The past 15 years have seen major leaps in our understanding of the molecular genetic mutations that act as drivers of acute myeloid leukemia (AML). Clinical trials of agents against specific mutant proteins, such as FLT3-internal tandem duplications (ITDs) and isocitrate dehydrogenase mutations (IDHs) are ongoing. This review discusses agents in clinical trials that target specific gene mutations and/or epigenetic targets.

Learning Objectives
  • A variety of pharmaceuticals against specific genetic and epigenetic targets are in clinical trials

  • Clinical trials using these targeted agents show promising early results

  • The future treatment of AML will likely incorporate novel agents in combination or with chemotherapy

Acute myeloid leukemia (AML) is an acquired disease of chromosomal translocations and somatic mutations, with single or multiple genetic events operating alone or together to produce the phenotype of differentiation block and proliferation at the myeloblast stage of hematopoiesis. The revolution in techniques and the reduction in the costs of next-generation sequencing (NGS) of DNA allows us to examine with extraordinary detail, the contribution of gene mutations to the pathogenesis of newly diagnosed, refractory, and relapsed AML. Techniques, such as CHIP-seq, have given us unprecedented ability to understand the epigenetic state of leukemic myeloblasts. Unlike solid malignancies, patients with AML have relatively few, perhaps 30, common, recurrent, genetic alterations.1,2  The genetic alterations with multiple retrospective studies supporting their prognostic import, NPM1, FLT3, and CEBPA, are incorporated into the current World Health Organization guidelines on the diagnosis of AML.3  Mutations found in genes such as c-KIT, isocitrate dehydrogenase (IDH1 and IDH2) and FLT-3 provide druggable molecular targets in patients with AML that may improve rates of complete remission (CR), disease-free survival (DFS), and overall survival (OS). In addition, inhibitors of proteins involved in regulating the epigenetic state of cells, such as the histone methyltransferase DOT1L and inhibitors of BET proteins show evidence of clinical activity in ongoing phase 1 studies (Table 1).

Table 1.

Response rate in clinical trials of targeted agents for the treatment of AML

Response rate in clinical trials of targeted agents for the treatment of AML
Response rate in clinical trials of targeted agents for the treatment of AML

Note that response criteria between trials do not necessarily use the international working group criteria and therefore, between trial comparisons are difficult to make.

The therapeutic limitations of our current approach to treating newly diagnosed “fit” patients with AML using induction chemotherapy are well known; multiple international retrospective studies show that only 40% of patients younger than age 60 survive >5 years.4  Indeed, even “favorable risk” core binding factor leukemias have an unacceptably high mortality rate of 40% at 5 years.5  The prognosis for patients who relapse after achieving a CR is dismal and patients older than age 60 fare little better. The de facto pharmacologic options for “unfit” patients, low-dose cytarabine or hypomethylating agents, are modestly effective but inadequate.6,7  The inhibition of molecular genetic pathways in AML is in its infancy, but they provide an early look at how we may use small molecule inhibitors to treat AML in the next few years.

In this brief monograph, I present an overview of the most exciting targeted molecular therapies that are currently in clinical trials and give a roadmap to what the treatment of leukemia may look like 5 years from now. This review is not meant to be exhaustive; the landscape of targets is well known and new drugs available against these targets are introduced frequently. Rather, I focus on therapies that have clinical data available and that I believe may make it into clinical practice as single agents in the next 5 years. Targets not addressed in this review are summarized in Table 2.

Table 2.

Other molecular targets with agents in clinical trials

Other molecular targets with agents in clinical trials
Other molecular targets with agents in clinical trials

FLT3 internal tandem duplications (FLT3-ITDs) are seen in ∼30% of patients with de novo AML. Patients with FLT3-ITD have a poor prognosis.8,9  Given the prevalence of FLT3-ITD and the suboptimal outcomes, the development of inhibitors of mutant FLT3 is seen as a priority in curing patients with FLT3–positive disease. Efforts to target AML with FLT-3-ITD are ongoing. Despite this effort and a number of “first-generation” FLT-3 inhibitors, both those that are used off-label (Sorafenib and Sunitinib) and have been explored in late stage clinical trials (Lestartinib, Midostaurin), the search for a novel inhibitor of FLT-3 that leads to an overall survival benefit compared with chemotherapy continues. Previous ASH education sessions provided a comprehensive review of treating FLT-3–positive disease.10  The state-of-the-art, however, and excitement, revolves around three drugs in clinical trials, Quizartinib, Crenolanib, and ASP-2215.

Formerly known as AC220, Quizartinib was developed expressly to treat FLT3 mutation-positive AML as a “second-generation” FLT3 inhibitor.11  It is both a highly selective and potent inhibitor of FLT3 mutated AML in vitro and in xenograft models of disease. A phase I study of Quizartinib in patients with relapsed or refractory AML, regardless of FLT3 mutation status, was completed using both intermittent (2 weeks on and 2 weeks off) and continuous dosing in sequential 28 day cycles. The maximum tolerated dose was not reached in the intermittent dosing cohorts and was determined to be 200 mg/day in the continuous dosing cohort.12  A variety of phase II studies have been conducted using Quizartinib in the relapsed and refractory setting (see References).13-15  What is remarkable about all of these studies, whether patients were in first relapse or later, young or old, is the relatively remarkable composite complete remission rate, partial remission rate, and median duration of response. In all of the phase II studies using Quizartinib as a single agent for relapsed/refractory disease the CRc (composite complete remission rate that includes CR, CRi, and CRp) ranged between 44% and 54%, whereas the overall response rate (CRc + PR) was between 61% and 72%. Of crucial importance, the PR definition did not require normalization of platelet and neutrophil counts as defined in the International Working Group Criteria. Median duration of response ranged between 11.3 and 12.7 weeks.

Despite the single-agent activity of Quizartinib, 50% of patients relapse within 3 months. Further studies suggest that the mechanism of resistance to Quizartinib is the development of acquired mutations in the tyrosine kinase domain of the FLT2 gene, including mutations in D835 and F691. Because of this, agents that can overcome this resistance and lead to a longer duration of response are seen as crucial to the development of targeted inhibitors of mutant FLT3.

The tyrosine kinase inhibitor, Crenolanib, initially developed as an inhibitor of PDGFR, was demonstrated to have activity against mutations in the activation loop of FLT3, the most common being a substitution at amino acid D835.16  As the primary mechanism of resistance to the FLT-3 inhibitor Quizartinib (AC220) is development of TKD mutations in the activation loop, it was hypothesized that Crenolanib would be a pan selective FLT-3 inhibitor that would overcome Quizartinib resistance. A phase II study of Crenolanib in patients with relapsed or refractory AML and a FLT3-ITD or FLT3-TKD was presented at the ASH annual meeting in 2014.17  Crenolanib was administered at a dose of 200 mg/m2/d, 3 times/d in continuous 28 day cycles, and patients were stratified based on whether they had received prior FLT3 directed therapy (eg, Quizartinib, Midostaurin, Sorafenib, PLX3397).

Crenolanib induced a complete remission with incomplete blood count recovery (CRi) in 23% of patients who were FLT3 inhibitor naïve, but in only 5% of patients who had received prior FLT3 therapy. Although both treatment groups had similar degrees of hematologic improvement (31% and 33%, respectively), it is unclear whether that translated into clinically significant benefit for the patients. Crenolanib is now being investigated in combination with induction chemotherapy in patients with newly diagnosed AML with a FLT3-ITD or TKD (NCT02283177).

ASP-2215 is a potent inhibitor of both FLT3-ITD and FLT3-TKD mutations. A phase I/II trial was initiated in 2013 and interim results were reported in 2015.18  One-hundred sixty-six patients were enrolled in a combination of the dose escalation and in-parallel dose expansion cohorts and the maximum tolerated dose was 300 mg daily. Of note, both FLT3 wild-type and FLT3 mutant patients were enrolled on the study. FLT3 wild-type patients derived minimal benefit from the study drug with a composite complete remission rate (encompassing CR, CRi, and CRp) of 8% and a PR rate of 3%. In the FLT3 mutant patient population, the overall response rate was 57% with a composite CR rate of 43% and a partial remission rate of 15%. A phase 1 study of ASP2215 in combination with induction consolidation chemotherapy is ongoing (NCT02236013) and a randomized phase III study of ASP2215 versus salvage chemotherapy is planned (NCT02421939).

IDH, the enzyme that converts isocitrate to alpha-ketoglutarate in the mitochondria (IDH2) or the cytoplasm (IDH1) as part of the citric acid cycle, is mutated in a subset of patients with de novo adult AML in retrospective analyses of large clinical trial databases. IDH2 is mutated in 10%-15% of patients and IDH1 is mutated in 5%-10% of adult AML.19-21  The prevalence of both IDH mutations appears to increase as patients age; IDH mutations have not been found, to date, in any patients with pediatric AML. Mutations in IDH2 are enriched in patients with normal karyotypes, but up to 30% of patients with IDH2 mutations have abnormal cytogenetics at the time of diagnosis that generally fall into the intermediate or unfavorable cytogenetic risk groups as defined by the National Comprehensive Cancer Network (NCCN). Elegant preclinical work done over the past 5 years has shown that the mutant IDH enzymes acquire neomorphic activity and catalyze the conversion of alpha-ketoglutarate into beta-hydroxygutarate (2-HG), elevate levels of 2-HG, and lead to dysregulation of target genes in the affected myeloblasts, leading to a block in differentiation and clinical AML.22-24  Inhibitors of mutant IDH1 and mutant IDH2 are currently in phase 1 clinical trials (NCT02381886, NCT01915498, NCT02074839) and early results have demonstrated that these agents have encouraging efficacy in patients with relapsed disease.

Interim results of a phase I/II study of AG-221 (Agios/Celgene), the first IDH-2 inhibitor currently in clinical trials, presented at the annual meeting of the European Hematology Association in 2015 demonstrated an overall response rate of 41%, in patients with relapsed/refractory AML.48 Twenty-seven percent of patients cleared their bone marrow of blasts with various levels of count recovery (true CR, CRi, and morphologic leukemia-free states) and 18% of patients achieved a true CR. An additional 14% of the patients had a true PR with normalization of platelet count and absolute neutrophil count (ANC). Seventy-six percent of the responding patients have been on treatment for at least 6 months. Interestingly, an additional 44% of patients had clinical stable disease, defined as a stable or decreased blast percentage in the bone marrow that does not meet the criteria for PR. Some of these stable disease patients are red cell transfusion independent, have normal platelet counts and a normal ANC, despite persistence of blasts in the bone marrow and peripheral blood. The number of patients with clinically meaningful stable disease, and those who are simply smoldering AML is an area of active investigation as it has implications for the OS benefit that may be observed in these patients.

Inhibitors of mutant IDH1 in clinical development include AG-120 (Agios) and IDH305 (Novartis). Early results of AG-120 in patients with relapsed AML have shown similar evidence of efficacy as the IDH-2 inhibitor, with an overall response rate of 31% and a true CR rate of 15%. An additional 27 patients had stable disease. Dose escalation continues and expansion cohorts in patients with relapsed/refractory AML have been initiated.

A variety of agents against non-mutated molecular targets are in the midst of clinical trials including BET inhibitors, DOT1L inhibitors, and the BCL-2 inhibitor ABT-199. The early results for these agents have show responses, but their activity is modest. Because of that, it is uncertain if these compounds will move forward as single agents for the treatment of AML. For example, the BCL-2 inhibitor ABT-199 showed a CR/CRi in 5 of 32 patients, the majority of whom had relapsed or refractory disease. Interestingly, 3 of the 5 patients with a CR/CRi had an IDH mutation.25  Although there is preclinical data suggesting that IDH1 and IDH2 mutations induce BCL-2 dependence in AML and inhibition of BCL-2 may be therapeutically effective, the small numbers of responders in this early study makes any statement about the clinical effectiveness of BCL-2 inhibition in IDH mutant AML premature.26 

Similarly, preclinical studies of DOT1L inhibition in acute leukemia associated with translocations involving the mixed lineage leukemia (MLL) gene, show remarkable effectiveness in preclinical studies. Translating these remarkable results to patients has been more difficult. Inhibition of DOT1L with the small molecule EPZ-5676 produced a complete remission in 2 of 34 patients with an MLL rearrangement or MLL-partial tandem duplication. In 1 patient, not only did morphologic evidence of myeloid disease disappear, but leukemia cutis resolved and the patient achieved a cytogenetic remission.27 

Similar to the excitement around DOT1L inhibition, small molecule inhibition of BET proteins has a robust preclinical rationale that is currently being tested in multiple clinical trials for AML (NCT02158858, NCT02308761, NCT01943851).28-30  To date, the only report from a clinical trial has been of the bromodomain inhibitor OTX015.31  In this study, 36 patients with relapsed and refractory leukemia (33 with AML, 2 with ALL, and 1 with MDS) were enrolled on this phase 1 dose escalation study and 28 were evaluable for dose limiting toxicity. Of these, 1 patient had a CRp and 1 patient had a true CR. Three other patients had evidence of clinical activity (decrease in blast percentage and resolution of gingival hypertrophy).

The results above speak to the difficulty of translating robust preclinical studies into therapeutically effective treatments in the clinic. The path forward for each of these drugs is either to identify a biomarker of response, or to consider combining these agents with other drugs that are synergistic.

Clonal evolution

When thinking about molecularly targeted therapies for AML we encounter the issue described in the introduction: AML is molecularly heterogeneous. The proliferative advantage of leukemic myeloblasts may shift away from the target that is suppressed by a novel agent, to a new molecular genetic alteration. This leads to the potential of a never-ending cascade of new gene mutations arising that need to be suppressed and old mutations that have been suppressed re-emerging to cause a relapse. Indeed, a classic example of this seen in routine clinical practice is the emergence of overt FLT3-ITD mutations in patients with relapsed AML. In addition, many patients with FLT3-ITD mutations treated with FLT-3 inhibitors are unable to achieve a true complete remission because FLT-3 is one of many drivers of disease. Finally, the clone that needs to be targeted, the so called “founder clone,” may have genetic alterations that are not discoverable with routine clinical assays.

Our ability to identify and target the founder clone may change in the next decade. With current technology, there are 2 potential ways to overcome therapeutic resistance. The first is to use targeted molecular agents in combination rather than sequentially. For example, patients with a FLT-3 ITD and IDH mutations can be targeted with both FLT3 and IDH inhibitors. This “off the shelf” approach of mixing and matching targets is attractive intellectually but realistically problematic. This, primarily, because we do not have the agents for many of the targets. Although these drugs will hopefully be developed over the next 5-10 years, a comprehensive approach to clinical trials that involves multiple pharmaceutical companies that test combination therapies is needed to quickly develop phase 1 trials that test combinations.

Given the inherent limitations of targeted therapy for AML, perhaps the ideal way to eradicate leukemia and produce cures is combining targeted therapy with chemotherapy or hypomethylating agents. The historical precedent for this suggestion is the example of the differentiating agent, all-trans retinoic acid (ATRA) for APL. Interestingly, single agent use of ATRA is remarkably effective, but the 3 year overall survival rate with ATRA alone is only 71%; this in a disease where the founder clone contains the t(15:17) translocation.32  Until the recent introduction of ATRA in combination with ATO the most effective way of curing APL was combining ATRA with traditional cytotoxic chemotherapy. Given the precedent, those patients with de novo AML, fit for induction chemotherapy, should be enrolled on clinical studies that combine induction chemotherapy with an inhibitor of a desired molecular target.

With varying degrees of potency, efficacy and durability, inhibitors of FLT-3, IDH1, IDH2, DOT1L, BET proteins, and ABT-199 demonstrate proof of concept that the molecular genetic alterations that drive AML, are targetable. However, AML is rarely driven by 1 genetic event and targeting a single mutation is unlikely to lead to clinical cure. Because AML arises from a complex set of molecular genetic events with competing clones that vie for a selective advantage, it is extraordinarily unlikely that we will ever treat AML with a single, imatinib-like drug. In addition, clonal evolution between diagnosis and relapse makes it unlikely that those same molecularly targeted drugs that put a patient into remission at the time of diagnosis will produce a CR at the time of relapse. In addition, with any of these molecularly targeted agents, especially those that lead to differentiation, will these agents cure AML, or simply turn an acute disease in to a chronic disease.

Eytan M. Stein, Leukemia Service, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065; Phone: 212-639-3314; Fax: 212-772-8550; e-mail: steine@mskcc.org.

1
Patel
 
JP
Gonen
 
M
Figueroa
 
ME
, et al. 
Prognostic relevance of integrated genetic profiling in acute myeloid leukemia
N Engl J Med
2012
, vol. 
366
 
12
(pg. 
1079
-
1089
)
2
Lindsley
 
RC
Mar
 
BG
Mazzola
 
E
, et al. 
Acute myeloid leukemia ontogeny is defined by distinct somatic mutations
Blood
2015
, vol. 
125
 
9
(pg. 
1367
-
1376
)
3
Vardiman
 
JW
Thiele
 
J
Arber
 
DA
, et al. 
The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes
Blood
2009
, vol. 
114
 
5
(pg. 
937
-
951
)
4
Burnett
 
A
Wetzler
 
M
Lowenberg
 
B
Therapeutic advances in acute myeloid leukemia
J Clin Oncol
2011
, vol. 
29
 
5
(pg. 
487
-
494
)
5
Bloomfield
 
CD
Lawrence
 
D
Byrd
 
JC
, et al. 
Frequency of prolonged remission duration after high-dose cytarabine intensification in acute myeloid leukemia varies by cytogenetic subtype
Cancer Res
1998
, vol. 
58
 
18
(pg. 
4173
-
4179
)
6
Kantarjian
 
HM
Thomas
 
XG
Dmoszynska
 
A
, et al. 
Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia
J Clin Oncol
2012
, vol. 
30
 
21
(pg. 
2670
-
2677
)
7
Dombret
 
H
Seymour
 
JF
Butrym
 
A
, et al. 
International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts
Blood
2015
, vol. 
126
 
3
(pg. 
291
-
299
)
8
Kottaridis
 
PD
Gale
 
RE
Frew
 
ME
, et al. 
The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials
Blood
2001
, vol. 
98
 
6
(pg. 
1752
-
1759
)
9
Frohling
 
S
Schlenk
 
RF
Breitruck
 
J
, et al. 
Prognostic significance of activating FLT3 mutations in younger adults (16 to 60 years) with acute myeloid leukemia and normal cytogenetics: a study of the AML Study Group Ulm
Blood
2002
, vol. 
100
 
13
(pg. 
4372
-
4380
)
10
Levis
 
M
FLT3 mutations in acute myeloid leukemia: what is the best approach in 2013?
Hematology Am Soc Hematol Educ Program
2013
, vol. 
2013
 
1
(pg. 
220
-
226
)
11
Zarrinkar
 
PP
Gunawardane
 
RN
Cramer
 
MD
, et al. 
AC220 is a uniquely potent and selective inhibitor of FLT3 for the treatment of acute myeloid leukemia (AML)
Blood
2009
, vol. 
114
 
14
(pg. 
2984
-
2992
)
12
Cortes
 
JE
Kantarjian
 
H
Foran
 
JM
, et al. 
Phase I study of quizartinib administered daily to patients with relapsed or refractory acute myeloid leukemia irrespective of FMS-like tyrosine kinase 3-internal tandem duplication status
J Clin Oncol
2013
, vol. 
31
 
29
(pg. 
3681
-
3687
)
13
Levis
 
MJ
Perl
 
AE
Dombret
 
H
, et al. 
Final results of a phase 2 open-label, monotherapy efficacy and safety study of quizartinib (AC220) in patients with FLT3-ITD positive or negative relapsed/refractory acute myeloid leukemia after second-line chemotherapy or hematopoietic stem cell transplantation
ASH Annual Meeting Abstracts
2012
, vol. 
120
 
21
 
Abstract 673
14
Cortes
 
JE
Perl
 
AE
Dombret
 
H
, et al. 
Final results of a phase 2 open-label, monotherapy efficacy and safety study of quizartinib (AC220) in patients ≥60 years of age with FLT3 ITD positive or negative relapsed/refractory acute myeloid leukemia
ASH Annual Meeting Abstracts
2012
, vol. 
120
 
21
 
Abstract 48
15
Schiller
 
GJ
Tallman
 
MS
Goldberg
 
SL
, et al. 
Final results of a randomized phase 2 study showing the clinical benefit of quizartinib (AC220) in patients with FLT3-ITD positive relapsed or refractory acute myeloid leukemia
J Clin Oncol
2014
, vol. 
32
 
Suppl
pg. 
5s
  
Abstract 7100
16
Smith
 
CC
Lasater
 
EA
Lin
 
KC
, et al. 
Crenolanib is a selective type I pan-FLT3 inhibitor
Proc Natl Acad Sci U S A
2014
, vol. 
111
 
14
(pg. 
5319
-
5324
)
17
Randhawa
 
JK
Kantarjian
 
HM
Borthakur
 
G
, et al. 
Results of a phase ii study of crenolanib in relapsed/refractory acute myeloid leukemia patients (Pts) with activating FLT3 mutations
Blood
2014
, vol. 
124
 
21
pg. 
389
 
18
Levis
 
MJ
Pearl
 
AE
Altman
 
JK
, et al. 
Results of a first-in-human, phase I/II trial of ASP2215, a selective, potent inhibitor of FLT3/Axl in patients with relapsed or refractory (R/R) acute myeloid leukemia (AML)
J Clin Oncol
2015
, vol. 
33
 
suppl
 
Abstract 7003
19
Green
 
CL
Evans
 
CM
Zhao
 
L
, et al. 
The prognostic significance of IDH2 mutations in AML depends on the location of the mutation
Blood
2011
, vol. 
118
 
2
(pg. 
409
-
412
)
20
Paschka
 
P
Schlenk
 
RF
Gaidzik
 
VI
, et al. 
IDH1 and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and confer adverse prognosis in cytogenetically normal acute myeloid leukemia with NPM1 mutation without FLT3 internal tandem duplication
J Clin Oncol
2010
, vol. 
28
 
22
(pg. 
3636
-
3643
)
21
Marcucci
 
G
Maharry
 
K
Wu
 
YZ
, et al. 
IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study
J Clin Oncol
2010
, vol. 
28
 
14
(pg. 
2348
-
2355
)
22
Ward
 
PS
Patel
 
J
Wise
 
DR
, et al. 
The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate
Cancer Cell
2010
, vol. 
17
 
3
(pg. 
225
-
234
)
23
Figueroa
 
ME
Abdel-Wahab
 
O
Lu
 
C
, et al. 
Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation
Cancer Cell
2010
, vol. 
18
 
6
(pg. 
553
-
567
)
24
Lu
 
C
Ward
 
PS
Kapoor
 
GS
, et al. 
IDH mutation impairs histone demethylation and results in a block to cell differentiation
Nature
2012
, vol. 
483
 
7390
(pg. 
474
-
478
)
25
Konopleva
 
M
Pollyea
 
DA
Potluri
 
J
, et al. 
A phase 2 study of ABT-199 (GDC-0199) in patients with acute myelogenous leukemia (AML)
Blood
2014
, vol. 
124
 
21
pg. 
118
 
26
Chan
 
SM
Thomas
 
D
Corces-Zimmerman
 
MR
, et al. 
Isocitrate dehydrogenase 1 and 2 mutations induce BCL-2 dependence in acute myeloid leukemia
Nat Med
2015
, vol. 
21
 
2
(pg. 
178
-
184
)
27
Stein
 
EM
Garcia-Manero
 
G
Rizzieri
 
DA
, et al. 
The DOT1L inhibitor EPZ-5676: safety and activity in relapsed/refractory patients with MLL-rearranged leukemia
Blood
2014
, vol. 
124
 
21
pg. 
387
 
28
Zuber
 
J
Shi
 
J
Wang
 
E
, et al. 
RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia
Nature
2011
, vol. 
478
 
7370
(pg. 
524
-
528
)
29
Blobel
 
GA
Kalota
 
A
Sanchez
 
PV
Carroll
 
M
Short hairpin RNA screen reveals bromodomain proteins as novel targets in acute myeloid leukemia
Cancer Cell
2011
, vol. 
20
 
3
(pg. 
287
-
288
)
30
Dawson
 
MA
Prinjha
 
RK
Dittmann
 
A
, et al. 
Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia
Nature
2011
, vol. 
478
 
7370
(pg. 
529
-
533
)
31
Dombret
 
H
Preudhomme
 
C
Berthon
 
C
, et al. 
A phase 1study of the BET-bromodomain inhibitor OTX015 in patients with advanced acute leukemia
Blood
2014
, vol. 
124
 
21
pg. 
117
 
32
Tallman
 
MS
Andersen
 
JW
Schiffer
 
CA
, et al. 
All-trans-retinoic acid in acute promyelocytic leukemia
N Engl J Med
1997
, vol. 
337
 
15
(pg. 
1021
-
1028
)

Competing Interests

Conflict-of-interest disclosure: The author has consulted for Agios Pharmaceuticals and Seattle Genetics.

Author notes

Off-label drug use: None disclosed.