechardeur D, Raeissi S, Hidalgo IJ,
et al. Polarized transport of docetaxel and vinblastine mediated by Pglycoprotein in human intestinal epithelial cell monolayers. Biochem
Pharmacol 1994;48:152830.
Dalton WS, Durie BG, Alberts DS, Gerlach JH, Cress AE. Characterization of a new drug-resistant human myeloma cell line that expresses
P-glycoprotein. Cancer Res 1986;46:512530.
Kimiya K, Naito S, Soejima T, Sakamoto N, Kotoh S, Kumazawa
J, et al. Establishment and characterization of doxorubicin-resistant
human bladder cancer cell line, KK47/ADM. J Urol 1992;148:4415.
Pastan I, Gottesman MM, Ueda K, Lovelace E, Rutherford AV, Willingham MC. A retrovirus carrying an MDR1 cDNA confers multidrug
resistance and polarized expression of P-glycoprotein in MDCK cells.
Proc Natl Acad Sci U S A 1988;85:448690.
Tebbi CK, Chervinsky D, Baker RM. Modulation of drug resistance in homoharringtonine-resistant C-1300 neuroblastoma cells
with cyclosporine A and dipyridamole. J Cell Physiol 1991;148:
46471.
Jansen WJ, Hulscher TM, van Ark-Otte J, Giaccone G, Pinedo HM,
Boven E. CPT-11 sensitivity in relation to the expression of P170glycoprotein and multidrug resistance-associated protein. Br J Cancer
1998;77:35965.
Webster L, Linsenmeyer M, Millward M, Morton C, Bishop J, Woodcock D. Measurement of cremophor EL following taxol: plasma
levels sufficient to reverse drug exclusion mediated by the multidrugresistant phenotype. J Natl Cancer Inst 1993;85:168590.
Minato K, Kanzawa F, Nishio K, Nakagawa K, Fujiwara Y, Saijo
N. Characterization of an etoposide-resistant human small-cell lung
cancer cell line. Cancer Chemother Pharmacol 1990;26:3137.
Hendricks CB, Rowinsky EK, Grochow LB, Donehower RC, Kaufmann SH. Effect of P-glycoprotein expression on the accumulation
and cytotoxicity of topotecan (SK&F 104864), a new camptothecin
analogue. Cancer Res 1992;52:226878.
Zhou XJ, Rahmani R. Preclinical and clinical pharmacology of vinca
alkaloids. Drugs 1992;44(Suppl. 4):116 [discussion 66-9].
Aoki S, Chen ZS, Higasiyama K, Setiawan A, Akiyama S, Kobayashi
M. Reversing effect of agosterol A, a spongean sterol acetate, on
multidrug resistance in human carcinoma cells. Jpn J Cancer Res
2001;92:88695.
Consoli U, Priebe W, Ling YH, Mahadevia R, Griffin M, Zhao
S, et al. The novel anthracycline annamycin is not affected
by P-glycoprotein-related multidrug resistance: comparison with
idarubicin and doxorubicin in HL-60 leukemia cell lines. Blood
1996;88:63344.
Labrie P, Maddaford SP, Lacroix J, Catalano C, Lee DK, Rakhit
S, et al. In vitro activity of novel dual action MDR anthranilamide
modulators with inhibitory activity at CYP-450. Bioorg Med Chem
2006;14:797287.
Riffkin CD, Chung R, Wall DM, Zalcberg JR, Cowman AF,
Foley M, et al. Modulation of the function of human MDR1
P-glycoprotein by the antimalarial drug mefloquine. Biochem
Pharmacol 1996;52:154552.
Pires MM, Emmert D, Hrycyna CA, Chmielewski J. Inhibition of
P-glycoprotein-mediated paclitaxel resistance by reversibly linked
quinine homodimers. Mol Pharmacol 2009;75:92100.
Mi YJ, Liang YJ, Huang HB, Zhao HY, Wu CP, Wang F, et al. Apatinib (YN968D1) reverses multidrug resistance by inhibiting the efflux
function of multiple ATP-binding cassette transporters. Cancer Res
2010;70:798191.
Zhang DM, Shu C, Chen JJ, Sodani K, Wang J, Bhatnagar J, et al.
BBA, a derivative of 23-hydroxybetulinic acid, potently reverses

[416]

[417]

[418]

[419]

[420]

[421]

[422]

[423]

[424]

[425]

[426]

[427]

[428]

[429]

[430]

[431]

253

ABCB1-mediated drug resistance in vitro and in vivo. Mol Pharm


2012;9:314759.
Robey RW, Shukla S, Finley EM, Oldham RK, Barnett D, Ambudkar SV, et al. Inhibition of P-glycoprotein (ABCB1)- and multidrug
resistance-associated protein 1 (ABCC1)-mediated transport by
the orally administered inhibitor, CBT-1((R)). Biochem Pharmacol
2008;75:130212.
Pawarode A, Shukla S, Minderman H, Fricke SM, Pinder EM,
OLoughlin KL, et al. Differential effects of the immunosuppressive agents cyclosporin A, tacrolimus and sirolimus on drug transport
by multidrug resistance proteins. Cancer Chemother Pharmacol
2007;60:17988.
Sauna ZE, Peng XH, Nandigama K, Tekle S, Ambudkar SV. The
molecular basis of the action of disulfiram as a modulator of the
multidrug resistance-linked ATP binding cassette transporters MDR1
(ABCB1) and MRP1 (ABCC1). Mol Pharmacol 2004;65:67584.
Katayama R, Koike S, Sato S, Sugimoto Y, Tsuruo T, Fujita N. Dofequidar fumarate sensitizes cancer stem-like side population cells to
chemotherapeutic drugs by inhibiting ABCG2/BCRP-mediated drug
export. Cancer Sci 2009;100:20608.
Evers R, Kool M, Smith AJ, van Deemter L, de Haas M, Borst P.
Inhibitory effect of the reversal agents V-104, GF120918 and Pluronic
L61 on MDR1 Pgp-, MRP1- and MRP2-mediated transport. Br J
Cancer 2000;83:36674.
Dai CL, Liang YJ, Chen LM, Zhang X, Deng WJ, Su XD, et al. Sensitization of ABCB1 overexpressing cells to chemotherapeutic agents
by FG020326 via binding to ABCB1 and inhibiting its function.
Biochem Pharmacol 2009;78:35564.
Gyemant N, Tanaka M, Antus S, Hohmann J, Csuka O, Mandoky L,
et al. In vitro search for synergy between flavonoids and epirubicin
on multidrug-resistant cancer cells. In Vivo 2005;19:36774.
Zhang J, Zhou F, Wu X, Gu Y, Ai H, Zheng Y, et al. 20(S)ginsenoside Rh2 noncompetitively inhibits P-glycoprotein in vitro
and in vivo: a case for herb-drug interactions. Drug Metab Dispos
2010;38:217987.
Caceres G, Robey RW, Sokol L, McGraw KL, Clark J, Lawrence
NJ, et al. HG-829 is a potent noncompetitive inhibitor of the ATPbinding cassette multidrug resistance transporter ABCB1. Cancer Res
2012;72:420413.
Matsson P, Pedersen JM, Norinder U, Bergstrom CA, Artursson P.
Identification of novel specific and general inhibitors of the three
major human ATP-binding cassette transporters P-gp, BCRP and
MRP2 among registered drugs. Pharm Res 2009;26:181631.
Dai CL, Tiwari AK, Wu CP, Su XD, Wang SR, Liu DG, et al. Lapatinib
(Tykerb, GW572016) reverses multidrug resistance in cancer cells by
inhibiting the activity of ATP-binding cassette subfamily B member
1 and G member 2. Cancer Res 2008;68:790514.
Duarte N, Gyemant N, Abreu PM, Molnar J, Ferreira MJ. New macrocyclic lathyrane diterpenes, from Euphorbia lagascae, as inhibitors
of multidrug resistance of tumour cells. Planta Med 2006;72:
1628.
Bates SE, Shieh CY, Mickley LA, Dichek HL, Gazdar A, Loriaux
DL, et al. Mitotane enhances cytotoxicity of chemotherapy in cell lines
expressing a multidrug resistance gene (mdr-1/P-glycoprotein) which
is also expressed by adrenocortical carcinomas. J Clin Endocrinol
Metab 1991;73:1829.
Yoshida N, Takagi A, Kitazawa H, Kawakami J, Adachi I. Inhibition
of P-glycoprotein-mediated transport by extracts of and monoterpenoids contained in Zanthoxyli fructus. Toxicol Appl Pharmacol
2005;209:16773.
Sato W, Fukazawa N, Nakanishi O, Baba M, Suzuki T, Yano O, et al.
Reversal of multidrug resistance by a novel quinoline derivative, MS209. Cancer Chemother Pharmacol 1995;35:2717.
Zhao XQ, Xie JD, Chen XG, Sim HM, Zhang X, Liang YJ, et al. Neratinib reverses ATP-binding cassette B1-mediated chemotherapeutic
drug resistance in vitro, in vivo, and ex vivo. Mol Pharmacol
2012;82:4758.

254

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

[432] Newman MJ, Rodarte JC, Benbatoul KD, Romano SJ, Zhang C,
Krane S, et al. Discovery and characterization of OC144-093, a novel
inhibitor of P-glycoprotein-mediated multidrug resistance. Cancer
Res 2000;60:296472.
[433] Li S, Lei Y, Jia Y, Li N, Wink M, Ma Y. Piperine, a piperidine alkaloid
from Piper nigrum re-sensitizes P-gp, MRP1 and BCRP dependent
multidrug resistant cancer cells. Phytomedicine 2011;19:837.
[434] Walter RB, Pirga JL, Cronk MR, Mayer S, Appelbaum FR, Banker
DE. PK11195, a peripheral benzodiazepine receptor (pBR) ligand,
broadly blocks drug efflux to chemosensitize leukemia and myeloma
cells by a pBR-independent, direct transporter-modulating mechanism. Blood 2005;106:358493.
[435] Baba M, Nakanishi O, Sato W, Saito A, Miyama Y, Yano O, et al.
Relationship between multidrug resistant gene expression and multidrug resistant-reversing effect of MS-209 in various tumor cells.
Cancer Chemother Pharmacol 1995;36:3617.
[436] Bates SF, Chen C, Robey R, Kang M, Figg WD, Fojo T. Reversal of
multidrug resistance: lessons from clinical oncology. Novartis Found
Symp 2002;243:8396 [discussion 96102, 1805].
[437] Bichat F, Solis-Recendez G, Poullain MG, Poupon MF, Khayat D,
Bastian G. S9788 modulation of P-glycoprotein- and multidrugrelated protein-mediated multidrug resistance by Servier 9788
in doxorubicin-resistant MCF7 cells. Biochem Pharmacol
1998;56:497502.
[438] Boesch D, Gaveriaux C, Jachez B, Pourtier-Manzanedo A, Bollinger
P, Loor F. In vivo circumvention of P-glycoprotein-mediated multidrug resistance of tumor cells with SDZ PSC 833. Cancer Res
1991;51:422633.
[439] Fojo AT, Shen DW, Mickley LA, Pastan I, Gottesman MM. Intrinsic
drug resistance in human kidney cancer is associated with expression of a human multidrug-resistance gene. J Clin Oncol 1987;5:
19227.
[440] Minderman H, OLoughlin KL, Pendyala L, Baer MR. VX-710
(biricodar) increases drug retention and enhances chemosensitivity
in resistant cells overexpressing P-glycoprotein, multidrug resistance protein, and breast cancer resistance protein. Clin Cancer Res
2004;10:182634.
[441] Jekerle V, Klinkhammer W, Scollard DA, Breitbach K, Reilly RM,
Piquette-Miller M, et al. In vitro and in vivo evaluation of WK-X-34,
a novel inhibitor of P-glycoprotein and BCRP, using radio imaging
techniques. Int J Cancer 2006;119:41422.
[442] Shepard RL, Cao J, Starling JJ, Dantzig AH. Modulation of Pglycoprotein but not MRP1- or BCRP-mediated drug resistance by
LY335979. Int J Cancer 2003;103:1215.
[443] Smith AJ, van Helvoort A, van Meer G, Szabo K, Welker E, Szakacs
G, et al. MDR3 P-glycoprotein, a phosphatidylcholine translocase,
transports several cytotoxic drugs and directly interacts with drugs
as judged by interference with nucleotide trapping. J Biol Chem
2000;275:235309.
[444] Barnouin K, Leier I, Jedlitschky G, Pourtier-Manzanedo A, Konig J,
Lehmann WD, et al. Multidrug resistance protein-mediated transport
of chlorambucil and melphalan conjugated to glutathione. Br J Cancer
1998;77:2019.
[445] Priebe W, Krawczyk M, Kuo MT, Yamane Y, Savaraj N, Ishikawa
T. Doxorubicin- and daunorubicin-glutathione conjugates, but not
unconjugated drugs, competitively inhibit leukotriene C4 transport
mediated by MRP/GS-X pump. Biochem Biophys Res Commun
1998;247:85963.
[446] Su GM, Davey MW, Davey RA. Induction of broad drug resistance in
small cell lung cancer cells and its reversal by paclitaxel. Int J Cancer
1998;76:7028.
[447] Deeley RG, Cole SP. Substrate recognition and transport by multidrug
resistance protein 1 (ABCC1). FEBS Lett 2006;580:110311.
[448] Zeng H, Chen ZS, Belinsky MG, Rea PA, Kruh GD. Transport
of methotrexate (MTX) and folates by multidrug resistance protein
(MRP) 3 and MRP1: effect of polyglutamylation on MTX transport.
Cancer Res 2001;61:722532.

[449] Sharp SY, Smith V, Hobbs S, Kelland LR. Lack of a role for MRP1
in platinum drug resistance in human ovarian cancer cell lines. Br J
Cancer 1998;78:17580.
[450] Li XQ, Li J, Shi SB, Chen P, Yu LC, Bao QL. Expression of MRP1,
BCRP, LRP and ERCC1 as prognostic factors in non-small cell lung
cancer patients receiving postoperative cisplatin-based chemotherapy.
Int J Biol Markers 2009;24:2307.
[451] Perez-Soler R, Neamati N, Zou Y, Schneider E, Doyle LA, Andreeff
M, et al. Annamycin circumvents resistance mediated by the multidrug resistance-associated protein (MRP) in breast MCF-7 and
small-cell lung UMCC-1 cancer cell lines selected for resistance to
etoposide. Int J Cancer 1997;71:3541.
[452] Wu CP, Klokouzas A, Hladky SB, Ambudkar SV, Barrand MA. Interactions of mefloquine with ABC proteins, MRP1 (ABCC1) and MRP4
(ABCC4) that are present in human red cell membranes. Biochem
Pharmacol 2005;70:50010.
[453] Germann UA, Ford PJ, Shlyakhter D, Mason VS, Harding MW.
Chemosensitization and drug accumulation effects of VX-710, verapamil, cyclosporin A, MS-209 and GF120918 in multidrug resistant
HL60/ADR cells expressing the multidrug resistance-associated protein MRP. Anticancer Drugs 1997;8:14155.
[454] Klokouzas A, Barrand MA, Hladky SB. Effects of clotrimazole
on transport mediated by multidrug resistance associated protein 1
(MRP1) in human erythrocytes and tumour cells. Eur J Biochem
2001;268:656977.
[455] Hooijberg JH, Broxterman HJ, Kool M, Assaraf YG, Peters GJ,
Noordhuis P, et al. Antifolate resistance mediated by the multidrug resistance proteins MRP1 and MRP2. Cancer Res 1999;59:
25325.
[456] Payen L, Delugin L, Courtois A, Trinquart Y, Guillouzo A, Fardel O.
The sulphonylurea glibenclamide inhibits multidrug resistance protein (MRP1) activity in human lung cancer cells. Br J Pharmacol
2001;132:77884.
[457] Benyahia B, Huguet S, Decleves X, Mokhtari K, Criniere E,
Bernaudin JF, et al. Multidrug resistance-associated protein MRP1
expression in human gliomas: chemosensitization to vincristine and
etoposide by indomethacin in human glioma cell lines overexpressing
MRP1. J Neurooncol 2004;66:6570.
[458] Maeno K, Nakajima A, Conseil G, Rothnie A, Deeley RG, Cole SP.
Molecular basis for reduced estrone sulfate transport and altered modulator sensitivity of transmembrane helix (TM) 6 and TM17 mutants
of multidrug resistance protein 1 (ABCC1). Drug Metab Dispos
2009;37:141120.
[459] Qian YM, Grant CE, Westlake CJ, Zhang DW, Lander PA, Shepard
RL, et al. Photolabeling of human and murine multidrug resistance protein 1 with the high affinity inhibitor [125I]LY475776 and
azidophenacyl-[35S]glutathione. J Biol Chem 2002;277:3522531.
[460] Weiss J, Theile D, Ketabi-Kiyanvash N, Lindenmaier H, Haefeli WE.
Inhibition of MRP1/ABCC1, MRP2/ABCC2, and MRP3/ABCC3
by nucleoside, nucleotide, and non-nucleoside reverse transcriptase
inhibitors. Drug Metab Dispos 2007;35:3404.
[461] Wang S, Folkes A, Chuckowree I, Cockcroft X, Sohal S, Miller
W, et al. Studies on pyrrolopyrimidines as selective inhibitors of
multidrug-resistance-associated protein in multidrug resistance. J
Med Chem 2004;47:132938.
[462] Liedert B, Materna V, Schadendorf D, Thomale J, Lage H.
Overexpression of cMOAT (MRP2/ABCC2) is associated with
decreased formation of platinum-DNA adducts and decreased G2arrest in melanoma cells resistant to cisplatin. J Invest Dermatol
2003;121:1726.
[463] Jedlitschky G, Hoffmann U, Kroemer HK. Structure and function of
the MRP2 (ABCC2) protein and its role in drug disposition. Expert
Opin Drug Metab Toxicol 2006;2:35166.
[464] Horikawa M, Kato Y, Tyson CA, Sugiyama Y. The potential for an
interaction between MRP2 (ABCC2) and various therapeutic agents:
probenecid as a candidate inhibitor of the biliary excretion of irinotecan metabolites. Drug Metab Pharmacokinet 2002;17:2333.

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256


[465] Hu Y, Stephen AG, Cao J, Tanzer LR, Slapak CA, Harrison SD, et al. A
very early induction of major vault protein accompanied by increased
drug resistance in U-937 cells. Int J Cancer 2002;97:14956.
[466] Evers R, de Haas M, Sparidans R, Beijnen J, Wielinga PR, Lankelma
J, et al. Vinblastine and sulfinpyrazone export by the multidrug resistance protein MRP2 is associated with glutathione export. Br J Cancer
2000;83:37583.
[467] Chen ZS, Kawabe T, Ono M, Aoki S, Sumizawa T, Furukawa T,
et al. Effect of multidrug resistance-reversing agents on transporting
activity of human canalicular multispecific organic anion transporter.
Mol Pharmacol 1999;56:121928.
[468] Zhu X, Zhang X, Ma G, Yan J, Wang H, Yang Q. Transport characteristics of tryptanthrin and its inhibitory effect on P-gp and MRP2 in
Caco-2 cells. J Pharm Pharm Sci 2011;14:32535.
[469] Zelcer N, Saeki T, Reid G, Beijnen JH, Borst P. Characterization of
drug transport by the human multidrug resistance protein 3 (ABCC3).
J Biol Chem 2001;276:464007.
[470] Zhang G, Wang Z, Qian F, Zhao C, Sun C. Silencing of the ABCC4
gene by RNA interference reverses multidrug resistance in human
gastric cancer. Oncol Rep 2015;33:114754.
[471] Ritter CA, Jedlitschky G, Meyer zu Schwabedissen H, Grube M, Kock
K, Kroemer HK. Cellular export of drugs and signaling molecules by
the ATP-binding cassette transporters MRP4 (ABCC4) and MRP5
(ABCC5). Drug Metab Rev 2005;37:25378.
[472] Sauna ZE, Nandigama K, Ambudkar SV. Multidrug resistance protein
4 (ABCC4)-mediated ATP hydrolysis: effect of transport substrates
and characterization of the post-hydrolysis transition state. J Biol
Chem 2004;279:4885564.
[473] Reid G, Wielinga P, Zelcer N, De Haas M, Van Deemter L, Wijnholds
J, et al. Characterization of the transport of nucleoside analog drugs
by the human multidrug resistance proteins MRP4 and MRP5. Mol
Pharmacol 2003;63:1094103.
[474] Wielinga P, Hooijberg JH, Gunnarsdottir S, Kathmann I, Reid G, Zelcer N, et al. The human multidrug resistance protein MRP5 transports
folates and can mediate cellular resistance against antifolates. Cancer
Res 2005;65:442530.
[475] Belinsky MG, Chen ZS, Shchaveleva I, Zeng H, Kruh GD. Characterization of the drug resistance and transport properties of multidrug
resistance protein 6 (MRP6, ABCC6). Cancer Res 2002;62:61727.
[476] Bessho Y, Oguri T, Ozasa H, Uemura T, Sakamoto H, Miyazaki M,
et al. ABCC10/MRP7 is associated with vinorelbine resistance in
non-small cell lung cancer. Oncol Rep 2009;21:2638.
[477] Kuang YH, Shen T, Chen X, Sodani K, Hopper-Borge E, Tiwari
AK, et al. Lapatinib and erlotinib are potent reversal agents for
MRP7 (ABCC10)-mediated multidrug resistance. Biochem Pharmacol 2010;79:15461.
[478] Shen T, Kuang YH, Ashby CR, Lei Y, Chen A, Zhou Y, et al. Imatinib
and nilotinib reverse multidrug resistance in cancer cells by inhibiting
the efflux activity of the MRP7 (ABCC10). PLoS ONE 2009;4:e7520.
[479] Deng W, Dai CL, Chen JJ, Kathawala RJ, Sun YL, Chen HF, et al.
Tandutinib (MLN518) reverses multidrug resistance by inhibiting the
efflux activity of the multidrug resistance protein 7 (ABCC10). Oncol
Rep 2013;29:247985.
[480] Sun YL, Chen JJ, Kumar P, Chen K, Sodani K, Patel A, et al. Reversal of MRP7 (ABCC10)-mediated multidrug resistance by tariquidar.
PLOS ONE 2013;8:e55576.
[481] Guo Y, Kotova E, Chen ZS, Lee K, Hopper-Borge E, Belinsky
MG, et al. MRP8, ATP-binding cassette C11 (ABCC11), is a cyclic
nucleotide efflux pump and a resistance factor for fluoropyrimidines
2 ,3 -dideoxycytidine and 9 -(2 -phosphonylmethoxyethyl)adenine. J
Biol Chem 2003;278:2950914.
[482] Uemura T, Oguri T, Ozasa H, Takakuwa O, Miyazaki M, Maeno K,
et al. ABCC11/MRP8 confers pemetrexed resistance in lung cancer.
Cancer Sci 2010;101:240410.
[483] Kara G, Tuncer S, Turk M, Denkbas EB. Downregulation of ABCE1
via siRNA affects the sensitivity of A549 cells against chemotherapeutic agents. Med Oncol 2015;32:557.

255

[484] Yang X, Liu Y, Zong Z, Tian D. The Rho kinase inhibitor fasudil
inhibits the migratory behaviour of 95-D lung carcinoma cells.
Biomed Pharmacother 2010;64:5862.
[485] Litman T, Brangi M, Hudson E, Fetsch P, Abati A, Ross DD, et al. The
multidrug-resistant phenotype associated with overexpression of the
new ABC half-transporter, MXR (ABCG2). J Cell Sci 2000;113(Pt
11):201121.
[486] Doyle LA, Yang W, Abruzzo LV, Krogmann T, Gao Y, Rishi AK, et al.
A multidrug resistance transporter from human MCF-7 breast cancer
cells. Proc Natl Acad Sci U S A 1998;95:1566570.
[487] Burger H, Foekens JA, Look MP, Meijer-van Gelder ME, Klijn JG,
Wiemer EA, et al. RNA expression of breast cancer resistance protein, lung resistance-related protein, multidrug resistance-associated
proteins 1 and 2, and multidrug resistance gene 1 in breast cancer: correlation with chemotherapeutic response. Clin Cancer Res
2003;9:82736.
[488] Allen JD, Brinkhuis RF, Wijnholds J, Schinkel AH. The mouse
Bcrp1/Mxr/Abcp gene: amplification and overexpression in cell lines
selected for resistance to topotecan, mitoxantrone, or doxorubicin.
Cancer Res 1999;59:423741.
[489] Allen JD, van Loevezijn A, Lakhai JM, van der Valk M, van Tellingen
O, Reid G, et al. Potent and specific inhibition of the breast cancer resistance protein multidrug transporter in vitro and in mouse
intestine by a novel analogue of fumitremorgin C. Mol Cancer Ther
2002;1:41725.
[490] Yanase K, Tsukahara S, Asada S, Ishikawa E, Imai Y, Sugimoto
Y. Gefitinib reverses breast cancer resistance protein-mediated drug
resistance. Mol Cancer Ther 2004;3:111925.
[491] Jin J, Shahi S, Kang HK, van Veen HW, Fan TP. Metabolites of ginsenosides as novel BCRP inhibitors. Biochem Biophys Res Commun
2006;345:130814.
[492] Shiozawa K, Oka M, Soda H, Yoshikawa M, Ikegami Y, Tsurutani J,
et al. Reversal of breast cancer resistance protein (BCRP/ABCG2)mediated drug resistance by novobiocin, a coumermycin antibiotic.
Int J Cancer 2004;108:14651.
[493] Kannan P, Telu S, Shukla S, Ambudkar SV, Pike VW, Halldin C, et al.
The specific P-glycoprotein inhibitor Tariquidar is also a substrate
and an inhibitor for breast cancer resistance protein (BCRP/ABCG2).
ACS Chem Neurosci 2011;2:829.
[494] Bauer S, Ochoa-Puentes C, Sun Q, Bause M, Bernhardt G, Konig B,
et al. Quinoline carboxamide-type ABCG2 modulators: indole and
quinoline moieties as anilide replacements. ChemMedChem 2013.
[495] Materna V, Pleger J, Hoffmann U, Lage H. RNA expression of
MDR1/P-glycoprotein, DNA-topoisomerase I, and MRP2 in ovarian carcinoma patients: correlation with chemotherapeutic response.
Gynecol Oncol 2004;94:15260.
[496] Ween MP, Lokman NA, Hoffmann P, Rodgers RJ, Ricciardelli
C, Oehler MK. Transforming growth factor-beta-induced protein
secreted by peritoneal cells increases the metastatic potential of ovarian cancer cells. Int J Cancer 2011;128:157084.

Biographies
Miranda P. Ween, PhD received her PhD in 2010 at the
University of Adelaide (Australia) with her thesis dedicated
to understanding the role of extracellular matrix in ovarian
cancer progression. Her focus then changed to the role of
ABC transporters in chemoresistance, before moving to the
Research Centre for Infectious Diseases at the University of
Adelaide to study ABC transporters, and most recently to
SA Pathology to investigate the role of metals in diseases
affecting the lung.

256

M.P. Ween et al. / Critical Reviews in Oncology/Hematology 96 (2015) 220256

Mark Armstrong received his MRes in 2013 at the University of Exeter (United Kingdom) with his thesis dedicated to
using a transcriptomics approach for identifying possible cancer biomarkers in gastric tissue samples. He now works for
the Data Management and Analysis Centre at the University
of Adelaide (Australia).
Martin K. Oehler, MD PhD FRANZCOG CGO is
the Director of Gynaecological Oncology, Royal Adelaide Hospital and Group Leader for Reproductive Cancer
Research, School of Paediatrics and Reproductive Health,
Robinson Research Institute, University of Adelaide and

investigates on topics related to gynaecological oncology, early detection of ovarian cancer and mechanisms of
chemoresistance.
Carmela Ricciardelli, PhD is cancer cell biologist
and Group leader for Reproductive Cancer Research,
School of Paediatrics and Reproductive Health, Robinson
Research Institute, University of Adelaide. Her research
focuses on the understanding the cross-talk between cancer cells and the tumour microenvironment, early detection
of ovarian cancer and mechanisms of metastasis and
chemoresistance.

You might also like