Over the past decade, there has been exponential growth in the number of genome sequencing studies performed across a spectrum of human diseases as sequencing technologies and analytic pipelines improve and costs decline. Pediatric hematologic malignancies have been no exception, with a multitude of next generation sequencing studies conducted on large cohorts of patients in recent years. These efforts have defined the mutational landscape of a number of leukemia subtypes and also identified germ-line genetic variants biologically and clinically relevant to pediatric leukemias. The findings have deepened our understanding of the biology of many childhood leukemias. Additionally, a number of recent discoveries may positively impact the care of pediatric leukemia patients through refinement of risk stratification, identification of targetable genetic lesions, and determination of risk for therapy-related toxicity. Although incredibly promising, many questions remain, including the biologic significance of identified genetic lesions and their clinical implications in the context of contemporary therapy. Importantly, the identification of germ-line mutations and variants with possible implications for members of the patient’s family raises challenging ethical questions. Here, we review emerging genomic data germane to pediatric hematologic malignancies.

Learning Objectives
  • Understand the genomic lesions currently used for risk stratification, targeted therapies, and individualization of chemotherapy dosing for pediatric patients with hematologic malignancies

  • Highlight several newly identified somatic and germ-line genetic lesions and variants with potential implications for prognostication, targeted therapeutic intervention, and determination of risk of pediatric hematologic malignancy development

The outcomes of children with most hematologic malignancies have steadily improved over recent decades. However, certain diseases and specific subsets of patients still have suboptimal outcomes with current standard of care treatment. Additionally, standard chemotherapy can be associated with a high burden of toxicity, both immediately and lifelong, for childhood cancer survivors. These challenges have fueled the pursuit of “precision medicine” for the care of children with hematologic malignancies. As broadly defined, precision medicine includes precise assignment of patients to risk-based therapy, identification of targetable genetic lesions, and individualization of chemotherapy dosing. Recent advances have facilitated routine performance of next generation sequencing assays in clinical environments. This has facilitated the translation of genomic profiling studies of large, well-annotated cohorts of pediatric patients with hematologic malignancies being uniformly treated on clinical trials.

Here, we will review well-established and newly identified genetic lesions in pediatric hematologic malignancies. We will discuss the potential prognostic and therapeutic implications of the described somatic genetic lesions. We will also discuss germ-line genetic mutations and polymorphisms associated with childhood leukemia risk and chemotherapy-induced toxicities.

Recurrent somatic genetic lesions are an integral component of risk stratification algorithms for pediatric B-lymphoblastic leukemia (B-ALL) for most large pediatric cancer consortia (Table 1). The majority of these lesions are structural chromosomal alterations that are associated with the development of disease and have prognostic implications.

Table 1.

Selected recurrent genetic alterations in childhood B-ALL

Genetic lesionIncidence in pediatric B-ALL, %OutcomePotential therapeutic implicationsComments
Aneuploidy     
 Hyperdiploidy DNA Index >1.16 or >50 chromosomes 20-25 Favorable  Some consortia consider specific trisomies, +4 and +10 considered favorable by COG 
 Hypodiploidy <44 chromosomes or DNA index <0.81 1-2 Unfavorable; consideration of HSCT in first CR Given common Ras pathway mutations; potential role for MEK or PI3K inhibitors Worsening prognosis with fewer chromosomes; frequent TP53 mutations in low hypodiploid (32-39 chromosome); Ras pathway mutations common 
Recurrent structural chromosomal aberrations     
 t(12;21)(p13;q22) (cryptic); ETV6-RUNX1 fusion 20-25 Favorable  Less common with increasing age 
 t(v;11)(v;q23) or t(11;v)(q23;v); KMT2A rearrangements ∼3 noninfant B-ALL; >75 infant B-ALL Unfavorable; noninfant improved with intensification of therapy; infant KMT2A-r dismal outcome regardless of therapy intensity Hypomethylating agents;DOT1L inhibitors; Menin-KMT2A protein-protein interaction inhibitors;PRMT5 inhibitors; LSD1 inhibitors KMT2A-AF4 most common fusion in B-ALL 
 +hsr(21)(q22); iAMP21 1-3 Unfavorable; improved with intensification of therapy  ≥5 copies of RUNX1 
 t(17;19)(q22;p13); TCF3-HLF <1 Very poor BCL2 inhibitor venetoclax Associated with hypercalcemia and coagulopathy 
 t(1;19)(q23;p13); TCF3-PBX1 Neutral with contemporary therapy  Poor outcome in older studies; higher incidence CNS disease and CNS relapse 
 t(5;14)(q31;q32); IL3-IGH 1-2 Neutral  Associated with peripheral eosinophilia 
 t(9;22)(q34;q11); BCR-ABL1 ∼5 Unfavorable with chemotherapy alone, greatly improved with TKI ABL-targeting TKIs (eg, imatinib, dasatinib, etc.) Incidence increases with age 
 Ph-like kinase fusions ∼20 of HR B-ALL Unfavorable ABL class (ABL1, ABL2, PDGFRB, CSF1R rearranged): imatinib/dasatinib; JAK activating (CRLF2, JAK2, EPOR rearrangements; IL7R indels/mutations, SH3B deletion): Ruxolitinib, other JAK inhibitors; NTRK3 fusions: Crizotinib, Larotrectinib; PTK2B fusion: FAK inhibitor Ongoing clinical trials investigating safety/efficacy of incorporation of TKIs into therapy 
 IGH-DUX4 3-7 Favorable  Associated with dysregulation of ETS transcription factor, ERG; IKZF1 deletion common 
 MEF2D rearranged (MEF2D-BCL9, MEF2D -HNRNPUL1, MEF2D-SS18, others) 3-6 Possibly unfavorable Potential HDAC inhibitors  
 ZNF384 rearranged (EP300-ZNF384, ARID1B-ZNF384, CREBBP-ZNF384, TCF3-ZNF384, others) 3-5 Neutral Potential HDAC inhibitors  
Common molecular lesions     
 IKZF1 deletion/mutation 15 B-ALL;30 HR B-ALL;60-80 Ph+;50-60 Ph-like;30-40 DUX4/ERG dysregulated Poor (except in DUX4/ERG dysregulated) FAK inhibition plus TKI (if other ABL class lesion present);retinoic acid Enriched at relapse; associated with glucocorticoid and TKI resistance 
 PAX5 deletions/mutations ∼30 B-ALL Neutral   
 TP53 mutations ∼5 B-ALL; 10-20 of relapsed B-ALL; >90 low-hypodiploid B-ALL (32-39 chromosomes) Poor  Somatic mutations enriched at relapse; >50% TP53 mutations in low-hypodiploid B-ALL are germ line; germ-line TP53 mutations associated with poor EFS/OS and increased risk for second malignancy 
 NT5C2 mutations 20 of relapsed B-ALL and T-ALL   Enzyme involved in nucleoside analog metabolism; gain of function mutations likely lead to decreased sensitivity to antimetabolite therapy 
 Ras pathway mutations At diagnosis incidence varies by type of B-ALL; ∼50 of relapsed B-ALL  MEK inhibitors;PI3K inhibitors  
 CREBBP mutations 20 of relapsed B-ALL   Associated with glucocorticoid resistance 
Genetic lesionIncidence in pediatric B-ALL, %OutcomePotential therapeutic implicationsComments
Aneuploidy     
 Hyperdiploidy DNA Index >1.16 or >50 chromosomes 20-25 Favorable  Some consortia consider specific trisomies, +4 and +10 considered favorable by COG 
 Hypodiploidy <44 chromosomes or DNA index <0.81 1-2 Unfavorable; consideration of HSCT in first CR Given common Ras pathway mutations; potential role for MEK or PI3K inhibitors Worsening prognosis with fewer chromosomes; frequent TP53 mutations in low hypodiploid (32-39 chromosome); Ras pathway mutations common 
Recurrent structural chromosomal aberrations     
 t(12;21)(p13;q22) (cryptic); ETV6-RUNX1 fusion 20-25 Favorable  Less common with increasing age 
 t(v;11)(v;q23) or t(11;v)(q23;v); KMT2A rearrangements ∼3 noninfant B-ALL; >75 infant B-ALL Unfavorable; noninfant improved with intensification of therapy; infant KMT2A-r dismal outcome regardless of therapy intensity Hypomethylating agents;DOT1L inhibitors; Menin-KMT2A protein-protein interaction inhibitors;PRMT5 inhibitors; LSD1 inhibitors KMT2A-AF4 most common fusion in B-ALL 
 +hsr(21)(q22); iAMP21 1-3 Unfavorable; improved with intensification of therapy  ≥5 copies of RUNX1 
 t(17;19)(q22;p13); TCF3-HLF <1 Very poor BCL2 inhibitor venetoclax Associated with hypercalcemia and coagulopathy 
 t(1;19)(q23;p13); TCF3-PBX1 Neutral with contemporary therapy  Poor outcome in older studies; higher incidence CNS disease and CNS relapse 
 t(5;14)(q31;q32); IL3-IGH 1-2 Neutral  Associated with peripheral eosinophilia 
 t(9;22)(q34;q11); BCR-ABL1 ∼5 Unfavorable with chemotherapy alone, greatly improved with TKI ABL-targeting TKIs (eg, imatinib, dasatinib, etc.) Incidence increases with age 
 Ph-like kinase fusions ∼20 of HR B-ALL Unfavorable ABL class (ABL1, ABL2, PDGFRB, CSF1R rearranged): imatinib/dasatinib; JAK activating (CRLF2, JAK2, EPOR rearrangements; IL7R indels/mutations, SH3B deletion): Ruxolitinib, other JAK inhibitors; NTRK3 fusions: Crizotinib, Larotrectinib; PTK2B fusion: FAK inhibitor Ongoing clinical trials investigating safety/efficacy of incorporation of TKIs into therapy 
 IGH-DUX4 3-7 Favorable  Associated with dysregulation of ETS transcription factor, ERG; IKZF1 deletion common 
 MEF2D rearranged (MEF2D-BCL9, MEF2D -HNRNPUL1, MEF2D-SS18, others) 3-6 Possibly unfavorable Potential HDAC inhibitors  
 ZNF384 rearranged (EP300-ZNF384, ARID1B-ZNF384, CREBBP-ZNF384, TCF3-ZNF384, others) 3-5 Neutral Potential HDAC inhibitors  
Common molecular lesions     
 IKZF1 deletion/mutation 15 B-ALL;30 HR B-ALL;60-80 Ph+;50-60 Ph-like;30-40 DUX4/ERG dysregulated Poor (except in DUX4/ERG dysregulated) FAK inhibition plus TKI (if other ABL class lesion present);retinoic acid Enriched at relapse; associated with glucocorticoid and TKI resistance 
 PAX5 deletions/mutations ∼30 B-ALL Neutral   
 TP53 mutations ∼5 B-ALL; 10-20 of relapsed B-ALL; >90 low-hypodiploid B-ALL (32-39 chromosomes) Poor  Somatic mutations enriched at relapse; >50% TP53 mutations in low-hypodiploid B-ALL are germ line; germ-line TP53 mutations associated with poor EFS/OS and increased risk for second malignancy 
 NT5C2 mutations 20 of relapsed B-ALL and T-ALL   Enzyme involved in nucleoside analog metabolism; gain of function mutations likely lead to decreased sensitivity to antimetabolite therapy 
 Ras pathway mutations At diagnosis incidence varies by type of B-ALL; ∼50 of relapsed B-ALL  MEK inhibitors;PI3K inhibitors  
 CREBBP mutations 20 of relapsed B-ALL   Associated with glucocorticoid resistance 

CNS, central nervous system; COG, Children’s Oncology Group; CR, complete remission; EFS, event-free survival; ETS, erythroblast transforming specific; HDAC, histone deacetylase inhibitor; HR, high risk; HSCT, hematopoietic stem cell transplant; iAMP21, intrachromosomal amplification of chromosome 21; IL7R, interleukin-7 receptor; OS, overall survival; Ph+, Philadelphia chromosome; T-ALL, T-cell acute lymphoblastic leukemia; TKI, tyrosine kinase inhibitor.

Hyperdiploidy (modal chromosome numbers 51-65 or DNA index of >1.16) is common in B-ALL, occurring in 20% to 25% of pediatric patients and decreasing in frequency with increasing age. Patients with hyperdiploidy generally do well, with studies from the Children’s Oncology Group (COG) finding that specific trisomies (trisomy of chromosomes 4 and 10) in particular are linked to a favorable outcome1  (Table 1).

Conversely, hypodiploidy with modal chromosome number <44 or DNA index of <0.81 has been associated with a dismal outcome, resulting in hematopoietic stem cell transplant (HSCT) in first complete remission (CR).2  However, recent data from a small series of patients treated at a single institution suggest that, if a patient with hypodiploidy has a bone marrow that is negative for minimal residual disease (MRD) by the end of induction therapy, chemotherapy alone may be curative.3  Recently analyzed data from the COG indicate that, although patients with an end of induction MRD < 0.01% fare better than those with MRD-positive disease, their overall outcomes with chemotherapy alone were still suboptimal, with a 5-year disease-free survival of 60.3% ± 9.2%. However, this study found that HSCT in first CR did not confer a survival benefit, suggesting a need for the development of novel therapeutic strategies for this poor prognosis subset.4 

In addition to aneuploidy, a number of chromosomal translocations and other structural chromosomal aberrations with prognostic impact are common in B-ALL. The most common translocation in pediatric B-ALL is t(12;21), which results in the ETV6-RUNX1 fusion gene. ETV6-RUNX1 fusions are present in 20% to 25% of pediatric B-ALL cases, and they are associated with an excellent outcome.5  Conversely, rearrangements involving the KMT2A gene (formerly MLL) on chromosome 11q23 are associated with higher levels of residual disease at the end of induction, which when present, are associated with inferior outcomes. Approximately 80% of infants with B-ALL have KMT2A-r compared with 3% to 5% in older children with B-ALL. Whereas infants with KMT2A rearrangements have a dismal prognosis,6  older patients have a more favorable outcome with appropriate intensification of therapy.7  Additionally, intrachromosomal amplification of chromosome 21 is a recurrent lesion in 1% to 3% of pediatric B-ALL cases that is associated with increased risk of relapse. However, treating affected patients with intensification of chemotherapy can mitigate this risk.7-9  Also, B-ALL harboring the translocation t(17;19), which results in the TCF3-HLF fusion gene, is characterized by hypercalcemia, coagulopathy, and a dismal outcome. Although rare (<1% of B-ALL patients), the recognition of t(17;19) is critical, because such patients warrant early consideration of HSCT and are candidates for novel agents—recent preclinical data suggest that these patients may be sensitive to BCL-2 inhibition.10 

Rearrangements involving monocyte enhancer factor D2 (MEF2D) occur in 3% to 4% of pediatric B-ALL and tend to occur in older children.11 MEF2D rearrangements result in fusion with multiple partners, all leading to similar gene expression profiles, including overexpression of the MEF2D target HDAC9, and implicating HDAC inhibition as a potential therapeutic strategy for MEF2D-rearranged patients. Retrospective studies of small cohorts of patients have found an association between MEF2D fusions and inferior outcome, although prospective studies of larger cohorts are needed.11,12 

Additionally, rearrangements of the zinc finger protein 384 gene (ZNF384) have been identified in 4% to 5% of pediatric B-ALL patients.13,14  These rearrangements lead to the fusion of ZNF384 with multiple partners, including TCF3, EWS1, CREBBP, and EP300.13,14 ZNF384-rearranged B-ALL is characterized by weak CD10 expression and aberrant expression of myeloid markers and a gene expression signature enriched for stem cell–related genes.13  Based on small cohorts thus far examined, it seems that the outcomes of patients with ZNF384 fusions are not statistically different than those without, although fusion partner-specific effects may exist. Hirabayashi et al13  reported that patients with TCF3-ZNF384 fusions were characterized by younger age at diagnosis, relative resistance to corticosteroids, and higher risk of relapse. Additional studies are needed to definitively determine the impact on outcome of these fusions.

Perhaps the best characterized structural chromosomal alteration of B-ALL is the t(9;22) Philadelphia chromosome (Ph+), leading to the BCR-ABL1 fusion gene.15  Although historically associated with a very poor outcome, incorporation of continuously administered tyrosine kinase inhibitors (TKIs) targeting BCR-ABL1, such as imatinib and dasatinib, into standard chemotherapy without HSCT has dramatically improved outcomes of Ph+ B-ALL patients in recent years and is now the well-accepted standard of care treatment of this disease.16-18 

Global gene expression analyses of B-ALL cohorts identified a group of patients whose disease had a gene expression profile largely overlapping with Ph+ B-ALL, despite lacking the BCR-ABL1 fusion gene. Such cases are classified as Ph-like B-ALL (or BCR-ABL1–like B-ALL), and as a group, they have an extremely poor prognosis. Ph-like disease is common in high-risk B-ALL, including in over 20% of National Cancer Institute (NCI) high-risk patients19,20  (Figure 1). The incidence of Ph-like disease increases with age from ∼10% in NCI standard-risk patients to over 20% of adolescents, with an apparent peak in young adults with B-ALL, over 30% of whom will have Ph-like disease.20-22 

Figure 1.

Frequencies of kinase subgroups in NCI high-risk (HR) and standard-risk (SR) patients who are Ph like. Reprinted from Reshmi et al20  with permission.

Figure 1.

Frequencies of kinase subgroups in NCI high-risk (HR) and standard-risk (SR) patients who are Ph like. Reprinted from Reshmi et al20  with permission.

Close modal

Extensive genomic studies have now identified a number of lesions driving Ph-like B-ALL, comprising a host of genomic alterations and leading to activation of kinase signaling pathways.20-27  Importantly, many of the identified kinase fusions and mutations are targetable by clinically available small molecule inhibitors of tyrosine kinases, offering hope that targeted therapy may improve outcome in affected patients akin to the improved outcomes in Ph+ acute lymphoblastic leukemia (ALL).

Approximately 1/2 of all Ph-like ALL cases harbor lesions leading to oncogenic overexpression of the CLRF2 gene, which encodes the thymic stromal lymphopoietin receptor that, together with the interleukin-7 receptor (IL7R), activates the JAK/STAT pathway on ligand binding (Figure 1). Therefore, overexpression of CRLF2 leads to aberrant hyperactivation of the JAK/STAT pathway. Curiously, ∼1/2 of CRLF2-overexpressing B-ALLs also have a concomitant activating mutation of JAK1 or JAK2. Additionally, rearrangements of the erythropoietin receptor (EPOR) that generate a truncated protein lacking the negative regulatory domain drive a small subset of Ph-like B-ALL via constitutive activation of the JAK/STAT signaling pathway.28  These, along with a handful of other JAK/STAT-activating lesions (including JAK2 fusions, IL7R insertion/deletions and mutations, and SH2B3 deletions), point toward the JAK pathway as an attractive potential target for the treatment of this prominent subset of Ph-like B-ALL. A number of clinical trials including pediatric patients are currently ongoing to assess the safety, tolerability, and efficacy of the combination of the JAK1/2 inhibitor, ruxolitinib, with standard chemotherapy for JAK-activated Ph-like B-ALL (NCT02723994, NCT03117751, and NCT02420717).

Additionally, a number of kinase fusions leading to activation of ABL class kinases, such as ABL1, ABL2, CSF1R, PDGFRA/B, and LYN, occur in 3% to 5% of pediatric B-ALL cases.20-22  Such fusions are predicted to be targetable by ABL kinase inhibitors, such as imatinib and dasatinib. Given the known safety and tolerability of these TKIs administered continuously on a standard intensified chemotherapy backbone, a number of clinical trial groups are now studying the efficacy of continuous dasatinib plus standard chemotherapy for Ph-like patients with ABL class lesions (NCT02883049, NCT03117751, and NCT02420717). Lesions of other activating signaling kinases, such as NTRK3, FLT3, and FGFR1, are rare but represent additional potentially targetable lesions in Ph-like ALL. Promising preclinical data and anecdotal reports of patient responses to TKI therapy provide hope that a precision medicine approach to Ph-like B-ALL will ultimately improve the outcome of this clinically challenging subset of patients.

A boon of recent genomic studies has greatly expanded our view of the genomic landscape of B-ALL (Table 1). Such studies have identified a number of recurrent genomic lesions with potential prognostic and therapeutic implications. Analyses of DNA copy neutral alteration by single-nucleotide polymorphism array studies have identified recurrent deletion of several transcription factors critical to normal B-cell development, including PAX5, EBF1, and IKZF1.29  Although deletion of each of these genes likely contributes to the development of B-ALL, only IKZF1 deletion seems to impact prognosis, with IKZF1 deletions and mutations being associated with a poor prognosis.30-32  The prognostic strength of IKZF1 status can be further refined by integration of additional genomic lesions. In a large study of patients treated on the Associazione Italiana Ematologia ed Oncologia Pediatrica-Berlin-Frankfurt-Muenster ALL 2000 trial, patients with IKZF1 deletion and concomitant deletion of CDKN2A, CKN2B, PAR1, or PAX5 without deletion of ERG (termed IKZF1plus) had a significantly worse 5-year event-free survival (EFS) than those with IKZF1 deletions without the IKZF1plus status, and those without IKZF1 deletions (53% ± 6% vs 79% ± 5% vs 87% ± 1%, respectively).33 IKZF1 lesions are highly enriched in high-risk subsets of B-ALL, including over 60% of Ph+ ALL and high frequency in Ph-like B-ALL, which suggests a specific interaction with these activating kinase lesions.19,30,34,35  Work to investigate the features of IKZF1 lesions that lead to a poor prognosis has found that loss of IKAROS (the protein encoded by IKZF1) function leads to a stem cell–like gene expression signature and increased adhesion to the bone marrow niche. In addition to potential use as a prognostic marker, preclinical work in Ph+ ALL has identified potential targeted therapeutic interventions for B-ALL with IKZF1 lesions. The efficacy of retinoids as well as pharmacologic inhibition of the FAK kinase in combination with TKIs has been shown in preclinical studies of IKZF1-deleted/mutated Ph+ B-ALL.36,37 

More recently identified recurrent genomic lesions in B-ALL with distinct transcriptional signatures and potential prognostic impact include combined deregulation of DUX4, a homeobox transcription factor, and ERG, a member of the erythroblast transforming–specific transcription factor family. These occur in 7% of pediatric B-ALL, and they are associated with favorable outcome.38  Interestingly, DUX4-ERG–dysregulated B-ALL frequently co-occurs with deletions of IKZF1, but unlike in other B-ALL subtypes, concomitant IKZF1 deletion does not seem to negatively impact outcome.38 

In contrast to B-ALL, recurrent genomic lesions are not widely incorporated into the risk stratification schemas for children with T-cell acute lymphoblastic leukemia (T-ALL) (Table 2). Clinical data suggest limited impact of any given genomic lesion on outcome with modern therapy. Therefore, current risk stratification for therapy generally relies on clinical features, such as extramedullary involvement and early response to induction therapy. Indeed, even white blood cell count and age at diagnosis, known to be prognostically significant for B-ALL, are not considered for patients with T-ALL. However, integration of genetic information and MRD for risk stratification is being explored, and potentially targetable genetic lesions are common; thus, possible future applications for genomics of T-ALL are on the horizon.

Table 2.

Selected recurrent genetic alterations in childhood T-ALL

Genetic lesionIncidence in pediatric T-ALL, %OutcomePotential therapeutic implicationsComments
Recurrent structural chromosomal aberrations     
 t(1;14)(p32;q11); t(1;7)(p32)(q34);TAL1 fusions 30-40 Possibly unfavorable  Associated with PTEN/PI3K mutations and 6q deletion 
 t(10;14)(q24;q11); TLX1 fusion 5-10 Possibly favorable Given co-occurrence of lesions, resulting signaling pathway activation, possible utility of JAK inhibitors and MEK inhibitors Associated with PHF6 mutations, JAK/STAT- and Ras-activated signaling mutations; NUP214-ABL1 fusions that activate STAT5 and Ras-MAPK pathways 
 t(11;14)(p15;q11), t(5;14)(q35;q32); TLX3 fusions 20-25 Possibly unfavorable Given co-occurrence of lesions, resulting signaling pathway activation, possible utility of JAK inhibitors and MEK inhibitors Associated with near–ETP-ALL; association with WT1 mutations, PHF6 mutations, JAK/STAT- and Ras-activated signaling mutations; NUP214-ABL1 fusions that activate STAT5 and Ras-MAPK pathways 
 t(11;14)(p15;q11); LMO1 fusion <5 Possibly unfavorable  Usually occurs with LYL1 or TAL1 fusions 
 t(11;13)(p15;q11); LMO2 fusion 3-5 Possibly unfavorable  Usually occurs with LYL1 or TAL1 fusions; highly enriched for ETP-ALL 
 7p15 translocations; HOXA overexpression 2-3 Possibly unfavorable  Associated with PHF6 and EZH2 mutations 
 11q23 translocations; KMT2A rearrangements Some studies possibly unfavorable, some possibly favorable  Included in HOXA-overexpressing subgroup 
 t(10;11)(p13;q21); PICALM-MLLT10 5-10 Possibly unfavorable  Included in HOXA-overexpressing subgroup; associated with EZH2 mutations 
 Inv14(q11;q13); NKX2-1 fusion ∼5 Neutral  Associated with LEF1 deletion/mutation 
Common molecular lesions     
 NOTCH1 mutations >70 Possibly favorable γ secretase inhibitors Associated with CDKN2A/B deletions and FBXW7 deletions/mutation; less common in TAL1 T-ALL 
 CDKN2A/B deletion 50-60 Possibly favorable  Associated with NOTCH1 mutations; less common in ETP-ALL 
 FBXW7 mutations/deletions ∼20 Possibly favorable  Negative regulator of NOTCH1; mutations/deletions associated with NOTCH1 mutations 
 PTEN mutations ∼20 Possibly unfavorable  Associated with TAL1 T-ALL 
 PHF6 deletions/mutations 20-25   Epigenetic regulator; mutations/deletions common in TLX1/3, HOXA-overexpressing, TAL1, and NKX2-1 T-ALL 
 Ras pathway mutations  Possibly unfavorable MEK and PI3K inhibitors Associated with ETP-ALL 
Genetic lesionIncidence in pediatric T-ALL, %OutcomePotential therapeutic implicationsComments
Recurrent structural chromosomal aberrations     
 t(1;14)(p32;q11); t(1;7)(p32)(q34);TAL1 fusions 30-40 Possibly unfavorable  Associated with PTEN/PI3K mutations and 6q deletion 
 t(10;14)(q24;q11); TLX1 fusion 5-10 Possibly favorable Given co-occurrence of lesions, resulting signaling pathway activation, possible utility of JAK inhibitors and MEK inhibitors Associated with PHF6 mutations, JAK/STAT- and Ras-activated signaling mutations; NUP214-ABL1 fusions that activate STAT5 and Ras-MAPK pathways 
 t(11;14)(p15;q11), t(5;14)(q35;q32); TLX3 fusions 20-25 Possibly unfavorable Given co-occurrence of lesions, resulting signaling pathway activation, possible utility of JAK inhibitors and MEK inhibitors Associated with near–ETP-ALL; association with WT1 mutations, PHF6 mutations, JAK/STAT- and Ras-activated signaling mutations; NUP214-ABL1 fusions that activate STAT5 and Ras-MAPK pathways 
 t(11;14)(p15;q11); LMO1 fusion <5 Possibly unfavorable  Usually occurs with LYL1 or TAL1 fusions 
 t(11;13)(p15;q11); LMO2 fusion 3-5 Possibly unfavorable  Usually occurs with LYL1 or TAL1 fusions; highly enriched for ETP-ALL 
 7p15 translocations; HOXA overexpression 2-3 Possibly unfavorable  Associated with PHF6 and EZH2 mutations 
 11q23 translocations; KMT2A rearrangements Some studies possibly unfavorable, some possibly favorable  Included in HOXA-overexpressing subgroup 
 t(10;11)(p13;q21); PICALM-MLLT10 5-10 Possibly unfavorable  Included in HOXA-overexpressing subgroup; associated with EZH2 mutations 
 Inv14(q11;q13); NKX2-1 fusion ∼5 Neutral  Associated with LEF1 deletion/mutation 
Common molecular lesions     
 NOTCH1 mutations >70 Possibly favorable γ secretase inhibitors Associated with CDKN2A/B deletions and FBXW7 deletions/mutation; less common in TAL1 T-ALL 
 CDKN2A/B deletion 50-60 Possibly favorable  Associated with NOTCH1 mutations; less common in ETP-ALL 
 FBXW7 mutations/deletions ∼20 Possibly favorable  Negative regulator of NOTCH1; mutations/deletions associated with NOTCH1 mutations 
 PTEN mutations ∼20 Possibly unfavorable  Associated with TAL1 T-ALL 
 PHF6 deletions/mutations 20-25   Epigenetic regulator; mutations/deletions common in TLX1/3, HOXA-overexpressing, TAL1, and NKX2-1 T-ALL 
 Ras pathway mutations  Possibly unfavorable MEK and PI3K inhibitors Associated with ETP-ALL 

ETP-ALL, early thymic precursor or early T-cell precursor acute lymphoblastic leukemia.

Structural chromosomal aberrations are not prognostic in T-ALL, including rearrangements involving TAL1, KMT2A, MLLT10, TLX1, and TLX3. Deletions of CDKN2A/B and PTEN are present in ∼50% and ∼20%, respectively. The most commonly mutated gene in T-ALL is the transmembrane receptor involved in normal T-cell development, NOTCH1, present in >70% of patients. Mutations of the ubiquitin ligase gene, FBXW7, activated signaling pathways, and epigenetic regulators are also common.39 

Efforts to incorporate genetic lesions to refine risk stratification are being explored. One example is a recent study by the pediatric French Acute Lymphoblastic Leukemia Study Group.40 

The researchers defined patients as genetic low risk (gLoR) if they had a mutation of either NOTCH1 or FBXW7 and lacked mutations of the Ras pathway and PTEN. All other patients were classified as genetic high risk (gHiR). Combining genetic risk and end of induction MRD at a threshold of 10−4 allowed for a refinement of risk stratification, distinguishing a group with an excellent outcome (gLoR: MRD < 10−4 and low cumulative risk of relapse [CIR] =4%) from a group with a very poor expected outcome (gHiR: MRD > 10−4, CIR = 43%). Other patient groups (gLoR: MRD > 10−4 and gHiR: MRD < 10−4) had intermediate outcomes.40  Thus, if these results are validated in additional cohorts, adopting an approach integrating clinical features, genetic lesions, and early response to therapy may allow for improved risk stratification of T-ALL.40 

The frequency of activating NOTCH1 mutations and loss of function mutations of the negative NOTCH1 regulator, FBXW7, in T-ALL makes the NOTCH1 signaling pathway an attractive potential therapeutic target. Targeting NOTCH1 signaling using γ-secretase inhibitors that prevent release of the transcriptional activating intracellular portion of NOTCH1 from the membrane has shown promising preclinical activity but thus far, limited clinical efficacy and high rates of dose-limiting gastrointestinal toxicity.41  NOTCH1 inhibitory antibodies are also being explored as an NOTCH1 targeting strategy. Additionally, NOTCH1 activation directly upregulates MYC; thus, targeting MYC may be an effective strategy for T-ALL with NOTCH1 activation.41 

Early thymic precursor or early T-cell precursor (ETP)-ALL is a distinct subtype of T-ALL characterized by a stem progenitor cell gene expression signature and an immature immunophenotype. Initial reports found that pediatric patients with ETP-ALL were a high-risk subset associated with poor response to therapy and high risk of relapse.42  More recent pediatric data suggest that, despite having higher rates of MRD positivity at early time points, with MRD-directed intensification of therapy, outcomes are not significantly different than typical T-ALL.43-45  Therefore, ETP status is currently not universally incorporated into risk stratification for pediatric T-ALL.

Although not independently prognostic, the requisite for intensive therapy for cure in ETP-ALL indicates that this subset of patients could benefit from more targeted approaches to therapy. The molecular landscape of ETP-ALL reveals a number of potentially targetable opportunities, including mutations of activated signaling pathways.38 

Recurrent structural chromosomal and molecular lesions are the most powerful prognostic predictors for patients with acute myeloid leukemia (AML) (Table 3). However, current risk stratification of pediatric AML takes into consideration only a limited number of somatic genetic lesions, many of which, although common in adult AML, affect only a small fraction of pediatric patients.46,47  Therefore, the majority of pediatric patients lack any prognostic genetic lesion. Recent genome-sequencing efforts of large cohorts of pediatric AML cases, including the Therapeutically Applicable Research to Generate Effective Treatments AML initiative and the St. Jude Children’s Research Hospital-Washington University Pediatric Cancer Genome Project, have identified genetic lesions enriched in pediatric AML, including novel fusion genes, mutations, and deletions that may have important clinical implications specific to pediatric AML.46,47 

Table 3.

Select recurrent genetic lesions in childhood AML

Genetic lesionIncidence in pediatric AML, %OutcomePotential therapeutic implicationsComments
Recurrent structural chromosomal aberrations     
 t(15;17)(q22;q12) and other rare variants; PML-RARA or other RARA fusion; rare RARB and RARG fusions ∼5 Favorable ATRA; arsenic Acute promyelocytic leukemia; associated with life-threatening coagulopathy; differentiation syndrome with ATRA 
 t(8;21)(q22;q22); RUNX1-RUNX1T1 ∼15 Favorable Dasatinib (targeting KIT kinase) Associated with mutations of activated signaling, most commonly KIT and NRAS/KRAS; associated with -X/-Y, mutations of chromatin modifiers and cohesin complex members 
 Inv(16)/t(16;16)(p13.1;q22); CBFβ-MYH11 10-15  Dasatinib (targeting KIT kinase) Often M4Eo morphology; associated with mutations of activated signaling, most commonly KIT and NRAS/KRAS 
 t(11;v)(q23;v);t(v;11)(v;q23); KMT2A rearrangements; KMT2A-AF9 most common 10-15 children and adolescents; 35-50 infants Overall neutral but fusion-specific impact on prognosis Hypomethylating agents; DOT1L inhibitors; menin-KMT2A protein-protein interaction inhibitors; PRMT5 inhibitors; LSD1 inhibitors Also common in topo II inhibitor–associated t-AML 
 Monosomy 7, del(7q) 1-3 Unfavorable  Del(7q) is relatively common in CBF AML but is not associated with worse outcome 
 Monosomy 5, del(5q) 1-2 Unfavorable   
 11q15 cryptic translocations; NUP98 fusions most commonly NUP98-NSD1 4-5 Possibly unfavorable; combined FLT3-ITD and NUP98-NSD1 fusion associated with poor prognosis  Most common in children 3-14 y; associated with FLT3-ITD and WT1 mutations; HOX overexpression; NUP98-KDM5A common in AMKL, where it is highly associated with mono- and biallelic RB1 deletion 
 t(1;22)(p13;q13); RBM15-MKL1 fusion ∼10 of AMKL Intermediate   
 Inv(16)(p13.3q24.3); CBFA2T3-GLS2 fusion 15-20 AMKL Unfavorable GLI inhibitors (GANT61)  
 HOX gene fusions ∼15 AMKL Intermediate  Associated with CTCF/Cohesin, MPL, and activated signaling pathway lesions 
Common molecular lesions     
 FLT3-ITD mutations 15-20 High–allelic ratio FLT3-ITD mutation associated with poor outcome; FLT3-ITD plus NPM1 mutations favorable; FLT3-ITD plus WT1 or NUP98-NSD1 unfavorable FLT3-targeting TKIs (eg, sorafenib, quizartinib, midostaurin) Associated with NPM1 and WT1 mutations and NUP98-NSD1 fusion 
 Other FLT3 mutations 10-15 Possibly poor outcome Non-D835Y mutations may be sensitive to FLT3-targeting TKIs  
 NPM1 mutations ∼10 Favorable DOT1L inhibitors; menin-KMT2A inhibitors Exceedingly rare in children <3 y, increased incidence with age; associated with FLT3-ITD mutations; trilineage dysplasia in 25% 
 CEBPA mutations 5-10 Favorable  More common in adolescents; associated with normal karyotype 
 Ras pathway mutations 40-50 Neutral MEK inhibitors; PI3K inhibitors Highly prevalent in infant AML, decreased incidence with age; associated with CBF AML 
 KIT mutations 10-15; 20-25 of CBF AML Possibly worse outcome for CBF patients with KIT mutations Dasatinib  
 WT1 mutations 15 Neutral but combination of FLT3-ITD and WT1 mutation associated with poor prognosis  Associated with FLT3-ITD mutations 
Genetic lesionIncidence in pediatric AML, %OutcomePotential therapeutic implicationsComments
Recurrent structural chromosomal aberrations     
 t(15;17)(q22;q12) and other rare variants; PML-RARA or other RARA fusion; rare RARB and RARG fusions ∼5 Favorable ATRA; arsenic Acute promyelocytic leukemia; associated with life-threatening coagulopathy; differentiation syndrome with ATRA 
 t(8;21)(q22;q22); RUNX1-RUNX1T1 ∼15 Favorable Dasatinib (targeting KIT kinase) Associated with mutations of activated signaling, most commonly KIT and NRAS/KRAS; associated with -X/-Y, mutations of chromatin modifiers and cohesin complex members 
 Inv(16)/t(16;16)(p13.1;q22); CBFβ-MYH11 10-15  Dasatinib (targeting KIT kinase) Often M4Eo morphology; associated with mutations of activated signaling, most commonly KIT and NRAS/KRAS 
 t(11;v)(q23;v);t(v;11)(v;q23); KMT2A rearrangements; KMT2A-AF9 most common 10-15 children and adolescents; 35-50 infants Overall neutral but fusion-specific impact on prognosis Hypomethylating agents; DOT1L inhibitors; menin-KMT2A protein-protein interaction inhibitors; PRMT5 inhibitors; LSD1 inhibitors Also common in topo II inhibitor–associated t-AML 
 Monosomy 7, del(7q) 1-3 Unfavorable  Del(7q) is relatively common in CBF AML but is not associated with worse outcome 
 Monosomy 5, del(5q) 1-2 Unfavorable   
 11q15 cryptic translocations; NUP98 fusions most commonly NUP98-NSD1 4-5 Possibly unfavorable; combined FLT3-ITD and NUP98-NSD1 fusion associated with poor prognosis  Most common in children 3-14 y; associated with FLT3-ITD and WT1 mutations; HOX overexpression; NUP98-KDM5A common in AMKL, where it is highly associated with mono- and biallelic RB1 deletion 
 t(1;22)(p13;q13); RBM15-MKL1 fusion ∼10 of AMKL Intermediate   
 Inv(16)(p13.3q24.3); CBFA2T3-GLS2 fusion 15-20 AMKL Unfavorable GLI inhibitors (GANT61)  
 HOX gene fusions ∼15 AMKL Intermediate  Associated with CTCF/Cohesin, MPL, and activated signaling pathway lesions 
Common molecular lesions     
 FLT3-ITD mutations 15-20 High–allelic ratio FLT3-ITD mutation associated with poor outcome; FLT3-ITD plus NPM1 mutations favorable; FLT3-ITD plus WT1 or NUP98-NSD1 unfavorable FLT3-targeting TKIs (eg, sorafenib, quizartinib, midostaurin) Associated with NPM1 and WT1 mutations and NUP98-NSD1 fusion 
 Other FLT3 mutations 10-15 Possibly poor outcome Non-D835Y mutations may be sensitive to FLT3-targeting TKIs  
 NPM1 mutations ∼10 Favorable DOT1L inhibitors; menin-KMT2A inhibitors Exceedingly rare in children <3 y, increased incidence with age; associated with FLT3-ITD mutations; trilineage dysplasia in 25% 
 CEBPA mutations 5-10 Favorable  More common in adolescents; associated with normal karyotype 
 Ras pathway mutations 40-50 Neutral MEK inhibitors; PI3K inhibitors Highly prevalent in infant AML, decreased incidence with age; associated with CBF AML 
 KIT mutations 10-15; 20-25 of CBF AML Possibly worse outcome for CBF patients with KIT mutations Dasatinib  
 WT1 mutations 15 Neutral but combination of FLT3-ITD and WT1 mutation associated with poor prognosis  Associated with FLT3-ITD mutations 

AMKL, acute megakaryoblastic leukemia; ATRA, all-trans-retinoic acid; CBF, core binding factor; ITD, internal tandem duplication; M4Eo, monocytic acute myeloid leukemia with eosinophilia; NUP98, nucleoporin 98-kDa; RARA, retinoic acid receptor α; RARB, retinoic acid receptor β; RARG, retinoic acid receptor γ; TKI, tyrosine kinase inhibitor.

Structural chromosomal aberrations creating fusion genes affecting the hematopoiesis-regulating transcription factor core binding factor complex are present in ∼25% of pediatric AML. The α subunit of this transcription factor complex, RUNX1, is fused to the RUNX1T1 gene in t(8;21) AML, whereas the β cofactor subunit of the complex, CBFb, is fused to MHY11 in inv(16) or t(16;16) AML. Collectively, these lesions are associated with a relatively favorable outcome, and therefore, they are not routinely offered HSCT in first CR. Conversely, the recurrent structural chromosomal aberrations of monosomy 5, 5q- and monosomy 7 are associated with particularly poor outcomes, with HSCT in first CR a generally accepted standard of care, although such lesions are quite uncommon in pediatric patients.48  Complex karyotypes (≥3 structural chromosomal lesions) are relatively common in pediatric AML; however, in contrast to adult disease, they are not correlated with outcome, and therefore, they are not routinely incorporated into risk classification for children.48  Another set of well-characterized structural chromosomal lesions in AML is lesions of 11q23 leading to fusion of the KMT2A gene with a host of different partners. Although the prognostic impact of KMT2A-r as a group has not been consistently shown , clinical data suggest that certain KMT2A rearrangements, such as t(10;11)(p11.2;q23) and t(6;11)(q27;q23), may portend a particularly poor prognosis, whereas the t(1;11)(q21;q23) translocation has been associated with an excellent outcome.49 

A number of recent studies have identified recurrent cryptic translocations involving the nucleoporin 98-kDa (NUP98) gene on chromosome 11p15 in AML in ∼4% of pediatric AML patients, with decreasing frequency with increasing age.50  Although the most common fusion partner is the histone methyltransferase gene, NSD1, over 30 different fusion partners have been identified to date. NUP98-rearranged AML occurs almost exclusively in cytogenetically normal AML with frequent concomitant FLT3-internal tandem duplication (-ITD) and WT1 mutations and is characterized by aberrant HOX gene expression.46,50-52  Several retrospective studies have found that patients with NUP98 rearrangement have a particularly poor outcome, suggesting the possible utility of incorporating NUP98 status into risk classification.50-52  However, as discussed below, the constellation of co-occurring mutations likely drives the prognostic impact rather than NUP98 status considered in isolation.46,50 

Specific fusions involving the retinoic acid receptor α (RARA) due to balanced translocations, most commonly, t(15;17)(q22;q12) producing PML-RARA and very rarely, fusions involving retinoic acid receptor β and retinoic acid receptor γ, cause acute promyelocytic leukemia (APL), a clinically and biologically distinct form of AML.53,54  Patients with APL generally have an excellent outcome, but they have relatively high rates of early death due to severe coagulopathy and differentiation syndrome. The RARA fusion genes result in the production of an abnormal retinoid acid receptor that causes the repression of RARA target genes, leading to blocked differentiation at the promyelocytic stage. This differentiation block can be overcome by treatment with all-trans-retinoic acid (ATRA), and treatment regimens combining ATRA and standard AML-directed chemotherapy are associated with excellent outcomes in APL, with CR rates up to 95% and survival >80%. Additionally, arsenic trioxide is a particularly active agent in APL, thought to work by binding to the PML portion of the PML-RARA fusion and causing its degradation. Adult studies have shown that, for low- and standard-risk APL patients, the combination of ATRA and arsenic without chemotherapy is as efficacious as standard chemotherapy but with much less toxicity.55  Studies of this combination for the upfront treatment of pediatric APL patients are ongoing (NCT02339740 and NCT01409161).

One of the most commonly mutated genes in AML is the receptor tyrosine kinase gene, FLT3. ITD mutations of the juxtamembrane domain (JMD) and point mutations of Asp835 in the tyrosine kinase domain (TKD) are the most common mutations of FLT3 in AML. FLT3-TKD mutations occur in around 10% of adult and pediatric AML patients, whereas FLT3-ITD mutations are far more common in adults, present in ∼35% compared with only 10% to 15% of pediatric patients.56  Although FLT3-TKD mutations do not seem to significantly impact prognosis, dominant FLT3-ITD lesions, as evidenced by an allelic ratio >0.4, are associated with a poor outcome.56 

Patients with high allelic ratio FLT3-ITD lesions are generally categorized as high-risk patients, warranting HSCT in first CR. In addition to its prognostic implications, as a receptor tyrosine kinase, FLT3 is potentially targetable by small molecule inhibitors.57  A number have been used in clinical trials, including sorafenib in a current AML trial recently run by the COG (NCT01371981). Adult studies looking at other TKIs targeting FLT3 have had promising results, most prominently the improved overall survival and EFS observed in adults with FLT3-mutated AML treated with the multikinase inhibitor midostaurin, which is now Food and Drug Administration approved for this use.58,59  Resistance to TKIs can arise through a variety of cell intrinsic and extrinsic mechanisms, including the emergence of resistance-conferring mutations of the TKD of FLT3.60,61  Many of the commonly occurring TKD mutations remain sensitive to some of the newer TKIs, such as crenolanib and gilteritinib, and therefore, they could be efficacious for patients in whom resistance to first generation TKIs has developed.62,63  Although FLT3-ITD mutations are less frequent in pediatric AML patients compared with adult AML patients, pediatric-specific FLT3 mutations have been recently identified. Novel point mutations and small insertions/deletions of not only the TKD but also, the JMD and trans-membrane domain have been identified in children.46,64  These mutations are associated with poor response to standard therapy but display exquisite sensitivity to FLT3-targeting TKIs in in vitro studies.46,64  Thus, identification of such mutations at diagnosis could inform risk stratification and point toward targeted therapeutic intervention.46,64 

In contrast to FLT3-ITD mutations, recurrent mutations of NPM1 and CEBPA genes are associated with a favorable outcome in pediatric AML.65,66  Mutations of NPM1 constitute one of the most frequent mutations in adult AML but are relatively rare in pediatric disease, occurring in <10% of patients.46,65 CEBPA mutations are also uncommon in pediatric AML, found in ∼5% of patients, and they are more common in older children compared with infant and young children.46,66  Both NPM1-mutant and CEBPA-mutant AML are now provisional entities in the World Health Organization classification of myeloid malignancies. Although rare, routine screening for NPM1 and CEBPA mutations is recommended, because such patients are candidates for treatment with chemotherapy alone. Additionally, novel therapeutic strategies for the treatment of NPM1-mutant AML are being explored. Mutant NPM1 seems to disrupt normal chromatin structure, leading to aberrant HOX gene expression, and targeting of histone modifications, including DOT1L inhibitors and inhibitors of the menin-KMT2A interaction, has shown promise in preclinical investigations.67  Interestingly, a small clinical trial in adults with NPM1-mutant AML showed dactinomycin as a promising agent, likely working because NPM1-mutant cells are more vulnerable to the dactinomycin-induced nucleolar stress response.68 

Other recurrent mutations in pediatric AML include mutations of the zinc finger transcription factor gene, WT1. Mutations of this gene are found in around 10% of children. Although patients with WT1 mutations have a worse EFS and overall survival, when combined with FLT3 status and cytogenetics, WT1 status has no clear prognostic impact.69  Additionally, a number of molecular lesions common in adult AML, including mutations of epigenetic modifiers DNMT3A, TET2, and IDH1/2, are rare in pediatric disease.46,70,71  Other significant differences in the frequency and spectrum of mutations of genes, such as WT1, MYC, GATA2, CBL, NRAS, and KRAS, exist between adult and pediatric AML, including a number of mutations exclusive to pediatric disease.46  Focal deletions of ELF1, ZEB2, and MBNL1 genes have also been identified predominantly in pediatric cases.46  Defining the prognostic impact of these pediatric-specific lesions could ultimately help refine pediatric AML risk classification and point toward potential pharmacologically targetable lesions.46 

Consideration of combinations of mutations seems to more precisely define prognostic subgroups in AML rather than consideration of individual lesions in isolation.46  Evaluation of multiple cohorts of pediatric AML patients found that those with both an FLT3-ITD and concomitant NPM1 mutation had a particularly favorable outcome, despite numerous prior studies showing a negative impact of FLT3-ITD mutations alone in AML. Conversely, patients with an FLT3-ITD and either a WT1 or NUP98-NSD1 fusion had considerably worse outcomes than those patients with FLT3-ITD mutation alone.46  Thus, additional analysis of large, well-annotated pediatric cohorts could establish a pediatric-specific genomic classification system akin to those used for adult disease.47,72  Of course, the potential tradeoff for such precision is complexity. Pediatric AML in general is a rare disease, and if divided into small subgroups of genomically identified patients, the power to detect significant differences in outcome could be lost. Ultimately, perhaps the most practical means to adequately risk stratify pediatric AML patients will be integration of cytogenetic and molecular risk for group and MRD determination.73,74  This will allow for the further refinement of classification for patients with well-defined genetic classifiers and an opportunity to risk stratify the large group of patients lacking such classifiers.46 

Focused sequencing of specific AML subsets common in pediatric AML has identified distinct genomic subgroups with prognostic impact. Perhaps the best example of this is in acute megakaryoblastic leukemia (AMKL), a rare disease in adults (<1% adult AML75 ) but relatively common among pediatric patients (60%-90% of pediatric Down syndrome [DS] AML76,77  and 5%-7% of pediatric non-DS AML78,79 ). DS-AMKL has a favorable outcome, as most such patients are curable with relatively low-intensity chemotherapy.80-83  Conversely, AMKL in non-DS patients is often associated with a poor prognosis. RNA sequencing and exome sequencing of a large cohort of non-DS AMKL patients identified genetic subgroups with prognostic impact.84  Patients with CBFA2T3-GLIS2 fusions had a dismal prognosis, with an overall survival of 14%-16%, whereas remarkably, no patients with GATA1 mutations relapsed.84-86  Patients with KMT2A rearrangements and NUP98-KDM5A fusions also did poorly, whereas those with HOX gene rearrangements and RMB15-MKL1 fusions had an intermediate prognosis.84  It was recently discovered that CBFA2T3-GLIS2 fusions AMKL is characterized by the overexpression of a number of Hedgehog-related genes and that targeting of this pathway using inhibitors of the downstream effectors of the Hedgehog pathway, GLI, could be an effective therapeutic strategy for this extremely poor prognosis subset.87 

Although many disease-specific genetic lesions characterize pediatric hematologic malignancies, a handful of lesions occur across multiple disease types, and thus, effectively targeting such lesions could have a broad impact on childhood leukemia.

The Ras/MAPK pathway is perhaps the most commonly mutated pathway in pediatric hematologic malignancies and human cancer in general. Ras-activating mutations are the genetic hallmark of juvenile myelomonocytic leukemia,88  but they also commonly occur in subsets of lymphoid and other myeloid malignancies. Ras pathway mutations occur in a spectrum of B-ALL subtypes and are particularly common in near-haploid B-ALL in patients with DS and high-hyperdiploid ALL.89-92  Activating RAS mutations are enriched at B-ALL relapse, suggesting that they may confer resistance to standard chemotherapy.90,93  Furthermore, around 15% of pediatric T-ALL patients harbor Ras pathway mutations,39  and mutations of the Ras pathway are common in pediatric AML, particularly in children <3 years of age.46  Although potentially promising, targeting Ras has proven difficult. Initially believed to be “undruggable,” direct Ras-targeting small molecules have recently shown promising preclinical activity.94,95  Most recent efforts have instead focused on targeting the downstream MAPK and PI3K signaling pathways. MEK and PI3K inhibitors are in various stages of clinical development for relapsed/refractory pediatric leukemias.

Rearrangements of KMT2A also occur across a spectrum of pediatric hematologic malignancies, including over 75% of infant ALL. KMT2A-r infant ALL is associated with a dismal outcome.6 KMT2A rearrangements have also been associated with a poor prognosis in noninfant B-ALL,7,96  T-ALL,91  and AML49,84  and are common in the poor prognosis mixed phenotype acute leukemia and therapy-related AML.97  Thus, effective targeted therapy for KMT2A-rearranged leukemia is a critical need. Global gene expression analysis of KMT2A-r ALL identified overexpression of FLT3, and preclinical work suggested potential efficacy of FLT3 inhibition in KMT2A-r ALL98 ; however, a trial combining the FLT3 inhibitor lestaurtinib failed to show a survival benefit in infant KMT2A-rearranged ALL.99 KMT2A-r leukemia is characterized by potentially targetable epigenomic aberrations as well. The histone methyltransferase, DOT1L, was shown to be critical to KMT2A-r leukemias, and preclinical data of small molecule inhibitors of DOT1L generated tremendous enthusiasm.100,101  However, a phase 1 study of the DOT1L inhibitor, pinometostat (EPZ5676), showed good tolerability but failed to show an efficacy signal in pediatric KMT2A-r leukemia patients.102  Additional strategies targeting the epigenetic aberrations that underlie KMT2A-r leukemia are being investigated, including agents targeting the menin-KMT2A interaction,103  the arginine methyltransferase PMRT5,104  and the histone demethylase LSD1.105 KMT2A-r leukemias are also characterized by aberrant DNA hypermethylation; thus, incorporation of the hypomethylating agent azacitidine into standard chemotherapy is being explored in a clinical trial of infant KMT2A-r leukemia (NCT02828358).

Genomic studies of matched diagnostic, remission, and relapse samples have allowed for exploration of the clonal dynamics of pediatric leukemias. At diagnosis, there is significant genetic heterogeneity with clonal evolution throughout disease progression. These important studies have identified specific genetic lesions that are specific to or enriched at relapse. Such lesions likely confer resistance to chemotherapy, allowing for the survival of a subclone that then expands and acquires new mutations to drive disease relapse.

Mutations of the gene, NT5C2, are among the most commonly occurring relapse-specific mutations in ALL. NT5C2 is a 5′-nucleotidase important in maintaining intracellular nucleotide levels. The mutations occurring in ALL are gain of function mutations, producing an activated NT5C2 protein that leads to 6-mercaptopurine (6-MP) and 6-thioguanine resistance through the inactivation of the purine nucleoside analogs.106 NT5C2 mutations are frequent in both relapsed B-ALL and T-ALL, with all patients harboring such mutations relapsing within 36 months of diagnosis.107,108  Screening for the emergence of NT5C2 mutations could identify patients who would benefit from an altered therapeutic approach, such as the introduction of additional noncross-reactive chemotherapeutic agents during maintenance therapy.109  Additionally, targeted therapeutic intervention may be possible, with inhibitors of NT5C2 being explored and data to support the use of inosine-5′-monophosphate dehydrogenase inhibition to mitigate the chemoresistance induced by NT5C2 mutations.106,110 

Additional relapse-enriched mutations in ALL have been identified, and the mechanisms by which they emerge under the selective pressure of chemotherapy are being elucidated. For example, mutations of the epigenetic regulator CREBBP are common at relapse in pediatric ALL.90,93,111,112 CREBBP encodes histone acetyltransferase CREB binding protein, a transcriptional coactivator. The genetic lesions that occur in ALL are loss of function mutations or deletions, which lead to aberrant transcriptional regulation of CREBBP targets, including glucocorticoid response elements. Thus, these mutations likely contribute to relapse via resistance to glucocorticoid therapy.90,112  Mutations of a number of additional epigenetic regulators are highly enriched at relapse in ALL, including SETD2, WHSC1, KMT2D, EP300, KDM6A, and MSH6, strongly implicating epigenetic dysregulation as a key mediator of resistance to chemotherapy.93,111,113  The frequency of such mutations also points toward the possibility of incorporating epigenetically targeted agents as a means to overcome chemoresistance in ALL. Additionally, deletions and mutation of IKZF1 are enriched at relapse along with activating Ras pathway mutations.90,93,111 

Likewise, in AML, selective pressure of chemotherapy leads to clonal evolution.114-117  Elegant work in adult AML revealed that mutations in genes involved in regulation of global chromatin structure, such as DNA methylation regulators, histone modifiers, and chromatin looping, arise in preleukemic stem/progenitor cells with malignant transformation after the acquisition of additional cooperative mutations. These preleukemic stem cells often survive chemotherapy and serve as reservoirs for relapse.114  The evolution of pediatric AML is likely different, because the founding epigenetic modifier mutations common in adult AML are rare in pediatric disease. However, sequencing studies of relapsed pediatric disease have identified relapse-enriched mutations of epigenetic regulators, such as CREBBP, SETD2, and ASXL3, indicating a role for epigenetic aberration in the persistence of a relapse-initiating clone in pediatric AML as well.115,116  Additionally, signaling mutations, including activating PTEN, NRAS, and TKD mutations of FLT3, seem to be enriched at relapse in pediatric AML and may represent targetable lesions, although larger studies are warranted.116,117 

Pharmacogenomics

Tolerance of standard chemotherapeutic agents can vary substantially. Recent work has uncovered a number of genetic variants in pediatric leukemia patients associated with poor tolerance of specific chemotherapies and increased risk for certain chemotherapy-induced toxicities. Such studies lay the groundwork for individualized administration of chemotherapeutic agents.

The antimetabolite 6-MP is a critical component of therapy for ALL, but vast interindividual tolerance exists. Polymorphisms of the thiopurine metabolism gene, thiopurine S-methyltransferase (TMPT), are a well-established risk factor for 6-MP toxicity,118  and more recently, identified coding variants of the nudix hydrolase 15 (NUDT15) gene are also highly associated with increased 6-MP–induced hematologic toxicity.119  It is now generally accepted routine practice to screen all ALL patients for the TMPT genotype, and newer guidelines have been established to screen individual in the highest-risk ethnic groups for NUDT15 toxicity–associated variants and dose adjust 6-MP accordingly.119  This type of individualized dosing should reduce toxicity and minimize the associated delays in therapy.

Germ-line polymorphisms associated with asparaginase toxicities, vincristine neuropathy, and corticosteroid-induced osteonecrosis have also been identified through recent genome-wide association studies (GWAS) of pediatric leukemia patients.120-123  The clinical application of these polymorphisms is less well defined than for the 6-MP metabolism–altering variants. However, one can envision that the prospective identification of these risk alleles could be used to identify patients in need of closer monitoring for specific toxicities and tailor enhanced supportive care measures in individuals with toxicity-associated polymorphisms.

Genetic susceptibility

Although cancer predisposition syndromes are thought to be rare underlying causes of childhood hematologic malignancies, recent studies have identified some important exceptions. Sequencing of paired tumor and germ-line material from pediatric patients with hypodiploid B-ALL identified mutation of TP53 in over 90% of B-ALL patients with low hypodiploid (32-39 chromosomes), 50% of which were present in the germ line.89  This seminal finding has important clinical and potential ethical implications. As a high-risk subset of B-ALL, such patients are frequently offered HSCT in first CR. The presence of a familial germ-line TP53 mutation in a matched sibling would make them an unsuitable donor, necessitating screening of siblings. Additionally, given the high risk of cancer development in patients with germ-line TP53 mutations, surveillance guidelines exist. Therefore, screening of other family members is advisable. Additionally, rare germ-line mutations in genes, such as ETV6 and PAX5, have been identified in familial as well as sporadic B-ALL.124-127  Similarly, germ-line GATA2 mutations have recently been identified in a high percentage of adolescent patients with myelodysplastic syndrome (MDS) with monosomy 7.128  Screening of potential sibling donors and other family members would also need to be discussed with the families of affected patients. Additional MDS/AML predisposition germ-line mutations are also well described, including ANKRD26, CEBPA, DDX41, ETV6, RUNX1, SRP72, SAMD9, and SAMD9L genes.129  Thus, kindred with histories of multiple family members with childhood- and young adult–onset hematologic malignancies should be referred for genetic screening.130,131  This raises a number of potential issues for affected families, including implications for the genetic testing and medical care of siblings and parents, the psychological impact of cancer surveillance, and reproductive planning regarding risk for future offspring. Additionally, families may feel pressure to contact estranged relatives, and genetic test results may reveal the presence of the mutation in a family member who wishes not to know their status. Furthermore, in the event of a patient’s death, reporting of results is complex.132  Thus, in addition to the medical challenges that they are facing, genetic testing potentially raises social, psychological, and ethical issues for affected families.124-127,131 

In addition to these bona fide cancer predisposition syndromes, a number of recent GWAS of childhood leukemia have identified a number of germ-line genetic variants highly associated with risk of pediatric ALL, including common variants in genes such as ARID5B, CEBPE, and IKZF1. Additionally, GATA3 germ-line variants are specifically associated with adolescent and young adult (AYA) Ph-like and non–Ph-like B-ALL.133,134  These studies have dramatically expanded our view of the biology of ALL development but also raise important ethical questions. Are there patients who should be screened for these germ-line variants, particularly subsets strongly linked to a particular risk allele, like AYA patients with Ph-like disease? Patients and their parents often ask if other family members are at risk or if the risk of leukemia development is something that they may pass on to their children. We now know that, in a portion of cases, potentially heritable risk does exist; therefore, should we seek to identify families carrying these risk alleles? Given the lack of certainty, routine screening of patients and families is not currently recommended, but with more genomic data emerging, these questions must be considered. Referral to a cancer genetics clinic should be considered for families with multiple cases of childhood leukemia.130,131 

In conclusion, genetic profiling of pediatric hematologic malignancies has led to improved risk stratification and identified a number of potentially targetable genetic vulnerabilities. Such efforts have translated into improved outcomes in specific subsets of patients (eg, Ph+ B-ALL patients treated with TKIs plus chemotherapy), with great hope for similar improvements in others (eg, Ph-like B-ALL). Interrogating the germ line of patients and unaffected controls has identified risk alleles for the development of pediatric leukemias as well as variants associated with differing susceptibility to a number of chemotherapeutic agents. Although these successes rightly generate enthusiasm, much work remains. The prognostic impact of genetic lesions identified in retrospective cohorts must be validated in patients treated with modern era therapy and integrated into the existing risk stratification schemas. Identified potentially targetable lesions must be validated as true molecular dependencies, and the safety and efficacy of incorporating targeted therapies into standard chemotherapy backbones will need to be determined. Additionally, although therapeutic effect is ideally determined through the conduct of appropriately powered clinical trials, this will not be possible for potentially targetable genomic lesions that occur in rare subsets of patients. Thus, we will likely need to reconsider how we test for and define efficacy of specific targeted agents. The growing number of genetic variants associated with increased risk of pediatric hematologic malignancies raises clinical and ethical questions, which will need to be addressed by pediatric oncologists, geneticists, and genetic counselors. Although formidable, taken together, these challenges are worth tackling given the large potential payout of improved outcomes for pediatric leukemia patients.

Mignon L. Loh, Department of Pediatrics, Benioff Children’s Hospital and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, 1450 3rd St, San Francisco, CA 94158; e-mail: mignon.loh@ucsf.edu.

1.
Borowitz
MJ
,
Devidas
M
,
Hunger
SP
, et al
;
Children’s Oncology Group
.
Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia and its relationship to other prognostic factors: a Children’s Oncology Group study
.
Blood
.
2008
;
111
(
12
):
5477
-
5485
.
2.
Schultz
KR
,
Devidas
M
,
Bowman
WP
, et al
;
Children’s Oncology Group
.
Philadelphia chromosome-negative very high-risk acute lymphoblastic leukemia in children and adolescents: results from Children’s Oncology Group Study AALL0031
.
Leukemia
.
2014
;
28
(
4
):
964
-
967
.
3.
Mullighan
CG
,
Jeha
S
,
Pei
D
, et al
.
Outcome of children with hypodiploid ALL treated with risk-directed therapy based on MRD levels
.
Blood
.
2015
;
126
(
26
):
2896
-
2899
.
4.
McNeer
JL
,
Dai
Y
,
Larsen
E
, et al
.
Poor outcome for children, adolescents, and young adults with hypodiploid all is not improved by hematopoietic stem cell transplant: a report from the Children’s Oncology Group (COG)
.
Pediatr Blood Cancer
.
2018
;
65
(
suppl 1
):e27057.
5.
Borowitz
MJ
,
Pullen
DJ
,
Shuster
JJ
, et al
;
Children’s Oncology Group study
.
Minimal residual disease detection in childhood precursor-B-cell acute lymphoblastic leukemia: relation to other risk factors. A Children’s Oncology Group study
.
Leukemia
.
2003
;
17
(
8
):
1566
-
1572
.
6.
Hilden
JM
,
Dinndorf
PA
,
Meerbaum
SO
, et al
;
Children’s Oncology Group
.
Analysis of prognostic factors of acute lymphoblastic leukemia in infants: report on CCG 1953 from the Children’s Oncology Group
.
Blood
.
2006
;
108
(
2
):
441
-
451
.
7.
Loh
ML
,
Devidas
M
,
Dai
Y
, et al
.
Outcomes of children, adolescents, and young adults with acute lymphoblastic leukemia based on blast genotype at diagnosis: a report from the Children’s Oncology Group
.
Blood
.
2016
;
128
(
22
):451.
8.
Harrison
CJ
,
Moorman
AV
,
Schwab
C
, et al
;
Ponte di Legno International Workshop in Childhood Acute Lymphoblastic Leukemia
.
An international study of intrachromosomal amplification of chromosome 21 (iAMP21): cytogenetic characterization and outcome
.
Leukemia
.
2014
;
28
(
5
):
1015
-
1021
.
9.
Heerema
NA
,
Carroll
AJ
,
Devidas
M
, et al
.
Intrachromosomal amplification of chromosome 21 is associated with inferior outcomes in children with acute lymphoblastic leukemia treated in contemporary standard-risk children’s oncology group studies: a report from the children’s oncology group
.
J Clin Oncol
.
2013
;
31
(
27
):
3397
-
3402
.
10.
Fischer
U
,
Forster
M
,
Rinaldi
A
, et al
.
Genomics and drug profiling of fatal TCF3-HLF-positive acute lymphoblastic leukemia identifies recurrent mutation patterns and therapeutic options
.
Nat Genet
.
2015
;
47
(
9
):
1020
-
1029
.
11.
Liu
YF
,
Wang
BY
,
Zhang
WN
, et al
.
Genomic profiling of adult and pediatric B-cell acute lymphoblastic leukemia
.
EBioMedicine
.
2016
;
8
:
173
-
183
.
12.
Gu
Z
,
Churchman
M
,
Roberts
K
, et al
.
Genomic analyses identify recurrent MEF2D fusions in acute lymphoblastic leukaemia
.
Nat Commun
.
2016
;
7
:
13331
.
13.
Hirabayashi
S
,
Ohki
K
,
Nakabayashi
K
, et al
;
Tokyo Children’s Cancer Study Group (TCCSG)
.
ZNF384-related fusion genes define a subgroup of childhood B-cell precursor acute lymphoblastic leukemia with a characteristic immunotype
.
Haematologica
.
2017
;
102
(
1
):
118
-
129
.
14.
Qian
M
,
Zhang
H
,
Kham
SK
, et al
.
Whole-transcriptome sequencing identifies a distinct subtype of acute lymphoblastic leukemia with predominant genomic abnormalities of EP300 and CREBBP
.
Genome Res
.
2017
;
27
(
2
):
185
-
195
.
15.
Nowell
PC
.
Discovery of the Philadelphia chromosome: a personal perspective
.
J Clin Invest
.
2007
;
117
(
8
):
2033
-
2035
.
16.
Schultz
KR
,
Bowman
WP
,
Aledo
A
, et al
.
Improved early event-free survival with imatinib in Philadelphia chromosome-positive acute lymphoblastic leukemia: a children’s oncology group study
.
J Clin Oncol
.
2009
;
27
(
31
):
5175
-
5181
.
17.
Schultz
KR
,
Carroll
A
,
Heerema
NA
, et al
;
Children’s Oncology Group
.
Long-term follow-up of imatinib in pediatric Philadelphia chromosome-positive acute lymphoblastic leukemia: Children’s Oncology Group study AALL0031
.
Leukemia
.
2014
;
28
(
7
):
1467
-
1471
.
18.
Biondi
A
,
Schrappe
M
,
De Lorenzo
P
, et al
.
Imatinib after induction for treatment of children and adolescents with Philadelphia-chromosome-positive acute lymphoblastic leukaemia (EsPhALL): a randomised, open-label, intergroup study
.
Lancet Oncol
.
2012
;
13
(
9
):
936
-
945
.
19.
Den Boer
ML
,
van Slegtenhorst
M
,
De Menezes
RX
, et al
.
A subtype of childhood acute lymphoblastic leukaemia with poor treatment outcome: a genome-wide classification study
.
Lancet Oncol
.
2009
;
10
(
2
):
125
-
134
.
20.
Reshmi
SC
,
Harvey
RC
,
Roberts
KG
, et al
.
Targetable kinase gene fusions in high-risk B-ALL: a study from the Children’s Oncology Group
.
Blood
.
2017
;
129
(
25
):
3352
-
3361
.
21.
Roberts
KG
,
Li
Y
,
Payne-Turner
D
, et al
.
Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia
.
N Engl J Med
.
2014
;
371
(
11
):
1005
-
1015
.
22.
Tasian
SK
,
Loh
ML
,
Hunger
SP
.
Philadelphia chromosome-like acute lymphoblastic leukemia
.
Blood
.
2017
;
130
(
19
):
2064
-
2072
.
23.
Loh
ML
,
Zhang
J
,
Harvey
RC
, et al
.
Tyrosine kinome sequencing of pediatric acute lymphoblastic leukemia: a report from the Children’s Oncology Group TARGET Project
.
Blood
.
2013
;
121
(
3
):
485
-
488
.
24.
Roberts
KG
,
Morin
RD
,
Zhang
J
, et al
.
Genetic alterations activating kinase and cytokine receptor signaling in high-risk acute lymphoblastic leukemia
.
Cancer Cell
.
2012
;
22
(
2
):
153
-
166
.
25.
Boer
JM
,
Steeghs
EM
,
Marchante
JR
, et al
.
Tyrosine kinase fusion genes in pediatric BCR-ABL1-like acute lymphoblastic leukemia
.
Oncotarget
.
2017
;
8
(
3
):
4618
-
4628
.
26.
Harvey
RC
,
Mullighan
CG
,
Chen
IM
, et al
.
Rearrangement of CRLF2 is associated with mutation of JAK kinases, alteration of IKZF1, Hispanic/Latino ethnicity, and a poor outcome in pediatric B-progenitor acute lymphoblastic leukemia
.
Blood
.
2010
;
115
(
26
):
5312
-
5321
.
27.
Harvey
RC
,
Mullighan
CG
,
Wang
X
, et al
.
Identification of novel cluster groups in pediatric high-risk B-precursor acute lymphoblastic leukemia with gene expression profiling: correlation with genome-wide DNA copy number alterations, clinical characteristics, and outcome
.
Blood
.
2010
;
116
(
23
):
4874
-
4884
.
28.
Iacobucci
I
,
Li
Y
,
Roberts
KG
, et al
.
Truncating erythropoietin receptor rearrangements in acute lymphoblastic leukemia
.
Cancer Cell
.
2016
;
29
(
2
):
186
-
200
.
29.
Mullighan
CG
,
Goorha
S
,
Radtke
I
, et al
.
Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia
.
Nature
.
2007
;
446
(
7137
):
758
-
764
.
30.
Mullighan
CG
,
Su
X
,
Zhang
J
, et al
;
Children’s Oncology Group
.
Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia
.
N Engl J Med
.
2009
;
360
(
5
):
470
-
480
.
31.
Boer
JM
,
van der Veer
A
,
Rizopoulos
D
, et al
.
Prognostic value of rare IKZF1 deletion in childhood B-cell precursor acute lymphoblastic leukemia: an international collaborative study
.
Leukemia
.
2016
;
30
(
1
):
32
-
38
.
32.
Martinelli
G
,
Iacobucci
I
,
Storlazzi
CT
, et al
.
IKZF1 (Ikaros) deletions in BCR-ABL1-positive acute lymphoblastic leukemia are associated with short disease-free survival and high rate of cumulative incidence of relapse: a GIMEMA AL WP report
.
J Clin Oncol
.
2009
;
27
(
31
):
5202
-
5207
.
33.
Stanulla
M
,
Dagdan
E
,
Zaliova
M
, et al
;
International BFM Study Group
.
IKZF1plus defines a new minimal residual disease-dependent very-poor prognostic profile in pediatric B-cell precursor acute lymphoblastic leukemia
.
J Clin Oncol
.
2018
;
36
(
12
):
1240
-
1249
.
34.
Mullighan
CG
,
Miller
CB
,
Radtke
I
, et al
.
BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros
.
Nature
.
2008
;
453
(
7191
):
110
-
114
.
35.
Iacobucci
I
,
Storlazzi
CT
,
Cilloni
D
, et al
.
Identification and molecular characterization of recurrent genomic deletions on 7p12 in the IKZF1 gene in a large cohort of BCR-ABL1-positive acute lymphoblastic leukemia patients: on behalf of Gruppo Italiano Malattie Ematologiche dell’Adulto Acute Leukemia Working Party (GIMEMA AL WP)
.
Blood
.
2009
;
114
(
10
):
2159
-
2167
.
36.
Churchman
ML
,
Evans
K
,
Richmond
J
, et al
.
Synergism of FAK and tyrosine kinase inhibition in Ph+ B-ALL
.
JCI Insight
.
2016
;
1
(
4
):
e86082
.
37.
Churchman
ML
,
Low
J
,
Qu
C
, et al
.
Efficacy of Retinoids in IKZF1-Mutated BCR-ABL1 Acute Lymphoblastic Leukemia
.
Cancer Cell
.
2015
;
28
(
3
):
343
-
356
.
38.
Zhang
J
,
McCastlain
K
,
Yoshihara
H
, et al
;
St. Jude Children’s Research Hospital–Washington University Pediatric Cancer Genome Project
.
Deregulation of DUX4 and ERG in acute lymphoblastic leukemia
.
Nat Genet
.
2016
;
48
(
12
):
1481
-
1489
.
39.
Liu
Y
,
Easton
J
,
Shao
Y
, et al
.
The genomic landscape of pediatric and young adult T-lineage acute lymphoblastic leukemia
.
Nat Genet
.
2017
;
49
(
8
):
1211
-
1218
.
40.
Petit
A
,
Trinquand
A
,
Chevret
S
, et al
;
French Acute Lymphoblastic Leukemia Study Group (FRALLE)
.
Oncogenetic mutations combined with MRD improve outcome prediction in pediatric T-cell acute lymphoblastic leukemia
.
Blood
.
2018
;
131
(
3
):
289
-
300
.
41.
Sanchez-Martin
M
,
Ferrando
A
.
The NOTCH1-MYC highway toward T-cell acute lymphoblastic leukemia
.
Blood
.
2017
;
129
(
9
):
1124
-
1133
.
42.
Coustan-Smith
E
,
Mullighan
CG
,
Onciu
M
, et al
.
Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia
.
Lancet Oncol
.
2009
;
10
(
2
):
147
-
156
.
43.
Patrick
K
,
Wade
R
,
Goulden
N
, et al
.
Outcome for children and young people with Early T-cell precursor acute lymphoblastic leukaemia treated on a contemporary protocol, UKALL 2003
.
Br J Haematol
.
2014
;
166
(
3
):
421
-
424
.
44.
Conter
V
,
Valsecchi
MG
,
Buldini
B
, et al
.
Early T-cell precursor acute lymphoblastic leukaemia in children treated in AIEOP centres with AIEOP-BFM protocols: a retrospective analysis
.
Lancet Haematol
.
2016
;
3
(
2
):
e80
-
e86
.
45.
Wood
BL
,
Dunsmore
KP
,
Devidas
M
, et al
.
T-lymphoblastic leukemia (T-ALL) shows excellent outcome, lack of significance of the early thymic precursor (ETP) immunophenotype, and validation of the prognostic value of end-induction minimal residual disease (MRD) in Children’s Oncology Group (COG) study AALL0434
.
Blood
.
2014
;
124
(
21
):1.
46.
Bolouri
H
,
Farrar
JE
,
Triche
T
Jr
, et al
.
The molecular landscape of pediatric acute myeloid leukemia reveals recurrent structural alterations and age-specific mutational interactions [published correction appears in Nat Med. 2018;24(4):526]
.
Nat Med
.
2018
;
24
(
1
):
103
-
112
.
47.
Papaemmanuil
E
,
Gerstung
M
,
Bullinger
L
, et al
.
Genomic classification and prognosis in acute myeloid leukemia
.
N Engl J Med
.
2016
;
374
(
23
):
2209
-
2221
.
48.
Creutzig
U
,
Zimmermann
M
,
Reinhardt
D
, et al
.
Changes in cytogenetics and molecular genetics in acute myeloid leukemia from childhood to adult age groups
.
Cancer
.
2016
;
122
(
24
):
3821
-
3830
.
49.
Balgobind
BV
,
Raimondi
SC
,
Harbott
J
, et al
.
Novel prognostic subgroups in childhood 11q23/MLL-rearranged acute myeloid leukemia: results of an international retrospective study
.
Blood
.
2009
;
114
(
12
):
2489
-
2496
.
50.
Struski
S
,
Lagarde
S
,
Bories
P
, et al
.
NUP98 is rearranged in 3.8% of pediatric AML forming a clinical and molecular homogenous group with a poor prognosis
.
Leukemia
.
2017
;
31
(
3
):
565
-
572
.
51.
Bisio
V
,
Zampini
M
,
Tregnago
C
, et al
.
NUP98-fusion transcripts characterize different biological entities within acute myeloid leukemia: a report from the AIEOP-AML group
.
Leukemia
.
2017
;
31
(
4
):
974
-
977
.
52.
Hollink
IH
,
van den Heuvel-Eibrink
MM
,
Arentsen-Peters
ST
, et al
.
NUP98/NSD1 characterizes a novel poor prognostic group in acute myeloid leukemia with a distinct HOX gene expression pattern
.
Blood
.
2011
;
118
(
13
):
3645
-
3656
.
53.
Osumi
T
,
Tsujimoto
SI
,
Tamura
M
, et al
.
Recurrent RARB translocations in acute promyelocytic leukemia lacking RARA translocation
.
Cancer Res
.
2018
;
78
(
16
):
4452
-
4458
.
54.
Qiu
JJ
,
Zeisig
BB
,
Li
S
, et al
.
Critical role of retinoid/rexinoid signaling in mediating transformation and therapeutic response of NUP98-RARG leukemia
.
Leukemia
.
2015
;
29
(
5
):
1153
-
1162
.
55.
Lo-Coco
F
,
Avvisati
G
,
Vignetti
M
, et al
;
Study Alliance Leukemia
.
Retinoic acid and arsenic trioxide for acute promyelocytic leukemia
.
N Engl J Med
.
2013
;
369
(
2
):
111
-
121
.
56.
Meshinchi
S
,
Alonzo
TA
,
Stirewalt
DL
, et al
.
Clinical implications of FLT3 mutations in pediatric AML
.
Blood
.
2006
;
108
(
12
):
3654
-
3661
.
57.
Grunwald
MR
,
Levis
MJ
.
FLT3 tyrosine kinase inhibition as a paradigm for targeted drug development in acute myeloid leukemia
.
Semin Hematol
.
2015
;
52
(
3
):
193
-
199
.
58.
Levis
M
.
Midostaurin approved for FLT3-mutated AML
.
Blood
.
2017
;
129
(
26
):
3403
-
3406
.
59.
Stone
RM
,
Manley
PW
,
Larson
RA
,
Capdeville
R
.
Midostaurin: its odyssey from discovery to approval for treating acute myeloid leukemia and advanced systemic mastocytosis
.
Blood Adv
.
2018
;
2
(
4
):
444
-
453
.
60.
Grunwald
MR
,
Levis
MJ
.
FLT3 inhibitors for acute myeloid leukemia: a review of their efficacy and mechanisms of resistance
.
Int J Hematol
.
2013
;
97
(
6
):
683
-
694
.
61.
von Bubnoff
N
,
Engh
RA
,
Aberg
E
,
Sänger
J
,
Peschel
C
,
Duyster
J
.
FMS-like tyrosine kinase 3-internal tandem duplication tyrosine kinase inhibitors display a nonoverlapping profile of resistance mutations in vitro
.
Cancer Res
.
2009
;
69
(
7
):
3032
-
3041
.
62.
Galanis
A
,
Ma
H
,
Rajkhowa
T
, et al
.
Crenolanib is a potent inhibitor of FLT3 with activity against resistance-conferring point mutants
.
Blood
.
2014
;
123
(
1
):
94
-
100
.
63.
Mori
M
,
Kaneko
N
,
Ueno
Y
, et al
.
Gilteritinib, a FLT3/AXL inhibitor, shows antileukemic activity in mouse models of FLT3 mutated acute myeloid leukemia
.
Invest New Drugs
.
2017
;
35
(
5
):
556
-
565
.
64.
Tarlock
K
,
Hylkema
T
,
Ries
R
, et al
.
Discovery and functional validation of novel pediatric specific flt3 activating mutations in acute myeloid leukemia: results from the COG/NCI target initiative
.
Blood
.
2015
;
126
(
23
):
87
.
65.
Brown
P
,
McIntyre
E
,
Rau
R
, et al
.
The incidence and clinical significance of nucleophosmin mutations in childhood AML
.
Blood
.
2007
;
110
(
3
):
979
-
985
.
66.
Ho
PA
,
Alonzo
TA
,
Gerbing
RB
, et al
.
Prevalence and prognostic implications of CEBPA mutations in pediatric acute myeloid leukemia (AML): a report from the Children’s Oncology Group
.
Blood
.
2009
;
113
(
26
):
6558
-
6566
.
67.
Kühn
MW
,
Song
E
,
Feng
Z
, et al
.
Targeting chromatin regulators inhibits leukemogenic gene expression in NPM1 mutant leukemia
.
Cancer Discov
.
2016
;
6
(
10
):
1166
-
1181
.
68.
Falini
B
,
Brunetti
L
,
Martelli
MP
.
Dactinomycin in NPM1-mutated acute myeloid leukemia
.
N Engl J Med
.
2015
;
373
(
12
):
1180
-
1182
.
69.
Ho
PA
,
Zeng
R
,
Alonzo
TA
, et al
.
Prevalence and prognostic implications of WT1 mutations in pediatric acute myeloid leukemia (AML): a report from the Children’s Oncology Group
.
Blood
.
2010
;
116
(
5
):
702
-
710
.
70.
Ho
PA
,
Kutny
MA
,
Alonzo
TA
, et al
.
Leukemic mutations in the methylation-associated genes DNMT3A and IDH2 are rare events in pediatric AML: a report from the Children’s Oncology Group
.
Pediatr Blood Cancer
.
2011
;
57
(
2
):
204
-
209
.
71.
Shih
AH
,
Abdel-Wahab
O
,
Patel
JP
,
Levine
RL
.
The role of mutations in epigenetic regulators in myeloid malignancies
.
Nat Rev Cancer
.
2012
;
12
(
9
):
599
-
612
.
72.
Döhner
H
,
Estey
E
,
Grimwade
D
, et al
.
Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel
.
Blood
.
2017
;
129
(
4
):
424
-
447
.
73.
Buldini
B
,
Rizzati
F
,
Masetti
R
, et al
.
Prognostic significance of flow-cytometry evaluation of minimal residual disease in children with acute myeloid leukaemia treated according to the AIEOP-AML 2002/01 study protocol
.
Br J Haematol
.
2017
;
177
(
1
):
116
-
126
.
74.
Rubnitz
JE
,
Inaba
H
,
Dahl
G
, et al
.
Minimal residual disease-directed therapy for childhood acute myeloid leukaemia: results of the AML02 multicentre trial
.
Lancet Oncol
.
2010
;
11
(
6
):
543
-
552
.
75.
Pagano
L
,
Pulsoni
A
,
Vignetti
M
, et al
;
GIMEMA
.
Acute megakaryoblastic leukemia: experience of GIMEMA trials
.
Leukemia
.
2002
;
16
(
9
):
1622
-
1626
.
76.
Taub
JW
,
Berman
JN
,
Hitzler
JK
, et al
.
Improved outcomes for myeloid leukemia of Down syndrome: a report from the Children’s Oncology Group AAML0431 trial
.
Blood
.
2017
;
129
(
25
):
3304
-
3313
.
77.
Uffmann
M
,
Rasche
M
,
Zimmermann
M
, et al
.
Therapy reduction in patients with Down syndrome and myeloid leukemia: the international ML-DS 2006 trial
.
Blood
.
2017
;
129
(
25
):
3314
-
3321
.
78.
Gamis
AS
,
Alonzo
TA
,
Meshinchi
S
, et al
.
Gemtuzumab ozogamicin in children and adolescents with de novo acute myeloid leukemia improves event-free survival by reducing relapse risk: results from the randomized phase III Children’s Oncology Group trial AAML0531
.
J Clin Oncol
.
2014
;
32
(
27
):
3021
-
3032
.
79.
Schweitzer
J
,
Zimmermann
M
,
Rasche
M
, et al
.
Improved outcome of pediatric patients with acute megakaryoblastic leukemia in the AML-BFM 04 trial
.
Ann Hematol
.
2015
;
94
(
8
):
1327
-
1336
.
80.
Creutzig
U
,
Reinhardt
D
,
Diekamp
S
,
Dworzak
M
,
Stary
J
,
Zimmermann
M
.
AML patients with Down syndrome have a high cure rate with AML-BFM therapy with reduced dose intensity
.
Leukemia
.
2005
;
19
(
8
):
1355
-
1360
.
81.
Gamis
AS
.
Acute myeloid leukemia and Down syndrome evolution of modern therapy–state of the art review
.
Pediatr Blood Cancer
.
2005
;
44
(
1
):
13
-
20
.
82.
Hitzler
JK
,
Zipursky
A
.
Origins of leukaemia in children with Down syndrome
.
Nat Rev Cancer
.
2005
;
5
(
1
):
11
-
20
.
83.
Rao
A
,
Hills
RK
,
Stiller
C
, et al
.
Treatment for myeloid leukaemia of Down syndrome: population-based experience in the UK and results from the Medical Research Council AML 10 and AML 12 trials
.
Br J Haematol
.
2006
;
132
(
5
):
576
-
583
.
84.
de Rooij
JD
,
Branstetter
C
,
Ma
J
, et al
.
Pediatric non-Down syndrome acute megakaryoblastic leukemia is characterized by distinct genomic subsets with varying outcomes
.
Nat Genet
.
2017
;
49
(
3
):
451
-
456
.
85.
Hara
Y
,
Shiba
N
,
Ohki
K
, et al
.
Prognostic impact of specific molecular profiles in pediatric acute megakaryoblastic leukemia in non-Down syndrome
.
Genes Chromosomes Cancer
.
2017
;
56
(
5
):
394
-
404
.
86.
de Rooij
JD
,
Masetti
R
,
van den Heuvel-Eibrink
MM
, et al
.
Recurrent abnormalities can be used for risk group stratification in pediatric AMKL: a retrospective intergroup study
.
Blood
.
2016
;
127
(
26
):
3424
-
3430
.
87.
Masetti
R
,
Bertuccio
SN
,
Astolfi
A
, et al
.
Hh/Gli antagonist in acute myeloid leukemia with CBFA2T3-GLIS2 fusion gene
.
J Hematol Oncol
.
2017
;
10
(
1
):
26
.
88.
Stieglitz
E
,
Taylor-Weiner
AN
,
Chang
TY
, et al
.
The genomic landscape of juvenile myelomonocytic leukemia [published correction appears in Nat Genet. 2015;47(11):1333]
.
Nat Genet
.
2015
;
47
(
11
):
1326
-
1333
.
89.
Holmfeldt
L
,
Wei
L
,
Diaz-Flores
E
, et al
.
The genomic landscape of hypodiploid acute lymphoblastic leukemia
.
Nat Genet
.
2013
;
45
(
3
):
242
-
252
.
90.
Malinowska-Ozdowy
K
,
Frech
C
,
Schönegger
A
, et al
.
KRAS and CREBBP mutations: a relapse-linked malicious liaison in childhood high hyperdiploid acute lymphoblastic leukemia
.
Leukemia
.
2015
;
29
(
8
):
1656
-
1667
.
91.
Matlawska-Wasowska
K
,
Kang
H
,
Devidas
M
, et al
.
MLL rearrangements impact outcome in HOXA-deregulated T-lineage acute lymphoblastic leukemia: a Children’s Oncology Group Study
.
Leukemia
.
2016
;
30
(
9
):
1909
-
1912
.
92.
Nikolaev
SI
,
Garieri
M
,
Santoni
F
, et al
.
Frequent cases of RAS-mutated Down syndrome acute lymphoblastic leukaemia lack JAK2 mutations
.
Nat Commun
.
2014
;
5
(
1
):
4654
.
93.
Ma
X
,
Edmonson
M
,
Yergeau
D
, et al
.
Rise and fall of subclones from diagnosis to relapse in pediatric B-acute lymphoblastic leukaemia
.
Nat Commun
.
2015
;
6
(
1
):
6604
.
94.
Ostrem
JM
,
Shokat
KM
.
Direct small-molecule inhibitors of KRAS: from structural insights to mechanism-based design
.
Nat Rev Drug Discov
.
2016
;
15
(
11
):
771
-
785
.
95.
Welsch
ME
,
Kaplan
A
,
Chambers
JM
, et al
.
Multivalent small-molecule pan-RAS inhibitors
.
Cell
.
2017
;
168
(
5
):
878
-
889.e29
.
96.
Moorman
AV
,
Ensor
HM
,
Richards
SM
, et al
.
Prognostic effect of chromosomal abnormalities in childhood B-cell precursor acute lymphoblastic leukaemia: results from the UK Medical Research Council ALL97/99 randomised trial
.
Lancet Oncol
.
2010
;
11
(
5
):
429
-
438
.
97.
Winters
AC
,
Bernt
KM
.
MLL-rearranged leukemias—an update on science and clinical approaches
.
Front Pediatr
.
2017
;
5
:
4
.
98.
Brown
P
,
Levis
M
,
McIntyre
E
,
Griesemer
M
,
Small
D
.
Combinations of the FLT3 inhibitor CEP-701 and chemotherapy synergistically kill infant and childhood MLL-rearranged ALL cells in a sequence-dependent manner
.
Leukemia
.
2006
;
20
(
8
):
1368
-
1376
.
99.
Brown
P
,
Kairalla
J
,
Wang
C
, et al
.
Addition of FLT3 inhibitor lestaurtinib to post-induction chemotherapy does not improve outcomes in MLL-rearranged infant acute lymphoblastic leukemia (ALL): AALL0631, A Children’s Oncology Group Study
.
Pediatr Blood Cancer
.
2016
;
63
(
suppl 3
):
S7
.
100.
Bernt
KM
,
Zhu
N
,
Sinha
AU
, et al
.
MLL-rearranged leukemia is dependent on aberrant H3K79 methylation by DOT1L
.
Cancer Cell
.
2011
;
20
(
1
):
66
-
78
.
101.
Daigle
SR
,
Olhava
EJ
,
Therkelsen
CA
, et al
.
Potent inhibition of DOT1L as treatment of MLL-fusion leukemia
.
Blood
.
2013
;
122
(
6
):
1017
-
1025
.
102.
Neerav Shukla
CW
,
O’Brien
MM
,
Silverman
LB
, et al
.
Final report of phase 1 study of the DOT1L inhibitor, pinometostat (EPZ-5676), in children with relapsed or refractory MLL-r acute leukemia
.
Blood
.
2016
;
128
(
22
):
2780
.
103.
He
S
,
Malik
B
,
Borkin
D
, et al
.
Menin-MLL inhibitors block oncogenic transformation by MLL-fusion proteins in a fusion partner-independent manner
.
Leukemia
.
2016
;
30
(
2
):
508
-
513
.
104.
Kaushik
S
,
Liu
F
,
Veazey
KJ
, et al
.
Genetic deletion or small-molecule inhibition of the arginine methyltransferase PRMT5 exhibit anti-tumoral activity in mouse models of MLL-rearranged AML
.
Leukemia
.
2018
;
32
(
2
):
499
-
509
.
105.
Feng
Z
,
Yao
Y
,
Zhou
C
, et al
.
Pharmacological inhibition of LSD1 for the treatment of MLL-rearranged leukemia
.
J Hematol Oncol
.
2016
;
9
(
1
):
24
.
106.
Tzoneva
G
,
Dieck
CL
,
Oshima
K
, et al
.
Clonal evolution mechanisms in NT5C2 mutant-relapsed acute lymphoblastic leukaemia
.
Nature
.
2018
;
553
(
7689
):
511
-
514
.
107.
Meyer
JA
,
Wang
J
,
Hogan
LE
, et al
.
Relapse-specific mutations in NT5C2 in childhood acute lymphoblastic leukemia
.
Nat Genet
.
2013
;
45
(
3
):
290
-
294
.
108.
Tzoneva
G
,
Perez-Garcia
A
,
Carpenter
Z
, et al
.
Activating mutations in the NT5C2 nucleotidase gene drive chemotherapy resistance in relapsed ALL
.
Nat Med
.
2013
;
19
(
3
):
368
-
371
.
109.
Meyer
JA
,
Carroll
WL
,
Bhatla
T
.
Screening for gene mutations: will identification of NT5C2 mutations help predict the chance of relapse in acute lymphoblastic leukemia?
Expert Rev Hematol
.
2013
;
6
(
3
):
223
-
224
.
110.
Jordheim
LP
,
Marton
Z
,
Rhimi
M
, et al
.
Identification and characterization of inhibitors of cytoplasmic 5′-nucleotidase cN-II issued from virtual screening
.
Biochem Pharmacol
.
2013
;
85
(
4
):
497
-
506
.
111.
Ding
LW
,
Sun
QY
,
Tan
KT
, et al
.
Mutational landscape of pediatric acute lymphoblastic leukemia [published correction appears in Cancer Res. 2017;77(8):2174]
.
Cancer Res
.
2017
;
77
(
2
):
390
-
400
.
112.
Mullighan
CG
,
Zhang
J
,
Kasper
LH
, et al
.
CREBBP mutations in relapsed acute lymphoblastic leukaemia
.
Nature
.
2011
;
471
(
7337
):
235
-
239
.
113.
Mar
BG
,
Bullinger
LB
,
McLean
KM
, et al
.
Mutations in epigenetic regulators including SETD2 are gained during relapse in paediatric acute lymphoblastic leukaemia
.
Nat Commun
.
2014
;
5
(
1
):
3469
.
114.
Corces-Zimmerman
MR
,
Hong
WJ
,
Weissman
IL
,
Medeiros
BC
,
Majeti
R
.
Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission
.
Proc Natl Acad Sci USA
.
2014
;
111
(
7
):
2548
-
2553
.
115.
Farrar
JE
,
Schuback
HL
,
Ries
RE
, et al
.
Genomic profiling of pediatric acute myeloid leukemia reveals a changing mutational landscape from disease diagnosis to relapse
.
Cancer Res
.
2016
;
76
(
8
):
2197
-
2205
.
116.
Masetti
R
,
Castelli
I
,
Astolfi
A
, et al
.
Genomic complexity and dynamics of clonal evolution in childhood acute myeloid leukemia studied with whole-exome sequencing
.
Oncotarget
.
2016
;
7
(
35
):
56746
-
56757
.
117.
Shiba
N
,
Yoshida
K
,
Shiraishi
Y
, et al
.
Whole-exome sequencing reveals the spectrum of gene mutations and the clonal evolution patterns in paediatric acute myeloid leukaemia
.
Br J Haematol
.
2016
;
175
(
3
):
476
-
489
.
118.
Relling
MV
,
Hancock
ML
,
Rivera
GK
, et al
.
Mercaptopurine therapy intolerance and heterozygosity at the thiopurine S-methyltransferase gene locus
.
J Natl Cancer Inst
.
1999
;
91
(
23
):
2001
-
2008
.
119.
Yang
JJ
,
Landier
W
,
Yang
W
, et al
.
Inherited NUDT15 variant is a genetic determinant of mercaptopurine intolerance in children with acute lymphoblastic leukemia
.
J Clin Oncol
.
2015
;
33
(
11
):
1235
-
1242
.
120.
Abaji
R
,
Gagné
V
,
Xu
CJ
, et al
.
Whole-exome sequencing identified genetic risk factors for asparaginase-related complications in childhood ALL patients
.
Oncotarget
.
2017
;
8
(
27
):
43752
-
43767
.
121.
Fernandez
CA
,
Smith
C
,
Yang
W
, et al
.
Genome-wide analysis links NFATC2 with asparaginase hypersensitivity
.
Blood
.
2015
;
126
(
1
):
69
-
75
.
122.
Relling
MV
,
Yang
W
,
Das
S
, et al
.
Pharmacogenetic risk factors for osteonecrosis of the hip among children with leukemia
.
J Clin Oncol
.
2004
;
22
(
19
):
3930
-
3936
.
123.
Lee
SHR
,
Yang
JJ
.
Pharmacogenomics in acute lymphoblastic leukemia
.
Best Pract Res Clin Haematol
.
2017
;
30
(
3
):
229
-
236
.
124.
Churchman
ML
,
Qian
M
,
Te Kronnie
G
, et al
.
Germline genetic IKZF1 variation and predisposition to childhood acute lymphoblastic leukemia
.
Cancer Cell
.
2018
;
33
(
5
):
937
-
948.e8
.
125.
Moriyama
T
,
Metzger
ML
,
Wu
G
, et al
.
Germline genetic variation in ETV6 and risk of childhood acute lymphoblastic leukaemia: a systematic genetic study
.
Lancet Oncol
.
2015
;
16
(
16
):
1659
-
1666
.
126.
Qian
M
,
Cao
X
,
Devidas
M
, et al
.
TP53 germline variations influence the predisposition and prognosis of B-cell acute lymphoblastic leukemia in children
.
J Clin Oncol
.
2018
;
36
(
6
):
591
-
599
.
127.
Treviño
LR
,
Yang
W
,
French
D
, et al
.
Germline genomic variants associated with childhood acute lymphoblastic leukemia
.
Nat Genet
.
2009
;
41
(
9
):
1001
-
1005
.
128.
Wlodarski
MW
,
Hirabayashi
S
,
Pastor
V
, et al
;
EWOG-MDS
.
Prevalence, clinical characteristics, and prognosis of GATA2-related myelodysplastic syndromes in children and adolescents
.
Blood
.
2016
;
127
(
11
):
1387
-
1397
.
129.
Babushok
DV
,
Bessler
M
,
Olson
TS
.
Genetic predisposition to myelodysplastic syndrome and acute myeloid leukemia in children and young adults
.
Leuk Lymphoma
.
2016
;
57
(
3
):
520
-
536
.
130.
Porter
CC
,
Druley
TE
,
Erez
A
, et al
.
Recommendations for surveillance for children with leukemia-predisposing conditions
.
Clin Cancer Res
.
2017
;
23
(
11
):
e14
-
e22
.
131.
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
.
132.
Scollon
S
,
Bergstrom
K
,
McCullough
LB
, et al
.
Pediatric cancer genetics research and an evolving preventive ethics approach for return of results after death of the subject
.
J Law Med Ethics
.
2015
;
43
(
3
):
529
-
537
.
133.
Perez-Andreu
V
,
Roberts
KG
,
Harvey
RC
, et al
.
Inherited GATA3 variants are associated with Ph-like childhood acute lymphoblastic leukemia and risk of relapse
.
Nat Genet
.
2013
;
45
(
12
):
1494
-
1498
.
134.
Perez-Andreu
V
,
Roberts
KG
,
Xu
H
, et al
.
A genome-wide association study of susceptibility to acute lymphoblastic leukemia in adolescents and young adults
.
Blood
.
2015
;
125
(
4
):
680
-
686
.

Competing Interests

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Author notes

Off-label drug use: None disclosed.