Leukemia in infants is rare but generates tremendous interest due to its aggressive clinical presentation in a uniquely vulnerable host, its poor response to current therapies, and its fascinating biology. Increasingly, these biological insights are pointing the way toward novel therapeutic approaches. Using representative clinical case presentations, we review the key clinical, pathologic, and epidemiologic features of infant leukemia, including the high frequency of KMT2A gene rearrangements. We describe the current approach to risk-stratified treatment of infant leukemia in the major international cooperative groups. We highlight recent discoveries that elucidate the molecular biology of infant leukemia and suggest novel targeted therapeutic strategies, including modulation of aberrant epigenetic programs, inhibition of signaling pathways, and immunotherapeutics. Finally, we underscore the need for increased global collaboration to translate these discoveries into improved outcomes.

Patient 1

A 4-week-old boy was admitted to the hospital with fever, feeding problems, and lethargy. Physical examination revealed hepatosplenomegaly, pallor, and petechiae. Laboratory examination showed striking leukocytosis (white blood cells [WBCs], 1100 × 109/L), anemia (hemoglobin [Hb], 3.1 g/dL), and thrombocytopenia (platelets [PLTs], 10 × 109/L]. Bone marrow examination showed 98% blast cells with a very immature B-cell phenotype (CD19+, CD10, cytoplasmic immunoglobulin-negative, CD20) and a KMT2A-MLLT1 gene fusion (Table 1). Cerebrospinal fluid (CSF) examination showed WBCs at 38 × 109/L, and cytospin showed lymphoblasts morphologically. He was enrolled in the Interfant-06 protocol (clinicaltrials.gov NCT00550992). During induction (Figure 1), he had febrile neutropenia and received broad-spectrum empiric antibiotics. He achieved morphological remission after induction.

Table 1.

Nomenclature of infant leukemia genetics

Gene nameAliasChromosomeTranslocationFusion proteinAlias
KMT2A MLL 11q23 t(v;11q23) Variable  
Fusion partners (selected)      
 AFF1 AF4 4q21 t(4;11)(q21;q23) KMT2A-AFF1 MLL-AF4 
 MLLT1 ENL 19p13.3 t(11;19)(q23;p13.3) KMT2A-MLLT1 MLL-ENL 
 MLLT3 AF9 9p21 t(9;11)(p21;q23) KMT2A-MLLT3 MLL-AF9 
 MLLT10 AF10 10p12 t(10;11)(p12;q23) KMT2A-MLLT10 MLL-AF10 
 ELL — 19p13.1 t(11;19)(q23;p13.1) KMT2A-ELL MLL-ELL 
Gene nameAliasChromosomeTranslocationFusion proteinAlias
KMT2A MLL 11q23 t(v;11q23) Variable  
Fusion partners (selected)      
 AFF1 AF4 4q21 t(4;11)(q21;q23) KMT2A-AFF1 MLL-AF4 
 MLLT1 ENL 19p13.3 t(11;19)(q23;p13.3) KMT2A-MLLT1 MLL-ENL 
 MLLT3 AF9 9p21 t(9;11)(p21;q23) KMT2A-MLLT3 MLL-AF9 
 MLLT10 AF10 10p12 t(10;11)(p12;q23) KMT2A-MLLT10 MLL-AF10 
 ELL — 19p13.1 t(11;19)(q23;p13.1) KMT2A-ELL MLL-ELL 

—, no alias; v, variable chromosome.

Figure 1.

Interfant induction, now used by all 3 cooperative groups (Interfant, COG, and JPLSG). ARA-C, cytarabine; BMP, bone marrow procedure; DEXA, dexamethasone; DNR, daunorubicin; ITH, intrathecal; MTX, methotrexate; PRED, prednisone; VCR, vincristine.

Figure 1.

Interfant induction, now used by all 3 cooperative groups (Interfant, COG, and JPLSG). ARA-C, cytarabine; BMP, bone marrow procedure; DEXA, dexamethasone; DNR, daunorubicin; ITH, intrathecal; MTX, methotrexate; PRED, prednisone; VCR, vincristine.

Close modal

Patient 2

A 4-month-old boy was admitted to the hospital because he was not feeding well and had pale gray skin. Physical examination revealed pallor and hepatosplenomegaly. Laboratory examination showed leukocytosis (WBCs, 250 × 109/L), anemia (Hb, 5.5 g/dL), and thrombocytopenia (PLTs, 28 × 109/L). Bone marrow examination showed 99% blast cells with very immature B-cell phenotype (CD19+, CD10, cytoplasmic immunoglobulin-negative, CD20) and a KMT2A-AFF1 gene fusion. CSF examination showed WBCs at 3 × 109/L, and cytospin showed lymphoblasts morphologically. He was enrolled in the Children’s Oncology Group (COG) protocol AALL0631 (clinicaltrials.gov NCT00557193). During induction, he had febrile neutropenia and received broad-spectrum empiric antibiotics. He achieved morphological remission after induction.

Patient 3

A 10-month-old girl was admitted to the hospital with irritability, fever, poor appetite, and rash. Physical examination revealed petechiae, pallor, and tenderness to palpation of the extremities. Laboratory examination showed leukopenia and neutropenia (WBCs, 1.2 × 109/L with 30% neutrophils), anemia (Hb, 6.7 g/dL), and thrombocytopenia (PLTs, 47 × 109/L). Bone marrow examination showed 99% blast cells with immature B-cell phenotype (CD19+, CD10+, cytoplasmic immunoglobulin-negative, CD20) and normal cytogenetics. CSF examination showed WBCs at 1 × 109/L, and cytospin showed no lymphoblasts morphologically. She was enrolled in the Interfant-06 protocol. She had no significant complications during induction, and achieved morphological remission after induction.

Patient 4

A 7-month-old girl was admitted to the hospital with a progressive rash, fever, and poor feeding. Physical examination revealed scattered palpable blue cutaneous nodules and hepatosplenomegaly. Laboratory examination showed leukocytosis (WBCs, 42 × 109/L), anemia (Hb, 5.9 g/dL), and thrombocytopenia (PLTs, 28 × 109/L). Bone marrow examination showed 78% blast cells with a monoblastic phenotype (MPO+, CD34, HLA-DR+, CD33+, CD11b+) and KMT2A-MLLT3 fusion. CSF examination showed WBCs at 22 × 109/L, and cytospin showed monoblasts morphologically. Skin biopsy showed leukemia cutis. She was enrolled in the COG AAML1031 protocol (clinicaltrials.gov NCT01371981). During induction with cytarabine, daunorubicin, and etoposide (ADE), she developed gram-negative sepsis, but fully recovered. She achieved morphological remission after induction, including resolution of leukemia cutis.

Patient 5

A 4-month-old boy was admitted with poor feeding, pale skin, and enlarging abdomen. Physical examination revealed pallor and hepatosplenomegaly. Laboratory examination showed leukopenia and neutropenia (WBCs, 2.1 × 109/L with 25% neutrophils), anemia (Hb, 5.4 g/dL), and thrombocytopenia (PLTs, 56 × 109/L). Bone marrow was difficult to aspirate, but sufficient material was obtained for flow cytometry, showing 23% blast cells with a megakaryoblastic phenotype (myeloperoxidase-negative [MPO], CD41a+, CD61+) and RBM15-MKL1 fusion. Bone marrow biopsy showed striking fibrosis. CSF examination showed WBCs at 0 × 109/L, and cytospin showed no blasts morphologically. He was enrolled in the COG AAML1031 protocol. During induction, he developed a presumed fungal pneumonia (nodular infiltrates on computed tomography with a positive serum galactomannan) that resolved with liposomal amphotericin B. The end-induction bone marrow showed 15% residual megakaryoblasts.

Patient 6

A 7-month-old girl was admitted to the hospital because of a worsening rash, fever, and poor feeding. Physical examination revealed petechiae, pallor, and diffuse adenopathy. Laboratory examination showed leukocytosis (WBCs, 95 × 109/L), anemia (Hb, 6.5 g/dL), and thrombocytopenia (PLTs, 14 × 109/L). Bone marrow examination showed 94% blast cells with 2 discrete populations of blasts: ∼60% of the blasts had an immature B-cell phenotype (CD19+, CD10, cytoplasmic immunoglobulin-negative, CD20) and ∼40% had a monoblastic phenotype (MPO+, CD34, HLA-DR+, CD33+, CD11b+). In addition, a KMT2A-MLLT10 gene fusion was detected. CSF examination showed WBCs at 13 × 109/L, and cytospin showed heterogeneous blasts morphologically. She was treated with induction according to the Interfant-06 protocol. During induction, she had febrile neutropenia and received broad-spectrum empiric antibiotics. She achieved morphological remission after induction.

A summary of the 6 patient cases is provided in Table 2.

Table 2.

Summary of patient cases

PatientDiagnostic categoryAge, mo/SexMolecular abnormalityInductionComplicationsResponse
High-risk KMT2A-rearranged B-ALL 1/M KMT2A-MLLT1 Interfant Febrile neutropenia Remission 
Intermediate-risk KMT2A-rearranged B-ALL 4/M KMT2A-AFF1 Interfant Febrile neutropenia Remission 
Low-risk wt-KMT2A B-ALL 10/F None Interfant None Remission 
KMT2A-rearranged AML 7/F KMT2A-MLLT3 ADE* Gram-negative sepsis Remission 
wt-KMT2A megakaryoblastic AML 4/M RBM15-MKL1 ADE* Fungal pneumonia Induction failure 
KMT2A-rearranged MPAL (B/myeloid) 7/F KMT2A-MLLT10 Interfant Febrile neutropenia Remission 
PatientDiagnostic categoryAge, mo/SexMolecular abnormalityInductionComplicationsResponse
High-risk KMT2A-rearranged B-ALL 1/M KMT2A-MLLT1 Interfant Febrile neutropenia Remission 
Intermediate-risk KMT2A-rearranged B-ALL 4/M KMT2A-AFF1 Interfant Febrile neutropenia Remission 
Low-risk wt-KMT2A B-ALL 10/F None Interfant None Remission 
KMT2A-rearranged AML 7/F KMT2A-MLLT3 ADE* Gram-negative sepsis Remission 
wt-KMT2A megakaryoblastic AML 4/M RBM15-MKL1 ADE* Fungal pneumonia Induction failure 
KMT2A-rearranged MPAL (B/myeloid) 7/F KMT2A-MLLT10 Interfant Febrile neutropenia Remission 

F, female; Interfant, Interfant induction (Figure 1); M, male; MPAL, mixed phenotype acute leukemia.

*

ADE indicates cytarabine 100 mg/m2 every 12 hours days 1 to 10; daunorubicin 50 mg/m2 days 1, 3, and 5; etoposide 100 mg/m2 days 1 to 5.

Definition and epidemiology

“Infant leukemia” refers to acute leukemia diagnosed prior to 1 year of age. Fortunately, infant leukemia is rare, with estimated incidence of 41 cases per million in the United States, which equates to ∼160 cases of infant leukemia per year. Within the infant age group, neuroblastoma, brain tumors, and acute leukemia all occur with similar frequency. There is a slight predominance of lymphoid over myeloid cases within infant leukemia, and of the lymphoid cases, nearly all are B-lineage with <5% T-lineage. The incidence of acute lymphoblastic leukemia (ALL) in infants is lower than in children aged 1 to 14 years old and approximately the same as the incidence of ALL in adolescents. In contrast, the incidence of acute myeloid leukemia (AML) in infants is about twice that of the incidence of AML in older children and adolescents. Interestingly, infant leukemia demonstrates a female predominance, in contrast to the male predominance in leukemia diagnosed beyond the first birthday.1 

Clinical features

Compared with older children, infants with acute leukemia tend to present with more aggressive features, including high WBC counts, hepatosplenomegaly, central nervous system (CNS) involvement, and leukemia cutis (skin infiltration).2,3 

Prognosis

The prognostic significance of infant age differs between ALL and AML. In ALL, infants fare far worse than older children. The 4-year event-free survival (EFS) in Interfant-99, the largest trial of infant ALL to date, was 47%.4  Recent trials for childhood ALL report long-term EFS rates exceeding 85%.5,6  In AML, outcomes for infants are similar to those for older children.3 

KMT2A (formerly MLL) gene rearrangements

A high proportion of infant leukemias are characterized cytogenetically by balanced chromosomal translocations involving the histone lysine methyltransferase 2A gene (KMT2A, formerly known as mixed lineage leukemia [MLL] gene) at chromosome 11q23. KMT2A rearrangements (KMT2A-r) occur in ∼5% of childhood ALL cases overall,7  but in 70% to 80% of ALL in infants.2,4  In childhood AML, KMT2A-r is more common overall (15%-20%), but is also particularly common in the infant age group (∼50%).8 

KMT2A-r results in the fusion of the N terminus of the KMT2A gene with the C terminus of a partner gene. Remarkably, 94 different KMT2A partner genes have now been identified.9  In infant ALL, 4 partner genes account for 93% of cases: AFF1 (formerly AF4; 49%), MLLT1 (formerly ENL; 22%), MLLT3 (formerly AF9; 17%), and MLLT10 (formerly AF10; 5%). In infant AML, 3 partner genes account for 66% of cases: MLLT3 (22%), MLLT10 (27%), and ELL (17%).

Various lines of evidence (eg, retrospective analyses of neonatal samples10  and twin concordance studies11 ) have shown that KMT2A rearrangements are acquired in hematopoietic precursors in utero, and, compared with other oncogenic fusions such as ETV6-RUNX1, initiate a strikingly rapid progression to leukemia. One intriguing aspect of leukemia epidemiology is that KMT2A-r leukemias occur with high frequency in 2 very different clinical situations: (1) infants with de novo acute leukemia and (2) patients with treatment-related secondary myelodysplastic syndrome/AML after exposure to potent DNA topoisomerase II (DNAt2) inhibitors (eg, etoposide). This has led to a hypothesis, with supporting evidence from case-control12,13  and laboratory14  studies, that maternal exposure to environmental DNAt2 inhibitors (eg, dietary flavonoids) during pregnancy may contribute to the risk of KMT2A-r infant leukemia. Germline genetic susceptibility may also play a role, as candidate gene studies15,16  and genome-wide association studies17,18  have identified a number of single-nucleotide polymorphisms that correlate with risk of infant leukemia.

In ALL, KMT2A-r is associated with CD10 negativity and coexpression of 1 or more myeloid antigens, suggesting that these leukemias arise from very immature lymphoid progenitors.19  In AML, KMT2A-r is associated with monocytic differentiation.3  Infant leukemia cases can be of ambiguous lineage, either due to a mixed phenotype (mixed phenotype acute leukemia [MPAL], as shown in patient 6) or to lack of differentiation markers (acute undifferentiated leukemia).

KMT2A-r has different prognostic implications in infant ALL than infant AML. In infant ALL, KMT2A-r is clearly associated with poorer outcome. In the Children’s Cancer Group protocol CCG-1953, 5-year EFS for KMT2A-r infants was 34% vs 60% with germline (wild type) KMT2A (wt-KMT2A).2  In Interfant-99, 4-year EFS in KMT2A-r and wt-KMT2A infants was 37% and 74%, respectively.4  In infant AML, KMT2A-r is not a significant risk factor. In a combined analysis of AML-BFM-98 and -2004, the 5-year EFS was 43% and 52% for KMT2A-r and wt-KMT2A infants, respectively (P = .59).3  There is, however, evidence in pediatric AML generally that within KMT2A-r cases, certain KMT2A fusion partners may be associated with favorable (eg, MLLT11) or unfavorable (eg, MLLT10) prognosis, and there are plans to incorporate these into future pediatric AML risk-stratification algorithms.20,21 

Among infants with KMT2A-r ALL, additional independent prognostic factors include age and WBCs at diagnosis, with the younger infants and those with the higher WBCs having poorer outcomes.2,4,22  In the context of a 7-day “prophase” of single-agent prednisone given prior to intensive induction chemotherapy in the Interfant-99 protocol, a poor response (≥1000 blasts per microliter in the peripheral blood on day 8) was also an independent negative prognostic factor.4 

Induction therapy and risk stratification: infant AML

Given the similar prognosis and response to therapy of infants with AML compared with older children, infants are generally treated on the same clinical trial protocols as older children, which typically include intensive multiagent chemotherapy to induce remission followed by consolidation with either additional chemotherapy courses (for patients with favorable prognostic features) or allogeneic hematopoietic stem cell transplantation (HSCT; for patients with unfavorable prognostic features). Gemtuzumab is likely to be added to chemotherapy in future protocols based on favorable results, both in pediatrics overall23  and infants specifically.24  Cases of AML in infants are relatively unlikely to harbor cytogenetic or molecular abnormalities that confer unfavorable risk or favorable risk, and risk stratification is typically dictated by end-induction minimal residual disease (MRD) testing, although the KMT2A fusion partner may be incorporated in future protocols.

One notable exception to this, however, is illustrated in patient 5. Among pediatric cases of acute megakaryoblastic leukemia, the vast majority occur in patients with Down syndrome (constitutional trisomy 21), typically characterized by presentation at age 2 to 4 years, a history of transient myeloproliferative disorder in the newborn period, and a favorable prognosis.25  When acute megakaryoblastic leukemia occurs in children without Down syndrome, on the other hand, the patient is nearly always an infant. A variety of oncogenic fusions, of which the first to be described was the RBM15-MKL1 fusion resulting from the reciprocal translocation t(1;22)(p13;q13), drive this rare subset of infant leukemia. The prognosis is generally unfavorable due to a high risk of chemotherapy resistance and relapse, although outcomes do vary according to the specific oncogenic fusion.26 

Induction therapy and risk stratification: infant ALL

Treatment of infant ALL is quite different than that for childhood ALL generally. There are 3 major cooperative groups conducting specific clinical trials for infant ALL: Interfant (based in Europe), COG (based in North America), and the Japanese Pediatric Leukemia Study Group (JPLSG). All have adopted an identical induction strategy based on Interfant-99 (Figure 1).4  Until recently, COG trials used a more intensive induction regimen, but this was abandoned in favor of the Interfant induction due to excessive toxicity.27  In recently completed trials, all used a prospective risk-stratified approach that incorporates KMT2A-r status and age (Table 3).

Table 3.

Risk stratification of infant ALL

RiskInterfantCOGJPLSGApproximate EFS, %
High KMT2A-r and age <6 mo and WBCs ≥ 300 000/μL KMT2A-r and age <3 mo KMT2A-r and (age <6 mo or CNS leukemia) 20 
Intermediate KMT2A-r and not high risk KMT2A-r and not high risk KMT2A-r and not high risk 50 
Low wt-KMT2A wt-KMT2A wt-KMT2A 75 
RiskInterfantCOGJPLSGApproximate EFS, %
High KMT2A-r and age <6 mo and WBCs ≥ 300 000/μL KMT2A-r and age <3 mo KMT2A-r and (age <6 mo or CNS leukemia) 20 
Intermediate KMT2A-r and not high risk KMT2A-r and not high risk KMT2A-r and not high risk 50 
Low wt-KMT2A wt-KMT2A wt-KMT2A 75 

Among infant ALL patients, wt-KMT2A patients are considered low risk, and have favorable clinical features (eg, lower WBCs, older age at presentation). However, the outcome of wt-KMT2A infants is clearly inferior to ALL patients diagnosed after 1 year of age. This is likely due, in part, to differences in distribution of favorable genetic features, as infants with wt-KMT2A ALL are much less likely to harbor the favorable genetic features high hyperdiploidy and ETV6–RUNX1 fusions compared with older children.28,29  Moving forward, wt-KMT2A infant ALL patients are likely to be treated on general frontline ALL protocols, and therefore subject to the same risk-group stratification strategies, including the prospective use of MRD monitoring.

Induction-therapy complications

The infant patient’s unique vulnerability to complications and toxicities presents a challenge in treating infant acute leukemia. There are limited data to guide how the distinct and rapidly changing physiology of infants (in terms of body composition, binding of drugs by plasma proteins, cytochrome p450 activity, renal function, immunocompetence, etc) should be considered in designing chemotherapy treatment protocols. The data that do exist differ by chemotherapy drug. For example, no age dependency was found for the pharmacokinetics of daunorubicin30  whereas the systemic clearance rate of methotrexate tended to increase with age during infancy.31  Infant leukemia protocols have encountered problems with excessive toxicity, both in ALL27  and AML.32  Survivors of infant leukemia also demonstrate an increased risk of late effects, particularly in cases where treatment included cranial radiation or HSCT.25 

Patient 1

Postinduction, patient 1 was randomized to receive consolidation with multiagent chemotherapy blocks protocol IB and MARMA. His MRD level post-MARMA was <10e−4 (0.01%) by polymerase chain reaction. He received allogeneic HSCT in first remission with an umbilical cord blood donor after a preparatory regimen with busulfan, cyclophosphamide, and melphalan. He relapsed 7 months after HSCT. His leukemia was refractory to several attempts at reinduction with various multiagent salvage chemotherapy regimens and he died of overwhelming sepsis and multiorgan failure at age 17 months.

Patient 2

Postinduction, patient 2 was randomized to receive consolidation with multiagent chemotherapy blocks (reinduction, consolidation, continuation) without the FLT3 inhibitor lestaurtinib, followed by maintenance therapy for a total duration of 2 years. He is now 5 years after initial diagnosis and remains in complete remission.

Patient 3

Postinduction, patient 3 received multiagent chemotherapy blocks protocol IB, MARMA, and OCTADAD, followed by maintenance for a total duration of 2 years. She is now 7 years after initial diagnosis and remains in complete remission.

Patient 4

Postinduction, patient 4 received 3 additional blocks of intensive multiagent chemotherapy. During recovery from the third and final postinduction block, she developed a rash that was biopsied and shown to represent relapsed leukemia cutis. Bone marrow and CSF also revealed recurrent disease (8% blasts in the bone marrow, WBCs at 7 × 109/L in CSF with blasts on cytospin). She received a salvage regimen with azacitidine and fludarabine, cytarabine, and granulocyte colony-stimulating factor (FLAG) chemotherapy. She achieved a second remission and proceeded to allogeneic HSCT with an HLA-haploidentical bone marrow donor after a preparatory regimen with busulfan and cyclophosphamide. She is now 2 years post-HSCT and remains in second remission.

Patient 5

Because patient 5 was refractory to induction therapy, he received a salvage chemotherapy regimen with mitoxantrone and high-dose cytarabine. He achieved a morphologic remission, but still had scattered detectable megakaryoblasts by morphology and 3% megakaryoblasts by flow cytometry. He received allogeneic HSCT with an umbilical cord blood donor after a preparatory regimen with busulfan, cyclophosphamide, and melphalan. He died of multiorgan failure in the setting of progressive fungal infection 4 months after HSCT. At autopsy, residual leukemia was seen in the bone marrow.

Patient 6

For patient 6, postinduction, flow cytometry revealed a residual population of 2.8% in the bone marrow that expressed the same monoblastic phenotype (MPO+, CD34, HLA-DR+, CD33+, CD11b+) that was detected as the minority population at diagnosis. She received a second round of induction therapy with ADE, complicated by Streptococcus viridans sepsis. She recovered and was in complete remission with negative flow cytometry. She received allogeneic HSCT with an HLA-identical sibling bone marrow donor after a preparatory regimen with busulfan and cyclophosphamide. She remains in remission 3 years after HSCT.

These cases are summarized in Table 2.

Postinduction therapy for infant leukemia

Postinduction management of infant leukemia is heterogeneous. Table 4 summarizes the postinduction treatment approaches for infant ALL by the major cooperative groups (see also, Table 3). Interfant-06 tested whether consolidation with “myeloid”-style chemotherapy with cytarabine, daunorubicin, mitoxantrone, and etoposide is superior to “lymphoid”-style consolidation with cyclophosphamide, cytarabine and 6-mercaptopurine in KMT2A-r infants. This stems from the hypothesis that these leukemias derive from an early hematopoietic precursor with myeloid differentiation potential and may therefore respond better to chemotherapy regimens developed for AML. AALL0631 tested whether the addition of an FLT3 tyrosine kinase inhibitor (lestaurtinib) to postinduction chemotherapy will enhance the effectiveness of chemotherapy, based on data showing aberrant activation of the FLT3 pathway in KMT2A-r ALL. Given the similarities in treatment approach and outcomes between the groups and the rarity of infant ALL, the 3 groups are currently developing a joint collaborative protocol to standardize treatment and enhance the ability to test novel treatment approaches based on recent discoveries regarding the unique molecular biology of KMT2A-r leukemias and ongoing pilot trials within the groups (detailed in subsequent sections).

Table 4.

Postinduction approaches for infant ALL in 3 major cooperative groups on most recent trials

Interfant*COGJPLSG
Trial Interfant-06 AALL0631 MLL-10 
Randomized postinduction intervention Protocol IB vs ADE/MAE ±Lestaurtinib (FLT3 tyrosine kinase inhibitor) None (single arm) 
HSCT All high risk, plus MRD+ after MARMA None All high risk 
Interfant*COGJPLSG
Trial Interfant-06 AALL0631 MLL-10 
Randomized postinduction intervention Protocol IB vs ADE/MAE ±Lestaurtinib (FLT3 tyrosine kinase inhibitor) None (single arm) 
HSCT All high risk, plus MRD+ after MARMA None All high risk 
*

Protocol IB consolidation indicates cyclophosphamide 1000 mg/m2 days 1 and 29; cytarabine 75 mg/m2 days 3 to 6, 10 to 13, 17 to 20, 24 to 27; 6-mercaptopurine 60 mg/m2 days 1 to 28 consolidation. ADE/MAE consolidation indicates cytarabine 100 mg/m2 every 12 hours days 1 to 10; daunorubicin 50 mg/m2 days 1, 3, and 5; etoposide 100 mg/m2 days 1 to 5/mitoxantrone 12 mg/m2 days 1, 3, and 5; cytarabine 100 mg/m2 every 12 hours days 1 to 10; etoposide 100 mg/m2 days 1 to 5. MARMA indicates methotrexate 5000 mg/m2 days 1 and 8; 6-mercaptopurine 25 mg/m2 days 1 to 14; cytarabine 3000 mg/m2 every 12 hours days 15, 16, 22 and 23; pegylated asparaginase 2500 IU/m2 day 23.

Postinduction treatment of infant AML is typically the same as for older children with AML. Patient 6 demonstrates a typical induction and postinduction course for an infant with MPAL. The decision of whether to proceed with induction treatment following an ALL or AML protocol is generally made based on whether the lymphoid or myeloid lineage appears to be predominant. Postinduction treatment should factor in the response to the chosen induction therapy.

HSCT in infant leukemia

The use of HSCT in infant leukemia is variable, reflecting uncertainty regarding the risk/benefit ratio of HSCT in this population. The published data (concisely reviewed by Sison and Brown33 ) are not conclusive. There does appear to be a small minority of KMT2A-r patients at high risk of relapse (very young age, very high WBCs, and persistence of MRD) who may benefit from HSCT in first remission.34,35 

Chemotherapy resistance and relapse in infant leukemia

Poor initial response to prednisone is significantly more common in infants than in older children with ALL, and infant KMT2A-r ALL cells demonstrate enhanced in vitro resistance to corticosteroids and asparaginase in assays using short-term exposure of bulk leukemia populations. Conversely, these cells appear to be particularly sensitive to nucleoside analogs like cytarabine, perhaps related to high expression levels of the membrane-bound nucleoside transporter ENT1.36,37  Infant KMT2A-r AML cells do not demonstrate enhanced resistance or sensitivity.38-40 

Clinical data do not support the hypothesis that bulk leukemia chemoresistance is responsible for poor outcomes. The typical pattern of failure for infant KMT2A-r ALL is to achieve rapid complete remission with induction chemotherapy, but then relapse several months later during active therapy. This suggests that the poor outcomes are due primarily to the emergence of a chemoresistant population over time. The low rates of second remission (∼40%) and very poor survival after relapse (∼20%) are consistent with this hypothesis.41,42 

Genomic studies of large cohorts of diagnostic KMT2A-r infant ALL specimens have revealed a relatively small number of cooperating genomic variants.43-46  A smaller number of paired diagnostic/relapse pairs have been studied, but there appears to be no significant increase or selection of mutations at relapse.44  Thus, it does not appear that relapse in KMT2A-r infant ALL can be explained by the appearance of, or selection for, subclones with resistance-inducing genomic mutations. As discussed in more detail in “Epigenetic agents,” KMT2A-r infant ALL is associated with aberrant epigenetic programs. Emerging data suggest that subclonal epigenetic changes may be responsible for chemoresistance and relapse in KMT2A-r infant ALL,47  raising the possibility that incorporating epigenetically targeted therapies may be able to prevent relapse.

Novel therapies

It is clear that our standard approaches to infant leukemia leave ample room for improvement. The unique molecular biology of KMT2A-r leukemia has suggested novel treatment approaches, several of which are in various stages of clinical investigation. The importance of collaborative and innovative clinical trials for this disease cannot be overemphasized. To that end, the 3 major cooperative groups in Tables 3 and 4 are developing a joint protocol to enhance the ability to test novel treatment approaches and improve the standard of care.

FLT3 inhibitors

KMT2A-r infant ALL is characterized by a distinct global gene-expression profile.48,49  A notable component of this profile is striking overexpression of FLT3.50  FLT3 signaling is constitutively activated in these cases, either by activating mutations51,52  or, more commonly, by autocrine activation via coexpressed FLT3 ligand.53  Moreover, FLT3 tyrosine kinase inhibition results in selective killing of these samples and synergizes with chemotherapy in a sequence-dependent manner.51,53-55  FLT3 overexpression has been shown to confer especially poor prognosis in KMT2A-r infant ALL.56,57  As shown in Table 4, COG trial AALL0631 was the first to incorporate a novel, molecularly targeted agent into frontline treatment of KMT2A-r infant ALL. Unfortunately, the trial failed to demonstrate a benefit for the addition of lestaurtinib, perhaps due in part to pharmacologic limitations.58  Nevertheless, this trial serves as proof of principle that novel targeted therapeutics can feasibly be tested in this high-risk group and has laid the groundwork for the international collaborative trial in development.

Epigenetic agents

Genomic studies have revealed a striking paucity of cooperating genetic alterations in infant KMT2A-r ALL compared with all other subsets of childhood ALL.43-46,59  Accordingly, KMT2A-r infant leukemia is increasingly recognized to be driven by aberrant epigenetic programs. As this complex network of interdependent epigenetic processes is elucidated, novel therapeutic strategies are emerging.

KMT2A translocations include partner genes that recruit multiprotein complexes with chromatin-modifying activity to KMT2A target genes, inducing dysregulated transcription of multiple genes. A required component of this aberrant epigenetic state and KMT2A-r leukemogenesis is the H3K79 methyltransferase DOT1L.60-63  Potent and highly selective small-molecule inhibitors of DOT1L showed promising activity in preclinical models of KMT2A-r leukemia.64,65  Unfortunately, the clinical activity of pinometostat, the first DOT1L inhibitor studied, was limited when used as monotherapy in relapsed adults and children with KMT2A-r leukemia.66,67 

BRD4 is one of a number of epigenetic “reader” proteins that binds acetylated histones and facilitates transcription downstream of MYC and other validated oncogenes. A nonbiased RNA interference screen of 243 chromatin-modifying genes identified BRD4 to be required for the maintenance of leukemia in an MLL-AF9 murine model.68  Potent and selective small-molecule inhibitors of BRD4 binding downregulated characteristic KMT2A-r and MYC target genes and demonstrated in vitro and in vivo antileukemic activity by inducing apoptosis and differentiation in murine models, leukemia cell lines, and, most importantly, in a cohort of primary KMT2A-r infant ALL cells.68,69  Early-phase clinical trials in adults with hematologic malignancies have been or are being conducted with bromodomain inhibitors such as OTX015 and CPI-0610. A phase 1 trial of OTX015 in adults with acute leukemia, including 2 with KMT2A-r, demonstrated modest activity as a single agent.70 

Also characteristic of KMT2A-r ALL is the epigenetic silencing of another set of genes with tumor-suppressor function via promoter region CpG island hypermethylation.71,72  Increasing degrees of promoter hypermethylation correlated with inferior survival in Interfant-99.72  Demethylating agents such as azacitidine, decitabine, and zebularine preferentially kill KMT2A-r ALL cells, and this correlates with the upregulation of several of the identified silenced genes.71-73  Hypermethylation of certain microRNAs (miRs), such as miR-152, also characterizes infant KMT2A-r ALL and has been associated with inferior outcomes.74,75  Based on these data, as well as the aforementioned report of the critical role of subclonal methylation changes in chemoresistance and relapse of infant KMT2A-r ALL,47  the COG (clinicaltrials.gov NCT02828358) and JPLSG (umin.ac.jp UMIN29275) are each conducting pilot trials of azacitidine in combination with the Interfant chemotherapy backbone.

Deacetylation of histone marks such as H3K9/14 is associated with gene silencing and can be modulated with histone deacetylase (HDAC) inhibitors. Connectivity maps have identified HDAC inhibitors as capable of reversing gene-expression profiles associated with chemotherapy resistance in relapsed childhood ALL76  and in infant KMT2A-r ALL.77  A high-throughput screen of a large panel of US Food and Drug Administration (FDA)-approved and investigational drugs in KMT2A-r infant ALL cell lines identified the HDAC inhibitor romidepsin as efficacious and synergistic in vivo with cytarabine.78  Finally, the HDAC inhibitor panobinostat was found to be effective in xenograft models of KMT2A-r ALL.79  Although demethylating agents and HDAC inhibitors are a promising strategy to reverse the inherent chemoresistance of KMT2A-r infant ALL, a pilot study of decitabine and vorinostat combined with reinduction chemotherapy in children with relapsed ALL led to excessive infectious toxicity,80  so safety has yet to be demonstrated.

Targeting microenvironment interactions

The pattern of remission and early relapse suggests that chemotherapy-resistant leukemia stem cells survive and can recapitulate the leukemia. Interactions between infant KMT2A-r ALL leukemia stem cells and the bone marrow stromal microenvironment via the CXCR4/SDF-1 axis have been shown to mediate survival and therapeutic resistance in KMT2A-r ALL.81  In xenograft models of KMT2A-r infant ALL, CXCR4 inhibition with plerixafor led to markedly enhanced in vivo efficacy of FLT3 inhibitors, suggesting that targeting leukemia-stroma interactions with CXCR4 inhibitors may represent a promising adjunctive therapy. Dynamic upregulation of CXCR4 expression on the surface of acute leukemias (including KMT2A-r cases) in response to cytotoxic chemotherapy may enhance this stroma-mediated resistance and identify patients for whom CXCR4 inhibition may be particularly effective.82  Subsequent preclinical studies have demonstrated the efficacy of small-molecule CXCR4 antagonism in reversing chemoresistance, both in vitro and in vivo,83,84  and these findings led to a clinical trial of plerixafor and chemotherapy in relapsed/refractory pediatric acute leukemia (clinicaltrials.gov NCT01319864). The combination was safe but showed modest clinical activity, although no KMT2A-r ALL patients were enrolled.85 

Immunotherapy

The recent FDA approval of blinatumomab86  and tisagenlecleucel87  has generated interest in immunotherapeutic approaches for high-risk B-ALL subsets, to include KMT2A-r infant ALL. A pilot trial of blinatumomab in combination with the Interfant chemotherapy backbone has been initiated by the Interfant group (trialregister.nl NTR6359). One potential limitation, however, is that CD19 antigen is not uniformly expressed in KMT2A-r leukemias. Case reports of immunologic “class switching” from CD19+ lymphoid to CD19 myeloid phenotypes have been reported with CD19-targeted immunotherapies,88-90  and xenograft studies demonstrate leukemia stem/initiating cell activity in CD19 subpopulations in KMT2A-r ALL cases.91-93  Low levels of CD22 expression in KMT2A-r ALL may also limit the activity of inotuzumab.94  Another challenge to chimeric antigen receptor (CAR) T-cell technology in infants is manufacturing product using autologous infant T cells. Recently, a gene-editing technique was used to simultaneously introduce the CD19 CAR construct and disrupt mediators of allogeneic rejection (T-cell receptor α chain and CD52) into healthy donor T cells, creating an “off-the-shelf” universal CAR-T product. Remarkably, 2 relapsed KMT2A-r ALL infants were treated and achieved molecular remission before proceeding to HSCT.95  Ultimately, the success of immunotherapy for infant leukemia will depend upon identifying the right antigen (or group of antigens) to target, because as discussed, CD19 and CD22 may not be optimal.

Targeting RAS pathway

Mutations in NRAS and KRAS have been found in 14% of KMT2A-r infant ALL.96  As has been found in other acute leukemias, RAS mutations tend to be subclonal and not stable between diagnosis and relapse, raising the question of whether they represent leukemia drivers.97,98  However, a cohort in which 70 patients were negative for RAS mutations and 9 were positive demonstrated a worse outcome among the RAS-mutant group,96  and preclinical studies suggest a potential role for MEK inhibition as a targeted therapeutic strategy.99,100 

Infant leukemia is one of most difficult clinical situations encountered in pediatric hematology/oncology. Standard approaches (consensus recommendations summarized in Table 5) are curative in a minority of patients, and participation in clinical trials of novel treatment approaches is strongly encouraged. Although most treatment failures are due to relapse, treatment-related mortality and life-limiting late effects in survivors are also problematic. Recent discoveries regarding the unique biology of these leukemias are fueling the development of a pipeline of exciting novel treatment strategies that are increasingly being incorporated into clinical trials and have the potential to reduce both relapse rates and treatment-related toxicities. Increasing collaboration among the major international cooperative groups will accelerate the translation of biological understanding into better outcomes.

Table 5.

Summary of consensus recommended treatment strategies for infant leukemia subtypes

Risk groupDefined on the basis ofRecommended treatment approach
Infant ALL   
 High KMT2A-r, younger age, late MRD clearance Interfant induction, then intensive chemotherapy consolidation, then strongly consider HSCT (prefer non–total body irradiation based, prefer age at HSCT ≥6 mo); continued consolidation and maintenance if HSCT unavailable 
 Intermediate KMT2A-r, older age, early MRD clearance Interfant induction, then intensive chemotherapy consolidation and maintenance 
 Low wt-KMT2A Interfant induction, then identical approach as pediatric ALL (risk-stratified chemotherapy based on genetics and MRD response) 
Infant AML Identical approach as pediatric AML (intensive chemotherapy/gemtuzumab induction, then risk-based consolidation with chemotherapy/gemtuzumab for low risk and HSCT for high risk) 
Risk groupDefined on the basis ofRecommended treatment approach
Infant ALL   
 High KMT2A-r, younger age, late MRD clearance Interfant induction, then intensive chemotherapy consolidation, then strongly consider HSCT (prefer non–total body irradiation based, prefer age at HSCT ≥6 mo); continued consolidation and maintenance if HSCT unavailable 
 Intermediate KMT2A-r, older age, early MRD clearance Interfant induction, then intensive chemotherapy consolidation and maintenance 
 Low wt-KMT2A Interfant induction, then identical approach as pediatric ALL (risk-stratified chemotherapy based on genetics and MRD response) 
Infant AML Identical approach as pediatric AML (intensive chemotherapy/gemtuzumab induction, then risk-based consolidation with chemotherapy/gemtuzumab for low risk and HSCT for high risk) 

A.B. was supported by Associazione Italiana per la Ricerca sul Cancro (20564, AIRC 5x1000) and Transcan2-189.

Contribution: P.B., R.B., and A.B. wrote the paper.

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

Correspondence: Patrick Brown, Johns Hopkins Oncology, 1650 Orleans St, CRB1 Room 2M51, Baltimore, MD 21231; e-mail: pbrown2@jhmi.edu.

1.
Howlader
N
,
Noone
AM
,
Krapcho
M
, et al
. SEER Cancer Statistics Review, 1975-2010.
Bethesda, MD
:
National Cancer Institute
; 2013.
2.
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
.
3.
Creutzig
U
,
Zimmermann
M
,
Bourquin
JP
, et al
.
Favorable outcome in infants with AML after intensive first- and second-line treatment: an AML-BFM study group report
.
Leukemia
.
2012
;
26
(
4
):
654
-
661
.
4.
Pieters
R
,
Schrappe
M
,
De Lorenzo
P
, et al
.
A treatment protocol for infants younger than 1 year with acute lymphoblastic leukaemia (Interfant-99): an observational study and a multicentre randomised trial
.
Lancet
.
2007
;
370
(
9583
):
240
-
250
.
5.
Hunger
SP
,
Lu
X
,
Devidas
M
, et al
.
Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: a report from the Children’s Oncology Group
.
J Clin Oncol
.
2012
;
30
(
14
):
1663
-
1669
.
6.
Pui
CH
,
Campana
D
,
Pei
D
, et al
.
Treating childhood acute lymphoblastic leukemia without cranial irradiation
.
N Engl J Med
.
2009
;
360
(
26
):
2730
-
2741
.
7.
Behm
FG
,
Raimondi
SC
,
Frestedt
JL
, et al
.
Rearrangement of the MLL gene confers a poor prognosis in childhood acute lymphoblastic leukemia, regardless of presenting age
.
Blood
.
1996
;
87
(
7
):
2870
-
2877
.
8.
Harrison
CJ
,
Hills
RK
,
Moorman
AV
, et al
.
Cytogenetics of childhood acute myeloid leukemia: United Kingdom Medical Research Council Treatment trials AML 10 and 12
.
J Clin Oncol
.
2010
;
28
(
16
):
2674
-
2681
.
9.
Meyer
C
,
Burmeister
T
,
Gröger
D
, et al
.
The MLL recombinome of acute leukemias in 2017
.
Leukemia
.
2018
;
32
(
2
):
273
-
284
.
10.
Gale
KB
,
Ford
AM
,
Repp
R
, et al
.
Backtracking leukemia to birth: identification of clonotypic gene fusion sequences in neonatal blood spots
.
Proc Natl Acad Sci USA
.
1997
;
94
(
25
):
13950
-
13954
.
11.
Ford
AM
,
Ridge
SA
,
Cabrera
ME
, et al
.
In utero rearrangements in the trithorax-related oncogene in infant leukaemias
.
Nature
.
1993
;
363
(
6427
):
358
-
360
.
12.
Alexander
FE
,
Patheal
SL
,
Biondi
A
, et al
.
Transplacental chemical exposure and risk of infant leukemia with MLL gene fusion
.
Cancer Res
.
2001
;
61
(
6
):
2542
-
2546
.
13.
Spector
LG
,
Xie
Y
,
Robison
LL
, et al
.
Maternal diet and infant leukemia: the DNA topoisomerase II inhibitor hypothesis: a report from the Children’s Oncology Group
.
Cancer Epidemiol Biomarkers Prev
.
2005
;
14
(
3
):
651
-
655
.
14.
Strick
R
,
Strissel
PL
,
Borgers
S
,
Smith
SL
,
Rowley
JD
.
Dietary bioflavonoids induce cleavage in the MLL gene and may contribute to infant leukemia
.
Proc Natl Acad Sci USA
.
2000
;
97
(
9
):
4790
-
4795
.
15.
Smith
MT
,
Wang
Y
,
Skibola
CF
, et al
.
Low NAD(P)H:quinone oxidoreductase activity is associated with increased risk of leukemia with MLL translocations in infants and children
.
Blood
.
2002
;
100
(
13
):
4590
-
4593
.
16.
Wiemels
JL
,
Smith
RN
,
Taylor
GM
,
Eden
OB
,
Alexander
FE
,
Greaves
MF
;
United Kingdom Childhood Cancer Study Investigators
.
Methylenetetrahydrofolate reductase (MTHFR) polymorphisms and risk of molecularly defined subtypes of childhood acute leukemia
.
Proc Natl Acad Sci USA
.
2001
;
98
(
7
):
4004
-
4009
.
17.
Ross
JA
,
Linabery
AM
,
Blommer
CN
, et al
.
Genetic variants modify susceptibility to leukemia in infants: a Children’s Oncology Group report
.
Pediatr Blood Cancer
.
2013
;
60
(
1
):
31
-
34
.
18.
Valentine
MC
,
Linabery
AM
,
Chasnoff
S
, et al
.
Excess congenital non-synonymous variation in leukemia-associated genes in MLL- infant leukemia: a Children’s Oncology Group report
.
Leukemia
.
2014
;
28
(
6
):
1235
-
1241
.
19.
Basso
G
,
Rondelli
R
,
Covezzoli
A
,
Putti
M
.
The role of immunophenotype in acute lymphoblastic leukemia of infant age
.
Leuk Lymphoma
.
1994
;
15
(
1-2
):
51
-
60
.
20.
Cooper
TM
,
Ries
RE
,
Alonzo
TA
, et al
.
Revised risk stratification criteria for children with newly diagnosed acute myeloid leukemia: a report from the Children’s Oncology Group
[abstract].
Blood
.
2017
;
130
(
suppl 1
). Abstract
407
.
21.
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
.
22.
Tomizawa
D
,
Koh
K
,
Sato
T
, et al
.
Outcome of risk-based therapy for infant acute lymphoblastic leukemia with or without an MLL gene rearrangement, with emphasis on late effects: a final report of two consecutive studies, MLL96 and MLL98, of the Japan Infant Leukemia Study Group
.
Leukemia
.
2007
;
21
(
11
):
2258
-
2263
.
23.
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
.
24.
Guest
EM
,
Aplenc
R
,
Sung
L
, et al
.
Gemtuzumab ozogamicin in infants with AML: results from the Children’s Oncology Group trials AAML03P1 and AAML0531
.
Blood
.
2017
;
130
(
7
):
943
-
945
.
25.
Hitzler
JK
,
Zipursky
A
.
Origins of leukaemia in children with Down syndrome
.
Nat Rev Cancer
.
2005
;
5
(
1
):
11
-
20
.
26.
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
.
27.
Salzer
WL
,
Jones
TL
,
Devidas
M
, et al
.
Decreased induction morbidity and mortality following modification to induction therapy in infants with acute lymphoblastic leukemia enrolled on AALL0631: a report from the Children’s Oncology Group
.
Pediatr Blood Cancer
.
2015
;
62
(
3
):
414
-
418
.
28.
De Lorenzo
P
,
Moorman
AV
,
Pieters
R
, et al
.
Cytogenetics and outcome of infants with acute lymphoblastic leukemia and absence of MLL rearrangements
.
Leukemia
.
2014
;
28
(
2
):
428
-
430
.
29.
van der Linden
MH
,
Boer
JM
,
Schneider
P
, et al
.
Clinical and molecular genetic characterization of wild-type MLL infant acute lymphoblastic leukemia identifies few recurrent abnormalities
.
Haematologica
.
2016
;
101
(
3
):
e95
-
e98
.
30.
Hempel
G
,
Relling
MV
,
de Rossi
G
, et al
.
Pharmacokinetics of daunorubicin and daunorubicinol in infants with leukemia treated in the interfant 99 protocol
.
Pediatr Blood Cancer
.
2010
;
54
(
3
):
355
-
360
.
31.
Lönnerholm
G
,
Valsecchi
MG
,
De Lorenzo
P
, et al
;
Interfant-99 Study Group
.
Pharmacokinetics of high-dose methotrexate in infants treated for acute lymphoblastic leukemia
.
Pediatr Blood Cancer
.
2009
;
52
(
5
):
596
-
601
.
32.
Gibson
BE
,
Webb
DK
,
Howman
AJ
,
De Graaf
SS
,
Harrison
CJ
,
Wheatley
K
;
United Kingdom Childhood Leukaemia Working Group and the Dutch Childhood Oncology Group
.
Results of a randomized trial in children with acute myeloid leukaemia: Medical Research Council AML12 trial
.
Br J Haematol
.
2011
;
155
(
3
):
366
-
376
.
33.
Sison
EA
,
Brown
P
.
Does hematopoietic stem cell transplantation benefit infants with acute leukemia?
Hematology Am Soc Hematol Educ Program
.
2013
;
2013
:
601
-
604
.
34.
Mann
G
,
Attarbaschi
A
,
Schrappe
M
, et al
;
Interfant-99 Study Group
.
Improved outcome with hematopoietic stem cell transplantation in a poor prognostic subgroup of infants with mixed-lineage-leukemia (MLL)-rearranged acute lymphoblastic leukemia: results from the Interfant-99 Study
.
Blood
.
2010
;
116
(
15
):
2644
-
2650
.
35.
Van der Velden
VH
,
Corral
L
,
Valsecchi
MG
, et al
;
Interfant-99 Study Group
.
Prognostic significance of minimal residual disease in infants with acute lymphoblastic leukemia treated within the Interfant-99 protocol
.
Leukemia
.
2009
;
23
(
6
):
1073
-
1079
.
36.
Stam
RW
,
den Boer
ML
,
Meijerink
JP
, et al
.
Differential mRNA expression of Ara-C-metabolizing enzymes explains Ara-C sensitivity in MLL gene-rearranged infant acute lymphoblastic leukemia
.
Blood
.
2003
;
101
(
4
):
1270
-
1276
.
37.
Stumpel
DJ
,
Schneider
P
,
Pieters
R
,
Stam
RW
.
The potential of clofarabine in MLL-rearranged infant acute lymphoblastic leukaemia
.
Eur J Cancer
.
2015
;
51
(
14
):
2008
-
2021
.
38.
Ramakers-van Woerden
NL
,
Beverloo
HB
,
Veerman
AJ
, et al
.
In vitro drug-resistance profile in infant acute lymphoblastic leukemia in relation to age, MLL rearrangements and immunophenotype
.
Leukemia
.
2004
;
18
(
3
):
521
-
529
.
39.
Palle
J
,
Frost
BM
,
Forestier
E
, et al
;
Nordic Society for Paediatric Haematology and Oncology
.
Cellular drug sensitivity in MLL-rearranged childhood acute leukaemia is correlated to partner genes and cell lineage
.
Br J Haematol
.
2005
;
129
(
2
):
189
-
198
.
40.
Zwaan
CM
,
Kaspers
GJ
,
Pieters
R
, et al
.
Cellular drug resistance in childhood acute myeloid leukemia is related to chromosomal abnormalities
.
Blood
.
2002
;
100
(
9
):
3352
-
3360
.
41.
Tomizawa
D
,
Koh
K
,
Hirayama
M
, et al
.
Outcome of recurrent or refractory acute lymphoblastic leukemia in infants with MLL gene rearrangements: a report from the Japan Infant Leukemia Study Group
.
Pediatr Blood Cancer
.
2009
;
52
(
7
):
808
-
813
.
42.
Driessen
EMC
,
de Lorenzo
P
,
Campbell
M
, et al
.
Outcome of relapsed infant acute lymphoblastic leukemia treated on the Interfant-99 protocol [published correction appears in Leukemia. 2017;31(12):2854]
.
Leukemia
.
2016
;
30
(
5
):
1184
-
1187
.
43.
Mullighan
CG
,
Goorha
S
,
Radtke
I
, et al
.
Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia
.
Nature
.
2007
;
446
(
7137
):
758
-
764
.
44.
Andersson
AK
,
Ma
J
,
Wang
J
, et al
;
St. Jude Children’s Research Hospital–Washington University Pediatric Cancer Genome Project
.
The landscape of somatic mutations in infant MLL-rearranged acute lymphoblastic leukemias
.
Nat Genet
.
2015
;
47
(
4
):
330
-
337
.
45.
Bardini
M
,
Galbiati
M
,
Lettieri
A
, et al
.
Implementation of array based whole-genome high-resolution technologies confirms the absence of secondary copy-number alterations in MLL-AF4-positive infant ALL patients
.
Leukemia
.
2011
;
25
(
1
):
175
-
178
.
46.
Dobbins
SE
,
Sherborne
AL
,
Ma
YP
, et al
.
The silent mutational landscape of infant MLL-AF4 pro-B acute lymphoblastic leukemia
.
Genes Chromosomes Cancer
.
2013
;
52
(
10
):
954
-
960
.
47.
Kostadinov
R
,
Scharpf
R
,
Brown
P
.
Identifying subclonal epigenetic changes driving chemoresistance in infant MLL-r acute lymphoblastic leukemias [abstract]
.
Blood
.
2015
;
126
(
23
). Abstract
809
.
48.
Kang
H
,
Wilson
CS
,
Harvey
RC
, et al
.
Gene expression profiles predictive of outcome and age in infant acute lymphoblastic leukemia: a Children’s Oncology Group study
.
Blood
.
2012
;
119
(
8
):
1872
-
1881
.
49.
Stam
RW
,
Schneider
P
,
Hagelstein
JA
, et al
.
Gene expression profiling-based dissection of MLL translocated and MLL germline acute lymphoblastic leukemia in infants
.
Blood
.
2010
;
115
(
14
):
2835
-
2844
.
50.
Armstrong
SA
,
Staunton
JE
,
Silverman
LB
, et al
.
MLL translocations specify a distinct gene expression profile that distinguishes a unique leukemia
.
Nat Genet
.
2002
;
30
(
1
):
41
-
47
.
51.
Armstrong
SA
,
Kung
AL
,
Mabon
ME
, et al
.
Inhibition of FLT3 in MLL. Validation of a therapeutic target identified by gene expression based classification
.
Cancer Cell
.
2003
;
3
(
2
):
173
-
183
.
52.
Taketani
T
,
Taki
T
,
Sugita
K
, et al
.
FLT3 mutations in the activation loop of tyrosine kinase domain are frequently found in infant ALL with MLL rearrangements and pediatric ALL with hyperdiploidy
.
Blood
.
2004
;
103
(
3
):
1085
-
1088
.
53.
Brown
P
,
Levis
M
,
Shurtleff
S
,
Campana
D
,
Downing
J
,
Small
D
.
FLT3 inhibition selectively kills childhood acute lymphoblastic leukemia cells with high levels of FLT3 expression
.
Blood
.
2005
;
105
(
2
):
812
-
820
.
54.
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
.
55.
Stam
RW
,
den Boer
ML
,
Schneider
P
, et al
.
Targeting FLT3 in primary MLL-gene-rearranged infant acute lymphoblastic leukemia
.
Blood
.
2005
;
106
(
7
):
2484
-
2490
.
56.
Chillón
MC
,
Gómez-Casares
MT
,
López-Jorge
CE
, et al
.
Prognostic significance of FLT3 mutational status and expression levels in MLL-AF4+ and MLL-germline acute lymphoblastic leukemia
.
Leukemia
.
2012
;
26
(
11
):
2360
-
2366
.
57.
Stam
RW
,
Schneider
P
,
de Lorenzo
P
,
Valsecchi
MG
,
den Boer
ML
,
Pieters
R
.
Prognostic significance of high-level FLT3 expression in MLL-rearranged infant acute lymphoblastic leukemia
.
Blood
.
2007
;
110
(
7
):
2774
-
2775
.
58.
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 [abstract]
.
Pediatr Blood Cancer
.
2016
;
63
(
suppl S3
). Abstract
S7
.
59.
Bardini
M
,
Spinelli
R
,
Bungaro
S
, et al
.
DNA copy-number abnormalities do not occur in infant ALL with t(4;11)/MLL-AF4
.
Leukemia
.
2010
;
24
(
1
):
169
-
176
.
60.
Chen
L
,
Deshpande
AJ
,
Banka
D
, et al
.
Abrogation of MLL-AF10 and CALM-AF10-mediated transformation through genetic inactivation or pharmacological inhibition of the H3K79 methyltransferase Dot1l
.
Leukemia
.
2013
;
27
(
4
):
813
-
822
.
61.
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
.
62.
Nguyen
AT
,
Taranova
O
,
He
J
,
Zhang
Y
.
DOT1L, the H3K79 methyltransferase, is required for MLL-AF9-mediated leukemogenesis
.
Blood
.
2011
;
117
(
25
):
6912
-
6922
.
63.
Deshpande
AJ
,
Chen
L
,
Fazio
M
, et al
.
Leukemic transformation by the MLL-AF6 fusion oncogene requires the H3K79 methyltransferase Dot1l
.
Blood
.
2013
;
121
(
13
):
2533
-
2541
.
64.
Daigle
SR
,
Olhava
EJ
,
Therkelsen
CA
, et al
.
Potent inhibition of DOT1L as treatment of MLL-fusion leukemia
.
Blood
.
2013
;
122
(
6
):
1017
-
1025
.
65.
Daigle
SR
,
Olhava
EJ
,
Therkelsen
CA
, et al
.
Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor
.
Cancer Cell
.
2011
;
20
(
1
):
53
-
65
.
66.
Stein
EM
,
Garcia-Manero
G
,
Rizzieri
DA
, et al
.
The DOT1L inhibitor EPZ-5676: safety and activity in relapsed/refractory patients with MLL-rearranged leukemia [abstract]
.
Blood
.
2014
;
124
(
21
). Abstract
387
.
67.
Shukla
N
,
O’Brien
MM
,
Silverman
LB
, et al
.
Preliminary report of the phase 1 study of the DOT1L inhibitor, pinometostat, EPZ-5676, in children with relapsed or refractory MLL-r acute leukemia: safety, exposure and target inhibition [abstract]
.
Blood
.
2015
;
126
(
23
). Abstract
3792
.
68.
Zuber
J
,
Shi
J
,
Wang
E
, et al
.
RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia
.
Nature
.
2011
;
478
(
7370
):
524
-
528
.
69.
Dawson
MA
,
Prinjha
RK
,
Dittmann
A
, et al
.
Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia
.
Nature
.
2011
;
478
(
7370
):
529
-
533
.
70.
Berthon
C
,
Raffoux
E
,
Thomas
X
, et al
.
Bromodomain inhibitor OTX015 in patients with acute leukaemia: a dose-escalation, phase 1 study
.
Lancet Haematol
.
2016
;
3
(
4
):
e186
-
e195
.
71.
Schafer
E
,
Irizarry
R
,
Negi
S
, et al
.
Promoter hypermethylation in MLL-r infant acute lymphoblastic leukemia: biology and therapeutic targeting
.
Blood
.
2010
;
115
(
23
):
4798
-
4809
.
72.
Stumpel
DJ
,
Schneider
P
,
van Roon
EH
, et al
.
Specific promoter methylation identifies different subgroups of MLL-rearranged infant acute lymphoblastic leukemia, influences clinical outcome, and provides therapeutic options
.
Blood
.
2009
;
114
(
27
):
5490
-
5498
.
73.
Stumpel
DJ
,
Schneider
P
,
van Roon
EH
,
Pieters
R
,
Stam
RW
.
Absence of global hypomethylation in promoter hypermethylated mixed lineage leukaemia-rearranged infant acute lymphoblastic leukaemia
.
Eur J Cancer
.
2013
;
49
(
1
):
175
-
184
.
74.
Stumpel
DJ
,
Schotte
D
,
Lange-Turenhout
EA
, et al
.
Hypermethylation of specific microRNA genes in MLL-rearranged infant acute lymphoblastic leukemia: major matters at a micro scale
.
Leukemia
.
2011
;
25
(
3
):
429
-
439
.
75.
Nishi
M
,
Eguchi-Ishimae
M
,
Wu
Z
, et al
.
Suppression of the let-7b microRNA pathway by DNA hypermethylation in infant acute lymphoblastic leukemia with MLL gene rearrangements
.
Leukemia
.
2013
;
27
(
2
):
389
-
397
.
76.
Bhatla
T
,
Wang
J
,
Morrison
DJ
, et al
.
Epigenetic reprogramming reverses the relapse-specific gene expression signature and restores chemosensitivity in childhood B-lymphoblastic leukemia
.
Blood
.
2012
;
119
(
22
):
5201
-
5210
.
77.
Stumpel
DJ
,
Schneider
P
,
Seslija
L
, et al
.
Connectivity mapping identifies HDAC inhibitors for the treatment of t(4;11)-positive infant acute lymphoblastic leukemia
.
Leukemia
.
2012
;
26
(
4
):
682
-
692
.
78.
Cruickshank
MN
,
Ford
J
,
Cheung
LC
, et al
.
Systematic chemical and molecular profiling of MLL-rearranged infant acute lymphoblastic leukemia reveals efficacy of romidepsin
.
Leukemia
.
2017
;
31
(
1
):
40
-
50
.
79.
Garrido Castro
P
,
van Roon
EHJ
,
Pinhanços
SS
, et al
.
The HDAC inhibitor panobinostat (LBH589) exerts in vivo anti-leukaemic activity against MLL-rearranged acute lymphoblastic leukaemia and involves the RNF20/RNF40/WAC-H2B ubiquitination axis
.
Leukemia
.
2018
;
32
(
2
):
323
-
331
.
80.
Burke
MJ
,
Brown
P
,
Gore
L
, et al
.
Invasive Candida infections in pediatric patients treated on the pilot study of decitabine and vorinostat with chemotherapy for relapsed ALL: a report from the Therapeutic Advances in Childhood Leukemia & Lymphoma (TACL) Consortium [abstract]
.
Blood
.
2014
;
124
(
21
). Abstract
3650
.
81.
Sison
EA
,
Rau
RE
,
McIntyre
E
,
Li
L
,
Small
D
,
Brown
P
.
MLL-rearranged acute lymphoblastic leukaemia stem cell interactions with bone marrow stroma promote survival and therapeutic resistance that can be overcome with CXCR4 antagonism
.
Br J Haematol
.
2013
;
160
(
6
):
785
-
797
.
82.
Sison
EA
,
McIntyre
E
,
Magoon
D
,
Brown
P
.
Dynamic chemotherapy-induced upregulation of CXCR4 expression: a mechanism of therapeutic resistance in pediatric AML
.
Mol Cancer Res
.
2013
;
11
(
9
):
1004
-
1016
.
83.
Sison
EA
,
Magoon
D
,
Li
L
, et al
.
Plerixafor as a chemosensitizing agent in pediatric acute lymphoblastic leukemia: efficacy and potential mechanisms of resistance to CXCR4 inhibition
.
Oncotarget
.
2014
;
5
(
19
):
8947
-
8958
.
84.
Sison
EA
,
Magoon
D
,
Li
L
, et al
.
POL5551, a novel and potent CXCR4 antagonist, enhances sensitivity to chemotherapy in pediatric ALL
.
Oncotarget
.
2015
;
6
(
31
):
30902
-
30918
.
85.
Cooper
TM
,
Sison
EAR
,
Baker
SD
, et al
.
A phase 1 study of the CXCR4 antagonist plerixafor in combination with high-dose cytarabine and etoposide in children with relapsed or refractory acute leukemias or myelodysplastic syndrome: a Pediatric Oncology Experimental Therapeutics Investigators’ Consortium study (POE 10-03)
.
Pediatr Blood Cancer
.
2017
;
64
(
8
):
e26414
.
86.
von Stackelberg
A
,
Locatelli
F
,
Zugmaier
G
, et al
.
Phase I/phase II study of blinatumomab in pediatric patients with relapsed/refractory acute lymphoblastic leukemia
.
J Clin Oncol
.
2016
;
34
(
36
):
4381
-
4389
.
87.
Maude
SL
,
Laetsch
TW
,
Buechner
J
, et al
.
Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia
.
N Engl J Med
.
2018
;
378
(
5
):
439
-
448
.
88.
Rayes
A
,
McMasters
RL
,
O’Brien
MM
.
Lineage switch in MLL-rearranged infant leukemia following CD19-directed therapy
.
Pediatr Blood Cancer
.
2016
;
63
(
6
):
1113
-
1115
.
89.
Mejstríková
E
,
Hrusak
O
,
Borowitz
MJ
, et al
.
CD19-negative relapse of pediatric B-cell precursor acute lymphoblastic leukemia following blinatumomab treatment
.
Blood Cancer J
.
2017
;
7
(
12
):
659
.
90.
Gardner
R
,
Wu
D
,
Cherian
S
, et al
.
Acquisition of a CD19-negative myeloid phenotype allows immune escape of MLL-rearranged B-ALL from CD19 CAR-T-cell therapy
.
Blood
.
2016
;
127
(
20
):
2406
-
2410
.
91.
Aoki
Y
,
Watanabe
T
,
Saito
Y
, et al
.
Identification of CD34+ and CD34- leukemia-initiating cells in MLL-rearranged human acute lymphoblastic leukemia
.
Blood
.
2015
;
125
(
6
):
967
-
980
.
92.
le Viseur
C
,
Hotfilder
M
,
Bomken
S
, et al
.
In childhood acute lymphoblastic leukemia, blasts at different stages of immunophenotypic maturation have stem cell properties
.
Cancer Cell
.
2008
;
14
(
1
):
47
-
58
.
93.
Cox
CV
,
Evely
RS
,
Oakhill
A
,
Pamphilon
DH
,
Goulden
NJ
,
Blair
A
.
Characterization of acute lymphoblastic leukemia progenitor cells
.
Blood
.
2004
;
104
(
9
):
2919
-
2925
.
94.
Shah
NN
,
Stevenson
MS
,
Yuan
CM
, et al
.
Characterization of CD22 expression in acute lymphoblastic leukemia
.
Pediatr Blood Cancer
.
2015
;
62
(
6
):
964
-
969
.
95.
Qasim
W
,
Zhan
H
,
Samarasinghe
S
, et al
.
Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells [published correction appears in Sci Transl Med. 2017;9(377)]
.
Sci Transl Med
.
2017
;
9
(
374
).
96.
Driessen
EM
,
van Roon
EH
,
Spijkers-Hagelstein
JA
, et al
.
Frequencies and prognostic impact of RAS mutations in MLL-rearranged acute lymphoblastic leukemia in infants
.
Haematologica
.
2013
;
98
(
6
):
937
-
944
.
97.
Emerenciano
M
,
Barbosa
TC
,
de Almeida Lopes
B
,
Meyer
C
,
Marschalek
R
,
Pombo-de-Oliveira
MS
.
Subclonality and prenatal origin of RAS mutations in KMT2A (MLL)-rearranged infant acute lymphoblastic leukaemia
.
Br J Haematol
.
2015
;
170
(
2
):
268
-
271
.
98.
Trentin
L
,
Bresolin
S
,
Giarin
E
, et al
.
Deciphering KRAS and NRAS mutated clone dynamics in MLL-AF4 paediatric leukaemia by ultra deep sequencing analysis
.
Sci Rep
.
2016
;
6
(
1
):
34449
.
99.
Kerstjens
M
,
Driessen
EM
,
Willekes
M
, et al
.
MEK inhibition is a promising therapeutic strategy for MLL-rearranged infant acute lymphoblastic leukemia patients carrying RAS mutations
.
Oncotarget
.
2017
;
8
(
9
):
14835
-
14846
.
100.
Kerstjens
M
,
Pinhancos
SS
,
Castro
PG
, et al
.
Trametinib inhibits RAS-mutant MLL-rearranged acute lymphoblastic leukemia at specific niche sites and reduces ERK phosphorylation in vivo
.
Haematologica
.
2018
;
103
(
4
):
e147
-
e150
.
Sign in via your Institution