touchONCOLOGY touchONCOLOGY
Haematological Malignancies, Multiple Myeloma
Read Time: 14 mins

Epigenetics in the Biology and Treatment of Multiple Myeloma

Copy Link
Published Online: Nov 28th 2016 European Oncology & Haematology, 2016;12(2):96–102 DOI: https://touchoncology.com/epigenetics-in-the-biology-and-treatment-of-multiple-myeloma/
Authors: Andrew Spencer, Sridurga Mithraprabhu
Quick Links:
Abstract
Article
Article Information
Abstract:
Overview

There is a critical need for more effective therapies in multiple myeloma (MM) since all patients eventually relapse following front-line treatment. A variety of both genetic and epigenetic abnormalities may be present in MM, the latter including DNA and histone methylation and histone deacetylation, and are thought to contribute to the pathogenesis of the disease. For example, global methylation analysis in MM has identified inactivated tumour suppressor genes that are prognostically important. Through their ability to acetylate histones and cytoplasmic proteins, histone deacetylases (HDAC) influence a wide variety of cellular functions, such as proliferation, differentiation and apoptosis. Increased class 1 HDAC expression has been linked in solid tumours with more locally advanced, de-differentiated and proliferative tumours, and with poor prognosis in MM. HDAC inhibitors, panobinostat and ricolinostat, have been demonstrated to be effective in combination with bortezomib and dexamethasone in newly diagnosed patients with MM and in heavily pre-treated patients with advanced MM. HDAC inhibitor–monoclonal antibody combinations are also being explored. The potential of HDAC inhibitors to improve outcome for patients with MM is evident but a greater understanding of their anti-tumour effects is needed.

Keywords

Multiple myeloma (MM), epigenetics, DNA methylation, histone modifications, histone deacetylases inhibitors, panobinostat, ricolinostat, combination strategies

Article:

Epigenetics encompasses heritable changes in the pattern of gene expression mediated by mechanisms other than alterations in primary nucleotide sequence. The epigenome is an inheritable record of changes to the DNA and histone proteins, such as methylation and nucleosome remodelling that directs which genes are to be silenced or expressed. Following early studies of abnormal gene expression in cancer, epigenetic modifications have been recognised to be of central importance in cancer pathophysiology.1 Mutations in regulators of the epigenome have been identified in cancer that have roles such as ‘writers’, ‘readers’, ‘erasers’ or ‘editors’ and have the potential to deregulate the expression of hundreds of genes genome-wide.2

Multiple myeloma (MM), a clonal expansion of plasma cells, is characterised by monoclonal protein production, end-organ damage and marked clinical and genetic heterogeneity.3–5 MM is the second most common haematological malignancy6 and accounts for approximately 20% of deaths from haematological malignancies7 and 0.8% of deaths from all cancers;8,9 in addition, MM is associated with high costs in healthcare provision.10 The median age of MM diagnosis is 74 years.11 In Europe, between 2005 and 2009, there was an over two-fold difference between the highest male cancer mortality in Hungary (235.2/100 000) and the lowest mortality in Sweden (112.9/100 000), and a 1.7-fold one in women (from 124.4 in Denmark to 71.0/100 000 in Spain).12Over the past decade, however, survival of patients with MM has improved dramatically with the advent of newer therapies.13

MM always follows the premalignant state of monoclonal gammopathy of undetermined significance (MGUS), although the precise molecular mechanisms involved in the progression from MGUS to MM are not properly understood. Various distinct genetic abnormalities have been reported in both MM and MGUS including epigenetic alterations such as DNA and histone methylation, and are known to contribute to the pathogenesis of the disease.14–17 Front-line treatment of MM can result in high response rates,18–21 although all patients eventually relapse and more effective therapies are needed. MM is heterogeneous and is associated with complex gene abnormalities and multiple signalling aberrations. A strategy of targeting single signalling pathways, genes or individual gene products may not therefore be inadequate to supress MM cell growth.22 The aim of this review is to describe epigenetic modifications and therapeutic combination strategies in MM.

Mb>DNA methylation
DNA methylation, which is the best characterised epigenetic modification, occurs in cytosinecontaining nucleotides that are immediately followed by nucleotide sequences containing guanine (i.e., cytosine-phosphodiester bond-guanine [CpG] islands). Located in 60% of promoters, CpG islands, when methylated usually results in silencing of tumour suppressor genes.23 These events are more frequent than tumour suppressor gene mutations. The CpG island methylator phenotype (CIMP), was first identified in colorectal cancer24 and has been observed extensively in a wide variety of tumours,25 although CIMP has rarely been reported in MM.26 MM is however characterised by a level of heterogeneity in DNA methylation that exceeds that described in several solid cancers.27,28

The DNA methylome, which is the map of DNA methylation modifications, has been described in normal plasma cells (NPCs) and plasma cells from MGUS and MM patient samples (Figure 1).29 Aberrant methylation patterns are related to disease stage in MM with significant difference and heterogeneity in the global methylation patterns in the malignant cell types (MM, primary plasma cell leukaemia and in human myeloma cell lines) compared to the non-malignant cells (non-plasma cell disorders and MGUS).17 DNA methylation at transcription start sites and gene promoters has been associated with the formation of heterochromatin and long-term gene silencing.30,31 Methylation is also an inactivating mechanism of the tumour suppressor p16 gene in MM, which is associated with high plasma cell proliferation and short survival.32 Furthermore, p16 methylation may be a marker for overall epigenetic changes associated with disease progression.33 Global methylation analysis in MM has pinpointed inactivated tumour suppressor genes that are prognostically important.28 In an analysis of the association of differential DNA methylation with prognosis in 159 patients with myeloma, 195 genes with changes in DNA methylation were identified. Hypermethylation of the genes GPX3, RBP1, SPARC and TGFBI was associated with significantly shorter overall survival (OS); this association was independent of age, International Staging System score, and adverse cytogenetics.28

The most comprehensive methylation study to date has demonstrated that B-cell specific intronic enhancers undergo DNA hyper-methylation, resulting in enhanced decommissioning of genes related to B-cell differentiation consistent with MM acquiring or preserving a ‘stem cell-like’ methylome.30 Finally, the expression of DNA (cytosine-5-)- methyltransferase 1 (DNMT1) is also known to be upregulated during disease pathogenesis compared to NPCs and silencing of DNMT1 significantly reduced the methylation of p16.34,35

MicroRNA are a class of short non-coding RNA molecules with RNA silencing functions.36 Epigenetic dysregulation of tumour-suppressor microRNA genes via DNA methylation has also been implicated in MM.37

Histone modifications
Histone modifications, including methylation, de/acetylation, phosphorylation, ubiquitination, adenosine diphosphate (ADP)-ribosylation and isomerisation, modulate gene expression by altering chromatin structure or recruiting histone modifiers.38 MMSET domain (MMSET) is a histone methyltransferase that is overexpressed in t(4;14)+ MM patients and is considered a key driving factor in the pathogenesis of this subtype of MM. MMSET catalyses dimethylation of lysine 36 on histone H3 (H3K36me2), and its overexpression causes a global increase in H3K36me2 and a decrease in lysine 27 methylation, resulting in gene expression changes promoting disease progression and tumoregenicity.39–43 Conversely, suppression of MMSET affects MM cell survival through regulation of genes associated with cell cycle progression, apopotosis and cell adhesion.44

Histone deacetylases (HDAC) are a highly conserved group of enzymes, currently grouped into four classes, that influence a wide variety of cellular functions via their ability to acetylate both histones and cytoplasmic proteins.45 These functions include proliferation, differentiation and apoptosis. Moreover, overexpression of Class I HDAC expression has

been associated, in most but not all cases, with locally advanced, dedifferentiated, proliferative tumours46 and with poor prognosis in MM.47 In contrast, expression of Class II HDACs has been found to be reduced in tumours and high expression of these isoforms has been linked in some instances to improved patient outcome in a range of both solid and haematological malignancies highlighting the contextual impact of HDAC dysregulation in cancer.46 In addition, the Class I specific HDAC inhibitor romidepsin has been shown to induce histone acetylation by promoting Caspase 8-mediated destruction of a critical Class I HDAC transcription factor Sp1.48 The combination of bortezomib and romidepsin thus promoting histone hyper-acetylation coincident with the putative disruption of protein homeostasis that such a combination would also induce (see Figure 2).

Therapeutic strategies in multiple myeloma
DNA and histone methylation
During cell division, DNMT1 maintains the DNA methylation pattern of the newly synthesised daughter strand, whereas the histone–lysine methyltransferase G9a (EHMT2) methylates histone H3 Lys9, with direct interaction between DNMT1 and G9a known to coordinate DNA- and histone-methylation during replication.49 Dual and reversible inhibitors are thus being developed to target G9a and DNMT1. Currently available DNMT inhibitors, 5-azacytidine and 5-aza-2’deoxycytidine have been evaluated pre-clinically in both in vitro and in vivo studies against MM and the DNA methylation score was shown to be predictive of MM cell sensitivity to 5-azacitidine.50,51 Oral 5-azacitidine in combination with lenalidomide and dexamethasone has been demonstrated to be effective and well tolerated in a group of heavily pre-treated patients with relapsed and/or refractory MM (RRMM) refractory to prior lenalidomide therapy.52 The Histone methyltransferase MMSET is known to be upregulated in t(4;14) patients and silencing of MMSET results in anti-MM activity,41,44,53 and while MMSET is currently not targetable, histone-methylation modifying drugs are under development.

HDAC inhibition
In vitro evaluation of a range of HDAC inhibitors (HDACi) has demonstrated that Class I HDAC inhibition is required by HDACi to induce maximal MM cell death.54 Major HDACi can be divided in to five categories on the basis of their chemical structure (Table 1). 55 The single-agent pan-HDACi, panobinostat (LBH589), in an interim analysis of a Phase II study was shown to induce deeper responses following high-dose chemotherapy conditioned autologous stem cell transplantation as demonstrated by both the time taken to achieve best response (Figure 3A) and response status according to duration of treatment with panobinostat (Figure 3B).56 Panobinostat has recently been approved by the US Food and Drugs Association, and European Union marketing authorisation was granted on 28 August 2015 for use in patients with MM who had received ≥2 prior therapeutic regimens that included bortezomib and an immunomodulatory drug. To avoid side effects associated with broad HDAC inhibition, class- and/or isoform-selective HDACi have been developed such as ricolinostat (ACY-1215) a selective HDAC6 inhibitor, which, coupled with bortezomib, as discussed below, targets ubitquinated protein degradation in proteasomes and lysosomes

Combination strategies
Histone deacetylase inhibition and bortezomib
Recent studies have demonstrated an alternative system to the proteasome for degradation of polyubiquitinated misfolded and/ or unfolded proteins: the aggresome (Figure 4).57,58 HDAC6 has an essential role in a variety of cellular processes, including aggresomal protein degradation.59 Selective inhibition of HDAC6 leads to increased acetylation of its substrate, tubulin, and inhibition of the proteasome pathway, resulting in accumulation of polyubiquitinated proteins and apoptosis in MM cells.60 Inhibition of both proteasomal and aggresomal protein degradation systems by bortezomib and tubacin (a HDAC6 inhibitor), respectively, have been shown to induce synergistic cytotoxicity in MM cell lines and freshly isolated bone marrow plasma cells (BMPCs) from MM patients.60 This finding provides the rationale for clinical protocols of this combination to improve patient outcome in MM. Further, the synergistic activity of HDACi and proteasome inhibitors has been demonstrated in two mouse models that were created via subcutaneously injected human MM and intravenously injected luciferase-expressing human MM.61 Tumour growth was delayed significantly and OS was significantly prolonged in mice treated with the combination therapy. Ricolinostat (ACY-1215) is a HDAC6 inhibitor. In a phase Ib study, the ricolinostat– bortezomib–dexamethasone combination induced a treatment response in a group of heavily pre-treated patients with RRMM and had a manageable tolerability profile.62 Forty-two patients were enrolled to eight combination cohorts: the first cohort received ricolinostat 40 mg once daily (QD) with bortezomib 1.0 mg/m2; subsequent cohorts were treated with bortezomib 1.3 mg/m2 with ricolinostat doses of 40, 80, 160 and 240 mg QD, and 160 mg twice daily (BID). Ricolinostat was administered orally on days 1–5 and 8–12 of a 21- day cycle, with bortezomib on days 1, 4, 8, 11 and dexamethasone on days 1, 2, 4, 5, 8, 9, 11 and 12. Among the 25 patients evaluable for response, the overall response rate was 44%: two patients had a very good partial response, nine had a partial response, and two had a

minimal response, giving a clinical benefit rate of 52%. Diarrhoea was the only dose-related adverse event.

The HDACi, vorinostat (suberoylanilide hydroxamic acid, Zolinza®, Merck, New Jersey, US), in combination with bortezomib, showed limited clinical benefit in the multicentre, randomised, double-blind study, VANTAGE 088.63 Patients were allocated randomly to receive 21-day cycles of bortezomib (1.3 mg/m2 intravenously on days 1, 4, 8, and 11) in combination with oral vorinostat (400 mg; n=315) or matching placebo (n=320) QD on days 1–14. Median progression-free survival (PFS) was 7.63 months (95% confidence interval [CI] 6.9–8.4) for vorinostat + bortezomib and 6.83 (95% CI 5.7–7.7) for placebo + bortezomib with a hazard ratio (HR) of 0.774 (0.64–0.94) p=0.01. The most common grade 3–4 adverse events were thrombocytopenia (143 [45%] patients in the vorinostat group versus 77 [24%] patients in the placebo group), neutropenia (89 [28%] versus 80 [25%]), and anaemia (53 [17%] versus 40 [13%]). In total, 312 (99%) of 315 patients in the vorinostat group and 315 (98%) of 320 patients in the placebo group had adverse events.

VANTAGE 095, an open-label, single-arm, multicentre, phase IIb study, was designed to investigate the efficacy and tolerability of oral vorinostat with bortezomib in patients with MM that was considered refractory to novel myeloma agents.64 Patients received 21-day cycles of bortezomib (1.3 mg/m2 intravenously on days 1, 4, 8, and 11) and oral vorinostat (400 mg/d on days 1-14). The overall response rate (ORR) was 11.3% (95% confidence interval, 6.6%-17.7%), and the median duration of response was 211 days (range, 64-550 days); the median OS duration was 11.2 months (95% CI, 8.5-14.4 months), with a 2-year survival rate of 32%. Frequently reported adverse events included thrombocytopenia (69.7%), nausea (57.0%), diarrhoea (53.5%), anaemia (52.1%) and fatigue (48.6%).

PANORAMA 2 was a phase II trial of panobinostat in combination with bortezomib and dexamethasone to treat patients (n=55) with RRMM (with ≥2 prior lines of therapy, including an immunomodulatory drug, and patients who had progressed on or within 60 days of the last bortezomibbased therapy).65 Patients were treated in two phases: Phase I treatment consisted of 8 three-week cycles of oral panobinostat (20 mg) three times per week on weeks 1 and 2, bortezomib (1.3 mg/m2 intravenously) two times per week on weeks 1 and 2, and oral dexamethasone (20 mg) four times per week on weeks 1 and 2 on days of and after bortezomib use. Patients who showed clinical benefit in phase I treatment continued study therapy in phase II, which consisted of 6-week cycles of panobinostat three times per week on weeks 1, 2, 4 and 5; bortezomib once per week on weeks 1, 2, 4 and 5; and dexamethasone on the days of and after bortezomib until disease progression, death, toxicity or treatment

withdrawal (Figure 5). The efficacy outcomes (Figure 6) showed that panobinostat, when combined with bortezomib and dexamethasone, can recapture responses in heavily pre-treated, bortezomib-refractory MM patients.

PANORAMA 1 was a multicentre, randomised, placebo-controlled, doubleblind phase III trial of patients (n=768) with RRMM who had received between one and three prior lines of therapy (Figure 7).66 Patients were randomised to receive 21-day cycles of placebo or panobinostat (20 mg; on days 1, 3, 5, 8, 10, 12, orally), both in combination with bortezomib (1.3 mg/m2 on days 1, 4, 8 and 11, intravenously) and dexamethasone (20 mg on days 1, 2, 4, 5, 8, 9, 11, 12, orally). The primary endpoint of PFS was met (p<0.0001) with a clinically relevant increase in median PFS of 3.9 months for the panobinostat–bortezomib–dexamethasone arm. Serious adverse events (SAEs) were reported in 228 (60%) of 381 patients in the panobinostat–bortezomib–dexamethasone group and 157 (42%) of 377 patients in the placebo–bortezomib–dexamethasone group. Common grade 3–4 laboratory abnormalities and adverse events (irrespective of association with study drug) included thrombocytopenia (256 [67%] in the panobinostat group versus 118 [31%] in the placebo group), lymphopenia (202 [53%] versus 150 [40%]), diarrhoea (97 [26%] versus 30 [8%]), asthenia or fatigue (91 [24%] versus 45 [12%]), and peripheral neuropathy (67 [18%] versus 55 [15%]). Nearly half (48.4%) had received ≥2 prior lines of therapy. A post-hoc subgroup analysis revealed a relatively greater increase in PFS in heavily pre-treated patients who had received ≥2 prior lines of therapy and both prior bortezomib and an immunomodulatory drug with a median improvement in PFS associated with the use of panobinostat of 7.8 months.67

HDACi and immunomodulatory drug combinations
In a Phase II trial in RRMM (n=20), the all oral combination of panobinostat– lenalidomide–dexamethasone demonstrated durable responses, even in high risk lenalidomide refractory patients, indicating the possible capacity of panobinostat to overcome acquired drug resistance.68 The treatment regimen consisted of panobinostat 20 mg on days 1, 3,

5, 15, 17, 19 together with lenalidomide 25 mg qd on days 1–21 and dexamethasone 40 mg on days 1, 8, 15 in a 28-day cycle. The primary endpoint, ORR, was 45%. Common grade 3–4 toxicities were neutropenia (55%), thrombocytopenia (40%) and infection (20%) and common grade 3–4 diarrhoea occurred in 15%. One patient discontinued due to an adverse event (asymptomatic QTc prolongation).

Ricolinostat in combination with lenalidomide and dexamethasone also demonstrated efficacy in patients (n=25) with RRMM.69 In the first phase of the study, patients were treated with escalating doses of ricolinostat on days 1–5 and 8–12 of a 28-day cycle with lenalidomide 25 mg on days 1–21 and dexamethasone 40 mg weekly. In the second phase, ricolinostat was also given on days 15–19. Doses up to 240 mg QD and 160 mg BID were explored. The ORR was 64% and 50% in the full cohort and in a lenalidomide-refractory patient cohort, respectively. Adverse events were generally low grade and manageable; common grade 3–4 toxicities included neutropenia and fatigue. Two dose-limiting toxicities have been reported to date (grade 3 syncope and grade 3 muscle cramps).

Panobinostat and multidrug combinations
The combination of bortezomib–thalidomide–dexamethasone–panobinostat combination is being evaluated in an ongoing Phase I/II study (n=55).70 The ORR was 88.9% in the dose escalation phase and currently >75% for patients being treated at the recommended phase II dose (RP2D) (Figure 8). The regimen comprised bortezomib 1.3 mg/m2 subcutaneously days 1 and 8, thalidomide 100 mg/daily (50 mg if pre-existing neuropathy), dexamethasone 20 mg days 1, 2, 8, 9 and panobinostat days 1, 3, 5, 8, 10, 12) every 3 weeks. Grade 3–4 adverse events were primarily haematological: hypophosphatemia (16.7%), neutropenia (14.6%), abnormal liver tests (6.3%) (none observed at the non-recommended dose); thrombocytopenia (6.3%), peripheral neuropathy (2.1%), diarrhoea (2.1%) and hyperglycaemia (2.1%). The predominant grade 1–2 toxicities were: fatigue (64.5%), neuropathy (28, 58.3%), constipation (22, 45.8%) and diarrhoea (35.4%). Nineteen SAEs were reported by 12 patients at the RP2D, of which four were treatment related and predominantly gastrointestinal. The RP2D for panobinostat in combination with thalidomide, dexamethasone and bortezomib was 20 mg. Only one dose-limiting toxicity was observed; therefore, the maximum tolerated dose was not reached. The panobinostat–bortezomib–lenalidomide–dexamethasone combination has been investigated in patients (n=22) with newly diagnosed MM.71 The maximum tolerated dose without supportive care was determined to be: lenalidomide 25 mg; bortezomib 1.3 mg/m2; dexamethasone 20 mg and panobinostat 10 mg. This multidrug combination led to rapid, high-depth responses with a near complete response (CR)/CR rate of 50% and was very well tolerated with limited grade 3–4 adverse events. One patient experienced grade 4 thrombocytopenia, and a second patient had grade 3 diarrhoea without supportive measures, for < 12 hours that subsequently resolved with supportive measures. This is the first trial that has combined panobinostat with subcutaneous bortezomib, which potentially contributed to the favourable adverse event profile.

HDAC inhibitors and carfilzomib
In vitro, ricolinostat and carfilzomib combination treatment led to synergistic anti-MM effects and an in vivo mouse xenograft study confirmed a decrease in tumour volume, associated with apoptosis, providing additional support for the combination of deacetylase and proteasome inhibitors.72 The combination of panobinostat and carfilzomib was shown to be feasible and effective in patients with RRMM in a phase I/II study.73 ORR in all patients (n=35) was 77% and common grade 3–4 haematological adverse events were thrombocytopenia (47%) and neutropenia (8%). Two additional ongoing studies are evaluating the combination of panobinostat and carfilzomib (ClinicalTrials.gov Identifiers: NCT01549431 and NCT01496118) and are expected to complete late-2016.74,75

HDAC inhibitors and pomalidomide
In vitrodata have suggested synergism between the HDACi, ricolinostat, and both lenalidomide and pomalidomide.76 Two clinical trials of ricolinostat in combination with pomalidomide and dexamethasone in RRMM (planned n=20 and n=100) are ongoing (NCT 02189343 and NCT 01997840).

Epigenetic therapies in combination with monoclonal antibodies
HDACi have been shown to modulate the anti-tumour immune responses through the engagement of natural killer group 2D (NKG2D) receptors with their ligands.77,78 HDACi also upregulate antigen expression on tumour cells.79,80 It has been hypothesised that HDAC inhibition will increase antibody-dependent cell-mediated cytotoxicity by upregulating NKG2D ligand and CD38 expression on MM cells thereby contributing to improved efficacy of monoclonal antibody treatment during induction and especially during consolidation and maintenance. Finally, panobinostat has been shown to upregulate the MM-specific kappa myeloma antigen in vitro and subsequently enhance antibodydependent cellular cytotoxicity with the developmental monoclonal antibody MDX1097.81

In mice studies, both anti-PD-1 and anti-cytotoxic T-lymphocyteassociated antigen-4 (CTLA-4) antibodies were unable to eradicate large, modestly immunogenic CT26 tumours or metastatic 4T1 tumours.82 Co-treatment with epigenetic-modulating drugs (5-azactytidine, a DNA methyltransferase inhibitor) and entinostat, (MS-275/SNDX-275; a Class I HDAC inhibitor) and checkpoint inhibitors improved treatment outcomes substantially, curing >80% of the tumour-bearing mice. The investigators revealed by functional studies that the main targets of the epigenetic modulators were myeloid-derived suppressor cells.

Conclusions and future directions
The treatment of MM with combination approaches has been described as entering a new era, where even target relapsed/refractory cases can be targeted.83,84 Increased understanding of genetic and epigenetic interactions in MM may ultimately facilitate the development of novel, individualised treatment strategies.63 Panobinostat and ricolinostat have shown efficacy in combination with bortezomib and dexamethasone in both newly diagnosed and heavily pre-treated patients with advanced MM. Panobinostat in combination with bortezomib and dexamethasone became the first HDACi approved for the treatment of MM, specifically for relapsed or RRMM in patients with ≥2 prior regimens including bortezomib and an immunomodulatory drug. HDAC inhibition in combination with lenalidomide and dexamethasone has proven effective even in patients who are refractory to lenalidomide, suggesting that panobinostat may be able to overcome drug resistance to IMIDS thus restoring lenalidomide sensitivity. The promising results in relapsed or RRMM from the PANORAMA 1 trial and other combination trials discussed here have paved the way to explore novel panobinostat combinations (panobinostat–carfilzomib or panobinostat–pomalidomide) as well as panobinostat combinations in other settings, such as front-line or maintenance therapy.

Preclinical evidence suggests that there is potential for HDACi–monoclonal antibody combinations in a clinical setting. There are also preclinical data supporting the basis for combining novel immunotherapeutics targeting programmed cell death protein 1 (PD-1/PD-ligand 1 pathway with HDACi.85 However, to progress this opportunity in a rational manner future preclinical work needs to focus on understanding the interplay between epigenetic regulation and the expression of antibody target antigens on MM cells and defining the immune modulatory capacity of HDACi in MM. This will inform the design of combinatorial therapeutic strategies to enhance monoclonal antibody-induced antibodydependent cell-mediated cytotoxicity in vitro and in vivo. There may also be an opportunity for a sequential therapeutic strategy to “prime” MM cells for monoclonal antibodies, chimeric antigen receptor-T cell therapy or other immune targeted therapies in development. While the prospects for enhancing MM outcome through the incorporation of HDACi into the therapeutic armamentarium are becoming evident it is clear that the optimal use of these agents requires further understanding of their pleiotropic anti-tumour capacity through future carefully designed preclinical and correlative studies.

Trial Names
PANORAMA 1 = Panobinostat or Placebo With Bortezomib and Dexamethasone in Patients With Relapsed Multiple Myeloma
PANORAMA 2 = Panobinostat in Combination with Bortezomib and Dexamethasone in Patients with Relapsed and Bortezomib-refractory Myeloma
VANTAGE = VorinostAt CliNical Trials in HemAtologic and solid maliGnanciEs

Article Information:
Disclosure

Andrew Spencer and Sridurga Mithraprabhu have nothing to disclose in relation to this article. This study involves a review of the literature and did not involve any studies with human or animal subjects performed by any of the authors.
Authorship: All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship of this manuscript, take responsibility for the integrity of the work as a whole, and have given final approval to the version to be published.

Correspondence

Andrew Spencer, Alfred Health, 55 Commercial Road, Melbourne, Victoria 3004, Australia. E: aspencer@netspace.net.au

Support

The publication of this article was supported by Novartis. The views and opinions expressed are those of the authors and do not necessarily reflect those of Novartis. The authors provided Novartis with the opportunity to review the article for scientific accuracy before submission. Any resulting changes were made at the author’s discretion.

Access

This article is published under the Creative Commons Attribution Noncommercial License, which permits any non-commercial use, distribution, adaptation and reproduction provided the original author(s) and source are given appropriate credit.

Acknowledgements

Medical writing assistance was provided by Catherine Amey at Touch Medical Media, UK, supported by Novartis.

Received

2016-08-03T00:00:00

References

1. Esteller M, Cancer epigenomics: DNA methylomes and histonemodification maps, Nat Rev Genet, 2007;8:286–98. 2. Plass C, Pfister SM, Lindroth AM, et al., Mutations in regulators of the epigenome and their connections to global chromatin patterns in cancer, Nat Rev Genet, 2013;14:765–80. 3. Palumbo A, Anderson K, Multiple myeloma, N Engl J Med, 2011;364:1046–60. 4. Morgan GJ, Walker BA, Davies FE. The genetic architecture of multiple myeloma. Nature reviews, Cancer, 2012;12(5):335–48. 5. Bergsagel PL, Mateos MV, Gutierrez NC, et al., Improving overall survival and overcoming adverse prognosis in the treatment of cytogenetically high-risk multiple myeloma, Blood, 2013;121:884–92. 6. Munshi NC, Anderson KC, New strategies in the treatment of multiple myeloma, Clin Cancer Res, 2013;19:3337–44. 7. Zweegman S, Palumbo A, Bringhen S, et al., Age and aging in blood disorders: multiple myeloma, Haematologica, 2014;99:1133–7. 8. Katzel JA, Hari P, Vesole DH, Multiple myeloma: charging toward a bright future, CA Cancer J Clin, 2007;57:301–18. 9. Parkin DM, Bray F, Ferlay J, et al., Global cancer statistics, 2002, CA Cancer J Clin, 2005;55:74–108. 10. Cook R, Economic and clinical impact of multiple myeloma to managed care, J Manag Care Pharm, 2008;14(7 Suppl):19–25. 11. Turesson I, Velez R, Kristinsson SY, et al., Patterns of multiple myeloma during the past 5 decades: stable incidence rates for all age groups in the population but rapidly changing age distribution in the clinic, Mayo Clin Proc, 2010;85:225–30. 12. Bosetti C, Bertuccio P, Malvezzi M, et al., Cancer mortality in Europe, 2005–2009, and an overview of trends since 1980, Ann Oncol, 2013;24:2657–71. 13. Kumar SK, Rajkumar SV, Dispenzieri A, et al., Improved survival in multiple myeloma and the impact of novel therapies, Blood, 2008;111:2516–20. 14. Bergsagel PL, Kuehl WM, Molecular pathogenesis and a consequent classification of multiple myeloma, J Clin Oncol, 2005;23:6333–8. 15. Chng WJ, Santana-Davila R, Van Wier SA, et al., Prognostic factors for hyperdiploid-myeloma: effects of chromosome 13 deletions and IgH translocations, Leukemia, 2006;20:807–13. 16. Chapman MA, Lawrence MS, Keats JJ, et al., Initial genome sequencing and analysis of multiple myeloma, Nature, 2011;471(7339):467–72. 17. Walker BA, Wardell CP, Chiecchio L, et al., Aberrant global methylation patterns affect the molecular pathogenesis and prognosis of multiple myeloma, Blood, 2011;117:553–62. 18. Rajkumar SV, Blood E, Vesole D, et al., Phase III clinical trial of thalidomide plus dexamethasone compared with dexamethasone alone in newly diagnosed multiple myeloma: a clinical trial coordinated by the Eastern Cooperative Oncology Group, J Clin Oncol, 2006;24:431–6. 19. Rajkumar SV, Hayman S, Gertz MA, et al., Combination therapy with thalidomide plus dexamethasone for newly diagnosed myeloma, J Clin Oncol, 2002;20:4319–23. 20. Kumar S, Flinn I, Richardson PG, et al., Randomized, multicenter, phase 2 study (EVOLUTION) of combinations of bortezomib, dexamethasone, cyclophosphamide, and lenalidomide in previously untreated multiple myeloma, Blood, 2012;119:4375–82. 21. Richardson PG, Weller E, Lonial S, et al., Lenalidomide, bortezomib, and dexamethasone combination therapy in patients with newly diagnosed multiple myeloma, Blood, 2010;116:679–86. 22. Mimura N, Hideshima T, Anderson KC, Novel therapeutic strategies for multiple myeloma, Exp Hematol, 2015;43:732–41. 23. Smith EM, Boyd K, Davies FE, The potential role of epigenetic therapy in multiple myeloma, Br J Haematol, 2010;148:702–13. 24. Toyota M, Ahuja N, Ohe-Toyota M, et al., CpG island methylator phenotype in colorectal cancer,Proc Natl Acad Sci U S A, 1999;96:8681–6. 25. Issa JP, CpG island methylator phenotype in cancer, Nature Rev Cancer, 2004;4:988–93. 26. Martin P, Garcia-Cosio M, Santon A, et al., Aberrant gene promoter methylation in plasma cell dyscrasias, Experimental and molecular pathology, 2008;84:256–61. 27. Kim JH, Karnovsky A, Mahavisno V, et al., LRpath analysis reveals common pathways dysregulated via DNA methylation across cancer types, BMC Genomics, 2012;13:526. 28. Kaiser MF, Johnson DC, Wu P, et al., Global methylation analysis identifies prognostically important epigenetically inactivated tumor suppressor genes in multiple myeloma, Blood, 2013;122:219–26. 29. Agirre X, Castellano G, Pascual M, et al., Whole-epigenome analysis in multiple myeloma reveals DNA hypermethylation of B cell-specific enhancers, Genome Res, 2015;25:478–87. 30. Jones PA, Functions of DNA methylation: islands, start sites, gene bodies and beyond, Nat Rev Genet, 2012;13:484–92. 31. Raynal NJ, Si J, Taby RF, et al., DNA methylation does not stably lock gene expression but instead serves as a molecular mark for gene silencing memory, Cancer Res, 2012;72:1170–81. 32. Mateos MV, Garcia-Sanz R, Lopez-Perez R, et al., Methylation is an inactivating mechanism of the p16 gene in multiple myeloma associated with high plasma cell proliferation and short survival, Br J Haematol, 2002;118:1034–40. 33. Gonzalez-Paz N, Chng WJ, McClure RF, et al., Tumor suppressor p16 methylation in multiple myeloma: biological and clinical implications, Blood, 2007;109:1228–32. 34. Zhou W, Chen H, Hong X, et al., Knockdown of DNA methyltransferase-1 inhibits proliferation and derepresses tumor suppressor genes in myeloma cells, Oncol Lett, 2014;8:2130–34. 35. Zhan F, Hardin J, Kordsmeier B, et al., Global gene expression profiling of multiple myeloma, monoclonal gammopathy of undetermined significance, and normal bone marrow plasma cells, Blood, 2002;99:1745–57. 36. Macfarlane LA, Murphy PR, MicroRNA: Biogenesis, Function and Role in Cancer, Curr Genomics, 2010;11:537–61. 37. Wong KY, Huang X, Chim CS, DNA methylation of microRNA genes in multiple myeloma, Carcinogenesis, 2012;33:1629–38. 38. Cohen I, Poreba E, Kamieniarz K, et al., Histone modifiers in cancer: friends or foes?, Genes Cancer, 2011;2:631–47. 39. Lauring J, Abukhdeir AM, Konishi H, et al., The multiple myeloma associated MMSET gene contributes to cellular adhesion, clonogenic growth, and tumorigenicity, Blood, 2008;111:856–64. 40. Mirabella F, Wu P, Wardell CP, et al., MMSET is the key molecular target in t(4;14) myeloma, Blood Cancer J, 2013;3:e114. 41. Min DJ, Ezponda T, Kim MK, et al., MMSET stimulates myeloma cell growth through microRNA-mediated modulation of c-MYC, Leukemia, 2013;27:686–94. 42. Wu SP, Pfeiffer RM, Ahn I, et al., Impact of genes highly correlated with MMSET myeloma on survival of Non-MMSET myeloma patients, Clin Cancer Res, 2016;22:4039–44. 43. Martinez-Garcia E, Popovic R, Min DJ, et al., The MMSET histone methyl transferase switches global histone methylation and alters gene expression in t(4;14) multiple myeloma cells, Blood, 2011;117:211–20. 44. Brito JL, Walker B, Jenner M, et al., MMSET deregulation affects cell cycle progression and adhesion regulons in t(4;14) myeloma plasma cells, Haematologica, 2009;94:78–86. 45. Cea M, Cagnetta A, Gobbi M, et al., New insights into the treatment of multiple myeloma with histone deacetylase inhibitors, Curr Pharm Des, 2013;19:734–44. 46. Weichert W, HDAC expression and clinical prognosis in human malignancies, Cancer Lett, 2009;280:168–76. 47. Mithraprabhu S, Kalff A, Chow A, et al., Dysregulated Class I histone deacetylases are indicators of poor prognosis in multiple myeloma, Epigenetics, 2014;9:1511–20. 48. Kikuchi J, Wada T, Shimizu R, et al., Histone deacetylases are critical targets of bortezomib-induced cytotoxicity in multiple myeloma, Blood, 2010;116:406–17. 49. Esteve PO, Chin HG, Smallwood A, et al., Direct interaction between DNMT1 and G9a coordinates DNA and histone methylation during replication, Genes Dev, 2006;20:3089–103. 50. Khong T, Sharkey J, Spencer A, The effect of azacitidine on interleukin-6 signaling and nuclear factor-kappaB activation and its in vitro and in vivo activity against multiple myeloma, Haematologica, 2008;93:860–9. 51. Moreaux J, Bruyer A, Veyrune JL, et al., DNA methylation score is predictive of myeloma cell sensitivity to 5-azacitidine, Br J Haematol, 2014;164:613–6. 52. Kalff A, Reed K, Khong TT, et al., 3033 Roar: A Phase Ib Trial of Oral Azacitidine in Combination with Lenalidomide and Dexamethasone for Relapsed Multiple Myeloma (MM) Patients Refractory to Prior Lenalidomide, Presented at: American Society of Hematology 57th Annual Meeting & Exposition. Orlando, FL, USA, 5–8 December 2015. 53. Xie Z, Bi C, Chooi JY, et al., MMSET regulates expression of IRF4 in t(4;14) myeloma and its silencing potentiates the effect of bortezomib, Leukemia, 2015;29:2347–54. 54. Mithraprabhu S, Khong T, Jones SS, et al., Histone deacetylase (HDAC) inhibitors as single agents induce multiple myeloma cell death principally through the inhibition of class I HDAC, Br J Haematol, 2013;162:559–62. 55. Tandon N, Ramakrishnan V, Kumar SK, Clinical use and applications of histone deacetylase inhibitors in multiple myeloma, Clin Pharmacol, 2016;8:35–44. 56. Kalff A, Khong T, Mithrapabhu S, et al., A 2-Stage Phase II study of panobinostat consolidation in myeloma patients with < CR following high-dose chemotherapy (HDT) conditioned autologous stem cell transplantation (ASCT), Clin Lymphoma Myeloma Leuk, 2015;15(Supplement 3):e132. 57. Kopito RR, Aggresomes, inclusion bodies and protein aggregation, Trends Cell Biol,2000;10:524–30. 58. Bennett EJ, Bence NF, Jayakumar R, et al., Global impairment of the ubiquitin-proteasome system by nuclear or cytoplasmic protein aggregates precedes inclusion body formation, Mol Cell, 2005;17:351–65. 59. Kawaguchi Y, Kovacs JJ, McLaurin A, et al., The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress, Cell, 2003;115:727–38. 60. Hideshima T, Bradner JE, Wong J, et al., Small-molecule inhibition of proteasome and aggresome function induces synergistic antitumor activity in multiple myeloma, Proc Natl Acad Sci U S A, 2005;102:8567–72. 61. Santo L, Hideshima T, Kung AL, et al., Preclinical activity, pharmacodynamic, and pharmacokinetic properties of a selective HDAC6 inhibitor, ACY-1215, in combination with bortezomib in multiple myeloma, Blood, 2012;119:2579–89. 62. Vogl DT, Raje N, Hari P, et al., Phase 1b results of ricolinostat (ACY-1215) combination therapy with bortezomib and dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma (MM) [abstract 4764], Blood, 2014;124:4764. 63. Dimopoulos M, Siegel DS, Lonial S, et al., Vorinostat or placebo in combination with bortezomib in patients with multiple myeloma (VANTAGE 088): a multicentre, randomised, doubleblind study, Lancet Oncol, 2013;14:1129–40. 64. Siegel DS, Dimopoulos M, Jagannath S, et al., VANTAGE 095: An International, Multicenter, Open-Label Study of Vorinostat (MK-0683) in Combination With Bortezomib in Patients With Relapsed and Refractory Multiple Myeloma, Clin Lymphoma Myeloma Leuk, 2016;16:329–34. 65. Richardson PG, Schlossman RL, Alsina M, et al., PANORAMA 2: panobinostat in combination with bortezomib and dexamethasone in patients with relapsed and bortezomibrefractory myeloma, Blood, 2013;122:2331–7. 66. 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;15:1195–206. 67. San-Miguel JF, Hungria VT, Yoon SS, et al., Panobinostat plus bortezomib and dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma who received prior bortezomib and IMiDs: A predefined subgroup analysis of PANORAMA 1, J Clin Oncol, 2015;33(suppl; abstr 8526). 68. Chari A, Cho HJ, Parekh SS, et al., A phase II study of panobinostat with lenalidomide and weekly dexamethasone in myeloma, J Clin Oncol, 2015;33(suppl; abstr 8528). 69. Yee AJ, Voorhees PM, Bensinger W, et al., Ricolinostat (ACY- 1215), a Selective HDAC6 Inhibitor, in Combination with Lenalidomide and Dexamethasone: Results of a Phase 1b Trial in Relapsed and Relapsed Refractory Multiple Myeloma, Presented at: 56th American Society of Hematology (ASH) Annual Meeting and Exposition, San Francisco, CA, USA, 6–9 December 2014. 70. Popat R, Brown S, Flanagan LM, et al., Thalidomide, Dexamethasone and Panobinostat (VTD-P) for Patients with Relapsed and Relapsed/ Refractory Myeloma: Preliminary Results of the Muk-Six Phase I/IIa Trial, Presented at: 56th American Society of Hematology (ASH) Annual Meeting and Exposition. San Francisco, CA, USA, 6–9 December 2014. 71. Shah JJ, Feng L, Manasanch EE, et al., Phase I/Ib Trial of the Efficacy and Safety of Combination Therapy with Lenalidomide/ Bortezomib/Dexamethasone (RVD) and Panobinostat in Transplant-Eligible Patients with Newly Diagnosed Multiple Myeloma. Presented at: 56th American Society of Hematology (ASH) Annual Meeting and Exposition. San Francisco, CA, USA, 6–9 December 2014. 72. Mishima Y, Santo L, Eda H, et al., Ricolinostat (ACY-1215) induced inhibition of aggresome formation accelerates carfilzomib-induced multiple myeloma cell death,Br J Haematol, 2015;169:423–34. 73. Berdeja JG, Hart LL, Mace JR, et al., Phase I/II study of the combination of panobinostat and carfilzomib in patients with relapsed/refractory multiple myeloma, Haematologica, 2015;100:670–6. 74. Shah JJ, Thomas SK, Weber DM, et al., Phase 1/1b Study of the Efficacy and Safety of the Combination of Panobinostat + Carfilzomib in Patients with Relapsed and/or Refractory Multiple Myeloma, Presented at: 54th American Society of Hematology (ASH) Annual Meeting and Exposition. Atlanta, GA, USA, 8–11 December 2012. 75. Kaufman JL, Zimmerman T, Rosenbaum CA, et al., Phase I Study of the Combination of Carfilzomib and Panobinostat for Patients with Relapsed and Refractory Myeloma: A Multiple Myeloma Research Consortium (MMRC) Clinical Trial, Presented at: 56th American Society of Hematology (ASH) Annual Meeting and Exposition. San Francisco, CA, USA, 6–9 December 2014. 76. Quayle SN, Jones SS, ACY-1215, a First-In-Class Selective Inhibitor Of HDAC6, Demonstrates Significant Synergy With Immunomodulatory Drugs (IMiDs) In Preclinical Models Of Multiple Myeloma (MM), Presented at: 55th Annual Society of Hematology (ASH) Annual Meeting and Exposition. New Orleans, LA, USA, 7–10 December 2013. 77. Wu X, Tao Y, Hou J, et al., Valproic acid upregulates NKG2D ligand expression through an ERK-dependent mechanism and potentially enhances NK cell-mediated lysis of myeloma. Neoplasia, 2012;14:1178–89. 78. Huang B, Sikorski R, Sampath P, et al., Modulation of NKG2Dligand cell surface expression enhances immune cell therapy of cancer, J Immunother, 2011;34:289–96. 79. Kuster K, Grotzinger C, Koschel A, et al., Sodium butyrate increases expression of the coxsackie and adenovirus receptor in colon cancer cells, Cancer Invest, 2010;28:268–74. 80. Canes D, Chiang GJ, Billmeyer BR, et al., Histone deacetylase inhibitors upregulate plakoglobin expression in bladder carcinoma cells and display antineoplastic activity in vitro and in vivo, Int J Cancer, 2005;113:841–8. 81. Asvadi P, Cuddihy A, Dunn RD, et al., MDX-1097 induces antibody-dependent cellular cytotoxicity against kappa multiple myeloma cells and its activity is augmented by lenalidomide, Br J Haematol, 2015;169:333–43. 82. Kim K, Skora AD, Li Z, et al., Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells, Proc Natl Acad Sci U S A, 2014;111:11774–9. 83. Naymagon L, Abdul-Hay M, Novel agents in the treatment of multiple myeloma: a review about the future, J Hematol Oncol, 2016;9:52. 84. Nooka AK, Lonial S, Novel Combination Treatments in Multiple Myeloma, Oncology, 2016;30:451–65. 85. Ray A, Sharma D, Song Y, et al., Combination of Anti-PD-L1 Antibody with IMiD® Immunomodulatory Drugs, HDAC Inhibitor ACY-1215, Bortezomib, or Toll-like Receptor 9 Agonist Enhances Anti-Tumor Immunity and Cytotoxicity in Multiple Myeloma, Presented at: 57th American Society of Hematology (ASH) ANnual Meeting. Orlando, FL, USA, 5–8 December 2015.

Further Resources

Share this Article
Related Content In Multiple Myeloma
  • Copied to clipboard!
    accredited arrow-down-editablearrow-downarrow_leftarrow-right-bluearrow-right-dark-bluearrow-right-greenarrow-right-greyarrow-right-orangearrow-right-whitearrow-right-bluearrow-up-orangeavatarcalendarchevron-down consultant-pathologist-nurseconsultant-pathologistcrosscrossdownloademailexclaimationfeedbackfiltergraph-arrowinterviewslinkmdt_iconmenumore_dots nurse-consultantpadlock patient-advocate-pathologistpatient-consultantpatientperson pharmacist-nurseplay_buttonplay-colour-tmcplay-colourAsset 1podcastprinter scenerysearch share single-doctor social_facebooksocial_googleplussocial_instagramsocial_linkedin_altsocial_linkedin_altsocial_pinterestlogo-twitter-glyph-32social_youtubeshape-star (1)tick-bluetick-orangetick-red tick-whiteticktimetranscriptup-arrowwebinar Sponsored Department Location NEW TMM Corporate Services Icons-07NEW TMM Corporate Services Icons-08NEW TMM Corporate Services Icons-09NEW TMM Corporate Services Icons-10NEW TMM Corporate Services Icons-11NEW TMM Corporate Services Icons-12Salary £ TMM-Corp-Site-Icons-01TMM-Corp-Site-Icons-02TMM-Corp-Site-Icons-03TMM-Corp-Site-Icons-04TMM-Corp-Site-Icons-05TMM-Corp-Site-Icons-06TMM-Corp-Site-Icons-07TMM-Corp-Site-Icons-08TMM-Corp-Site-Icons-09TMM-Corp-Site-Icons-10TMM-Corp-Site-Icons-11TMM-Corp-Site-Icons-12TMM-Corp-Site-Icons-13TMM-Corp-Site-Icons-14TMM-Corp-Site-Icons-15TMM-Corp-Site-Icons-16TMM-Corp-Site-Icons-17TMM-Corp-Site-Icons-18TMM-Corp-Site-Icons-19TMM-Corp-Site-Icons-20TMM-Corp-Site-Icons-21TMM-Corp-Site-Icons-22TMM-Corp-Site-Icons-23TMM-Corp-Site-Icons-24TMM-Corp-Site-Icons-25TMM-Corp-Site-Icons-26TMM-Corp-Site-Icons-27TMM-Corp-Site-Icons-28TMM-Corp-Site-Icons-29TMM-Corp-Site-Icons-30TMM-Corp-Site-Icons-31TMM-Corp-Site-Icons-32TMM-Corp-Site-Icons-33TMM-Corp-Site-Icons-34TMM-Corp-Site-Icons-35TMM-Corp-Site-Icons-36TMM-Corp-Site-Icons-37TMM-Corp-Site-Icons-38TMM-Corp-Site-Icons-39TMM-Corp-Site-Icons-40TMM-Corp-Site-Icons-41TMM-Corp-Site-Icons-42TMM-Corp-Site-Icons-43TMM-Corp-Site-Icons-44TMM-Corp-Site-Icons-45TMM-Corp-Site-Icons-46TMM-Corp-Site-Icons-47TMM-Corp-Site-Icons-48TMM-Corp-Site-Icons-49TMM-Corp-Site-Icons-50TMM-Corp-Site-Icons-51TMM-Corp-Site-Icons-52TMM-Corp-Site-Icons-53TMM-Corp-Site-Icons-54TMM-Corp-Site-Icons-55TMM-Corp-Site-Icons-56TMM-Corp-Site-Icons-57TMM-Corp-Site-Icons-58TMM-Corp-Site-Icons-59TMM-Corp-Site-Icons-60TMM-Corp-Site-Icons-61TMM-Corp-Site-Icons-62TMM-Corp-Site-Icons-63TMM-Corp-Site-Icons-64TMM-Corp-Site-Icons-65TMM-Corp-Site-Icons-66TMM-Corp-Site-Icons-67TMM-Corp-Site-Icons-68TMM-Corp-Site-Icons-69TMM-Corp-Site-Icons-70TMM-Corp-Site-Icons-71TMM-Corp-Site-Icons-72