You are on page 1of 31

Leukemia and

Ly m p h o m a
R. Paul Guillerman, MDa,*, Stephan D. Voss, MD, PhDb,
Bruce R. Parker, MDa

KEYWORDS
 Lymphoma  Leukemia  Hodgkin lymphoma
 Non-Hodgkin lymphoma  Acute lymphoblastic leukemia
 Acute myeloid leukemia

Leukemia and lymphoma are the most common and LEUKEMIA


third most common pediatric malignancies, respec- Classification and Epidemiology
tively, and together account for nearly half of all
Leukemia is the most common childhood malig-
cases of childhood cancer. Although childhood
nancy, accounting for one-quarter to one-third of
leukemia and lymphoma share similar cell lineage
childhood malignancy cases. Nearly all childhood
origins and both are treated with risk-stratified
leukemia cases are the acute form. Acute
protocols entailing cytotoxic chemotherapy, the
leukemia is classified by the morphology, immuno-
clinical manifestations and imaging indications for
phenotype, and cytogenetics of the leukemic cells
these malignancies vary substantially, with some
into acute lymphoblastic leukemia (ALL) and acute
overlap. Advances in imaging have played a particu-
myeloid leukemia (AML). ALL and AML account
larly important role in improving the assessment of
for three-quarters and one-fifth of childhood leu-
lymphoma at the time of diagnosis, during treat-
kemia cases, respectively. Chronic myelogenous
ment, and following therapy. Imaging has also
leukemia (CML) accounts for less than 5% of
helped guide the design of clinical trials evaluating
cases of childhood leukemia, whereas juvenile
novel treatment strategies.
myelomonocytic leukemia (JMML), a myelodys-
Along with providing relevant details on current
plastic-myeloproliferative syndrome, accounts for
classification, epidemiology, and treatment, this
less than 1% of cases of childhood leukemia.1
article reviews the current roles of imaging in the
There is a sharp peak in ALL incidences among
management of pediatric patients with leukemia
children 2 to 3 years of age, with evidence that
and lymphoma, with attention to diagnosis, stag-
ALL initiates in utero2 AML rates are highest in
ing, risk stratification, therapy response assess-
the first 2 years of life, decline to a nadir at 6 years
ment, and surveillance for disease relapse and
of age, and slowly increase during the adolescent
adverse effects of therapy. Advances in functional
years.3
imaging and integration of clinical imaging re-
ALL is subtyped by the World Health Organization
search into cancer cooperative group study
(WHO) by immunophenotype as B-lymphoblastic or
protocols are also discussed to provide insights
T-lymphoblastic.4 Precursor B-cell ALL accounts
into future applications of imaging in the man-
for 80% to 85% of childhood ALL. About 12% of
agement of pediatric leukemia and lymphoma
ALL is T-cell lineage, which is associated with older
patients.
age, male gender, leukocytosis, and a mediastinal
radiologic.theclinics.com

a
Department of Pediatric Radiology, Texas Childrens Hospital, 6701 Fannin Street, Suite 470, Houston, TX
77030, USA
b
Department of Radiology, Childrens Hospital Boston, Harvard Medical School, 300 Longwood Avenue,
Boston, MA 02115, USA
* Corresponding author.
E-mail address: rpguille@texaschildrens.org

Radiol Clin N Am 49 (2011) 767797


doi:10.1016/j.rcl.2011.05.004
0033-8389/11/$ see front matter 2011 Elsevier Inc. All rights reserved.
768 Guillerman et al

mass. T-cell lineage ALL may be regarded as mortality. The duration of remission is prolonged
a disseminated form of T-cell lymphoblastic by chemotherapy intensification and/or HSCT,
lymphoma in terms of malignant phenotype, although HSCT may not be necessary in those
approach to therapy, and patterns of relapse. About with complete remission and favorable prognostic
2% of ALL is mature B-cell and the disseminated factors.8,9
form of Burkitt or Burkitt-like lymphoma.1 AML has
traditionally been subtyped into M0 to M7 forms
Imaging Features
according to the French-American-British (FAB)
Cooperative Group morphologic-immunohisto- Fever, petechiae, lethargy, and pallor caused by
chemical classification system. Acute promyelo- bone marrow suppression by leukemic cells are
cytic leukemia (APL), the M3 subtype of AML, is common at presentation. These symptoms and
notable for bleeding complications related to severe signs often prompt a chest radiograph. Chest radi-
coagulopathy. A newer classification of AML by the ography may reveal a mediastinal mass (especially
WHO incorporates cytogenetic abnormalities and from thymic infiltration in T-cell ALL), cardiomegaly
specific gene mutations and provides more reliable and pulmonary vascular plethora (related to
prognostic information.4 anemia), pulmonary air space opacification (related
An increased risk of leukemia is associated with to infection, hemorrhage, or leukostasis), pleural
certain genetic disorders, including trisomy 21, thickening (especially in JMML), splenomegaly
monosomy 7, and neurofibromatosis type 1 (partic- (present in 75%), or skeletal abnormalities.10
ularly JMML), and DNA repair disorders such as More than a third of children with leukemia
ataxia-telangiectasia. Of special interest to the present with limping, bone pain, arthralgia, or other
radiologist is the reported increased risk of complaints referable to the extremities or spine.
leukemia from prenatal or postnatal radiation expo- Approximately 40% of children presenting with
sure, although the magnitude of the risk is subject acute leukemia have at least 1 radiographic skeletal
to considerable uncertainty and debate.5 abnormality.11,12 The number of bones involved on
radiographs correlates with bone pain severity,
but symptoms correlate poorly with the location of
Treatment
skeletal lesions on radiographs and asymptomatic
Risk-based treatment assignment is used in chil- involvement is common, especially in nonweight-
dren with leukemia so that those children who bearing areas.12,13 Findings of leukemia on skeletal
have a good outcome with modest therapy can radiographs include transverse lucent metaphyseal
be spared more intensive and toxic treatment, bands, diffuse demineralization, subperiosteal
whereas a more aggressive, and potentially more cortical bone erosion, periosteal reaction, lytic
toxic, therapeutic approach can be provided for bone lesions, osteosclerosis, and pathologic frac-
patients who have a lower probability of long- tures. Transverse metaphyseal lucent bands, also
term survival. The 10-year event-free survival known as leukemic lines, are attributable to distur-
(EFS) rate is 67% to 78% for standard and higher bance of endosteal mineralization resulting in
risk childhood ALL with risk-adapted combination abnormally small trabeculae adjacent to the zone
chemotherapy and central nervous system (CNS) of provisional calcification. In preschool-aged chil-
prophylactic therapy (intrathecal chemotherapy dren, transverse lucent metaphyseal bands are
with or without cranial radiation).6 More intensive more specific for leukemia than for other diseases
chemotherapy regimens or hematopoietic stem and are most conspicuous at sites of rapid skeletal
cell transplantation (HSCT) may be pursued for growth such as the distal femur, proximal tibia,
certain high-risk ALL groups and marrow relapse. proximal humerus, and distal radius (Fig. 1).12
Children with AML have a wide range in outcome Diffuse demineralization is common, particularly
depending on specific biologic factors, with a 5- after therapy, whereas osteosclerosis is uncommon
year EFS rate of 40% to 58%.7 Infection, severe and usually a late manifestation after therapy. Path-
hemorrhage, hyperleukocytosis-related leukosta- ologic fractures are most commonly observed as
sis, and resistant leukemia lead to the high mortality. vertebral compression deformities in the setting of
The mainstay of AML treatment is systemic combi- diffuse demineralization. Lytic bone lesions are
nation chemotherapy, with some form of CNS- usually metadiaphyseal and geographic or perme-
directed therapy incorporated into most protocols. ative. Subperiosteal resorption of the medial cortex
Induction of profound bone marrow aplasia is of the proximal humerus is commonly visible on
generally necessary to achieve remission, and chest radiographs at the time of presentation, but
leukocyte growth factors such as G-CSF or GM- is nonspecific and can also be observed in the
CSF are often administered to reduce the duration setting of Gaucher disease, sickle cell disease,
of neutropenia, but have no significant effect on neuroblastoma, or lymphoma (Fig. 2).14
Leukemia and Lymphoma 769

Technetium (Tc)-99m phosphonatebased bone


scintigraphy is abnormal in 75% of patients with
ALL at diagnosis. The most common abnormality
is symmetric increased uptake in the metadiaphy-
ses of the lower limbs (Fig. 3). Other patterns
include diffuse increased uptake in a superscan
pattern with accentuation of the long bone meta-
physes, focal increased uptake at sites of cortical
bone destruction or pathologic fracture, and focal
decreased uptake at sites of osteonecrosis.15 The
addition of early phase whole-body scintigraphy
may increase the sensitivity for detection of
abnormal uptake from leukemia in the long bone
metadiaphyses, spine, and pelvis compared with
that of delayed-phase whole-body scintigraphy
alone.16 The number of regions with abnormal
uptake is positively correlated with age. There is
only modest correlation between the sites of
abnormal uptake, radiographic abnormalities, and
clinical signs and symptoms.17
On magnetic resonance (MR) imaging, malig-
nant infiltration of the bone marrow by leukemia
typically manifests as increased signal intensity
on fat-suppressed T2-weighted and short-tau
inversion recovery (STIR) sequences and de-
creased signal intensity on T1-weighted images.18
Prolongation of the T1 relaxation time correlates
with the proportion of blast cells in the marrow.19
The infiltration is usually diffuse, including involve-
ment of the epiphyses (Fig. 4). The findings are
less conspicuous in hematopoietic marrow than
Fig. 1. An ankle radiograph from a 7-year-old girl in fatty marrow and consequently are more difficult
with precursor B-cell ALL shows transverse lucent to appreciate in younger children before the phys-
bands (leukemic lines) along the distal tibial and iologic conversion of hematopoietic to fatty
fibular metaphyses just proximal to the zones of
marrow.20 ALL and AML cannot be reliably
provisional calcification.

Fig. 2. Anteroposterior (A) and lateral (B) chest radiographs of a 7-year-old boy with precursor B-cell ALL reveal
subperiosteal resorption of the medial cortex and subtle transverse lucent metaphyseal bands of the proximal
humeri, as well as diffuse demineralization of the vertebral bodies.
770 Guillerman et al

Leukemic involvement of the solid viscera,


especially the spleen and thymus, is common,
and manifests as organomegaly from diffuse infil-
tration or as focal masses. Diffuse infiltration of
the thymus is characteristic of T-cell ALL (Fig. 5).
Splenomegaly and a mediastinal mass from
thymic infiltration at presentation of childhood
ALL are independent predictors of tumor lysis
syndrome.22 The constellation of splenomegaly,
hepatomegaly, lymphadenopathy, and skin rash
is characteristic of JMML.23 Nephromegaly at
presentation is usually caused by leukemic cell
infiltration, but can also be caused by renal vein
thrombosis from intravascular leukostasis.24 Focal
renal leukemic masses at presentation are usually
multifocal, bilateral, low attenuation on computed
tomography (CT), and hypoechoic on ultrasound,
and must be differentiated from lymphoma,
Fig. 3. A delayed-phase Tc-99m methylene diphosph- nephroblastomatosis, and infection. Renal infiltra-
onate bone scintigraphy image of the lower extremi- tion is most frequent with T-cell ALL and the M4
ties of a child with precursor B-cell ALL depicts and M5 subtypes of AML, and is often accom-
symmetric increased radiopharmaceutical uptake in panied by extramedullary involvement at other
the long bone metadiaphyses.
sites.25 Leukemic infiltration of the kidneys is
rarely associated with acute renal failure or renal
tubular dysfunction at presentation.26,27 Pancre-
atic enlargement from leukemic infiltration is
distinguished from MR imaging or MR spectros- unusual.28
copy. The MR imaging appearance associated Brain atrophy, at least borderline in degree and
with marrow infiltration by acute leukemia is not of unclear cause, can be seen by CT in 40% of
specific and can also be seen in settings of hema- children with ALL at diagnosis.29 Even in the pres-
topoietic marrow hyperplasia and infiltrative ence of neurologic symptoms, CNS imaging find-
metastases from solid tumors such as neuroblas- ings other than atrophy are uncommon. Head CT
toma, rhabdomyosarcoma, and Ewing sarcoma.21 or MR imaging can reveal hemorrhage or infarction

Fig. 4. An MR imaging examination of the lower extremities performed on a 3-year-old boy with refusal to walk
shows diffuse abnormal low signal intensity of the bone marrow on a T1-weighted image (A) and diffuse
abnormal high intensity of the bone marrow on a STIR (B) image caused by marrow infiltration by precursor
B-cell ALL.
Leukemia and Lymphoma 771

Fig. 5. A coronal chest CT image of 13-year-old


patient with T-cell ALL shows a characteristic medias-
tinal mass from diffuse leukemic infiltration of the
thymus.

caused by intravascular leukostasis or throm-


bocytopenia.30 Cerebral hemorrhage is more
common than subdural or subarachnoid hemor-
rhage. Abnormal enhancement of the meninges
or nerve roots in a child with leukemia suggests Fig. 6. A coronal T1-weighted image from an MR
leptomeningeal involvement, even if CSF cytologic imaging examination in an 11-year-old girl with
AML shows a soft tissue mass of the right maxillary
studies are negative. As many as half of all cases
sinus, zygoma, and inferior orbit that is isointense to
of acute leukemia involve ocular manifestations, the marrow of the diploe of the skull, consistent
and the most frequent finding is retinal hemor- with a chloroma.
rhage. Retinal hemorrhages related to leukemia
are usually bilateral and located in the posterior
pole.31 associated with AML and is rare, occurring as an
A potential diagnostic pitfall is aleukemic or sub- isolated finding in less than 1% of cases of AML
leukemic leukemia. From one-quarter to one-third and in 11% of cases along with marrow disease
of cases of leukemia present with anemia and at the time of diagnosis.36
thrombocytopenia but no leukocytosis or leukemic A definitive diagnosis of leukemia is usually es-
blasts on peripheral blood smear.11,32,33 Many of tablished by bone marrow aspiration or biopsy
these patients have bone or joint pain that can revealing malignant cells of myeloid or lymphoid
masquerade clinically as osteomyelitis or arthritis lineage. Definitive diagnosis can also be estab-
for several months and prompt referral for muscu- lished by biopsy of an extramedullary mass of
loskeletal MR imaging examinations. Appropriate leukemic cells.
recognition of bone marrow infiltration on MR
imaging can suggest the correct diagnosis of
Staging and Risk Stratification
leukemia before leukocytosis or blasts in the
peripheral blood are noted.34 Leukemia is conceptualized as a disseminated
Extramedullary leukemia (EML), also known as malignancy of the hematopoietic system and there
granulocytic sarcoma or chloroma, describes is no role for traditional staging based on imaging
a mass of leukemic cells outside the bone marrow. findings as for lymphoma and solid tumors, even
EML is more common in infants than in older chil- for children with isolated EML who must be treated
dren and can precede the blast phase by up to 4 as if there is systemic disease. ALL is classified as
years.35 The skin, orbits, CNS, and spine are the low risk, standard risk, high risk, and very high risk
most common sites, and symptoms relate to from clinical and biologic features and early treat-
mass effect. On CT, EML masses show variable ment response. Higher risk ALL groups include
enhancement and can be confused with other infants, adolescents, those with high leukocyte
neoplasms, hematoma, or abscess. On MR ima- counts, those with CNS disease, those with initial
ging, EML masses show isointensity to hypercellu- induction failure or high levels of end-induction
lar bone marrow (Fig. 6). EML is most commonly minimal residual disease (MRD), and those with
772 Guillerman et al

T-cell lineage, hypodiploidy or certain chromo- whereas a low marrow fat fraction persists in the
some translocations.37 In AML, risk category defi- setting of unresponsive disease. In children with
nitions are in evolution. Adolescent and obese ALL who enter remission, marrow T1 relaxation
patients are in a poorer outcome group and leuko- time normalizes, whereas the marrow T1 relaxa-
cyte count at diagnosis is inversely related to tion time remains prolonged in those who do not
survival, whereas trisomy 21, APL subtype, and enter remission. However, the specificity of MR
early response to therapy are favorable factors.38 imaging is limited by the difficulty in differentiating
The prognostic significance of radiographic and viable neoplasm from effects of therapy, including
scintigraphic bone abnormalities is uncertain, relat- hematopoietic marrow regeneration (particularly
ing to conflicting reports in the literature. It has been with G-CSF or GM-CSF therapy), hematopoietic
reported that multiple bone involvement on radio- marrow reconstitution following stem cell trans-
graphs portends a shorter duration of remission plantation, marrow iron overload from transfu-
and survival39; that there is no correlation of radio- sional hemosiderosis, and marrow infarction and
logical or scintigraphic extent of bone disease and fibrosis.4446 Because of these limitations, MR
duration of remission or survival13,17; that children imaging has not replaced marrow aspirate or
without radiographic abnormalities have an aggres- biopsy for assessment of therapeutic response in
sive form of leukemia, whereas those with a few leukemia.47
bone lesions have an indolent form40; and that those
with radiographic bone lesions represent a subset
Surveillance for Relapse
with a better prognosis.33
Nephromegaly at the time of presentation is The most common site of ALL relapse is the
reportedly an adverse prognostic factor for marrow, followed by the CNS and testes. Isolated
ALL.41 Nephromegaly in childhood ALL is also marrow involvement occurs in 48% of cases of
correlated with subsequent renal damage detect- relapsed ALL in children at a median time of 26
able by renal MAG-3 or dimercaptosuccinic acid months, whereas the incidence of isolated CNS
(DMSA) scintigraphy.42 Overt testicular involve- relapse is less than 5% and the incidence of iso-
ment at diagnosis has been considered an lated testicular relapse is less than 2%. Outcome
adverse prognostic factor for ALL, but this may of ALL is poorer with early relapse and with iso-
no longer be the case with aggressive initial lated bone marrow relapse than with later relapse
therapy. Neither the presence of a mediastinal and combined marrow and testicular or CNS
mass at the time of diagnosis nor an incomplete relapse.48 Most AML relapses occur in the
response with a residual mediastinal mass on marrow, with CNS relapse being uncommon.
chest radiograph at day 35 or 70 of therapy Survival is substantially lower in those with shorter
predicts a worse prognosis in T-cell ALL.43 remissions, and relapsed leukemia is still the
primary cause of death in patients with AML.49
In some instances, relapse of leukemia in the
Therapy Response Evaluation
vertebral marrow can be detected by MR imaging
Imaging is not currently relied on to evaluate several weeks before relapse is detected by iliac
response to therapy for childhood leukemia. bone marrow aspirate or biopsy, reflecting the
However, some trends may be observed on patchy nature of relapsed leukemia and effects
skeletal radiographs. The transverse metaphyseal of sampling bias.50 Unlike the diffuse marrow
lucent bands usually resolve quickly with treat- abnormality typical of leukemia on MR imaging at
ment, whereas periostitis, cortical erosion, and presentation, early relapsed ALL can manifest as
lytic bone lesions resolve more slowly, and osteo- well-defined nodules of low signal intensity on
penia may worsen, related to steroids. Bone scle- T1-weighted sequences and high signal intensity
rosis and osteonecrosis may also develop after on T2-weighted and STIR sequences. In this
therapy. setting, directed marrow lesional biopsy may be
The high sensitivity of MR imaging for bone required to avoid false-negative iliac marrow
marrow abnormalities has led to investigation of sampling.51 Although time to relapse is an impor-
MR imaging as a method for therapy response tant predictor of outcome, evidence that early
evaluation. During chemotherapy for leukemia, detection of relapse by frequent surveillance
the bone marrow becomes hypocellular and ede- improves outcome is lacking.52
matous. Following chemotherapy, there is progres- Extramedullary involvement is more common at
sive regeneration of normal hematopoietic cells relapse than at presentation. Sanctuary sites for
and fat. Marked increase in the marrow fat fraction leukemic cells during therapy, where relapse can
is observed by chemical shift MR imaging in the occur even in the presence of bone marrow remis-
marrow of patients responding to chemotherapy, sion, include the CNS, testes, and kidneys. CNS
Leukemia and Lymphoma 773

prophylaxis has greatly reduced the incidence of childhood and adolescent/young adult forms of
CNS relapse, and surveillance imaging of the HL.54,55
CNS is not warranted. A possible exception is HL is characterized by a variable number of char-
EML, for which relapse is extramedullary in nearly acteristic clonal multinucleated giant cells (Reid-
40% of cases and most often in the CNS.36 Sternberg [RS] cells) in an inflammatory milieu.
Patients with suspected testicular relapse gener- The WHO classification separates the uncommon
ally go to biopsy without imaging, but occasionally nodular lymphocyte-predominant form of HL
ultrasound is requested and may show hypoe- (NLPHL) from the common form, designated
choic, enlarged testicles with or without focal classic HL. WHO subtypes of classic HL are nodular
lesions (Fig. 7). Isolated testicular relapse is rare sclerosis (NS), lymphocyte rich (LR), mixed cellu-
and testicular biopsy at the end of therapy has larity (MC), and lymphocyte depleted (LD).56 The
failed to show a survival benefit for patients with NS subtype of HL accounts for greater than 85%
early detection of occult disease,53 arguing of pediatric HL and is characterized by lymph nodes
against a role for surveillance imaging of the that have thickened capsules and dense collage-
testes. nous bands that separate the nodes into macro-
Surveillance chest radiographs are sometimes nodules. The presence of collagen and fibrous
obtained to evaluate for mediastinal relapse in stroma contributes to the presence of residual
patients with T-cell ALL with a prior mediastinal mediastinal soft tissue that is commonly seen early
mass. However, a beneficial impact of this prac- after completion of therapy, even after no viable
tice on outcome has not been established. disease remains. The MC subtype accounts for
30% of the cases in young children and can be
confused for NHL.
The characteristic RS cell in classic HL is
LYMPHOMA
believed to arise from preapoptotic germinal
Classification and Epidemiology
center B-cells that cannot synthesize immuno-
Pediatric lymphoma, including Hodgkin lymphoma globulin and show constitutive activation of the
(HL) and non-Hodgkin lymphoma (NHL), is the nuclear factor k-B pathway, conferring resistance
third most common malignant neoplasm in child- to apoptotic stimuli. EBV is associated with 15%
hood and adolescence. HL is unusual in patients to 25% of HL in developed countries and up to
younger than 4 years of age, and typically occurs 90% in developing countries, most commonly in
in older children and adolescents.3 In patients younger patients with MC histology. Despite this,
younger than 10 years of age, there is a male EBV serologic status does not seem to be a prog-
predominance of HL; beyond this age the relative nostic factor in pediatric patients with HL,57 in
incidence begins to equilibrate between the contrast with patients with NHL.
genders and patients older than 15 years of age NHL of childhood is a heterogeneous collection
have an approximately equal incidence between of lymphoid neoplasms that are not classified as
boys and girls.54 There are some studies suggest- HL. Although a large number of forms of NHL are
ing an association between chronic Epstein-Barr recognized, the 4 most commonly occurring in
virus (EBV) and HL and this has led some investi- children include Burkitt lymphoma (BL), diffuse
gators to propose a distinction between the large B-cell lymphoma (DLBCL), anaplastic large

Fig. 7. Testicular relapse of precursor B-cell ALL in a 3-year-old boy manifests as testicular enlargement by ill-
defined masses on ultrasonography.
774 Guillerman et al

cell lymphoma (ALCL), and precursor T-cell and B- diagnosed with HL is around 95%.7 This high
cell lymphoblastic lymphoma (LL). In contrast with cure rate has led to renewed interest in the role
HL, NHL is more common in children younger than that treatment-related toxicities and long-term
10 years of age. There is also a male predomi- consequences of therapy play in overall morbidity
nance, particularly in older patients.58 Age- and mortality.63,64 The risk of death caused by
specific incidence also varies according to disease disease at 20 years from diagnosis almost equals
classification, with LL occurring with a fairly the risk of death from other causes, including
constant rate across all age groups, whereas treatment-related effects.63
ALCL and DLBCL predominate in older adoles- In the 1960s and 1970s, extended field radiation
cents. Although the cause of NHL is uncertain, therapy improved disease-free and overall survival
there is an increased incidence of lymphomas in rates amongst patients with HL.55 However, this
immunosuppressed patients. Other studies have so-called mantle radiation resulted in significant
shown a role for EBV in the pathogenesis of lym- late effects in the irradiated tissues. Subsequent
phoproliferative disease and NHL. Taken together, development of a combination of chemotherapy
these findings suggest that disordered immuno- regimens showed that disease-free survival could
regulation, with resultant clonal proliferation of be improved with lower dose radiation therapy
immature cells that have failed to differentiate, regimens, and, in certain patients with HL, elimina-
contributes to malignant transformation in NHL.59 tion of radiation therapy altogether. In patients with
Each NHL subtype has characteristic pathologic low-stage HL, overall survival was no different for
features, and recent molecular and translational patients whose initial therapy was chemotherapy
investigations have led to new understanding of alone, because of effective salvage regimens.65,66
the pathobiology of NHL.54,59 BL and DLBCL are In patients with advanced-stage HL, EFS was
believed to derive from lymph node germinal center higher for those who received initial chemotherapy
regions where proliferating B-cell lymphoblasts and radiation therapy compared with those with
normally differentiate. The activation of proto- chemotherapy alone.54,67 As with current standard
oncogenes and/or disruption of tumor suppressor regimens, these early treatment regimens were
genes or hypermutation of proto-oncogenes are risk adapted: those with favorable-risk disease,
believed to result in the malignant transformation defined by low stage and low bulk disease,
of these germinal center lymphoblasts. Consistent typically receive 2 to 4 cycles of multiagent che-
with this interpretation, B-lineage cell surface motherapy with either low-dose involved field radi-
antigen CD20 shows increased expression on ation or no radiation, whereas those patients with
both BL and DLBCL. ALCL shows expression of higher risk disease are stratified to receive more
CD30 and is characterized by overexpression of intensive chemotherapy before involved field
the anaplastic lymphoma kinase (ALK) tyrosine radiotherapy.55
kinase, which is believed to play a role in ALCL These risk-adapted approaches do not take into
tumor genesis. LL, in contrast with the other pedi- account initial disease response, in contrast with
atric NHL subtypes, predominantly arises from response-adapted approaches, in which the over-
immature T-cells, corresponding to defined stages all treatment intensity is modulated during the
of thymocyte differentiation. Less than 10% of LL is course of therapy based on initial response. This
of B-cell origin.58 When greater than 25% of the latter approach is emerging as a potential means
bone marrow is infiltrated with lymphoblasts, the of further reducing therapy and potentially reduc-
disease is termed ALL rather than LL. Posttrans- ing late effects for those patients with HL in
plant lymphoproliferative disease (PTLD) is seen whom cure is likely, while maintaining high cure
in the setting of both solid organ and bone marrow rates and aggressive treatment of those patients
transplantation, and results directly from host at higher risk of relapse.64,67
immunosuppression.60,61 PTLD, which is usually Patients with NHL have lower overall survival
EBV-related, is not initially classified as a malignant rates compared with HL. With current treatment
lymphoma and frequently responds to reduction in approaches, more than 85% of children and 75%
immunosuppression. However, the lymphoproli- of adolescents with NHL survive at least 5 years.7
ferative disease may progress to an aggressive Treatment of childhood NHL depends on localized
B-cell lymphoma, resulting in widespread malig- versus disseminated disease. Localized disease is
nant disease.62 typically defined as stage I or II disease, whereas
stage III or IV disease is generally considered to
be disseminated. In most children, NHL is widely
Treatment
disseminated from the outset, and systemic treat-
With current treatment approaches, the 5-year ment with aggressive combination chemotherapy
survival rate for children and adolescents is usually recommended for most patients.54,58
Leukemia and Lymphoma 775

Children with refractory or relapsed NHL have The diagnostic evaluation should always include
a worse outcome than newly diagnosed patients, imaging of the chest and neck up to the level of
and thus aggressive up-front therapy and early the Waldeyer ring. Large neck and mediastinal
remission remain the goal of new treatment masses may compress the airway and central
regimens.68 vascular structures. Care must therefore be taken
The outcome for LL is excellent, with longer leu- in sedating these patients before imaging.71 After
kemialike therapy consisting of induction, consoli- a careful physiologic and radiographic evaluation
dation, and maintenance therapy. In contrast, of the patient has been performed, the least inva-
nonlymphoblastic NHL has superior outcome sive procedure should be used to establish the
with short, intensive pulse therapy. For recurrent diagnosis. If possible, peripheral lymph node
or refractory B-lineage NHL or LL, survival is low biopsy is preferable. Aspiration cytology is not
(10%20%), emphasizing the importance of sufficient because of the absence of stromal
achieving cure during the initial therapy. For recur- tissue, and core needle biopsies are necessary.
rent or refractory ALCL, as many as 60% of Surgical staging has been largely replaced by
patients can ultimately be salvaged and achieve imaging; however, mediastinoscopy or thoraco-
long-term survival.69 PTLD can involve multiple scopy may be needed when other modalities fail
organs and systems, and responds variably to to establish the diagnosis or if questionable areas
conventional lymphoma therapies.6062 of involvement result in upstaging the patient and
Radiation therapy plays little role in the routine histologic confirmation is required.
management of pediatric NHL, in contrast with Lung involvement is seen in less than 5% of chil-
HL.58 Mediastinal radiation is not commonly used dren younger than 10 years of age and 15% of
for patients with mediastinal masses, except in the adolescents with HL.70,72 Nodules greater than 1
emergent treatment of symptomatic superior vena cm are the most common pulmonary finding during
cava obstruction or airway obstruction. Even in staging of patients with HL, although diffuse intersti-
this instance, low-dose radiation is usually used. tial thickening and lobar or segmental consolidation
are other pulmonary manifestations of disease. The
Imaging Features presence of pulmonary disease usually occurs in
association with ipsilateral hilar or mediastinal
In HL, chest radiographs obtained for upper respi- lymphadenopathy (Fig. 10). The most common
ratory symptoms and/or vague constitutional mechanisms of disease spreading into the lungs
symptoms, such as fever or night sweats, often are hematogenous and lymphangitic spread, and
prompt the initial diagnosis. At the time of diag- less frequent, direct invasion. Pleural and pericardial
nosis, a mediastinal mass is present in more than effusions are infrequent findings in HL and usually
two-thirds of patients with HL (Figs. 8 and 9).70 result from lymphatic obstruction.70 Pleural effu-
sions are typically transudative and usually negative
for malignant cells. Pericardial effusion, when
present, may suggest tumor involvement of the peri-
cardium from direct extension of the adjacent medi-
astinal mass. Where pericardial involvement is
suspected, MR imaging may be superior to CT,
particularly with the advent of respiratory and
cardiac gated MR imaging.
Liver involvement by HL is almost always
associated with splenic involvement, and splenic
involvement without associated para-aortic lym-
phadenopathy is unusual (Fig. 11). Splenic in-
volvement occurs in 30% to 40% of Hodgkin
disease (HD). Splenic size is unreliable for predict-
ing splenic HD involvement.73
At diagnosis, bone marrow involvement is
unusual in HL. MR imaging and [18F]fluorodeoxy-
glucose (FDG)positron emission tomography
Fig. 8. An 18-year-old patient with HL. On the upright (PET) are more sensitive than conventional CT for
posteroanterior (PA) chest radiograph, the transverse detecting bone marrow involvement.72 Cortical
width of the mediastinal mass exceeds one-third of bone involvement is similarly rare in HL. When
the thoracic diameter, meeting the criterion for bulk present, lesions are typically lytic and may have
disease. accompanying periosteal reaction.70
776 Guillerman et al

Fig. 9. Chest CT images of an 18-year-old girl (see chest radiograph in Fig. 8) with HL depict characteristic
conglomerate lobular mediastinal lymphadenopathy.

Historically, gallium (67Ga) scintigraphy was the have access to FDG-PET scanning and there is
mainstay of functional imaging in lymphoma but currently little justification for gallium scanning.
has now usually been replaced by FDG-PET. FDG-PET has been studied extensively in adult
FDG-PET has been shown to be more sensitive lymphoma77 and, to a lesser extent, in pediatric
than gallium scintigraphy for determining lung, populations.72,78 The overall consensus from
bone, and nodal involvement and has a much lower multiple studies is that FDG-PET is more sensitive
radiation dose than gallium scintigraphy.7476 than CT for involvement of normal-sized lymph no-
Gallium has prolonged retention in the bowel, des and extranodal disease, including the spleen,
further limiting its usefulness in evaluating intra- liver, and bone marrow (see Fig. 11; Fig. 12).7984
abdominal disease. Although gallium scintigraphy Combined FDG-PET/CT imaging retains the high
approaches the sensitivity of FDG-PET for diag- sensitivity of FDG-PET for detecting disease, but
nosis and staging of neck and mediastinal/chest improves the specificity, with coregistered fused
nodal disease in pediatric HL, nearly all institutions images resulting in the highest sensitivity and
specificity at detecting disease and correlating
sites of abnormal FDG uptake with specific
anatomic regions.80,82,85,86
False-positive FDG uptake is well recognized
and can result from rebound thymic hyperplasia,
hypermetabolic brown fat, muscle, hyperplastic/
recovering marrow and/or spleen, gonadal and
breast cyclic hormonal stimulation, and sites of
recent surgery or infection (Fig. 13).87,88 The char-
acteristic patterns of nonspecific uptake are well
recognized by experienced radiologists and nu-
clear medicine physicians and the use of coregis-
tered FDG-PET/CT has proved invaluable for
identifying and eliminating areas of false-positive
uptake from the diagnostic evaluation.88 In some
instances, background uptake can be reduced.
For example, brown fat uptake can be largely elim-
inated by warming the patient, and, in challenging
cases, premedicating the patient with fentanyl
and/or benzodiazepines.89
Among pediatric NHL, BL most commonly
presents with intra-abdominal visceral disease,
and widespread extranodal involvement is often
present. The initial imaging evaluation is usually
directed at assessment of symptoms referable to
abdominal involvement. Involvement of the perito-
neum, solid abdominal organs, and bowel with
Fig. 10. A 14-year-old girl with HL. CT images show complicating intussusception can be seen (Figs.
pulmonary, mediastinal, and bilateral hilar involvement. 14 and 15).
Leukemia and Lymphoma 777

Fig. 11. Axial and coronal contrast-enhanced CT images of the chest and abdomen, respectively, show a large
mediastinal mass with accompanying diffuse splenic involvement and mesenteric lymph node enlargement in
this patient with HL. The accompanying FDG-PET image shows diffuse FDG uptake throughout the mass in
both the chest and abdomen.

As many as 75% of patients with LL present with and bone,54 and often presents with extensive
a mediastinal mass and dyspnea, wheezing, multifocal disease (Figs. 17 and 18).
stridor, or dysphagia.58 The mediastinal enlarge- DLBCL has a less characteristic clinical pattern
ment in LL is attributable to diffuse thymic involve- compared with other NHL subtypes. Most patients
ment and does not show the typical nodular with DLBCL present with localized disease. Anterior
heterogeneous appearance more characteristic mediastinal and/or bulky cervical/supraclavicular
of HL and other forms of NHL (Fig. 16). These lymphadenopathy are more characteristic of DLBCL
tumors grow rapidly and patients with LL may than of the other NHL subtypes. Up to 70% of these
deteriorate quickly because of airway compres- patients have a mediastinal mass, which may
sion and impairment of venous return, particularly produce airway obstruction and superior vena
when patients are placed supine or sedated. In this cava syndrome (Fig. 19).54,72 In general, these
population of patients, diagnostic imaging may not tumors are more aggressive than HL and malignant
be possible because of the tenuous clinical status pericardial effusions resulting from direct pericardial
of the patient, and pleural fluid and/or bone invasion, malignant pleural effusions, and pulmo-
marrow aspiration may be the only means of accu- nary involvement may be seen (Fig. 20). Patients
rately diagnosing these patients. with DLBCL with disease localized to the medias-
ALCL is often associated with systemic symp- tinum may be difficult to distinguish from HL based
toms and signs such as fever and weight loss, on imaging (see Fig. 19).54 The presence of meta-
and a prolonged waxing and waning course before chronous peripheral lymphadenopathy or bone
diagnosis. A mediastinal mass may be seen in up involvement makes DLBCL the more likely diag-
to 40% of patients with ALCL, and, when present, nosis. About 20% of pediatric patients with DLBCL
is often accompanied by large pleural and pericar- present with primary mediastinal disease (primary
dial effusions. ALCL can also involve lung, skin, mediastinal B-cell lymphoma).90 This presentation
778 Guillerman et al

Fig. 12. Axial CT, coronal FDG-PET, and fused axial PET/CT show extensive FDG-avid HL. Retroperitoneal lymph
node involvement was shown by FDG-PET and PET/CT fusion, whereas the same site shown on the CT alone could
have been interpreted as unopacified bowel. This patient is the same 14 year old who is shown in Fig. 9, who also
had extensive pulmonary nodules, confirmed as FDG-avid sites of disease.

is more common in older children and adolescents. of disease that indicate hematogenous spread.
These tumors are more aggressive and are associ- Unique to HL is the designation of extralymphatic
ated with a worse outcome compared with other disease that results from direct extension of an
pediatric large B-cell lymphomas. Growth into adja- involved lymph node region, which can be chal-
cent structures is common and there is a character- lenging at the time of staging. For example, contig-
istic tendency for focal involvement of the kidneys uous involvement of lung adjacent to a large
(see Fig. 20). mediastinal mass may be considered stage IIE
PTLD can involve multiple organs or have focal rather than stage IV disease. However, the presence
involvement. The use of FDG-PET to stage the of a malignant pleural effusion that is cytologically
extent of disease and assess response to therapy positive for HL would be considered stage IV. For
in PTLD is increasingly advocated (Fig. 21).62,91 areas of questionable noncontiguous extralym-
phatic involvement, pathologic confirmation may
be required before assignment to stage IV. The pres-
Staging and Risk Stratification
ence of B symptoms (fever, night sweats, weight
The Ann Arbor system for staging of HL was devel- loss) is included in the staging of the patient, and
oped to classify anatomic sites of disease based influences whether the patient is assigned to a low
on a combination of clinical, surgical, and imaging or intermediate/high-risk treatment regimen.
findings. The Cotswold modifications of the Ann The posteroanterior (PA) upright chest radio-
Arbor staging system incorporated the prognostic graph is still used for determining the presence of
implications of tumor bulkiness and number of bulk disease (mediastinal mass > one-third of the
disease sites into the staging system.92 The revised maximal transthoracic diameter) (see Fig. 8).55
Ann Arbor staging system is shown in Box 1. However, the Cotswolds modification of the Ann
Staging is largely based on identifying disease Arbor classification also defines lymph nodes
above or below the diaphragm and at identifying greater than 10 cm in maximal dimension on CT
noncontiguous involvement of extralymphatic sites imaging as bulky. Despite various attempts, the
Leukemia and Lymphoma 779

Fig. 13. A 15-year-old patient with stage IV HL. Baseline FDG-PET and CT show extensive disease, including pulmo-
nary lesions. At the end of therapy, FDG uptake has resolved, except for presumed background brown fat uptake
in the neck. Residual CT abnormalities were interpreted as scar tissue in view of the negative FDG-PET scan. By 4
months after completion of therapy the patients pulmonary relapse was obvious by CT and confirmed by FDG-
PET. Rx, treatment.

Fig. 14. Sporadic BL has a proclivity for widespread extranodal involvement, as illustrated by the presence of
masses in the pancreas and right kidney on an abdominal CT image (A), proximal right tibial bone marrow on
an T1-weighted MR image (B), and lumbosacral epidural space on a sagittal STIR MR image (C) in this 8-year-
old boy.
780 Guillerman et al

Fig. 15. An 8-year-old girl with BL. Coronal CT reconstruction shows thickening of the distal ileum and cecum
(arrows). Two days later, the patient presented with abdominal pain. An ultrasound examination showed an
intussusception. A contrast enema confirmed the intussusception, which was successfully reduced. When the
intussusception recurred, she was taken to the operating room where the diagnosis of BL was established.

current definitions of bulky disease are not stan- as defined by PA chest radiograph and CT of nodal
dardized and frequently depend on the clinical trial disease, is still recognized in the risk stratification of
protocol and the cooperative group from which patients enrolled in Childrens Oncology Group
the protocol derives. For example, bulky disease, trials and St Jude Consortium trials. However,
results from the German-Austrian Pediatric Multi-
center trial suggest that bulk disease alone is not
a prognostic factor for outcome with a risk-
adapted treatment strategy.93 Contrast-enhanced
CT and FDG-PET imaging complete the contribu-
tion of imaging to staging and risk stratification.
There is currently no role for bone scintigraphy in
the routine staging evaluation.94 Increasingly,
particularly in Europe, the use of MR imaging,
including diffusion-weighted imaging, is being
advocated to reduce radiation exposure in these
heavily imaged and treated patients.95,96
Combined FDG-PET/CT imaging provides the
most sensitive and specific means of accurately
Fig. 16. Diffuse infiltration of the thymus resulting in
a smoothly marginated homogeneous anterior medias- staging patients with HL (see Figs. 1113). For
tinal mass that compresses the airway and occludes the example, in one study of children and adolescents
left brachiocephalic vein or superior vena cava is with HL, FDG-PET changed the staging in 15% of
a common presentation of T-cell lymphoblastic lym- the patients, most of whom were upstaged. Most
phoma, as shown in this chest CT image. of the false negatives not detected by FDG-PET
Leukemia and Lymphoma 781

Fig. 17. An 11-year-old patient with paraspinal ALCL. CT and MR images show paraspinal mass with bone destruc-
tion and invasion of the spinal canal. An MR imaging and FDG-PET scan obtained while the patient was on
therapy showed a decrease in size of the mass. Although the patient did not have a baseline FDG-PET scan,
the presence of FDG uptake after completion of consolidation chemotherapy suggested residual active disease.
Rx, treatment.

had tiny pulmonary nodules shown by chest CT.97 is based on tumor burden, and has served clini-
Most discordance between CT and FDG-PET cians well for many years. In the past, even with
occurs at extranodal sites, such as the lung, in less effective chemotherapy, the St Jude system
which CT is superior for tiny nodules, and the provided a sound basis for treatment stratification.
spleen and bone marrow, in which FDG-PET is Children with NHL, in contrast with HL, frequently
superior.98 The highest diagnostic accuracy is present with disseminated disease. As with HL,
therefore achieved using a combination of FDG- CT scanning is most commonly used for the initial
PET and CT. staging of NHL. For specific sites of bone or CNS
Despite these advances in functional imaging, disease, MR imaging is used. FDG-PET detects
staging with conventional imaging modalities (CT additional disease in a small proportion of pedi-
and/or MR imaging) alone as the standard initial atric patients with NHL (see Figs. 17, 18 and 20;
staging procedure for risk and treatment stratifica- Figs. 22 and 23), but it has not been shown to
tion has historically been sufficient to achieve cure detect additional sites of disease that would result
rates greater than 90% in pediatric HL. Therefore, in frequent alterations of patient stage, nor has it
the ultimate impact of additional whole-body been generally shown to result in any modification
imaging with either MR imaging or FDG-PET, of treatment.100 High cure rates result from risk-
although promising, may be modest in terms of adapted intensive chemotherapy without radiation
changing overall cure rates of pediatric HL. therapy, and it has not been necessary to map
However, the incorporation of FDG-PET/CT find- precisely every small site of nodal disease, be-
ings into radiation treatment planning may be signif- cause patients are being treated intensively and
icant in treatment-related toxicity, by guiding systemically.58 As a result, in contrast with the
treatment dose and target treatment volume.55,63,99 adult situation, FDG-PET is not routinely included
One study found that involved field radiation therapy in the diagnostic staging of childhood NHL.101
(IFRT) volumes needed to be adjusted from FDG- However, NHL is often disseminated at the time
PET findings in 70% of pediatric patients with HL.98 of initial disease presentation and, in this setting,
The St Jude (Murphy) classification is used for early response assessment may be more useful
the staging of pediatric NHL (Box 2). This system in predicting ultimate patient outcome rather than
782 Guillerman et al

Fig. 18. A 12-year-old boy with ALCL. He initially presented with testicular pain, swelling, and left testicular
enlargement. Ultrasonography (not shown) revealed heterogeneous testicular echogenicity, but no focal mass;
testicular biopsy was negative for malignancy. The CT scan shown here, obtained for subsequent abdominal
pain, reveals diffuse mesenteric and retroperitoneal lymphadenopathy. Occlusion of the inferior vena cava and
obliteration of the left renal vein likely accounted for the initial left testicular complaints. FDG-PET scan shows
uptake in the primary mass, as well as a left supraclavicular site. No uptake was present in the scrotum.

extent of disease and overall tumor burden at (see Figs. 17 and 18). Sites of involvement that are
diagnosis (see Fig. 23). For these patients, diag- unusual in childhood lymphoma, such as skin,
nostic FDG-PET imaging is obtained to provide lung, and bone, are common in ALCL. LL is another
a baseline for subsequent response assessment. example of the limitations of the NHL staging
ALCL is an example of disease distribution not system. Most patients are stage III, with few pre-
fitting well into the St Jude NHL staging system senting with either stage I or stage II disease.
Furthermore, the outcome of patients with stage
IV disease (usually the result of bone marrow
involvement), differs little from those with stage III
disease. Because of the overall excellent response
of these patients, features such as tumor bulk,
pleural effusion, and respiratory obstruction do
not ultimately influence overall outcome.54,58 As
with LL, BL responds rapidly to aggressive che-
motherapy.58 Patients with localized disease have
an excellent outcome after a short course of aggres-
sive chemotherapy, and, with improvements in
treatment, even patients with advanced-stage
disease at diagnosis have an excellent overall
outcome. Although FDG-PET scanning may be per-
formed during staging of patients with BL, given the
Fig. 19. Primary mediastinal large B-cell lymphoma speed with which these tumors grow and enlarge,
often assumes a lobular morphology like HL and tends there is little evidence that functional imaging
to occlude the superior vena cava, as shown on this affects staging or outcome in this population of
chest CT image. patients.101
Leukemia and Lymphoma 783

Fig. 20. (A) Axial CT images in a patient with mediastinal DLBCL, showing mediastinal mass, near occlusion of the
brachiocephalic vein, tracheal narrowing, pulmonary metastases, and bilateral renal lesions. (B) FDG-PET shows
uptake in the mediastinal mass, pulmonary lesions, and renal lesions. In addition, FDG-PET clearly shows 2 foci
of disease in the pancreas that are difficult to resolve by CT.

Therapy Response Evaluation and determining initial disease bulk in an effort to


stratify patients into treatment groups. Early
Initial efforts to develop objective measurement
criteria for response were also developed, and
criteria for assessing solid tumors were put
led to categories ranging from complete response
forward by the WHO and used bidirectional
(CR) to progressive disease, and included stable/
measurement techniques.102 These measurement
no change or partial response (PR) in the classifi-
techniques focused primarily on disease staging
cation scheme, depending on the estimates of
784 Guillerman et al

Fig. 21. A 12-year-old heart transplant recipient with fever and increased EBV titers. CT showed extensive lymph
node enlargement in the retroperitoneum, supraclavicular regions, and mediastinum, as well as splenic involve-
ment, consistent with PTLD. Disease did not respond to reduction in immunosuppression. FDG-PET confirmed
extensive FDG-avid PTLD. One month after chemotherapy there is no residual disease.

change in tumor size. The WHO criteria presented as an in vivo chemosensitivity test, even when
many challenges, particularly for tumors such as significant changes in tumor volume or complete
HL, which frequently leave measurable residual resolution of the tumor mass are not yet seen on
posttreatment scar tissue. Minimum lesion size morphologic imaging (Fig. 24). Studies showing
and numbers of lesions to be recorded were not that significant reductions in tumor volume and
specified and, depending on the location of tumor, FDG-PET negativity are associated with favorable
size measurements of lymph nodes may have outcome in both pediatric HL and NHL suggest
been based on physical examination estimates. that rapid and homogeneous cytotoxic chemo-
In addition, these methods were devised before therapy responsiveness may lead to improved
the advent of the multiplanar cross-sectional disease-free survival.103
imaging techniques that are in common use today Several studies of FDG-PET for response as-
(CT and MR imaging), and underestimates of sessment in pediatric lymphoma have been
disease burden and the choice of measurement reported.86,104107 The use of FDG-PET in assess-
technique often led to errors in establishing ing response during therapy for tumors that are
disease progression or response. FDG avid at the time of staging is being investigated.
Developing objective measures of treatment In one study of pediatric and young adult patients
response is essential to having evaluable prospec- with HL or NHL, the negative predictive value of
tive end points in early phase clinical trials and FDG-PET during therapy was 96%, whereas the
determining whether new agents warrant further positive predictive value was 100%.86 This obser-
testing. The challenge is developing surrogate vation, substantiated by other studies, suggests
end points that accurately reflect the disease that interim FDG-PET scanning during therapy is
process and response to therapy at a time when an excellent prognostic indicator for predicting clin-
other indicators of response (eg, change in clinical ical outcome.
status) may not reflect treatment response. For An ongoing major European study in childhood
example, a brisk response to chemotherapy is an and adolescent HL is evaluating the role of interim
indirect determinant of biologic homogeneity FDG-PET in determining the need for involved field
within the tumor, which, in turn, translates into radiotherapy in patients who have a good early
more uniform chemosensitivity across the entire response to induction chemotherapy (ie, those
tumor volume. As a result, changes in FDG uptake patients who are in complete remission or in partial
that occur soon after the initiation of therapy serve remission based on CT, but FDG-PET negative).100
Leukemia and Lymphoma 785

Box 1 Box 2
Modified Ann Arbor staging system for St Jude staging system for childhood NHL
childhood HL
Stage I: a single tumor (extranodal) or single anatomic
Stage I: involvement of single lymph node region (I) or site (nodal), excluding mediastinum or abdomen
localized involvement of a single extralymphatic
Stage II: a single tumor (extranodal) with regional
organ or site (IE)
node involvement; 2 or more nodal sites on the same
Stage II: involvement of 2 or more lymph node regions side of the diaphragm; 2 single (extranodal) tumors
on the same side of the diaphragm (II) or localized with or without regional node involvement on the
contiguous involvement of a single extralymphatic same side of the diaphragm; a primary
organ or site and its regional lymph node(s) with gastrointestinal tract tumor, with or without
involvement of 1 or more lymph node regions on associated mesenteric nodes, grossly completely
the same side of the diaphragm (IIE) resected
Stage III: involvement of lymph node regions on both Stage III: 2 single tumors (extranodal) on opposite
sides of the diaphragm (III), which may also be sides of the diaphragm; 2 or more nodal areas above
accompanied by localized contiguous involvement of and below the diaphragm; primary intrathoracic
an extralymphatic organ or site (IIIE), by involvement tumors (mediastinal, pleural, thymic); extensive
of the spleen (IIIS), or both (IIIE1S) primary intra-abdominal disease, unresectable;
paraspinal or epidural tumors
Stage IV: disseminated (multifocal) involvement of 1
or more extralymphatic organs or tissues, with or Stage IV: any of stages IIII with initial CNS or bone
without associated lymph node involvement, or marrow involvement (<25%)
isolated extralymphatic organ involvement with
distant (nonregional) nodal involvement

Anatomic lymph node regions for the purpose of HL


staging are Waldeyer ring, cervical/supraclavicular/ NHL.77,108 Although the focus of this project was
occipital/preauricular, infraclavicular, axillary/pectoral,
epitrochlear/brachial, mediastinal, hilar, splenic/splenic
adult lymphoma, the criteria proposed can, for
hilar, mesenteric, para-aortic/celiac/periportal/retro- the most part, be applied to pediatric lymphomas.
crural,iliac, inguinal/femoral, and popliteal. A summary of these criteria is shown in Table 1. As
before, measurable extranodal disease should be
The designation A is for asymptomatic disease and B is
for the presence of unexplained fever, weight loss, or assessed in a manner similar to that for nodal
night sweats. The designation E is for minimal extra- disease. For HL, the spleen is still considered
lymphatic disease from direct extension of an involved a site of nodal disease (see Fig. 11). Disease loci
lymph node region, originally devised to indicate ex- that are assessable but not measurable (eg,
tralymphatic disease limited enough to be subjected
pleural effusions and bone lesions) are recorded
to definitive treatment by radiation therapy. The
designation E is not appropriate for cases of wide- as present or absent unless it is proved by biopsy
spread or diffuse extralymphatic disease (eg, a large to be negative. The major change to the response
pleural effusion that is cytologically positive), which criteria is the emphasis placed on FDG-PET in
should be considered stage IV. determining response.
The most significant change of the IHP criteria
from the earlier criteria used to determine re-
This represents an early effort to incorporate a sponse is the definition of CR. CR is defined by
response-based treatment algorithm into ongoing the IHP as disappearance of all evidence of
clinical trials of pediatric lymphoma patients. Most disease, but is now primarily based on FDG-PET.
of the data have been obtained after 2 cycles of In patients with typically FDG-avid lymphomas
chemotherapy, although there is no evidence to (nearly all patients with HL and most patients
suggest that a response evaluation after 2 or 3 with NHL), a posttreatment residual mass of any
cycles is either superior or inferior to that performed size is permitted by CT or MR imaging, as long
after 1 cycle. A very early response to therapy after 1 as it is FDG-PET negative (see Fig. 24); this is
cycle may be more predictive than responses true for patients in whom the FDG-PET was posi-
measured after more prolonged therapy. tive before therapy, and even in patients with no
As noted earlier, the use of two-dimensional pretreatment FDG-PET scan, provided they have
measurement techniques does not account for typically FDG-avid lymphomas (such as HL). Vari-
functional and metabolic changes in the tumor. ably FDG-avid lymphomas or those in which FDG
The International Harmonization Project (IHP) avidity is unknown must have their diseased lymph
was convened to address this issue in adult nodes and nodal masses regress to normal size
lymphoma and issued revised criteria in 2007 for (not >1.5 cm in greatest transverse diameter, or
complete remission, partial remission, progressive not more than 1.0 cm in short axis for lymph nodes
disease, and stable disease (SD) both for HL and less than 1.5 cm in size at diagnosis). These
786 Guillerman et al

Fig. 22. A 16-year-old patient with DLBCL. Axial CT and FDG-PET show uptake in the mediastinal mass and
multiple diaphragmatic lymph nodes. An additional focus of disease in a small aortocaval lymph node (arrow)
would not have been detected without the use of FDG-PET.

criteria have been developed for adults but should and spleen. Splenic/hepatic nodules must regress
be applicable for most pediatric patients, although by not less than 50% in size. If bone marrow was
some clinical judgment is necessary in cases with involved before therapy and clinical CR was ac-
borderline enlarged lymph nodes. Splenic and/or hieved by other criteria, but there is persistent
liver involvement, either as diffuse enlargement marrow involvement, the patient is still considered
or focal nodules, should return to normal size a partial responder. For partial responders, post-
and nodules should disappear. However, the treatment FDG-PET should be positive in at least
determination of splenic involvement may be chal- 1 previously involved site. If FDG avidity at base-
lenging and multiple imaging modalities may be line is unknown, then the CT criteria are used.
used to unequivocally establish the presence or SD is defined by the IHP as failure to attain either
absence of residual abdominal visceral disease CR or PR, but not meeting criteria for progressive
(see Fig. 24). Bone marrow involvement, if present, disease. For FDG-avid lymphomas, the FDG-PET
must have cleared based on repeat bone marrow scan should be positive at prior sites of disease
biopsy. Residual bone marrow abnormalities by with no new areas of involvement on the posttreat-
imaging (ie, FDG-PET or MR imaging) should be ment CT or FDG-PET. If FDG-PET is not available,
confirmed by biopsy in order for a patient to be there must be no change in the size of the previous
considered in CR. lesions on the posttreatment CT scan.
PR is defined by the IHP as regression of The appearance of any new lesion at the end of
measurable disease with no new sites of disease. therapy should be considered relapsed (after CR)
There must be at least a 50% decrease in the sum or progressive disease (after PR or SD) according
of the product of the diameters of up to 6 largest to the IHP unless otherwise confirmed by histo-
dominant nodes or nodal masses, which should logic evaluation. Increased FDG uptake at a previ-
be clearly measurable in 2 dimensions. There ously unaffected site or at a previously involved
should be no increase in size of other nodes, liver, site that had responded should also be considered
Leukemia and Lymphoma 787

Fig. 23. An 8-year-old patient with hip pain. MR imaging shows diffuse marrow replacement on T2-weighted and
T1-weighted images. CT shows extensive visceral and intra-abdominal involvement. FDG-PET confirms diffuse
abnormal uptake throughout the abdomen and bone marrow. Bone marrow aspirate showed greater than
25% infiltration of the marrow by Burkitt cells, indicating Burkitt leukemia.

Fig. 24. This patient with HL received 2 4-week cycles of therapy. Compared with the baseline examination (see
Fig. 11) there is still residual mediastinal soft tissue abnormality, although decreased from the baseline examina-
tion. The spleen is now normal in size; however, punctate hypodensities are still present in the spleen. Despite
these residual CT findings, the accompanying FDG-PET shows complete resolution of abnormal FDG uptake in
the mediastinal mass and in the abdomen.
788 Guillerman et al

is too small to be detected by PET (<1.5 cm in long


Table 1
Summary of new Harmonization Project axis, or 1.0 cm with newer scanners).
criteria for PET and CT in determining Restaging at completion of therapy uses the
response in lymphomaa same response criteria used for early interim
response assessments, with CR reserved for
Response Criteria those patients with absence of disease by clinical
CR FDG-PET completely negative examination and imaging studies. As before,
Residual lymph nodes/nodal residual abnormalities on cross-sectional imaging
masses allowed, if FDG are allowed, provided they remain FDG negative
negative and no new sites of FDG activity are identified.
Bone marrow biopsy negative Currently, visual assessment is considered
Splenic/liver involvement must adequate for interpreting FDG-PET findings as
disappear positive or negative when assessing response
No new sites of disease after completion of therapy. The use of standard-
PR PDG positivity should be ized uptake value (SUV) measurements is still
present in at least 1 experimental.109 The use of mediastinal blood
previously involved site pool activity is currently recommended as the
Regression of measurable
reference background activity to define FDG-PET
disease; no new sites of
disease positivity for residual masses,108,109 although this
50% decrease in SPD of 6 can be challenging (Fig. 25), and a recent review
dominant LNs/nodal masses emphasizes the importance of establishing stan-
50% reduction in splenic/ dards for background activity to avoid stage
hepatic nodules, if present migration purely as a result of improvements in
Even if CR by other criteria, technology.110 The use of FDG-PET and functional
positive bone marrow response assessment has clearly revolutionized
biopsy is considered PR the response evaluation in both HD and NHL.
SD Failure to achieve PR, but not Studies have shown that tumor responsiveness
meeting PD criteria as determined by FDG-PET activity after 1 to 2
PD/relapse Any lesion increased in size by cycles of treatment can identify patients at
50% from nadir increased risk of relapse. However, studies are still
Any new lesion ongoing or in development in which early FDG
PET should be positive in new/ response assessment is or will be used to dictate
progressed lesions if
treatment approach.
1.5 cm
If it is accepted that FDG-PET imaging is a valu-
Notes: New criteria include PET in definition of CR. PET able surrogate biomarker for assessing a response
considered positive if uptake is greater than mediastinal to therapy in pediatric lymphoma, then the next
blood pool (lesions >2 cm), or more than local background challenge is how to use this surrogate to effectively
(lesions <2 cm). guide therapy. The goal is to move from a risk-
Abbreviations: CR, complete response; LNs, lymph no-
des; PD, progressive disease; PR, partial response; SD, stable adapted prediction of outcome based on staging
disease; SPD. and baseline symptoms to a response-directed
a
Based on work from the IHP. treatment paradigm.55 To validate functional im-
Data from Juweid ME, Stroobants S, Hoekstra OS, et al. aging as a useful biomarker of response, the next
Use of positron emission tomography for response ass-
era of clinical trials in pediatric lymphoma will likely
essment of lymphoma: consensus of the Imaging Sub-
committee of International Harmonization Project in require that treatment decisions be made based
Lymphoma. J Clin Oncol 2007;25:57178; and Cheson BD, solely on functional imaging biomarker findings,
Pfistner B, Juweid ME, et al. Revised response criteria for irrespective of residual abnormalities seen on
malignant lymphoma. J Clin Oncol 2007;25:57986. conventional cross-sectional imaging. There will
be challenges. At least 1 randomized study has
shown that the addition of radiation therapy in
relapsed or progressive disease (see Fig. 13). patients with bulky HL and FDG-negative postche-
Lymph nodes are considered abnormal if their motherapy residual masses resulted in improved
short axis is greater than 1.0 cm. In most patients EFS compared with patients who underwent
with prior pulmonary nodules, new lung nodules chemotherapy alone.111 Despite development of
identified by CT are typically benign and should the new harmonization criteria for adults, there
be histologically confirmed to establish relapse/ are no data as yet in children to direct how to incor-
progressive disease. Sites of relapse or progres- porate these new response assessment guidelines
sive disease should be FDG avid unless the lesion in pediatric treatment protocols. For example, it is
Leukemia and Lymphoma 789

Fig. 25. (A) PET/CT showing increased FDG uptake in the mediastinal nodal masses of a patient with HL at diag-
nosis. The uptake is clearly greater than in the mediastinal blood pool or liver. (B) After 2 cycles of therapy, a medi-
astinal soft tissue mass persists. There is low-level FDG uptake in the mass, as great as in the mediastinal blood
pool but less than in the liver, which emphasizes the challenge in interpreting residual FDG uptake in patients
completing their up-front chemotherapy.
790 Guillerman et al

unclear whether radiation treatment can be re- presents as an enlarging thymic mass within 6
duced to initial involved sites of nodal activity or to 8 months of completion of chemotherapy
eliminated entirely for those patients with HL who (Fig. 26). Rebound thymic hyperplasia can show
have no residual nodal activity early after response avidity for FDG, usually in a pattern of mild, diffuse
to therapy.55 One approach, for example, would be uptake, in contrast with the intense, discrete
restricting radiation to sites of residual FDG uptake. uptake usually associated with lymphoma. If a child
The goal would be to titrate therapy using the prog- or adolescent has imaging findings compatible
nostic value of early treatment response to reduce with rebound thymic hyperplasia, continued sur-
treatment intensity in those patients with rapid early veillance is advised rather than biopsy, especially
responses and thereby reduce toxicity while, at the if there is no other evidence of recurrence or prior
same time, intensifying treatment of those with neoplastic involvement of the thymus.112
slow early responses in an effort to improve disease In a recent pediatric intermediate-stage and
control. advanced-stage HL study, relapses occurred in
These approaches, all of which require confirma- 10.6% of the patients, with a median time to relapse
tion in clinical trial settings, are intended to develop of 7 months.113 Most of these relapses occurred
response-adapted therapy to identify patients with within 18 months after completion of therapy and
favorable chemotherapy-sensitive disease who most of these relapses were local, at original sites
can be treated with abbreviated chemotherapy of disease. Nearly two-thirds of these relapses
and low-dose IFRT or no radiation therapy at all. were detected based on clinical symptoms, labora-
Alternatively, rather than entirely eliminating radia- tory tests, or physical examination findings. Only
tion therapy, radiation therapy volumes may be 17% of the patients, all of whom relapsed more
restricted to lymph node regions that were initially than 1 year after therapy, were asymptomatic and
involved with disease rather than the entire regional had disease detected solely from surveillance
nodal group.55,99 Such an approach has the poten- imaging. A review of the number of imaging studies
tial to significantly reduce the irradiated volume of performed to identify these relapses revealed that
normal tissues compared with involved field radia- more than 400 CT scans were mandated by
tion and, coupled with functional imaging response protocol to detect these asymptomatic relapses.
assessment, could produce significant reductions Based on this, it has been suggested that CT, and
in radiation-related late effects. imaging of any kind, is overused in the routine post-
treatment surveillance of patients with HL, and
modifications in surveillance protocol are indicated
Surveillance for Relapse
for routine long-term surveillance. A recent report
Relapse occurs in approximately 20% of pediatric has further emphasized the considerable increase
patients with HL.63 Most of these occur within the in radiation exposure to these patients attributable
first 3 years. It has been suggested that relapses to routine surveillance imaging.114
occurring beyond 1 year after the completion of As has been reported in adult studies, most
therapy have a favorable prognosis relative to relapses occur in areas of initial disease and within
those occurring early after completion of therapy the first year after completion of therapy, which indi-
and therefore surveillance imaging and identifica- cates that the frequency of screening should be
tion of relapse has remained an important goal of greatest in the early posttherapy years. The role of
observation of therapy. FDG-PET as a surveillance tool to detect relapse
Rebound thymic hyperplasia, a potential mimic in asymptomatic patients has not been established.
of mediastinal relapse, most characteristically In particular, the problem of false-positive findings

Fig. 26. A chest CT image (A) obtained in an asymptomatic 15-year-old patient 5 months after completion of
therapy for HL shows enlargement of an anterior mediastinal soft tissue structure compared with a chest CT
image (B) obtained at the end of therapy, representing rebound thymic hyperplasia.
Leukemia and Lymphoma 791

remains and current recommendations do not within radiation fields, are all of concern.55 Historical
include the use of FDG-PET for routine surveil- treatment regimens for lymphoma, which used high
lance.77 However, if sites of disease are detected radiation doses and intense chemotherapy regi-
by other imaging modalities or are suspected clini- mens, had well-established rates of secondary
cally, there may be a role for FDG-PET imaging in malignancy.54 The risk of secondary malignancy
confirming relapse, but not as an integral part of after low-dose radiation is not well described,
routine surveillance in either HL or NHL.100 In one because this became the standard treatment of
study, the use of FDG-PET/CT to identify recurrent children only in the mid 1980s and for adolescents
disease in asymptomatic patients with HL and NHL in the early 1990s. With unknown latency periods
led to false-positive results in 63% and 41% of for developing second malignancies after low-
patients, respectively, for a positive predictive dose radiation exposure and reduced chemo-
value of only 53%.106 Although the negative predic- therapy, there is no clear role for routine surveillance
tive value was greater than 99% in this study, the imaging in these patients.
high frequency of false positives does not allow
appropriate treatment decisions to be confidently FUTURE DIRECTIONS
made based solely on FDG-PET surveillance
imaging. With isotropic voxel acquisition of cross-sectional
There will be understandable reluctance to imaging data, the ability to generate three-
reducing the intensity of surveillance imaging at dimensional tumor representations has improved
a time when treatment intensity and duration of and accurate tumor volume calculations are now
therapy are also being reduced. Nonetheless, all feasible, although automated measurement tech-
of the available evidence suggests that aggressive niques remain elusive. Nonetheless, changes in tu-
monitoring early after therapy combined with judi- mor volume, particularly with extensive multifocal
cious imaging and close physical examination and sites of bulky lymphoma, may be an important vari-
laboratory monitoring during the surveillance able to correlate with FDG-PET response. It seems
period is the most effective means of following overly simplistic to assume that all patients who
these patients. become FDG negative after 2 cycles of therapy,
even in the presence of large residual masses, will
TREATMENT-RELATED COMPLICATIONS be uniformly free of disease progression or relapse.
The ability to provide other criteria for response to
Overall survival rates are excellent for ALL, HL, local- develop a multivariant array of imaging criteria
ized low-stage NHL, and even for some advanced- should allow us to best identify those patients truly
stage NHL and AML. The goal of reducing toxic manifesting a good response to chemotherapy,
effects of therapy is now a focus of the next genera- without overlooking those patients in whom more
tion of treatment protocols.55,115 Imaging plays aggressive treatment is mandated.
an important role in diagnosing treatment-related In the past, MR imaging of the thoracic and
complications of leukemia and lymphoma. Many of abdominal cavity has been limited by motion arti-
the complications associated with leukemia and fact and long examination times. However, with
lymphoma are shared because of the treatment of increasing availability of faster MR imaging scan-
both with cytotoxic chemotherapy with associated ning techniques and respiratory and cardiac gat-
marrow suppression. The complications can be ing capabilities, a more routine role of MR imaging
acute or late in onset, and can involve virtually any in evaluating pediatric lymphoma is becoming
organ system. Among the complications amenable feasible (Fig. 27). Whole-body MR imaging has
to diagnosis by imaging are opportunistic infecti- shown good agreement with FDG-PET/CT for
ons, cerebral hemorrhage/infarction, methotrexate- both nodal and extranodal staging of lymphoma.116
induced leukoencephalopathy, venous thrombosis, MR imaging false negatives occur with normal-
anthracycline-induced cardiomyopathy, bleomycin- sized involved lymph nodes and spleen, disease
induced pulmonary fibrosis, bronchiolitis obliterans, that is detectable by FDG-PET. MR imaging pro-
radiation pneumonitis, radiation pericarditis, typhli- vides an alternative imaging method to CT for
tis, asparaginase-associated pancreatitis, hepatic anatomic disease assessment at staging and re-
veno-occlusive disease, graft-versus-host disease, staging without ionizing radiation exposure. Fur-
hemorrhagic cystitis, posttransplant lymphoprolifer- thermore, surveillance by MR imaging may be the
ative disorder, osteonecrosis, and osteoporosis. A imaging modality of choice, particularly in patients
detailed discussion of the imaging of these compli- with lymphoma in whom sustained PR or CR has
cations is beyond the scope of this article. occurred.
Second malignant neoplasms, including AML, Because of high tumor cellularity and high
NHL, and malignancies of breast, lung, and thyroid nuclear/cytoplasm ratios, most forms of lymphoma
792 Guillerman et al

Fig. 27. A 16-year-old boy with stage IIA HL. Initial presentation with cough led to discovery of an anterior medi-
astinal mass, for which a broad differential diagnosis existed. Chest CT, T2-weighted chest MR imaging, and FDG-
PET scan show an FDG-avid mass that abuts the pericardium, consistent with neoplasm. Gated cardiac MR imaging
(not shown) sequences showed that the mass was not adherent to pericardium. The mass was subsequently
excised, confirming the diagnosis of HL.

that have been studied have high signal intensity finding in the pelvis and spine of children limit the
(ie, restricted diffusion) on diffusion-weighted MR specificity of this technique, raising the risk of
images.95,96 In one study, diffusion-weighted MR false-positive interpretations.121
imaging matched FDG-PET/CT findings in 94% of
the lymph node regions studied.117 Furthermore, SUMMARY
changes in diffusion characteristics may provide
an additional means of evaluating residual nodal As the most common childhood malignancy,
masses, because nodal apparent diffusion coeffi- leukemia is frequently encountered as an under-
cient has been shown to increase following lying condition in subjects of pediatric imaging
successful chemotherapy.118 The use of whole- studies. The most frequent indication for imaging
body MR imaging with diffusion-weighted imaging of children with leukemia is to evaluate for complica-
with background signal suppression sequences tions of treatment. Occasionally, imaging findings
has also been shown to provide better tissue suggest a previously unsuspected diagnosis of
contrast in detecting malignant nodal involvement leukemia, particularly in children with nonspecific
compared with conventional MR imaging se- musculoskeletal complaints or unexplained fever.
quences.119 As these techniques are incorporated There is no current routine role of imaging in risk
into the evaluation of pediatric lymphoma patients, stratification, therapy response assessment, or re-
it seems likely that changes in tumor characteris- lapse surveillance for childhood leukemia.
tics as manifested by changes in diffusion or The use of imaging in guiding diagnostic
changes in enhancement may provide additional procedures, risk stratification, therapy response
surrogates of response to help further develop assessment, and relapse surveillance in childhood
a response assessment profile. Whole-body MR lymphoma has evolved in the last 20 to 30 years.
imaging with diffusion-weighted imaging may From a time when nearly all patients with lym-
also serve as a more sensitive method to rapidly phoma were surgically staged to a time when
evaluate leukemic infiltration of the bone marrow nearly all patients have multiple imaging studies,
for therapeutic response to cytotoxic chemo- each of which provides complimentary informa-
therapy.120 However, widespread interindividual tion, current lymphoma management requires the
variation and restricted diffusion as a normal integration of imaging at all phases of treatment.
Leukemia and Lymphoma 793

Current approaches should include a combination 10. Siegel MJ, Shackelford GD, McAlister WH. Pleural
of anatomic and functional imaging techniques to thickening. An unusual feature of childhood leukemia.
predict which patients will benefit from less toxic Radiology 1981;138:3679.
treatment regimens and which patients will require 11. Abbas AA, Baker DL, Felimban SK, et al. Musculo-
augmented therapy. Patient-specific and disease- skeletal and radiological manifestations of child-
specific imaging biomarkers to provide specific hood acute leukaemia: a clinical review. Haema
indicators of disease activity are needed to guide 2004;7:44855.
the evolution from risk-adapted therapy to 12. Sinigaglia R, Gigante C, Bisinella G, et al. Musculo-
response-based therapy. The ongoing challenge skeletal manifestations in pediatric acute leukemia.
is to optimize available imaging techniques and J Pediatr Orthop 2008;28:208.
develop a reproducible set of validated biomarkers 13. Hann IM, Gupta S, Palmer MK, et al. The prognostic
and image-processing tools to best accomplish significance of radiological and symptomatic bone
these goals. involvement in childhood acute lymphoblastic
leukaemia. Med Pediatr Oncol 1979;6:515.
14. Melhem RE, Saber TJ. Erosion of the medial cortex
of the proximal humerus. A sign of leukemia on the
REFERENCES chest radiograph. Radiology 1980;137:779.
15. Bernard EJ, Nicholls WD, Howman-Giles RB, et al.
1. Smith MA, Ries LAG, Gurney JG, et al. Leukemia. Patterns of abnormality on bone scans in acute
In: Ries LA, Smith MA, Gurney JG, editors. Cancer childhood leukemia. J Nucl Med 1998;39:19836.
incidence and survival among children and adoles- 16. Shalaby-Rana E, Majd M. (99m)Tc-MDP scinti-
cents: United States SEER Program 19751995. graphic findings in children with leukemia: value
Bethesda (MD): National Cancer Institute; 1999. of early and delayed whole-body imaging. J Nucl
p. 1734. Med 2001;42:87883.
2. Greaves M. In utero origins of childhood 17. Clausen N, Gotze H, Pedersen A, et al. Skeletal
leukaemia. Early Hum Dev 2005;81:1239. scintigraphy and radiography at onset of acute
3. Altekruse SF, Kosary CL, Krapcho M, et al. SEER lymphocytic leukemia in children. Med Pediatr On-
cancer statistics review, 19752007, National Cancer col 1983;11:2916.
Institute. Bethesda (MD), based on November 2009 18. Moulopoulos LA, Dimopoulos MA. Magnetic reso-
SEER data submission, posted to the SEER Web nance imaging of the bone marrow in hematologic
site, 2010. Available at: http://seer.cancer.gov/csr/ malignancies. Blood 1997;90:212747.
1975_2007/. Accessed September 7, 2010. 19. Moore SG, Gooding CA, Brasch RC, et al. Bone
4. Swerdlow SH, Campo E, Harris NL, et al. WHO marrow in children with acute lymphocytic leukemia:
classification of tumours of haematopoietic and MR relaxation times. Radiology 1986;160:23740.
lymphoid tissues. 4th edition. Lyon (France): Inter- 20. Babyn PS, Ranson M, McCarville ME. Normal bone
national Agency for Research on Cancer; 2008. marrow: signal characteristics and fatty conver-
5. Dainiak N. Hematologic consequences of expo- sion. Magn Reson Imaging Clin N Am 1998;6:
sure to ionizing radiation. Exp Hematol 2002;30: 47395.
51328. 21. Ruzal-Shapiro C, Berdon WE, Cohen MD, et al. MR
6. Gaynon PS, Angiolillo AL, Carroll WL, et al. Long-term imaging of diffuse bone marrow replacement in pedi-
results of the Childrens Cancer Group studies for chi- atric patients with cancer. Radiology 1991;181:5879.
ldhood acute lymphoblastic leukemia 19832002: 22. Truong TH, Beyene J, Hitzler J, et al. Features at
a Childrens Oncology Group Report. Leukemia presentation predict children with acute lympho-
2010;24:28597. blastic leukemia at low risk for tumor lysis syndrome.
7. Smith MA, Seibel NL, Altekruse SF, et al. Outcomes Cancer 2007;110:18329.
for children and adolescents with cancer: chal- 23. Niemeyer CM, Arico M, Basso G, et al. Chronic
lenges for the twenty-first century. J Clin Oncol myelomonocytic leukemia in childhood: a retro-
2010;28:262534. spective analysis of 110 cases. European Working
8. Creutzig U, Reinhardt D. Current controversies: Group on Myelodysplastic Syndromes in Child-
which patients with acute myeloid leukaemia hood (EWOG-MDS). Blood 1997;89:353443.
should receive a bone marrow transplantation?A 24. Murray JC, Dorfman SR, Brandt ML, et al. Renal
European view. Br J Haematol 2002;118:36577. venous thrombosis complicating acute myeloid
9. Lange BJ, Smith FO, Feusner J, et al. Outcomes in leukemia with hyperleukocytosis. J Pediatr Hematol
CCG2961, a childrens oncology group phase 3 Oncol 1996;18:32730.
trial for untreated pediatric acute myeloid leukemia: 25. Hilmes MA, Dillman JR, Mody RJ, et al. Pediatric
a report from the childrens oncology group. Blood renal leukemia: spectrum of CT imaging findings.
2008;111:104453. Pediatr Radiol 2008;38:42430.
794 Guillerman et al

26. Sato A, Imaizumi M, Chikaoka S, et al. Acute renal childhood acute lymphoblastic leukaemia. Arch
failure due to leukemic cell infiltration followed by Dis Child 1977;52:5303.
relapse at multiple extramedullary sites in a child 40. Heinrich SD, Gallagher D, Warrior R, et al. The
with acute lymphoblastic leukemia. Leuk Lymphoma prognostic significance of the skeletal manifesta-
2004;45:8258. tions of acute lymphoblastic leukemia of childhood.
27. Hayek M, Srinivasan A. Acute lymphoblastic J Pediatr Orthop 1994;14:10511.
leukemia presenting with lactic acidosis and renal 41. DAngelo P, Mura R, Rizzari C, et al. Prognostic
tubular dysfunction. J Pediatr Hematol Oncol 2003; value of nephromegaly at diagnosis of childhood
25:48890. acute lymphoblastic leukemia. Acta Haematol
28. Rausch DR, Norton KI, Glass RB, et al. Infantile 1995;94:849.
leukemia presenting with cholestasis secondary 42. Yetgin S, Olgar S, Aras T, et al. Evaluation of kidney
to massive pancreatic infiltration. Pediatr Radiol damage in patients with acute lymphoblastic leu-
2002;32:3601. kemia in long-term follow-up: value of renal scan.
29. Jankovic M, Zanetto F, Conter V, et al. Cranial Am J Hematol 2004;77:1329.
computed tomography findings in children with 43. Attarbaschi A, Mann G, Dworzak M, et al. Medias-
acute lymphoblastic leukemia at diagnosis. Am J tinal mass in childhood T-cell acute lymphoblastic
Pediatr Hematol Oncol 1989;11:3279. leukemia: significance and therapy response.
30. Vazquez E, Lucaya J, Castellote A, et al. Neuroi- Med Pediatr Oncol 2002;39:55865.
maging in pediatric leukemia and lymphoma: 44. Vande Berg BC, Lecouvet FE, Michaux L, et al.
differential diagnosis. Radiographics 2002;22: Magnetic resonance imaging of the bone marrow
141128. in hematological malignancies. Eur Radiol 1998;8:
31. Ulu EM, Tore HG, Bayrak A, et al. MRI of central ner- 133544.
vous system abnormalities in childhood leukemia. 45. Fletcher BD, Wall JE, Hanna SL. Effect of hemato-
Diagn Interv Radiol 2009;15:8692. poietic growth factors on MR images of bone
32. Shackelford GD, Bloomberg G, McAlister WH. The marrow in children undergoing chemotherapy.
value of roentgenography in differentiating aplastic Radiology 1993;189:74551.
anemia from leukemia masquerading as aplastic 46. Emy PY, Levin TL, Sheth SS, et al. Iron overload in retic-
anemia. Am J Roentgenol Radium Ther Nucl Med uloendothelial systems of pediatric oncology patients
1972;116:6514. who have undergone transfusions: MR observations.
33. Kai T, Ishii E, Matsuzaki A, et al. Clinical and prog- AJR Am J Roentgenol 1997;168:10115.
nostic implications of bone lesions in childhood 47. Benz-Bohm G, Gross-Fengels W, Bohndorf K, et al.
leukemia at diagnosis. Leuk Lymphoma 1996;23: MRI of the knee region in leukemic children. Part II.
11923. Follow up: responder, non-responder, relapse. Pe-
34. Guillerman RP. Normal and abnormal bone marrow. diatr Radiol 1990;20:2726.
In: Slovis T, editor. Caffeys pediatric diagnostic 48. Gaynon PS, Qu RP, Chappell RJ, et al. Survival
imaging. 11th edition. Philadelphia: Elsevier; 2008. after relapse in childhood acute lymphoblastic
p. 297096. leukemia: impact of site and time to first relapse
35. Bulas RB, Laine FJ, Das Narla L. Bilateral orbital the Childrens Cancer Group experience. Cancer
granulocytic sarcoma (chloroma) preceding the 1998;82:138795.
blast phase of acute myelogenous leukemia: CT 49. Webb DK. Management of relapsed acute myeloid
findings. Pediatr Radiol 1995;25:4889. leukaemia. Br J Haematol 1999;106:8519.
36. Dusenbery KE, Howells WB, Arthur DC, et al. Extra- 50. Jensen KE, Thomsen C, Henriksen O, et al.
medullary leukemia in children with newly diag- Changes in T1 relaxation processes in the bone
nosed acute myeloid leukemia: a report from the marrow following treatment in children with acute
Childrens Cancer Group. J Pediatr Hematol Oncol lymphoblastic leukemia. A magnetic resonance
2003;25:7608. imaging study. Pediatr Radiol 1990;20:4648.
37. Schultz KR, Pullen DJ, Sather HN, et al. Risk- 51. Kan JH, Hernanz-Schulman M, Frangoul HA, et al.
and response-based classification of childhood B- MRI diagnosis of bone marrow relapse in children
precursoracute lymphoblastic leukemia: a combined with ALL. Pediatr Radiol 2008;38:7681.
analysis of prognostic markers from the Pediatric 52. Rubnitz JE, Hijiya N, Zhou Y, et al. Lack of benefit of
Oncology Group (POG) and Childrens Cancer early detection of relapse after completion of
Group (CCG). Blood 2007;109:92635. therapy for acute lymphoblastic leukemia. Pediatr
38. Creutzig U, Zimmermann M, Ritter J, et al. Defini- Blood Cancer 2005;44:13841.
tion of a standard-risk group in children with 53. Trigg ME, Steinherz PG, Chappell R, et al. Early
AML. Br J Haematol 1999;104:6309. testicular biopsy in males with acute lymphoblastic
39. Masera G, Carnelli V, Ferrari M, et al. Prognostic leukemia: lack of impact on subsequent event-free
significance of radiological bone involvement in survival. J Pediatr Hematol Oncol 2000;22:2733.
Leukemia and Lymphoma 795

54. Reiter A, Ferrando AA. Malignant lymphomas 69. Attarbaschi A, Dworzak M, Steiner M, et al. Outcome
and lymphadenopathies. In: Orkin SH, Fisher DE, of children with primary resistant or relapsed non-
Look AT, et al, editors. Oncology of infancy and child- Hodgkin lymphoma and mature B-cell leukemia
hood. Philadelphia: Elsevier; 2009. p. 417508. after intensive first-line treatment: a population-
55. Hodgson DC, Hudson MM, Constine LS. Pediatric based analysis of the Austrian Cooperative Study
Hodgkin lymphoma: maximizing efficacy and mini- Group. Pediatr Blood Cancer 2005;44:706.
mizing toxicity. Semin Radiat Oncol 2007;17:23042. 70. Abramson SJ, Price AP. Imaging of pediatric
56. Jaffe ES, Harris NL, Stein H, et al. Pathology and lymphomas. Radiol Clin North Am 2008;46:31338, ix.
genetics of tumours of hematopoietic and lymphoid 71. Shamberger RC. Preanesthetic evaluation of chil-
tissues. World Health Organization Classification of dren with anterior mediastinal masses. Semin Pe-
Tumours of Haemtopoietic and Lymphoid Tissues. diatr Surg 1999;8:618.
Lyon (France): IARC Press; 2001. 72. Toma P, Granata C, Rossi A, et al. Multimodality
57. Claviez A, Tiemann M, Luders H, et al. Impact of imaging of Hodgkin disease and non-Hodgkin
latent Epstein-Barr virus infection on outcome in chil- lymphomas in children. Radiographics 2007;27:
dren and adolescents with Hodgkins lymphoma. 133554.
J Clin Oncol 2005;23:404856. 73. Aygun B, Karakas SP, Leonidas J, et al. Reliability
58. Link MP, Weinstein HJ. Malignant non-Hodgkin of splenic index to assess splenic involvement in
lymphomas in children. In: Pizzo PA, Poplack DG, pediatric Hodgkins disease. J Pediatr Hematol On-
editors. Principles and practice of pediatric col 2004;26:746.
oncology. 5th edition. Philadelphia: Lippincott Wil- 74. Hines-Thomas M, Kaste SC, Hudson MM, et al.
liams & Wilkins; 2006. p. 72247. Comparison of gallium and PET scans at diagnosis
59. Cairo MS, Raetz E, Lim MS, et al. Childhood and and follow-up of pediatric patients with Hodgkin
adolescent non-Hodgkin lymphoma: new insights lymphoma. Pediatr Blood Cancer 2008;51:198203.
in biology and critical challenges for the future. Pe- 75. Rini JN, Nunez R, Nichols K, et al. Coincidence-
diatr Blood Cancer 2005;45:75369. detection FDG-PET versus gallium in children and
60. Gottschalk S, Rooney CM, Heslop HE. Post-trans- young adults with newly diagnosed Hodgkins
plant lymphoproliferative disorders. Annu Rev disease. Pediatr Radiol 2005;35:16978.
Med 2005;56:2944. 76. Mody RJ, Bui C, Hutchinson RJ, et al. Comparison of
61. Shroff R, Rees L. The post-transplant lymphoproli- (18)F flurodeoxyglucose PET with Ga-67 scintig-
ferative disordera literature review. Pediatr Neph- raphy and conventional imaging modalities in pedi-
rol 2004;19:36977. atric lymphoma. Leuk Lymphoma 2007;48:699707.
62. Bakker NA, van Imhoff GW, Verschuuren EA, et al. 77. Juweid ME, Stroobants S, Hoekstra OS, et al. Use of
Presentation and early detection of post-transplant positron emission tomography for response assess-
lymphoproliferative disorder after solid organ trans- ment of lymphoma: consensus of the Imaging
plantation. Transpl Int 2007;20:20718. Subcommittee of International Harmonization Pro-
63. Schwartz CL. Special issues in pediatric Hodgkins ject in Lymphoma. J Clin Oncol 2007;25:5718.
disease. Eur J Haematol Suppl 2005;(66):5562. 78. Hudson MM, Krasin MJ, Kaste SC. PET imaging in
64. Punnett A, Tsang RW, Hodgson DC. Hodgkin pediatric Hodgkins lymphoma. Pediatr Radiol
lymphoma across the age spectrum: epidemi- 2004;34:1908.
ology, therapy, and late effects. Semin Radiat On- 79. Wickmann L, Luders H, Dorffel W. 18-FDG-PET-
col 2010;20:3044. findings in children and adolescents with Hodgkins
65. Ruhl U, Albrecht M, Dieckmann K, et al. Response- disease: retrospective evaluation of the correlation
adapted radiotherapy in the treatment of pediatric to other imaging procedures in initial staging and
Hodgkins disease: an interim report at 5 years of to the predictive value of follow up examinations.
the German GPOH-HD 95 trial. Int J Radiat Oncol Klin Padiatr 2003;215:14650 [in German].
Biol Phys 2001;51:120918. 80. Furth C, Denecke T, Steffen I, et al. Correlative
66. Nachman JB, Sposto R, Herzog P, et al. Randomized imaging strategies implementing CT, MRI, and
comparison of low-dose involved-field radiotherapy PET for staging of childhood Hodgkin disease.
and no radiotherapy for children with Hodgkins J Pediatr Hematol Oncol 2006;28:50112.
disease who achieve a complete response to 81. Hermann S, Wormanns D, Pixberg M, et al. Staging
chemotherapy. J Clin Oncol 2002;20:376571. in childhood lymphoma: differences between FDG-
67. Aleman BM, Re D, Diehl V. The role of radiation PET and CT. Nuklearmedizin 2005;44:17.
therapy in patients with Hodgkins lymphoma. 82. Riad R, Omar W, Kotb M, et al. Role of PET/CT in
Curr Hematol Malig Rep 2007;2:15160. malignant pediatric lymphoma. Eur J Nucl Med
68. Bradley MB, Cairo MS. Stem cell transplantation for Mol Imaging 2010;37:31929.
pediatric lymphoma: past, present and future. 83. Paes FM, Kalkanis DG, Sideras PA, et al. FDG PET/
Bone Marrow Transplant 2008;41:14958. CT of extranodal involvement in non-Hodgkin
796 Guillerman et al

lymphoma and Hodgkin disease. Radiographics involvement in malignant lymphoma. Br J Haematol


2010;30:26991. 2010;149:62830.
84. Hernandez-Pampaloni M, Takalkar A, Yu JQ, et al. 97. Kabickova E, Sumerauer D, Cumlivska E, et al.
F-18 FDG-PET imaging and correlation with CT in Comparison of 18F-FDG-PET and standard proce-
staging and follow-up of pediatric lymphomas. Pe- dures for the pretreatment staging of children and
diatr Radiol 2006;36:52431. adolescents with Hodgkins disease. Eur J Nucl
85. Hutchings M, Loft A, Hansen M, et al. Position Med Mol Imaging 2006;33:102531.
emission tomography with or without computed 98. Robertson VL, Anderson CS, Keller FG, et al. Role
tomography in the primary staging of Hodgkins of FDG-PET in the definition of involved-field radia-
lymphoma. Haematologica 2006;91:4829. tion therapy and management for pediatric Hodg-
86. Miller E, Metser U, Avrahami G, et al. Role of 18F- kins lymphoma. Int J Radiat Oncol Biol Phys
FDG PET/CT in staging and follow-up of lymphoma 2011;80(2):32432.
in pediatric and young adult patients. J Comput 99. Krasin MJ, Hudson MM, Kaste SC. Positron emis-
Assist Tomogr 2006;30:68994. sion tomography in pediatric radiation oncology:
87. Shammas A, Lim R, Charron M. Pediatric FDG integration in the treatment-planning process. Pe-
PET/CT: physiologic uptake, normal variants, diatr Radiol 2004;34:21421.
and benign conditions. Radiographics 2009;29: 100. Shankar A, Fiumara F, Pinkerton R. Role of FDG
146786. PET in the management of childhood lymphomas-
88. Kaste SC, Howard SC, McCarville EB, et al. 18F- case proven or is the jury still out? Eur J Cancer
FDG-avid sites mimicking active disease in pedi- 2008;44:66373.
atric Hodgkins. Pediatr Radiol 2005;35:14154. 101. Pinkerton R. Continuing challenges in childhood
89. Gelfand MJ, OHara SM, Curtwright LA, et al. Pre- non-Hodgkins lymphoma. Br J Haematol 2005;
medication to block [(18)F]FDG uptake in the 130:4808.
brown adipose tissue of pediatric and adolescent 102. Miller AB, Hoogstraten B, Staquet M, et al. Reporting
patients. Pediatr Radiol 2005;35:98490. results of cancer treatment. Cancer 1981;47:20714.
90. Bea S, Zettl A, Wright G, et al. Diffuse large B-cell 103. Hutchings M, Barrington SF. PET/CT for therapy
lymphoma subgroups have distinct genetic profiles response assessment in lymphoma. J Nucl Med
that influence tumor biology and improve gene- 2009;50(Suppl 1):21S30S.
expression-based survival prediction. Blood 2005; 104. Meany HJ, Gidvani VK, Minniti CP. Utility of PET
106:318390. scans to predict disease relapse in pediatric
91. von Falck C, Maecker B, Schirg E, et al. Post trans- patients with Hodgkin lymphoma. Pediatr Blood
plant lymphoproliferative disease in pediatric solid Cancer 2007;48:399402.
organ transplant patients: a possible role for [18F]- 105. Montravers F, McNamara D, Landman-Parker J,
FDG-PET(/CT) in initial staging and therapy moni- et al. [(18)F]FDG in childhood lymphoma: clinical
toring. Eur J Radiol 2007;63:42735. utility and impact on management. Eur J Nucl
92. Crowther D, Lister TA. The Cotswolds report on the Med Mol Imaging 2002;29:115565.
investigation and staging of Hodgkins disease. Br 106. Rhodes MM, Delbeke D, Whitlock JA, et al. Utility of
J Cancer 1990;62:5512. FDG-PET/CT in follow-up of children treated for
93. Dieckmann K, Potter R, Hofmann J, et al. Does bulky Hodgkin and non-Hodgkin lymphoma. J Pediatr
disease at diagnosis influence outcome in child- Hematol Oncol 2006;28:3006.
hood Hodgkins disease and require higher radia- 107. Furth C, Steffen IG, Amthauer H, et al. Early and
tion doses? Results from the German-Austrian late therapy response assessment with [18F]fluoro-
Pediatric Multicenter Trial DAL-HD-90. Int J Radiat deoxyglucose positron emission tomography in
Oncol Biol Phys 2003;56:64452. pediatric Hodgkins lymphoma: analysis of
94. Shulkin BL, Goodin GS, McCarville MB, et al. Bone a prospective multicenter trial. J Clin Oncol 2009;
and [18F]fluorodeoxyglucose positron-emission 27:438591.
tomography/computed tomography scanning for 108. Cheson BD, Pfistner B, Juweid ME, et al. Revised
the assessment of osseous involvement in Hodgkin response criteria for malignant lymphoma. J Clin
lymphoma in children and young adults. Leuk Oncol 2007;25:57986.
Lymphoma 2009;50:1794802. 109. Delbeke D, Stroobants S, de Kerviler E, et al.
95. Kwee TC, Takahara T, Vermoolen MA, et al. Whole- Expert opinions on positron emission tomography
body diffusion-weighted imaging for staging malig- and computed tomography imaging in lymphoma.
nant lymphoma in children. Pediatr Radiol 2010; Oncologist 2009;14(Suppl 2):3040.
40(10):1592602. 110. Gallamini A, Fiore F, Sorasio R, et al. Interim positron
96. Kwee TC, Fijnheer R, Ludwig I, et al. Whole-body emission tomography scan in Hodgkin lymphoma:
magnetic resonance imaging, including diffusion- definitions, interpretation rules, and clinical valida-
weighted imaging, for diagnosing bone marrow tion. Leuk Lymphoma 2009;50:17614.
Leukemia and Lymphoma 797

111. Picardi M, De Renzo A, Pane F, et al. Randomized 117. Lin C, Luciani A, Itti E, et al. Whole-body diffusion-
comparison of consolidation radiation versus obser- weighted magnetic resonance imaging with
vation in bulky Hodgkins lymphoma with post- apparent diffusion coefficient mapping for staging
chemotherapy negative positron emission tomog- patients with diffuse large B-cell lymphoma. Eur
raphy scans. Leuk Lymphoma 2007;48:17217. Radiol 2010;20:202738.
112. Guillerman RP, Parker BR. Pediatric lymphoma. In: 118. Punwani S, Shankar A, Daw S, et al. Derivation of his-
Guermazi A, editor. Radiological imaging in hemato- tographic apparent diffusion coefficient changes
logical malignancies. Berlin; Heidelberg: Springer- associated with successful response to first line
Verlag; 2004. p. 24788. chemotherapy of adolescent and childhood lym-
113. Voss SD, Constine LS, London WB, et al. Evaluation of phoma. Pediatr Radiol 2010;40:1098.
surveillance CT imaging for the detection of relapse in 119. Savelli S, Maurizio MD, Mortilla M, et al. Whole-body
Hodgkin lymphoma. Pediatr Radiol 2010;40:590. MRI with diffusion-weighted whole-body imaging
114. Chong AL, Grant RM, Ahmed BA, et al. Imaging in with background signal suppression sequences
pediatric patients: time to think again about surveil- for disease detection and staging of pediatric onco-
lance. Pediatr Blood Cancer 2010;55:40713. logical patients. Pediatr Radiol 2010;40:1097.
115. Metzger ML, Hudson MM. Balancing efficacy and 120. Ballon D, Watts R, Dyke JP, et al. Imaging thera-
safety in the treatment of adolescents with Hodg- peutic response in human bone marrow using
kins lymphoma. J Clin Oncol 2009;27:60713. rapid whole-body MRI. Magn Reson Med 2004;
116. Punwani S, Taylor SA, Bainbridge A, et al. Pediatric 52:12348.
and adolescent lymphoma: comparison of whole- 121. Ording Muller LS, Avenarius D, Olsen OE. High
body STIR half-Fourier RARE MR imaging with an signal in bone marrow at diffusion-weighted imaging
enhanced PET/CT reference for initial staging. with body background suppression (DWIBS) in
Radiology 2010;255:18290. healthy children. Pediatr Radiol 2011;41(2):2216.

You might also like