Pediatric cancer outcomes have significantly improved, and yet this success is not spread equally across cancer types or patients. Disparities data in pediatric oncology highlight needed improvements in access to care, including clinical trials and advanced testing for all patients. For cancers such as brain tumors and sarcomas, continued advancement in understanding the biology of tumor heterogeneity is an essential step toward finding new therapeutic combinations to improve outcomes. Pediatric cancer survivors need access to emerging technologies aimed at reducing or better managing toxicities from therapy. With advances in treatment and survival, pediatric oncology patients continue to need longitudinal, multidisciplinary subspecialty care. Refining the communication between pediatric oncologists, primary pediatricians, survivorship clinics, and adult primary care is key in ensuring the best lifelong care of pediatric cancer survivors. In this State-of-The-Art review, we discuss 5 major domains in pediatric oncology: reducing toxicity, cancer biology, novel therapies, detection and monitoring, and access to care, to highlight recent advances and areas for continued improvement.

In the past 50 years, pediatric cancer outcomes have greatly improved. Some cancers, such as pediatric acute lymphoblastic leukemia, have demonstrated steady improvements in outcome throughout this time, with a 3-year survival in 1975 of ∼59% and a 5-year survival in 2020 of ∼90%.14  Immunotherapy continues to revolutionize the treatment of aggressive pediatric leukemias and lymphomas.5,6  In contrast, after initial improvements in outcome because of the discovery and use of chemotherapeutic agents such as doxorubicin and pediatric cancers such as bone sarcomas, some brain tumors have not achieved meaningful survival improvements with new therapies in decades.7  Broadly speaking, survival differences have inspired 2 major research focuses in pediatric oncology: (1) for patients with excellent (90%+) survival, we now seek ways to reduce or modify therapy to minimize long-term treatment complications, and (2) for patients diagnosed with tumors stuck in a survival “plateau” (continued poor outcomes), we turn to innovations in biology to determine new therapeutic vulnerabilities, enhance disease monitoring, and better understand tumor heterogeneity, metastasis, and treatment resistance.

Primary pediatricians are often the physicians first uncovering the atypical symptoms, abnormal physical exam findings, and concerning laboratory values that ultimately lead to the workup and diagnosis of cancer in children. Suspected malignant solid tumors in children most often require a biopsy to determine a definitive diagnosis. Historically, the majority of these diagnostic biopsies were open biopsies performed by pediatric or orthopedic surgeons. Given innovations in image-guided percutaneous biopsy techniques, diagnostic tumor core biopsies are performed by interventional radiologists and at many institutions are now more common than open biopsies.8  Tumor molecular profiling at the time of diagnosis is also now more prevalent and helps drive personalized medicine efforts in pediatric oncology.9,10 

Although some children with specific subtypes of cancer have benefitted from targeted therapies or immunotherapies, the vast majority of children diagnosed with cancer today are still treated with standard chemotherapy and sometimes surgery and/or radiation therapy. As more children survive pediatric cancer, primary care pediatricians and pediatricians across subspecialties increasingly interact with childhood cancer survivors, children living with cancer, and their families (Fig 1). Pediatric cancer affects almost every facet of child and adolescent health. This State-of-the-Art article aims to provide pediatricians with an update on the state of pediatric cancer care. We highlight the following domains: reducing toxicity, cancer biology, novel therapies, detection and monitoring, and access to care, because these domains both represent areas of need for children with cancer and core topics in general pediatric medicine, such as long-term health, medical research and advancement, and health disparities.

FIGURE 1

Overview of 5 major areas of advancement in pediatric oncology. Figures created with BioRender.com.

FIGURE 1

Overview of 5 major areas of advancement in pediatric oncology. Figures created with BioRender.com.

Close modal

Thanks to cooperative national clinical trial efforts over decades, some pediatric patients diagnosed with cancer, such as newly diagnosed standard risk B-cell lymphoblastic leukemia, now have excellent outcomes (>90% event-free and overall survival).11  Despite these excellent outcomes, current treatment regimens result in significant risk of both short-term (during therapy) and long-term (after therapy completion) side effects, and many pediatric cancer survivors are diagnosed with 1 or more chronic health issues.1214  In the setting of overall excellent outcomes, a logical next-step is to determine how to better mitigate or treat toxicities while maintaining excellent overall survival. Table 1 highlights examples of only some of the toxicities encountered during cancer directed therapy to highlight the range of approaches to reduce or avoid those long-term effects. These approaches include future lifestyle modifications, protective medications, utilizing different chemotherapy regimens, and broadly reducing exposure to agents by optimizing risk stratification (what patients can still have excellent outcomes with less chemotherapy or radiation exposure).

TABLE 1

Examples of Long-term Side Effects From Pediatric Cancer Treatment and Interventions

Cancer TypeExamples of Long-term Side Effects From Therapy63–66Examples of Interventions to Reduce Long-term Side Effects63–66
Hodgkin lymphoma • Gonadal dysfunction: potential for reduced fertility or infertility in both males and females
• Thyroid complication: hypothyroidism; increased risk of thyroid cancer 
• Discussing options for fertility preservation at diagnosis and during long-term follow-up visits
• Thyroid complications: decreased risk by reducing utilization of radiation therapy 
Acute lymphoblastic leukemia • Growth impairment including short stature, precocious puberty, or delayed puberty
• Metabolic syndrome, obesity 
• Growth impairment: improved by the replacement of cranial radiotherapy with intrathecal chemotherapy
• Dietary and exercise interventions 
Neuroblastoma • Hearing loss: related to exposure to platinum compounds
• Cataracts 
• Reduce the duration and intensity of treatment when possible through risk stratification efforts 
Wilms tumor • Cardiac toxicity: related to anthracyclines, radiation involving heart
• Renal dysfunction: glomerular and tubular damage; end-stage renal disease in patients with bilateral Wilms tumor or receiving radiation therapy in unilateral disease 
• Cardiac toxicity: using cardioprotective agents (dexrazoxane), and minimizing chemotherapy and radiation when possible
• Renal dysfunction: nephron-sparing surgery for bilateral disease, avoiding nephrotoxic agents (for example, non-steroidal anti-inflammatories) 
Cancer TypeExamples of Long-term Side Effects From Therapy63–66Examples of Interventions to Reduce Long-term Side Effects63–66
Hodgkin lymphoma • Gonadal dysfunction: potential for reduced fertility or infertility in both males and females
• Thyroid complication: hypothyroidism; increased risk of thyroid cancer 
• Discussing options for fertility preservation at diagnosis and during long-term follow-up visits
• Thyroid complications: decreased risk by reducing utilization of radiation therapy 
Acute lymphoblastic leukemia • Growth impairment including short stature, precocious puberty, or delayed puberty
• Metabolic syndrome, obesity 
• Growth impairment: improved by the replacement of cranial radiotherapy with intrathecal chemotherapy
• Dietary and exercise interventions 
Neuroblastoma • Hearing loss: related to exposure to platinum compounds
• Cataracts 
• Reduce the duration and intensity of treatment when possible through risk stratification efforts 
Wilms tumor • Cardiac toxicity: related to anthracyclines, radiation involving heart
• Renal dysfunction: glomerular and tubular damage; end-stage renal disease in patients with bilateral Wilms tumor or receiving radiation therapy in unilateral disease 
• Cardiac toxicity: using cardioprotective agents (dexrazoxane), and minimizing chemotherapy and radiation when possible
• Renal dysfunction: nephron-sparing surgery for bilateral disease, avoiding nephrotoxic agents (for example, non-steroidal anti-inflammatories) 

One approach to toxicity reduction is decreasing cumulative doses of chemotherapy. Again, using leukemia as an example, chemotherapy is assigned by risk group. Patients with standard risk leukemia are exposed to less cumulative anthracyclines as compared with patients with high-risk leukemia.15,16  Further, historically boys received an additional year of maintenance chemotherapy for the treatment of B-cell lymphoblastic leukemia on some cooperative treatment protocols. Current practice has eliminated this extra year of therapy for boys as with modern upfront intensive treatment regimens, the risks of this practice outweigh the benefits.17  Reduction or elimination of radiation therapy is another example of therapy modifications that can reduce late effects such as secondary malignancies. Recent data have demonstrated that radiation therapy can be eliminated for patients with Hodgkin lymphoma that demonstrate adequate response to chemotherapy.18 

New chemotherapy formulations aimed at maintaining antitumor activity while reducing damage to normal tissue are emerging. Liposomal formulations of chemotherapy, such as doxorubicin, are in various stages of preclinical testing and clinical trials. In addition to less toxic formulations of traditional chemotherapy, immunotherapies have emerged as treatments with reduced—or different—toxicity profiles as compared with DNA damage directed chemotherapies. To date, immunotherapies have been most successful in the treatment of pediatric leukemias and lymphomas, and the long-term toxicity profiles of these treatments continue to be examined.1921 

Large sequencing efforts in pediatric oncology over the past decade have greatly enhanced understanding of germline alterations in children with cancer.2224  This is important not only for cancer predisposition and tumor biology but also for the way in which a child’s genetic make-up may alter how they metabolize drugs. Such pharmacogenomic analyses may reveal the need to dose reduce certain medications or prompt the use of certain drugs in patients demonstrating genetic factors that may make a medicine more efficacious. As ∼20% of children with cancer will be admitted to the hospital for the treatment of an adverse drug reaction,25  this is an emerging area of importance given the steady rise in patient access to tumors and germline sequencing. Further, early prediction of patients whose tumors may not respond to a drug could help physicians consider alternatives earlier. One example of this is response to platinum agents being reduced by mutations in genes of the ERCC family of DNA excision repair proteins.25 

Until new treatment options emerge, some side effects are currently unavoidable, such as the likelihood of some treatments affecting fertility. Patients experiencing life-threatening oncologic medical emergencies often do not have time to undergo fertility preservation before initiation of gonadotoxic treatments. Cumulative exposure to cyclophosphamide and equivalent drugs greatly reduces a child’s future ability to have children.26,27  Postpubertal females and males have the option for pretreatment oocyte or sperm cryopreservation.28  Recent advances in fertility preservation have emerged and provide a way to enhance chances of future fertility while still successfully treating a child’s cancer. For example, prepubertal females at some specialized centers now have access to research protocols attempting ovarian cryopreservation for fertility preservation.29,30  Infertility can have a major impact on an adult’s quality of life. Continued advances in fertility preservation—and affordable, reliable access to fertility counseling and preservation services—have the potential to significantly improve the long-term well-being of children and adolescents with cancer.31 

Pediatric oncology patients routinely follow-up in survivorship clinics annually after completion of initial therapy and undergo active disease surveillance for a number of years. Predicted side effects from therapy are monitored over time. Efforts to optimize communication with primary care pediatricians and include pediatricians as an active member of the postcancer care team is a priority. Patients and families often move or re-establish care and, therefore, efforts have focused on establishing survivorship portals that can “move” with the patient.32  A wealth of educational information and care guidelines are also provided, www.survivorshipguidelines.org.33  Accurate and up to date communication regarding the overall health monitoring of survivors is also needed to ensure the successful transition to adult medicine care. Awareness and monitoring are key components to successful management of long-term toxicities for childhood cancer survivors (Fig 2).

FIGURE 2

Overview of care transitions and collaborations for children with cancer. A child and their siblings (if applicable) receive primary pediatric care. When a primary pediatrician notes abnormal symptoms, etc and a child is diagnosed with cancer, care shifts to the pediatric oncology team. After therapy, the child with cancer transitions back to their primary pediatrician and both short- and long-term survivorship care plans are developed.

FIGURE 2

Overview of care transitions and collaborations for children with cancer. A child and their siblings (if applicable) receive primary pediatric care. When a primary pediatrician notes abnormal symptoms, etc and a child is diagnosed with cancer, care shifts to the pediatric oncology team. After therapy, the child with cancer transitions back to their primary pediatrician and both short- and long-term survivorship care plans are developed.

Close modal

An unfortunate reality of pediatric cancer is that some children still do not survive. Children diagnosed with brain tumors such as diffuse intrinsic pontine glioma (DIPG) and bone tumors such as osteosarcoma and metastatic Ewing sarcoma have not seen meaningful improvements in outcomes in decades. For these cancers, researchers strive to continue to better understand the biology of these tumors (heterogeneity, metastatic potential, immune evasion, treatment resistance, and stemness—a cancer cell’s ability to undergo “self-renewal and differentiation”34 ) and corresponding interventions that may “move the needle” toward improving survival. As these cancer types are less common than pediatric leukemias, some of the challenges in understanding these tumors derives from the fact that they are relatively rare, thereby making tumor subpopulations more challenging to study.

For cancers with >50% survival at diagnosis such as bone sarcomas, a logical question to ask is: what biologic features make some primary tumors more aggressive than others? Pediatric sequencing efforts have clearly demonstrated the genetic profiles across the spectrum of pediatric cancers. Tumor subsets exist and there is a clear need to understand which tumor subsets are associated with inferior clinical outcomes. Aggressive tumor subtypes can then be studied to determine specific vulnerabilities, and ultimately provide clues to new treatments to test in patients with aggressive tumor subtypes. Neuroblastoma is an example of a solid tumor where ALK mutations and N-Myc amplification have been found to drive cancer progression and be associated with worse outcomes.35,36  ALK inhibitors (eg, crizotinib, lorlatinib, etc) are being studied in the treatment of ALK mutated high risk neuroblastoma, an example of biologic understanding driving treatment change.37  Dedicated efforts for defining risk groups for bone sarcomas are underway.

Before any compound being tested in clinical trials, preclinical testing is needed. Although cell lines are often useful for dissecting the effects of a compound on signaling pathways, cell lines often poorly mimic the complexity of the in vivo tumor microenvironment. With current preclinical models of many pediatric cancers, it is still challenging to predict clinical effectiveness of an agent.38  We strive to use patient derived xenografts and immunocompetent models of cancer and to represent tumor subsets in preclinical studies. Advances in the generation of humanized mice (mice transplanted with human immune cells) allows investigators to now better understand the complex relationships between the tumor and the body’s immune system. Given the rise in immunotherapeutic approaches to cancer treatment in the past decade, immunocompetent modeling is a key emerging component in studying the efficacy of specific immunotherapies.39 

For rare tumors, every sample is valuable. National and international efforts to enhance biospecimen banking of difficult to treat cancers is a priority. The field continues to rely on the generosity and willingness of pediatric patients and their families to participate in research studies to advance our understanding of these cancers.40  Optimizing sample storage conditions and annotation through data harmonization is an ongoing priority. For cancers such as DIPG, biopsy samples were historically not obtained and diagnosis was made based on clinical or MRI findings alone. Recent advances in neurosurgical stereotactic biopsy approaches now provide an opportunity for biopsy material from suspected DIPGs to be safely obtained.41  Such diagnostic biopsy material allows for a richer understanding of DIPG biology and researchers hope such samples will start to provide clues for new therapeutic approaches.

In addition to obtaining material at the time of original diagnostic biopsies, biopsy material at the time of relapse is also invaluable. Tumors change over time and after exposure to cytotoxic chemotherapy. Individual tumor subpopulations can develop resistance, ultimately resulting in disease relapse.42,43  Historically, pediatric cancers were not always biopsied at the time of relapse to spare children from additional procedures. Given the need to better understand relapsed disease to improve outcomes and the ability to participate in clinical trials, for example, the National Cancer Institute-Children’s Oncology Group Pediatric MATCH trial (“matching” tumor alterations to possible targeted therapy), there has been a shift in practice and today, pediatric patients are more likely to be offered rebiopsy at the time of suspected disease progression.44  The MATCH trial has demonstrated the feasibility of identifying personalized therapeutic options for pediatric patients with difficult to treat cancer.45 

New treatment approaches are needed to continue to improve upon long-term morbidity and mortality for pediatric patients with cancer. Examples of targeted agents (antibodies, cellular therapies, kinase inhibitors, antibody-drug conjugates, etc) used in the treatment of pediatric cancer are listed in Table 2.

TABLE 2

Examples of Targeted Agents for the Treatment of Pediatric, Adolescent, and Young Adult Cancers

Cancer TypeExample of Actionable Target35,46,47,6773 Example Targeted Agents35,46,47,6773 
Acute lymphoblastic leukemia • Expression of CD19 • Blinatumomab (CD19-CD3 bispecific T-cell engager, BiTE)
• CD19 chimeric antigen receptor (CAR) T cells 
Infantile fibrosarcoma • NTRK fusion • NTRK inhibitors (larotrectinib, etc) 
Neuroblastoma • High expression of GD2
• ALK mutation 
• Dinutuximab (monoclonal antibody against GD2)
• ALK inhibitors (lorlatinib, etc) 
Low grade gliomas • BRAF mutations • RAF inhibitors, pan-RAF inhibitors (tovorafenib), and MEK inhibitors 
Hodgkin lymphoma • CD30 • Brentuximab vedotin (CD30 targeting antibody drug conjugate. Chemotherapy agent linked to CD30 is monomethyl auristatin E [MMAE]) 
Mature B cell lymphomas • CD20 • Rituximab (CD20 chimeric antibody) 
Epithelioid sarcoma • SMARCB1/INI1 mutations • Tazemetostat (EZH2 inhibitor) 
Cancer TypeExample of Actionable Target35,46,47,6773 Example Targeted Agents35,46,47,6773 
Acute lymphoblastic leukemia • Expression of CD19 • Blinatumomab (CD19-CD3 bispecific T-cell engager, BiTE)
• CD19 chimeric antigen receptor (CAR) T cells 
Infantile fibrosarcoma • NTRK fusion • NTRK inhibitors (larotrectinib, etc) 
Neuroblastoma • High expression of GD2
• ALK mutation 
• Dinutuximab (monoclonal antibody against GD2)
• ALK inhibitors (lorlatinib, etc) 
Low grade gliomas • BRAF mutations • RAF inhibitors, pan-RAF inhibitors (tovorafenib), and MEK inhibitors 
Hodgkin lymphoma • CD30 • Brentuximab vedotin (CD30 targeting antibody drug conjugate. Chemotherapy agent linked to CD30 is monomethyl auristatin E [MMAE]) 
Mature B cell lymphomas • CD20 • Rituximab (CD20 chimeric antibody) 
Epithelioid sarcoma • SMARCB1/INI1 mutations • Tazemetostat (EZH2 inhibitor) 

The goal of precision oncology is to identify drugs predicted to work against tumors with specific mutations, etc. As noted above, The MATCH trial has demonstrated the feasibility of identifying personalized therapeutic options for pediatric patients with difficult to treat cancer.45  Examples of recent precision oncology success stories in pediatric oncology include the utilization of NTRK inhibitors for rare NTRK pediatric solid tumors, such as infantile fibrosarcoma and RAF/MEK inhibitors for the treatment of low-grade gliomas in children.4648 

Hundreds of immunotherapies for the treatment of cancer now exist. Broadly, these therapies include engineered cells, such as CD19 CAR-T cells for the treatment of pediatric B-cell lymphoblastic leukemia, checkpoint inhibitors aimed at “reawakening” the body’s own immune system, cytokine manipulation, and agents aimed at remodeling the tumor microenvironment. Although challenges exist in the utilization of CAR-T cells for the treatment of solid tumors, exciting progress has been made in demonstrating the feasibility of CAR-T cell utilization for the treatment of currently incurable pediatric cancers such as DIPG (diffuse intrinsic pontine glioma).49  Given the rapid rise in immunotherapies being developed in adults, there is significant future potential in immunomanipulation for the treatment of pediatric cancers. Currently, few children with cancer are treated with immunotherapies. In addition, we are only beginning to understand the long-term side effects of immunomanipulation on the health of children and adolescents. As these data emerge, continued communication with general pediatricians is essential to ensure that these effects are recognized and effectively addressed.

Tumors are known to be heterogeneous and develop treatment resistance, including resistance to targeted agents. Tumors rarely demonstrate long-term responses to single agents. Combination approaches to “attack” tumors using different methods is one way to circumvent the resistance. Combining DNA damaging agents with immunotherapies and utilizing antibody-drug conjugates are 2 examples of emerging multimodality therapies that have the potential to continue to change the landscape of pediatric cancer treatment. Immunotherapy timing (the delivery order of agents and/or severity of immunosuppression when agents are given) is especially critical when adding immunotherapies to existing treatment regimens. Preclinical data specifically addressing immunotherapy timing is an emerging topic of great interest.50,51 

Disease monitoring is conducted both on-therapy and after completion of initial therapy for the treatment of cancer in children. Child exposure to radiation, such as that delivered by chest radiographs and CT scans, should always be avoided or minimized when possible. New technologies are emerging that likely will revolutionize the way tumor responses and relapses are monitored.

When tumor cells die, fragments of DNA from the tumor cells are released into the bloodstream of the patient. This circulating tumor DNA (ctDNA) can be detected in blood from patients with cancer52  and provides many future opportunities: (1) more frequent monitoring of disease relapse at a level that a scan may not be able to detect, (2) reduce exposure to radiation, and (3) detect emerging resistant subpopulations of tumor cells while on therapy. One large question that ctDNA may be able to help address is whether earlier detection and treatment of relapses will improve outcomes. As ctDNA monitoring (“liquid biopsies”) is increasingly included on prospective clinical trials, we will come to more fully understand the potential of this technology. A recent example of the power of ctDNA was demonstrated in intermediate risk, fusion negative rhabdomyosarcoma, where investigators demonstrate that the presence of ctDNA in a patient’s serum at diagnosis is associated with a worse outcome (statistically significant inferior event-free and overall survival).53 

In addition to tumor sequencing, germline sequencing of pediatric patients with cancer has also increased and can complement and enhance knowledge derived from tumor-only sequencing.54,55  As discussed above, there may be potential pharmacogenomic benefits of this information. Additionally, should a patient have a germline pathogenic variant in a cancer predisposition gene discovered, cancer screening recommendations may be suggested for the patient.56,57  We know that some children develop secondary malignancies after their primary cancer therapy. We still do not fully understand which pathogenic germline findings may increase the likelihood of children developing a treatment-related secondary malignancy. Family members of children with cancer may be referred for genetic counseling and testing depending on the germline result found in the child. Many variants of unknown significance are detected during germline testing, and as more data emerge, screening and risk recommendation are updated over time. It is especially important to remember this for survivors who may not have had access to germline testing when initially diagnosed or treated, as they may desire testing. If testing did occur, a re-examination of the interpretation of the findings may be warranted given variant reclassifications over time. Many children’s hospitals nationally now have cancer predisposition programs where referrals for genetic counseling and testing are accepted both for patients with cancer and their family members.58 

Pediatricians are well aware that inequities in access to care exist. Pediatric oncology is not spared from this reality.

In the United States, Black children diagnosed with cancer continue to have inferior overall survival compared with white children. One example of many is that lower survival rates are observed among Black children with acute myeloid leukemia when compared with non-Hispanic white children.59  The reasons for this are understudied and multifactorial, though minority populations remain underrepresented in cancer research.59  Rates of relapse among minority children and those living in poverty also remain higher across disease groups,59  highlighting the need for social determinants of health to be incorporated into treatment plans. Globally, overall survival for children with cancer varies based on the income status of the country in which they reside. Issues with access to cancer drugs, clinical trials, expert opinions for rare tumors, supportive care measures, and advanced testing such as tumor sequencing all contribute to the noted disparities.59,60 

When considering clinical trials, an emerging approach in oncology is the integrated use of real-world data (RWD) or real world evidence. Although randomized control trials are the gold standard in pediatric oncology, for rare tumors and understanding subpopulations of patients, this is not always feasible. Examples of RWD include electronic medical record data, billing data, social media platforms, patient or family surveys, and information from tumor registries. For example, use of real world evidence helped lay the groundwork for the accelerated US Food and Drug Administration approval of blinatumomab for the treatment of relapsed or refractory B cell acute lymphoblastic leukemia in adults.61  Incorporation of RWD may help bridge some gaps in disparities in clinical trial access and more timely access to emerging drugs in pediatric oncology.

For survivors, access to specialty services, such as physical and occupational therapy, ongoing oncofertility and reproductive health resources, neuropsychologic testing, and behavioral health services are inconsistent. For example, survivors and their parents often experience post-traumatic stress disorder and anxiety, and lack of access to behavioral health resources can have a significant, lifelong negative impact on long-term health for cancer survivors.62  Partnerships between pediatric oncologists and general pediatricians are key in advocating for both the needs of individual patients and for the health of children more broadly. A dedicated effort must be made to improve survival, mitigate toxicities, and promote lifelong health in all children with cancer.

A simplified statement of goals in pediatric oncology is to finally “break” the survival plateau for some solid tumors and to minimize toxicity for all children and adolescents diagnosed with cancer. Here, we highlighted several ways in which innovations in drug delivery, monitoring, novel therapeutics, and access to care continue to help move the field of pediatric oncology forward. Children with cancer need excellent longitudinal care both during and after completion of cancer directed therapy. Continuing to strengthen the partnership and communication between the patient’s pediatric oncology team and primary care pediatrician is essential in this mission.

Drs Helms and Guimera contributed to the manuscript draft and figure editing; Drs Janeway and Bailey conceptualized the article and contributed to the manuscript draft; and all authors critically reviewed and revised the manuscript, approved the final manuscript as submitted, and agree to be accountable for all aspects of the work.

FUNDING: Dr Bailey is currently supported by the National Institutes of Health (NCI K08CA252178) and would like to thank the UPMC Children’s Hospital Foundation for their support. The other authors received no additional funding.

CONFLICT OF INTEREST DISCLOSURES: Dr Janeway has served as a consultant for Bayer, Ipsen, and Illumina; the other authors have indicated they have no potential conflicts of interest to disclose.

ctDNA

circulating tumor DNA

DIPG

diffuse intrinsic pontine glioma

RWD

real world data

1
Islami
F
,
Ward
EM
,
Sung
H
, et al
.
Annual report to the nation on the status of cancer, part 1: national cancer statistics
.
J Natl Cancer Inst
.
2021
;
113
(
12
):
1648
1669
2
O’Leary
M
,
Krailo
M
,
Anderson
JR
,
Reaman
GH
; Children’s Oncology Group.
Progress in childhood cancer: 50 years of research collaboration, a report from the Children’s Oncology Group
.
Semin Oncol
.
2008
;
35
(
5
):
484
493
3
Siegel
DA
,
Richardson
LC
,
Henley
SJ
, et al
.
Pediatric cancer mortality and survival in the United States, 2001–2016
.
Cancer
.
2020
;
126
(
19
):
4379
4389
4
Smith
MA
,
Seibel
NL
,
Altekruse
SF
, et al
.
Outcomes for children and adolescents with cancer: challenges for the twenty-first century
.
J Clin Oncol
.
2010
;
28
(
15
):
2625
2634
5
Foster
JB
,
Maude
SL
.
New developments in immunotherapy for pediatric leukemia
.
Curr Opin Pediatr
.
2018
;
30
(
1
):
25
29
6
Salzer
E
,
Attarbaschi
A
.
The value of immunotherapy in pediatric leukemia and lymphoma
.
Magazine of European Medical Oncology
.
2021
;
14
:
397
401
7
Cores
EP
,
Holland
JF
,
Wang
JJ
,
Sinks
LF
.
Doxorubicin in disseminated osteosarcoma
.
JAMA
.
1972
;
221
(
10
):
1132
1138
8
Bruinsma
RS
,
Nievelstein
RAJ
,
Littooij
AS
, et al
.
Diagnostic accuracy of image-guided core needle biopsy of non-central nervous system tumors in children
.
Pediatr Blood Cancer
.
2021
;
68
(
10
):
e29179
9
Harris
MH
,
DuBois
SG
,
Glade Bender
JL
, et al
.
Multicenter feasibility study of tumor molecular profiling to inform therapeutic decisions in advanced pediatric solid tumors: The Individualized Cancer Therapy (iCat) Study
.
JAMA Oncol
.
2016
;
2
(
5
):
608
615
10
Marron
JM
,
DuBois
SG
,
Glade Bender
J
, et al
.
Patient/parent perspectives on genomic tumor profiling of pediatric solid tumors: The Individualized Cancer Therapy (iCat) experience
.
Pediatr Blood Cancer
.
2016
;
63
(
11
):
1974
1982
11
Angiolillo
AL
,
Schore
RJ
,
Kairalla
JA
, et al
.
Excellent outcomes with reduced frequency of vincristine and dexamethasone pulses in standard-risk B-lymphoblastic leukemia: results from Children’s Oncology Group AALL0932
.
J Clin Oncol
.
2021
;
39
(
13
):
1437
1447
12
Chehab
L
,
Doody
DR
,
Esbenshade
AJ
, et al
.
A population-based study of the long-term risk of infections associated with hospitalization in childhood cancer survivors
.
J Clin Oncol
.
2023
;
41
(
2
):
364
372
13
Prasad
M
,
Goswami
S
,
Chinnaswamy
G
,
Banavali
SD
,
Kurkure
PA
.
Long-term outcomes in survivors of childhood cancer: a 30-year experience from India
.
JCO Glob Oncol
.
2022
;
8
:
e2200044
14
Williams
AM
,
Liu
Q
,
Bhakta
N
, et al
.
Rethinking success in pediatric oncology: beyond 5-year survival
.
J Clin Oncol
.
2021
;
39
(
20
):
2227
2231
15
Möricke
A
,
Reiter
A
,
Zimmermann
M
, et al
;
German-Austrian-Swiss ALL-BFM Study Group
.
Risk-adjusted therapy of acute lymphoblastic leukemia can decrease treatment burden and improve survival: treatment results of 2169 unselected pediatric and adolescent patients enrolled in the trial ALL-BFM 95
.
Blood
.
2008
;
111
(
9
):
4477
4489
16
Schore
RJ
,
Angiolillo
AJ
,
Kairalla
JA
, et al
.
Outcomes with reduced intensity therapy in a low-risk subset of children with National Cancer Institute (NCI) standard-risk (SR) B-lymphoblastic leukemia (B-ALL): a report from Children’s Oncology Group (COG) AALL0932
.
J Clin Oncol
. Meeting Abstract,
2020
;
38
(
15
)
17
Teachey
DT
,
Hunger
SP
,
Loh
ML
.
Optimizing therapy in the modern age: differences in length of maintenance therapy in acute lymphoblastic leukemia
.
Blood
.
2021
;
137
(
2
):
168
177
18
Mauz-Körholz
C
,
Landman-Parker
J
,
Balwierz
W
, et al
.
Response-adapted omission of radiotherapy and comparison of consolidation chemotherapy in children and adolescents with intermediate-stage and advanced-stage classical Hodgkin lymphoma (EuroNet-PHL-C1): a titration study with an open-label, embedded, multinational, non-inferiority, randomised controlled trial
.
Lancet Oncol
.
2022
;
23
(
1
):
125
137
19
Conroy
M
,
Naidoo
J
.
Immune-related adverse events and the balancing act of immunotherapy
.
Nat Commun
.
2022
;
13
(
1
):
392
20
Jain
T
,
Litzow
MR
.
No free rides: management of toxicities of novel immunotherapies in ALL, including financial
.
Hematology (Am Soc Hematol Educ Program)
.
2018
;
2018
(
1
):
25
34
21
Schubert
ML
,
Schmitt
M
,
Wang
L
, et al
.
Side-effect management of chimeric antigen receptor (CAR) T-cell therapy
.
Ann Oncol
.
2021
;
32
(
1
):
34
48
22
Gillani
R
,
Camp
SY
,
Han
S
, et al
.
Germline predisposition to pediatric Ewing sarcoma is characterized by inherited pathogenic variants in DNA damage repair genes
.
Am J Hum Genet
.
2022
;
109
(
6
):
1026
1037
23
Mody
RJ
,
Prensner
JR
,
Everett
J
,
Parsons
DW
,
Chinnaiyan
AM
.
Precision medicine in pediatric oncology: lessons learned and next steps
.
Pediatr Blood Cancer
.
2017
;
64
(
3
):
10.1002/pbc.26288
24
Parsons
DW
,
Roy
A
,
Yang
Y
, et al
.
Diagnostic yield of clinical tumor and germline whole-exome sequencing for children with solid tumors
.
JAMA Oncol
.
2016
;
2
(
5
):
616
624
25
Elzagallaai
AA
,
Carleton
BC
,
Rieder
MJ
.
Pharmacogenomics in pediatric oncology: mitigating adverse drug reactions while preserving efficacy
.
Annu Rev Pharmacol Toxicol
.
2021
;
61
:
679
699
26
Green
DM
,
Liu
W
,
Kutteh
WH
, et al
.
Cumulative alkylating agent exposure and semen parameters in adult survivors of childhood cancer: a report from the St Jude Lifetime Cohort Study
.
Lancet Oncol
.
2014
;
15
(
11
):
1215
1223
27
Green
DM
,
Nolan
VG
,
Goodman
PJ
, et al
.
The cyclophosphamide equivalent dose as an approach for quantifying alkylating agent exposure: a report from the Childhood Cancer Survivor Study
.
Pediatr Blood Cancer
.
2014
;
61
(
1
):
53
67
28
Lambertini
M
,
Peccatori
FA
,
Demeestere
I
, et al
;
ESMO Guidelines Committee.
Fertility preservation and post-treatment pregnancies in post-pubertal cancer patients: ESMO clinical practice guidelines
.
Ann Oncol
.
2020
;
31
(
12
):
1664
1678
29
Hinkle
K
,
Orwig
KE
,
Valli-Pulaski
H
, et al
.
Cryopreservation of ovarian tissue for pediatric fertility
.
Biopreserv Biobank
.
2021
;
19
(
2
):
130
135
30
Oktay
K
,
Harvey
BE
,
Partridge
AH
, et al
.
Fertility preservation in patients with cancer: ASCO clinical practice guideline update
.
J Clin Oncol
.
2018
;
36
(
19
):
1994
2001
31
Poorvu
PD
,
Frazier
AL
,
Feraco
AM
, et al
.
Cancer treatment-related infertility: a critical review of the evidence
.
JNCI Cancer Spectr
.
2019
;
3
(
1
):
pkz008
32
Poplack
DG
,
Fordis
M
,
Landier
W
,
Bhatia
S
,
Hudson
MM
,
Horowitz
ME
.
Childhood cancer survivor care: development of the Passport for Care
.
Nat Rev Clin Oncol
.
2014
;
11
(
12
):
740
750
33
Hudson
MM
,
Bhatia
S
,
Casillas
J
,
Landier
W
;
Section on Hematology and Oncology
,
Children’s Oncology Group
,
American Society of Pediatric Hematology and Oncology
.
Long-term follow-up care for childhood, adolescent, and young adult cancer survivors
.
Pediatrics
.
2021
;
148
(
3
):
e2021053127
34
Clever
H
.
Cancer therapy: defining stemness
.
Nature
.
2016
;
534
:
176
177
35
Mossé
YP
,
Laudenslager
M
,
Longo
L
, et al
.
Identification of ALK as a major familial neuroblastoma predisposition gene
.
Nature
.
2008
;
455
(
7215
):
930
935
36
Wang
LL
,
Teshiba
R
,
Ikegaki
N
, et al
.
Augmented expression of MYC and/or MYCN protein defines highly aggressive MYC-driven neuroblastoma: a Children’s Oncology Group study
.
Br J Cancer
.
2015
;
113
(
1
):
57
63
37
Foster
JH
,
Voss
SD
,
Hall
DC
, et al
.
Activity of crizotinib in patients with ALK-aberrant relapsed/refractory neuroblastoma: A Children’s Oncology Group Study (ADVL0912)
.
Clin Cancer Res
.
2021
;
27
(
13
):
3543
3548
38
Gengenbacher
N
,
Singhal
M
,
Augustin
HG
.
Preclinical mouse solid tumour models: status quo, challenges and perspectives
.
Nat Rev Cancer
.
2017
;
17
(
12
):
751
765
39
Nguyen
R
,
Patel
AG
,
Griffiths
LM
, et al
.
Next-generation humanized patient-derived xenograft mouse model for pre-clinical antibody studies in neuroblastoma
.
Cancer Immunol Immunother
.
2021
;
70
(
3
):
721
732
40
Venkatramani
R
,
Piao
J
,
Krailo
MD
, et al
.
Carcinomas in the children and young adults: A report form Children’s Oncology Group APEC14B1 study
.
Journal of Clinical Oncology
.
2019
;
37
(
15
):
e21505-e
41
Hersh
DS
,
Kumar
R
,
Moore
KA
, et al
.
Safety and efficacy of brainstem biopsy in children and young adults
.
J Neurosurg Pediatr
.
2020
;
26
(
5
):
552
562
42
Byron
SA
,
Hendricks
WPD
,
Nagulapally
AB
, et al
.
Genomic and transcriptomic analysis of relapsed and refractory childhood solid tumors reveals a diverse molecular landscape and mechanisms of immune evasion
.
Cancer Res
.
2021
;
81
(
23
):
5818
5832
43
Waanders
E
,
Gu
Z
,
Dobson
SM
, et al
.
Mutational landscape and patterns of clonal evolution in relapsed pediatric acute lymphoblastic leukemia
.
Blood Cancer Discov
.
2020
;
1
(
1
):
96
111
44
Cohen
B
,
Roth
M
,
Marron
JM
, et al
.
Pediatric oncology provider views on performing a biopsy of solid tumors in children with relapsed or refractory disease for the purpose of genomic profiling
.
Ann Surg Oncol
.
2016
;
23
(
Suppl 5
):
990
997
45
Parsons
DW
,
Janeway
KA
,
Patton
DR
, et al
;
NCI-COG Pediatric MATCH Team
.
Actionable tumor alterations and treatment protocol enrollment of pediatric and young adult patients with refractory cancers in the National Cancer Institute-Children’s Oncology Group Pediatric MATCH Trial
.
J Clin Oncol
.
2022
;
40
(
20
):
2224
2234
46
Blauel
ER
,
Laetsch
TW
.
The promise of TRK inhibitors in pediatric cancers with NTRK fusions
.
Cancer Genet
.
2022
;
262-263
:
71
79
47
Schreck
KC
,
Grossman
SA
,
Pratilas
CA
.
BRAF mutations and the utility of RAF and MEK inhibitors in primary brain tumors
.
Cancers (Basel)
.
2019
;
11
(
9
):
1262
48
Shulman
DS
,
DuBois
SG
.
The evolving diagnostic and treatment landscape of NTRK-fusion-driven pediatric cancers
.
Paediatr Drugs
.
2020
;
22
(
2
):
189
197
49
Majzner
RG
,
Ramakrishna
S
,
Yeom
KW
, et al
.
GD2-CAR T cell therapy for H3K27M-mutated diffuse midline gliomas
.
Nature
.
2022
;
603
(
7903
):
934
941
50
Qian
DC
,
Kleber
T
,
Brammer
B
, et al
.
Effect of immunotherapy time-of-day infusion on overall survival among patients with advanced melanoma in the USA (MEMOIR): a propensity score-matched analysis of a single-centre, longitudinal study
.
Lancet Oncol
.
2021
;
22
(
12
):
1777
1786
51
Williamson
CW
,
Sherer
MV
,
Zamarin
D
, et al
.
Immunotherapy and radiation therapy sequencing: State of the data on timing, efficacy, and safety
.
Cancer
.
2021
;
127
(
10
):
1553
1567
52
Keller
L
,
Belloum
Y
,
Wikman
H
,
Pantel
K
.
Clinical relevance of blood-based ctDNA analysis: mutation detection and beyond
.
Br J Cancer
.
2021
;
124
(
2
):
345
358
53
Abbou
S
,
Klega
K
,
Tsuji
J
, et al
.
Circulating tumor DNA is prognostic in intermediate-risk rhabdomyosarcoma: a report from the Children’s Oncology Group
.
J Clin Oncol
.
2023
;
41
(
13
):
2382
2393
54
Schienda
J
,
Church
AJ
,
Corson
LB
, et al
.
Germline sequencing improves tumor-only sequencing interpretation in a precision genomic study of patients with pediatric solid tumor
.
JCO Precis Oncol
.
2021
;
5
:
PO.21.00281
55
Villani
A
,
Davidson
S
,
Kanwar
N
, et al
.
The clinical utility of integrative genomics in childhood cancer extends beyond targetable mutations
.
Nat Cancer
.
2022
56
Brodeur
GM
,
Nichols
KE
,
Plon
SE
,
Schiffman
JD
,
Malkin
D
.
Pediatric cancer predisposition and surveillance: an overview, and a tribute to Alfred G. Knudson Jr
.
Clin Cancer Res
.
2017
;
23
(
11
):
e1
e5
57
Druker
H
,
Zelley
K
,
McGee
RB
, et al
.
Genetic counselor recommendations for cancer predisposition evaluation and surveillance in the pediatric oncology patient
.
Clin Cancer Res
.
2017
;
23
(
13
):
e91
e97
58
Venier
RE
,
Grubs
RE
,
Kessler
E
,
Cooper
KL
,
Bailey
KM
,
Meade
J
.
Evaluation of barriers to referral for cancer predisposition syndromes in pediatric oncology patients in the United States
.
J Genet Couns
.
2022
;
31
(
4
):
901
911
59
Aristizabal
P
,
Winestone
LE
,
Umaretiya
P
,
Bona
K
.
Disparities in pediatric oncology: the 21st century opportunity to improve outcomes for children and adolescents with cancer
.
Am Soc Clin Oncol Educ Book
.
2021
;
41
:
e315
e326
60
Gupta
S
,
Dai
Y
,
Chen
Z
, et al
.
Racial and ethnic disparities in childhood and young adult acute lymphocytic leukaemia: secondary analyses of eight Children’s Oncology Group cohort trials
.
Lancet Haematol
.
2023
;
10
(
2
):
e129
e141
61
Raphael
MJ
,
Gyawali
B
,
Booth
CM
.
Real-world evidence and regulatory drug approval
.
Nat Rev Clin Oncol
.
2020
;
17
(
5
):
271
272
62
Zeltzer
LK
,
Recklitis
C
,
Buchbinder
D
, et al
.
Psychological status in childhood cancer survivors: a report from the Childhood Cancer Survivor Study
.
J Clin Oncol
.
2009
;
27
(
14
):
2396
2404
63
Bruzzi
P
,
Bigi
E
,
Predieri
B
, et al
.
Long-term effects on growth, development, and metabolism of ALL treatment in childhood
.
Expert Rev Endocrinol Metab
.
2019
;
14
(
1
):
49
61
64
van Dorp
W
,
van Beek
RD
,
Laven
JS
,
Pieters
R
,
de Muinck Keizer-Schrama
SM
,
van den Heuvel-Eibrink
MM
.
Long-term endocrine side effects of childhood Hodgkin’s lymphoma treatment: a review
.
Hum Reprod Update
.
2012
;
18
(
1
):
12
28
65
Whittle
SB
,
Smith
V
,
Doherty
E
,
Zhao
S
,
McCarty
S
,
Zage
PE
.
Overview and recent advances in the treatment of neuroblastoma
.
Expert Rev Anticancer Ther
.
2017
;
17
(
4
):
369
386
66
Wright
KD
,
Green
DM
,
Daw
NC
.
Late effects of treatment for wilms tumor
.
Pediatr Hematol Oncol
.
2009
;
26
(
6
):
407
413
67
First EZH2 Inhibitor Approved-for Rare Sarcoma
.
First EZH2 inhibitor approved-for rare sarcoma
.
Cancer Discov
.
2020
;
10
(
3
):
333
334
68
Czarnecka
AM
,
Sobczuk
P
,
Kostrzanowski
M
, et al
.
Epithelioid sarcoma-from genetics to clinical practice
.
Cancers (Basel)
.
2020
;
12
(
8
):
2112
69
Deng
C
,
Pan
B
,
O’Connor
OA
.
Brentuximab vedotin
.
Clin Cancer Res
.
2013
;
19
(
1
):
22
27
70
Elsallab
M
,
Ellithi
M
,
Hempel
S
,
Abdel-Azim
H
,
Abou-El-Enein
M
.
Long-term response to autologous anti-CD19 chimeric antigen receptor T cells in relapsed or refractory B cell acute lymphoblastic leukemia: a systematic review and meta-analysis
.
Cancer Gene Ther
.
2023
;
30
(
6
):
845
854
71
Handgretinger
R
,
Baader
P
,
Dopfer
R
, et al
.
A phase I study of neuroblastoma with the anti-ganglioside GD2 antibody 14.G2a
.
Cancer Immunol Immunother
.
1992
;
35
(
3
):
199
204
72
Meinhardt
A
,
Burkhardt
B
,
Zimmermann
M
, et al
;
Berlin-Frankfurt-Münster group
.
Phase II window study on rituximab in newly diagnosed pediatric mature B-cell non-Hodgkin’s lymphoma and Burkitt leukemia
.
J Clin Oncol
.
2010
;
28
(
19
):
3115
3121
73
van der Sluis
IM
,
de Lorenzo
P
,
Kotecha
RS
, et al
.
Blinatumomab added to chemotherapy in infant lymphoblastic leukemia
.
N Engl J Med
.
2023
;
388
(
17
):
1572
1581
This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License (https://creativecommons.org/licenses/by-nc-nd/4.0/), which permits noncommercial, distribution, and reproduction in any medium, provided the original author and source are credited.