Rearrangements of the Mixed-Lineage Leukemia (MLL) gene are found in > 70% of infant leukemia, ∼ 10% of adult acute myelogenous leukemia (AML), and many cases of secondary acute leukemias. The presence of an MLL rearrangement generally confers a poor prognosis. There are more than 60 known fusion partners of MLL having some correlation with disease phenotype and prognosis. The most common fusion proteins induce the inappropriate expression of homeotic (Hox) genes, which, during normal hematopoiesis, are maintained by wild-type MLL. MLL-rearranged leukemias display remarkable genomic stability, with very few gains or losses of chromosomal regions. This may be explained by recent studies suggesting that MLL-rearranged leukemias are largely driven by epigenetic dysregulation. Several epigenetic regulators that modify DNA or histones have been implicated in MLL-fusion driven leukemogenesis, including DNA methylation, histone acetylation, and histone methylation. The histone methyltransferase DOT1L has emerged as an important mediator of MLL-fusion–mediated leukemic transformation. The clinical development of targeted inhibitors of these epigenetic regulators may therefore hold promise for the treatment of MLL-rearranged leukemia.

Leukemias bearing translocations involving chromosome 11q23 are of particular interest due to unique clinical and biological characteristics. 11q23 rearrangements are found in > 70% of leukemias in infants < 1 year of age whether the immunophenotype is more consistent with acute lymphoblastic leukemia (ALL) or acute myelogenous leukemia (AML).1  Infants diagnosed with ALL harboring an 11q23 translocation have a particularly poor prognosis, with an overall survival of ∼ 50%, whereas children with ALL that does not harbor a MLL translocation have an overall survival of > 80%. In contrast to other poor-prognosis ALL, the prognosis of most infant MLL-rearranged leukemia is not improved by the use of allogeneic stem cell transplantation. A subgroup of infants with additional high-risk features may benefit from hematopoietic stem cell transplantation (HSCT) during the first complete remission, but the 5-year event-free and overall survival rates in this group remain in the 50% range.2  Leukemias that develop as a result of treatment with topoisomerase II inhibitors frequently harbor 11q23 rearrangements and also have a poor prognosis.3  Some infant leukemias express cell-surface antigens characteristic of both lymphoblasts and monoblasts, and are often designated mixed phenotype acute leukemia (MPAL). The association of 11q23 rearrangements with ALL, AML, or MPAL is unique in that most other chromosomal rearrangements tend to be associated with leukemias of a particular hematopoietic lineage. These observations led to the name Mixed-Lineage Leukemia (MLL) for the gene that resides on 11q23.

The most common MLL translocation found in lymphoblastic leukemias is t(4;11)(q21;q23). Multiple investigators cloned the MLL-AF4 gene formed as a result of this translocation in the early 1990s.4–7 MLL-AF4 encodes a protein of 2304 amino acids, with the NH2-terminal 1439 amino acids derived from MLL on chromosome 11 and the COOH-terminal 865 amino acids from the AF4 gene on chromosome 4. Subsequently, > 60 different translocations have been identified, all of which appear to produce a fusion protein possessing the NH2-terminus of MLL fused in-frame to the COOH-terminus of the fusion partner.8,9  Whereas MLL translocations can be found in either ALL or AML, particular translocations show lineage specificity, with the t(4;11) found most often in ALL, the t(9;11)(p21;q23) found most often in AML, and the t(11;16) found most often in myelodysplastic syndrome and secondary leukemia. A recent retrospective analysis demonstrated that the prognosis of MLL-rearranged leukemias may also be influenced by the fusion partner. Survival associated with the rare t(1;11)(q21;q23) translocation was good, in contrast to very poor outcomes in with the more frequent t(4;11)(q21;q23), t(10;11)(p12;q23) (MLL-AF10), t(10;11)(p11.2;q23) (MLL-ABI-1), and t(6;11)(q27;q23) translocations in this series.10  This points to the translocation partner as having a role in the disease phenotype and functional heterogeneity of MLL fusions, but the molecular details of these associations are unclear.

The MLL gene, located on the human chromosome 11q23, is the ortholog of the Drosophila melanogaster trithorax gene.4–7  Trithorax group (trxG) and polycomb protein (PcG) genes were discovered in Drosophila as activators and repressors of homeobox (Hox) genes. Hox genes are a group of transcription factors involved in the development of cell identity and anteroposterior patterning in mammals, which also play a critical role in hematopoiesis. Mll is also fundamentally involved in the development of the axial skeleton and the hematopoietic system, at least in part via direct regulation of Hox gene expression.11–13  The MLL gene encodes a 3969–amino acid DNA-binding protein that possesses multiple recognizable protein motifs, including an NH2-terminal DNA-binding domain, transcriptional activation and repression domains, and a COOH-terminal SET domain that contains histone H3 lysine 4 (H3K4) methyltransferase activity.14,15  Recent biochemical studies have identified MLL as a member of a large multiprotein complex that contains proteins involved in chromatin modification/remodeling. The complex notably includes histone deacetylases (HDACs) and members of the Swi/Snf chromatin-remodeling complex.15  In addition, MLL is recruited to the promoters of select cell cycle–regulatory genes by the protein product of the MEN1 tumor-suppressor gene, suggesting a role for MLL in tumor suppression and cell-cycle control.16–18  These data support the hypothesis that the MLL protein regulates gene expression and cell-cycle control via chromatin modification.

Analysis of Mll-knockout mice suggests that Mll plays an important role in development and hematopoiesis through maintenance of appropriate homeotic (Hox) gene expression.11,19  Detailed studies assessing the specific role of Mll in hematopoietic development have shown that Mll is necessary for definitive hematopoiesis and expansion of hematopoietic progenitors and stem cells found in the aorta-gonad-mesonephros region of the developing embryo.13,20  In addition, Mll plays a critical role in adult hematopoietic stem cells.21  The defect in hematopoietic progenitor expansion can be rescued by reexpression of Hox genes, confirming the importance of Mll-mediated Hox gene expression during hematopoiesis.

Multiple studies have demonstrated the ability of Hox genes to induce leukemia in mice,22  and the t(7;11)(p15;p15) translocation found in some human AML cases results in a fusion of the HOXA9 gene to the nucleoporin NUP98.23,24  Given the apparent importance of Hox genes in leukemogenesis, and the fact that MLL-fusion proteins generated by MLL translocations activate Hox gene expression (as described below), hox gene activation is likely a critical part of the leukemogenic program driven by MLL-fusion proteins. Further support for HOX genes as central regulators of MLL-induced leukemogenesis comes from gene-expression studies finding multiple HOXA cluster genes more highly expressed in MLL-rearranged myelogenous and lymphoblastic leukemias compared with MLL-germline leukemias.25–28 

Gene-expression studies of human MLL-rearranged B-precursor ALL demonstrated that hundreds of genes are differentially expressed compared with other B-precursor ALLs.25–27,29  Based on the magnitude of the differences in gene expression, MLL translocations appear to specify a unique lymphoblastic leukemia. Other large gene-expression studies have also shown that ALLs with distinct chromosomal rearrangements have unique gene-expression profiles, which provides support for this concept.27  The genes that are relatively highly expressed in MLL-rearranged B-precursor ALL are those associated with hematopoietic progenitors and developing myeloid cells, whereas the genes expressed at lower level are genes associated with lymphoid identity. Specific gene-expression signatures have also been associated with MLL translocations in primary human AML blasts.28  Even though there are clear differences between MLL-rearranged ALL and MLL-rearranged AML in the expression of lineage-associated genes, there appears to be a core gene-expression profile found in all MLL-rearranged human leukemias independent of the lineage markers.28  Presumably, MLL-fusion proteins directly regulate a subset of these genes. This is further supported by the fact that MLL-associated signatures across all studies and phenotypes contain multiple highly expressed HOX genes.

Mouse models of leukemia predict multiple genetic events are necessary for the development of acute leukemias. “Knock-in” models of MLL-rearranged AML have been developed, in which the fusion genes generated by translocations found in human leukemias are under the control of the endogenous promoter. Mice containing an MLL-AF9 fusion gene under control of the Mll promoter spontaneously develop AML with a latency of 4 months to > 1 year.30  This is widely interpreted as a requirement for a second genetic event during leukemogenesis. A multistep model of leukemogenesis is further supported by mouse models such as an Mll-Cbp knock-in or an Aml1-Eto knock-in model, which do not spontaneously develop leukemia.31,32  In these models, either irradiation or chemical mutagenesis is necessary to induce leukemias, thus they clearly require multiple events for the development of leukemia. Several lines of evidence suggest that a multistep model of leukemogenesis also applies to human acute leukemia. Elegant epidemiologic studies suggest that childhood leukemias require at least 2, and probably more, genetic events for leukemia development. In particular, lymphoblastic leukemias with TEL-AML1 rearrangements appear to develop after a multistep process. TEL-AML1 rearrangements are often detected in blood taken at birth from a child who will develop ALL 3-5 years later.33  A recently described twin pair, one with TEL-AML1 ALL, and one clinically healthy, allowed the isolation of a clearly abnormal, TEL-AML1–bearing preleukemic clone that up to that point had failed to cause clinically overt leukemia in the unaffected twin.34  This suggests that TEL-AML1 rearrangements are the first genetic event, but that additional mutations are required for the development of ALL. Similar studies performed on newborn blood spots from children who subsequently develop MLL-rearranged ALL show that MLL-translocations clearly arise in utero and are detectable at birth, whereas the clinical disease develops some time in the first year of life.35 

Mutant signaling molecules are attractive candidates for cooperation with translocation-associated fusion proteins during leukemogenesis. Ever-increasing evidence suggests that activated kinases play a central role in the pathogenesis of leukemias and myeloproliferative syndrome. The most dramatic evidence for such a role is activation of the ABL tyrosine kinase by the BCR-ABL fusion produced by the t(9;22) and its inhibition by imatinib (Gleevec).36,37  Other mutant kinases frequently identified in AML are the receptor tyrosine kinases FLT3 and c-KIT.38  Recent mouse experiments support the hypothesis that DNA-binding fusion proteins generated by leukemia-associated translocations perform different functions than activated tyrosine kinases.39  DNA-binding fusions induce either a block in differentiation or activate self-renewal in developing hematopoietic progenitors, whereas activated kinases may provide a survival or proliferation signal. These data prompted the hypothesis that at least 2 different classes of mutations are necessary for leukemogenesis.40  The identification of FLT3 mutations and RAS mutations in MLL-rearranged ALLs suggests that a multistep process may also be necessary.41,42 

Given the widespread epigenetic dysregulation (described below), the question arises as to whether the model of multistep pathogenesis might not apply to MLL-rearranged leukemias. Rather, epigenetic changes alone may be sufficient for oncogenesis driven by MLL-fusion proteins. In this scenario, mutations such as those described in FLT3 or RAS are acquired after the development of leukemia and represent clonal outgrowth of leukemia cells that either proliferate more rapidly or are less likely to undergo apoptosis. This interpretation is supported by careful tracking of the clonal evolution of (non-MLL-rearranged) ALL, which has revealed that bulk leukemias are comprised of several clones and subclones with different cytogenetic abnormalities that over time contribute to various extents to the clinical disease.43,44  Multiple mutations might therefore influence the disease phenotype or confer a selective advantage to the respective subclone, but not be necessary for initial leukemia development. Recent genome-wide studies have demonstrated very few gains or losses of chromosomal regions in MLL-rearranged ALLs, providing further support for the concept that MLL-rearranged leukemias may be largely a result of epigenetic dysregulation.45  If MLL-rearranged leukemias are largely driven by epigenetic changes, then therapeutics targeting chromatin structure might be particularly useful in this disease. Possible approaches that focus on targeting epigenetic modifications are described below.

Recent studies have focused on the role of DNA methylation at CpG dinucleotides in normal hematopoiesis, leukemia development, and therapeutic response. The majority of DNA methylation found in cells is controlled by the de novo methyltransferases DNMT3A and DNMT3B and by the maintenance methyltransferase DNMT1, whereas a recently described form of DNA methylation, 5-hydroxymethylation, is generated by the proteins TET1-TET3.46  These DNA modifications likely play multiple roles in the control of gene expression and/or genome integrity via modulation of chromatin state and access of transcriptional complexes to their target genes. However, the most well-described function of DNA methylation is repression of gene expression as a result of methylation at CpG islands located near genes. Recent studies in mice have demonstrated a critical role for DNA methylation in the development and maintenance of normal hematopoietic stem cells and murine leukemias through genetic inactivation of Dnmt1.47,48  Inactivation of Dnmt1 led to inappropriate expression of myeloid cell developmental programs in hematopoietic stem and progenitor cells, leading to inappropriate cell fate decisions and stem cell loss. Inactivation of DNMT1 also delayed the onset of MLL-AF9–mediated AML and suppressed the development of MYC-induced leukemia/lymphomas.48  These initial studies demonstrate that DNA methylation is indeed critical for normal hematopoiesis and leukemia development, providing support for therapeutic approaches that target DNA methylation in leukemia.

Recent clinical studies have, with some success, assessed the therapeutic activity of hypomethylating agents in patients with myelodysplastic syndrome.49,50  Similar studies are ongoing in patients with various leukemias and solid tumors.51,52  Given that MLL-fusion proteins transform cells via aberrant epigenetic programs, as described below, it is possible that modifying the DNA-methylation state in MLL-rearranged leukemia cells could lead to relevant changes in gene expression and thus have therapeutic efficacy. Indeed, studies in cell lines have shown that treatment of MLL-rearranged ALL cells with hypomethylating agents leads to the reactivation of tumor suppressor gene expression and the inhibition of cell growth and proliferation.53  Furthermore, DNA-methylation profiling of MLL-rearranged ALLs showed unique profiles for these leukemias compared with other ALLs and also an association between elevated DNA methylation and therapeutic outcome.54,55  Whereas there are little data on the use of hypomethylating agents against MLL-rearranged leukemias in vivo, MLL-rearranged leukemia cell lines do appear to be more sensitive to hypomethylating agents in vitro than some other subtypes of leukemia.55  In addition, TET1, the founding member of the TET family of proteins that are responsible for 5-hydroxymethylation of DNA, was initially identified in a leukemia possessing a t(10;11)(q22;q23) translocation that generates an MLL-TET1 fusion protein. This points to hydroxymethylation as potentially relevant in some subsets of leukemias with MLL translocations.56  Whereas further study is clearly required to fully understand the relevance of DNA methylation in MLL-rearranged leukemias, targeting DNA methylation is an approach worthy of further evaluation.

An initial hint that histone acetylation might play a role in MLL-rearranged leukemia stems from the binding of HDACs to the MLL wild-type protein.57  A recent study demonstrated that the recruitment of HDACs to MLL-bound promoters is mediated by the MLL-PHD3-Bromo cassette stabilized by CyP33, resulting in silencing of the respective genes. This may represent an important mechanism for the physiological repression of MLL-regulated gene-expression programs as cells differentiate.58  In MLL-rearranged leukemias, the PHD3-Bromo cassette is replaced by the fusion partner, and loss of the MLL-PHD domain has been confirmed to be required for the transforming activity.59,60  The loss of HDAC-mediated silencing of MLL-target genes may contribute to MLL-fusion–mediated leukemogenesis.

Whereas locus-specific loss of HDAC activity may contribute to leukemogenesis, more global modulation of histone acetylation might reorganize the chromatin state such that critical epigenetic programs are lost. Several HDAC inhibitors have entered clinical trials for hematologic malignancies and have shown responses in selected patients.52,61  In preclinical studies, pharmacologic inhibition of HDACs with various pan-HDAC inhibitors was reported to exhibit in vitro antiproliferative and pro-apoptotic effects on a panel of B-ALL cell lines, including several bearing an MLL rearrangement, and in primary MLL-rearranged ALL samples.62,63  Analysis of HDAC isoform–specific inhibitors revealed that compounds selectively targeting class I HDACs also inhibited proliferation and induced apoptosis in MLL-rearranged B-ALL cell lines in a dose range that is consistent with specific target inhibition. Compounds with even further specificity for HDAC1 and HDAC2 also exhibited toxicity toward the B-ALL cell line panel, including those with MLL rearrangements. A phase 1A/2 study of the pan-deacetylase inhibitor panobinostat (LBH589) showed 5 responses in 26 response-evaluable patients with AML. One of the 2 observed complete remissions was achieved in a patient with an MLL-CBP fusion (t(11;16)(q23;p13).64  CBP (also called CREB binding protein or CEBBP) is a histone acetyltransferase and transcriptional coactivator. Recurrent mutations of CBP have also been described recently in samples of relapsed ALL (18.3%, including one case with an MLL rearrangement).65  Functional in vitro studies suggest that CBP mutations may negatively affect glucocorticoid responsiveness. Glucocorticoid response has been validated in multiple clinical studies as an independent prognostic marker in MLL-rearranged infant ALL. These findings suggest that HDAC inhibitors may be interesting therapeutic approaches for MLL-rearranged leukemias, possibly those with MLL-CBP or MLL-p300 rearrangements or perhaps glucocorticoid-resistant MLL-rearranged infant ALL in particular.

Biochemical studies have shown that MLL-fusion oncoproteins copurify with protein complexes normally involved in transcriptional elongation. This suggests that MLL-fusion proteins induce leukemia via unregulated transcriptional regulation at MLL-fusion target genes.66  At least 3 related complexes, PAFc, DOT1L, and pTEFb (also designated AEP or SEC), have been shown to bind to MLL-fusion proteins. The polymerase-associated factor complex (PAFc) associates with the NH3-terminal portion of wild-type MLL and MLL-fusion proteins and regulates RNA polymerase II.67  The pTEFb complex (CDK9/cyclinT) releases stalled RNA polymerase II and thus stimulates transcriptional elongation. pTEFb interacts with MLL-fusion partners such as ENL, ELL (as well as ELL2, ELL3), AF4, and AF5,68,69  and copurifies with the respective MLL fusions. The third, the DOT1L complex, consists of DOT1L, a histone methyltransferase that modifies histone H3 on lysine 79 (H3K79), and multiple MLL-fusion partners such as AF9, ENL, AF17, and AF10.70–75  Based on biochemical interactions, it appears that MLL fusions regulate transcription via recruitment of one or several of these protein complexes. ENL has been identified as a core member of both the pTEFb and Dot1l complexes. However, whether and how these complexes interact, and the extent to which each contributes to MLL-fusion–mediated leukemia development is just beginning to be understood. Another example of the recruitment of a histone-modifying enzyme by an MLL-fusion protein is the MLL-EEN fusion, which requires an arginine methyltransferase for its transforming activity.76  Because studies have linked histone methylation and particularly methylation of H3K79 to positive transcriptional regulation,77  the aberrant recruitment of H3K79 methyltransferase activity or arginine methyltransferase activity might directly influence gene expression. Therefore, at least for these fusion proteins, association with a unique histone methyltransferase may be important for leukemogenic transformation.

The association of MLL-fusion proteins with the histone methyltransferase DOT1L has prompted assessment of H3K79 methylation in MLL-rearranged patient cells and leukemia models. Early evidence that MLL fusions may regulate gene expression via histone modification came from studies assessing the MLL-ENL fusion. Upon expression of MLL-ENL in immortalized murine hematopoietic progenitors, H3K79 levels associated with the HoxA9 and Meis1 promoters increased.78  Further studies demonstrated enhanced H3K79 methylation associated with genes overexpressed in MLL-rearranged leukemia cell lines and murine leukemia models on a genome-wide level.8,79,80  These observations hold true for primary human leukemia samples. Similar to the mouse model, enhanced H3K79 methylation is associated with the HOXA cluster in human leukemia cells.79,80  Genome-wide analysis of primary MLL-rearranged leukemia samples has revealed a distinct H3K79 methylation profile and a unique transcription profile that can distinguish MLL-rearranged leukemias from leukemias with germline MLL.25,28,79  Furthermore, there is a strong correlation between elevated H3K79 methylation and abnormal gene expression in human MLL-rearranged primary ALL samples on a genome-wide basis.8,79,80  Because H3K79 methylation is a histone mark associated with actively transcribed genes, it has been hypothesized that DOT1L may play an active role in the maintenance of MLL-fusion–mediated gene-expression programs.8,70 

In addition to being associated with actively transcribed genes, H3K79 methylation appears to be associated with direct MLL-fusion target genes. A recent study determined the genome-wide promoter occupancy of the MLL-AF4 protein and showed that H3K79 methylation was associated with MLL-AF4-target genes in a human MLL-rearranged cell line.80  Similarly, aberrant H3K79 methylation has been demonstrated on direct MLL-AF9 targets in a murine model of MLL-AF9 fusion–driven leukemia.81  In this study, detailed analysis of multiple epigenetic modifications and correlation with expression levels revealed abnormally high peaks and broader spread of H3K79 methylation, specifically on fusion target genes. This was specific to MLL-AF9 leukemia cells, because H3K79 methylation profiles were normal when the same loci were expressed under physiologic regulation in normal hematopoietic progenitors.

Functional studies that suppress or inactivate Dot1L expression also support a critical role for H3K79 methylation in MLL-fusion–mediated transformation and leukemogenesis. shRNA-mediated suppression of DOT1L in human leukemia cell lines has been shown to suppress MLL-fusion–mediated target gene expression,79  and this suppression leads to growth arrest and apoptosis in MLL-AF4 ALL and MLL-AF9 AML cell lines.82  Several groups of investigators have recently developed conditional loss-of-function mouse models and have shown that inactivation of DOT1L in MLL-fusion–transformed cells leads to differentiation and apoptosis in vitro.83,84  Furthermore, inactivation of Dot1L in mouse MLL-AF9–mediated AML inhibits leukemia development.81,85  On a molecular level, loss of Dot1lL and H3K79 methylation led to the collapse of a leukemia-associated gene-expression signature that contained many of the MLL-AF9 target genes. In contrast, global gene expression levels did not change despite association of H3K79 methylation with as many as 11% of all genes. The notion that the loss of widespread chromatin marks may not affect the majority of genes associated with this modification, but rather may induce very specific gene-expression changes on a small subset of genes, is supported by recent studies on epigenetic regulation in yeast. It is therefore possible that the recruitment of Dot1l and the institution of abnormal histone methylation patterns at key loci of leukemogenic transcriptional programs is a crucial step in MLL-fusion–mediated transformation and in the maintenance of MLL-fusion protein–mediated gene expression. Initial studies on hematopoiesis in conditional Dot1l-knockout mice show that hematopoietic cells can develop in neonatal mice in the absence of Dot1L, albeit less efficiently than in wild-type animals81  and that inactivation of Dot1L in adult mice leads to pancytopenia, within 1-2 months.83  Furthermore, hematopoietic cells transformed by other leukemogenic oncogenes such as HoxA9 and Meis1 do not appear to be affected in their growth. These studies support the hypothesis that DOT1L is a potential therapeutic target in this disease and that continued development of DOT1L inhibitors is warranted.

The recent development of a specific small-molecule inhibitor of DOT1L will be instrumental in addressing some fundamental questions about the therapeutic potential for inhibition of H3K79 methylation, in addition to providing proof of principle for the clinical development of DOT1L inhibitors. EPZ004777 is a competitive inhibitor of the methyl donor S-adenosyl-methionine.86  Analysis of this compound on a panel of human leukemia cell lines revealed antiproliferative activity in the nanomolar or low micromolar range that was remarkably selective for cell lines bearing MLL rearrangements. On a molecular level, treatment with the inhibitor causes a decrease in mRNA expression of known MLL-fusion target genes, including HOXA9 and MEIS1. In addition, inhibitor-treated, MLL-rearranged cell lines exhibited cell-cycle arrest in G0/G1, an increase in expression of differentiation markers in MLL-rearranged AML cells, and death by apoptosis. These data provide a very compelling rationale for the clinical development of DOT1L inhibitors as targeted therapeutics for MLL-rearranged leukemias.

Conflict-of-interest disclosure: S.A.A. has consulted for Epizyme. K.M.B. declares no competing financial interests. Off-label drug use: None disclosed.

Scott A. Armstrong, Children's Hospital Boston, Karp Family Research Laboratories, 1 Blackfan Cir, Boston, MA 02215; Phone: (617) 919-2508; e-mail: Scott.Armstrong@childrens.harvard.edu.

1
Biondi
 
A
Cimino
 
G
Pieters
 
R
Pui
 
CH
Biological and therapeutic aspects of infant leukemia
Blood
2000
, vol. 
96
 
1
(pg. 
24
-
33
)
2
Mann
 
G
Attarbaschi
 
A
Schrappe
 
M
, et al. 
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
, vol. 
116
 
15
(pg. 
2644
-
2650
)
3
Felix
 
CA
Hosler
 
MR
Winick
 
NJ
Masterson
 
M
Wilson
 
AE
Lange
 
BJ
ALL-1 gene rearrangements in DNA topoisomerase II inhibitor-related leukemia in children
Blood
1995
, vol. 
85
 
11
(pg. 
3250
-
3256
)
4
Domer
 
PH
Fakharzadeh
 
SS
Chen
 
CS
, et al. 
Acute mixed-lineage leukemia t(4;11)(q21;q23) generates an MLL-AF4 fusion product
Proc Natl Acad Sci U S A
1993
, vol. 
90
 
16
(pg. 
7884
-
7888
)
5
Gu
 
Y
Nakamura
 
T
Alder
 
H
, et al. 
The t(4;11) chromosome translocation of human acute leukemias fuses the ALL-1 gene, related to Drosophila trithorax, to the AF-4 gene
Cell
1992
, vol. 
71
 
4
(pg. 
701
-
708
)
6
Tkachuk
 
DC
Kohler
 
S
Cleary
 
ML
Involvement of a homolog of Drosophila trithorax by 11q23 chromosomal translocations in acute leukemias
Cell
1992
, vol. 
71
 
4
(pg. 
691
-
700
)
7
Ziemin-van der Poel
 
S
McCabe
 
NR
Gill
 
HJ
, et al. 
Identification of a gene, MLL, that spans the breakpoint in 11q23 translocations associated with human leukemias
Proc Natl Acad Sci U S A
1991
, vol. 
88
 
23
(pg. 
10735
-
10739
)
8
Krivtsov
 
AV
Armstrong
 
SA
MLL translocations, histone modifications and leukaemia stem-cell development
Nat Rev Cancer
2007
, vol. 
7
 
11
(pg. 
823
-
833
)
9
Ayton
 
PM
Cleary
 
ML
Molecular mechanisms of leukemogenesis mediated by MLL fusion proteins
Oncogene
2001
, vol. 
20
 
40
(pg. 
5695
-
5707
)
10
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
, vol. 
114
 
12
(pg. 
2489
-
2496
)
11
Yu
 
BD
Hess
 
JL
Horning
 
SE
Brown
 
GA
Korsmeyer
 
SJ
Altered Hox expression and segmental identity in Mll-mutant mice
Nature
1995
, vol. 
378
 
6556
(pg. 
505
-
508
)
12
Yagi
 
H.
Deguchi
 
K.
Aono
 
A.
Tani
 
Y.
Kishimoto
 
T.
Komori
 
T.
Growth disturbance in fetal liver hematopoiesis of Mll-mutant mice
Blood
1998
, vol. 
92
 
1
(pg. 
108
-
117
)
13
Ernst
 
P
Fisher
 
JK
Avery
 
W
Wade
 
S
Foy
 
D
Korsmeyer
 
SJ
Definitive hematopoiesis requires the mixed-lineage leukemia gene
Dev Cell
2004
, vol. 
6
 
3
(pg. 
437
-
443
)
14
Milne
 
TA
Briggs
 
SD
Brock
 
HW
, et al. 
MLL targets SET domain methyltransferase activity to Hox gene promoters
Mol Cell
2002
, vol. 
10
 
5
(pg. 
1107
-
1117
)
15
Nakamura
 
T
Mori
 
T
Tada
 
S
, et al. 
ALL-1 is a histone methyltransferase that assembles a supercomplex of proteins involved in transcriptional regulation
Mol Cell
2002
, vol. 
10
 
5
(pg. 
1119
-
1128
)
16
Hughes
 
CM
Rozenblatt-Rosen
 
O
Milne
 
TA
, et al. 
Menin associates with a trithorax family histone methyltransferase complex and with the hoxc8 locus
Mol Cell
2004
, vol. 
13
 
4
(pg. 
587
-
597
)
17
Milne
 
TA
Hughes
 
CM
Lloyd
 
R
, et al. 
Menin and MLL cooperatively regulate expression of cyclin-dependent kinase inhibitors
Proc Natl Acad Sci U S A
2005
, vol. 
102
 
3
(pg. 
749
-
754
)
18
Liu
 
H
Takeda
 
S
Kumar
 
R
, et al. 
Phosphorylation of MLL by ATR is required for execution of mammalian S-phase checkpoint
Nature
2010
, vol. 
467
 
7313
(pg. 
343
-
346
)
19
Hess
 
JL
Yu
 
BD
Li
 
B
Hanson
 
R
Korsmeyer
 
SJ
Defects in yolk sac hematopoiesis in Mll-null embryos
Blood
1997
, vol. 
90
 
5
(pg. 
1799
-
1806
)
20
Ernst
 
P
Mabon
 
M
Davidson
 
AJ
Zon
 
LI
Korsmeyer
 
SJ
An Mll-dependent Hox program drives hematopoietic progenitor expansion
Curr Biol
2004
, vol. 
14
 
22
(pg. 
2063
-
2069
)
21
Jude
 
CD
Climer
 
L
Xu
 
D
Artinger
 
E
Fisher
 
JK
Ernst
 
P
Unique and independent roles for MLL in adult hematopoietic stem cells and progenitors
Cell Stem Cell
2007
, vol. 
1
 
3
(pg. 
324
-
337
)
22
Owens
 
BM
Hawley
 
RG
HOX and non-HOX homeobox genes in leukemic hematopoiesis
Stem Cells
2002
, vol. 
20
 
5
(pg. 
364
-
379
)
23
Borrow
 
J
Shearman
 
AM
Stanton
 
VP
, et al. 
The t(7;11)(p15;p15) translocation in acute myeloid leukaemia fuses the genes for nucleoporin NUP98 and class I homeoprotein HOXA9
Nat Genet
1996
, vol. 
12
 
2
(pg. 
159
-
167
)
24
Nakamura
 
T
Largaespada
 
DA
Lee
 
MP
, et al. 
Fusion of the nucleoporin gene NUP98 to HOXA9 by the chromosome translocation t(7;11)(p15;p15) in human myeloid leukaemia
Nat Genet
1996
, vol. 
12
 
2
(pg. 
154
-
158
)
25
Armstrong
 
SA
Staunton
 
JE
Silverman
 
LB
, et al. 
MLL translocation specify a distinct gene expression profile that distinguishes a unique leukemia
Nat Genet
2002
, vol. 
30
 (pg. 
41
-
47
)
26
Ferrando
 
AA
Armstrong
 
SA
Neuberg
 
DS
, et al. 
Gene expression signatures of MLL-rearranged T-lineage and B-precursor acute leukemias: dominance of HOX dysregulation
Blood
2003
, vol. 
102
 (pg. 
262
-
268
)
27
Yeoh
 
EJ
Ross
 
ME
Shurtleff
 
SA
, et al. 
Classification, subtype discovery, and prediction of outcome in pediatric acute lymphoblastic leukemia by gene expression profiling
Cancer Cell
2002
, vol. 
1
 (pg. 
133
-
143
)
28
Ross
 
ME
Mahfouz
 
R
Onciu
 
M
, et al. 
Gene expression profiling of pediatric acute myelogenous leukemia
Blood
2004
, vol. 
104
 
12
(pg. 
3679
-
3687
)
29
Ross
 
ME
Zhou
 
X
Song
 
G
, et al. 
Classification of pediatric acute lymphoblastic leukemia by gene expression profiling
Blood
2003
, vol. 
102
 (pg. 
2951
-
2959
)
30
Corral
 
J
Lavenir
 
I
Impey
 
H
, et al. 
An Mll-AF9 fusion gene made by homologous recombination causes acute leukemia in chimeric mice: a method to create fusion oncogenes
Cell
1996
, vol. 
85
 
6
(pg. 
853
-
861
)
31
Higuchi
 
M
O'Brien
 
D
Kumaravelu
 
P
, et al. 
Expression of a conditional AML1-ETO oncogene bypasses embryonic lethality and establishes a murine model of human t(8;21) acute myeloid leukemia
Cancer Cell
2002
, vol. 
1
 
1
(pg. 
63
-
74
)
32
Wang
 
J
Iwasaki
 
H
Krivtsov
 
A
, et al. 
Conditional MLL-CBP targets GMP and models therapy-related myeloproliferative disease
EMBO J
2005
, vol. 
24
 
2
(pg. 
368
-
381
)
33
Wiemels
 
JL
Cazzaniga
 
G
Daniotti
 
M
, et al. 
Prenatal origin of acute lymphoblastic leukaemia in children
Lancet
1999
, vol. 
354
 
9189
(pg. 
1499
-
1503
)
34
Hong
 
D
Gupta
 
R
Ancliff
 
P
, et al. 
Initiating and cancer-propagating cells in TEL-AML1-associated childhood leukemia
Science
2008
, vol. 
319
 
5861
(pg. 
336
-
339
)
35
Ford
 
AM
Ridge
 
SA
Cabrera
 
ME
, et al. 
In utero rearrangements in the trithorax-related oncogene in infant leukaemias
Nature
1993
, vol. 
363
 
6427
(pg. 
358
-
360
)
36
Sawyers
 
CL
Rational therapeutic intervention in cancer: kinases as drug targets
Curr Opin Genet Dev
2002
, vol. 
12
 
1
(pg. 
111
-
115
)
37
Druker
 
BJ
Talpaz
 
M
Resta
 
DJ
, et al. 
Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia
N Engl J Med
2001
, vol. 
344
 
14
(pg. 
1031
-
1037
)
38
Scheijen
 
B
Griffin
 
JD
Tyrosine kinase oncogenes in normal hematopoiesis and hematological disease
Oncogene
2002
, vol. 
21
 
21
(pg. 
3314
-
3333
)
39
Huntly
 
BJ
Shigematsu
 
H
Deguchi
 
K
, et al. 
MOZ-TIF2, but not BCR-ABL, confers properties of leukemic stem cells to committed murine hematopoietic progenitors
Cancer Cell
2004
, vol. 
6
 
6
(pg. 
587
-
596
)
40
Kelly
 
LM
Gilliland
 
DG
Genetics of myeloid leukemias
Annu Rev Genomics Hum Genet
2002
, vol. 
3
 (pg. 
179
-
198
)
41
Armstrong
 
SA
Kung
 
AL
Mabon
 
ME
, et al. 
Validation of a therapeutic target identified by gene expression based classification
Cancer Cell
2003
, vol. 
3
 
2
(pg. 
173
-
183
)
42
Mahgoub
 
N
Parker
 
RI
Hosler
 
MR
, et al. 
RAS mutations in pediatric leukemias with MLL gene rearrangements
Genes Chromosomes Cancer
1998
, vol. 
21
 
3
(pg. 
270
-
275
)
43
Anderson
 
K
Lutz
 
C
van Delft
 
FW
, et al. 
Genetic variegation of clonal architecture and propagating cells in leukaemia
Nature
2011
, vol. 
469
 
7330
(pg. 
356
-
361
)
44
Notta
 
F
Mullighan
 
CG
Wang
 
JC
, et al. 
Evolution of human BCR-ABL1 lymphoblastic leukaemia-initiating cells
Nature
2011
, vol. 
469
 
7330
(pg. 
362
-
367
)
45
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
)
46
Tahiliani
 
M
Koh
 
KP
Shen
 
Y
, et al. 
Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1
Science
2009
, vol. 
324
 
5929
(pg. 
930
-
935
)
47
Trowbridge
 
JJ
Snow
 
JW
Kim
 
J
Orkin
 
SH
DNA methyltransferase 1 is essential for and uniquely regulates hematopoietic stem and progenitor cells
Cell Stem Cell
2009
, vol. 
5
 
4
(pg. 
442
-
449
)
48
Bröske
 
AM
Vockentanz
 
L
Kharazi
 
S
, et al. 
DNA methylation protects hematopoietic stem cell multipotency from myeloerythroid restriction
Nat Genet
2009
, vol. 
41
 
11
(pg. 
1207
-
1215
)
49
Kantarjian
 
H
Issa
 
JP
Rosenfeld
 
CS
, et al. 
Decitabine improves patient outcomes in myelodysplastic syndromes: results of a phase III randomized study
Cancer
2006
, vol. 
106
 
8
(pg. 
1794
-
1803
)
50
Fenaux
 
P
Mufti
 
GJ
Hellstrom-Lindberg
 
E
, et al. 
International Vidaza High-Risk MDS Survival Study Group. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study
Lancet Oncol
2009
, vol. 
10
 
3
(pg. 
223
-
232
)
51
Boumber
 
Y
Issa
 
JP
Epigenetics in cancer: what's the future?
Oncology (Williston Park)
2011
, vol. 
25
 
3
(pg. 
220
-
226
)pg. 
228
 
52
Fandy
 
TE
Carraway
 
H
Gore
 
SD
DNA demethylating agents and histone deacetylase inhibitors in hematologic malignancies
Cancer J
2007
, vol. 
13
 
1
(pg. 
40
-
48
)
53
Stam
 
RW
den Boer
 
ML
Passier
 
MM
, et al. 
Silencing of the tumor suppressor gene FHIT is highly characteristic for MLL gene rearranged infant acute lymphoblastic leukemia
Leukemia
2006
, vol. 
20
 
2
(pg. 
264
-
271
)
54
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
)
55
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
)
56
Lorsbach
 
RB
Moore
 
J
Mathew
 
S
Raimondi
 
SC
Mukatira
 
ST
Downing
 
JR
TET1, a member of a novel protein family, is fused to MLL in acute myeloid leukemia containing the t(10;11)(q22;q23)
Leukemia
2003
, vol. 
17
 
3
(pg. 
637
-
641
)
57
Xia
 
ZB
Anderson
 
M
Diaz
 
MO
Zeleznik-Le
 
NJ
MLL repression domain interacts with histone deacetylases, the polycomb group proteins HPC2 and BMI-1, and the corepressor C-terminal-binding protein
Proc Natl Acad Sci U S A
2003
, vol. 
100
 
14
(pg. 
8342
-
8347
)
58
Wang
 
Z
Song
 
J
Milne
 
TA
, et al. 
Pro isomerization in MLL1 PHD3-bromo cassette connects H3K4me readout to CyP33 and HDAC-mediated repression
Cell
2010
, vol. 
141
 
7
(pg. 
1183
-
1194
)
59
Chen
 
J
Santillan
 
DA
Koonce
 
M
, et al. 
Loss of MLL PHD finger 3 is necessary for MLL-ENL-induced hematopoietic stem cell immortalization
Cancer Res
2008
, vol. 
68
 
15
(pg. 
6199
-
6207
)
60
Muntean
 
AG
Giannola
 
D
Udager
 
AM
Hess
 
JL
The PHD fingers of MLL block MLL fusion protein-mediated transformation
Blood
2008
, vol. 
112
 
12
(pg. 
4690
-
4693
)
61
Lane
 
AA
Chabner
 
BA
Histone deacetylase inhibitors in cancer therapy
J Clin Oncol
2009
, vol. 
27
 
32
(pg. 
5459
-
5468
)
62
Stumpel
 
DJPM
Schneider
 
P
Seslija
 
L
, et al. 
Connectivity mapping identifies HDAC inhibitors as suitable candidates for the treatment of t(4;11)-positive acute lymphoblastic leukemia in infants [Abstract]
Blood
2010
, vol. 
116
 
21
pg. 
1322
 
63
Stubbs
 
MC
Kim
 
W-I
Davis
 
T
, et al. 
Selective inhibition of HDAC1 and HDAC2 is a potential therapeutic option for B-ALL65279 [Abstract]
Blood
2010
, vol. 
116
 
21
pg. 
1194
 
64
Ottmann
 
OG
Spencer
 
A
Prince
 
HM
, et al. 
Phase IA/II study of oral panobinostat (LBH589), a novel pan-deacetylase inhibitor (DACi) demonstrating efficacy in patients with advanced hematologic malignancies [Abstract]
Blood
2008
, vol. 
112
 
11
pg. 
958
 
65
Mullighan
 
CG
Zhang
 
J
Kasper
 
LH
, et al. 
CREBBP mutations in relapsed acute lymphoblastic leukaemia
Nature
2011
, vol. 
471
 
7337
(pg. 
235
-
239
)
66
Mohan
 
M
Lin
 
C
Guest
 
E
Shilatifard
 
A
Licensed to elongate: a molecular mechanism for MLL-based leukaemogenesis
Nat Rev Cancer
2010
, vol. 
10
 
10
(pg. 
721
-
728
)
67
Muntean
 
AG
Tan
 
J
Sitwala
 
K
Huang
 
Y
, et al. 
The PAF complex synergizes with MLL fusion proteins at HOX loci to promote leukemogenesis
Cancer Cell
2010
, vol. 
17
 
6
(pg. 
609
-
621
)
68
Yokoyama
 
A
Lin
 
M
Naresh
 
A
Kitabayashi
 
I
Cleary
 
ML
A higher-order complex containing AF4 and ENL family proteins with P-TEFb facilitates oncogenic and physiologic MLL-dependent transcription
Cancer Cell
2010
, vol. 
17
 
2
(pg. 
198
-
212
)
69
Lin
 
C
Smith
 
ER
Takahashi
 
H
, et al. 
AFF4, a component of the ELL/P-TEFb elongation complex and a shared subunit of MLL chimeras, can link transcription elongation to leukemia
Mol Cell
2010
, vol. 
37
 
3
(pg. 
429
-
437
)
70
Okada
 
Y
Feng
 
Q
Lin
 
Y
, et al. 
hDOT1L links histone methylation to leukemogenesis
Cell
2005
, vol. 
121
 
2
(pg. 
167
-
178
)
71
Bitoun
 
E
Oliver
 
PL
Davies
 
KE
The mixed-lineage leukemia fusion partner AF4 stimulates RNA polymerase II transcriptional elongation and mediates coordinated chromatin remodeling
Hum Mol Genet
2007
, vol. 
16
 
1
(pg. 
92
-
106
)
72
Mueller
 
D
Bach
 
C
Zeisig
 
D
, et al. 
A role for the MLL fusion partner ENL in transcriptional elongation and chromatin modification
Blood
2007
, vol. 
110
 
13
(pg. 
4445
-
4454
)
73
Mueller
 
D
García-Cuéllar
 
MP
Bach
 
C
Buhl
 
S
Maethner
 
E
Slany
 
RK
Misguided transcriptional elongation causes mixed lineage leukemia
PLoS Biol
2009
, vol. 
7
 
11
pg. 
e1000249
 
74
Zhang
 
W
Xia
 
X
Reisenauer
 
MR
Hemenway
 
CS
Kone
 
BC
Dot1a-AF9 complex mediates histone H3 Lys-79 hypermethylation and repression of ENaCalpha in an aldosterone-sensitive manner
J Biol Chem
2006
, vol. 
281
 
26
(pg. 
18059
-
18068
)
75
Mohan
 
M
Herz
 
HM
Takahashi
 
YH
, et al. 
Linking H3K79 trimethylation to Wnt signaling through a novel Dot1-containing complex (DotCom)
Genes Dev
2010
, vol. 
24
 
6
(pg. 
574
-
589
)
76
Cheung
 
N
Chan
 
LC
Thompson
 
A
Cleary
 
ML
So
 
CW
Protein arginine-methyltransferase-dependent oncogenesis
Nat Cell Biol
2007
, vol. 
9
 
10
(pg. 
1208
-
1215
)
77
Steger
 
DJ
Lefterova
 
MI
Ying
 
L
, et al. 
DOT1L/KMT4 recruitment and H3K79 methylation are ubiquitously coupled with gene transcription in mammalian cells
Mol Cell Biol
2008
, vol. 
28
 
8
(pg. 
2825
-
2839
)
78
Milne
 
TA
Martin
 
ME
Brock
 
HW
Slany
 
RK
Hess
 
JL
Leukemogenic MLL fusion proteins bind across a broad region of the Hox a9 locus, promoting transcription and multiple histone modifications
Cancer Res
2005
, vol. 
65
 
24
(pg. 
11367
-
11374
)
79
Krivtsov
 
AV
Feng
 
Z
Lemieux
 
ME
, et al. 
H3K79 methylation profiles define murine and human MLL-AF4 leukemias
Cancer Cell
2008
, vol. 
14
 
5
(pg. 
355
-
368
)
80
Guenther
 
MG
Lawton
 
LN
Rozovskaia
 
T
, et al. 
Aberrant chromatin at genes encoding stem cell regulators in human mixed-lineage leukemia
Genes Dev
2008
, vol. 
22
 
24
(pg. 
3403
-
3408
)
81
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
)
82
Bernt
 
K
Faber
 
J
Davis
 
TN
Kung
 
A
Armstrong
 
S
DOT1L and histone H3 Lysine79 methylation as a therapeutic target in mixed lineage leukemia [Abstract]
Blood
2009
, vol. 
114
 
22
pg. 
93
 
83
Chang
 
MJ
Wu
 
H
Achille
 
NJ
, et al. 
Histone H3 lysine 79 methyltransferase Dot1 is required for immortalization by MLL oncogenes
Cancer Res
2010
, vol. 
70
 
24
(pg. 
10234
-
10242
)
84
Jo
 
SY
Granowicz
 
EM
Maillard
 
I
Thomas
 
D
Hess
 
JL
Requirement for Dot1l in murine postnatal hematopoiesis and leukemogenesis by MLL translocation
Blood
2011
, vol. 
117
 
18
(pg. 
4759
-
4768
)
85
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
)
86
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
)