Sie sind auf Seite 1von 12

From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

Review Series

ADVANCES IN ACUTE MYELOID LEUKEMIA

Epigenetics and approaches to targeted epigenetic therapy in acute


myeloid leukemia
Bas J. Wouters and Ruud Delwel
Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands

Acute myeloid leukemia (AML) is the most epigenetic mechanisms in the pathogene- overview of the current state of epigenetics
common type of acute leukemia in adults. sis of the disease. In contrast to genetic and epigenetic therapy in AML and will
AML is a heterogeneous malignancy char- changes, epigenetic modifications are fre- describe perspectives on how to identify
acterized by distinct genetic abnormalities. quently reversible, which provides opportu- promising new approaches for epigenetic
Recent discoveries have highlighted an nities for targeted treatment using specific targeted treatment. (Blood. 2016;127(1):
additional important role of dysregulated inhibitors. In this review, we will provide an 42-52)

Introduction
Acute myeloid leukemia (AML) is the most common type of acute Modes of epigenetic (dys)regulation in AML
leukemia in adults.1 Despite current treatment protocols involving
intensive chemotherapy and the judicious use of stem cell trans- We first provide an outline of various modes of epigenetic regulation in
plantation, AML is still fatal in approximately half of younger patients hematopoietic cells and how these may be disturbed in AML. This has
and in about 80% of patients over the age of 60 as a result of primary been summarized in Table 1. Several recent reviews have provided
refractoriness, relapse, or treatment-related mortality.2 Consequently, excellent descriptions of the various abnormalities in epigenetic-
there is a need for more specific and less toxic drugs that are rationally regulating enzymes in hematopoietic malignancies in general and AML
designed to target leukemia-specific abnormalities. in particular, to which the reader is referred for more detail.4-6
The term epigenetics refers to changes in gene expression that are
inheritable by cell division but not caused by changes in the DNA DNA methylation
sequence itself.3 Examples include DNA modifications, such as cyto- The process of DNA methylation involves the addition of a methyl
sine methylation, modifications of histone proteins, or RNA-associated group to the 5-carbon position of cytosines in CpG dinucleotides,
gene silencing. Recent discoveries have shed light on the important role yielding 5-methylcytosine. Aberrant patterns of DNA methylation
of dysregulated epigenetic mechanisms in the pathogenesis of AML.4-6 in malignancies were initially particularly studied in the context of
Several genes playing key roles in epigenetic regulation show recurrent so-called “CpG islands” in gene promoters. Hypermethylation of
somatic alterations in AML. It is however apparent that epigenetic cytosines in CpG islands is associated with silencing of tumor-
dysregulation in AML is more widespread than can be explained by suppressor genes.47 It is now increasingly clear that aberrant DNA
recurrent somatic mutations alone.7 For instance, disturbed genome- methylation patterns outside CpG islands may be equally important
wide patterns of DNA methylation also exist in AML subtypes not in leukemogenesis, and that hypomethylation may be as relevant as
linked to mutations in known epigenetic modifiers.8 In addition to hypermethylation. Newly identified regions of interest include gene
somatic changes in coding regions of genes, alterations in enhancer bodies and so-called “CpG shores” (regions at 2 kb at either side of CpG
elements have been reported that perturb normal regulation of gene islands). In addition, even (long distant) enhancer regions are frequently
expression.9 regulated by methylation. DNA methylation is established by DNA
Epigenetic modifications are frequently reversible. Because of this methyltransferases (DNMTs). Recurrent mutations in DNMT3A, one of
inherent plasticity, epigenetic dysregulation offers potential avenues for the de novo DNA methyltransferases, are found in 6% to 36% of AML
targeted treatment using specific inhibitors of histone-modifying pro- patients and are most commonly heterozygous (Table 1). DNMT3A
teins or proteins driving or maintaining DNA methylation. In support of mutations appear to be early events in leukemogenesis. This is evident
this, both experimental evidence as well as clinical observations under- by studies demonstrating that DNMT3A mutations in AML patients
line the concept that epigenetic mutations and alterations contribute to a may also be present in T-lymphocytes derived from the same patient.48
preleukemic state but are usually not sufficient to cause full-blown acute These findings indicate that the mutation can arise in a primitive cell that
leukemia.10 This would indicate that these defects are already present is still able to give rise to both myeloid as well as lymphoid cells. In
in early clones and targeting those abnormalities may help to eradicate addition, it was recently demonstrated that DNMT3A mutations can
the disease. be found in (elderly) individuals without an apparent hematologic
In this review, we give an overview of the current state of epigenetics malignancy, indicating that these mutations may be involved in clonal
and epigenetic therapy in AML and will describe perspectives on how hematopoiesis, which in some individuals precedes the development of
to identify promising new approaches for epigenetic-targeted treatment. overt leukemia.10

Submitted July 22, 2015; accepted September 25, 2015. Prepublished online © 2016 by The American Society of Hematology
as Blood First Edition paper, December 10, 2015; DOI 10.1182/blood-2015-
07-604512.

42 BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1


From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1 EPIGENETICS IN AML 43

Table 1. Recurrently mutated or translocated genes with epigenetic function in AML


Type of abnormalities described
Gene Epigenetic function (percentage of AML) References Remarks
DNMT3A De novo DNA methylation Mostly frameshifts; rare missense- and 7,11-16 Associated with normal karyotype;
non-sense mutations (6-36%) may be associated with poor prognosis
TET2 Conversion of 5-methylcytosine to Frameshift, nonsense and missense 7,15-17,18-21 Mutually exclusive with IDH1/2 mutations
5-hydroxymethylcytosine mutations (8-27%)
IDH1 and IDH2 Enzymes that convert isocitrate to Missense mutations (5-16% for IDH1; 7,15,16,22-24 Mutually exclusive with TET2 mutations.
a-ketoglutarate (a-KG), a cofactor 6-19% for IDH2) Mutations result in production of
for TET2 2-hydroxyglutarate, which inhibits
TET2 function
CREBBP (CBP) Histone lysine acetyltransferase Rearrangements: fusion genes 7,25 Rare
KAT6A (MYST3/MOZ) Histone lysine acetyltransferase Rearrangements: fusion genes 7,26 Rare
EP300 (p300) Histone lysine acetyltransferase Rearrangements: fusion genes 27,28 Rare
HDAC2 and HDAC3 Histone deacetylase Missense mutations 7 Rare
KMT2A (MLL/MLL1) H3K4 methyltransferase Rearrangements: fusion genes (1-10%); 7,15,16,29-32 More than 50 fusion partners reported in
partial tandem duplications (4-7%) acute leukemias
EZH2 H3K27 methyltransferase, enzymatic Mutations (2%) 7,33
component of PRC2
NSD1 H3K36 methyltransferase Rearrangement involving NUP98 (2-5%) 7,34,35
ASXL1 Recruitment of PRC2 to target loci Mostly frameshifts or nonsense 7,15,16,36,37-41 More common in elderly patients; poor
mutations (3-25%) prognosis particularly in association with
RUNX1 mutations
ASXL2 Homolog of ASXL1; function unknown Mutations (23% of AML with 42 Mutually exclusive with ASXL1 mutations
RUNX1-RUNX1T1)
JARID2 Recruitment of PRC2 to target loci Deletion in transformation of MDS 43
or MPN to AML
SUZ12 Member of PRC2 Missense mutations, insertions and 7,43,44 Sporadically mutated in progression severe
deletions congenital neutropenia to AML; deleted
in transformation MDS/MPN to AML
KDM5A (JARID1) Histone lysine demethylase Rearrangement involving NUP98 45 10% of pediatric acute megakaryoblastic
leukemia
KDM6A (UTX) Histone lysine demethylase Missense mutations 7,46 Rare

7
Sporadic mutations from TCGA extracted using COSMIC (http://cancer.sanger.ac.uk/).
MDS, myelodysplastic syndrome; MPN, myeloproliferative neoplasm; PRC, polycomb repressor complex.

The mechanism by which mutant DNMT3A contributes to leuke- emphasizes the relationship between defective transcriptional regula-
mic transformation is not completely clear. It is hypothesized that mu- tors and epigenetic alterations. In fact, evidence for a role of onco-
tant DNMT3A acts as a dominant negative over wild-type DNMT3A. genic transcription factors in directing the activity of DNMTs and
AML cells with the R882H mutation have been reported to have pro- changing the DNA methylation patterns has been proposed for
found reduction of de novo methyltransferase activity.49 Mice trans- EVI1,54 PML-RARA55 and RUNX1-RUNX1T1.56
planted with Dnmt3a-null hematopoietic stem cells (HSCs) in a
competitive transplantation setting do not develop myeloid malignan- DNA hydroxymethylation
cies.50 However, when the same Dnmt3a-null HSCs are transplanted
The oxidation of 5-methylcytosine yields 5-hydroxymethylcytosine
in irradiated mice, not in a competitive setting, preleukemic disorders
(5hmC). This process is catalyzed by the Ten-Eleven-Translocation
are frequently observed.51 Mouse modeling experiments show that the
(TET)-enzymes and is dependent on a-ketoglutarate. Mutations in
R882H variant, which is the most common DNMT3A mutation, is able TET2 are observed in 8% to 27% of patients with AML (Table 1). These
to drive the development of myeloproliferative disorders in vivo as mutations are associated with reduced levels of 5hmC.57 TET2 muta-
well. These data support the hypothesis that mutant DNMT3A acts as tions confer a poor prognosis in intermediate-risk AML.17 In addition,
a dominant negative.52 Mice that develop full-blown AML generally mutations in genes encoding the enzymes called isocitrate dehydro-
acquire cooperating activating mutations in signaling factors such as genase 1 and 2 (IDH1 and IDH2) are frequently observed in AML
c-Kit.52 This is similar to what is observed in human AML patients with (Table 1). They appear more prevalent in patients with normal cyto-
DNMT3A mutations, who frequently carry concomitant FLT3-ITD genetics. IDH proteins catalyze the conversion of isocitrate to
mutations. Recent work suggests that the presence of DNMT3A a-ketoglutarate. Mutations in IDH1 and IDH2 result in the production
R882H promotes chemoresistance, which appears to be associated of an aberrant metabolite, 2-hydroxyglutarate.58 This molecule has
with impaired DNA damage sensing and impaired chromatin re- been demonstrated to act as a competitive inhibitor of a-ketoglutarate,
modeling capacities.53 resulting in the inhibition of TET2. TET2 mutations and IDH1/IDH2
Specific patterns of genome-wide cytosine methylation are associ- mutations are mutually exclusive in AML.4 In addition, IDH1/IDH2
ated with distinct subtypes of AML. Using genome-wide profiling, mutations are associated with global cytosine hypermethylation signa-
unique patterns of cytosine methylation are associated with AMLs tures. Patients with TET2 mutations display overlapping hypermeth-
characterized by specific translocations and fusion genes (eg, RUNX1- ylation signatures. TET2 mutations are observed in healthy aging
RUNX1T1 [AML1-ETO], PML-RARA, mutations in CEBPA, and individuals, supporting the concept of clonal hematopoiesis that may
mutations in NPM1).8 The fact that mutations in genes encoding tran- precede leukemogenesis.10,59 Recently, Levine and colleagues reported
scription factors are associated with unique DNA methylation patterns that mutations in the WT1 gene are mutually exclusive with mutations
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

44 WOUTERS and DELWEL BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1

in IDH1/IDH2 or TET2.60 Increased global levels of 5hmC were ob- Histone lysine methylation
served upon WT1 overexpression, whereas reduced 5hmC levels were
Methylation of histone lysine residues can result in mono-, di-, or
observed when WT1 was silenced. Although the mechanism of 5hmC
decrease upon WT silencing or mutation is not understood, the inves- trimethylation. This is processed by lysine methyltransferases (KMTs).
tigators demonstrated that WT1 physically interacts with TET2. Histone methylation alters the affinity of reader proteins to the meth-
Decreased WT1 levels and absence of TET2 resulted in a similar ylated histones. Histone methylation can result in activation or re-
hematopoietic differentiation phenotype. The authors conclude that pression, the actual effect being dependent on its context.77 Marks
TET2, IDH1/IDH2, and WT1 mutations define an AML subtype char- associated with activation include methylation of H3K4, H3H36, and
acterized by dysregulated DNA hydroxymethylation. H3K79. Marks associated with silenced gene transcription include
Murine models to study the mechanism of TET2 and IDH1/2 methylation of H3K9, H3K27, and H4K20. Methylation state (eg,
mutations have recently been developed. Experiments performed in mono- vs trimethylation) may have different functional consequences
conditional Tet2 knockout mice point to a role for Tet2 in the regulation in the same lysine residue.77
of self-renewal of hematopoietic stem cells. These mice do not develop In AML, KMTs that are recurrently involved in translocations or are
leukemia, but the presence of cooperating Flt3-ITD mutations does lead mutated include mixed-lineage leukemia (MLL) proteins and compo-
to increased incidence of leukemia.61 Leukemogenesis may be depen- nents of the polycomb repressor complexes (PRCs) (Table 1). KMT2A,
dent on the downregulation of Gata2 by hypermethylation, an effect that more frequently referred to as MLL, is a member of the family of SET
can be overcome by reintroduction of Gata2.61 It is predicted that mice domain–containing KMTs. MLL targets H3K4 and leaves marks of
expressing conditional mutant Idh1 or Idh2 alleles in the presence of transcriptional activation. MLL is involved in translocations in approx-
Flt3-ITD or Nras mutations will develop AML in a comparable manner. imately 5% to 10% of AML.29 Partial tandem duplications of MLL are
In addition to 5-methylcytosine and 5hmC, two other DNA found in 5% to 7% of de novo AML. MLL translocations result in
modifications have been recognized (ie, 5-formylcytosine and fusion proteins that lack the wild-type SET domain and are frequently
5-carboxylcytosine).62 Similar to 5hmC, both represent oxidative der- replaced by members of genes encoding super elongation complex
ivates of methylated cytosines and can be the result of the activity of nuclear proteins, such as AF9, AF10, and ENL. This complex also
TET enzymes. The biological role of 5hmC and other derivates remains contains DOT1L, a KMT that targets H3K79. MLL-transformed AML
to be established. It has been proposed that they may not merely be samples show aberrant H3K79 methylation, leading to ongoing ex-
intermediates toward CG demethylation, but may have distinct biolog- pression of target genes, among which the homeobox genes are par-
ical roles, for instance in the regulation of gene expression.62 ticularly involved.78
Enhancer of zeste 2 polycomb repressive complex 2 subunit
(EZH2) is an H3K27 methyltransferase that is part of the PRC2
Histone acetylation
polycomb repressor complex. EZH2 catalyzes di- and trimethylation
Histone acetylation involves the transfer of acetyl groups to lysine res- of H3K27, which results in transcriptional repression. Aberrant gain of
idues in histone proteins. The processes of acetylation and deacetylation action of EZH2 has been associated with transcriptional repression of
are governed by histone lysine acetyltransferases (KATs) and histone critical target genes in various types of cancer. In myeloid malignancies,
deacetylases (HDACs), respectively. Acetylation of lysine residues re- however, EZH2 mutations lead to loss of function (Table 1).79,80 The
sults in open chromatin confirmations, whereas deacetylation results functional basis for this difference is unclear, but some preliminary
in condensed and closed chromatin. observations have been made. In a myelodysplastic syndrome (MDS)
Sporadic translocations involving KATs have been described in mouse model, knockout of Ezh2 promoted the development of MDS
myeloid malignancies, and mutations in HDACs have only rarely been by impairing normal HSCs via activating inflammatory cytokine re-
identified (Table 1). However, there is evidence that HDACs may be sponses, while at the same time preventing the transformation to AML
aberrantly recruited by myeloid oncoproteins, such as EVI1 or PML- via PRC1-mediated repression of Hoxa9.81 In a study focusing on
RARA.63,64 These oncoproteins frequently interact with scaffold MLL-AF9 leukemia, EZH2 was not required for leukemogenesis, but
proteins or major complexes, which recruit HDACs. This leads to was required for tumor progression.82 A recent study highlights another
chromatin remodeling, in particular deacetylation of lysines at histone mechanism of interference with EZH2 function. Mutations in the
tails near the transcription factor binding sites. These changes affect splicing factor gene SRSF2, frequently found in MDS, cause mis-
transcription of putative target genes.65-67 splicing of EZH2, resulting in nonsense-mediated decay.83
Acetylated lysine residues in histones can be recognized by reader Other members of the PRC2 complex include additional sex
proteins containing bromodomains, such as the bromodomain and extra combs like transcriptional regulator 1 (ASXL1), Jumonji AT-rich
terminal (BET) proteins BRD2, BRD3, and BRD4. These proteins interactive domain 2 (JARID2), and SUZ12 polycomb repressive
contain 2 so-called bromodomains, which bind to acetylated histones. complex 2 subunit (SUZ12), all of which have been implicated in the
BRD4 is involved in a complex including Mediator and pTEF-b, and development of AML (Table 1). Most of the mutations in these genes
through this complex links histone acetylation to transcription.68,69 result in loss of function of the complex. The PRC2-associated pro-
Although BRD4 and other BET proteins are ubiquitously present at tein ASXL1 has an important role in the recruitment of the complex
gene promoters and enhancers, inhibition of BET proteins results in to target loci. Similar to loss-of-function mutations in EZH2, muta-
disproportionately large changes in expression of particular genes.70-72 tions in ASXL1 result in loss of PRC2-mediated H3K27 methyl-
Evidence is accumulating that these cell type–specific transcriptional ation. ASXL1 mutations may be associated with adverse prognosis,
effects of BET inhibition can to a large extent be explained by the an effect that seems to be correlated to the presence of RUNX1
association of BRD4 with exceptionally large enhancer elements, mutations.36 Mutations in ASXL2 are frequently observed in patients
called “super-enhancers,” which are involved in lineage-specific gene with the RUNX1-RUNX1T1 fusion gene and are mutually exclusive
regulation.73-76 In several types of malignancy, including AML, with ASXL1 mutations.42
disease-specific oncogenic super-enhancers, also referred to as Similar to ASXL1, JARID2 recruits PRC2 to specific target loci.
“stretched enhancers” or “locus control regions,” can be recognized Deletions of JARID2 have been reported in patients showing leukemic
that drive expression of critical oncogenes, such as MYC. transformation of chronic myeloid malignancies.43 SUZ12 has been
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1 EPIGENETICS IN AML 45

reported to be mutated in progression from severe congenital neutro- approved for the treatment of MDS as well as for AML with low blast
penia to AML.44 Thus, in most if not all mutations found in PRC2 count. In Europe, decitabine is also approved for the treatment of elderly
complex members in AML, the ultimate effect is loss of function re- AML patients with blast counts .30% who are not eligible for intensive
sulting in loss of H3K27 methylation. These effects are thus suggested treatment. Azacitidine and decitabine are thought to exert their effects
to cause increased expression of important target genes. by reversing aberrant DNA hypermethylation and thereby restoring
Another polycomb repressor group gene that has been linked to expression of critical (tumor-suppressor) genes. There are increasing
AML is BMI1. BMI1 is part of the PRC1 polycomb repressor complex. data to support the efficacy of these hypomethylating compounds in the
Increased BMI1 expression has been associated with poor prognosis in treatment of patients not eligible for intensive chemotherapy.107-110
AML and MDS.84,85 There are currently no established biomarkers that predict the response
Several AML-related fusion proteins may interfere with normal to these drugs, although it would be likely that not all patients are
PRC functioning. The PML-RARA fusion protein can engage in a equally susceptible. This hypothesis is supported by the identification
complex with several PRC2 members, including SUZ12, EZH2, and of factors that predict for response to azacitidine in MDS and methyl-
EED, thereby recruiting them to specific genomic loci.86 Another ation signatures that predict for response to decitabine in chronic
fusion protein, PZLF-RARA, has been shown to interact with PRC1 myelomonocytic leukemia, respectively.111,112 In MDS, particular
components.87 gene mutations, such as those in TET2 and DNMT3A, may predict for
Demethylation of histone proteins is governed by lysine demeth- better responsiveness to treatment with inhibitors of DNMTs.113 Com-
ylases (KDMs). Two classes of KDMs are known: the amine oxidases, parable studies need to be carried out to dissect which AML subtypes
which include lysine-specific demethylase 1 (LSD1/KDM1A), and the may benefit from treatment with DNMT inhibitors. One experimental
a-ketoglutarate–dependent Jumonji domain (JmjC) containing pro- approach would be to test the sensitivity to these drugs in cohorts of
teins. LSD1 has specificity for H3K4 and H3K9 and has a role both as AML samples, taking into account subtypes that are particularly de-
a transcriptional activator as well as a transcriptional repressor, de- fined by unique DNA-methylation signatures. We previously demon-
pending on the cellular context.88 KDM5A (JARID1), a JmjC lysine strated that leukemias with well-defined molecular abnormalities can be
demethylase, is involved in a fusion protein with NUP98 in ;10% of discriminated based on these epigenetic signatures.8 Moreover, with
children with acute megakaryoblastic leukemia.45,89 KDM6A (UTX) DNA methylation profiling we could identify AML subgroups that did
is another JmjC family member and can be altered by inactivating not harbor any known recurrent genetic defect. It is possible that certain
mutations in AML (Table 1).46 epigenetically defined AML subtypes are more responsive to DNA
methyltransferase inhibitors than others. Responsiveness studies may
Other histone modifications be carried out in vitro using short-term assays testing cellular viability.
These experiments could provide an indication of response to the bulk
In addition to the histone modifications described so far, other modi-
of cells to these agents. Whether the compounds will target the leukemia
fications are known. Methylation of arginine residues in histones is me-
stem cells needs to be established using in vivo models. Mouse models
diated by the family of protein arginine methyltransferases (PRMTs), of
have been generated for a number of AML subtypes carrying the distinct
which 9 have been identified. Arginine methylation may be associated
mutations, which allow investigators to study the effects of hypomethy-
both with transcriptional activation as well as with silencing.90,91
lating agents either alone or in combination with chemotherapy in vivo.
PRMT5, which symmetrically dimethylates arginine residues and is
One should keep in mind that murine models may not fully represent the
considered to be a transcriptional repressor, is required for sustaining
actual human situation. Xenotransplant models in which human leukemia
normal hematopoiesis.92 Dysregulated expression of several PRMTs,
cells are transplanted in immunodeficient mice provide an opportunity to
including PRMT5, has been observed in malignancies such as leuke-
study the response of the human leukemias. The combination of these
mia. Tyrosine phosphorylation at histone tails has been reported, but
different approaches may, at least in a subset of AML subtypes, provide
the mechanism by which this may occur and the function of histone
insight into responsiveness to DNMT inhibitors. Obviously the same
phosphorylation is poorly understood.93
strategy can be applied for other newly derived compounds or com-
binations of agents, either with or without chemotherapy.
Analyzing altered DNA methylation patterns of AML cells may
help to uncover critical target genes that upon methylation have become
Epigenetic targeted treatment of AML silenced in certain patient groups. Shih and colleagues recently dem-
onstrated how animal models can help to uncover such mechanisms.61
Taking into account the inherent reversibility of epigenetic marks,
In their study, Tet2-null mice that also carried Flt3-ITD mutations
disordered epigenetic regulation poses the chance to use targeted drugs.
developed AML and expressed a unique methylation signature. Gata2
The translation of such drugs to standard clinical use is still in an early
appeared to be silenced by methylation in those animals. Upon re-
stage. Compounds in various stages of preclinical and clinical devel-
expression of Gata2, the investigators reported reprogramming of the
opment are summarized in Table 2. We first describe drugs that are
tumor cells. It will be of interest to study the effects of DNMT inhibitors
currently available and then embark on approaches to identify novel
in this model and to study reprogramming of these Tet22/2/Flt3-ITD
targets.
leukemia cells. No specific therapy for leukemias with TET2 mutations
Drugs targeting disordered patterns of DNA cytosine has been developed yet; however, the observed hypermethylation sig-
methylation and hydroxymethylation
nature in patients as well as in animal models suggests that in particular
in this subtype of AML hypomethylating agents may be effective.114
Two epigenetic compounds currently approved for clinical use in Further studies in xenotransplant models may help to unravel the most
myeloid malignancies are 5-azacytidine (azacitidine) and 5-aza-29- optimal combination of drugs, which may also include compounds that
deoxycytidine (decitabine). Both are pyrimidine analogs that function target uncontrolled signaling by FLT3-ITD.
as inhibitors of DNA methyltransferases (Table 2). Azacitidine can be As indicated, IDH1/2 mutations are also associated with a dis-
converted into decitabine, which is incorporated into DNA. Azacitidine tinct methylation signature. IDH1/2 inhibitors have been devel-
itself is predominantly incorporated into RNA.106 Both drugs have been oped and are already being tested in early clinical trials (Table 2).
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

46 WOUTERS and DELWEL BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1

Table 2. Examples of epigenetic targeted therapy in AML in various stages of development


Class of epigenetic
regulator Target Compound Phase of development
DNA DNMTs Azacitidine Approved (see text)
methyltransferase Decitabine Approved (see text)
Rationally designed novel inhibitors Preclinical and clinical94,95
Regulator of IDH1, IDH2 Inhibitors of mutant IDH1/2 Clinical trials ongoing with compounds including IDH305 (ClinicalTrials.gov
methylation identifier: NCT02381886; targeted at IDH1 R132 mutation), AG-221
(NCT01915498; targeted at mutant IDH2), AG-120 (NCT02074839;
targeted at mutant IDH1)
Histone lysine CREBBP (CBP) CREBBP inhibitor Preclinical96
acetyltransferase EP300 (p300) EP300 inhibitor Preclinical97
Histone deacetylase HDACs HDAC inhibitors Several clinical trials ongoing, often in combination with other treatment
modalities (eg, with DNMT inhibitors [examples ClinicalTrials.gov
identifiers NCT01617226 and NCT00867672], conventional
chemotherapy [example NCT01802333], or in conjunction with allogeneic
stem cell transplantation [examples NCT01451268 and http://www.hovon.
nl/studies/studies-per-ziektebeeld/aml.html?
action5showstudie&studie_id5104&categorie_id54])
Histone acetyl Bromodomain BET inhibitors Several clinical trials ongoing with compounds including OTX-015
reader containing proteins (ClinicalTrials.gov identifier: NCT01713582), CPI-0610 (NCT02158858),
(BET proteins) TEN-010 (NCT02308761), GSK525762 (NCT01943851)
Histone lysine EZH2 EZH2 inhibitors Preclinical98,99
methyltransferase MLL-complexes DOT1L inhibitors Clinical trial with compounds including EPZ-5676 (ClinicalTrials.gov
identifier: NCT01684150)
Inhibitors of MLL-Menin interface Preclinical100
Inhibitors of MLL-LEDGF interface Preclinical101
Histone lysine LSD1 LSD1 inhibitors Clinical trials with compounds including GSK2879552 (ClinicalTrials.gov
demethylase identifier: NCT02177812) and tranylcypromine in combination with
tretinoine (NCT02261779)
Jumonji family of Small molecular inhibitors competitive for Preclinical102,103
KDMs 2-oxoglutarate
Histone arginine PRMTs PRMT inhibitors Preclinical104,105
methyltransferase

The outcomes of these studies are pending, but preliminary data are It will be challenging to find the optimal combinations of HDAC
promising.115 It was shown that AGI-6780, a selective inhibitor of the inhibitors with other agents. It is also likely that timing of treatment (eg,
IDH2 R140Q mutation, acts by binding an allosteric site located within concurrent vs subsequent administration) may affect responses—an
the dimer interface and induces differentiation of AML cell lines in aspect that will in fact be crucial for other epigenetic drugs as well.
vitro.116 Idh1/2-mutant animal models have been developed as well, Answering these questions will require proper in vivo models as
providing another opportunity to study the effects of the specific agents discussed earlier. It is also possible that targeting altered histone
at a molecular level. There is also experimental evidence that these acetylation or the consequences thereof has to be carried out in a
mutations may be targeted by alternative modes of action, such as completely different manner. This is supported by recent findings
BCL2 inhibitors.117 In addition, IDH1 R132H mutations result in a involving the targeting of transcription elongation factors such as
tumor-specific potential neoantigen that may offer a possibility of bromodomains proteins using small molecules. Bromodomain pro-
development of vaccination strategies that induce mutation-specific teins, including BET proteins, exert their function by binding acetylated
T-cell responses.118 lysine residues in histone proteins, such as H3K27. There are to date no
known mutations in bromodomain proteins in AML. However, BET
Drugs targeting disordered patterns of histone acetylation inhibitors showed therapeutic effects in various AML mouse models as
HDAC inhibitors were among the first epigenetic drugs developed, and well as in cell lines and small series of primary samples.9,70,127,128 BET
several have been tested in clinical trials for malignancies, including inhibitors may be effective in AML associated with MLL transloca-
AML.119 Responses to HDAC inhibitors as a single agent in AML or tions.70 Furthermore, it was recently demonstrated that in AML with
MDS appear to be modest.120 More recent work has therefore focused chromosome 3q abnormalities, a subtype with an adverse prognosis,
on combining these compounds with other drugs. A phase 2 study the leukemic translocation results in the formation of a novel super-
testing a combination of vorinostat with idarubicin and cytarabine sug- enhancer that drives overexpression of the EVI1 proto-oncogene.9
gested that the addition of an HDAC inhibitor to standard chemother- Indeed, AML t(3;3) or inv(3;3) cell lines appear sensitive to the BET
apy may lead to improved response rates in patients with newly inhibitor JQ1. Not surprisingly, given the large molecular heterogeneity
diagnosed AML and MDS.121 More recently, combinations of DNMT among AML, previous studies have shown that BET inhibitors are not
inhibitors and HDAC inhibitors have been tested.122-124 In a phase 2 uniformly active against all leukemia cell lines and patient samples.129
randomized trial, combination of the HDAC inhibitor valproic acid and BET inhibitors have entered phase 1/2 clinical trials (Table 2).130
low-dose decitabine did not lead to improved outcome in MDS and Studies in larger series will be needed to identify molecular features
AML patients.125 Moreover, in another phase 2 study, the HDAC linked to response to these drugs and to further clarify the mechanisms
inhibitor entinostat appeared even to be antagonistic when administered underlying the efficacy of BET inhibition in AML and its target genes
concurrently with azacitidine.126 and proteins. An important question that needs to be answered is how
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1 EPIGENETICS IN AML 47

to identify factors that differ between normal super-enhancers, specific epigenetic modulators may be predicted to be targetable in
which direct normal cellular functions, and oncogenic super- certain AML subtypes. Biochemical, molecular, and cell biological
enhancers, which are unique to particular types of malignancy. studies should be applied to uncover mechanisms of action of the
Insights into these differences would enable the development of identified compounds. An example of a compound that was iden-
more selective treatment approaches. A recent study describes tified to target a specific AML subset is the BET inhibitor JQ1.
phthalimide conjugation to the BET inhibitor JQ1 as a modified BET inhibitors in general show a higher affinity for super-
method to BET inhibition. In this approach, the BET inhibitor is used enhancers compared with normal (small) enhancers. Molecular and
to target BRD4, whereas the appended phthalimide moiety results in biochemical analyses revealed that JQ1 particularly targets the
hijacking of the cereblon E3 ubiquitin ligase complex, resulting in oncogenic super-enhancer that causes overexpression of the EVI1
targeted BRD4 degradation.131 transforming gene.9 The effects of BET inhibitors on leukemic
stem cells of super-enhancer–driven EVI1 AMLs should be tested
Drugs targeting disordered patterns of histone methylation in mouse models or in xenotransplant models.
A complementary approach involves the use of genetic tools
The histone demethylase LSD1 is highly expressed in hematopoietic
such as shRNA libraries or CRISPR/Cas9 technology targeting
cells and is specifically required for the terminal differentiation of
epigenetic regulators. These studies may be carried out in vitro
myeloid cells.132 LSD1 may be overexpressed in AML and appears to
as well as in vivo. Using an inducible shRNA library targeting
show a higher level of expression in less differentiated subtypes of the
approximately 250 epigenetic modulators in MLL-fusion mouse
disease. Recent reports have highlighted a potential application of
AML cells, Zuber and colleagues identified Brd4 as a potential
LSD1 inhibition in AML, and early clinical trials have been initiated
target for treatment in vivo.127 These studies pointed to the
(Table 2).133,134 MLL translocations result in fusion proteins that lack
potential usage of BET inhibitors for this specific AML subtype.
the wild-type SET domain and instead frequently recruit another lysine
As one may anticipate based on the partial response of leukemia
methyltransferase, DOT1L, which targets H3K79. DOT1L can be
cells to particular drugs, a third approach to identify novel targets
targeted by small-molecule inhibition.78,135 The first clinical trials in-
would be to combine the use of selected compounds (eg, BET
volving these compounds have been started and results have yet to
inhibitors) with genetic screening approaches such as shRNA.
follow (Table 2).
2. An alternative route would be to start with detailed maps of genetic
Together, single epigenetic drugs such as inhibitors of BET pro-
and epigenetic marks in particular AML subtypes. This way, one
teins, DOT1L, mutant IDH1/2, or DNMTs may show some significant
could prioritize certain drugs to be tested in distinct subtypes of the
effects. However, these responses willmostlikely be partial or transient.
disease. For instance, TET2 mutations have been associated with
Therefore, these novel compounds, derived from functional epige-
increased levels of cytosine methylation. This may be a rationale
nomics, need to be further studied in representative models in com-
for specifically testing DNMT inhibitors in those patients
bination with other drugs that target complementary pathways or with
(Figure 1). Indeed, patients with TET2 mutations may be more sus-
chemotherapy. Molecular and epigenetic studies are essential for a
ceptible to azacitidine than AML patients in general.114 Similarly,
better understanding of epigenetic changes that lead to leukemic trans-
one could compile genome-wide maps of super-enhancers by
formation, and these investigations can lead to novel drugs that target
applying ChIP-sequencing using antibodies directed against BRD4
those lesions. We therefore believe that further searching for novel
and H3K27. Identifying AML cases with particular super-enhancer
targets and molecules that recognize those elements will be crucial.
profiles could provide leads to test BET inhibitors specifically in
subsets of the disease. By combining genetic mapping studies with
screening studies using extensive libraries of compounds or genetic
libraries, it should be possible to match particular drugs to patient
How to identify novel targets for subgroups and to identify biomarkers.
epigenetic treatment? 3. A third approach is to use chemical-based engineering tech-
niques to design novel molecules predicted to specifically interfere
As was pointed out, the application of targeted epigenetic treatment is
with certain epigenetic processes. These types of structural ap-
still in an early phase. Still, there are already multiple examples of AML
proaches heavily rely on chemical methods such as crystallogra-
with unique epigenetic alterations, identified using integrated genetic
phy, frequently in combination with computer-based modeling.
and epigenetic analysis, that may be matched to appropriate compounds
This could result in the development of inhibitors with a relatively
(Figure 1). A number of strategies can be applied to identify novel tar-
high affinity for mutated proteins, as has been done for IDH1/2
gets and to match those to appropriate modes of targeted treatment.
mutations.116 Alternatively, the development of BET inhibitors and
1. A first strategy is to apply high-throughput compound screening derivates illustrates that also inhibitors of wild-type proteins may
approaches. Numerous compounds and small molecules are cur- be applicable in certain situations.131,136
rently available that interfere with particular known epigenetic
processes. Large-scale titration sensitivity experiments of these
molecules in representative series of primary AML specimens in
vitro, for instance using tritiated thymidine- or MTT assays, will Optimal applications of epigenetic therapy
provide compound response signatures of each AML sample.
AML patients may be classified based on these response signa- Despite great progress in the understanding of the molecular basis
tures (Figure 2). Because epigenetic changes in response to spe- of AML in the past years, treatment of the disease has essentially not
cific compounds may need several rounds of cell cycle, several shown major changes in the past decades. This review highlights the
time points should be assessed if possible (eg, 3, 7, and 14 days potential of compounds targeting epigenetic regulators in AML.
of culture). These experiments should be carried out under well- An important question is what the role of such compounds should be
defined conditions using appropriate combinations of hematopoi- in clinical practice. There are several challenges that may hamper the
etic growth stimuli. Based on the outcome of these experiments, clinical introduction of novel targeted therapies in general.137 Some of
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

48 WOUTERS and DELWEL BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1

Figure 1. Examples of integrated genetic and epi-


genetic analysis of human AML leading to potential
epigenetic treatment. The integrated analysis of gene
mutations, DNA methylation, and histone modifications
(A) has revealed multiple AML subtypes with specific
mutations in association with specific epigenetic alter-
ations. Four examples are indicated (B). Mutations in
IDH1/2 and TET2 result in aberrant patterns of DNA
cytosine methylation and hydroxymethylation (C),
whereas rearrangements affecting the EVI and MLL
loci result in alterations of chromatin marks (C). For
each of these 4 examples, certain epigenetic inhibitors
have been designed or are predicted to be active, of-
fering the potential to translate these findings to clinical
trials in human patients (D). The various inhibitors are
discussed in more detail in the text.

these challenges include the inherent problems in the translation of will be key, which may involve either combination with standard
preclinical findings to the clinic, the presence of multiple coactive chemotherapy or with other targeted therapies that are predicted to
deregulated pathways in the disease, and questions related to the target parallel critical pathways.
optimal design of clinical trials (how to best select patients to Together, epigenetic targeted treatment holds great promise. At the
include and which end points to address). In particular, the testing same time, there are various challenges that will have to be dealt with.
of novel targeted treatment in an isolated fashion may be problem- In the first place, many of the types of targeted treatment that will be
atic and may in fact underestimate the effectiveness of these novel applied will not be mutation-specific, but will rather target particular
compounds. It is reasonable to assume that combination therapy epigenetic regulating systems that are also active in nontransformed

Figure 2. Epigenetic modifier screen for human AML.


Numerous compounds and small molecules are currently
available that interfere with particular known epigenetic
processes. Large-scale titration sensitivity experiments of
these molecules in primary AML specimens in vitro can
provide compound response signatures of each AML
sample (A). Using those assays, AML subtypes may be
uncovered that uniformly respond to specific epigenetic
compounds (B). The molecular, epigenetic, and biological
effects to these AML cells may subsequently be studied in
vitro and in vivo (C). The outcomes of such experiments
should lead to the translation to clinical trials in human
patients (D).
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1 EPIGENETICS IN AML 49

cells, posing the risk of toxicity. Furthermore, the action of epigenetic


marks and regulators is often cell type–specific, the actual result being Acknowledgments
dependent on particular protein-protein interactions, which also carries
risks of off-target toxicity. Along the same lines, many chromatin- The authors thank Egied Simons for help with preparing the figures
modifying enzymes have more diverse functions, including a role in and Dr Peter Valk for critically reading the manuscript. They
modifying nonhistone substrates. The identification of biomarkers that apologize to authors whose work could not be cited due to space
can predict the susceptibility to certain epigenetic compounds will be limitations.
essential for a proper choice of epigenetic treatment of the distinct AML B.J.W. is a recipient of a Fellowship Award by the Dutch Cancer
subtypes.138 An additional challenge in the treatment of AML that has Society (K.W.F.) and a Special Fellowship Award by the Leukemia
emerged is the concept of clones and subclones.139,140 Targeted therapy & Lymphoma Society.
may be successful in eradicating some subclones, but may not be able
to eradicate subclones that do not harbor the particular epigenetic
abnormality. In this respect, it is promising to note that recent work has
indicated that many of the mutations that can be found in clonal
hematopoiesis are in epigenetic regulators such as TET2 and DNTM3A. Authorship
These mutations seem to enhance the self-renewal capacity of hema-
topoietic progenitor cells and may predispose individuals to the devel- Contribution: B.J.W. and R.D. wrote the manuscript.
opment of leukemia. Targeting such early lesions may eradicate the Conflict-of-interest disclosure: The authors declare no competing
founding clones. Still, it is most likely that the combination of financial interests.
several types of treatment (eg, epigenetic therapy in combination Correspondence: Ruud Delwel, Erasmus Medical Center, De-
with conventional chemotherapy) will have the best potential to be partment of Hematology, P.O. Box 2040, 3000 CA Rotterdam, The
successful. Netherlands; e-mail: h.delwel@erasmusmc.nl.

References
1. Ferrara F, Schiffer CA. Acute myeloid leukaemia monocytic leukemia. Nat Genet. 2011;43(4): without FLT3 internal tandem duplication. J Clin
in adults. Lancet. 2013;381(9865):484-495. 309-315. Oncol. 2010;28(22):3636-3643.
2. Burnett A, Wetzler M, Löwenberg B. Therapeutic 14. Marcucci G, Metzeler KH, Schwind S, et al. Age- 24. Marcucci G, Maharry K, Wu YZ, et al. IDH1 and
advances in acute myeloid leukemia. J Clin related prognostic impact of different types of IDH2 gene mutations identify novel molecular
Oncol. 2011;29(5):487-494. DNMT3A mutations in adults with primary subsets within de novo cytogenetically normal
cytogenetically normal acute myeloid leukemia. acute myeloid leukemia: a Cancer and Leukemia
3. Egger G, Liang G, Aparicio A, Jones PA.
J Clin Oncol. 2012;30(7):742-750. Group B study. J Clin Oncol. 2010;28(14):
Epigenetics in human disease and prospects for
2348-2355.
epigenetic therapy. Nature. 2004;429(6990): 15. Patel JP, Gönen M, Figueroa ME, et al.
457-463. Prognostic relevance of integrated genetic 25. Rozman M, Camós M, Colomer D, et al. Type I
profiling in acute myeloid leukemia. N Engl J MOZ/CBP (MYST3/CREBBP) is the most
4. Abdel-Wahab O, Levine RL. Mutations in
Med. 2012;366(12):1079-1089. common chimeric transcript in acute myeloid
epigenetic modifiers in the pathogenesis and
leukemia with t(8;16)(p11;p13) translocation.
therapy of acute myeloid leukemia. Blood. 2013; 16. Shen Y, Zhu YM, Fan X, et al. Gene mutation
Genes Chromosomes Cancer. 2004;40(2):
121(18):3563-3572. patterns and their prognostic impact in a cohort
140-145.
of 1185 patients with acute myeloid leukemia.
5. Fong CY, Morison J, Dawson MA. Epigenetics in
Blood. 2011;118(20):5593-5603. 26. Gervais C, Murati A, Helias C, et al; Groupe
the hematologic malignancies. Haematologica.
Francophone de Cytogénétique Hématologique.
2014;99(12):1772-1783. 17. Metzeler KH, Maharry K, Radmacher MD, et al.
Acute myeloid leukaemia with 8p11 (MYST3)
TET2 mutations improve the new European
6. Greenblatt SM, Nimer SD. Chromatin modifiers rearrangement: an integrated cytologic,
LeukemiaNet risk classification of acute myeloid
and the promise of epigenetic therapy in acute cytogenetic and molecular study by the groupe
leukemia: a Cancer and Leukemia Group B
leukemia. Leukemia. 2014;28(7):1396-1406. francophone de cytogénétique hématologique.
study. J Clin Oncol. 2011;29(10):1373-1381.
7. Cancer Genome Atlas Research Network. Leukemia. 2008;22(8):1567-1575.
18. Delhommeau F, Dupont S, Della Valle V, et al.
Genomic and epigenomic landscapes of adult 27. Ida K, Kitabayashi I, Taki T, et al. Adenoviral
Mutation in TET2 in myeloid cancers. N Engl J
de novo acute myeloid leukemia. N Engl J Med. E1A-associated protein p300 is involved in acute
Med. 2009;360(22):2289-2301.
2013;368(22):2059-2074. myeloid leukemia with t(11;22)(q23;q13). Blood.
19. Chou WC, Chou SC, Liu CY, et al. TET2 1997;90(12):4699-4704.
8. Figueroa ME, Lugthart S, Li Y, et al. DNA
mutation is an unfavorable prognostic factor
methylation signatures identify biologically 28. Rowley JD, Reshmi S, Sobulo O, et al. All
in acute myeloid leukemia patients with
distinct subtypes in acute myeloid leukemia. patients with the T(11;16)(q23;p13.3) that
intermediate-risk cytogenetics. Blood. 2011;
Cancer Cell. 2010;17(1):13-27. involves MLL and CBP have treatment-related
118(14):3803-3810.
hematologic disorders. Blood. 1997;90(2):
9. Gröschel S, Sanders MA, Hoogenboezem R,
20. Weissmann S, Alpermann T, Grossmann V, 535-541.
et al. A single oncogenic enhancer
et al. Landscape of TET2 mutations in acute
rearrangement causes concomitant EVI1 and 29. Krivtsov AV, Armstrong SA. MLL translocations,
myeloid leukemia. Leukemia. 2012;26(5):
GATA2 deregulation in leukemia. Cell. 2014; histone modifications and leukaemia stem-cell
934-942.
157(2):369-381. development. Nat Rev Cancer. 2007;7(11):
21. Gaidzik VI, Paschka P, Späth D, et al. TET2 823-833.
10. Jaiswal S, Fontanillas P, Flannick J, et al.
mutations in acute myeloid leukemia (AML):
Age-related clonal hematopoiesis associated 30. Byrd JC, Mrózek K, Dodge RK, et al; Cancer and
results from a comprehensive genetic and
with adverse outcome. N Engl J Med. Leukemia Group B (CALGB 8461). Pretreatment
clinical analysis of the AML study group. J Clin
2014;371(26):2488-2498. cytogenetic abnormalities are predictive of
Oncol. 2012;30(12):1350-1357.
induction success, cumulative incidence of
11. Ribeiro AF, Pratcorona M, Erpelinck-
22. Abbas S, Lugthart S, Kavelaars FG, et al. relapse, and overall survival in adult patients with
Verschueren C, et al. Mutant DNMT3A: a marker
Acquired mutations in the genes encoding IDH1 de novo acute myeloid leukemia: results from
of poor prognosis in acute myeloid leukemia.
and IDH2 both are recurrent aberrations in acute Cancer and Leukemia Group B (CALGB 8461).
Blood. 2012;119(24):5824-5831.
myeloid leukemia: prevalence and prognostic Blood. 2002;100(13):4325-4336.
12. Ley TJ, Ding L, Walter MJ, et al. DNMT3A value. Blood. 2010;116(12):2122-2126. 31. Grimwade D, Walker H, Harrison G, et al;
mutations in acute myeloid leukemia. N Engl J
23. Paschka P, Schlenk RF, Gaidzik VI, et al. IDH1 Medical Research Council Adult Leukemia
Med. 2010;363(25):2424-2433.
and IDH2 mutations are frequent genetic Working Party. The predictive value of
13. Yan XJ, Xu J, Gu ZH, et al. Exome sequencing alterations in acute myeloid leukemia and confer hierarchical cytogenetic classification in older
identifies somatic mutations of DNA adverse prognosis in cytogenetically normal adults with acute myeloid leukemia (AML):
methyltransferase gene DNMT3A in acute acute myeloid leukemia with NPM1 mutation analysis of 1065 patients entered into the United
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

50 WOUTERS and DELWEL BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1

Kingdom Medical Research Council AML11 trial. implications. Nat Rev Cancer. 2011;11(10): 65. Villa R, Morey L, Raker VA, et al. The methyl-
Blood. 2001;98(5):1312-1320. 726-734. CpG binding protein MBD1 is required for PML-
32. Slovak ML, Kopecky KJ, Cassileth PA, et al. RARalpha function. Proc Natl Acad Sci USA.
48. Shlush LI, Zandi S, Mitchell A, et al; HALT Pan-
Karyotypic analysis predicts outcome of 2006;103(5):1400-1405.
Leukemia Gene Panel Consortium. Identification
preremission and postremission therapy in adult of pre-leukaemic haematopoietic stem cells in 66. Santoro F, Botrugno OA, Dal Zuffo R, et al.
acute myeloid leukemia: a Southwest Oncology acute leukaemia. Nature. 2014;506(7488): A dual role for Hdac1: oncosuppressor in
Group/Eastern Cooperative Oncology Group 328-333. tumorigenesis, oncogene in tumor maintenance.
Study. Blood. 2000;96(13):4075-4083. Blood. 2013;121(17):3459-3468.
49. Russler-Germain DA, Spencer DH, Young MA,
33. Wang X, Dai H, Wang Q, et al. EZH2 mutations et al. The R882H DNMT3A mutation associated 67. Spensberger D, Vermeulen M, Le Guezennec X,
are related to low blast percentage in bone with AML dominantly inhibits wild-type DNMT3A et al. Myeloid transforming protein Evi1 interacts
marrow and -7/del(7q) in de novo acute myeloid by blocking its ability to form active tetramers. with methyl-CpG binding domain protein 3 and
leukemia. PLoS One. 2013;8(4):e61341. Cancer Cell. 2014;25(4):442-454. inhibits in vitro histone deacetylation by Mbd3/
34. Brown J, Jawad M, Twigg SR, et al. A cryptic t(5; Mi-2/NuRD. Biochemistry. 2008;47(24):
50. Challen GA, Sun D, Jeong M, et al. Dnmt3a
11)(q35;p15.5) in 2 children with acute myeloid 6418-6426.
is essential for hematopoietic stem cell
leukemia with apparently normal karyotypes, differentiation. Nat Genet. 2012;44(1):23-31. 68. Belkina AC, Denis GV. BET domain
identified by a multiplex fluorescence in situ co-regulators in obesity, inflammation
51. Mayle A, Yang L, Rodriguez B, et al. Dnmt3a
hybridization telomere assay. Blood. 2002;99(7): and cancer. Nat Rev Cancer. 2012;12(7):
loss predisposes murine hematopoietic stem
2526-2531. 465-477.
cells to malignant transformation. Blood. 2015;
35. Cerveira N, Correia C, Dória S, et al. Frequency 125(4):629-638. 69. Wu SY, Chiang CM. The double bromodomain-
of NUP98-NSD1 fusion transcript in childhood containing chromatin adaptor Brd4 and
52. Celik H, Mallaney C, Kothari A, et al. Enforced transcriptional regulation. J Biol Chem. 2007;
acute myeloid leukaemia. Leukemia. 2003;
differentiation of Dnmt3a-null bone marrow leads 282(18):13141-13145.
17(11):2244-2247.
to failure with c-Kit mutations driving leukemic
36. Paschka P, Schlenk RF, Gaidzik VI, et al. ASXL1 transformation. Blood. 2015;125(4):619-628. 70. Dawson MA, Prinjha RK, Dittmann A, et al.
mutations in younger adult patients with acute Inhibition of BET recruitment to chromatin as
myeloid leukemia: a study by the German- 53. Guryanova O, Shank K, Garrett-Bakelman F, an effective treatment for MLL-fusion leukaemia.
Austrian Acute Myeloid Leukemia Study Group. et al. DNMT3A R882 mutation promote Nature. 2011;478(7370):529-533.
Haematologica. 2015;100(3):324-330. chemoresistance and therapeutic relapse
through impaired DNA-damage sensing. EHA 71. Filippakopoulos P, Qi J, Picaud S, et al.
37. Chou WC, Huang HH, Hou HA, et al. Distinct Annual Meeting, Vienna, Austria, 2015. Selective inhibition of BET bromodomains.
clinical and biological features of de novo acute Nature. 2010;468(7327):1067-1073.
myeloid leukemia with additional sex comb-like 54. Lugthart S, Figueroa ME, Bindels E, et al.
Aberrant DNA hypermethylation signature in 72. Nicodeme E, Jeffrey KL, Schaefer U, et al.
1 (ASXL1) mutations. Blood. 2010;116(20): Suppression of inflammation by a synthetic
4086-4094. acute myeloid leukemia directed by EVI1. Blood.
2011;117(1):234-241. histone mimic. Nature. 2010;468(7327):
38. Metzeler KH, Becker H, Maharry K, et al. ASXL1 1119-1123.
mutations identify a high-risk subgroup of older 55. Subramanyam D, Belair CD, Barry-Holson KQ,
et al. PML-RARalpha and Dnmt3a1 cooperate in 73. Valent P, Zuber J. BRD4: a BET(ter) target for
patients with primary cytogenetically normal AML the treatment of AML? Cell Cycle. 2014;13(5):
within the ELN Favorable genetic category. vivo to promote acute promyelocytic leukemia.
Cancer Res. 2010;70(21):8792-8801. 689-690.
Blood. 2011;118(26):6920-6929.
56. Liu S, Shen T, Huynh L, et al. Interplay of 74. Shi J, Vakoc CR. The mechanisms behind the
39. Schnittger S, Eder C, Jeromin S, et al. ASXL1 therapeutic activity of BET bromodomain
exon 12 mutations are frequent in AML with RUNX1/MTG8 and DNA methyltransferase 1
in acute myeloid leukemia. Cancer Res. 2005; inhibition. Mol Cell. 2014;54(5):728-736.
intermediate risk karyotype and are
independently associated with an adverse 65(4):1277-1284. 75. Lovén J, Hoke HA, Lin CY, et al. Selective
outcome. Leukemia. 2013;27(1):82-91. inhibition of tumor oncogenes by disruption
57. Figueroa ME, Abdel-Wahab O, Lu C, et al.
of super-enhancers. Cell. 2013;153(2):320-334.
40. El-Sharkawi D, Ali A, Evans CM, et al. ASXL1 Leukemic IDH1 and IDH2 mutations result in
mutations are infrequent in young patients with a hypermethylation phenotype, disrupt TET2 76. Whyte WA, Orlando DA, Hnisz D, et al. Master
primary acute myeloid leukemia and their function, and impair hematopoietic transcription factors and mediator establish
detection has a limited role in therapeutic risk differentiation. Cancer Cell. 2010;18(6):553-567. super-enhancers at key cell identity genes. Cell.
stratification. Leuk Lymphoma. 2014;55(6): 2013;153(2):307-319.
58. Ward PS, Patel J, Wise DR, et al. The common
1326-1331. feature of leukemia-associated IDH1 and IDH2 77. Barski A, Cuddapah S, Cui K, et al. High-
41. Boultwood J, Perry J, Pellagatti A, et al. Frequent mutations is a neomorphic enzyme activity resolution profiling of histone methylations in the
mutation of the polycomb-associated gene converting alpha-ketoglutarate to 2- human genome. Cell. 2007;129(4):823-837.
ASXL1 in the myelodysplastic syndromes and in hydroxyglutarate. Cancer Cell. 2010;17(3): 78. Bernt KM, Zhu N, Sinha AU, et al. MLL-
acute myeloid leukemia. Leukemia. 2010;24(5): 225-234. rearranged leukemia is dependent on aberrant
1062-1065. 59. Busque L, Patel JP, Figueroa ME, et al. H3K79 methylation by DOT1L. Cancer Cell.
42. Micol JB, Duployez N, Boissel N, et al. Frequent Recurrent somatic TET2 mutations in normal 2011;20(1):66-78.
ASXL2 mutations in acute myeloid leukemia elderly individuals with clonal hematopoiesis. Nat 79. Ernst T, Chase AJ, Score J, et al. Inactivating
patients with t(8;21)/RUNX1-RUNX1T1 Genet. 2012;44(11):1179-1181. mutations of the histone methyltransferase gene
chromosomal translocations. Blood. 2014; 60. Rampal R, Alkalin A, Madzo J, et al. DNA EZH2 in myeloid disorders. Nat Genet. 2010;
124(9):1445-1449. hydroxymethylation profiling reveals that WT1 42(8):722-726.
43. Puda A, Milosevic JD, Berg T, et al. Frequent mutations result in loss of TET2 function in acute 80. Nikoloski G, Langemeijer SM, Kuiper RP, et al.
deletions of JARID2 in leukemic transformation myeloid leukemia. Cell Reports. 2014;9(5): Somatic mutations of the histone methyltransferase
of chronic myeloid malignancies. Am J Hematol. 1841-1855. gene EZH2 in myelodysplastic syndromes. Nat
2012;87(3):245-250. 61. Shih AH, Jiang Y, Meydan C, et al. Mutational Genet. 2010;42(8):665-667.
44. Beekman R, Valkhof MG, Sanders MA, et al. cooperativity linked to combinatorial epigenetic 81. Sashida G, Harada H, Matsui H, et al. Ezh2 loss
Sequential gain of mutations in severe gain of function in acute myeloid leukemia. promotes development of myelodysplastic
congenital neutropenia progressing to acute Cancer Cell. 2015;27(4):502-515. syndrome but attenuates its predisposition to
myeloid leukemia. Blood. 2012;119(22): 62. Wu H, Zhang Y. Mechanisms and functions of leukaemic transformation. Nat Commun. 2014;5:
5071-5077. Tet protein-mediated 5-methylcytosine oxidation. 4177.
45. de Rooij JD, Hollink IH, Arentsen-Peters ST, Genes Dev. 2011;25(23):2436-2452. 82. Neff T, Sinha AU, Kluk MJ, et al. Polycomb
et al. NUP98/JARID1A is a novel recurrent 63. Izutsu K, Kurokawa M, Imai Y, Maki K, Mitani K, repressive complex 2 is required for MLL-AF9
abnormality in pediatric acute megakaryoblastic Hirai H. The corepressor CtBP interacts with Evi- leukemia. Proc Natl Acad Sci USA. 2012;
leukemia with a distinct HOX gene expression 1 to repress transforming growth factor beta 109(13):5028-5033.
pattern. Leukemia. 2013;27(12):2280-2288. signaling. Blood. 2001;97(9):2815-2822. 83. Dixon JR, Jung I, Selvaraj S, et al. Chromatin
46. van Haaften G, Dalgliesh GL, Davies H, et al. 64. Senyuk V, Chakraborty S, Mikhail FM, Zhao R, architecture reorganization during stem cell
Somatic mutations of the histone H3K27 Chi Y, Nucifora G. The leukemia-associated differentiation. Nature. 2015;518(7539):
demethylase gene UTX in human cancer. Nat transcription repressor AML1/MDS1/EVI1 331-336.
Genet. 2009;41(5):521-523. requires CtBP to induce abnormal growth and 84. Chowdhury M, Mihara K, Yasunaga S, Ohtaki M,
47. Baylin SB, Jones PA. A decade of exploring the differentiation of murine hematopoietic cells. Takihara Y, Kimura A Expression of Polycomb-
cancer epigenome - biological and translational Oncogene. 2002;21(20):3232-3240. group. Expression of Polycomb-group (PcG)
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1 EPIGENETICS IN AML 51

protein BMI-1 predicts prognosis in patients with MLL-dependent leukemia. Cancer Res. 2014; myeloid leukemia. Nat Med. 2015;21(2):
acute myeloid leukemia. Leukemia. 2007;21(5): 74(18):5139-5151. 178-184.
1116-1122. 102. Sayegh J, Cao J, Zou MR, et al. Identification 118. Schumacher T, Bunse L, Pusch S, et al.
85. Mihara K, Chowdhury M, Nakaju N, et al. Bmi-1 of small molecule inhibitors of Jumonji AT-rich A vaccine targeting mutant IDH1 induces
is useful as a novel molecular marker for interactive domain 1B (JARID1B) histone antitumour immunity. Nature. 2014;512(7514):
predicting progression of myelodysplastic demethylase by a sensitive high throughput 324-327.
syndrome and patient prognosis. Blood. 2006; screen. J Biol Chem. 2013;288(13):
119. Wagner JM, Hackanson B, Lübbert M,
107(1):305-308. 9408-9417.
Jung M. Histone deacetylase (HDAC) inhibitors
86. Villa R, Pasini D, Gutierrez A, et al. Role of the 103. Kruidenier L, Chung CW, Cheng Z, et al. A in recent clinical trials for cancer therapy. Clin
polycomb repressive complex 2 in acute selective jumonji H3K27 demethylase inhibitor Epigenetics. 2010;1(3-4):117-136.
promyelocytic leukemia. Cancer Cell. 2007; modulates the proinflammatory macrophage
120. Quintás-Cardama A, Santos FP, Garcia-Manero
11(6):513-525. response. Nature. 2012;488(7411):404-408.
G. Histone deacetylase inhibitors for the
87. Boukarabila H, Saurin AJ, Batsché E, et al. The 104. Chan-Penebre E, Kuplast KG, Majer CR, et al. treatment of myelodysplastic syndrome and
PRC1 Polycomb group complex interacts with A selective inhibitor of PRMT5 with in vivo and in acute myeloid leukemia. Leukemia. 2011;25(2):
PLZF/RARA to mediate leukemic transformation. vitro potency in MCL models. Nat Chem Biol. 226-235.
Genes Dev. 2009;23(10):1195-1206. 2015;11(6):432-437.
121. Garcia-Manero G, Tambaro FP, Bekele NB,
88. Shi Y, Lan F, Matson C, et al. Histone 105. Alinari L, Mahasenan KV, Yan F, et al. et al. Phase II trial of vorinostat with idarubicin
demethylation mediated by the nuclear amine Selective inhibition of protein arginine and cytarabine for patients with newly diagnosed
oxidase homolog LSD1. Cell. 2004;119(7): methyltransferase 5 blocks initiation and acute myelogenous leukemia or myelodysplastic
941-953. maintenance of B-cell transformation. Blood. syndrome. J Clin Oncol. 2012;30(18):
89. Gruber TA, Larson Gedman A, Zhang J, et al. An 2015;125(16):2530-2543. 2204-2210.
Inv(16)(p13.3q24.3)-encoded CBFA2T3-GLIS2 106. Navada SC, Steinmann J, Lübbert M, Silverman 122. Lübbert M, Kuendgen A. Combining DNA
fusion protein defines an aggressive subtype LR. Clinical development of demethylating methyltransferase and histone deacetylase
of pediatric acute megakaryoblastic leukemia. agents in hematology. J Clin Invest. 2014; inhibition to treat acute myeloid leukemia/
Cancer Cell. 2012;22(5):683-697. 124(1):40-46. myelodysplastic syndrome: achievements
90. Cheung N, Chan LC, Thompson A, Cleary ML, 107. Huls G. Azacitidine in AML: a treatment option? and challenges. Cancer. 2015;121(4):
So CW. Protein arginine-methyltransferase- Blood. 2015;126(3):283-284. 498-501.
dependent oncogenesis. Nat Cell Biol. 2007; 123. Kirschbaum M, Gojo I, Goldberg SL, et al.
108. Dombret H, Seymour JF, Butrym A, et al.
9(10):1208-1215. A phase 1 clinical trial of vorinostat in
International phase 3 study of azacitidine vs
91. Yang Y, Bedford MT. Protein arginine conventional care regimens in older patients with combination with decitabine in patients with
methyltransferases and cancer. Nat Rev Cancer. newly diagnosed AML with .30% blasts. Blood. acute myeloid leukaemia or myelodysplastic
2013;13(1):37-50. 2015;126(3):291-299. syndrome. Br J Haematol. 2014;167(2):
185-193.
92. Liu F, Cheng G, Hamard PJ, et al. Arginine 109. Blum W, Garzon R, Klisovic RB, et al. Clinical
methyltransferase PRMT5 is essential for response and miR-29b predictive significance 124. Strati P, Kantarjian H, Ravandi F, et al. Phase I/II
sustaining normal adult hematopoiesis. J Clin in older AML patients treated with a 10-day trial of the combination of midostaurin (PKC412)
Invest. 2015;125(9):3532-3544. schedule of decitabine. Proc Natl Acad Sci USA. and 5-azacytidine for patients with acute myeloid
93. Dawson MA, Bannister AJ, Göttgens B, et al. 2010;107(16):7473-7478. leukemia and myelodysplastic syndrome. Am J
JAK2 phosphorylates histone H3Y41 and Hematol. 2015;90(4):276-281.
110. He J, Xiu L, De Porre P, Dass R, Thomas X.
excludes HP1alpha from chromatin. Nature. Decitabine reduces transfusion dependence in 125. Issa JP, Garcia-Manero G, Huang X, et al.
2009;461(7265):819-822. older patients with acute myeloid leukemia: Results of phase 2 randomized study of low-
94. Issa JP, Roboz G, Rizzieri D, et al. Safety and results from a post hoc analysis of a randomized dose decitabine with or without valproic acid in
tolerability of guadecitabine (SGI-110) in patients phase III study. Leuk Lymphoma. 2015;56(4): patients with myelodysplastic syndrome and
with myelodysplastic syndrome and acute 1033-1042. acute myelogenous leukemia. Cancer. 2015;
myeloid leukaemia: a multicentre, randomised, 121(4):556-561.
111. Itzykson R, Thépot S, Quesnel B, et al; Groupe
dose-escalation phase 1 study. Lancet Oncol. Francophone des Myelodysplasies(GFM). 126. Prebet T, Sun Z, Figueroa ME, et al. Prolonged
2015;16(9):1099-1110. Prognostic factors for response and overall administration of azacitidine with or without
95. Chen S, Wang Y, Zhou W, et al. Identifying novel survival in 282 patients with higher-risk entinostat for myelodysplastic syndrome and
selective non-nucleoside DNA methyltransferase myelodysplastic syndromes treated with acute myeloid leukemia with myelodysplasia-
1 inhibitors through docking-based virtual azacitidine. Blood. 2011;117(2):403-411. related changes: results of the US Leukemia
screening. J Med Chem. 2014;57(21):9028-9041. Intergroup trial E1905. J Clin Oncol. 2014;
112. Meldi K, Qin T, Buchi F, et al. Specific molecular
32(12):1242-1248.
96. Gang EJ, Hsieh YT, Pham J, et al. Small- signatures predict decitabine response in
molecule inhibition of CBP/catenin interactions chronic myelomonocytic leukemia. J Clin Invest. 127. Zuber J, Shi J, Wang E, et al. RNAi screen
eliminates drug-resistant clones in acute 2015;125(5):1857-1872. identifies Brd4 as a therapeutic target in acute
lymphoblastic leukemia. Oncogene. 2014; myeloid leukaemia. Nature. 2011;478(7370):
113. Traina F, Visconte V, Elson P, et al. Impact of
33(17):2169-2178. 524-528.
molecular mutations on treatment response to
97. Gao XN, Lin J, Ning QY, et al. A histone DNMT inhibitors in myelodysplasia and 128. Herrmann H, Blatt K, Shi J, et al. Small-molecule
acetyltransferase p300 inhibitor C646 induces related neoplasms. Leukemia. 2014;28(1): inhibition of BRD4 as a new potent approach to
cell cycle arrest and apoptosis selectively in 78-87. eliminate leukemic stem- and progenitor cells
AML1-ETO-positive AML cells. PLoS One. 2013; in acute myeloid leukemia AML. Oncotarget.
114. Itzykson R, Kosmider O, Cluzeau T, et al;
8(2):e55481. 2012;3(12):1588-1599.
Groupe Francophone des Myelodysplasies
98. Knutson SK, Wigle TJ, Warholic NM, et al. (GFM). Impact of TET2 mutations on response 129. Dawson MA, Gudgin EJ, Horton SJ, et al.
A selective inhibitor of EZH2 blocks H3K27 rate to azacitidine in myelodysplastic syndromes Recurrent mutations, including NPM1c, activate
methylation and kills mutant lymphoma cells. Nat and low blast count acute myeloid leukemias. a BRD4-dependent core transcriptional program
Chem Biol. 2012;8(11):890-896. Leukemia. 2011;25(7):1147-1152. in acute myeloid leukemia. Leukemia. 2014;
28(2):311-320.
99. Girard N, Bazille C, Lhuissier E, et al. 115. Stein EM, Altman JK, Collins R, et al. AG-221,
3-Deazaneplanocin A (DZNep), an inhibitor of an oral, selective, first-in-class, potent inhibitor 130. Dombret H, Preudhomme C, Berthon C, et al.
the histone methyltransferase EZH2, induces of the IDH2 mutant metabolic enzyme, induces A phase 1 study of the BET-bromodomain
apoptosis and reduces cell migration in durable remissions in a phase I study in patients inhibitor OTX015 in patients with advanced
chondrosarcoma cells. PLoS One. 2014;9(5): with IDH2 mutation positive advanced acute leukemia. Blood. 2014;124(21):
e98176. hematologic malignancies. Blood. 2014;124(21): 117-117.
115-115. 131. Winter GE, Buckley DL, Paulk J, et al. Phthalimide
100. Grembecka J, He S, Shi A, et al. Menin-MLL
inhibitors reverse oncogenic activity of MLL 116. Wang F, Travins J, DeLaBarre B, et al. Targeted conjugation as a strategy for in vivo target
fusion proteins in leukemia. Nat Chem Biol. inhibition of mutant IDH2 in leukemia cells protein degradation. Science. 2015;348(6241):
2012;8(3):277-284. induces cellular differentiation. Science. 2013; 1376-1381.
101. Cerm áková K, Tesina P, Demeulemeester J, 340(6132):622-626. 132. Lynch JT, Harris WJ, Somervaille TC. LSD1
et al. Validation and structural 117. Chan SM, Thomas D, Corces-Zimmerman MR, inhibition: a therapeutic strategy in cancer?
characterization of the LEDGF/p75-MLL et al. Isocitrate dehydrogenase 1 and 2 Expert Opin Ther Targets. 2012;16(12):
interface as a new target for the treatment of mutations induce BCL-2 dependence in acute 1239-1249.
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

52 WOUTERS and DELWEL BLOOD, 7 JANUARY 2016 x VOLUME 127, NUMBER 1

133. Lokken AA, Zeleznik-Le NJ. Breaking the LSD1/ by a potent small-molecule DOT1L inhibitor. 138. Treppendahl MB, Kristensen LS, Grønbæk K.
KDM1A addiction: therapeutic targeting of the Cancer Cell. 2011;20(1):53-65. Predicting response to epigenetic therapy.
epigenetic modifier in AML. Cancer Cell. 2012; J Clin Invest. 2014;124(1):47-55.
136. Delmore JE, Issa GC, Lemieux ME, et al. BET
21(4):451-453.
bromodomain inhibition as a therapeutic 139. Klco JM, Spencer DH, Miller CA, et al.
134. Schenk T, Chen WC, Göllner S, et al. strategy to target c-Myc. Cell. 2011;146(6): Functional heterogeneity of genetically defined
Inhibition of the LSD1 (KDM1A) demethylase 904-917. subclones in acute myeloid leukemia. Cancer
reactivates the all-trans-retinoic acid Cell. 2014;25(3):379-392.
137. Estey E, Levine RL, L öwenberg B. Current
differentiation pathway in acute myeloid
challenges in clinical development of 140. Wong TN, Ramsingh G, Young AL, et al. Role of
leukemia. Nat Med. 2012;18(4):605-611.
“targeted therapies”: the case of acute TP53 mutations in the origin and evolution
135. Daigle SR, Olhava EJ, Therkelsen CA, et al. myeloid leukemia. Blood. 2015;125(16): of therapy-related acute myeloid leukaemia.
Selective killing of mixed lineage leukemia cells 2461-2466. Nature. 2015;518(7540):552-555.
From www.bloodjournal.org by guest on July 24, 2016. For personal use only.

2016 127: 42-52


doi:10.1182/blood-2015-07-604512 originally published
online December 10, 2015

Epigenetics and approaches to targeted epigenetic therapy in acute


myeloid leukemia
Bas J. Wouters and Ruud Delwel

Updated information and services can be found at:


http://www.bloodjournal.org/content/127/1/42.full.html
Articles on similar topics can be found in the following Blood collections
Clinical Trials and Observations (4361 articles)
Free Research Articles (3946 articles)
Myeloid Neoplasia (1519 articles)
Review Articles (645 articles)
Review Series (109 articles)

Information about reproducing this article in parts or in its entirety may be found online at:
http://www.bloodjournal.org/site/misc/rights.xhtml#repub_requests

Information about ordering reprints may be found online at:


http://www.bloodjournal.org/site/misc/rights.xhtml#reprints

Information about subscriptions and ASH membership may be found online at:
http://www.bloodjournal.org/site/subscriptions/index.xhtml

Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly by the American Society
of Hematology, 2021 L St, NW, Suite 900, Washington DC 20036.
Copyright 2011 by The American Society of Hematology; all rights reserved.

Das könnte Ihnen auch gefallen