2,001
Views
8
CrossRef citations to date
0
Altmetric
Original Articles: Research

Phase 1 dose-escalation study of oral abexinostat for the treatment of patients with relapsed/refractory higher-risk myelodysplastic syndromes, acute myeloid leukemia, or acute lymphoblastic leukemia

, ORCID Icon, , , , , , , , & show all
Pages 1880-1886 | Received 30 Aug 2016, Accepted 18 Nov 2016, Published online: 02 Dec 2016

Abstract

Histone deacetylase (HDAC) inhibitor abexinostat is under investigation for the treatment of various cancers. Epigenetic changes including aberrant HDAC activity are associated with cancers, including myelodysplastic syndromes (MDS), acute myeloid leukemia (AML), and acute lymphoblastic leukemia (ALL). In this phase 1 dose-escalation study, 17 patients with relapsed/refractory higher-risk MDS, AML, or ALL received oral abexinostat (60, 80 [starting dose], 100, or 120 mg) twice daily (bid) on Days 1–14 of 21-day cycles. The most common treatment-related grade ≥3 adverse events were thrombocytopenia (29%) and neutropenia (24%), none of which led to discontinuation. Maximum-tolerated dose was not reached. Of 12 evaluable patients, best response was stable disease in 1 patient. This study was closed due to limited clinical benefit. Future development of oral abexinostat 100 mg bid in patients with MDS, AML, or ALL should focus on combination regimens.

ISRCTN registry: 99680465

Introduction

Epigenetic changes have been recognized as drivers of malignant phenotypes in various cancers,[Citation1] including myelodysplastic syndromes (MDS),[Citation2] acute myeloid leukemia (AML),[Citation3] and acute lymphoblastic leukemia (ALL).[Citation4] For example, equilibrium between histone acetyltransferase and deacetylase (HAT, HDAC) activity is needed for normal cell growth and function, and perturbation of that equilibrium due to aberrant expression, function, and/or recruitment of HDAC genes has been associated with various cancers.[Citation5] Acetylation of histones by HATs leads to a more open chromatin conformation, promoting gene transcription, while removal of acetyl groups by HDACs results in tighter histone–DNA interactions and inhibition of transcription.[Citation6] HDACs are also known to regulate the acetylation (and therefore function) of many non-histone proteins important for cell growth and differentiation, including tumor suppressors.[Citation7]

Inhibition of HDACs has been associated with various downstream anticancer effects,[Citation5] and several HDAC inhibitors with varying structural class, specificity, and potency are under investigation as anticancer agents.[Citation5,Citation7] The pleotropic biological effects of HDAC inhibitors are related to their effects on both histone and non-histone proteins.[Citation8] Histone acetylation results in the re-expression of a variety of genes involved in cell growth (through induction of p21 and G1 arrest), differentiation, and survival. Non-histone proteins targeted by HDAC inhibitors include proteins involved in the regulation of gene expression, pathways of extrinsic and intrinsic apoptosis, cell cycle progression, redox pathways, mitotic division, DNA repair, cell migration, and angiogenesis. HDAC inhibitors have also been show to display immuno-modulatory effects.[Citation9]

Four HDAC inhibitors are currently approved by the US Food and Drug Administration: vorinostat, romidepsin, and belinostat for the treatment of relapsed/refractory T-cell lymphomas [Citation10–12] and panobinostat for multiple myeloma;[Citation13] panobinostat is also approved by the European Medicines Agency.[Citation14] Epigenetic rationale and preclinical data with HDAC inhibitors [Citation15–17] led to clinical studies of various HDAC inhibitors in patients with MDS, AML, and/or ALL,[Citation18–28] though reported single-agent efficacy has been limited. This stresses the need for new HDAC inhibitors.

Abexinostat (S 78454, PCI-24781) is an oral hydroxamate-based Class I and II HDAC inhibitor that has demonstrated potent preclinical activity in hematologic and solid tumors [Citation29,Citation30] and is under investigation as a single agent and in combination therapies for the treatment of various cancers. Early studies of single-agent abexinostat in patients with relapsed/refractory lymphomas and chronic lymphocytic leukemia have shown clinical activity and manageable toxicities.[Citation31,Citation32] Abexinostat has been shown to potently inhibit the growth of myeloid and lymphoid malignant cell lines.[Citation29] The primary objective of this study was to evaluate the safety and tolerability of oral abexinostat in patients with relapsed/refractory higher-risk MDS, AML, or ALL. Secondary objectives included assessment of the pharmacokinetic (PK) and pharmacodynamic profiles and disease responses to oral abexinostat.

Materials and methods

Study design

This was multicenter, open-label, phase 1, dose-escalation study of abexinostat in patients with relapsed/refractory higher-risk MDS, AML, or ALL. Patients received oral abexinostat twice daily (bid), 4 hours apart, for 14 consecutive days in a 21-day cycle for 3 cycles or until disease progression (PD) or unacceptable toxicity. After Cycle 3, patients could continue to receive abexinostat at investigator’s discretion until PD, relapse, safety concerns, or patient decision. Dose escalation followed a traditional algorithm-based 3 + 3 design. The starting dose was 80 mg bid, with de-escalation to 60 mg bid or escalation to 100 mg bid and 120 mg bid as tolerated. The study was registered in the ISRCTN registry (99680465).

The primary endpoints were the maximum tolerated dose (MTD), dose-limiting toxicities (DLTs), and the safety/tolerability profile of abexinostat. Secondary endpoints included PK and pharmacodynamic parameters, response rates, time to remission, remission duration, relapse-free survival, event-free survival (EFS), and time to blood count recoveries (platelets, neutrophils, and hemoglobin).

Key eligibility criteria

Patients with AML, MDS, or ALL were enrolled in the study. Patients with AML (excluding acute promyelocytic leukemia) were eligible; those aged 18 to <60 years must have ≥2 prior lines of therapy with maximum duration after last complete response (CR) ≤12 months and those aged ≥60 years must have ≥1 prior lines of therapy with maximum duration after first CR ≤12 months. Adult patients with International Prognostic Scoring System [Citation33] intermediate-2 or high-risk MDS that had failed hypomethylating therapy, or with histologically or cytologically confirmed B-cell ALL (excluding Philadelphia chromosome-positive ALL and B-cell ALL 3 Burkitt like) that had failed conventional or investigational therapy, were also eligible. All diagnoses were made according to the World Health Organization 2008 classifications.[Citation34]

Estimated life expectancy >8 weeks, Eastern Cooperative Oncology Group performance status ≤2, and adequate renal and hepatic function were required for inclusion. Patients with AML with white blood cell count ≥30 G/L who received hydroxyurea up to 24 hours prior to first study drug administration to stabilize count to <30 G/L were eligible. The study was performed in accordance with the ethical principles stated in the Declaration of Helsinki. Trial documents were approved by an independent Ethics Committee in accordance with local regulations, and all patients provided written informed consent.

Toxicity assessments

At each study visit, patients underwent clinical, biological, and safety evaluations. Adverse events (AEs) were assessed according to the National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0. Relationships of AEs to study drug were per investigator’s judgment. Assessment of DLTs occurred at the end of Cycle 1. Nonhematologic DLTs included any grade ≥3 biological toxicity lasting >7 consecutive days (except electrolyte abnormalities that responded to supplementation), any grade ≥3 AEs that caused inability to administer abexinostat for >7 consecutive days despite effective supportive care, or any grade ≥3 prolongation of the corrected QT (QTc) interval or grade ≥2 prolongation of the QTc interval persisting >14 days. Hematologic DLTs included any grade 4 neutropenia or thrombocytopenia not related to PD and persisting through the end of Cycle 1; Cycle 1 could be extended up to Day 42 without additional abexinostat doses until peripheral hematologic parameter recovery. Grade ≥3 anemia was considered a DLT if deemed a hemolytic process secondary to abexinostat, and grade ≥3 lymphopenia was considered a DLT if clinically significant. Triplicate electrocardiograms (ECGs) were performed at baseline and at various time points during Cycle 1, Days 1, 8, and 14. Thereafter, predose ECGs were performed on Day 1 of each cycle and at the end of study visit. All ECGs were centrally reviewed by an independent cardiologist at Cardiabase (Nancy, France).

Response assessments

Responses were evaluated based on recommendations from the European LeukemiaNet [Citation35] for AML and ALL and the International Working Group criteria [Citation36] for MDS. Patients evaluable for response had received ≥1 abexinostat dose and had ≥1 baseline and ≥1 postbaseline tumor evaluation.

Pharmacokinetic/pharmacodynamic assessments

Blood samplings for PK assessments were taken on Day 1 (predose, 0.5, 1, 2, 3, and 4 hours after first dose, and 0.5, 1, 2, 3, 4, and 6 hours after second dose), Day 2 (predose), Day 8 (predose), and Day 14 (predose, 0.5, 1, 2, 3, and 4 hours after first dose, and 0.5, 1, 2, 3, 4, and 6 hours after second dose) of Cycle 1, as well as on Day 1 (predose) of Cycle 2. Plasma samples were transferred to a central laboratory for analysis. Concentrations of abexinostat and its metabolites were measured by liquid chromatography with tandem mass spectrometry detection. Blood from a subset of the PK sampling times were also used for pharmacodynamic assessments via measurement of histone H3 acetylation in peripheral blood mononuclear cells.

Results

Patient characteristics and disposition

A total of 17 patients were enrolled, treated with abexinostat, and included in the safety analysis (). The median age was 71 years, and the most common diagnosis was AML (13 patients, 76%; 35% de novo, 41% secondary); 2 patients each had MDS and ALL (12% each). Patients had a median of 3 prior lines of chemotherapy (range, 1–10). Baseline characteristics were similar among cohorts and reflective of the target populations.

Table 1. Baseline characteristics.

All 17 patients withdrew from the study, at a median treatment duration of 3.0 weeks (). Reasons for withdrawal included PD (82%, n = 14) and AEs (18%, n = 3, each at 100 mg bid). All planned dose levels were explored. Patients were treated with abexinostat 80 mg bid (n = 3), 100 mg bid (n = 11), and 120 mg bid (n = 3). The number of patients who could be evaluated for DLT was 3/3, 6/11, and 2/3 in the 3 dose levels, respectively. Among the 6 patients who were not evaluable for DLT, 4 had discontinued during Cycle 1 for reasons other than a DLT, 1 did not undergo a hematologic DLT assessment during Cycle 1, and 1 did not receive ≥22 of 28 treatment doses.

Table 2. Patient disposition.

Safety

Grade ≥3 AEs were reported for all 17 patients and were considered treatment-related in 9 patients (53%). The most common (reported in >1 patient) treatment-related hematologic AEs were thrombocytopenia (n = 6), neutropenia (n = 4), and anemia (n = 4), and nonhematologic AEs were nausea (n = 12), asthenia (n = 8), vomiting (n = 6), diarrhea (n = 4), decreased appetite (n = 4), abdominal pain upper (n = 3), and abdominal pain (n = 2). Treatment-related grade ≥3 AEs reported in >1 patient were thrombocytopenia (n = 5), neutropenia (n = 4), anemia (n = 3), and asthenia (n = 3).

Of 11 DLT-evaluable patients, 1 of 6 treated at the 100 mg bid dose experienced a DLT. This 69-year-old woman had a history of atrial fibrillation, diabetes mellitus, hypertension, venous insufficiency, and mitral regurgitation. On Day 12, she died at home, and the reported cause of death was cardiac arrest; no autopsy was performed. As such, the event was assessed as drug-related as this possibility could not be excluded.

Serious AEs (SAEs) were reported in 16 patients (94%). All grade SAEs reported in >1 patient included thrombocytopenia (n = 6), neutropenia (n = 4), febrile neutropenia (n = 2), vomiting (n = 2), general physical health deterioration (n = 2), and staphylococcal sepsis (n = 2). Thrombocytopenia was the only drug-related SAE reported in >1 patient (n = 5). Four patients discontinued treatment due to AEs unrelated to abexinostat: 1 with leukocytosis that resolved after discontinuation and 3 who died, with septic shock, general physical health deterioration, and progression of malignant neoplasm (1 patient each).

No enrolled patients had baseline ECG abnormalities, and 2 of 17 patients developed clinically significant emergent ECG abnormalities during the treatment period. One patient treated at 100 mg bid experienced an isolated event of ventricular premature beat (monomorphic), and 1 patient treated at 120 mg bid experienced several events of atrial fibrillation and abnormal T pattern in precordial leads. QTc according to Bazett’s (QTcB) or Fridericia’s (QTcF) formulas were both increased >30–60 msec in 8 patients (47%); 2 (12%) and 3 (18%) patients, respectively, had increases >60 msec in QTcB or QTcF. Two (12%) patients had maximum absolute QTcB and QTcF >480 msec and no patients had QTcB or QTcF >500 msec.

Pharmacokinetics and pharmacodynamics

Abexinostat was rapidly absorbed and eliminated with 80, 100, and 120 mg bid dosing (). Though data were limited, mean exposures after the first abexinostat dose on Days 1 and 14 appeared to increase in a greater than dose-proportional manner. Slight accumulation of abexinostat was observed after 14 days of treatment at all dose levels. Abexinostat was extensively converted to metabolites S 78730 and S 78731 and accumulation of metabolites was observed after 14 days of dosing at all dose levels. Histone H3 acetylation showed high interpatient variability in peripheral blood mononuclear cells of evaluable patients (n = 7). No robust or reproducible effect was observed on the ratio of histone H3 acetylation versus total histone H3, with only a moderate increase in ratio from pretreatment in selected patients (Supplemental Figure).

Table 3. Pharmacokinetic parameters for abexinostat.

Efficacy

Of 12 patients evaluable for response, the best response was stable disease in 1 patient with MDS treated with 120 mg bid, with an EFS of 9.29 weeks. The median EFS for all 12 patients was 3.14 weeks (range, 2.57–9.29 weeks). Bone marrow blast changes from baseline were reported in 9 patients; decreases in blast involvement were reported at ≥1 postbaseline time point in 3 of 9 patients, with the highest decrease of 22% in 1 patient.

Discussion

Study results demonstrated the tolerability of oral abexinostat in patients with relapsed/refractory higher-risk MDS, AML, and ALL. Based on previous clinical experience with abexinostat, no unexpected safety concerns were observed in this study. Grade ≥3 AEs were primarily hematologic; the most common treatment-related grade ≥3 AEs reported were thrombocytopenia (29%), neutropenia (24%), anemia (18%), and asthenia (18%), which did not result in treatment discontinuation. Thrombocytopenia is a constant toxic effect of HDAC inhibitors, including abexinostat, and its mechanisms are not yet fully understood. They include reduction of platelet production by megakaryocytes which could be reversed by thrombopoietin administration in a mouse model.[Citation37] A more recent study showed that in addition to a defect in proplatelet formation which was mainly p53-independent, abexinostat also inhibited megakaryocyte differentiation by inducing progenitor and precursor apoptosis through the induction of p53.[Citation38] ECG changes including QTc prolongation have been reported with other HDAC inhibitors and a potential class effect has been suggested.[Citation39,Citation40] However, in this study and others of abexinostat,[Citation31,Citation32,Citation41] treatment with abexinostat did not result in clinically relevant ECG changes. The MTD was not identified. Limited clinical benefit was seen in the 12 patients evaluable for response (best response of stable disease in 1 patient), and the study was closed. Abexinostat was rapidly absorbed and eliminated, with tmax 0.5–1 hour in most cohorts, supporting the use of bid dosing separated by 4 hours. In pharmacodynamic analyzes, histone H3 acetylation varied highly across patients; however, analyzes were limited (n = 7). Efficacy results were in concordance with what has been reported with other single-agent HDAC inhibitors in these patient populations, with hematologic improvements seen in some patients but limited clinical responses.[Citation18–28]

HDAC inhibitors have shown preclinical additive or synergistic effects with a variety of both novel agents and chemotherapies;[Citation42] however, preclinical effects have not consistently translated to clinical results. Combinations with DNA methyltransferase inhibitors are among the most appealing since they are associated with an increased reactivation of transcription as well as an increased efficacy on tumor cells as compared with single agents.[Citation43] Results from the preliminary phase 1/2 studies of vorinostat and azacitidine or decitabine showed promising activity but were not confirmed by randomized studies, which did not show significant improvements with the addition of HDAC inhibition.[Citation44–46] However, results may have been limited by suboptimal dosing and schedule, as preclinical studies have shown that synergy is highly dependent on sequencing of the hypomethylator and HDAC inhibitor,[Citation47] and doses used as single agents may be limited by overlapping toxicities, leading to short exposure to treatment. Additional randomized studies are ongoing. Some studies of chemotherapies combined with HDAC inhibitors in MDS/AML or ALL have shown encouraging results, but no randomized studies have been published to date.[Citation44,Citation48,Citation49]. Our results show that abexinostat can be administered safely on Days 1–14 of 21-day cycles in patients with MDS and acute leukemias. As with other HDAC inhibitors, the safety profile of abexinostat is compatible with combination regimens for MDS and AML, which should focus on DNA methyltransferase inhibitor combinations exploring new dosing and schedules.

Potential conflict of interest

Disclosure forms provided by the authors are available with the full text of this article online at http://dx.doi.org/10.1080/10428194.2016.1263843.

Supplemental material

ICMJE Forms for Disclosure of Potential Conflicts of Interest

Download Zip (5.3 MB)

Supplemental Figure

Download MS Word (125.6 KB)

Acknowledgements

We thank the patients who participated in the study, their supporters, and the investigators and clinical research staff from the study centers. This study was sponsored by Pharmacyclics LLC, an AbbVie Company. This manuscript was developed with editorial support from Stacey Rose, PhD, and funded by Pharmacyclics LLC, an AbbVie Company.

References

  • Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007;128:683–692.
  • Issa JP. Epigenetic changes in the myelodysplastic syndrome. Hematol Oncol Clin North Am. 2010;24:317–330.
  • Conway O'Brien E, Prideaux S, Chevassut T. The epigenetic landscape of acute myeloid leukemia. Adv Hematol. 2014;2014:103175.
  • Peirs S, Van der Meulen J, Van de Walle I, et al. Epigenetics in T-cell acute lymphoblastic leukemia. Immunol Rev. 2015;263:50–67.
  • New M, Olzscha H, La Thangue NB. HDAC inhibitor-based therapies: can we interpret the code? Mol Oncol. 2012;6:637–656.
  • Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov. 2006;5:769–784.
  • Khan O, La Thangue NB. HDAC inhibitors in cancer biology: emerging mechanisms and clinical applications. Immunol Cell Biol. 2012;90:85–94.
  • West AC, Johnstone RW. New and emerging HDAC inhibitors for cancer treatment. J Clin Investig. 2014;124:30–39.
  • Tang J, Yan H, Zhuang S. Histone deacetylases as targets for treatment of multiple diseases. Clin Sci (Lond). 2013;124:651–662.
  • Zolinza (vorinostat) [prescribing information]. Whitehouse Station, NJ: Merck & Co., Inc.; 2015.
  • Istodax (romidepsin) [prescribing information]. Summit, NJ: Celgene Corporation; 2014.
  • Beleodaq (belinostat) [prescribing information]. Irvine, CA: Spectrum, Pharmaceuticals, Inc.; 2014.
  • Farydak (panobinostat). East Hanover, NJ: Novartis Pharmaceuticals Corporation; 2015.
  • Farydak (panobinostat) [summary of product characteristics]. Nuremberg, Germany: Novartis Pharma GmbH; 2015.
  • Chambers AE, Banerjee S, Chaplin T, et al. Histone acetylation-mediated regulation of genes in leukaemic cells. Eur J Cancer. 2003;39:1165–1175.
  • Einsiedel HG, Kawan L, Eckert C, et al. Histone deacetylase inhibitors have antitumor activity in two NOD/SCID mouse models of B-cell precursor childhood acute lymphoblastic leukemia. Leukemia. 2006;20:1435–1436.
  • Scuto A, Kirschbaum M, Kowolik C, et al. The novel histone deacetylase inhibitor, LBH589, induces expression of DNA damage response genes and apoptosis in Ph- acute lymphoblastic leukemia cells. Blood. 2008;111:5093–5100.
  • Gore SD, Weng LJ, Figg WD, et al. Impact of prolonged infusions of the putative differentiating agent sodium phenylbutyrate on myelodysplastic syndromes and acute myeloid leukemia. Clin Cancer Res. 2002;8:963–970.
  • Kuendgen A, Schmid M, Schlenk R, et al. The histone deacetylase (HDAC) inhibitor valproic acid as monotherapy or in combination with all-trans retinoic acid in patients with acute myeloid leukemia. Cancer. 2006;106:112–119.
  • Garcia-Manero G, Assouline S, Cortes J, et al. Phase 1 study of the oral isotype specific histone deacetylase inhibitor MGCD0103 in leukemia. Blood. 2008;112:981–989.
  • Garcia-Manero G, Yang H, Bueso-Ramos C, et al. Phase 1 study of the histone deacetylase inhibitor vorinostat (suberoylanilide hydroxamic acid [SAHA]) in patients with advanced leukemias and myelodysplastic syndromes. Blood. 2008;111:1060–1066.
  • Schaefer EW, Loaiza-Bonilla A, Juckett M, et al. A phase 2 study of vorinostat in acute myeloid leukemia. Haematologica. 2009;94:1375–1382.
  • Byrd JC, Marcucci G, Parthun MR, et al. A phase 1 and pharmacodynamic study of depsipeptide (FK228) in chronic lymphocytic leukemia and acute myeloid leukemia. Blood. 2005;105:959–967.
  • Klimek VM, Fircanis S, Maslak P, et al. Tolerability, pharmacodynamics, and pharmacokinetics studies of depsipeptide (romidepsin) in patients with acute myelogenous leukemia or advanced myelodysplastic syndromes. Clin Cancer Res. 2008;14:826–832.
  • Gojo I, Jiemjit A, Trepel JB, et al. Phase 1 and pharmacologic study of MS-275, a histone deacetylase inhibitor, in adults with refractory and relapsed acute leukemias. Blood. 2007;109:2781–2790.
  • DeAngelo DJ, Spencer A, Bhalla KN, et al. Phase Ia/II, two-arm, open-label, dose-escalation study of oral panobinostat administered via two dosing schedules in patients with advanced hematologic malignancies. Leukemia. 2013;27:1628–1636.
  • Giles F, Fischer T, Cortes J, et al. A phase I study of intravenous LBH589, a novel cinnamic hydroxamic acid analogue histone deacetylase inhibitor, in patients with refractory hematologic malignancies. Clin Cancer Res. 2006;12:4628–4635.
  • Odenike OM, Alkan S, Sher D, et al. Histone deacetylase inhibitor romidepsin has differential activity in core binding factor acute myeloid leukemia. Clin Cancer Res. 2008;14:7095–7101.
  • Rivera-Del Valle N, Gao S, Miller CP, et al. PCI-24781, a novel hydroxamic acid HDAC inhibitor, exerts cytotoxicity and histone alterations via caspase-8 and FADD in leukemia cells. Int J Cell Biol. 2010;2010:207420.
  • Buggy JJ, Cao ZA, Bass KE, et al. CRA-024781: a novel synthetic inhibitor of histone deacetylase enzymes with antitumor activity in vitro and in vivo. Mol Cancer Ther. 2006;5:1309–1317.
  • Evens AM, Balasubramanian S, Vose JM, et al. A phase I/II multicenter, open-label study of the oral histone deacetylase inhibitor abexinostat in relapsed/refractory lymphoma. Clin Cancer Res. 2016;22:1059–1066.
  • Morschhauser F, Terriou L, Coiffier B, et al. Phase 1 study of the oral histone deacetylase inhibitor abexinostat in patients with Hodgkin lymphoma, non-Hodgkin lymphoma, or chronic lymphocytic leukaemia. Investig New Drugs. 2015;33:423–431.
  • Greenberg P, Cox C, LeBeau MM, et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 1997;89:2079–2088.
  • 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;114:937–951.
  • Dohner H, Estey EH, Amadori S, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115:453–474.
  • Cheson BD, Greenberg PL, Bennett JM, et al. Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia. Blood. 2006;108:419–425.
  • Bishton MJ, Harrison SJ, Martin BP, et al. Deciphering the molecular and biologic processes that mediate histone deacetylase inhibitor-induced thrombocytopenia. Blood. 2011;117:3658–3668.
  • Ali A, Bluteau O, Messaoudi K, et al. Thrombocytopenia induced by the histone deacetylase inhibitor abexinostat involves p53-dependent and -independent mechanisms. Cell Death Dis. 2013;4:e738.
  • Kristeleit R, Fong P, Aherne GW, et al. Histone deacetylase inhibitors: emerging anticancer therapeutic agents? Clin Lung Cancer. 2005;7:S19–S30.
  • Molife R, Fong P, Scurr M, et al. HDAC inhibitors and cardiac safety. Clin Cancer Res. 2007;13:1068. Author reply 1068–1069.
  • Ribrag VK, Kim WS, Bouabdallah R, et al. Safety and efficacy of abexinostat, a pan-histone deacetylase (HDAC) inhibitor, in non-Hodgkin lymphoma and chronic lymphocytic leukemia: results of an ongoing phase 2 study [abstract]. Blood. 2015;126:Abstract 256.
  • Thurn KT, Thomas S, Moore A, et al. Rational therapeutic combinations with histone deacetylase inhibitors for the treatment of cancer. Future Oncol. 2011;7:263–283.
  • Gore SD, Baylin S, Sugar E, et al. Combined DNA methyltransferase and histone deacetylase inhibition in the treatment of myeloid neoplasms. Cancer Res. 2006;66:6361–6369.
  • Stahl M, Gore SD, Vey N, et al. Lost in translation? Ten years of development of histone deacetylase inhibitors in acute myeloid leukemia and myelodysplastic syndromes. Expert Opin Investig Drugs. 2016;25:307–317.
  • Prebet T, Sun Z, Figueroa ME, et al. Prolonged administration of azacitidine with or without entinostat for myelodysplastic syndrome and acute myeloid leukemia with myelodysplasia-related changes: results of the US Leukemia Intergroup trial E1905. J Clin Oncol. 2014;32:1242–1248.
  • Issa JP, Garcia-Manero G, Huang X, et al. Results of phase 2 randomized study of low-dose decitabine with or without valproic acid in patients with myelodysplastic syndrome and acute myelogenous leukemia. Cancer. 2015;121:556–561.
  • Cameron EE, Bachman KE, Myohanen S, et al. Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nat Genet. 1999;21:103–107.
  • Mummery A, Narendran A, Lee KY. Targeting epigenetics through histone deacetylase inhibitors in acute lymphoblastic leukemia. Curr Cancer Drug Targets. 2011;11:882–893.
  • Sekeres M, Othus M, List AF, et al. Additional analyses of a randomized phase II study of azacitidine combined with lenalidomide or with vorinostat vs. azacitidine monotherapy in higher-risk myelodysplastic syndromes and chronic myelomonocytic leukemia: North American Intergroup Study SWOG S1117 [abstract]. Blood. 2015;126:Abstract 908.