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 unique biology that is increasingly pointing the way toward novel therapeutic approaches. This review highlights the key clinical, pathologic, and epidemiologic features of infant leukemia, including the high frequency of mixed lineage leukemia (MLL) gene rearrangements. The state of the art with regard to current approaches to risk stratified treatment of infant leukemia in the major international cooperative groups is discussed. Finally, exciting recent discoveries elucidating the molecular biology of infant leukemia are reviewed and novel targeted therapeutic strategies, including FLT3 inhibition and modulation of aberrant epigenetic programs, are suggested.

Leukemia in infants is among the most vexing clinical problems in pediatric hematology/oncology. It is so rare that even the largest pediatric leukemia centers may see only a handful of cases per year and most centers will go a year or more between cases, precluding the widespread acquisition of expertise that occurs with more common diagnoses. Infants with leukemia tend to present with aggressive clinical features that make initial management particularly challenging, and this difficulty is amplified by the vulnerability of infants to complications and toxicity of the necessary procedures and treatments. Perhaps most importantly, the eventual outcomes for infants with leukemia in terms of relapse-free survival are poor compared with leukemia in older children despite the use of maximally intensified standard therapies (chemotherapy with or without hematopoietic stem cell transplantation [HSCT]). As daunting as these challenges are, there is real promise that better days lie ahead for infants with leukemia. The fascinating and ever-expanding discoveries regarding the unique molecular biology of infant leukemia are leading to novel therapeutic strategies that have generated exciting preclinical results and are advancing to clinical trials. There is a palpable sense of hope among caregivers for infants with leukemia that the emerging era of molecularly targeted therapy will be especially beneficial for these patients. Turning this hope into reality will require close collaboration between molecular biologists and clinical trialists and among the international cooperative groups that have painstakingly established the infrastructure for the systematic treatment of these patients.

The term “infant leukemia” generally refers to acute lymphoblastic leukemia (ALL) or acute myeloid leukemia (AML) diagnosed in a child before 1 year of age. The estimated incidence of acute leukemia in infants is 41 cases per million in the United States, which equates to ∼ 160 cases of infant leukemia per year, with ∼ 90 of the cases being ALL and 70 AML. Neuroblastoma and brain tumors occur with similar frequency as acute leukemia in the infant population. The incidence of ALL in infants is significantly lower than in children aged 1 to 14 years old and approximately the same as adolescents. In contrast, the incidence of AML in infants is approximately twice that of older children and adolescents. Interestingly, females have a higher risk of developing infant leukemia than males but a lower risk of developing leukemia beyond the first birthday.1 

Compared with older children, infants with acute leukemia tend to present with more aggressive features, including high WBC counts, hepatosplenomegaly, CNS involvement, and leukemia cutis (skin infiltration).2,3  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  Conversely, outcomes for infants with AML are similar to those for older children.2 

A high proportion of acute leukemias occurring in infants are characterized cytogenetically by balanced chromosomal translocations involving the mixed lineage leukemia (MLL) gene at chromosome 11q23. MLL rearrangements (MLL-r) occur in ∼ 5% of childhood ALL cases overall,7  but in 70% to 80% of ALL in infants.3,4  In childhood AML, MLL-r is more common overall (15%-20%), but is also particularly common in the infant age group (∼ 50%).8 

MLL-r results in the fusion of the N-terminus of the MLL gene with the C-terminus of a partner gene. Remarkably, 79 different MLL partner genes have now been identified.9  In infant ALL, 4 partner genes account for 93% of cases: AF4 (49%), ENL (22%), AF9 (17%), and AF10 (5%). In infant AML, 3 partner genes account for 66% of cases: AF9 (22%), AF10 (27%), and ELL (17%).

Various lines of evidence (eg, retrospective analyses of neonatal samples and twin concordance studies) have shown that MLL rearrangements are acquired in hematopoietic precursors in utero, and this initiates rapid progression to full blown leukemia. One intriguing aspect of leukemia epidemiology is that MLL-r leukemias occur with high frequency in 2 very different clinical situations: infants with de novo acute leukemia and 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-control studies10,11  and laboratory studies,12  that maternal exposure to environmental DNAt2 inhibitors (eg, dietary flavonoids) during pregnancy may contribute to the risk of MLL-r infant leukemia. Germline genetic susceptibility may also play a role, because candidate gene studies13,14  and genome-wide association studies15  have identified several single nucleotide polymorphisms that are correlated with risk of infant leukemia.

In ALL, MLL-r is associated with CD10 negativity and coexpression of one or more myeloid antigens, suggesting that these leukemias arise from very immature lymphoid progenitors.16  In AML, MLL-r is associated with monocytic differentiation.2 

The presence of MLL-r in infant acute leukemia has different prognostic implications in ALL than in AML. In infant ALL, MLL-r is clearly associated with poorer outcome. In the Children's Cancer Group protocol CCG-1953, the 5-year EFS for MLL-r infants was 34% compared with 60% with germline (wild-type) MLL (MLL-g).3  In Interfant-99, the 4-year EFS in MLL-r and MLL-g infants was 37% and 74%, respectively.4  Conversely, in infant AML, MLL-r is not a significant risk factor. In a combined analysis of AML-BFM-98 and AML-BFM-2004, the 5-year EFS was 43% and 52% for MLL-r and MLL-g infants, respectively (P = .59).2 

Among infants with MLL-r ALL, several additional independent prognostic factors have been identified. The most important of these are age and WBC at diagnosis, with the younger infants and those with the higher WBC having poorer outcomes.3,4,17  In the context of a 7-day “prophase” of single-agent prednisone given before intensive induction chemotherapy in the Interfant-99 protocol, a poor response (≥ 1000 blasts/μL in the peripheral blood on day 8) was also an independent negative prognostic factor.4 

Poor response to prednisone is significantly more common in infants than in older children with ALL, which has raised the question of whether the poor outcome for infants with leukemia may be partly due to enhanced chemoresistance. Indeed, infant MLL-r ALL cells demonstrate enhanced in vitro resistance to corticosteroids and asparaginase in assays using short-term exposure of bulk leukemia populations. Conversely, infant MLL-r AML cells do not demonstrate a more resistant phenotype.18-20  It is somewhat counterintuitive, then, that the typical pattern of failure for infant MLL-r ALL is to achieve rapid complete remission with induction chemotherapy, but then relapse during the first year of therapy. This would suggest that the poor outcomes are due primarily to the emergence of a chemoresistant population over time. The low rates of second remission reported by the Japanese Pediatric Leukemia Study Group (JPLSG) in patients relapsing after treatment for infant ALL are consistent with this hypothesis.21 

Another challenging aspect of the treatment of infants with acute leukemia is the infant patient's unique vulnerability to complications and toxicities. There are several complex physiologic processes that undergo rapid changes during the first year of life and there are very limited data to guide how the distinct 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. It is perhaps not surprising, then, that infant leukemia protocols have encountered problems with excessive toxicity. In the Children's Oncology Group (COG) infant ALL protocol P9407, for example, death from toxicity (primarily infectious) within the first 90 days of enrollment occurred in 25% of the first 68 patients. After the study was amended to substitute prednisone for dexamethasone, reduce the dose of daunorubicin, and enhance supportive care, the early death rate dropped to 6% for the next 141 patients.22  Remarkably, the early death rate increased again on the successor COG trial AALL0631, with 4 of the first 26 patients (15%) dying from infections, although the only change to induction therapy was to substitute a single dose of PEG asparaginase for native E coli asparaginase.23  Similar issues have been reported in infants with AML. Induction mortality on the Medical Research Council (MRC) protocols AML10 and AML12 was 12% and 3% for infants and older children, respectively.24  Survivors of infant leukemia also demonstrate an increased risk of late effects, particularly in cases in which treatment included cranial radiation or HSCT.25 

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 HSCT (for patients with unfavorable prognostic features). Cases of AML in infants are relatively unlikely to harbor cytogenetic or molecular abnormalities that confer unfavorable risk or favorable risk. Therefore, infants usually fall into the intermediate cytogenetic/molecular risk group, for which current stratification into favorable or unfavorable prognostic groups is dictated by end-induction minimal residual disease testing.

In contrast, treatment for infant ALL is quite different from that for childhood ALL generally. Currently, there are 3 major cooperative groups conducting specific clinical trials for infant ALL: Interfant (Interfant-06), COG (AALL0631), and JPLSG (MLL-10). All have adopted an identical induction strategy based on Interfant-99.4  All are using a prospective risk-stratified approach that incorporates MLL rearrangement status. Table 1 summarizes the key features of the 3 trials.

Table 1.
graphic
graphic

PPR indicates prednisone poor response; MRD, minimal residual disease; Protocol IB, cyclophosphamide, cytarabine, 6-mercaptopurine consolidation; ADE/MAE, cytarabine, daunorubicin, etoposide/mitoxantrone, cytarabine, etoposide consolidation; and TKI, tyrosine kinase inhibitor.

Interfant-06 is testing whether consolidation with “myeloid”-style chemotherapy with cytarabine, daunorubicin/mitoxantrone, and etoposide will prove superior to “lymphoid”-style consolidation with cyclophosphamide, cytarabine, and 6-mercaptopurine in MLL-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 is testing whether the addition of a FLT3 tyrosine kinase inhibitor to postinduction chemotherapy will enhance the effectiveness of chemotherapy based on data showing aberrant activation of the FLT3 pathway in MLL-r ALL (detailed below). The use of HSCT varies between the groups on these trials, reflecting the controversy that exists regarding the risk/benefit ratio of HSCT in this population. Sison reviews this important topic in the accompanying evidence-based review elsewhere in this publication.

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 MLL-r leukemias detailed in the following section.

MLL-r infant ALL is characterized by a distinct global gene expression profile.26,27  A notable component of this profile is striking overexpression of FLT3.28  FLT3 signaling is constitutively activated in these cases either by activating mutations29,30  or, more commonly, by autocrine activation by coexpressed FLT3 ligand.31  Moreover, FLT3 tyrosine kinase inhibition results in selective killing of these samples and synergizes with chemotherapy in a sequence-dependent manner.30-33  FLT3 overexpression has been shown to confer especially poor prognosis in MLL-r infant ALL.34,35  As discussed above, the ongoing COG trial AALL0631 is the first to incorporate a novel, molecularly targeted agent into frontline treatment of MLL-r infant ALL. A dose that is safe in combination with intensive chemotherapy and results in sustained pharmacodynamic FLT3 inhibition has been successfully determined,36  and a randomized evaluation of efficacy is ongoing. 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 by Interfant, COG, and JPLSG.

Genomic single nucleotide polymorphism and sequencing studies have revealed a striking paucity of cooperating genetic alterations in infant MLL-r ALL compared with all other subsets of childhood ALL.37  Accordingly, MLL-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.

The acquisition of a reciprocal MLL translocation initiates transformation in utero by the aberrant recruitment of multiprotein complexes with chromatin-modifying activity to MLL target genes via myriad translocation partner genes, inducing dysregulated transcription of multiple genes and a highly characteristic global gene expression profile. Several studies have now established that a required component of this aberrant epigenetic state and MLL-r leukemogenesis is the H3K79 methyltransferase DOT1L.38-41  It is of particular interest, then, that potent and highly selective small-molecule inhibitors of DOT1L are in clinical development after having shown promising activity in preclinical models of MLL-r leukemia.42,43 

BRD4 is one of several 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.44  Potent and selective small-molecule inhibitors of BRD4 binding down-regulated characteristic MLL-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 MLL-r infant ALL cells.44,45  A phase 1 clinical trial in adults with hematologic malignancies with a BRD2/3/4 inhibitor (OTX015) is under way and others are in development.

Complementing the widespread gene activation in MLL-r ALL is the epigenetic silencing of a specific set of genes with tumor suppressor function via promoter region CpG island hypermethylation and associated repressive histone modifications.46,47  A high level of promoter hypermethylation was correlated with inferior survival in Interfant-99.47  Demethylating agents such as decitabine and zebularine preferentially kill MLL-r lymphoblastic leukemia cells and this is correlated with the up-regulation of several of the identified silenced genes.46,47  Promoter hypermethylation and transcriptional silencing of certain microRNAs with likely tumor suppressor function also characterize infant MLL-r ALL cases.48,49  Of particular interest is hypermethylation of miR-152, which correlates with a poor prognosis in MLL-AF4+ infant ALL.48 

Synergistic reexpression of epigenetically silenced genes has been demonstrated by combining CpG demethylation with reversal of repressive chromatin states. Deacetylation of histone marks such as H3K9/14 is associated with gene silencing and can be modulated with histone deacetylase (HDAC) inhibitors. Two recent applications of the connectivity map concept have identified HDAC inhibitors as capable of reversing epigenetically determined global gene expression profiles associated with chemotherapy resistance, one in relapsed childhood ALL50  and one in infant MLL-r ALL.51  These findings support the testing of demethylating agents and HDAC inhibitors as a strategy to reverse the inherent chemoresistance of MLL-r infant ALL. A pilot study of decitabine and vorinostat in combination with reinduction chemotherapy in children with relapsed ALL is under way in the Therapeutic Advances in Childhood Leukemia (TACL) consortium and will hopefully lay the groundwork for subsequent frontline trials in infant ALL.

As noted above, although the majority of infants with ALL can achieve remission, most will suffer disease recurrence with a short latency to relapse. This suggests that chemotherapy-resistant leukemia stem cells survive and can recapitulate the leukemia. Interactions between infant MLL-r ALL leukemia stem cells and the BM stromal microenvironment via the CXCR4/SDF-1 axis have been shown to mediate survival and therapeutic resistance in MLL-r ALL.52  Moreover, in xenograft models of MLL-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 up-regulation of CXCR4 expression on the surface of acute leukemias (including MLL-r) in response to cytotoxic chemotherapy may enhance this stroma-mediated resistance and serve as a biomarker that can identify subsets of infants with leukemia for whom CXCR4 inhibition may be particularly effective.53 

It is notable that most of the therapeutically relevant biological insights described above are specific to infant ALL. This reflects a relative paucity of published data suggesting that AML in infants represents a biologic entity distinct from AML in older children.

Infant leukemia is one of most difficult clinical situations encountered in pediatric hematology/oncology. Standard approaches with maximally intensive and toxic regimens of chemotherapy and HSCT are curative in a minority of patients. 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.

Conflict-of-interest disclosure: The author has consulted for Epizyme. Off-label drug use: decitabine and vorinostat.

Patrick Brown, MD, Johns Hopkins Oncology, 1650 Orleans Street, CRB1 Room 2M49, Baltimore, MD 21231; Phone: 410-955-8817; Fax: 410-955-8897; e-mail: pbrown2@jhmi.edu.

1
Howlader
 
N
Noone
 
AM
Krapcho
 
M
, et al. 
SEER Cancer Statistics Review, 1975-2010
2013
Bethesda
National Cancer Institute
 
[Based on November 2012 SEER data submission posted to the SEER Web site, April 2013.]
2
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
, vol. 
26
 
4
(pg. 
654
-
661
)
3
Hilden
 
JM
Dinndorf
 
PA
Meerbaum
 
SO
, et al. 
Analysis of prognostic factors of acute lymphoblastic leukemia in infants: report on CCG 1953 from the Children's Oncology Group
Blood
2006
, vol. 
108
 
2
(pg. 
441
-
451
)
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
, vol. 
370
 
9583
(pg. 
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
, vol. 
30
 
14
(pg. 
1663
-
1669
)
6
Pui
 
CH
Campana
 
D
Pei
 
D
, et al. 
Treating childhood acute lymphoblastic leukemia without cranial irradiation
N Engl J Med
2009
, vol. 
360
 
26
(pg. 
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
, vol. 
87
 
7
(pg. 
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
, vol. 
28
 
16
(pg. 
2674
-
2681
)
9
Meyer
 
C
Hofmann
 
J
Burmeister
 
T
, et al. 
The MLL recombinome of acute leukemias in 2013
Leukemia
 
Published online ahead of print April 20, 2013. doi: 10.1038/leu.2013.135
10
Alexander
 
FE
Patheal
 
SL
Biondi
 
A
, et al. 
Transplacental chemical exposure and risk of infant leukemia with MLL gene fusion
Cancer Res
2001
, vol. 
61
 
6
(pg. 
2542
-
2546
)
11
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
, vol. 
14
 
3
(pg. 
651
-
655
)
12
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 U S A
2000
, vol. 
97
 
9
(pg. 
4790
-
4795
)
13
Wiemels
 
JL
Smith
 
RN
Taylor
 
GM
Eden
 
OB
Alexander
 
FE
Greaves
 
MF
Methylenetetrahydrofolate reductase (MTHFR) polymorphisms and risk of molecularly defined subtypes of childhood acute leukemia
Proc Natl Acad Sci U S A
2001
, vol. 
98
 
7
(pg. 
4004
-
4009
)
14
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
, vol. 
100
 
13
(pg. 
4590
-
4593
)
15
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
, vol. 
60
 
1
(pg. 
31
-
34
)
16
Basso
 
G
Rondelli
 
R
Covezzoli
 
A
Putti
 
M
The role of immunophenotype in acute lymphoblastic leukemia of infant age
Leuk Lymphoma
1994
, vol. 
15
 
1-2
(pg. 
51
-
60
)
17
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
, vol. 
21
 
11
(pg. 
2258
-
2263
)
18
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
, vol. 
18
 
3
(pg. 
521
-
529
)
19
Palle
 
J
Frost
 
BM
Forestier
 
E
, et al. 
Cellular drug sensitivity in MLL-rearranged childhood acute leukaemia is correlated to partner genes and cell lineage
Br J Haematol
2005
, vol. 
129
 
2
(pg. 
189
-
198
)
20
Zwaan
 
CM
Kaspers
 
GJ
Pieters
 
R
, et al. 
Cellular drug resistance in childhood acute myeloid leukemia is related to chromosomal abnormalities
Blood
2002
, vol. 
100
 
9
(pg. 
3352
-
3360
)
21
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
, vol. 
52
 
7
(pg. 
808
-
813
)
22
Salzer
 
WL
Jones
 
TL
Devidas
 
M
, et al. 
Modifications to induction therapy decrease risk of early death in infants with acute lymphoblastic leukemia treated on Children's Oncology Group P9407
Pediatr Blood Cancer
2012
, vol. 
59
 
5
(pg. 
834
-
839
)
23
Salzer
 
W
Jones
 
T
Dreyer
 
Z
, et al. 
Decreased induction morbidity and mortality with changes to induction therapy in infants with acute lymphoblastic leukemia enrolled on Children's Oncology Group (COG) trial AALL0631
Pediatr Blood Cancer
2012
, vol. 
58
 
7
pg. 
418a
 
24
Gibson
 
BE
Wheatley
 
K
Hann
 
IM
, et al. 
Treatment strategy and long-term results in paediatric patients treated in consecutive UK AML trials
Leukemia
2005
, vol. 
19
 
12
(pg. 
2130
-
2138
)
25
Leung
 
W
Hudson
 
M
Zhu
 
Y
, et al. 
Late effects in survivors of infant leukemia
Leukemia
2000
, vol. 
14
 
7
(pg. 
1185
-
1190
)
26
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
, vol. 
119
 
8
(pg. 
1872
-
1881
)
27
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
, vol. 
115
 
14
(pg. 
2835
-
2844
)
28
Armstrong
 
SA
Staunton
 
JE
Silverman
 
LB
, et al. 
MLL translocations specify a distinct gene expression profile that distinguishes a unique leukemia
Nat Genet
2002
, vol. 
30
 
1
(pg. 
41
-
47
)
29
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
, vol. 
103
 
3
(pg. 
1085
-
1088
)
30
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
, vol. 
3
 
2
(pg. 
173
-
183
)
31
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
, vol. 
105
 
2
(pg. 
812
-
820
)
32
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
, vol. 
20
 
8
(pg. 
1368
-
1376
)
33
Stam
 
RW
den Boer
 
ML
Schneider
 
P
, et al. 
Targeting FLT3 in primary MLL-gene-rearranged infant acute lymphoblastic leukemia
Blood
2005
, vol. 
106
 
7
(pg. 
2484
-
2490
)
34
Chillon
 
MC
Gomez-Casares
 
MT
Lopez-Jorge
 
CE
, et al. 
Prognostic significance of FLT3 mutational status and expression levels in MLL-AF4+ and MLL-germline acute lymphoblastic leukemia
Leukemia
2012
, vol. 
26
 
11
(pg. 
2360
-
2366
)
35
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
, vol. 
110
 
7
(pg. 
2774
-
2775
)
36
Sun
 
D
Kaeding
 
A
Magoon
 
D
, et al. 
Safety and biological activity of the FLT3 inhibitor lestaurtinib in infant MLL-rearranged (MLL-r) ALL: Children's Oncology Group protocol AALL0631
ASCO Meeting Abstracts
2012
, vol. 
30
 
15 Suppl
pg. 
9548
 
37
Mullighan
 
CG
Goorha
 
S
Radtke
 
I
, et al. 
Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia
Nature
2007
, vol. 
446
 
7137
(pg. 
758
-
764
)
38
Deshpande
 
AJ
Chen
 
L
Fazio
 
M
, et al. 
Leukemic transformation by the MLL-AF6 fusion oncogene requires the H3K79 methyltransferase Dot1l
Blood
2013
, vol. 
121
 
13
(pg. 
2533
-
2541
)
39
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
, vol. 
27
 
4
(pg. 
813
-
822
)
40
Bernt
 
KM
Zhu
 
N
Sinha
 
AU
, et al. 
MLL-rearranged leukemia is dependent on aberrant H3K79 methylation by DOT1L
Cancer Cell
2011
, vol. 
20
 
1
(pg. 
66
-
78
)
41
Nguyen
 
AT
Taranova
 
O
He
 
J
Zhang
 
Y
DOT1L, the H3K79 methyltransferase, is required for MLL-AF9-mediated leukemogenesis
Blood
2011
, vol. 
117
 
25
(pg. 
6912
-
6922
)
42
Daigle
 
SR
Olhava
 
EJ
Therkelsen
 
CA
, et al. 
Potent inhibition of DOT1L as treatment for MLL-fusion leukemia
Blood
2013
, vol. 
122
 
6
(pg. 
1017
-
1025
)
43
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
, vol. 
20
 
1
(pg. 
53
-
65
)
44
Zuber
 
J
Shi
 
J
Wang
 
E
, et al. 
RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia
Nature
2011
, vol. 
478
 
7370
(pg. 
524
-
528
)
45
Dawson
 
MA
Prinjha
 
RK
Dittmann
 
A
, et al. 
Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia
Nature
2011
, vol. 
478
 
7370
(pg. 
529
-
533
)
46
Schafer
 
E
Irizarry
 
R
Negi
 
S
, et al. 
Promoter hypermethylation in MLL-r infant acute lymphoblastic leukemia: biology and therapeutic targeting
Blood
2010
, vol. 
115
 
23
(pg. 
4798
-
4809
)
47
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
, vol. 
114
 
27
(pg. 
5490
-
5498
)
48
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
, vol. 
25
 
3
(pg. 
429
-
439
)
49
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
, vol. 
27
 
2
(pg. 
389
-
397
)
50
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
, vol. 
119
 
22
(pg. 
5201
-
5210
)
51
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
, vol. 
26
 
4
(pg. 
682
-
692
)
52
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
, vol. 
160
 
6
(pg. 
785
-
797
)
53
Sison
 
EA
McIntyre
 
E
Magoon
 
D
Brown
 
P
Dynamic chemotherapy-induced up-regulation of surface CXCR4 expression as a mechanism of chemotherapy resistance in pediatric acute myeloid leukemia
Mol Cancer Res
2013
, vol. 
11
 
9
(pg. 
1004
-
1016
)