You are on page 1of 10

SPELEMAN, PARK, AND HENDERSON

Neuroblastoma: A Tough Nut to Crack


Frank Speleman, PhD, Julie R. Park, MD, and Tara O. Henderson, MD, MPH

OVERVIEW

Neuroblastoma, an embryonal tumor arising from neural crest–derived progenitor cells, is the most common solid tumor in
childhood, with more than 700 cases diagnosed per year in the United States. In the past several decades, significant
advances have been made in the treatment of neuroblastoma. Treatment advances reflect improved understanding of the
biology of neuroblastoma. Although amplification of MYCN was discovered in the early 1980s, our understanding of
neuroblastoma oncogenesis has advanced in the last decade as a result of high-throughput genomic analysis, exome and
whole-genome sequencing, genome-wide association studies, and synthetic lethal drug screens. Our refined understanding
of neuroblastoma biology and genetics is reflected in improved prognostic stratification and appropriate tailoring of therapy
in recent clinical trials. Moreover, for high-risk neuroblastoma, a disease that was uniformly fatal 3 decades ago, recent
clinical trials incorporating autologous hematopoietic transplant and immunotherapy utilizing anti-GD2 antibody plus
cytokines have shown improved event-free and overall survival. These advances have resulted in a growing population of
long-term survivors of neuroblastoma. Examination of the late effects and second malignant neoplasms (SMNs) in both older
generations of survivors and more recently treated survivors will inform both design of future trials and surveillance
guidelines for long-term follow-up. As a consequence of advances in understanding of the biology of neuroblastoma,
successful clinical trials, and refined understanding of the late effects and SMNs of survivors, the promise of precision
medicine is becoming a reality for patients with neuroblastoma.

N euroblastoma is undoubtedly one of the most enigmatic


tumors, with remarkable heterogeneous clinical be-
havior ranging from spontaneous regression to metastatic
regulatory pathways controlling MYCN activity, bringing the
old enemy into the spotlight of our current and future
targeted drug efforts. Fortunately, our increasing knowledge
disease that is refractory to therapy (see the therapy update of neuroblastoma biology and genetics, together with access
section). Progress in understanding the molecular basis of to a rapidly increasing number of actionable drugs, is now
the biologic and clinical behavior of neuroblastoma has offering new hope for significantly increased cure rates and
moved forward at a slower pace compared with many other reduced toxicity.
tumor entities, such as childhood leukemias. Although am-
plification of the MYCN oncogene was discovered as early NEUROBLASTOMA BIOLOGY UPDATE
as 1983,1 the finding of further genes implicated in neu- Neuroblastoma Biology and Genetics: Progress in
roblastoma oncogenesis took much longer. Low-resolution Brief
whole-genome analyses revealed further highly recurrent In the recent past, several excellent reviews have described
and large genomic DNA copy-number alterations, such as progress in understanding neuroblastoma biology.3-8 Neu-
1p and 11q deletions and 17q gain.2 In the past decade, roblastoma, an embryonal tumor arising from neural
this picture has dramatically changed by virtue of high- crest–derived progenitor cells, has been considered as a
throughput genomic analysis such as transcriptome pro- developmental disorder that develops through deregulation
filing, exome and whole-genome sequencing, genome-wide of signaling pathways governing sympathetic nervous sys-
association studies, and synthetic lethal drug screens. tem development, such as neurothrophin receptor signal-
As a result, step by step, the promise of precision oncology is ing, and several key transcription factors orchestrating
now also approaching patients with neuroblastoma suf- the complex developmental lineage commitment and
fering from aggressive tumors at diagnosis or relapsed tu- differentiation. 6,9-11
mors with limited further treatment options.3 Interestingly, As for other cancers, a search for recurrent genetic alterations
many of the novel findings connect directly or indirectly to was a major path toward understanding neuroblastoma

From the Center for Medical Genetics Ghent, Cancer Research Institute Ghent, Ghent, Belgium; Seattle Children’s Hospital, Seattle, WA; Department of Pediatrics, University of
Washington School of Medicine, Seattle, WA; University of Chicago Comer Children’s Hospital, Chicago, IL.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Julie R. Park, MD, Seattle Children’s Hospital, 4800 Sandpoint Way NE, Seattle, WA 98105; email: julie.park@seattlechildrens.org.

© 2016 by American Society of Clinical Oncology.

e548 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


Downloaded from ascopubs.org by 181.53.178.179 on May 1, 2021 from 181.053.178.179
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
NEUROBLASTOMA BIOLOGY, THERAPY, AND SURVIVORSHIP

formation. Roughly three major subgroups were identified Untangling the MYCN Regulatory Network
using DNA copy-number analyses: favorable tumors among MYCN biology has been intensively studied for many years
children younger than 18 months without MYCN amplifi- using in vitro model systems and a TH-MYCN–driven mouse
cation with typically whole chromosome imbalances, and model, among others. More recently, new mouse and
high-risk tumors among older children with either MYCN zebrafish models were generated18,29-31 and novel pros-
amplification and 1p deletions or 11q deletions in the pects for drug MYCN activity have brought MYCN further
absence of MYCN amplifications, with both high-risk into the picture.32 MYCN controls multiple essential cellular
groups exhibiting almost invariably 17q gain.2,12,13 In view processes, including cell cycle, apoptosis, differentiation,
of the high frequency and large size of these segmental and metabolism, and it is involved in basic processes
aberrations, it has been proposed that dosage effects of connected to replication, transcription, and splicing.33-35
multiple genes located on the involved chromosomal To further unravel the underlying molecular basis of MYCN
regions are implicated in tumor formation. Unfortunately, transcriptional control, chromatin immunoprecipitation
the large size of these segments has precluded the identi- sequencing has been performed and many bona fide target
fication of the culprit genes on 11q and 17q, whereas a few genes have been identified, although MYC(N) overall can
genes are now considered to be involved for 1p, including bind to thousands of promotors. The nature of these target
CHD5, CAMTA1, and CASZ1.14-16 genes appears to be context dependent, and recent studies
In addition to MYCN, two other oncogenes, ALK and provided evidence of a so-called amplifier role for MYC and
LIN28B, were found to be amplified, albeit in very low MYCN, boosting the transcription of already active genes, a
frequency.17,18 Furthermore, activating ALK mutations were process that might be distinctly different between normal
identified as the major cause of familial neuroblastomas, and malignant cells. 36,37 MYCN both activates and re-
although somatic mutations were discovered in less than presses certain gene sets through promotor binding and
10% of neuroblastomas, opening the way for targeted association of activator or repressor protein complexes,
therapies.19-21 A remarkable productive approach was taken respectively. In this context, the binding of MYCN to EZH2,
by Maris et al using genome-wide association studies to which is overexpressed in neuroblastoma cells and is a core
genetic variants predisposing to neuroblastoma and pointing component of the PRC2 repressive complex, may play a key
toward involvement of multiple loci, including LINC00340, role in MYCN-controlled gene repression of neuronal dif-
BARD1, LMO1, DUSP12, DDX4, LIN28B, HACE1, NEFL, and ferentiation genes. Further dissection of the regulatory
TP53.22-28 protein complexes that are acting in concert with MYCN in
oncogenic regulation of DNA replication, transcription,
splicing, and so forth will be pivotal.38,39 Moreover, MYCN
also controls expression of many microRNAs, most nota-
bly the miR-17-92 cluster, targeting transforming growth
factor-beta pathway components and DKK3, among
KEY POINTS others.40-42

• Recent high-throughput genomics analyses have


provided a wealth of novel data that shed new light on The Novel Neuroblastoma Oncogenes, ALK and
complex neuroblastoma tumor biology and clinical LIN28B, Are Wired Into the MYCN Regulatory
behavior. Network
• Novel insights into tumor biology are opening the way
Given the central role of MYCN in neuroblastoma biology,
for precision medicine among patients with high-risk
and relapsed neuroblastoma.
understanding upstream regulators is also of key impor-
• Tumor biology and clinical characteristics determine tance. LIN28B has been shown to positively regulate MYCN
prognosis and treatment recommendations. levels through let-7 binding. In keeping with its impor-
• Therapy reduction has proven possible for patients tant upstream regulator role for MYCN, LIN28B itself
without high-risk neuroblastoma; for patients with has been recognized as a bona fide driver gene by mouse
high-risk neuroblastoma, dose-intensive multimodality modeling. 18 In addition, using an unbiased LIN28B-39
therapy, including dose-intensive induction untranslated region reporter screen, we found that miR-
chemotherapy, high-dose chemotherapy with 26a-5p and miR-26b-5p regulate LIN28B expression and
autologous stem cell rescue, external beam that MYCN indirectly affects the expression of miR-26a-5p,
radiotherapy, and immunotherapy has improved hence regulating LIN28B. Furthermore, MYCN directly
outcomes.
regulates LIN28B expression through LIN28B promoter
• Understanding of the late effects and second
malignancies of survivors is essential for ensuring the
binding. 43 Taken together, these data point to a com-
long-term health of survivors with neuroblastoma as plex regulatory relationship between both genes.44 Other
well as informing the design of future neuroblastoma MYCN upstream regulators include SIRT1, SIRT2, and
treatment trials aimed at improved survival along with a PRMT5. 45-47
concurrent reduction in late effects. The finding of ALK mutations and rare amplifications in
neuroblastomas was exciting to neuroblastoma researchers

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e549


Downloaded from ascopubs.org by 181.53.178.179 on May 1, 2021 from 181.053.178.179
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
SPELEMAN, PARK, AND HENDERSON

and pediatric oncologists because this represented an im- older than age 14. It was shown that most of the tumors
portant novel druggable target. Several small molecules with ATRX loss showed evidence of alternative lengthening
are now under clinical testing; although resistance to of telomeres. Two recent articles further showed mutual
such compounds has been reported, further in-depth in- exclusivity for ATRX inactivating events versus hTERT ac-
vestigation of the optimal design and potential powerful tivating rearrangements. hTERT is a known MYCN target
combination therapies must be explored to exploit the full gene and previous lower-resolution DNA copy-number
potential of this molecular target.48,49 In this context, we analyses revealed recurrent gains and rearrangements
performed an in-depth analysis of downstream mutant ALK affecting the hTERT locus.60 This type of alteration was
signaling to gain a deeper understanding of the rewired studied in more detail using whole-genome sequencing
oncogenic signaling in ALK mutant neuroblastoma cells and approaches and revealed that 5p15.33 rearrangements
to provide a molecular basis to gain insights into possible juxtapose the TERT coding sequence to strong enhancer
drug-resistance mechanisms and drug-induced compen- elements, resulting in massive chromatin remodeling and
satory pathways. This study provided a rich data set for DNA methylation of the affected region and transcriptional
exploration and showed mutant ALK-driven upregulation upregulation of hTERT.61,62 TERT rearrangements (23%),
of mitogen-activated protein kinase negative feedback ATRX deletions (11%), and MYCN amplifications (37%) iden-
regulators, among others, and upregulation of RET and RET- tified three almost nonoverlapping groups of high-stage
driven sympathetic neuronal markers of the cholinergic neuroblastoma, each associated with very poor prognosis.62
lineage.50 Although this most likely connects ALK and RET Of further interest, sequencing whole genomes of di-
signaling in a developmental context (in keeping with agnostic and relapsed tumors has provided both insight into
other observations),51,52 this observation also offers novel the nature of mutations driving therapy resistance and
venues for drug treatments that are currently under molecular evidence for the existence of minor subclones
investigation. that emerge during therapy (e.g., ALK mutant subclones
ALK has been connected to MYCN through multiple in otherwise ALK wild-type tumors at diagnosis, among
mechanisms. ALK was shown to drive MYCN expression others). 63,64
by promotor activation53 and activation through ERK5.54
In addition, activated ALK downstream signaling through
phosphoinositide 3-kinase/AKT controls glycogen synthase NEUROBLASTOMA THERAPY UPDATE
kinase 3 beta activity and MYCN protein stabilization. These Because of the heterogeneous biology of neuroblastoma, its
mechanisms explain the observed cooperative effect of clinical behavior and prognosis range from near uniform
mutant ALK accelerated tumor formation in MYCN trans- survival to high risk for fatal demise. Significant advances in
genic mice and zebrafish.29,31,55 our knowledge of neuroblastoma biology have led to im-
proved prognostic stratification and appropriate tailoring of
therapy, although our evolving molecular understanding has
The Whole Genome and Nothing But the Whole yet to translate fully into novel therapies. The International
Genome Risk Group (INRG) classification system uses clinical and
In keeping with recent findings in other embryonal tumors, biologic factors that have been shown to predict progno-
the overall mutation burden was low, with an average sis and risk of recurrence, including age, International
of fewer than 10 mutations. 56-59 No additional recur- Neuroblastoma Staging System stage histopathology, DNA
rent mutations with significant frequency, such as ALK- index (ploidy), segmental chromosomal gains or losses, and
activating mutations, were found; however, Molenaar MYCN amplification,65 to classify patients as having low-risk,
et al56 collectively demonstrated that an increase in im- intermediate-risk, and high-risk neuroblastoma. In addition, a
plication of low-frequency mutations of neuritogenesis National Cancer Institute–sponsored Clinical Trials Planning
genes was observed overall. In addition, chromothripsis, Meeting has proposed revisions to the International Response
a complex pattern of interchromosomal and/or intra- Criteria. These revised criteria will integrate tumor response
chromosomal rearrangements believed to arise through in the primary tumor, soft tissue and bone metastases, and
a single time-point event, was also demonstrated in sev- bone marrow. Primary and metastatic soft tissue sites will
eral cases. Perhaps the main power of whole-genome se- be assessed using RECIST and 123I-metaiodobenzylguanidine
quencing was the possibility to combine both mutation (MIBG) or 18F-fluorodeoxyglucose (FDG)–PET scans if the
and copy-number events at single base-pair resolution. By tumor is MIBG nonavid. 123I-MIBG or FDG-PET scans, for
combining such data, further recurrent events emerged, MIBG nonavid disease, replace 99technetium bone scans for
including the ARID1A and ARID1B epigenetic components osteomedullary metastases assessment. Bone marrow
of the SWI/SNF remodeling complex and, most notably, will be assessed by histology/immunohistochemistry and
ATRX inactivating events (mainly deletions) and hTERT cytology/immunocytology. Urinary catecholamine levels
activating structural rearrangements. ATRX encodes a SWI/ will not be included in response assessment. Overall re-
SNF chromatin-remodeling ATP-dependent helicase; re- sponse will be defined as complete response, partial re-
markably, ATRX mutations occur mainly in older children, sponse, minor response, stable disease, and progressive
with a frequency rising to greater than 40% in children disease. These international collaborations provide the

e550 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


Downloaded from ascopubs.org by 181.53.178.179 on May 1, 2021 from 181.053.178.179
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
NEUROBLASTOMA BIOLOGY, THERAPY, AND SURVIVORSHIP

opportunity for comparison across international clinical High-Risk Neuroblastoma: Beyond Chemotherapy
trials and have opened the door for international collab- Dose Intensity
orative trials. High-risk neuroblastoma is defined by presence of tumor
MYCN amplification or children with metastatic disease
older than age 18 months at diagnosis. High-risk neuroblas-
Neuroblastoma: When Less Is More toma is largely chemotherapy responsive, but approxi-
Low-risk disease is characterized by a localized primary mately one-half of patients still experience disease relapse
tumor (INRGSS L1) or INRGSS stage MS without the pres- despite improvements in overall response rates. Standard
ence of MYCN amplification. Survival rates for patients therapy for patients with high-risk neuroblastoma has
with low-risk neuroblastoma are excellent with surgery evolved through the results of randomized clinical trials
alone and rare recurrences can often be cured with sal- performed by international cooperative groups and in-
vage chemotherapy, resulting in survival rates of greater volves three components: multi-agent chemotherapy in-
than 95%. 66-68 Chemotherapy is reserved for patients duction and surgical resection, consolidation using high-
with localized neuroblastoma who have life- or organ- dose chemotherapy with autologous hematopoietic stem
threatening symptoms or the minority of patients who cell rescue, and postconsolidation/maintenance treatment
experience recurrence or progressive disease. Several of minimal residual disease using a biologic differentiating
clinical trials have further established the potential for agent (isotretinoin) and immunotherapy with antiganglioside
spontaneous regression of localized neuroblastoma, es- (GD2) antibody plus cytokines.
pecially among patients who are diagnosed at younger Dose intensity of chemotherapy is an important factor
than age 12 months.69-71 In addition, recent clinical trials in successful treatment of high-risk neuroblastoma. Dose-
support a continued reduction of chemotherapy expo- intensive multi-agent induction chemotherapy provided im-
sure and surgery for the majority of low-risk asymp- proved event-free survival compared with a less-intensive
tomatic patients, and studies to examine strategies to chemotherapy regimen.77 Additional randomized trials
biologically identify the rare patient with localized dis- demonstrated that the use of myeloablative therapy and
ease who will have a poor outcome are underway. autologous hematopoietic stem cell rescue further im-
Intermediate-risk classification encompasses a wide proved outcomes for high-risk neuroblastoma compared
spectrum of disease for which surgical resection and with use of conventionally dosed chemotherapy or
moderate-dose multi-agent chemotherapy are the back- observation.78-80 More recently, randomized clinical trials
bone of therapy. Intermediate risk includes subsets of demonstrated that the specific agents used for consolidation
patients with locoregional disease (INRGSS L2) without chemotherapy (busulfan/melphalan)81 or the intensity of
MYCN amplification and infants (younger than 18 months consolidation therapy (tandem transplant; J. Park, personal
at diagnosis) with metastatic disease and favorable tumor communication, April 2016) may further improve outcomes
biologic features including no MCYNA. Survival after for high-risk neuroblastoma.
moderate-dose chemotherapy including carboplatin or Randomized clinical trials have also highlighted the role
cisplatin, doxorubicin, etoposide, and cyclophosphamide is of treating minimal residual disease to improve outcomes
greater than 90% for patients whose tumors exhibit fa- for patients with high-risk neuroblastoma. Isotretinoin, a
noncytotoxic differentiating agent, led to improved event-
vorable clinical and biologic characteristics, including in-
free survival from high-risk neuroblastoma. Furthermore,
fants with stage M who lack MYCN amplification. 72,73
the addition of immunotherapy utilizing anti-GD2 antibody
Extent of surgical resection does not affect prognosis.72
plus cytokines improved event-free and overall survival,
Ongoing studies are investigating whether further reduction
becoming the first example of the benefits of immune-
(and, in some cases, elimination) of therapy for the majority
targeted therapy for treatment of a solid tumor in
of patients with favorable histology and genomics can be
childhood.
achieved.
Although outcomes for patients with high-risk neuro-
Despite the overall excellent prognosis for the majority of
blastoma have markedly improved over the past 2 decades,
patients with intermediate-risk neuroblastoma, the prog-
our current therapies for these patients remain subop-
nosis is less favorable for patients older than 18 months with timal. Chemotherapy and radiotherapy resistance remain
locoregional disease (INRGSS L2) and unfavorable histology the hallmark of failure. However, optimism remains as our
or segmental chromosomal aberrations or those infants expanding knowledge of tumor biology provides ample
with metastatic disease with unfavorable biologic charac- opportunities for the introduction of novel molecularly
teristics. Event-free survival for this rare cohort of patients targeted agents into an optimized backbone of chemo-
ranges from 40% to 70%72,74-76 with a wide range of therapy therapy and radiotherapy. Novel therapeutic approaches
intensity used. Improved understanding of the underlying may target more generalizable features of neuroblastoma,
tumor biology and international collaborative clinical trials including surface epitopes such as GD2 for immuno-
are needed to better define optimal therapy for this rare therapy 82 and targeted radionuclide delivery. Alterna-
cohort of patients. tively, the molecularly targeted therapy paradigm includes

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e551


Downloaded from ascopubs.org by 181.53.178.179 on May 1, 2021 from 181.053.178.179
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
SPELEMAN, PARK, AND HENDERSON

identification and pharmacologic antagonism of an onco- chemotherapy may augment activity and will be studied in
genic driver as a novel therapeutic. up-front treatment of children whose neuroblastoma har-
Ongoing and planned clinical trials will exploit the ra- bors ALK aberrations. Genetic studies of relapsed neuro-
diosensitivity of neuroblastoma by incorporating 131I-MIBG blastoma demonstrate genetic evolution of tumors and
targeted radiotherapy. Neuroblastoma is one of the most further support combination of molecularly targeted agents
radiosensitive tumors of childhood, resulting in a marked for treatment of relapsed disease.64
decreased in primary site-specific recurrence, yet 50% of
patients have recurrence in bone marrow and/or cortical
bone.83 Efforts to radiate all sites of metastatic disease NEUROBLASTOMA SURVIVORSHIP
using conventional total body irradiation (TBI) have not As described, there has been tremendous success in the
clearly benefited patients, in part because of toxicity- treatment of neuroblastoma. Five-year relative survival has
associated limitations of TBI beyond a dose of 1,000 to increased from 54% among patients diagnosed between
1,200 cGy and associated second malignancies.84 MIBG, 1975 and 1984 to 78% for those diagnosed between 2005
a norepinephrine analog, is concentrated selectively in sym- and 2011.99 Subsequently, there has been a growing pop-
pathetic tissues. Clinical trials have demonstrated the ulation of survivors of neuroblastoma. Consistent with other
safety of 131I-MIBG (12–18 mCi/kg) administered with childhood cancer survivors, these advances have not come
chemotherapy, and objective response rates of up to 25% without a cost.
have been documented among patients with recurrent and The long-term outcomes of 954 survivors of neuroblas-
refractory disease.85-87 131I-MIBG can be safely combined toma diagnosed between 1970 and 1986 who are partici-
with myeloablative chemotherapy for patients with a poor pants in the North American cohort, the Childhood Cancer
response to induction chemotherapy with additional an- Survivor Study (CCSS), were reported in 2009.100 The 25-year
titumor efficacy.88 cumulative late mortality was 6%. Compared with the sex-
Future therapies must also build on the success of anti- and age-matched general population, the risk of death was
GD2 monoclonal antibody immunotherapy and develop almost sixfold higher (standardized mortality ratio [SMR],
novel approaches to enhance antibody efficacy, such as the 5.6; 95% CI, 4.4–6.9). The most common cause of late
addition of lenolidomide,89 which increases cytokine levels mortality was recurrence, followed by an SMN (SMR, 10.9;
and activates natural killer cells, combination with cytotoxic 95% CI, 5.8–18.7), pulmonary complications (SMR,
therapy that may alter the inhibitory tumor microenviron- 11.4; 95% CI, 3.1–29.3), and cardiac complications (SMR, 5.0;
ment (R. Mody, personal communication, April 2016) or 95% CI, 1.0–14.5). The 20-year cumulative incidence of
active immunization with anti-idiotype antibodies are being a chronic health condition was 41.1%. Compared with
studied among patients with relapsed disease.90 Promising their siblings, survivors had an 8.3-fold risk of developing a
novel immunotherapies genetically modify a patient’s own chronic health condition. Survivors of neuroblastoma were
T cells to be redirected against tumor-associated antigens at increased risk for at least one of the following health
(e.g., GD2, L1CAM). Autologous T cells engineered to over- complications: neurologic, musculoskeletal, endocrine, and
express chimeric antigen receptors are infused and have sensory complications, with 20-year cumulative incidences
been shown to persist and demonstrate antitumor activity of 30%, 8%, 8%, and 9%, respectively. In the CCSS cohort,
among patients with neuroblastoma.91,92 30 survivors of neuroblastoma developed an SMN 5 years
Translation of our evolving understanding of neuroblas- or later after the neuroblastoma diagnosis. Of the patients
toma biology and the increasing availability of active clinical with SMNs, eight had thyroid cancer, five had renal cell
compounds has greatly enhanced the availability of mo- carcinoma, three had soft tissue sarcomas, two had AML,
lecularly targeted agents. However, these remain in early- two had breast cancer, one had a brain tumor, one had
phase clinical trials for patients with recurrent high-risk Hodgkin lymphoma, and seven had other SMNs. The risk of
neuroblastoma, with the ultimate goal to bring those with developing an SMN was eightfold higher among survivors of
the most encouraging activity into front-line therapy. For neuroblastoma than the general population (standardized
patients with MYCN-amplified neuroblastoma, preclinical incidence ratio, 8.0; 95% CI, 5.4–11.4). Similar to other
studies suggest that BET bromodomain inhibitors can induce childhood cancer survivors, risks of late effects and SMNs
cell death by interfering with MYCN transcription.93 Other were attributable to radiation exposure (e.g., SMN, thyroid
drugs that have effects on MYCN stability (aurora kinase A disease, or cardiac disease), cisplatin exposure (e.g., hearing
and mTOR inhibitors)5 as well as those that target MYC- loss or renal disease), alkylator exposure (e.g., renal dis-
dependent metabolic changes (difluoromethylornithine)94 ease, infertility, or risk of treatment-related acute myeloid
are being studied. ALK is a pharmacologically tractable target leukemia/myelodysplastic syndrome), and other treatment
and early experience suggests that its inhibition can induce exposures.
responses in pediatric tumors with activating ALK muta- Notably, the outcomes reported from the CCSS reflect
tions.95,96 Emergent resistance to targeted therapeutics outcomes associated with the treatment and survival suc-
is a concern.97,98 A major issue is that ALK mutations cess of an earlier era for neuroblastoma. Although data on
confer differential sensitivity to available ALK inhibitors.98 stage and/or risk group were not available in the CCSS
The combination of an ALK inhibitor with cytotoxic analysis, only 20 survivors had undergone bone marrow

e552 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


Downloaded from ascopubs.org by 181.53.178.179 on May 1, 2021 from 181.053.178.179
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
NEUROBLASTOMA BIOLOGY, THERAPY, AND SURVIVORSHIP

transplantation, suggesting that most of the participants did treatment with TBI.103,108 Survivors with growth failure
not have high-risk disease.100 Thus, these data do not reflect often exhibit suboptimal responses to growth hormone (GH)
the late effects or SMNs experienced by patients with re- therapy.109,110 The etiology of this striking growth failure
cently treated neuroblastoma, such as high-risk patients and inadequate GH response is unclear and may be at-
who received more modern therapies such as tandem he- tributable to compromised spine growth after radiation.
matopoietic stem cell transplantation, biologic therapies, Recent data have suggested that it may also be a conse-
and immunotherapy. Currently, survivorship researchers are quence of premature epiphyseal closure caused by 13-cis-
focused on the questions of whether risk-adapted therapies retinoic acid exposure.103,111 Other prevalent endocrine late
improve the late effect profiles of low- and intermediate-risk effects include hypothyroidism, abnormal glucose meta-
survivors and, importantly, identifying the long-term out- bolism, and delayed/abnormal pubertal development and
comes of high-risk survivors exposed to therapies never maturation.103,111,112 The high prevalence of hypothyroid-
given alone or in combination to any other population of ism in survivors of neuroblastoma reflects damage from
childhood cancer survivors. diagnostic exposure to radioactive iodine tracers (131I-MIBG)
Regarding the effects of risk-based therapy on the long- and, in some cases, therapeutic radiation therapy that
term outcomes of survivors of neuroblastoma, a recent involves the thyroid.113 A number of investigators reported
study examined second cancers in the Surveillance, Epide- laboratory evidence of diabetes and metabolic syndrome
miology, and End Results database across treatment eras. among survivors of high-risk neuroblastoma.112,114 Female
Among 2,801 patients with neuroblastoma treated between survivors may demonstrate abnormal pubertal progression
1973 and 2006 (era 1, 1973 to 1989; era 2, 1990 to 1996; and premature ovarian failure; puberty and gonadal func-
and era 3, 1997 to 2006), 34 patients developed an SMN tion has not been characterized in male individuals.103 In
(14 developed a carcinoma and 10 had a hematologic malig- addition to endocrine late effects, studies of survivors of
nancy). Importantly, there was no difference in the incidence of high-risk neuroblastoma suggest elevated rates of sensori-
SMNs in era 1 compared with era 3. Consistent with other neural hearing loss,104-108 cardiac dysfunction,103,104,108
institutional studies reporting a high incidence of SMNs in hypertension/renal dysfunction,104-108 and neurocognitive
high-risk survivors,101 the 30-year cumulative incidence of deficits including behavioral problems as well as speech
SMNs among high-risk patients was 10.4% (95% CI, 4.0–20.5) and/or learning disabilities.103,105 Importantly, these studies
compared with 3.6% among lower-risk patients.102 A large, were small institutional studies limiting investigators to
comprehensive analysis of SMNs among these high-risk have the power to describe true incidence and risk of late
survivors is necessary to examine the effect of modern effects and identify the risk factors associated with their
regimens and exposures (including the combination of ra- development.
diotherapy, chemotherapy, cis-retinoic acid, immunother- Recognizing the gap in knowledge regarding the com-
apy, as well as 131I-MIBG) on risk for SMNs. bined effects on morbidity and quality of life of high-dose
Current knowledge regarding the long-term outcomes of chemotherapy, radiation, and non-TBI–based stem cell
high-risk survivors of neuroblastoma treated with more transplantation in combination with cis-retinoic acid, im-
modern approaches is limited to small institutional series. munocytokines, and immunotherapy in high-risk survivors
These studies suggest a high burden of multiple late effects of neuroblastoma, researchers have identified a cohort
for these survivors (summarized in Table 1).103-108 Endocrine of 5-year survivors (. 700) of high-risk neuroblastoma
late effects are prominent, with most series identifying treated between 2000 and 2011 who were enrolled in the
growth failure and short stature even in the absence of Children’s Oncology Group (COG) Neuroblastoma Biology

TABLE 1. Prevalence of Late Effects Among Survivors of High-Risk Neuroblastoma


Late Effect Cohen, 2014103 Laverdière, 2005104 Moreno, 2013105 Perwein, 2011106 Hobbie, 2008108 Trahair, 2007107
No. of Survivors 51 63 57 16 13 23
Late Effect
Cardiac 10 (20) 5 (8) — — 1 (8) —
Pulmonary — 12 (19) — 4 (25) 7 (54) —
Hypertension/Renal — 6 (9) 8 (14) 10 (63) 8 (61) 10 of 21 (47)
Growth Abnormality 10 (20) 6 (9) 4 (7) 7 (44) 7 (54) 13 of 13 (100)
Gonadal Failure 9 (12) 13 (41)* 2 (4) 2 (13) 2 (15) 5 of 6 (83)
Hypothyroid 29 (49) 15 (24) 1 (2) 8 (50) 7 (54) 4 of 14 (29)
Hearing Loss 37 (73) 39 (62) 28 (49) 6 (38) 12 (92) 11 of 15 (73)
Neurocognitive 11 (38) 8 (13) 12 (21) — — —
Data are given as numbers (percentages).
*Of 32 female survivors, 13 had ovarian failure.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e553


Downloaded from ascopubs.org by 181.53.178.179 on May 1, 2021 from 181.053.178.179
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
SPELEMAN, PARK, AND HENDERSON

Study ANBL00B1.115 The COG LEAHRN (Late Effects After High EPILOGUE: NEW TECHNIQUES, NEW
Risk Neuroblastoma) study (ALTE 15N2) plans to open in 2016 DISCOVERIES, NEW IDEAS...IN THAT ORDER
and will recruit almost 400 of these survivors for a cross- Referencing the quote of Nobel prize winner Sydney Brenner
sectional study to characterize the survivors’ late effects and above, it is fair to say that the past decade of neuroblastoma
SMNs, assess for predictors of these late effects and SMNs, research has been tremendously fruitful and, most impor-
and identify factors predictive of decreased health-related tantly, many novel insights in biology and emerging novel
quality of life. Furthermore, ALTE 15N2 will serve to es- drugs, not the least of those targeting MYCN or MYCN-driven
tablish a cohort of high-risk survivors of neuroblastoma, with pathways, are opening new exciting possibilities.7,93,118-123
stored peripheral blood samples, as a resource for future Now, we are facing a novel wave of technological de-
investigation. velopments that will further fuel discoveries and new ideas
Understanding of the late effects and SMNs of survivors is and venues for drug treatment. These include the study of
essential for both ensuring the long-term health of survivors long noncoding RNAs124,125; single cell profiling at the DNA,
of neuroblastoma as well as informing the design of future RNA, and chromatin levels126; novel and very powerful
neuroblastoma treatment trials aimed at improved survival proteome and interactome methods; metabolome profiling;
along with a concurrent reduction in late effects. While we and, importantly, further exploration of zebrafish and mouse
continue to refine our understanding of the late effects models using powerful novel genome-editing technolo-
of modern neuroblastoma treatments, all neuroblastoma gies.127,128 These efforts must continue to enhance our
survivors should continue to participate in lifelong risk- understanding of neuroblastoma clinical behavior and in-
based health care with surveillance and early intervention form the design of optimal therapy for children with neu-
of late effects and SMNs, as recommended by the Institute roblastoma, therapy that will enhance survival while
of Medicine and the COG.116,117 minimizing late effects.

References
1. Schwab M, Alitalo K, Klempnauer KH, et al. Amplified DNA with limited 14. Henrich KO, Bauer T, Schulte J, et al. CAMTA1, a 1p36 tumor sup-
homology to myc cellular oncogene is shared by human neuroblas- pressor candidate, inhibits growth and activates differentiation
toma cell lines and a neuroblastoma tumour. Nature. 1983;305: programs in neuroblastoma cells. Cancer Res. 2011;71:3142-3151.
245-248. 15. Liu Z, Rader J, He S, et al. CASZ1 inhibits cell cycle progression
2. Speleman F, De Preter K, Vandesompele J. Neuroblastoma genetics in neuroblastoma by restoring pRb activity. Cell Cycle. 2013;12:
and phenotype: a tale of heterogeneity. Semin Cancer Biol. 2011;21: 2210-2218.
238-244. 16. Fujita T, Igarashi J, Okawa ER, et al. CHD5, a tumor suppressor gene
3. Irwin MS, Park JR. Neuroblastoma: paradigm for precision medicine. deleted from 1p36.31 in neuroblastomas. J Natl Cancer Inst. 2008;100:
Pediatr Clin North Am. 2015;62:225-256. 940-949.
4. Louis CU, Shohet JM. Neuroblastoma: molecular pathogenesis and 17. Mossé YP, Laudenslager M, Longo L, et al. Identification of ALK as a
therapy. Annu Rev Med. 2015;66:49-63. major familial neuroblastoma predisposition gene. Nature. 2008;455:
5. Barone G, Anderson J, Pearson AD, et al. New strategies in neuro- 930-935.
blastoma: therapeutic targeting of MYCN and ALK. Clin Cancer Res. 18. Molenaar JJ, Domingo-Fernández R, Ebus ME, et al. LIN28B induces
2013;19:5814-5821. neuroblastoma and enhances MYCN levels via let-7 suppression. Nat
6. Cheung NK, Dyer MA. Neuroblastoma: developmental biology, Genet. 2012;44:1199-1206.
cancer genomics and immunotherapy. Nat Rev Cancer. 2013;13: 19. Janoueix-Lerosey I, Lequin D, Brugières L, et al. Somatic and germline
397-411. activating mutations of the ALK kinase receptor in neuroblastoma.
7. Bagatell R, Cohn SL. Genetic discoveries and treatment advances in Nature. 2008;455:967-970.
neuroblastoma. Curr Opin Pediatr. 2016;28:19-25. 20. Chen Y, Takita J, Choi YL, et al. Oncogenic mutations of ALK kinase in
8. Schulte JH, Eggert A. Neuroblastoma. Crit Rev Oncog. 2015;20: neuroblastoma. Nature. 2008;455:971-974.
245-270. 21. George RE, Sanda T, Hanna M, et al. Activating mutations in ALK
9. Jiang M, Stanke J, Lahti JM. The connections between neural crest provide a therapeutic target in neuroblastoma. Nature. 2008;455:
development and neuroblastoma. Curr Top Dev Biol. 2011;94: 975-978.
77-127. 22. Capasso M, Diskin S, Cimmino F, et al. Common genetic variants in
10. Nakagawara A. Trk receptor tyrosine kinases: a bridge between cancer NEFL influence gene expression and neuroblastoma risk. Cancer Res.
and neural development. Cancer Lett. 2001;169:107-114. 2014;74:6913-6924.
11. Nakagawara A, Ohira M. Comprehensive genomics linking between 23. Diskin SJ, Capasso M, Diamond M, et al. Rare variants in TP53 and
neural development and cancer: neuroblastoma as a model. Cancer susceptibility to neuroblastoma. J Natl Cancer Inst. 2014;106:dju047.
Lett. 2004;204:213-224. 24. Maris JM, Mosse YP, Bradfield JP, et al. Chromosome 6p22 locus
12. Schleiermacher G, Janoueix-Lerosey I, Delattre O. Recent insights into associated with clinically aggressive neuroblastoma. N Engl J Med.
the biology of neuroblastoma. Int J Cancer. 2014;135:2249-2261. 2008;358:2585-2593.
13. Domingo-Fernandez R, Watters K, Piskareva O, et al. The role of 25. Diskin SJ, Capasso M, Schnepp RW, et al. Common variation at 6q16
genetic and epigenetic alterations in neuroblastoma disease patho- within HACE1 and LIN28B influences susceptibility to neuroblastoma.
genesis. Pediatr Surg Int. 2013;29:101-119. Nat Genet. 2012;44:1126-1130.

e554 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


Downloaded from ascopubs.org by 181.53.178.179 on May 1, 2021 from 181.053.178.179
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
NEUROBLASTOMA BIOLOGY, THERAPY, AND SURVIVORSHIP

26. Capasso M, Devoto M, Hou C, et al. Common variations in BARD1 49. Bresler SC, Weiser DA, Huwe PJ, et al. ALK mutations confer differ-
influence susceptibility to high-risk neuroblastoma. Nat Genet. 2009; ential oncogenic activation and sensitivity to ALK inhibition therapy in
41:718-723. neuroblastoma. Cancer Cell. 2014;26:682-694.
27. Diskin SJ, Hou C, Glessner JT, et al. Copy number variation at 1q21.1 50. Lambertz I, Kumps C, Claeys S, et al. Upregulation of MAPK negative
associated with neuroblastoma. Nature. 2009;459:987-991. feedback regulators and RET in mutant ALK neuroblastoma: impli-
28. Wang K, Diskin SJ, Zhang H, et al. Integrative genomics identifies LMO1 cations for targeted treatment. Clin Cancer Res. 2015;21:3327-3339.
as a neuroblastoma oncogene. Nature. 2011;469:216-220. 51. Cazes A, Lopez-Delisle L, Tsarovina K, et al. Activated Alk triggers
29. Zhu S, Lee JS, Guo F, et al. Activated ALK collaborates with MYCN in prolonged neurogenesis and Ret upregulation providing a thera-
neuroblastoma pathogenesis. Cancer Cell. 2012;21:362-373. peutic target in ALK-mutated neuroblastoma. Oncotarget. 2014;5:
30. Althoff K, Beckers A, Bell E, et al. A Cre-conditional MYCN-driven 2688-2702.
neuroblastoma mouse model as an improved tool for preclinical 52. Reiff T, Huber L, Kramer M, et al. Midkine and Alk signaling in
studies. Oncogene. 2015;34:3357-3368. sympathetic neuron proliferation and neuroblastoma predisposition.
31. Heukamp LC, Thor T, Schramm A, et al. Targeted expression of mu- Development. 2011;138:4699-4708.
tated ALK induces neuroblastoma in transgenic mice. Sci Transl Med. 53. Schönherr C, Ruuth K, Kamaraj S, et al. Anaplastic lymphoma kinase
2012;4:141ra91. (ALK) regulates initiation of transcription of MYCN in neuroblastoma
32. Henssen A, Althoff K, Odersky A, et al. Targeting MYCN-driven cells. Oncogene. 2012;31:5193-5200.
transcription by BET-bromodomain inhibition. Clin Cancer Res. 54. Umapathy G, El Wakil A, Witek B, et al. The kinase ALK stimulates the
Epub 2015 Dec 2. kinase ERK5 to promote the expression of the oncogene MYCN in
33. Huang M, Weiss WA. Neuroblastoma and MYCN. Cold Spring Harb neuroblastoma. Sci Signal. 2014;7:ra102.
Perspect Med. 2013;3:a014415. 55. Berry T, Luther W, Bhatnagar N, et al. The ALK(F1174L) mutation
34. Seton-Rogers S. RNA splicing: MYC maintains high-fidelity splicing. Nat potentiates the oncogenic activity of MYCN in neuroblastoma. Cancer
Rev Cancer. 2015;15:385. Cell. 2012;22:117-130.
35. Zhang S, Wei JS, Li SQ, et al. MYCN controls an alternative RNA splicing 56. Molenaar JJ, Koster J, Zwijnenburg DA, et al. Sequencing of neuro-
program in high-risk metastatic neuroblastoma. Cancer Lett. 2016; blastoma identifies chromothripsis and defects in neuritogenesis
371:214-224. genes. Nature. 2012;483:589-593.
36. Walz S, Lorenzin F, Morton J, et al. Activation and repression by 57. Pugh TJ, Morozova O, Attiyeh EF, et al. The genetic landscape of high-
oncogenic MYC shape tumour-specific gene expression profiles. risk neuroblastoma. Nat Genet. 2013;45:279-284.
Nature. 2014;511:483-487. 58. Cheung NK, Zhang J, Lu C, et al; St Jude Children’s Research
37. Kress TR, Sabò A, Amati B. MYC: connecting selective transcrip- Hospital–Washington University Pediatric Cancer Genome Project.
tional control to global RNA production. Nat Rev Cancer. 2015;15: Association of age at diagnosis and genetic mutations in patients with
593-607. neuroblastoma. JAMA. 2012;307:1062-1071.
38. Conacci-Sorrell M, McFerrin L, Eisenman RN. An overview of MYC 59. Sausen M, Leary RJ, Jones S, et al. Integrated genomic analyses identify
and its interactome. Cold Spring Harb Perspect Med. 2014;4: ARID1A and ARID1B alterations in the childhood cancer neuroblas-
a014357. toma. Nat Genet. 2013;45:12-17.
39. Wang C, Liu Z, Woo CW, et al. EZH2 mediates epigenetic silencing of 60. Kumps C, Fieuw A, Mestdagh P, et al. Focal DNA copy number changes
neuroblastoma suppressor genes CASZ1, CLU, RUNX3, and NGFR. in neuroblastoma target MYCN regulated genes. PLoS One. 2013;8:
Cancer Res. 2012;72:315-324. e52321.
40. Schulte JH, Horn S, Otto T, et al. MYCN regulates oncogenic microRNAs 61. Peifer M, Hertwig F, Roels F, et al. Telomerase activation by geno-
in neuroblastoma. Int J Cancer. 2008;122:699-704. mic rearrangements in high-risk neuroblastoma. Nature. 2015;526:
41. De Brouwer S, Mestdagh P, Lambertz I, et al. Dickkopf-3 is regulated by 700-704.
the MYCN-induced miR-17-92 cluster in neuroblastoma. Int J Cancer. 62. Valentijn LJ, Koster J, Zwijnenburg DA, et al. TERT rearrangements are
2012;130:2591-2598. frequent in neuroblastoma and identify aggressive tumors. Nat Genet.
42. Mestdagh P, Boström AK, Impens F, et al. The miR-17-92 microRNA 2015;47:1411-1414.
cluster regulates multiple components of the TGF-b pathway in 63. Schramm A, Köster J, Assenov Y, et al. Mutational dynamics between
neuroblastoma. Mol Cell. 2010;40:762-773. primary and relapse neuroblastomas. Nat Genet. 2015;47:872-877.
43. Beckers A, Van Peer G, Carter DR, et al. MYCN-targeting miRNAs are 64. Eleveld TF, Oldridge DA, Bernard V, et al. Relapsed neuroblastomas
predominantly downregulated during MYCN‑driven neuroblastoma show frequent RAS-MAPK pathway mutations. Nat Genet. 2015;47:
tumor formation. Oncotarget. 2015;6:5204-5216. 864-871.
44. Cotterman R, Knoepfler PS. N-Myc regulates expression of pluripo- 65. Cohn SL, Pearson AD, London WB, et al; INRG Task Force. The In-
tency genes in neuroblastoma including lif, klf2, klf4, and lin28b. PLoS ternational Neuroblastoma Risk Group (INRG) classification system:
One. 2009;4:e5799. an INRG Task Force report. J Clin Oncol. 2009;27:289-297.
45. Marshall GM, Liu PY, Gherardi S, et al. SIRT1 promotes N-Myc on- 66. Strother DR, London WB, Schmidt ML, et al. Outcome after surgery
cogenesis through a positive feedback loop involving the effects of alone or with restricted use of chemotherapy for patients with low-
MKP3 and ERK on N-Myc protein stability. PLoS Genet. 2011;7: risk neuroblastoma: results of Children’s Oncology Group study
e1002135. P9641. J Clin Oncol. 2012;30:1842-1848.
46. Liu PY, Xu N, Malyukova A, et al. The histone deacetylase SIRT2 67. Perez CA, Matthay KK, Atkinson JB, et al. Biologic variables in the
stabilizes Myc oncoproteins. Cell Death Differ. 2013;20:503-514. outcome of stages I and II neuroblastoma treated with surgery as
47. Park JH, Szemes M, Vieira GC, et al. Protein arginine methyltransferase primary therapy: a children’s cancer group study. J Clin Oncol. 2000;
5 is a key regulator of the MYCN oncoprotein in neuroblastoma cells. 18:18-26.
Mol Oncol. 2015;9:617-627. 68. Simon T, Spitz R, Faldum A, et al. New definition of low-risk neuro-
48. Crescenzo R, Inghirami G. Anaplastic lymphoma kinase inhibitors. Curr blastoma using stage, age, and 1p and MYCN status. J Pediatr Hematol
Opin Pharmacol. 2015;23:39-44. Oncol. 2004;26:791-796.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e555


Downloaded from ascopubs.org by 181.53.178.179 on May 1, 2021 from 181.053.178.179
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
SPELEMAN, PARK, AND HENDERSON

69. Nuchtern JG, London WB, Barnewolt CE, et al. A prospective study of 86. French S, DuBois SG, Horn B, et al. 131I-MIBG followed by consoli-
expectant observation as primary therapy for neuroblastoma in young dation with busulfan, melphalan and autologous stem cell trans-
infants: a Children’s Oncology Group study. Ann Surg. 2012;256: plantation for refractory neuroblastoma. Pediatr Blood Cancer.
573-580. 2013;60:879-884.
70. Rubie H, De Bernardi B, Gerrard M, et al. Excellent outcome with 87. Gaze MN, Chang YC, Flux GD, et al. Feasibility of dosimetry-based high-
reduced treatment in infants with nonmetastatic and unresectable dose 131I-meta-iodobenzylguanidine with topotecan as a radio-
neuroblastoma without MYCN amplification: results of the pro- sensitizer in children with metastatic neuroblastoma. Cancer Biother
spective INES 99.1. J Clin Oncol. 2011;29:449-455. Radiopharm. 2005;20:195-199.
71. Simon T, Hero B, Benz-Bohm G, et al. Review of image defined risk 88. Yanik GA, Villablanca JG, Maris JM, et al. 131I-metaiodobenzylguanidine
factors in localized neuroblastoma patients: results of the GPOH NB97 with intensive chemotherapy and autologous stem cell transplantation
trial. Pediatr Blood Cancer. 2008;50:965-969. for high-risk neuroblastoma. A new approaches to neuroblastoma
72. Baker DL, Schmidt ML, Cohn SL, et al; Children’s Oncology Group. therapy (NANT) phase II study. Biol Blood Marrow Transplant. 2015;
Outcome after reduced chemotherapy for intermediate-risk neuro- 21:673-681.
blastoma. N Engl J Med. 2010;363:1313-1323. 89. Xu Y, Sun J, Sheard MA, et al. Lenalidomide overcomes suppression of
73. De Bernardi B, Gerrard M, Boni L, et al. Excellent outcome with human natural killer cell anti-tumor functions by neuroblastoma
reduced treatment for infants with disseminated neuroblastoma microenvironment-associated IL-6 and TGFb1. Cancer Immunol
without MYCN gene amplification. J Clin Oncol. 2009;27:1034-1040. Immunother. 2013;62:1637-1648.
74. Oberthuer A, Juraeva D, Hero B, et al. Revised risk estimation and 90. Koehn TA, Trimble LL, Alderson KL, et al. Increasing the clinical
treatment stratification of low- and intermediate-risk neuroblastoma efficacy of NK and antibody-mediated cancer immunotherapy:
patients by integrating clinical and molecular prognostic markers. Clin potential predictors of successful clinical outcome based on ob-
Cancer Res. 2015;21:1904-1915. servations in high-risk neuroblastoma. Front Pharmacol. 2012;3:91.
75. Schleiermacher G, Mosseri V, London WB, et al. Segmental chro- 91. Park JR, Digiusto DL, Slovak M, et al. Adoptive transfer of chimeric
mosomal alterations have prognostic impact in neuroblastoma: antigen receptor re-directed cytolytic T lymphocyte clones in patients
a report from the INRG project. Br J Cancer. 2012;107:1418-1422. with neuroblastoma. Mol Ther. 2007;15:825-833.
76. Meany HJ, London WB, Ambros PF, et al. Significance of clinical and 92. Louis CU, Brenner MK. Cellular immunotherapy for neuroblastoma:
biologic features in stage 3 neuroblastoma: a report from the In- a review of current vaccine and adoptive T cell therapeutics. Curr
ternational Neuroblastoma Risk Group project. Pediatr Blood Cancer. Pharm Des. 2009;15:424-429.
2014;61:1932-1939. 93. Puissant A, Frumm SM, Alexe G, et al. Targeting MYCN in neuro-
77. Pearson AD, Pinkerton CR, Lewis IJ, et al; European Neuroblastoma blastoma by BET bromodomain inhibition. Cancer Discov. 2013;3:
Study Group; Children’s Cancer and Leukaemia Group (CCLG formerly 308-323.
United Kingdom Children’s Cancer Study Group). High-dose rapid and 94. Bassiri H, Benavides A, Haber M, et al. Translational development of
standard induction chemotherapy for patients aged over 1 year with difluoromethylornithine (DFMO) for the treatment of neuroblastoma.
stage 4 neuroblastoma: a randomised trial. Lancet Oncol. 2008;9: Transl Pediatr. 2015;4:226-238.
247-256. 95. Mossé YP, Lim MS, Voss SD, et al. Safety and activity of crizotinib for
78. Matthay KK, Villablanca JG, Seeger RC, et al; Children’s Cancer Group. paediatric patients with refractory solid tumours or anaplastic large-
Treatment of high-risk neuroblastoma with intensive chemotherapy, cell lymphoma: a Children’s Oncology Group phase 1 consortium
radiotherapy, autologous bone marrow transplantation, and 13- study. Lancet Oncol. 2013;14:472-480.
cis-retinoic acid. N Engl J Med. 1999;341:1165-1173. 96. Mossé YP, Wood A, Maris JM. Inhibition of ALK signaling for cancer
79. Berthold F, Boos J, Burdach S, et al. Myeloablative megatherapy with therapy. Clin Cancer Res. 2009;15:5609-5614.
autologous stem-cell rescue versus oral maintenance chemotherapy 97. Carpenter EL, Haglund EA, Mace EM, et al. Antibody targeting of
as consolidation treatment in patients with high-risk neuroblastoma: anaplastic lymphoma kinase induces cytotoxicity of human neuro-
a randomised controlled trial. Lancet Oncol. 2005;6:649-658. blastoma. Oncogene. 2012;31:4859-4867.
80. Pritchard J, Cotterill SJ, Germond SM, et al. High dose melphalan in the 98. Bresler SC, Wood AC, Haglund EA, et al. Differential inhibitor sensi-
treatment of advanced neuroblastoma: results of a randomised trial tivity of anaplastic lymphoma kinase variants found in neuroblastoma.
(ENSG-1) by the European Neuroblastoma Study Group. Pediatr Blood Sci Transl Med. 2011;3:108ra114.
Cancer. 2005;44:348-357. 99. Altekruse S, Krapcho M, Neyman N, et al. (eds). SEER Cancer Statistics
81. Ladenstein RL, Poetschger U, Luksch R, et al. Busulphan-melphalan Review 1975–2007. Bethesda, MD: National Cancer Institute; 2010.
as a myeloablative therapy (MAT) for high-risk neuroblastoma: results http://seer.cancer.gov/csr/1975_2007/,/csr/1975_2007/., based on
from the HR-NBL1/SIOPEN trial. J Clin Oncol. 2011;29 (suppl; abstr 2). November 2009 SEER data submission, posted to the SEER web site.
82. Yu AL, Gilman AL, Ozkaynak MF, et al; Children’s Oncology Group. Anti- Accessed April 7, 2016.
GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neu- 100. Laverdière C, Liu Q, Yasui Y, et al. Long-term outcomes in survivors of
roblastoma. N Engl J Med. 2010;363:1324-1334. neuroblastoma: a report from the Childhood Cancer Survivor Study.
83. Matthay KK, Atkinson JB, Stram DO, et al. Patterns of relapse after J Natl Cancer Inst. 2009;101:1131-1140.
autologous purged bone marrow transplantation for neuroblastoma: 101. Martin A, Schneiderman J, Helenowski IB, et al. Secondary malignant
a Childrens Cancer Group pilot study. J Clin Oncol. 1993;11:2226-2233. neoplasms after high-dose chemotherapy and autologous stem cell
84. Flandin I, Hartmann O, Michon J, et al. Impact of TBI on late effects in rescue for high-risk neuroblastoma. Pediatr Blood Cancer. 2014;61:
children treated by megatherapy for stage IV neuroblastoma. A study 1350-1356.
of the French Society of Pediatric Oncology. Int J Radiat Oncol Biol 102. Applebaum MA, Henderson TO, Lee SM, et al. Second malignancies in
Phys. 2006;64:1424-1431. patients with neuroblastoma: the effects of risk-based therapy.
85. DuBois SG, Allen S, Bent M, et al. Phase I/II study of (131)I-MIBG with Pediatr Blood Cancer. 2015;62:128-133.
vincristine and 5 days of irinotecan for advanced neuroblastoma. 103. Cohen LE, Gordon JH, Popovsky EY, et al. Late effects in children
Br J Cancer. 2015;112:644-649. treated with intensive multimodal therapy for high-risk neuroblastoma:

e556 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


Downloaded from ascopubs.org by 181.53.178.179 on May 1, 2021 from 181.053.178.179
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
NEUROBLASTOMA BIOLOGY, THERAPY, AND SURVIVORSHIP

high incidence of endocrine and growth problems. Bone Marrow Paper presented at: 46th Congress of the International Society of
Transplant. 2014;49:502-508. Paediatric Oncology; October 2014; Toronto, ON, Canada.
104. Laverdière C, Cheung NK, Kushner BH, et al. Long-term complications 116. Hewitt M, Weiner SL, Simone JV (eds); National Cancer Policy Board;
in survivors of advanced stage neuroblastoma. Pediatr Blood Cancer. Institute of Medicine; National Research Council. Childhood Cancer
2005;45:324-332. Survivorship: Improving Care and Quality of Life. Washington, DC:
105. Moreno L, Vaidya SJ, Pinkerton CR, et al; European Neuroblastoma National Academies Press; 2003.
Study Group; Children’s Cancer and Leukaemia Group (CCLG) (formerly 117. Children’s Oncology Group. Long-Term Follow-up Guidelines for
UKCCSG). Long-term follow-up of children with high-risk neuroblastoma: Survivors of Childhood, Adolescent, and Young Adult Cancers, version
the ENSG5 trial experience. Pediatr Blood Cancer. 2013;60:1135-1140. 4.0. Monrovia, CA: Children’s Oncology Group; 2014.
106. Perwein T, Lackner H, Sovinz P, et al. Survival and late effects in children 118. Ham J, Costa C, Sano R, et al. Exploitation of the apoptosis-primed
with stage 4 neuroblastoma. Pediatr Blood Cancer. 2011;57:629-635. state of MYCN-amplified neuroblastoma to develop a potent and
107. Trahair TN, Vowels MR, Johnston K, et al. Long-term outcomes in specific targeted therapy combination. Cancer Cell. 2016;29:159-172.
children with high-risk neuroblastoma treated with autologous stem 119. Chayka O, D’Acunto CW, Middleton O, et al. Identification and
cell transplantation. Bone Marrow Transplant. 2007;40:741-746. pharmacological inactivation of the MYCN gene network as a ther-
108. Hobbie WL, Moshang T, Carlson CA, et al. Late effects in survivors of apeutic strategy for neuroblastic tumor cells. J Biol Chem. 2015;290:
tandem peripheral blood stem cell transplant for high-risk neuro- 2198-2212.
blastoma. Pediatr Blood Cancer. 2008;51:679-683. 120. Sottile F, Gnemmi I, Cantilena S, et al. A chemical screen identifies the
109. Olshan JS, Gubernick J, Packer RJ, et al. The effects of adjuvant chemotherapeutic drug topotecan as a specific inhibitor of the
chemotherapy on growth in children with medulloblastoma. Cancer. B-MYB/MYCN axis in neuroblastoma. Oncotarget. 2012;3:535-545.
1992;70:2013-2017. 121. Molenaar JJ, Ebus ME, Geerts D, et al. Inactivation of CDK2 is syn-
110. Willi SM, Cooke K, Goldwein J, et al. Growth in children after bone thetically lethal to MYCN over-expressing cancer cells. Proc Natl Acad
marrow transplantation for advanced neuroblastoma compared with Sci USA. 2009;106:12968-12973.
growth after transplantation for leukemia or aplastic anemia. 122. He S, Liu Z, Oh DY, et al. MYCN and the epigenome. Front Oncol. 2013;
J Pediatr. 1992;120:726-732. 3:1.
111. Hobbie WL, Mostoufi SM, Carlson CA, et al. Prevalence of advanced 123. Moore NF, Azarova AM, Bhatnagar N, et al. Molecular rationale for
bone age in a cohort of patients who received cis-retinoic acid for the use of PI3K/AKT/mTOR pathway inhibitors in combination with
high-risk neuroblastoma. Pediatr Blood Cancer. 2011;56:474-476. crizotinib in ALK-mutated neuroblastoma. Oncotarget. 2014;5:
112. van Waas M, Neggers SJ, Raat H, et al. Abdominal radiotherapy: 8737-8749.
a major determinant of metabolic syndrome in nephroblastoma and 124. Mestdagh P, Fredlund E, Pattyn F, et al. An integrative genomics
neuroblastoma survivors. PLoS One. 2012;7:e52237. screen uncovers ncRNA T-UCR functions in neuroblastoma tumours.
113. van Santen HM, de Kraker J, van Eck BL, et al. High incidence of Oncogene. 2010;29:3583-3592.
thyroid dysfunction despite prophylaxis with potassium iodide during 125. Pandey GK, Kanduri C. Long noncoding RNAs and neuroblastoma.
(131)I-meta-iodobenzylguanidine treatment in children with neuro- Oncotarget. 2015;6:18265-18275.
blastoma. Cancer. 2002;94:2081-2089. 126. Gawad C, Koh W, Quake SR. Single-cell genome sequencing: current
114. Meacham LR, Sklar CA, Li S, et al. Diabetes mellitus in long-term state of the science. Nat Rev Genet. 2016;17:175-188.
survivors of childhood cancer. Increased risk associated with radiation 127. Olivares O, Däbritz JH, King A, et al. Research into cancer meta-
therapy: a report for the childhood cancer survivor study. Arch Intern bolomics: towards a clinical metamorphosis. Semin Cell Dev Biol. 2015;
Med. 2009;169:1381-1388. 43:52-64.
115. Mody R, London WB, Park JR, et al. Late recurrences, mortality and 128. White R, Rose K, Zon L. Zebrafish cancer: the state of the art and the
second cancers in five-year survivors of high risk neuroblastoma. path forward. Nat Rev Cancer. 2013;13:624-636.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e557


Downloaded from ascopubs.org by 181.53.178.179 on May 1, 2021 from 181.053.178.179
Copyright © 2021 American Society of Clinical Oncology. All rights reserved.

You might also like