Skip to main content
Therapeutic Advances in Hematology logoLink to Therapeutic Advances in Hematology
. 2019 Aug 6;10:2040620719866081. doi: 10.1177/2040620719866081

Insights into novel emerging epigenetic drugs in myeloid malignancies

Namrata S Chandhok 1, Thomas Prebet 2,
PMCID: PMC6685116  PMID: 31431820

Abstract

Epigenetics has been defined as ‘a stably heritable phenotype resulting from changes in a chromosome without alterations in the DNA sequence’ and several epigenetic regulators are recurrently mutated in hematological malignancies. Epigenetic modifications include changes such as DNA methylation, histone modifications and RNA associated gene silencing. Transcriptional regulation, chromosome stability, DNA replication and DNA repair are all controlled by these modifications. Mutations in genes encoding epigenetic modifiers are a frequent occurrence in hematologic malignancies and important in both the initiation and progression of cancer. Epigenetic modifications are also frequently reversible, allowing excellent opportunities for therapeutic intervention. The goal of epigenetic therapies is to reverse epigenetic dysregulation, restore the epigenetic balance, and revert malignant cells to a more normal condition. The role of epigenetic therapies thus far is most established in hematologic malignancies, with several agents already approved by the US Food and Drug Administration. In this review, we discuss pharmacological agents targeting epigenetic regulators.

Keywords: acute myeloid leukemia, BET inhibitors, demethylases, DOT1L inhibitors, epigenetic modifications, histone modification, hypomethylating agents, IDH inhibitors, LSD1 inhibitors, myelodysplastic syndrome, novel agents

Introduction

Epigenetics refers to regulatory mechanisms that lead to heritable changes in gene expression that are not due to a change in the DNA coding sequence. Differentiation in normal hematopoiesis is dependent on tightly regulated, ongoing epigenome remodeling.1,2 Dysregulation of epigenetic regulators such as direct-DNA modifications, histone tail modifications and noncoding RNAs can lead to widespread alterations in the normal patterns of gene expression.3 These alterations of genes regulating the cellular epigenome are known offenders in tumorigenesis and occur frequently in a spectrum of hematologic malignancy diseases. For example, in the landmark The Cancer Genome Atlas (TCGA) study analyzing the genome of 200 adult cases of de novo acute myeloid leukemia (AML) identified mutations in 44% of DNA methylation-related genes and 30% of chromatin-modifying genes.4 Owing to the prevalence of epigenetic dysregulation and inherent plasticity of the epigenome, several classes of cancer therapeutics targeting epigenetic aberrations have emerged.

In mammalian cells, genomic DNA is packaged into condensed complexes of DNA and histone proteins known as nucleosomes that are units of chromatin that protect DNA structure and sequence, as well as regulating gene expression and DNA replication.5 Nucleosomes are repeating units of chromatin that are composed of 147 base pairs of DNA wrapped around eight histone proteins that are very stable protein–DNA complexes, but remain dynamic and are tightly regulated.5 Chromatin can be reversibly altered in many ways (addition or removal of epigenetic marks on DNA or chromatin and RNA silencing) that lead to changes in gene activity and cellular phenotypes, and it reorganizes to regulate transcriptional activity at a particular locus based on intrinsic and extrinsic stimuli.6 Alterations in the epigenetic program of the cell are common in many human cancers and therefore are an attractive therapeutic target.7

In this review, we discuss both established and emerging epigenetic therapies in hematologic malignancies.

DNA methylation: targeting DNA methyltransferases, TET, and IDH mutations

Alteration in DNA methylation status is a characteristic epigenetic change in many cancers including hematological malignancies. DNA methylation is the addition of a methyl group to DNA, usually at the 5’ position of the cytosine ring within CpG (cytosine preceding guanine) dinucleotides that often modifies genes and noncoding genomic regions, affecting gene expression. Tightly regulated DNA methylation plays a key role in embryonic development, cellular differentiation, and genome stability, and disruption of methylation affects the expression of protein coding genes and noncoding RNAs resulting in tumorigenesis.810 Mutations in the DNA methylation regulators DNMT3A, TET1/2, and IDH1/2 are recurrent in hematologic malignancies.

DNA methyltransferases

DNA methylation is catalyzed by a group of enzymes known as DNA methyltransferases (DNMTs).8,11 The main DNMTs include DNMT1, which is responsible for the maintenance of existing methylation patterns by replicating CpG methylation patterns from the mother to daughter strand during DNA replication and antagonizing DNA demethylation, and DNMT3A and DNMT3B that are essential for de novo methyltransferase activity, targeting previously unmethylated CpG dinucleotides.12

Although mutations in all the above methyltransferases have been noted in cancer, recurrent mutations in DNMT3A are most prevalent by far, and have been noted in approximately 22% of patients with AML, 13% of patients with myelodysplastic syndrome (MDS), 9% of patients with myeloproliferative neoplasms (MPN), and 11% of patients with T cell lymphomas.1319 In AML, the DNMT3A mutations usually involve the p.882 codon, and are an early event in clonal hematopoiesis.20,21 Mutations in the p.882 codon of DNMT3A produces a hypomorphic protein that inhibits the remaining wild-type (WT) DNMT3A, thereby markedly reducing cellular DNMT activity, leading to global hypomethylation.20,22 Numerous studies agree that DNMT3A-mutant AML patients have a significantly lower overall survival (OS) compared with those with WT DNMT3A regardless of age, even though there is no statistically significant difference between complete remission (CR) and relapse-free survival (RFS) between the patients with or without the DNMT3A mutations.15,2326 Poor clinical outcomes of patients with DNMT3A-mutations are primarily due to relative anthracycline resistance; patients who received standard-dose daunorubicin-based induction therapy have poor outcomes, however the adverse prognostic impact of DNMT3A mutations is mitigated by daunorubicin dose intensification.2630 Patients with DNMT3A mutations have increased likelihood of CR in patients with MDS or previously untreated AML who receive hypomethylating agents (HMAs).31,32

Mutations in DNMT3A enzymes cannot be directly targeted at present, primarily due to the detrimental role of DNMT3A loss in hematopoiesis. However, the antineoplastic activity of HMAs azacitidine (AZA) and decitabine (DEC) in all patients with AML and MDS (regardless of mutational status) is clear.33 The exact mechanism of action of these HMAs is unclear, but the antileukemic effects are thought to be secondary to degradation of DNMTs that lead to global DNA hypomethylation, gene reactivation, DNA damage, and eventual cell death.34 AZA and DEC are US Food and Drug Administration (FDA) and European Medicines Agency (EMA) approved for the treatment of MDS, chronic myelomonocytic leukemia (CMML), and AML, with slight variations in specific approvals in Europe and the United States.35 For example, DEC has FDA, but not EMA, approval in the setting of MDS. In the setting of AML, DEC has EMA, but not FDA, approval.

Novel formulations of HMAs

Although the role of HMAs is well established in myeloid malignancies, advances in the field have come from new formulations that are designed to improve patient comfort and improve benefits by prolonging drug exposure that are now in clinical trials. These include oral AZA (CC-486), oral DEC with cedazuridine, a novel cytidine deaminase inhibitor (ASTX727) and subcutaneous guadecitabine (GDAC, SGI-110), a next-generation hypomethylating drug that has a longer half-life than its active metabolite DEC.

All these agents (GDAC (SGI-110), oral AZA (CC-486), and ASTX727) are in phase III assessment as single-agent therapy (ClinicalTrials.gov identifiers: NCT03306264, NCT02920008, and NCT01757535) in MDS, CMML and MDS in the treatment naïve setting, and are also being assessed after relapse with traditional HMA therapy. In the case of GDAC, the results of the randomized phase III study of GDAC [ASTRAL-1 (ClinicalTrials.gov identifier: NCT02348489] in treatment- naïve unfit patients did not meet its coprimary endpoints of superior CR rate and OS when compared with HMA or cytarabine.36 However, a recently published phase II study that included intermediate- and high-risk MDS and CMML both in the treatment-naïve of HMA relapsed or refractory setting demonstrated a 40–55% overall response rate (ORR) in all subgroups.37 Of the 53 HMA refractory patients included on this study, there were 2 complete responses to GDAC.37 Although toxicity was considered acceptable, 12% of patients withdrew from the study based on serious adverse events; the overall adverse event profile was similar to traditional HMA therapy.3739 ASTX727 is a fixed-dose combination of DEC and cedazuridine, a novel cytidine deaminase inhibitor. Phase I studies with previously treated or newly diagnosed MDS and CMML were published recently; ASTX727 was confirmed to have a safety profile similar to that of DEC.40 Phase II dose confirmation was presented at the 2017 ASH and EHA meetings, and a multicenter, phase III randomized, study of ASTX727 versus IV DEC for patients with MDS and CMML is currently active (ClinicalTrials.gov identifier: NCT03306264). Finally, the oral formulation of AZA, CC-486, has been evaluated in phase I and has been generally well tolerated even with extended dosing in patients with treatment-naïve and refractory AML and MDS.4143 It has also been evaluated in the post-transplant maintenance in AML and MDS.44 This agent is currently in later phase evaluation in studies such as QUAZAR AML-001 (ClinicalTrials.gov identifier: NCT01757535).

IDH mutations

Isocitrate dehydrogenase-1 and −2 (IDH1/2) mutations occur in ~20% of AML, and up to 12% of patients with MDS. IDH enzymes catalyze the conversion of isocitrate to α-ketoglutarate (aKG) in the citric acid cycle.45 There are three conserved mutational hotspots in the IDH enzymes that alter their function and lead to the production of (R)-2-hydroxyglutarate (2HG), an oncometabolite with numerous downstream effects, including impairment of normal TET catalytic activity, and histone lysine demethylases, both of which are crucial to epigenetic regulation.4648

Mutant IDH inhibitors

Recently 2-IDH inhibitors, enasidenib for mutant IDH2 and ivosidenib for mutant IDH1 were approved by the FDA for relapsed/refractory AML. The mIDH1 inhibitor ivosidenib gained FDA approval for the treatment of patients with relapsed/refractory IDH1 mutant AML based on the phase I, multicenter, open-label, dose-escalation, and dose-expansion study.49 In this study, that was most recently updated at ASCO annual meeting in 2018, 179 patients with IDH1-positive relapsed/refractory AML were treated 500 mg oral ivosidenib that resulted in CR in 24.7% (n = 43; 95% CI 18.5–31.8) and CR with partial hematologic improvement (CRi) rate in 8% (n = 14; 95% CI 4.5–13.1). The median duration of CR + CRi was 8.2 months (range, 5.6–12). The ORR was 41.9% (95% CI 34.6–49.5%) and median time to best response was 2.0 months (range, 0.9–5.6).Of the patients treated with ivosidenib, 21 patients were eventually received hematopoietic stem cell transplantation (HSCT).49 The most serious adverse effects included QT prolongation, and the potentially fatal differentiation syndrome that led to a boxed warning for the drug.49,50 This expansion cohort is ongoing (ClinicalTrials.gov identifier: NCT02074839) and based on the early success with this agent, ivosidenib is being examined in the upfront setting in AML who are candidates for nonintensive treatment [AGILE trial (ClinicalTrials.gov identifier: NCT03173248)], in high-risk MDS (ClinicalTrials.gov identifier: NCT03503409), and in numerous combination studies primarily in myeloid malignancies.

The IDH2 inhibitor enasidanib was the first in class IDH2 mutation-specific inhibitor. Similar to ivosidenib, in a phase I/II study of enasidenib in patients with relapsed/refractory AML with IDH2 mutations, 40.3% of patients responded to therapy, 19.3% achieved CR and the median OS was 9.3 months for all study participants, but 19.7 months for patients who achieved CR.51 Ivosidenib, the recently approved IDH1 mutation-specific inhibitor, demonstrated similar results with a 41.6% ORR, 30.4% CR/CRi rate including a 21.6% CR rate.49 IDHENTIFY (ClinicalTrials.gov identifier: NCT02577406), a phase III randomized, open-label study comparing single-agent enasidenib with conventional care regimens (CCR) in older patients relapsed or refractory IDH2 mutant AML is currently enrolling.52 This agent is also being examined in the upfront AML setting (ClinicalTrials.gov identifier: NCT02632708), MDS as a single agent (ClinicalTrials.gov identifiers: NCT03744390 and NCT03383575), in combination with other drugs such as AZA or venetoclax (ClinicalTrials.gov identifier: NCT02677922), and in the post-transplant setting as a maintenance agent (ClinicalTrials.gov identifier: NCT03728335). Although the above agents were the first in class to receive FDA approval, there are numerous others mutant IDH inhibitors such as FT-2102 (mIDH1), BAY 1436032 (pan mIDH1), and the combined IDH1 and IDH2 inhibitor AG-881 currently in clinical trials. Interestingly, recent publications have determined that patients who relapse almost uniformly develop either resistance mutations that impede IDH inhibitor binding or isoform switch from one IDH enzyme to the other.53,54

Histone modifications

In addition to changes in DNA methylation, modifications of the core of eight histone proteins play an important role in genetic expression and, thus, genomic regulation. There are several histone modifying processes including methylation, acetylation, ubiquitination, phosphorylation, sumoylation, and glycosylation that are involved in cancer pathogenesis.54 Histones themselves have an accessible lysine rich amino-terminal that can be both acetylated and methylated.

Histone methylation and demethylation

Histone methylation status is a dynamic process that is essential to the transcriptional program of a cell. Similar to DNA methylation, histone methylation is a process by which methyl groups are transferred to histones leading to structural modification of chromatin, transcriptional modification, and eventually alteration in gene expression. Histone methyltransferases are a class of catalytic enzymes responsible for the transfer of methyl groups from S-adenosyl methionine onto the lysine (lysine methyltransferase or KMTs) or arginine residues [protein arginine methyltransferases (PRMTs)] leading to transcriptional activation or repression depending on the degree and sites of methylation.55 Aberrations in histone methyltransferases have been noted in several hematological malignancies including myeloid malignancies as well as B and T cell lymphomas.5659 Conversely, histone demethylases remove methyl groups from histones and belong to two families of proteins: the lysine-specific demethylase (LSD) family and the JumonjiC (JMJC) family of histone demethylases.60

Histone methyltransferase (KMT) inhibitors: EZH2 inhibitors

Enhancer of zeste homolog 2 (EZH2) is the catalytic subunit of the Polycomb repressive complex 2 (PRC2) that is responsible for transcriptional repression of target genes by trimethylation of lysine 27 on histone H3 (H3K27me3).61 In hematopoiesis EZH2 is a regulator of adult hematopoietic stem cell (HSC) differentiation, proliferation and apoptosis.62,63 It regulates these pathways by repression of negative cell cycle regulators (such as CDKN2A), repression of differentiation transcription factors (such as BLIMP and IRF4), and repression of pro-apoptotic genes (such as NOX and p21).62,64,65

The fact that changes in expression, gain of function, and loss of function mutations in EZH2 contribute may contribute to the development of hematologic malignancies suggests that EZH2 can act not only as an oncogene, but also as a tumor suppressor, depending on the cell context. Gain of function mutations, or overexpression of EZH2, is frequently seen in patients with Burkitt lymphoma, high-grade follicular lymphomas, diffuse large B cell lymphomas (DLBCL), and multiple myeloma with 4;14 translocations.57,66,67 Loss of function mutations have been reported more frequently in myeloid malignancies such as MDS, atypical chronic myelogenous leukemia (CML) and myelofibrosis, and are generally associated with a poorer prognosis with shorter OS and event-free survival.68,69 Loss of function and deletions EZH2 mutations were also noted in up to a quarter of patients with T cell acute lymphoblastic leukemia (T-ALL).70

Several EZH2 inhibitors have been developed and a few are in early phase clinical trials, primarily for high grade lymphomas. Phase I studies were recently completed on tazemetostat (EPZ-6438), a potent and highly selective EZH2 inhibitor that had previously shown antiproliferative/antitumor activity in in vitro and in B cell non-Hodgkin lymphoma xenograph models bearing EZH2 activating mutations.71,72 The open-label, multicenter, dose-escalation, phase I study that included both solid tumors and relapsed/refractory B cell non-Hodgkin lymphoma that was recently published established the recommended phase II dose of 800 mg twice daily and demonstrated durable objective responses, including complete responses, were observed in 8/21 patients with B cell non-Hodgkin lymphoma.73 Phase II data in epithelioid sarcoma was presented at ASCO 2017, but is immature in the lymphoid malignancies. Studies with this drug were recently paused by the FDA based on a patient with a solid tumor malignancy enrolled in a phase I trial who developed a secondary T cell lymphoma but resumed as of September 2018. Other EZH inhibitors in clinical trial are listed in Table 1. Of note, recent work has demonstrated that dual inactivation of both EZH1 and EZH2, leading to complete inactivation disruption of PRC2 was effective at eliminating aggressive quiescent leukemic stem cells in MLL-AF9 leukemia.74 This has led to the development of novel EZH1/2 inhibitors that has shown promise in preclinical studies.75

Table 1.

Selected trials of novel emerging epitherapies.

Select trials targeting DNA modification
Drug Target ClinicalTrials.gov identifier Malignancy
Guadecitabine (SGI-110) DNMT NCT02907359
NCT02920008
NCT03075826
NCT02131597
NCT03075826
MDS, AML, CMML, MPN
Oral Azacitidine (CC-486) DNMT NCT03723135
NCT03703375
NCT03450343
MDS, AML, T cell lymphoma, DLBCL
Select trials targeting Histone modification
Drug Target NCT Malignancy
Tazemetostat (EPZ-6438) EZH2 NCT03456726
NCT03603951
NCT01897571
NCT03213665
NCT02875548
DLBCL, HL, NHL, histiocytic disorders
CPI-1205 EZH2 NCT02395601 B cell lymphoma
PF-06821497 EZH2 NCT03460977 Follicular lymphoma
MAK683 EZH2 NCT02900651 DLBCL
Pinometostat (EPZ-5676) DOT1L NCT03724084 AML-MLL
GSK2879552 LSD1 NCT02929498 MDS
Tranylcypromine LSD1 NCT02273102 AML
MDS
IMG-7289 LSD1 NCT03136185
NCT02842827
Myelofibrosis
AML
MDS
INCB059872 LSD1 NCT02712905 Hematologic malignancies
4SC-202 HDAC NCT01344707 Hematologic malignancies
Abexinostat (PCI24781) HDAC NCT01149668
NCT03600441
NCT00724984
NCT00473577
B cell lymphomas, any hematologic malignancy
Givinostat HDAC NCT01901432
NCT01761968
MPN
Mocetinostat HDAC NCT00431873 CLL
Resminostat HDAC NCT02953301 T cell lymphoma,
Rocilinostat HDAC NCT02091063
NCT01583283
Lymphoma, multiple myeloma
ABBV-075 BET NCT02391480 AML, NHL, multiple myeloma
CPI-0610 BET NCT02158858 Myelofibrosis
FT-1101 BET NCT02543879 Hematologic malignancies
GSK525762/I-BET762 BET NCT01943851 Hematologic malignancies
INCB054329 BET NCT02431260 Hematologic malignancies
PLX51107 BET NCT02683395 AML, MDS, NHL
RO6870810/TEN-010 BET NCT03068351
NCT03255096
Multiple myeloma, DLBCL

AML, acute myeloid leukemia; BET, bromodomain and extra-terminal domain; CLL, chronic lymphocytic leukemia; CMML, chronic myelomonocytic leukemia; DLBCL, diffuse large B cell lymphoma; EZH2, enhancer of zeste homolog 2; HDAC, histone deacetylase; HL, Hodgkin’s lymphoma; LSD, lysine-specific demethylase; MDS, myelodysplastic syndrome; MLL, mixed-lineage leukemia; MPN, myeloproliferative neoplasm; NHL, non-Hodgkin’s lymphoma.

Histone methyltransferase (KMT) inhibitors: DOT1L inhibitors

Disruptor of telomeric silencing 1-like (DOT1L) is the only known a methyltransferase that triggers histone H3 lysine 79 (H3K79) methylation. H3K79 methylation is the only known histone lysine methylation without at least one known corresponding histone demethylase to date and increasing H3K79 methylation has been noted as a part of aging. DOT1L also has known roles in DNA-repair mechanisms, cell-cycle regulation, and maintenance of genome stability.76 H3K79 methylation is implicated in several processes, including transcription elongation by RNA polymerase II, the DNA damage response, and cell cycle checkpoint activation.7779

Approximately 10% of all leukemias harbor MLL1 translocations, and these translocations are particularly enriched in pediatric leukemia and therapy-related leukemia secondary to etoposide. MML rearranged leukemias have with distinct clinical features and portend a poor prognosis. The majority of MLL translocations result in oncogenic fusion proteins in which the native methyltransferase domain is replaced by sequences that interact with DOT1L directly or indirectly.80,81 MLL-rearranged leukemia is dependent on aberrant H3K79 methylation by DOT1L, which make DOT1L inhibitors an attractive target.82 Pinometostat (EPZ-5676) was the first-in-class, small-molecule inhibitor of the histone methyltransferase DOT1L, is currently in clinical testing. Results from a phase I study with 51 patients with relapsed/refractory MLL leukemia of pinometostat were modest, with only 2/51 patients achieving CR with 9/51 experiencing grade 3 or higher drug-related events.83 Based on these data, it seems unlikely that DOT1L inhibition will be sufficient for management of MLL leukemia as a single agent, but may be incorporated into rational combination therapy.

Histone demethylase inhibitors targeting LSD1

Histone modifications are reversible and epigenetic marks, such as methylation or acetylation, can be removed. Enzymes capable of reversing methylation come in two classes: LSD1/LSD2 or KDM1A/KDM1B; and the larger family of JMJC domain-containing histone demethylases.81,84

LSD1 is a key regulator of self-renewal and differentiation in human embryonic stem cells, is pivotal to maintenance of hematopoietic stem cells and differentiation of granulopoiesis.85,86 It causes transcriptional repression by demethylating mono- or di-methylated H3K4, but can also stimulate transcription upon interactions with the androgen receptor.87 LSD1 can also demethylate many nonhistone substrates such as p53, DNMT1, and STAT3.88,89 LSD1 is overexpressed in a several malignancies including solid tumors (gastric, esophageal, breast, lung, colon, etc.), as well as hematopoietic and lymphoid neoplasms including AML, ALL, MPNs, CMML, and MDS.90 LSD1 inactivation inhibited cancer cell differentiation, proliferation, invasion and migration, and tumor growth in animal models and demonstrated the therapeutic potential of LSD1 inhibitors.9193

Numerous LSD inhibitors [trans-2-phenylcyclopropylamine derivatives, monoamine oxidase (MAO) inactivators, peptide-based, polyamine-based, and others] have been developed and fall into two categories: reversible or irreversible inhibition.91,94 Some of these inhibitors are currently in early phase clinical trials and there is no mature trial data. Myelosuppression from LSD1 inhibitors at therapeutic doses may prohibit their use in myeloid malignancies, owing to the role in normal hematopoiesis and terminal myeloid differentiation, but this remains to be seen. JMJC domain-containing histone demethylases, a much larger class of histone demethylation agents, are still being explored in the lab and although inhibitors are being developed, they have not entered the clinical realm.

Histone acetylation and deacetylation

Similar to methylation, histone acetylation status is an important mechanism that regulate of chromatin structure, transcription, and DNA repair. Histone lysine acetyltransferases (HATs) are responsible for acetylation of histones that relax chromatin structures, exposed promoter regions, and increased transcription. Mutations in HATS that affect their catalytic activity are frequent in lymphoid malignancies, occurring in up to 40% of DLBL, 60% of follicular lymphoma (FL), and less frequently in B cell ALL, T cell ALL, and cutaneous T cell lymphoma (CTCL).9599 HAT mutations are also notable in some myeloid malignancies, specifically with CBP mutations or translocations, however effectively targeting HAT activity remains in the laboratory for now.4,100,101

Histone deacetylases (HDACs) oppose this action by catalyzing deacetylation that leads to chromatin condensation and resultant gene silencing.102104 HDACs are a very diverse group of enzymes, that are subdivided into four classes (I, II, III, and IV) based on homology to yeast proteins, subcellular location, and enzymatic activities. They lead to tumorigenesis by repression of tumor suppressor gene expression or modification of oncogenic cell-signaling pathways.105107 In line with variety of HDAC proteins, HDAC inhibitors are a structurally varied class of medications that vary in their potency and specificity to different classes of HDACs including hydroxamates, cyclic peptides, aliphatic acids, benzamides, and electrophilic ketones. HDAC inhibitors induce cancer cell cycle arrest, differentiation, and cell death. They also reduce angiogenesis and modulate immune response.108 Although HATs have not yet proven themselves as druggable targets, the role of HDAC inhibition has been established in several hematologic malignancies. FDA-approved HDAC inhibitors (HDACi) include vorinostat for the management of CTCL, romidepsin for CTCL and peripheral T cell lymphoma (PTCL), belinostat for PTCL, and panobinostat is FDA approved for the management of multiple myeloma.109112

There are a plethora of other HDACis currently in early phase clinical trials, including the established agents mentioned above, as well as a whole host of newer agents (Abexinostat (PCI24781), AR42, Givinostat, Mocetinostat, Quisinostat (JNJ-26481585), Resminostat (4SC201), Rocilinostat (ACY1215), Tacedinaline (CI994), as well as others) in phase I and II clinical trials. Some ongoing trials of these novel agents are listed in Table 1. It is useful to note that although there are ongoing monotherapy trials in select hematologic malignancies, most trials at present are focused on combination therapy both for drug synergy and to overcome resistance to HDACi monotherapy.113

Epigenetic ‘readers’: bromodomain proteins

The previously described targets included epigenetic ‘writers’ or ‘erasers’ in that they were involved in the addition or removal of chemical groups onto either histone tails or the DNA. The bromodomain and extra-terminal domain (BET) family of adaptor proteins are epigenetic ‘readers’, or chromatin regulators that possess specialized domains that survey the epigenetic landscape and dock at specific regions within the genome.114 These proteins then serve as scaffolds for transcription factors and chromatin-modifying enzymes to assemble functional complexes onto specific loci and facilitate DNA-templated processes.

The BET family of epigenetic readers have two tandem bromodomains, an extra-terminal domain and a C-terminal domain.115 Bromodomains (BRDs) have acetyl–lysine binding pockets and bind to acetylated lysines on histone tails and recruit other chromatin factors and transcriptional machinery to regulate gene transcription.116 The BET family has four members, three of which (BRD2, BRD3, and BRD4) are expressed ubiquitously, and germ cell specific BRDT.114 In addition to transcriptional regulation where it is important for both initiating and continuing transcription, BET proteins also have an essential role in cell-cycle regulation.117

BET protein disruption is associated with cancer and the study of BET proteins, and BET inhibitors, has been an area of robust research over the past decade. Early studies employing BET inhibitors revealed the importance of BET in regulation of MYC an oncogenic driver.118 BET inhibition in the early studies led to downregulation of c-Myc, cell-cycle arrest and cellular senescence in myeloma models, and prolonged survival in Burkitt’s lymphoma and AML murine xenograft models.118,119 In lymphoma, BET proteins were found to preferentially bind in the proximity of critical lymphoma-related oncogenes.120 Early lymphoma cell line studies showed that although BET inhibitors were cytostatic in most cases, but induced apoptosis in a subgroup of cell lines.121 Preclinical efficacy of BET inhibitors has been demonstrated in AML while several other papers have documented similar efficacy in several hematologic malignancies.122126

Early phase clinical trials of BET inhibitor monotherapy in hematologic malignancies have shown that BET inhibitors are tolerated at therapeutic doses and there is some signal of limited efficacy.127129 In the acute leukemia cohort of 41 patients five patients demonstrated some degree of response to the BET inhibitor OTX015.128 Limited antitumor activity was also noted in patients with relapsed or refractory lymphomas in a phase I study of the BET inhibitor CPI-0610.130 Like with HDACis, thoughtful combinations utilizing BET inhibitors are more likely to derive clinical benefit than BET inhibitor monotherapy. Combinations with agents such as cell-cycle inhibitors, DNA damage repair inhibitors, apoptosis inhibitors, checkpoint inhibitors, or other epitherapies such as HDACi have demonstrated preclinical efficacy.117 Combinations may induce cytotoxicity and help to overcome resistance to a single targeted agent. Numerous trials currently ongoing, some of which are listed in Table 1.

Micro-RNA

MicroRNAs (miRNAs) are evolutionarily conserved 21–23-nucleotide single-stranded noncoding RNAs that typically destabilize messenger RNA and are crucial to regulating gene expression.131 Alterations in miRNA mediate processes in tumorigenesis such as inflammation, cell-cycle regulation, stress response, differentiation, apoptosis, and invasion, and are implicated in the generation or maintenance of cancer.132 Significant differences have been noted in the miRNA expression between normal and cancer tissues, where miRNAs acting as tumor suppressors or oncogenes.133 These changes in miRNA expressions may be due to mutations, translocations of other epigenetic changes, and are being assessed as prognostic markers in most hematologic malignancies.134

There are two classes of miRNA-targeted therapeutics have also entered early phase clinical trials (e.g. ClinicalTrials.gov identifiers: NCT03713320 and NCT02580552 for mycosis fungoides, chronic lymphocytic leukemia, DLBCL, or ATLL). MicroRNA mimics aim to restore lost miRNA expression (tumor suppressor miRNA) and miRNA inhibitors that bind to their target and inhibit the target oncogenic miRNA function.135 There have been early successes in preclinical models with several miRNA therapies, however these agents are yet to make a splash in cancer therapy and are in the early staged of clinical evaluation. We are likely to see these epitherapies in the future, potentially in combination with chemotherapy, as several miRNAs have been shown to be sensitive cancer cells to chemotherapy.136,137 Improved understanding of the miRNA targetome will also help refine the development of these agents.

Conclusion

Although there has been a relative boom in the emergence of novel epigenetic therapies, optimal implementation of these agents will require significant efforts. Targeting the cancer epigenome remains challenging because of several overlapping dependent and independent phenomena such as DNA modifications, covalent post-translational modifications of histones, histone variants, noncovalent remodeling of chromatin, and microRNA expression. Improved understanding of the epigenetic mechanisms in normal cells, and defective epigenetic mechanisms in neoplastic cells, will help in designing optimal combinations while minimizing drug-related toxicities in the future. Capitalizing on interdependent mechanisms in cancer, such as cancer epigenetics and cancer immunology or metabolism are also likely to lead to more significant gains.

Footnotes

Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.

Conflict of interest statement: The authors declare that there is no conflict of interest.

Ethical approval: Ethics approval was not required for this review.

Informed consent: Informed consent was not required for this review.

Contributor Information

Namrata S. Chandhok, Division of Hematology/Oncology, Smilow Cancer Center at Yale New Haven Hospital, New Haven, CT, USA

Thomas Prebet, Division of Hematology/Oncology, Smilow Cancer Center at Yale New Haven Hospital, 35 Park Street, New Haven, CT 06511, USA.

References

  • 1. Farlik M, Halbritter F, Müller F, et al. DNA methylation dynamics of human hematopoietic stem cell differentiation. Cell Stem Cell 2016; 19: 808–822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Cedar H, Bergman Y. Epigenetics of haematopoietic cell development. Nat Rev Immunol 2011; 11: 478. [DOI] [PubMed] [Google Scholar]
  • 3. Yoo CB, Jones PA. Epigenetic therapy of cancer: past, present and future. Nat Rev Drug Discov 2006; 5: 37. [DOI] [PubMed] [Google Scholar]
  • 4. The Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med 2013; 368: 2059–2074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Li B, Carey M, Workman JL. The role of chromatin during transcription. Cell 2007; 128: 707–719. [DOI] [PubMed] [Google Scholar]
  • 6. Flavahan WA, Gaskell E, Bernstein BE. Epigenetic plasticity and the hallmarks of cancer. Science 2017; 357: eaal2380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Pfister SX, Ashworth A. Marked for death: targeting epigenetic changes in cancer. Nat Rev Drug Discov 2017; 16: 241. [DOI] [PubMed] [Google Scholar]
  • 8. Reik W. Stability and flexibility of epigenetic gene regulation in mammalian development. Nature 2007; 447: 425. [DOI] [PubMed] [Google Scholar]
  • 9. Esteller M. Relevance of DNA methylation in the management of cancer. Lancet Oncol 2003; 4: 351–358. [DOI] [PubMed] [Google Scholar]
  • 10. Baylin SB, Jones PA. A decade of exploring the cancer epigenome-biological and translational implications. Nat Rev Cancer 2011; 11: 726–734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Bird A. DNA methylation patterns and epigenetic memory. Genes Dev 2002; 16: 6–21. [DOI] [PubMed] [Google Scholar]
  • 12. Rodríguez-Paredes M, Esteller M. Cancer epigenetics reaches mainstream oncology. Nat Med 2011; 17: 330. [DOI] [PubMed] [Google Scholar]
  • 13. Ley TJ, Miller C, Ding L, et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med 2013; 368: 2059–2074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Huether R, Dong L, Chen X, et al. The landscape of somatic mutations in epigenetic regulators across 1,000 paediatric cancer genomes. Nat Commun 2014; 5: 3630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Ley TJ, Ding L, Walter MJ, et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med 2010; 363: 2424–2433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Haferlach T, Nagata Y, Grossmann V, et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia 2014; 28: 241–247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Roller A, Grossmann V, Bacher U, et al. Landmark analysis of DNMT3A mutations in hematological malignancies. Leukemia 2013; 27: 1573–1578. [DOI] [PubMed] [Google Scholar]
  • 18. Couronne L, Bastard C, Bernard OA. TET2 and DNMT3A mutations in human T-cell lymphoma. N Engl J Med 2012; 366: 95–96. [DOI] [PubMed] [Google Scholar]
  • 19. Yang L, Rau R, Goodell MA. DNMT3A in haematological malignancies. Nat Rev Cancer 2015; 15: 152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Russler-Germain DA, Spencer DH, Young MA, et al. The R882H DNMT3A mutation associated with AML dominantly inhibits wild-type DNMT3A by blocking its ability to form active tetramers. Cancer Cell 2014; 25: 442–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Buscarlet M, Provost S, Feroz Zada Y, et al. DNMT3A and TET2 dominate clonal hematopoiesis, demonstrate benign phenotypes and different genetic predisposition. Blood 2017; 130: 753–762. [DOI] [PubMed] [Google Scholar]
  • 22. Kim SJ, Zhao H, Hardikar S, et al. A DNMT3A mutation common in AML exhibits dominant-negative effects in murine ES cells. Blood 2013; 122: 4086–4089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Thol F, Damm F, Ludeking A, et al. Incidence and prognostic influence of DNMT3A mutations in acute myeloid leukemia. J Clin Oncol 2011; 29: 2889–2896. [DOI] [PubMed] [Google Scholar]
  • 24. Marcucci G, Metzeler KH, Schwind S, et al. Age-related prognostic impact of different types of DNMT3A mutations in adults with primary cytogenetically normal acute myeloid leukemia. J Clin Oncol 2012; 30: 742–750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Shen Y, Zhu YM, Fan X, et al. Gene mutation patterns and their prognostic impact in a cohort of 1185 patients with acute myeloid leukemia. Blood 2011; 118: 5593–5603. [DOI] [PubMed] [Google Scholar]
  • 26. Ribeiro AF, Pratcorona M, Erpelinck-Verschueren C, et al. Mutant DNMT3A: a marker of poor prognosis in acute myeloid leukemia. Blood 2012; 119: 5824–5831. [DOI] [PubMed] [Google Scholar]
  • 27. Gaidzik VI, Schlenk RF, Paschka P, et al. Clinical impact of DNMT3A mutations in younger adult patients with acute myeloid leukemia: results of the AML Study Group (AMLSG). Blood 2013; 121: 4769–4777. [DOI] [PubMed] [Google Scholar]
  • 28. Ostronoff F, Othus M, Ho PA, et al. Mutations in the DNMT3A exon 23 independently predict poor outcome in older patients with acute myeloid leukemia: a SWOG report. Leukemia 2013; 27: 238–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Tie R, Zhang T, Fu H, et al. Association between DNMT3A mutations and prognosis of adults with de novo acute myeloid leukemia: a systematic review and meta-analysis. PLoS One 2014; 9: e93353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Guryanova OA, Shank K, Spitzer B, et al. DNMT3A mutations promote anthracycline resistance in acute myeloid leukemia via impaired nucleosome remodeling. Nat Med 2016; 22: 1488–1495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Metzeler KH, Walker A, Geyer S, et al. DNMT3A mutations and response to the hypomethylating agent decitabine in acute myeloid leukemia. Leukemia 2011; 26: 1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Traina F, Visconte V, Elson P, et al. Impact of molecular mutations on treatment response to DNMT inhibitors in myelodysplasia and related neoplasms. Leukemia 2013; 28: 78. [DOI] [PubMed] [Google Scholar]
  • 33. Da Costa EM, McInnes G, Beaudry A, et al. DNA methylation-targeted drugs. Cancer J 2017; 23: 270–276. [DOI] [PubMed] [Google Scholar]
  • 34. Giorgio C, Dell’Aversana C, Altucci L. Chapter 27 -Epigenetics in acute myeloid leukemia. In:Tollefsbol TO. (ed.) Medical epigenetics. Boston: Academic Press, 2016, pp.483–498. [Google Scholar]
  • 35. Derissen EJB, Beijnen JH, Schellens JHM. Concise drug review: azacitidine and decitabine. Oncologist 2013; 18: 619–624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Hansen R. Astex Pharmaceuticals and Otsuka announce results of the phase 3 ASTRAL-1 study of guadecitabine (SGI-110) in treatment-naïve AML patients ineligible to receive intense induction chemotherapy. CA: Astex Pharmaceuticals, Inc, 2018. [Google Scholar]
  • 37. Garcia-Manero G, Roboz G, Walsh K, et al. Guadecitabine (SGI-110) in patients with intermediate or high-risk myelodysplastic syndromes: phase 2 results from a multicentre, open-label, randomised, phase 1/2 trial. Lancet Haematol 2019; 6: e317–e327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Guillermo Garcia-Manero M, Ritchie EK, Walsh KJ, et al. Long term results of a randomized phase 2 dose-response study of guadecitabine, a novel subcutaneous (SC) hypomethylating agent (HMA), in 102 patients with intermediate or high risk myelodysplastic syndromes (MDS) or chronic myelomonocytic leukemia (CMML). Blood 2018; 132: 231. [Google Scholar]
  • 39. Garcia-Manero G, Sasaki K, Montalban-Bravo G, et al. Final report of a phase II study of guadecitabine (SGI-110) in patients (pts) with previously untreated myelodysplastic syndrome (MDS). Blood 2018; 132: 232.29776907 [Google Scholar]
  • 40. Savona MR, Odenike O, Amrein PC, et al. An oral fixed-dose combination of decitabine and cedazuridine in myelodysplastic syndromes: a multicentre, open-label, dose-escalation, phase 1 study. Lancet Haematol 2019; 6: e194–e203. [DOI] [PubMed] [Google Scholar]
  • 41. Savona MR, Gore SD, Kolibaba KS, et al. CC-486 (Oral Azacitidine) monotherapy in patients with acute myeloid leukemia (AML). Blood 2015; 126: 452. [Google Scholar]
  • 42. Garcia-Manero G, Gore SD, Cogle C, et al. Phase I study of oral azacitidine in myelodysplastic syndromes, chronic myelomonocytic leukemia, and acute myeloid leukemia. J Clin Oncol 2011; 29: 2521–2527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Savona MR, Kolibaba K, Conkling P, et al. Extended dosing with CC-486 (oral azacitidine) in patients with myeloid malignancies. Am J Hematol 2018; 93: 1199–1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. de Lima M, Oran B, Champlin RE, et al. CC-486 maintenance after stem cell transplantation in patients with acute myeloid leukemia or myelodysplastic syndromes. Biol Blood Marrow Transplant 2018; 24: 2017–2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. DiNardo CD, Ravandi F, Agresta S, et al. Characteristics, clinical outcome, and prognostic significance of IDH mutations in AML. Am J Hematol 2015; 90: 732–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Clark O, Yen K, Mellinghoff IK. Molecular pathways: isocitrate dehydrogenase mutations in cancer. Clin Cancer Res 2016; 22: 1837–1842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Xu W, Yang H, Liu Y, et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases. Cancer Cell 2011; 19: 17–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Chowdhury R, Yeoh KK, Tian YM, et al. The oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases. EMBO Rep 2011; 12: 463–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. DiNardo CD, Stein EM, de Botton S, et al. Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. N Engl J Med 2018; 378: 2386–2398. [DOI] [PubMed] [Google Scholar]
  • 50. Micheau O, Lens S, Gaide O, et al. NF-kappaB signals induce the expression of c-FLIP. Mol Cell Biol 2001; 21: 5299–5305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant-IDH2 relapsed or refractory acute myeloid leukemia. Blood 2017; 30: 722–731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Stein EM, Fathi AT, DiNardo CD, et al. Enasidenib (AG-221), a potent oral inhibitor of mutant isocitrate dehydrogenase 2 (IDH2) enzyme, induces hematologic responses in patients with myelodysplastic syndromes (MDS). Blood 2016; 128: 343. [Google Scholar]
  • 53. Harding JJ, Lowery MA, Shih AH, et al. Isoform switching as a mechanism of acquired resistance to mutant isocitrate dehydrogenase inhibition. Cancer Discov 2018: 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Intlekofer AM, Shih AH, Wang B, et al. Acquired resistance to IDH inhibition through trans or cis dimer-interface mutations. Nature 2018; 559: 125–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Rice JC, Briggs SD, Ueberheide B, et al. Histone methyltransferases direct different degrees of methylation to define distinct chromatin domains. Mol Cell 2003; 12: 1591–1598. [DOI] [PubMed] [Google Scholar]
  • 56. Ntziachristos P, Tsirigos A, Vlierberghe PV, et al. Genetic inactivation of the polycomb repressive complex 2 in T cell acute lymphoblastic leukemia. Nat Med 2012; 18: 298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Morin RD, Johnson NA, Severson TM, et al. Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B-cell lymphomas of germinal-center origin. Nat Genet 2010; 42: 181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Zhang J, Ding L, Holmfeldt L, et al. The genetic basis of early T-cell precursor acute lymphoblastic leukaemia. Nature 2012; 481: 157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Nikoloski G, Langemeijer SMC, Kuiper RP, et al. Somatic mutations of the histone methyltransferase gene EZH2 in myelodysplastic syndromes. Nat Genet 2010; 42: 665. [DOI] [PubMed] [Google Scholar]
  • 60. Morera L, Lübbert M, Jung M. Targeting histone methyltransferases and demethylases in clinical trials for cancer therapy. Clin Epigenetics 2016; 8: 57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Cao R, Wang L, Wang H, et al. Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science 2002; 298: 1039–1043. [DOI] [PubMed] [Google Scholar]
  • 62. Xie H, Xu J, Hsu JH, et al. Polycomb repressive complex 2 regulates normal hematopoietic stem cell function in a developmental-stage-specific manner. Cell Stem Cell 2014; 14: 68–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Mochizuki-Kashio M, Mishima Y, Miyagi S, et al. Dependency on the polycomb gene EZH2 distinguishes fetal from adult hematopoietic stem cells. Blood 2011; 118: 6553–6561. [DOI] [PubMed] [Google Scholar]
  • 64. Good-Jacobson KL. Regulation of germinal center, B-cell memory, and plasma cell formation by histone modifiers. Front Immunol 2014; 5: 596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Herviou L, Cavalli G, Cartron G, et al. EZH2 in normal hematopoiesis and hematological malignancies. Oncotarget 2015; 7: 2284–2296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Abd Al Kader L, Oka T, Takata K, et al. In aggressive variants of non-Hodgkin lymphomas, Ezh2 is strongly expressed and polycomb repressive complex PRC1.4 dominates over PRC1.2. Virchows Arch 2013; 463: 697–711. [DOI] [PubMed] [Google Scholar]
  • 67. Asangani IA, Ateeq B, Cao Q, et al. Characterization of the EZH2-MMSET histone methyltransferase regulatory axis in cancer. Mol Cell 2013; 49: 80–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Ernst T, Chase AJ, Score J, et al. Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nat Genet 2010; 42: 722–726. [DOI] [PubMed] [Google Scholar]
  • 69. Nikoloski G, Langemeijer SM, Kuiper RP, et al. Somatic mutations of the histone methyltransferase gene EZH2 in myelodysplastic syndromes. Nat Genet 2010; 42: 665–667. [DOI] [PubMed] [Google Scholar]
  • 70. Ntziachristos P, Tsirigos A, Van Vlierberghe P, et al. Genetic inactivation of the polycomb repressive complex 2 in T cell acute lymphoblastic leukemia. Nat Med 2012; 18: 298–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Knutson SK, Kawano S, Minoshima Y, et al. Selective inhibition of EZH2 by EPZ-6438 leads to potent antitumor activity in EZH2-mutant non-hodgkin lymphoma. Mol Cancer Ther 2014; 13: 842–854. [DOI] [PubMed] [Google Scholar]
  • 72. Brach D, Johnston-Blackwell D, Drew A, et al. EZH2 inhibition by tazemetostat results in altered dependency on B-cell activation signaling in DLBCL. Mol Cancer Ther 2017; 16: 2586–2597. [DOI] [PubMed] [Google Scholar]
  • 73. Italiano A, Soria JC, Toulmonde M, et al. Tazemetostat, an EZH2 inhibitor, in relapsed or refractory B-cell non-Hodgkin lymphoma and advanced solid tumours: a first-in-human, open-label, phase 1 study. Lancet Oncol 2018; 19: 649–659. [DOI] [PubMed] [Google Scholar]
  • 74. Neff T, Sinha AU, Kluk MJ, et al. Polycomb repressive complex 2 is required for MLL-AF9 leukemia. Proc Natl Acad Sci U S A 2012; 109: 5028–5033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Fujita S, Honma D, Adachi N, et al. Dual inhibition of EZH1/2 breaks the quiescence of leukemia stem cells in acute myeloid leukemia. Leukemia 2017; 32: 855. [DOI] [PubMed] [Google Scholar]
  • 76. Wood K, Tellier M, Murphy S. DOT1L and H3K79 methylation in transcription and genomic stability. Biomolecules 2018; 8, https://www.ncbi.nlm.nih.gov/pubmed/29495487 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Wood K, Tellier M, Murphy S. DOT1L and H3K79 Methylation in Transcription and Genomic Stability. Biomolecules 2018; 8: 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Kim W, Choi M, Kim JE. The histone methyltransferase Dot1/DOT1L as a critical regulator of the cell cycle. Cell Cycle 2014; 13: 726–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. McLean CM, Karemaker ID, van Leeuwen F. The emerging roles of DOT1L in leukemia and normal development. Leukemia 2014; 28: 2131. [DOI] [PubMed] [Google Scholar]
  • 80. Daigle SR, Olhava EJ, Therkelsen CA, et al. Potent inhibition of DOT1L as treatment of MLL-fusion leukemia. Blood 2013; 122: 1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Muntean AG, Hess JL. The pathogenesis of mixed-lineage leukemia. Annu Rev Pathol 2012; 7: 283–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Bernt Kathrin M, Zhu N, Sinha Amit U, et al. MLL-rearranged leukemia is dependent on aberrant H3K79 methylation by DOT1L. Cancer Cell 2011; 20: 66–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Stein EM, Garcia-Manero G, Rizzieri DA, et al. The DOT1L inhibitor pinometostat reduces H3K79 methylation and has modest clinical activity in adult acute leukemia. Blood 2018; 131: 2661–2669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Tsukada Y, Fang J, Erdjument-Bromage H, et al. Histone demethylation by a family of JmjC domain-containing proteins. Nature 2005; 439: 811. [DOI] [PubMed] [Google Scholar]
  • 85. Adamo A, Sese B, Boue S, et al. LSD1 regulates the balance between self-renewal and differentiation in human embryonic stem cells. Nat Cell Biol 2011; 13: 652–659. [DOI] [PubMed] [Google Scholar]
  • 86. Kerenyi MA, Shao Z, Hsu YJ, et al. Histone demethylase Lsd1 represses hematopoietic stem and progenitor cell signatures during blood cell maturation. Elife 2013; 2: e00633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Metzger E, Wissmann M, Yin N, et al. LSD1 demethylates repressive histone marks to promote androgen-receptor-dependent transcription. Nature 2005; 437: 436. [DOI] [PubMed] [Google Scholar]
  • 88. Hamamoto R, Saloura V, Nakamura Y. Critical roles of non-histone protein lysine methylation in human tumorigenesis. Nat Rev Cancer 2015; 15: 110–124. [DOI] [PubMed] [Google Scholar]
  • 89. Jin L, Hanigan CL, Wu Y, et al. Loss of LSD1 (lysine-specific demethylase 1) suppresses growth and alters gene expression of human colon cancer cells in a p53- and DNMT1(DNA methyltransferase 1)-independent manner. Biochem J 2013; 449: 459–468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Niebel D, Kirfel J, Janzen V, et al. Lysine-specific demethylase 1 (LSD1) in hematopoietic and lymphoid neoplasms. Blood 2014; 124: 151–152. [DOI] [PubMed] [Google Scholar]
  • 91. Zheng YC, Ma J, Wang Z, et al. A systematic review of histone lysine-specific demethylase 1 and its inhibitors. Med Res Rev 2015; 35: 1032–1071. [DOI] [PubMed] [Google Scholar]
  • 92. Zheng YC, Yu B, Chen ZS, et al. TCPs: privileged scaffolds for identifying potent LSD1 inhibitors for cancer therapy. Epigenomics 2016; 8: 651–666. [DOI] [PubMed] [Google Scholar]
  • 93. McAllister TE, England KS, Hopkinson RJ, et al. Recent progress in histone demethylase inhibitors. J Med Chem 2016; 59: 1308–1329. [DOI] [PubMed] [Google Scholar]
  • 94. Mould DP, McGonagle AE, Wiseman DH, et al. Reversible inhibitors of LSD1 as therapeutic agents in acute myeloid leukemia: clinical significance and progress to date. Med Res Rev 2015; 35: 586–618. [DOI] [PubMed] [Google Scholar]
  • 95. Pasqualucci L, Dominguez-Sola D, Chiarenza A, et al. Inactivating mutations of acetyltransferase genes in B-cell lymphoma. Nature 2011; 471: 189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Vicente C, Schwab C, Broux M, et al. Targeted sequencing identifies association between IL7R-JAK mutations and epigenetic modulators in T-cell acute lymphoblastic leukemia. Haematologica 2015; 100: 1301–1310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Okosun J, Bödör C, Wang J, et al. Integrated genomic analysis identifies recurrent mutations and evolution patterns driving the initiation and progression of follicular lymphoma. Nat Genet 2013; 46: 176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Mullighan CG, Zhang J, Kasper LH, et al. CREBBP mutations in relapsed acute lymphoblastic leukaemia. Nature 2011; 471: 235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. da Silva Almeida AC, Abate F, Khiabanian H, et al. The mutational landscape of cutaneous T cell lymphoma and Sézary syndrome. Nat Genet 2015; 47: 1465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Sobulo OM, Borrow J, Tomek R, et al. MLL is fused to CBP, a histone acetyltransferase, in therapy-related acute myeloid leukemia with a t(11;16)(q23;p13.3). Proc Natl Acad Sci U S A 1997; 94: 8732–8737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Borrow J, Stanton VP, Jr, Andresen JM, et al. The translocation t(8;16)(p11;p13) of acute myeloid leukaemia fuses a putative acetyltransferase to the CREB–binding protein. Nat Genet 1996; 14: 33. [DOI] [PubMed] [Google Scholar]
  • 102. Grunstein M. Histone acetylation in chromatin structure and transcription. Nature 1997; 389: 349. [DOI] [PubMed] [Google Scholar]
  • 103. Gallinari P, Di Marco S, Jones P, et al. HDACs, histone deacetylation and gene transcription: from molecular biology to cancer therapeutics. Cell Res 2007; 17: 195–211. [DOI] [PubMed] [Google Scholar]
  • 104. Kuo MH, Allis CD. Roles of histone acetyltransferases and deacetylases in gene regulation. Bioessays 1998; 20: 615–626. [DOI] [PubMed] [Google Scholar]
  • 105. Juan LJ, Shia WJ, Chen MH, et al. Histone deacetylases specifically down-regulate p53-dependent gene activation. J Biol Chem 2000; 275: 20436–20443. [DOI] [PubMed] [Google Scholar]
  • 106. Li Y, Seto E. HDACs and HDAC inhibitors in cancer development and therapy. Cold Spring Harb Perspect Med 2016; 6: a026831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Seto E, Yoshida M. Erasers of histone acetylation: the histone deacetylase enzymes. Cold Spring Harb Perspect Biol 2014; 6: a018713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Eckschlager T, Plch J, Stiborova M, et al. Histone deacetylase inhibitors as anticancer drugs. Int J Mol Sci 2017; 18: 1414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Mann BS, Johnson JR, Cohen MH, et al. FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist 2007; 12: 1247–1252. [DOI] [PubMed] [Google Scholar]
  • 110. Grant C, Rahman F, Piekarz R, et al. Romidepsin: a new therapy for cutaneous T-cell lymphoma and a potential therapy for solid tumors. Expert Rev Anticancer Ther 2010; 10: 997–1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Sawas A, Radeski D, O’Connor OA. Belinostat in patients with refractory or relapsed peripheral T-cell lymphoma: a perspective review. Ther Adv Hematol 2015; 6: 202–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. San-Miguel JF, Hungria VT, Yoon SS, et al. Panobinostat plus bortezomib and dexamethasone versus placebo plus bortezomib and dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma: a multicentre, randomised, double-blind phase 3 trial. Lancet Oncol 2014; 15: 1195–1206. [DOI] [PubMed] [Google Scholar]
  • 113. Suraweera A, O’Byrne KJ, Richard DJ. Combination therapy with histone deacetylase inhibitors (hdaci) for the treatment of cancer: achieving the full therapeutic potential of HDACi. Front Oncol 2018; 8: 92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Dawson MA, Kouzarides T, Huntly BJP. Targeting epigenetic readers in cancer. N Engl J Med 2012; 367: 647–657. [DOI] [PubMed] [Google Scholar]
  • 115. Stathis A, Bertoni F. BET proteins as targets for anticancer treatment. Cancer Discov 2018; 8: 24–36. [DOI] [PubMed] [Google Scholar]
  • 116. Pérez-Salvia M, Esteller M. Bromodomain inhibitors and cancer therapy: from structures to applications. Epigenetics 2016; 12: 323–339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Doroshow DB, Eder JP, LoRusso PM. BET inhibitors: a novel epigenetic approach. Ann Oncol 2017; 28: 1776–1787. [DOI] [PubMed] [Google Scholar]
  • 118. Delmore Jake E, Issa Ghayas C, Lemieux Madeleine E, et al. BET Bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 2011; 146: 904–917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Mertz JA, Conery AR, Bryant BM, et al. Targeting MYC dependence in cancer by inhibiting BET bromodomains. Proc Natl Acad Sci U S A 2011; 108: 16669–16674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Chapuy B, McKeown MR, Lin CY, et al. Discovery and characterization of super-enhancer-associated dependencies in diffuse large B cell lymphoma. Cancer Cell 2013; 24: 777–790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Boi M, Gaudio E, Bonetti P, et al. The BET Bromodomain inhibitor OTX015 affects pathogenetic pathways in pre-clinical B-cell tumor models and synergizes with targeted drugs. Clin Cancer Res 2015; 21, https://www.ncbi.nlm.nih.gov/pubmed/25623213 [DOI] [PubMed] [Google Scholar]
  • 122. Dawson MA, Gudgin EJ, Horton SJ, et al. Recurrent mutations, including NPM1c, activate a BRD4-dependent core transcriptional program in acute myeloid leukemia. Leukemia 2014; 28: 311–320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Dawson MA, Prinjha RK, Dittmann A, et al. Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 2011; 478: 529–533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Zuber J, Shi J, Wang E, et al. RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature 2011; 478: 524–528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Delmore JE, Issa GC, Lemieux ME, et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 2011; 146: 904–917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Ott CJ, Kopp N, Bird L, et al. BET bromodomain inhibition targets both c-MYC and IL7R in high-risk acute lymphoblastic leukemia. Blood 2012; 120: 2843–2852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Dickinson M, Kamdar M, Huntly BJP, et al. A phase I study of molibresib (GSK525762), a selective bromodomain (BRD) and extra terminal protein (BET) inhibitor: results from part 1 of a phase I/II open label single agent study in subjects with non-hodgkin’s lymphoma (NHL). Blood 2018; 132: 1682. [Google Scholar]
  • 128. Amorim S, Stathis A, Gleeson M, et al. Bromodomain inhibitor OTX015 in patients with lymphoma or multiple myeloma: a dose-escalation, open-label, pharmacokinetic, phase 1 study. Lancet Haematol 2016; 3: e196–e204. [DOI] [PubMed] [Google Scholar]
  • 129. Dawson M, Stein EM, Huntly BJP, et al. A phase I study of GSK525762, a selective bromodomain (BRD) and extra terminal protein (BET) inhibitor: results from part 1 of phase I/II open label single agent study in patients with acute myeloid leukemia (AML). Blood 2017; 130: 1377.28667012 [Google Scholar]
  • 130. Abramson JS, Blum KA, Flinn IW, et al. BET inhibitor CPI-0610 is well tolerated and induces responses in diffuse large B-cell lymphoma and follicular lymphoma: preliminary analysis of an ongoing phase 1 study. Blood 2015; 126: 1491. [Google Scholar]
  • 131. Li Z, Rana TM. Therapeutic targeting of microRNAs: current status and future challenges. Nat Rev Drug Discov 2014; 13: 622. [DOI] [PubMed] [Google Scholar]
  • 132. Farazi TA, Spitzer JI, Morozov P, et al. miRNAs in human cancer. J Pathol 2011; 223: 102–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Lu J, Getz G, Miska EA, et al. MicroRNA expression profiles classify human cancers. Nature 2005; 435: 834. [DOI] [PubMed] [Google Scholar]
  • 134. Szymczyk A, Macheta A, Podhorecka M. Abnormal microRNA expression in the course of hematological malignancies. Cancer Manag Res 2018; 10: 4267–4277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Rupaimoole R, Slack FJ. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov 2017; 16: 203. [DOI] [PubMed] [Google Scholar]
  • 136. Stahlhut C, Slack FJ. Combinatorial action of microRNAs let-7 and miR-34 effectively synergizes with erlotinib to suppress non-small cell lung cancer cell proliferation. Cell Cycle 2015; 14: 2171–2180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Kovalchuk O, Filkowski J, Meservy J, et al. Involvement of microRNA-451 in resistance of the MCF-7 breast cancer cells to chemotherapeutic drug doxorubicin. Mol Cancer Ther 2008; 7: 2152–2159. [DOI] [PubMed] [Google Scholar]

Articles from Therapeutic Advances in Hematology are provided here courtesy of SAGE Publications

RESOURCES