103
Views
19
CrossRef citations to date
0
Altmetric
Review

Clinical use and applications of histone deacetylase inhibitors in multiple myeloma

, &
Pages 35-44 | Published online: 06 May 2016

Abstract

The incorporation of various novel therapies has resulted in a significant survival benefit in newly diagnosed and relapsed patients with multiple myeloma (MM) over the past decade. Despite these advances, resistance to therapy leads to eventual relapse and fatal outcomes in the vast majority of patients. Hence, there is an unmet need for new safe and efficacious therapies for continued improvement in outcomes. Given the role of epigenetic aberrations in the pathogenesis and progression of MM and the success of histone deacetylase inhibitors (HDACi) in other malignancies, many HDACi have been tried in MM. Various preclinical studies helped us to understand the antimyeloma activity of different HDACi in MM as a single agent or in combination with conventional, novel, and immune therapies. The early clinical trials of HDACi depicted only modest single-agent activity, but recent studies have revealed encouraging clinical response rates in combination with other antimyeloma agents, especially proteasome inhibitors. This led to the approval of the combination of panobinostat and bortezomib for the treatment of relapsed/refractory MM patients with two prior lines of treatment by the US Food and Drug Administration. However, it remains yet to be defined how we can incorporate HDACi in the current therapeutic paradigms for MM that will help to achieve longer disease control and significant survival benefits. In addition, isoform-selective and/or class-selective HDAC inhibition to reduce unfavorable side effects needs further evaluation.

Introduction

Multiple myeloma (MM) is a plasma cell malignancy, characterized by an accumulation of high levels of monoclonal immunoglobulins or paraproteins in blood and/or urine and end organ damage, including anemia, renal failure, hypercalcemia, and bony lesions.Citation1 MM is the second most commonly diagnosed hematologic malignancy representing 1.6% of all new cancer cases in the US. The outcomes of these patients have not been satisfactory, and the 5-year survival is 46.6% according to surveillance, epidemiology, and end results analysis.Citation2

Over the last two decades, the treatment paradigm for MM has changed with the use of autologous stem cell transplantation (ASCT) and novel therapeutic options including proteasome inhibitors (PIs) and immunomodulatory drugs (IMiDs).Citation3 The incorporation of novel drugs, particularly thalidomide, lenalidomide (Len), and bortezomib (Btz), has resulted in a significant prolongation of overall survival (OS) in newly diagnosed and relapsed patients.Citation4 Despite these advances, acquired or intrinsic resistance to therapy leads to eventual relapse and fatal outcomes in vast majority of patients. In an analysis of 286 patients with relapsed MM, who were refractory to Btz and had relapsed following, or were refractory to or ineligible to receive an IMiD, OS and event-free survival were 9 and 5 months, respectively. These findings indicate the poor outcome of patients, once they become refractory to current modalities and an unmet need for safe and efficacious novel therapies.Citation5

MM is a biologically complex disease, with great heterogeneity in terms of genetic alterations, thereby giving rise to individual differences in overall response and survival of patients receiving the same treatment. In addition to genetic alterations such as point mutations, deletions, or translocations, epigenetic alterations and abnormal microRNA expression also contribute to the pathogenesis of MM.Citation6Citation9

Epigenetic aberrations are heritable changes in gene expression that occur independent of changes in the primary DNA sequence. Most of the epigenetic mechanisms occur at the level of chromatin. Chromatin is built up by nucleosomes that contain ~146 bp of DNA wrapped around an octamer consisting of four core histones (H3–H4 tetramer and two H2A–H2B dimers). The modifications of these nucleosomes play an important role in the transition of chromatin between open and closed states.Citation10 The N-terminal tail regions of histones undergo a wide variety of enzyme modifications, including acetylation, methylation, sumoylation, phosphorylation, and ubiquitination, which are crucial in modulating gene expression. This phenomenon is referred to as the “histone code” and has a significant effect on gene expression and chromatin structure.Citation11 Histone acetylation is controlled by histone acetyl transferases, which transfer acetyl groups to the side chain of lysine residues on N-terminal, and histone deacetylases (HDACs), which deacetylate and counterbalance activity of histone acetyl transferases.Citation12,Citation13

The HDAC family of enzymes regulates the acetylation level of histones in chromatin and various nonhistone substrates, including many proteins involved in tumor progression, cell cycle control, apoptosis, angiogenesis, and cell invasion.Citation14 The HDAC family consists of 18 genes that are grouped into classes I, II, III, and IV based on their homology to respective yeast orthologs Rpd3, HdaI, and Sir2.Citation15 Various classes of HDAC and their localization and function have been shown in . HDAC inhibitors (HDACi) typically target the “classical” classes I, II, and IV HDACs (which contain a Zn2þ catalytic ion in their active site) and not class III HDACs that use NAD+ as the essential cofactor.Citation14,Citation16

Table 1 Various classes of HDAC

This review focuses on the antimyeloma activity of different HDACi in preclinical and clinical settings.

Epigenetic changes in MM

Epigenetic aberrations play an important role in the initiation and progression of most of the malignancies, including MM, and this is largely attributed to alterations in the expression of histone-modifying enzymes.Citation17,Citation18

In cancer, global DNA hypomethylation of repetitive sequences (such as long interspersed nuclear element 1 [LINE-1] and Alu repeats), gene bodies, and intergenic regions has been observed. This contributes to genomic instability, transposon activation, proto-oncogene activation, and loss of normal imprinting patterns. In addition, site-specific CpG island hypermethylation of gene promoters such as tumor suppressor genes results in gene silencing.Citation19 Even in MM, there is increased global hypomethylation of the LINE-1 and Alu repetitive elements compared to normal control subjects.Citation20 This seems to be an early event in the pathogenesis, and the global methylation levels of repetitive elements decrease as the disease progresses from monoclonal gammopathy of unknown significance to MM.Citation20,Citation21

Epigenetic alterations also increase the vulnerability to genomic instability. LINE-1 hypomethylation is found to be associated with translocations of chromosome 14 and deletion of chromosome 13q.Citation21 Also, t(4;14) translocation showed more frequent hypermethylation that may underline the poor prognosis associated with its presence.Citation22

The most characteristic documentation of aberrations of histone modifications is in t(4;14) MM, which leads to overexpression of multiple myeloma SET domain containing protein (MMSET) (NSD-2), a histone methyltransferase. MMSET regulates genes involved in the p53 pathway, nuclear factor kappa B pathway, apoptosis, cell cycle regulation, DNA repair, and adhesion, and its upregulation enhances survival and adhesion of MM cells.Citation23,Citation24

In addition, epigenetic alterations may result in dysregulation of critical oncogenic pathways such as cyclin dependent kinase/retinoblastoma (CDK/Rb), Wnt/β-catenin, Janus kinase/signal transducer and activator of transcription protein (JAK/STAT), death associated protein kinase-1/p14-ARF/p53 (DAPK-1/p14 ARF/p53) pathways, which contribute to the pathogenesis of MM.Citation8

HDACs in MM

Overexpression of HDAC proteins, especially class I HDACs, has been observed in both solid and hematological malignancies.Citation25Citation29 In the majority of tumors, HDAC expression is associated with a poor prognosis.Citation30Citation32 However, HDAC expression is correlated with a better prognosis in breast cancer, acute lymphoblastic leukemia, and chronic lymphocytic leukemia.Citation33Citation35 Patients with MM with high transcript levels of HDACs 1, 2, 4, 6, and 11 show a shorter progression-free survival (PFS) than those expressing lower levels. However, when HDAC protein levels were examined, it was found that only increased HDAC1 expression correlated with poor PFS and OS.Citation36

HDACi in MM

Butyrate and trichostatin A were among the initial molecules identified as HDACi. Since then, various natural and synthetic HDACi have been developed and evaluated as anticancer agents in the preclinical and clinical settings. Major HDACi can be divided into five categories on the basis of their chemical structure (). The direct impact of HDAC inhibition on chromatin is hyperacetylation of histone proteins, which alters the chromatin structure and results in up- or downregulation of gene expression involved in cell cycle regulation, apoptosis, cytokine signaling, adhesion and migration, proteasomal degradation, drug resistance, and DNA damage.Citation37Citation39

Table 2 Classification of HDAC inhibitors

Preclinical activity of HDACi in MM

As a single agent

Microarray analysis has shown that HDACi induce transcriptional modulation of 7%–10% of the genes in myeloma and human lymphoid cell lines by acetylation of histones and nonhistone proteins.Citation38,Citation40 The pattern of gene alteration is quite similar across different HDACi in the same cell line.Citation41,Citation42 HDACi such as valproate, FK228, and ITF2357 affect the viability of interleukin (IL)-6-dependent and -independent MM cell lines, indicating that the antimyeloma activity of HDACi is not influenced by IL-6.Citation43Citation45 Moreover, coculturing MM cells with bone marrow stromal cells (BMSCs) do not protect them from death induced by LAQ824, ITF2357, LBH589, or KD5170, suggesting that HDACi could overcome the protective effect of the BMSCs.Citation45Citation48 The various possible mechanisms of the anti-myeloma activity of HDACi have been described later.

Cell cycle arrest

Almost all HDACi induce G0/SG1 arrest due to increase in histone acetylation and upregulation of cyclin-dependent kinase (CDK) inhibitor CDKN1A by p53-dependent and -independent ways, as observed in MM cell lines treated with valproate, NVP-LAQ824, LBH589, NaB, SAHA, and ITF2357.Citation43,Citation45,Citation46,Citation49Citation51 Other effects include upregulation of other CDK inhibitors such as p27 and p19 and/or the decrease of cyclins D1 and D2.Citation43,Citation52,Citation53

Apoptotic pathway

HDACi upregulate expression of proapoptotic Bcl-2 family proteins (Bax, Bak, and Bim) and downregulate antiapoptotic proteins (Bcl-2, Bcl-xL, MCL1, and XIAP). Overall, this triggers increased mitochondrial permeability and cytosolic release of cytochrome C and Smac followed by activation of intrinsic apoptotic pathway, as seen in MM cell lines treated with depsipeptide, ITF2357, LBH589, SAHA, and KD5170.Citation44,Citation45,Citation47,Citation50,Citation51 Extrinsic apoptotic pathway is activated by upregulation of death receptors and ligands, caspase-8 cleavage, and downregulation of Flice-like inhibitory protein (caspase-8 inhibitor) as seen in MM cells after valproate, suberoylanilide hydroxamic acid (SAHA), and LBH589 treatment.Citation50,Citation51,Citation54 Autophagy is a catabolic process involving the degradation of long-lived proteins or cytoplasmic organelles through the lysosomal machinery, which plays a role in valporate-induced cytotoxicity in human myeloma cell lines.Citation54

DNA damage and oxidative stress

HDACi interfere with the function of DNA-repair proteins such as Ku70, RAD51, RAD50, DNA-PKcs, BRCA1, and BRCA2, thus inducing double-stranded breaks in DNA.Citation55,Citation56 The HDACi PDX-101 and KD-5170 phosphorylate H2AX on ser139 and induce DNA damage.Citation57 Another HDACi, SDNX-275, could enhance the DNA damage response induced by the alkylating agent melphalan in MM cell lines.Citation58 Moreover, HDACi-induced chromatin hyperacetylation makes DNA more sensitive to drugs, radiation, and reactive oxygen species.Citation59 The production of reactive oxygen species observed after HDAC inhibition seems crucial as evidenced by the upregulation of several antioxidant genes such as glutathione S-transferase, glutathione reductase, and superoxide dismutase 1 and 2 on the treatment of U937 leukemic cells with vorinostat.Citation60

Ubiquitin–proteasome system

HDACi decrease activity of 20S proteasome and downregulate genes encoding 26S proteasome and ubiquitin conjugating enzymes in MM cells.Citation38,Citation46 Also, tubacin or pan HDACi such as SAHA or LBH589, hyperacetylate α-tubulin, accumulate polyubiquitinated proteins, leading to apoptosis subsequently.Citation48 HDAC inhibition enhances the cytotoxic effects of Btz both in vitro and in vivo,Citation38,Citation47,Citation48,Citation61,Citation62 which will be discussed later.

BMSC interaction

Multiple cytokines such as IL-6, IL-1, insulin-like growth factor-1 (IGF-1), tumor necrosis factor-α, vascular endothelial growth factor (VEGF), Dickkopf-related protein 1, and secreted frizzled-related protein are secreted at high levels by either the malignant plasma cells or the BMSCs. This then causes activation of signaling pathways in the MM cells and further promotes their interaction with cells in the tumor microenvironment such as BMSCs, endothelial cells, osteoblasts, and osteoclasts. The net result of such interaction is increased tumor growth, angiogenesis, bone disease, and drug resistance.Citation63,Citation64

HDACi downregulate the expression of genes involved in cytokine signaling such as IGF-1, IGF-1 receptor, and IL-6 receptor.Citation38 Mitsiades et alCitation38,Citation50 showed that vorinostat not only suppresses the expression of receptor genes involved in MM cell proliferation, survival, and/or migration such as IGF-1R, IL-6R, TNF-R, CD138 (syndecan-1), and CXCR-4.55 but also reduces the autocrine IGF-1 and paracrine IL-6 secretion of BMSC.

Antiangiogenesis

HDACi induce alteration of numerous pro- and antiangiogenic genes (angiopoietin, TIE2, eNOS, p53, pVHL, and thrombospondin 1), thereby targeting increased angiogenesis in MM.Citation65Citation67 Valporate decreases VEGF secretion and VEGF receptor expression, resulting in inhibition of the vascular tubule formation of endothelial cells in cocultures with MM cells.Citation43,Citation68,Citation69

In combination with PIs

HDACi have been tried in combination with a variety of agents for MM, but the most synergistic effects are seen with Btz. The precise mechanisms causing this synergy are not yet completely defined. The best understood mechanism is dual inhibition of the proteasomal and aggresomal protein degradation pathways, targeted by Btz and HDACi, respectively. Btz inhibits proteasome and causes accumulation of polyubiquitinated proteins that form an aggresome by a process dependent on the interaction of HDAC6 with tubulin and dynein complex. HDAC6 inhibition leads to increased hyperacetylation of tubulin and upregulation of polyubiquitinated proteins, resulting in apoptosis.Citation70,Citation71 In accordance with the above mentioned dual inhibition phenomenon, non-selective HDACi like vorinostat as well as selective HDAC6i like tubacin and ACY-1215 have been found to inhibit aggresome formation and induce caspase-mediated apoptosis in MM when combined with Btz.Citation72Citation74

In addition, HDAC1 overexpression causes resistance to Btz both in vitro and in vivo, which is reversed by the class I HDACi romidepsin. Moreover, Btz downregulates the expression of class I HDACs and enhances HDACi cytotoxicity.Citation75 Taken together, Btz and HDACi combination appears to be a promising therapeutic strategy that can overcome drug resistance.

In combination with other agents

Preclinical studies have shown that addition of vorinostat or panobinostat to MM cell lines and tumor cells derived from patients resistant to conventional therapies increases their susceptibility to IMiDs (such as pomalidomide or Len) and dexamethasone.Citation38,Citation76 Moreover, treatment of MM cells with vorinostat increases their sensitivity to DNA-damaging agents, such as doxorubicin or melphalan.Citation38,Citation77

Treatment of MM cell line with sodium butyrate in combination with DNA methyltransferase inhibitor, decitabine, resulted in increased expression of p16 gene and G1 arrest, a phenomenon not seen with either agent alone.Citation78 Furthermore, mTORC1 inhibitor RAD001 caused potent G0/G1 arrest, while LBH589 induced pronounced apoptosis, both of which were enhanced when the drugs were used in combination.Citation79

In addition, additive effects of HDACi have been seen in conjunction with RSK2 (Ser227) inhibitor BI-D1870 and heat shock protein-90 (alpha/beta) inhibitor NVP-AUY922 in preclinical studies.Citation80,Citation81 Also, HDACi-inducible Bim is primarily neutralized by Bcl-2 and Bcl-xL, thus providing a mechanistic framework by which Bcl-2 antagonists potentiate the lethality of HDACi.Citation82 Also, SAHA and trichostatin A induce G1 arrest by upregulating p21 and p27 and inhibiting E2F transcriptional activity. The tumor necrosis factor-related apoptosis-inducing ligand effect can be enhanced after HDACi pretreatment and is found to be consistent with the upregulation of proapoptotic Bim, Bak, Bax, Noxa, and p53 upregulated modulator of apoptosis (PUMA) and downregulation of antiapoptotic Bcl-2 and Bcl-xL.Citation83

In combination with immune therapies

In addition to all the above-mentioned combination therapies, HDACi enhance MHC classes I and II expression and tumor-associated antigens on tumor cells, inducing cell death mediated by natural killer cells and cytotoxic T-cells.Citation84Citation89 Also, vorinostat induces the secretion of adenosine triphosphate and high mobility group box 1 protein (HMGB-1) and expression of calreticulin on the tumor cell surface, which are important mediators of recognition and phagocytosis by dendritic cells.Citation88Citation90 However, the effect of HDACi on the immune cells is far from clear and has been reviewed in detail elsewhere. Overall, it appears that the HDACi could promote or inhibit the functions of regulatory T-cells, myeloid-derived suppressor cells, and tumor-associated macrophages.Citation91

HDACi have shown favorable responses in combination with immune therapies in preclinical settings. Christiansen et alCitation88 observed synergistic responses when vorinostat or panobinostat was used in combination with anti-CD40 and anti-CD137 antibodies in solid tumors. He also noted an important role for CD8+ cytotoxic T-cells and natural killer cells for the synergy observed.Citation88 In another study, LAQ824 induced synergistic cell death in combination with adoptive transfer of tumor-specific T-cells in melanoma.Citation92 However, the effect of this combination remains unexplored in MM. One preclinical study showed that LBH589 impairs the phenotype and function of dendritic cells by downregulating dendritic cell maturation, antigen presentation, and T-cell costimulation markers on immature and mature dendritic cells.Citation93 Thus, it is important to examine the immune status of patients with MM before and after HDACi treatment. Such studies will help us not only to better understand the effects of HDACi on immune cells but also identify potential combinations of HDACi with immune therapies.

Clinical trials using HDACi in MM

HDAC represents a very interesting clinical target for the development of novel antimyeloma therapy. The early clinical trials of different HDACi have revealed only modest single-agent activity, but encouraging clinical response rates have been reported in combination with other antimyeloma agents such as PIs, IMiDs, dexamethasone, and conventional cytotoxic therapy.

Vorinostat

Vorinostat (SAHA) is a potent nonselective HDACi with a hydroxamic acid moiety, which causes reversible inhibition of classes I and II HDACs. It was the first epigenetic agent used therapeutically in malignancy and was approved by the US Food and Drug Administration (FDA) for the treatment of cutaneous T-cell lymphoma in 2006.Citation94

In the initial dose-escalating Phase I trial of vorinostat in relapsed/refractory MM (RRMM), 13 patients with a median of three prior lines of therapy were included. The most common drug-related adverse effects (AEs) included fatigue, anorexia, dehydration, diarrhea, and nausea and were mostly grade ≤2. Among the ten evaluable patients, one had a minimal response and nine had stable disease (SD).Citation95

Based on the synergy with PIs depicted in preclinical studies, a Phase I trial evaluated vorinostat in combination with Btz in patients with RRMM. The 23 patients enrolled in the study had received a median of seven prior regimens with 20 patients post ASCT and 19 patients with prior Btz (nine of whom were Btz refractory). The dose-limiting toxicity was prolonged QT interval seen in two patients. The most common toxicities were myelosuppression, diarrhea, and fatigue. The overall response rate (ORR) was 42%, with two patients having very good partial response (VGPR) and seven patients having PR, including three patients who were Btz refractory.Citation96

VANTAGE 095 was a multicenter, open-label Phase IIB study in which 143 patients with RRMM (Btz refractory) received vorinostat in combination with Btz till progressive disease, unacceptable toxicities, or patient withdrawal. The ORR was 11%, while 47% of patients had SD. The median duration of response (DOR) was 7.0 months, and the median OS was 11.1 months. However, serious AEs were reported in 65% of patients, resulting in treatment discontinuations in 11% of patients.Citation97

On the basis of these encouraging responses, a multicenter, randomized, double-blind Phase III study, VANTAGE 088 trial, was conducted. They enrolled 637 patients with RRMM who had progressive disease after one to three prior antimyeloma treatments (but were Btz sensitive) and randomized them to receive Btz with vorinostat or placebo. The addition of vorinostat to Btz significantly improved the ORR (56% vs 41%) and clinical benefit rates (CBRs) (71% vs 53%). The median PFS also increased from 6.83 to 7.63 months, but the median OS was not significantly different between the two groups. More patients in the vorinostat group developed high-grade AEs, especially fatigue, myelosuppression, and gastrointestinal disorders compared to the placebo group. The authors concluded that though the study achieved the primary end point of prolonging the PFS, the clinical value of adding vorinostat to Btz needed further evaluation with regard to optimizing the dose of vorinostat to minimize toxicity.Citation98

Vorinostat has also been used in combination with carfilzomib in compassionate use setting for patients with RRMM and was well tolerated.Citation99 A Phase I dose-escalation trial of vorinostat with Len/dexamethasone in RRMM demonstrated an ORR of 47%. Serious AEs were reported in 45% of the patients and were considered to be study drug related in 22%.Citation100 Hence, this combination seems to be effective and needs further evaluation.

Panobinostat

Panobinostat (LBH589) is a cinnamic hydroxamic acid analog that exhibits tenfold higher inhibitory activity against classes I, II, and IV HDACs than vorinostat. A Phase II multicenter study of oral panobinostat in 38 heavily pretreated patients with RRMM showed that it was well tolerated and the most common AEs were nausea and fatigue. But the ORR was lower than what was seen in the preclinical studies with VGPR in one patient, mixed response (MR) in one patient, and SD in three patients.Citation101

In view of poor results with its use as monotherapy and preclinical data depicting synergy with Btz, a Phase Ib trial studied the use of panobinostat in combination with Btz in RRMM. Among the 47 patients enrolled in the dose-escalation phase, 76% of patients had ≥MR with responses seen in ten of 15 Btz refractory patients. Out of the 12 evaluable patients enrolled in the dose-expansion phase, MR was seen in 75% of patients.Citation102

PANORAMA 2 is a Phase II trial of panobinostat in combination with Btz and dexamethasone in patients with relapsed and Btz refractory MM with at least two prior lines of therapy. Fifty-five heavily pretreated patients with a median of four prior regimens were enrolled. The ORR was 34.5%, and the CBR was 52.7%. Median PFS was 5.4 months, and the median DOR was 6.0 months. Common grade 3/4 AEs included thrombocytopenia (63.6%), fatigue (20.0%), and diarrhea (20.0%).Citation103

PANORAMA 1 is a multicenter double-blind Phase III trial of patients with RRMM after one to three previous treatment regimens. Approximately 768 eligible patients were randomized to receive Btz and dexamethasone with panobinostat or placebo. It was demonstrated that though the ORR (60.7% vs 54.6%) was similar, the proportion of patients achieving complete response (CR) or near CR (27.6% vs 16.7%) was significantly higher with panobinostat compared to placebo. The addition of panobinostat prolonged the median DOR (13.14 vs 10.87 months), median PFS (11⋅99 vs 8⋅08 months), and median OS (33.6 vs 30.4 months). Serious AEs were higher in the panobinostat group (60% vs 42%). Common grade 3–4 AEs were thrombocytopenia, lymphopenia, diarrhea, asthenia, and peripheral neuropathy.Citation104 Recent subgroup analysis of PANORAMA 1 trial demonstrated a clear PFS benefit of 7.8 months for panobinostat–Btz–Dex among patients who received two or more prior regimens, including Btz and IMiD, a population with poorer prognosis and limited treatment options.Citation105

Collectively, the results of PANORAMA 1 and 2 show that the combination of panobinostat and Btz appears promising and has recently been approved by the FDA for the treatment of RRMM in patients with two prior treatments, including Btz and IMiDs.

Romidepsin

Romidepsin (FR901228 or FK228) is a depsipeptide derived from the bacterium Chromobacterium violaceum with activity mainly against class I HDAC. It was approved by the FDA for the treatment of relapsed cutaneous T-cell lymphoma in 2009.Citation106

A Phase II study evaluated the activity of romidepsin in heavily pretreated patients with MM who were refractory to therapies, including ASCT, Btz, and IMiDs. Although no objective responses were achieved, ~30% of patients exhibited stabilization of M-protein, resolution of hypercalcemia, and improvement in bone pain. The most common AEs were grade 1/2 and included nausea, fatigue, taste alteration, and clinically insignificant electrocardiographic abnormalities.Citation107

A Phase II trial used romidepsin with Btz and dexamethasone based on preclinical synergy. The incidence of grade 3 anemia and neutropenia was similar to that reported in previous trials using Btz–dexamethasone. PR was seen in 52% (VGPR in 28%) and CR was seen in 8% of the 25 patients enrolled. The median time to progression was 7.2 months, and the median OS was > 36 months.Citation108

A Phase I/II trial is evaluating the combination of romidepsin and Len in patients with relapsed/refractory lymphoma and myeloma. The study is ongoing, but the Phase I results suggest that the combination is well tolerated up to standard single-agent doses of each drug.Citation109

ACY-1215

ACY-1215 is an oral small molecule targeted against HDAC6. In view of responses seen in xenograft severe combined immunodeficiency mouse models,Citation60 a Phase I trial is evaluating ACY-1215 alone (part 1, Phase Ia) and in combination with Btz (part 2, Phase Ib) in patients with RRMM after at least two lines of treatment. In Phase Ia, no maximal tolerated dose was identified and AEs reported were elevated creatinine, fatigue, hypercalcemia, and upper respiratory tract infection (not attributed to ACY-1215). In Phase Ib, grade 3 or 4 gastrointestinal AEs were rare and hematologic AEs were manageable. The ORR was 25%, and the CBR was 60% in this heavily pretreated patient population.Citation110

Another ongoing trial is exploring the combination of ACY-1215 with Len/dexamethasone. ACY-1215 is found to be well tolerated, and no dose-limiting toxicity has been observed so far. The most common AEs, mainly grades 1/2, were fatigue, upper respiratory tract infections, and neutropenia. At the interim analysis, the ORR was 81%, including one CR and three VGPR.Citation111

Belinostat

Belinostat (PXD101) is a nonselective HDACi of hydroxamic acid class. A Phase II study enrolled 24 patients with RRMM who received belinostat as monotherapy and in combination with high dose of dexamethasone. This treatment was well tolerated, with minimal side effects, obtaining one MR and five SD.Citation112

Givinostat

Givinostat (ITF2357) is an orally active HDACi. In a Phase II trial, givinostat (alone or combined with dexamethasone) proved tolerable but showed only a modest clinical benefit. Only five of the 19 patients with advanced MM achieved SD. All patients experienced grade 3/4 thrombocytopenia, three had grade 3/4 gastrointestinal toxicity, and three had transient electrocardiographic abnormalities.Citation113

Conclusion

Epigenetic aberrations have now been recognized to contribute to the development and progression of various types of cancer, including MM. HDACi regulate the acetylation status of various histone and nonhistone proteins required for cellular processes, including gene expression, protein recycling, cell proliferation, and apoptosis, that are important for myeloma cell growth and survival. Preclinical evidence from studies of HDACi, alone or in combination with other antimyeloma agents, provides a strong scientific rationale for the evaluation of these regimens in the clinical setting. Results from early-stage clinical trials demonstrate that though HDACi show only modest activity as single agent, using them in combination with other anti-MM agents, especially Btz, show significant clinical responses. It must be noted that most of these trials were performed in patients relapsed on or refractory to Btz, and perhaps their utilization earlier in therapy, likely in combination with Btz, would be more effective. Hence, their precise role in the armamentarium of therapy for MM is yet to be defined. In addition, isoform-selective and/or class-selective HDAC inhibition needs further evaluation to reduce unfavorable side effects.

Disclosure

SKK has received research support from Novartis for clinical trials. The authors report no other conflicts of interest in this work.

References

  • KyleRARajkumarSVMultiple myelomaN Engl J Med2004351181860187315509819
  • National Cancer Institute [webpage on the Internet]SEER Stat Fact Sheets: Myeloma2016 Available from: http://seer.cancer.gov/statfacts/html/mulmy.htmlAccessed March 7, 2016
  • KristinssonSYLandgrenODickmanPWDerolfARBjorkholmMPatterns of survival in multiple myeloma: a population-based study of patients diagnosed in Sweden from 1973 to 2003J Clin Oncol200725151993199917420512
  • KumarSKRajkumarSVDispenzieriAImproved survival in multiple myeloma and the impact of novel therapiesBlood20081112516252017975015
  • KumarSKLeeJHLahuertaJJRisk of progression and survival in multiple myeloma relapsing after therapy with IMiDs and bortezomib: a multicenter international myeloma working group studyLeukemia20122614915721799510
  • SharmaAHeuckCJFazzariMJDNA methylation alterations in multiple myeloma as a model for epigenetic changes in cancerWiley Interdiscip Rev Syst Biol Med2010265466920890963
  • ChapmanMALawrenceMSKeatsJJInitial genome sequencing and analysis of multiple myelomaNature201147146747221430775
  • DimopoulosKGimsingPGronbaekKThe role of epigenetics in the biology of multiple myelomaBlood Cancer J20144e20724786391
  • DimopoulosKGimsingPGrønbaekKAberrant microRNA expression in multiple myelomaEur J Haematol2013919510523586898
  • BentleyGAFinchJTLewit-BentleyARothMThe crystal structure of the nucleosome core particle by contrast variationBasic Life Sci1984271051176712563
  • MoniotSWeyandMSteegbornCStructures, substrates, and regulators of mammalian sirtuins – opportunities and challenges for drug developmentFront Pharmacol201231622363286
  • KhanOLa ThangueNBDrug insight: histone deacetylase inhibitor-based therapies for cutaneous T-cell lymphomasNat Clin Pract Oncol2008571472618839006
  • KhanOLa ThangueNBHDAC inhibitors in cancer biology: emerging mechanisms and clinical applicationsImmunol Cell Biol201290859422124371
  • WittODeubzerHEMildeTOehmeIHDAC family: what are the cancer relevant targets?Cancer Lett200927782118824292
  • GregorettiIVLeeYMGoodsonHVMolecular evolution of the histone deacetylase family: functional implications of phylogenetic analysisJ Mol Biol2004338173115050820
  • LiuTLiuPYMarshallGMThe critical role of the class III histone deacetylase SIRT1 in cancerCancer Res2009691702170519244112
  • JonesPABaylinSBThe fundamental role of epigenetic events in cancerNat Rev Genet2002341542812042769
  • SmithEMBoydKDaviesFEThe potential role of epigenetic therapy in multiple myelomaBr J Haematol200914870271319912222
  • MaesKMenuEValckenborghEVRietIVVanderkerkenKBruynEDEpigenetic modulating agents as a new therapeutic approach in multiple myelomaCancers2013543046124216985
  • BollatiVFabrisSPegoraroVDifferential repetitive DNA methylation in multiple myeloma molecular subgroupsCarcinogenesis2009301330133519531770
  • AokiYNojimaMSuzukiHGenomic vulnerability to LINE-1 hypomethylation is a potential determinant of the clinicogenetic features of multiple myelomaGenome Med2012410123259664
  • WalkerBAWardellCPChiecchioLAberrant global methylation patterns affect the molecular pathogenesis and prognosis of multiple myelomaBlood201111755356220944071
  • Martinez-GarciaEPopovicRMinDJThe MMSET histone methyl transferase switches global histone methylation and alters gene expression in t(4;14) multiple myeloma cellsBlood201111721122020974671
  • BritoJLWalkerBJennerMMMSET deregulation affects cell cycle progression and adhesion regulons in t(4;14) myeloma plasma cellsHaematologica200994788619059936
  • WeichertWHDAC expression and clinical prognosis in human malignanciesCancer Lett2009280216817619103471
  • MarquardLGjerdrumLMChristensenIJJensenPBSehestedMRalfkiaerEPrognostic significance of the therapeutic targets histone deacetylase 1, 2, 6 and acetylated histone H4 in cutaneous T-cell lymphomaHistopathology200853326727718671804
  • SkovVLarsenTSThomassenMIncreased gene expression of histone deacetylases in patients with Philadelphia-negative chronic myeloproliferative neoplasmsLeuk Lymphoma201253112312921806350
  • WangJCChenCDumlaoTEnhanced histone deacetylase enzyme activity in primary myelofibrosisLeuk Lymphoma200849122321232719052980
  • MarquardLPoulsenCBGjerdrumLMHistone deacetylase 1, 2, 6 and acetylated histone H4 in B- and T-cell lymphomasHistopathology200954668869819438744
  • WeichertWRöskeAGekelerVAssociation of patterns of class I histone deacetylase expression with patient prognosis in gastric cancer: a retrospective analysisLancet Oncol20089213914818207460
  • WeichertWRöskeANiesporekSClass I histone deacetylase expression has independent prognostic impact in human colorectal cancer: specific role of class I histone deacetylases in vitro and in vivoClin Cancer Res20081461669167718347167
  • WeichertWDenkertCNoskeAExpression of class I histone deacetylases indicates poor prognosis in endometrioid subtypes of ovarian and endometrial carcinomasNeoplasia20081091021102718714364
  • ZhangZYamashitaHToyamaTQuantitation of HDAC1 mRNA expression in invasive carcinoma of the breastBreast Cancer Res Treat2005941111616172792
  • MorenoDAScrideliCACortezMADifferential expression of HDAC3, HDAC7 and HDAC9 is associated with prognosis and survival in childhood acute lymphoblastic leukaemiaBr J Haematol2010150666567320636436
  • Van DammeMCrompotEMeulemanNHDAC isoenzyme expression is deregulated in chronic lymphocytic leukemia B-cells and has a complex prognostic significanceEpigenetics20127121403141223108383
  • MithraprabhuSKalffAChowAKhongTSpencerADysregulated class I histone deacetylases are indicators of poor prognosis in multiple myelomaEpigenetics20149111511152025482492
  • HellerGSchmidtWMZieglerBGenome-wide transcriptional response to 5-aza-2′-deoxycytidine and trichostatin a in multiple myeloma cellsCancer Res200868445418172295
  • MitsiadesCSMitsiadesNSMcMullanCJTranscriptional signature of histone deacetylase inhibition in multiple myeloma: biological and clinical implicationsProc Natl Acad Sci U S A200410154054514695887
  • NeriPTagliaferriPdi MartinoMTIn vivo anti-myeloma activity and modulation of gene expression profile induced by valproic acid, a histone deacetylase inhibitorBr J Haematol200814352053118986388
  • Van LintCEmilianiSVerdinEThe expression of a small fraction of cellular genes is changed in response to histone hyperacetylationGene Expr199652452538723390
  • GraySGQianCNFurgeKGuoXTehBTMicroarray profiling of the effects of histone deacetylase inhibitors on gene expression in cancer cell linesInt J Oncol20042477379515010814
  • PeartMJSmythGKvan LaarRKIdentification and functional significance of genes regulated by structurally different histone deacetylase inhibitorsProc Natl Acad Sci USA20051023697370215738394
  • KaiserMZavrskiISterzJThe effects of the histone deacetylase inhibitor valproic acid on cell cycle, growth suppression and apoptosis in multiple myelomaHaematologica20069124825116461312
  • KhanSBMaududiTBartonKAyersJAlkanSAnalysis of histone deacetylase inhibitor, depsipeptide (FR901228), effect on multiple myelomaBr J Haematol200412515616115059137
  • GolayJCuppiniLLeoniFThe histone deacetylase inhibitor ITF2357 has anti-leukemic activity in vitro and in vivo and inhibits IL-6 and VEGF production by stromal cellsLeukemia2007211892190017637810
  • CatleyLWeisbergETaiYTNVPLAQ824 is a potent novel histone deacetylase inhibitor with significant activity against multiple myelomaBlood20031022615262212816865
  • FengRMaHHassigCAKD5170, a novel mercaptoketone-based histone deacetylase inhibitor, exerts antimyeloma effects by DNA damage and mitochondrial signalingMol Cancer Ther200871494150518566220
  • CatleyLWeisbergEKiziltepeTAggresome induction by proteasome inhibitor bortezomib and alpha-tubulin hyperacetylation by tubulin deacetylase (TDAC) inhibitor LBH589 are synergistic in myeloma cellsBlood20061083441344916728695
  • LavelleDChenYHHankewychMDe-SimoneJHistone deacetylase inhibitors increase p21(WAF1) and induce apoptosis of human myeloma cell lines independent of decreased IL-6 receptor expressionAm J Hematol20016817017811754398
  • MitsiadesNMitsiadesCSRichardsonPGMolecular sequelae of histone deacetylase inhibition in human malignant B cellsBlood20031014055406212531799
  • MaisoPCarvajal-VergaraXOcioEMThe histone deacetylase inhibitor LBH589 is a potent antimyeloma agent that overcomes drug resistanceCancer Res2006665781578916740717
  • DeleuSLemaireMArtsJThe effects of JNJ-26481585, a novel hydroxamate-based histone deacetylase inhibitor, on the development of multiple myeloma in the 5T2MM and 5T33MM murine modelsLeukemia2009231894190319494837
  • BaiLYOmarHAChiuCFChiZPHuJLWengJRAntitumor effects of (S)-HDAC42, a phenylbutyrate-derived histone deacetylase inhibitor, in multiple myeloma cellsCancer Chemother Pharmacol201035373379
  • SchwartzCPalissotVAoualiNValproic acid induces non-apoptotic cell death mechanisms in multiple myeloma cell linesInt J Oncol20073057358217273758
  • RosatoRRAlmenaraJAMaggioSCRole of histone deacetylase inhibitor-induced reactive oxygen species and DNA damage in LAQ-824/fludarabine antileukemic interactionsMol Cancer Ther20087103285329718852132
  • ChenCSWangYCYangHCHistone deacetylase inhibitors sensitize prostate cancer cells to agents that produce DNA double-strand breaks by targeting Ku70 acetylationCancer Res200767115318532717545612
  • FengROtonAMaparaMYAndersonGBelaniCLentzschSThe histone deacetylase inhibitor, PXD101, potentiates bortezomib-induced anti-multiple myeloma effect by induction of oxidative stress and DNA damageBr J Haematol200713938539717910628
  • LeeCKWangSHuangXRyderJLiuBHDAC inhibition synergistically enhances alkylator-induced DNA damage responses and apoptosis in multiple myeloma cellsCancer Lett201029623324020447761
  • BakkenistCJKastanMBDNA damage activates ATM through intermolecular autophosphorylation and dimer dissociationNature2003421692249950612556884
  • Quintas-CardamaASantosFPGarcia-ManeroGHistone deacetylase inhibitors for the treatment of myelodysplastic syndrome and acute myeloid leukemiaLeukemia201125222623521116282
  • PeiXYDaiYGrantSSynergistic induction of oxidative injury and apoptosis in human multiple myeloma cells by the proteasome inhibitor bortezomib and histone deacetylase inhibitorsClin Cancer Res2004103839385215173093
  • DeleuSLemaireMArtsJBortezomib alone or in combination with the histone deacetylase inhibitor JNJ-26481585: effect on myeloma bone disease in the 5T2MM murine model of myelomaCancer Res2009695307531119531653
  • MaesKMenuEVan ValckenborghEVan RietIVanderkerkenKDe BruyneEEpigenetic modulating agents as a new therapeutic approach in multiple myelomaCancers (Basel)20135243046124216985
  • LemaireMDeleuSde BruyneEvan ValckenborghEMenuEVanderkerkenKThe microenvironment and molecular biology of the multiple myeloma tumorAdv Cancer Res2012110194221704227
  • QianDZWangXKachhapSKThe histone deacetylase inhibitor NVP-LAQ824 inhibits angiogenesis and has a greater antitumor effect in combination with the vascular endothelial growth factor receptor tyrosine kinase inhibitor PTK787/ZK222584Cancer Res200464186626663415374977
  • EllisLHammersHPiliRTargeting tumor angiogenesis with histone deacetylase inhibitorsCancer Lett2009280214515319111391
  • LiuTKuljacaSTeeAMarshallGMHistone deacetylase inhibitors: multifunctional anticancer agentsCancer Treat Rev200632315716516516391
  • DongXFSongQLiLZZhaoCLWangLQHistone deacetylase inhibitor valproic acid inhibits proliferation and induces apoptosis in KM3 cells via downregulating VEGF receptorNeuro Endocrinol Lett20072877578018063935
  • KitazoeKAbeMHiasaMValproic acid exerts anti-tumor as well as antiangiogenic effects on myelomaInt J Hematol200989455719093163
  • DavenportELMooreHEDunlopASHeat shock protein inhibition is associated with activation of the unfolded protein response pathway in myeloma plasma cellsBlood20071102641264917525289
  • BaliPPranpatMBradnerJInhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90J Biol Chem20052802672934
  • RichonVMGarcia-VargasJHardwickJSDevelopment of vorinostat: current applications and future perspectives for cancer therapyCancer Lett200928020121019181442
  • HideshimaTBradnerJEWongJSmall-molecule inhibition of proteasome and aggresome function induces synergistic antitumor activity in multiple myelomaProc Natl Acad Sci U S A20051028567857215937109
  • SantoLHideshimaTKungALPreclinical activity, pharmacodynamic, and pharmacokinetic properties of a selective HDAC6 inhibitor, ACY-1215, in combination with bortezomib in multiple myelomaBlood20121192579258922262760
  • KikuchiJWadaTShimizuRHistone deacetylases are critical targets of bortezomib-induced cytotoxicity in multiple myelomaBlood201011640641720351311
  • OcioEMVilanovaDAtadjaPIn vitro and in vivo rationale for the triple combination of panobinostat (LBH589) and dexamethasone with either bortezomib or lenalidomide in multiple myelomaHaematologica20109579480319951978
  • CampbellRASanchezESteinbergJVorinostat enhances the antimyeloma effects of melphalan and bortezomibEur J Haematol20108420121119929977
  • DuHLRenLMChenHZhuYQiYRe-expression of p16 gene in the myeloma cell line U266 induced by synergy of sodium butyrate and 5-Aza-2′-deoxycytidineDi Yi Jun Yi Da Xue Xue Bao20022298198412433622
  • RamakrishnanVKimlingerTTimmMHaugJRajkumarSVKumarSMultiple mechanisms contribute to the synergistic anti-myeloma activity of the pan-histone deacetylase inhibitor LBH589 and the rapalog RAD001Leuk Res2014381358136625282334
  • ShimuraYKurodaJRiMRSK2(Ser227) at N-terminal kinase domain is a potential therapeutic target for multiple myelomaMol Cancer Ther201211122600260923012246
  • KaiserMLamottkeBMiethMSynergistic action of the novel HSP90 inhibitor NVP-AUY922 with histone deacetylase inhibitors, melphalan, or doxorubicin in multiple myelomaEur J Haematol20108433734420028416
  • ChenSDaiYPeiXYGrantSBim upregulation by histone deacetylase inhibitors mediates interactions with the Bcl-2 antagonist ABT-737: evidence for distinct roles for Bcl-2, Bcl-xL, and Mcl-1Mol Cell Biol2009296149616919805519
  • FandyTEShankarSRossDDSausvilleESrivastavaRKInteractive effects of HDAC inhibitors and TRAIL on apoptosis are associated with changes in mitochondrial functions and expressions of cell cycle regulatory genes in multiple myelomaNeoplasia20057764665716026644
  • MagnerWJKazimALStewartCActivation of MHC class I, II, and CD40 gene expression by histone deacetylase inhibitorsJ Immunol2000165127017702411120829
  • SkovSPedersenMTAndresenLStratenPTWoetmannAOdumNCancer cells become susceptible to natural killer cell killing after exposure to histone deacetylase inhibitors due to glycogen synthase kinase-3-dependent expression of MHC class I-related chain A and BCancer Res20056523111361114516322264
  • KhanANGregorieCJTomasiTBHistone deacetylase inhibitors induce TAP, LMP, Tapasin genes and MHC class I antigen presentation by melanoma cellsCancer Immunol Immunother200857564765418046553
  • ManningJIndrovaMLubyovaBInduction of MHC class I molecule cell surface expression and epigenetic activation of antigen-processing machinery components in a murine model for human papilloma virus 16-associated tumoursImmunology2008123221822717725605
  • ChristiansenAJWestABanksKMEradication of solid tumors using histone deacetylase inhibitors combined with immune-stimulating antibodiesProc Natl Acad Sci U S A2011108104141414621368108
  • WestACMattarolloSRShorttJAn intact immune system is required for the anticancer activities of histone deacetylase inhibitorsCancer Res201373247265727624158093
  • SonnemannJGressmannSBeckerSWittigSSchmuddeMBeckJFThe histone deacetylase inhibitor vorinostat induces calreticulin exposure in childhood brain tumour cells in vitroCancer Chemother Pharmacol201066361161620221600
  • KroesenMGielenPBrokICArmandariIHoogerbruggePMAdemaGJHDAC inhibitors and immunotherapy; a double edged sword?Oncotarget20145166558657225115382
  • VoDDPrinsRMBegleyJLEnhanced antitumor activity induced by adoptive T-cell transfer and adjunctive use of the histone deacetylase inhibitor LAQ824Cancer Res200969228693869919861533
  • SongWTaiYTTianZHDAC inhibition by LBH589 affects the phenotype and function of human myeloid dendritic cellsLeukemia201125116116821102427
  • MannBSJohnsonJRCohenMHJusticeRPazdurRFDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphomaOncologist200712101247125217962618
  • RichardsonPMitsiadesCColsonKPhase I trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) in patients with advanced multiple myelomaLeuk Lymphoma200849350250718297527
  • BadrosABurgerAMPhilipSPhase I study of vorinostat in combination with bortezomib for relapsed and refractory multiple myelomaClin Cancer Res200915165250525719671864
  • SiegelDSDimopoulosMAYoonS-SVantage 095: Vorinostat in Combination with Bortezomib in Salvage Multiple Myeloma Patients: Final Study Results of a Global Phase 2b TrialASHSan Diego2011
  • DimopoulosMASiegelDSLonialSVorinostat or placebo in combination with bortezomib in patients with multiple myeloma (VANTAGE 088): a multicentre, randomised, double-blind studyLancet Oncol201314111129114024055414
  • AlsayedYNairBPKauffmanMCarfilzomib (CFZ) in combination with DEX and other agents (Dox, Thal, DDP, vorinostat) for far advanced and refractory multiple myeloma (FARMM)J Clin Oncol201028e18504 ASCO Annual Meeting Abstracts
  • SiegelDSRichardsonPDimopoulosMVorinostat in combination with lenalidomide and dexamethasone in patients with relapsed or refractory multiple myelomaBlood Cancer J20144e18224562384
  • WolfLWSiegelDMatousJA phase II study of oral panobinostat (LBH589) in adult patients with advanced refractory multiple myelomaBlood200811112 ASH Annual Meeting Abstracts
  • San-MiguelJFRichardsonPGGSezerOA phase lb study of oral panobinostat and IV bortezomib in relapsed or relapsed and refractory multiple myelomaJ Clin Oncol20112915 abstr8075. ASCO Annual Meeting Proceedings
  • RichardsonPGSchlossmanRLAlsinaMPANORAMA 2: panobinostat in combination with bortezomib and dexamethasone in patients with relapsed and bortezomib-refractory myelomaBlood20131222331233723950178
  • San-MiguelJFHungriaVTMYoonSSPanobinostat 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 trialLancet Oncol2014151195120625242045
  • RichardsonPGHungriaVTMYoonSSPanobinostat plus bortezomib and dexamethasone in relapsed/relapsed and refractory myeloma: outcomes by prior treatmentBlood2016127671372126631116
  • BertinoaEMOttersonaGARomidepsin: a novel histone deacetylase inhibitor for cancerExpert Opin Investig Drugs201120811511158
  • NiesvizkyRElySMarkTPhase 2 trial of the histone deacetylase inhibitor romidepsin for the treatment of refractory multiple myelomaCancer2011117233634220862746
  • HarrisonSJQuachHLinkEA high rate of durable responses with romidepsin, bortezomib, and dexamethasone in relapsed or refractory multiple myelomaBlood2011118246274628321911830
  • LunningMARuanJNairSA phase I/II trial of the combination of romidepsin and lenalidomide in patients with relapsed/refractory lymphoma and myeloma: phase I resultsJ Clin Oncol2014325 supplabstr8582
  • RajeNVoglDTHariPNACY-1215, a Selective Histone Deacetylase (HDAC) 6 Inhibitor: Interim Results Of Combination Therapy With Bortezomib In Patients With Multiple Myeloma (MM)ASH 2013 Annual Meeting Abstract 759 (Oral Presentation)New Orleans, LA2013
  • YeeAVorheesPBensingerWIACY-1215, a Selective Histone Deacetylase (HDAC) 6 Inhibitor, In Combination With Lenalidomide and Dexamethasone (dex), Is Well Tolerated Without Dose Limiting Toxicity (DLT) In Patients (Pts) With Multiple Myeloma (MM) At Doses Demonstrating Biologic Activity: Interim Results Of a Phase 1b TrialASH 2013 Annual Meeting Abstract 3190 (Poster Presentation)New Orleans, LA2013
  • SullivanDSSSchusterMBerensonJA phase II study of PXD 101 in advanced multiple myelomaBlood200611108 ASH Annual Meeting Abstracts
  • GalliMSalmoiraghiSGolayJA phase II multiple dose clinical trial of histone deacetylase inhibitor ITF2357 in patients with relapsed or progressive multiple myelomaAnn Hematol201089218519019633847