Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jul 1.
Published in final edited form as: Clin Lymphoma Myeloma Leuk. 2019 Mar 25;19(7):397–405. doi: 10.1016/j.clml.2019.03.017

Future of personalized therapy targeting aberrant signaling pathways in multiple myeloma

Faiz Anwer 1,*, Kevin Mathew Gee 2, Ahmad Iftikhar 3, Mirza Baig 4, Atlantis Dawn Russ 3, Sabina Saeed 5, Muhammad Abu Zar 6, Faryal Razzaq 6, Jennifer Carew 6, Steffan Nawrocki 6, Hussam Al-Katab 7, Nadia Nunes Cavalcante 3, Ali McBride 8, Jason Valent 1, Christy Samaras 1
PMCID: PMC6626550  NIHMSID: NIHMS1525331  PMID: 31036508

Abstract

Multiple myeloma (MM) is genetically complex disease. Identification of mutations and aberrant signaling pathways that contribute to the progression of MM and drug resistance has potential to lead to specific targets and personalized treatment. Aberrant signal pathways include: RAS pathway activation due to RAS or BRAF mutations (targeted by vermurafenib alone or combined with cobimetinib), BCL2 overexpression especially in t(11:14) (targeted by venetoclax), JAK2 pathway activation (targeted by ruxolitinib), NF-kB pathway activation (treated with DANFIN combined with bortezomib), MDM2 overexpression (targeted by DS-3032b) and targeting the PI3K/mTOR pathway (targeted by BEZ235). Cyclin D1 (CCND1), and MYC are also emerging as key potential targets. In addition, histone deacetylase (HDAC) inhibitors are already in use for the treatment of MM in combination therapy and targeted inhibition of FGFR3 (AZD4547) is effective in myeloma cells with t(4;14) translocation. Bromodomain and extra terminal (BET) protein antagonists decrease the expression of MYC and have displayed promising anti-myeloma activity. A better understanding of the alterations in signaling pathways that promote MM progression will further inform the development of precision therapy for patients.

Keywords: Multiple Myeloma, Intracellular pathway, Signaling pathways, Mutations, Personalized therapy

Introduction:

Multiple myeloma (MM) is a genetically complex B-cell malignancy. Clonal plasma cells acquire increasing levels of genetic aberrations including copy number variations, point mutations, gene deletions, and translocations. Clinical guidelines define disease risk stratification based on cytogenetic aberrations and secondary mutational events in plasma cell populations of MM patients. Cytogenetic aberrations in MM can be divided into two main groups: hyperdiploid MM (H-MM) and non-hyperdiploid MM (NH-MM). H-MM are characterized by trisomies of chromosomes 3, 5, 7, 9, 11, 15, 19, and 21. NH-MM harbor IGH translocations, mainly t (4;14), t (6;14), t (11;14), t (14;16), and t (14;20).1,2,3 Secondary mutational events contribute to tumor progression and include MYC rearrangements, copy number variations (CNV), del(13q), dup(1q), del(1p) del(17p) and somatic mutations in KRAS, NRAS, BRAF, and P53 among many others.

MM is clinically divided into high, intermediate and standard risk based on interphase fluorescence in situ hybridization (I-FISH). High risk MM (HRMM) patients have del (17p), t(14;16), t(14;20), t(4;14), del 13, plasma cell labeling index (PCLI), a measure of plasma cell proliferation ≥ 3% and gene expression profiling (GEP) shows a high risk signature. The intermediate risk category includes t(14;4), and 1q gain. Standard risk patients carry t(11;14) and t(6;14) mutations.4,5 For disease staging, the International Myeloma Working Group (IMWG) combined the International Staging System (ISS) with chromosomal abnormalities detected by I-FISH after CD138 and serum lactate dehydrogenase (LDH) sorting for newly diagnosed MM (NDMM) and proposed a revised (R) staging system named R-ISS6. The Mayo Clinic’s mSMART risk stratification guidelines 5 rely on I-FISH and serum biomarkers (LDH, β2 microglobulin), however, many characterized aberrations are not adequately surveyed by these methods. The use of other sequencing and array technologies to identify changes to the genomic landscape such as whole exome sequencing (WES), whole genome sequencing (WGS), single nucleotide polymorphism (SNP) arrays, gene expression profiling (GEP), array comparative genomic hybridization (aCGH), in addition to historical genetic testing methods could identify new subgroups and novel targetable pathways in MM. Research identifying aberrant molecular pathways enhances our understanding of the mechanisms of disease in MM and offers opportunity for the development of new, more targeted therapeutic strategies.

Methods

We performed a focused review of the literature to compile current clinical data regarding novel genetic mutations, disrupted intracellular pathways, cancer driving aberrations and response to targeted therapies to summarize the potential for the advancement of personalized and precision therapy against MM.

Literature Search

We searched PubMed, and Clinicaltrials.gov for only English language studies. We did not limit our search to time period. We also cross checked bibliographies manually to avoid missing potential studies. We retrieved preclinical studies, phase I/II trials and data from clinicaltrials.gov on complete or still recruiting trials. We based our electronic database search on keywords like multiple myeloma, cytogenetics, molecular profiling, mutations, disrupted pathways and prognosis. A sample search strategy is “(multiple myeloma [MeSH Terms]) AND factor NF-kb, transcription [MeSH Terms]) AND “drug therapy” [MeSH Subheading])”. The final updated search was performed on 03/16/2018.

Eligibility Criteria

We included 1) Preclinical studies, 2) Case report & series, 3) Phase 1 or higher clinical trials, and 4) studies that had efficacy outcomes clearly documented. We excluded studies in which no objective efficacy outcomes were reported.

Results

The literature search yielded a total of 827 articles. Relevant articles which focused on the effect of drug therapy on MM cell lines and patients with specific mutation and aberrant intracellular pathways yielded 49 preclinical studies, case reports, case series and clinical trials. We present the summary of findings from these articles here and in Table 1.

Table 1. Emerging therapies targeting novel aberrant pathways in Multiple Myeloma.

Abbreviations: BCL-2=B cell lymphoma-2, JAK-STAT=Janus kinase Signal Transducer and Activator of Transcription proteins, MEK=Mitogen activated protein kinase kinase, FGFR=fibroblast growth factor receptor, NF-KB=nuclear factor kappa light chain enhancer of activated B cells, mTOR=mammalian target of rapamycin, MM=multiple myeloma, NSD2=nuclear receptor binding SET domain2, BET=bromodomain and extra terminal, BRD=bromodomain, MYC=Myeloctomatosis (oncogene), HDAC=histone deacetylase

Pathway Drug Name Mechanism of action Clinical Trial
BCL-2 overexpression
venetoclax (ABT-199) BCL-2 inhibitor NCT01794520
JAK-STAT
ruxolitinib JAK2 inhibitor NCT00639002
SC99 JAK2/STAT3 inhibitor
NF-KB
DANFIN Proteasome inhibitor-inhibits NF-KB activation and signaling
Ras/Raf/Mek/Erk
selumetinib (AZD6244) MEK 1/2 inhibitor -blocks the MEK immediately downstream of BRAF NCT01085214
sorafenib Multi kinase inhibitor-inhibits cell surface TKIs and downstream intracellular serine/threonine kinases
vemurafenib BRAF inhibitor NCT01524978
cobimetinib BRAF inhibitor NCT03312530
FGFR mutation
MFGR1877S Anti-FGFR monoclonal antibody
dovitinib (TKI258) Non selective FGFR inhibitor NCT02465060
BGJ398 Non selective FGFR inhibitor
AZD4547 FGFR (1-3) inhibitor
Cyclin D
palbociclib CDK4/6 inhibitor
PI3K/AKT/mTOR
clioquinol Hydroxyquinoline, downregulates expression of mTOR, inducing autophagy in MM cells NCT00963495
SC-06 Disrupts mTOR signaling pathway by downregulating Raptor, a key component of mTORC1 signaling
BEZ235 mTOR inhibitor
C98 PI3K/AKT inhibitor
C96 PI3K/AKT inhibitor
Epigenetic regulation
LEM06 MMSET/NSD2 inhibitor (histone methyl transferase inhibitor)
JQ-1 BET inhibitor
CPI-0601 BET inhibitor
OTX015/MK8628 BET inhibitor NCT01713582
CPI-203 BET inhibitor
CGI13250 Suppresses MYC transcription by blocking BRD4 binding to MYC promoter
panobinostat HDAC inhibitor NCT01651039
NCT01023308
vorinostat HDAC inhibitor NCT00773838
romidepsin HDAC inhibitor NCT00066638
NCT01755975
ricolinostat HDAC inhibitor NCT01323751

A). Developing Targets: Signaling pathways against which specific agents have been tested in clinical trials

Apoptosis in MM

BCL-2 proteins that regulate cell death are grouped according to their pro-apoptotic (i.e. BAX, BAK, BAD, BIK) or anti-apoptotic (i.e. BCL-2, BCL-XL, BCL-W, MCL-1) activity. MM cell lines and primary patient samples, with t(11;14), detected in up to 20% MM patients, have upregulation of cyclin D1, high BCL-2, low BCL-XL, and low MCL-1 profile.7 Patients with MM often have overexpression of BCL-2 or MCL-1, which makes them resistant to apoptosis.8 Therefore, inhibitors that target these anti-apoptotic proteins have been the focus of several studies.

BCL2 inhibition

Kumar et al. conducted a phase 1 study on 66 patients with relapsed refractory MM (RRMM) using venetoclax, a BCL-2 inhibitor. Venetoclax given weekly in dose-escalation (300, 600, 900, or 1200 mg) had an acceptable toxicity profile. The overall response rate (ORR) was 21% (14/66) and 15% achieved a partial response (PR) or better. The best response (12/14 [86%]) was seen in patients with t(11;14), a translocation that is associated with high levels of BCL-2. Those with t(11;14) had an ORR of 40% and PR of 27%.9 Moreau et al. in an open-label phase 1b study on 66 patients with RRMM used venetoclax in combination with the proteasome inhibitor bortezomib and dexamethasone, a steroid that has been reported to increase the expression of proapoptotic protein BIM.8 The treatment was well tolerated when considering adverse events, with an ORR of 67% (44/66) and a PR rate of 42% or better. The median time to progression was 9.5 months and the duration of response was 9.7 months.8

Ras/Raf/Mek/Erk

The RAS/RAF/MEK/ERK pathway is an important regulator of gene expression, cell survival, proliferation, migration and angiogenesis. KRAS/NRAS/BRAF mutations are detectable in up to 50% of MM patients10 and 45-81% of RRMM patients.11 RAS mutations are linked to a more aggressive clinical course leading to shorter survival.12 t(4;14) translocation can cause increased expression of FGFR3 which also stimulates RAS/MAPK pathway.13 IL-6 also triggers growth of cells via the RAS/MAPK pathway.14 Xu et al.15 noted a dominant mutation cluster in RAS/RAF genes in samples taken from MM patients, identifying RAS/RAF/MEK/ERK signaling as a therapeutic target. In comparison with NDMM, RRMM patients have statistically significant higher overall incidence of RAS/BRAF mutations (p =0.011), which are mostly driven by a higher prevalence of NRAS mutations (p =0.010). Mulligan et al16 observed that NRAS but not KRAS-mutant MM had significantly lower response rates (P = .016) and a shorter time to progression (P = .012) following treatment with bortezomib monotherapy. These data indicate that an important component of bortezomib’s antitumor activity acts at a level upstream of NRAS survival signaling and thus cannot effectively kill myeloma cells with this mutation. MEK inhibitors can kill MM cell lines that have MAF oncogenes and are resistant to other chemotherapeutic agents like lenalidomide, pomalidomide, bortezomib and dexamethasone by re-sensitizing MM cells to these agents.1719

BRAF and MEK inhibition

Selumetinib and sorafenib are two agents under investigation for use in MM with mutations in the Ras/Raf/Mek/Erk pathway. In a phase II trial of the MEK1/2 inhibitor selumetinib (AZD6244) for treating RRMM as a single-agent, AZD6244 resulted in minimal improvement with only 2 out of 36 heavily pretreated RRMM patients achieving very good partial response (VGPR).20 Another phase II trial for assessing the efficacy of sorafenib, a multi-kinase inhibitor, in RRMM patients showed 50% overall survival (95% CI 27-73%) at 12 months and median progression-free survival of 1.2 months (95% CI 1.0-5.4). 21

Approximately 4% of patients with MM have BRAF mutations.22,23 Patients who harbor an activating V600E mutation in the BRAF kinase have an aggressive clinical course, higher incidence of extra-medullary disease and shorter overall survival (OS).24 BRAF mutations are highly prevalent in melanoma and hairy cell leukemia and treatment with vemurafenib, a BRAF inhibitor, has shown to have clinical benefit.25,26 Andrulis et al reported a RRMM patient with BRAF V600E who was refractory to all approved treatments, but responded rapidly and had a durable response as stable remission to vermurafenib.24 Larger scale trials are needed to further explore the role of BRAF mutation inhibitors for treatment of MM patients harboring the V600E mutation.

Cobimetinib (C21H21F3IN3O2) is an FDA approved drug to treat melanoma with a BRAF V600E or V600K mutation, in combination with vemurafenib. Cobimetinib is a reversible, non-ATP-competitive MEK1/MEK2/MAPK. In a case report of RRMM with V600E mutation, Mey et al. reported a rapid and sustained response with a combination of vemurafenib and cobimetinib.27 A phase III study of combined inhibition of BRAF (vemurafenib) and MEK (cobimetinib) showed a statistically significant (p<0.001) improvement in progression-free survival (PFS) of patients with BRAF-mutated melanoma, compared to the usage of vemurafenib alone. The usage of vemurafenib alone showed progression-free survival (PFS) of 6.2 months, while the combination showed 9.9 months of progression-free survival, total of a 16-week difference.28 A phase Ib/II clinical trial is currently underway using cobimetinib as a single agent and in combination with venetoclax with or without atezolizumab in patients with RRMM.29

FGFR3 inhibition

FGFR3 mutations have been identified in MM.30 t(4;14) brings FGFR3 under the control of Ig heavy chain promoter, causing aberrant expression of FGFR3.30,31 Myeloma cell lines with t(4;14) chromosomal translocation are very sensitive to FGFR3 targeted inhibition.31 Monoclonal antibodies bind to the FGFR extracellular domain, compete with FGFs and as a result block FGF-FGFR association. Monoclonal antibodies targeting FGFR3 have been shown to have significant inhibitor effect on cellular proliferation in t(4;14) positive multiple myeloma cases.32 MFGR1877S, is a human anti-FGFR3 monoclonal antibody which was studied in a phase I trial. It was found to be well-tolerated overall in patients with multiple RRMM. No objective responses were observed; however, stable disease was observed in 3 out of 14 patients for 3–4 cycles.33

Dovitinib (TKI258) is a well-studied second-generation non-selective FGFR inhibitor. Among its other actions, it has been shown to inhibit the cellular activity of FGFR3 in t(4;14) MM in pre-clinical studies.34 In a phase II, open label, non-randomized trial, dovitinib did not demonstrate single-agent activity in RRMM, but may stabilize disease in some t(4;14)-positive patients.35

BGJ398 is a potent, pan-FGFR inhibitor which demonstrated preclinical antitumor activity in RT112 bladder cancer xenograft models overexpressing wild-type FGFR3. In a study of t(4;14)-positive myeloma cells, 36 enhanced expression of FGFR3 was observed. However, the cells were not sensitive to FGFR inhibitors (neither dovitinib nor BGJ398). While preclinical data was promising, this study concluded that single agent activity is modest and often independent of FGFR status.

AZD4547 is a selective, small molecule, oral FGFR (FGFR 1-3) inhibitor which has shown to be a potent inhibitor of proliferation in cell lines with activation of the FGFR pathway and also in tumor xenograft models 37. Inhibition by AZD4547 is also shown to cause a significant dose-dependent tumor growth inhibition and increased survival in gastric cancer carrying an FGFR2 gene amplification.38 A Phase I, open label, multicenter study that assessed the safety, tolerability, pharmacokinetics and preliminary anti-tumor activity of ascending doses of AZD4547 in patients with advanced solid tumors was completed in March 2015.39 It is currently in phase II trials, aimed to define its role in targeted therapy directed by genetic testing in patients with advanced refractory solid tumors, lymphomas, or MM.40

CDK inhibition

Dysregulation of cyclin D is an early and unifying event in MM pathogenesis.41 Post-transcriptional modifications play an important role in regulating CCND2 expression.42 Overexpression of cyclin D1 is seen in up to 60% of MM cells, which is associated with either t(11;14)(q13;q32), polysomy of chromosome 11 or induced expression from interaction of MM tumor cells with the surrounding bone marrow stroma.43,44 CCND1 overexpression is historically associated with poor prognosis in MM.45,46 In a small series of patients, CCND1 overexpression conferred improved response to bortezomib therapy.47,48 In a study of 45 relapsed MM patients who received bortezomib, CCND1 expression was associated with better prognosis (OR, −2.21; p = 0.07).49 In a study of 74 NDMM patients who received high-dose chemotherapy with autologous stem cell transplant (ASCT), patients who overexpressed cyclin-D1 had significantly longer duration of remission in comparison with patients who did not (41 vs. 26 months, respectively; p = 0.02) resulting in longer median event-free survival (33 vs. 24 months, respectively; p = 0.055). The risk for progression after bortezomib treatment was significantly decreased (HR 0.102, 95% CI 0.021-0.498, p = 0.0048) and progression-free survival prolonged (p = 0.0011).50 Palbociclib (C24H29N7O2), an FDA approved drug to treat breast cancer works by selectively inhibiting CDK4/6. In a phase II study, to evaluate the safety and efficacy of palbociclib in combination with bortezomib and dexamethasone in RRMM, objective responses were achieved in 5 (20%) patients and 11 (44%) achieved stable disease.51

PI3K/mTOR inhibition

The PI3K/AKT/mTOR pathway guards tumor growth and is involved in proliferation, survival and drug resistance in MM cells. This pathway also mediates the formation and activity of bone-forming osteoblasts and bone-resorbing osteoclasts.52 IL-6 can directly activate the proliferative effects of PI3K53and activate AKT hence playing an important role in MM pathogenesis.54,55

Dysregulated activation of mTOR signaling pathway is considered to be associated with drug resistance and poor prognosis of many cancers, including MM. 56 mTOR coordinates cell growth and proliferation in response to inputs from growth factors, nutrient status and energy stress, thus regulating cell cycle progress and survival.56 mTORC1 is a key modulator in MM cell proliferation, tumor development and chemoresistance. Disruption of this signaling could lead to MM cell apoptosis, tumor regression, and improved survival of MM patients.57

Feng et al. researched the effects of an organic compound, silybin, which decreased proliferation and ultimately led to apoptosis of MM cell lines by inhibiting the expression of PI3K, p-AKT and mTOR.58 With in vitro studies, a novel PI3K and mTOR inhibitor, BEZ235, showed potent antitumor effects as well as decreased osteoclast and increased osteoblast production and activity, respectively. This property of BEZ235 can be utilized against osteolytic lesions in MM patients.52 Oral PI3K/AKT inhibitors C98 and C96, have also been shown to inhibit proliferation and induce apoptosis of MM cells in both in vitro and xenograft studies. Importantly, the agents were very well tolerated in the animal studies.59,60

Clioquinol (5-chloro-7-iodo-8-hydroxyquinoline) is an anti-microbial drug, metal chelator, and potential anticancer drug. Clioquinol inhibits mTOR activity by disrupting the integrity of mTORC1 in MM.57 In preclinical studies, Clioquinol induced autophagy in leukemia and myeloma cells. Clioquinol has been shown to induce autophagy in association with the increase of the PI3KC3/Beclin 1 complex and dissociation of Beclin 1/Bcl-2.61 A phase I trial of Clioquinol was performed in patients with advanced hematological malignancies, including MM, identifying neuropathy and abdominal pain as dose limiting toxicities (NCT00963495).

A novel small molecule SC-06 also disrupts the mTOR signaling pathway, inducing cell apoptosis. In preclinical studies, SC06 showed promising results by disrupting mTOR signaling thus decreasing tumor volume in mouse xenograft models of MM.56 There have been no clinical trials to date with SC-06.

Histone deacetylase inhibition

Histone deacetylases (HDAC) modulate the organization of chromatin, thereby functioning as a controller over gene transcription. In MM, HDAC function is dysregulated, sometimes resulting in the upregulation of oncogenes. HDAC inhibitors represent a new therapeutic class in MM treatment that have the potential to control proliferation, differentiation, cell cycle arrest, and apoptosis. HDAC inhibitors are continuing to emerge as a promising therapy to inhibit MMSET complex assembly, because MMSET associates with HDACs in a large complex.6264 Several preclinical investigations and clinical trials have demonstrated the antimyeloma activity of HDAC inhibitors.65 In preclinical settings, HDAC inhibitors (panobinostat, vorinostat and romidepsin) showed remarkable anti-MM activities as single agents. However, they showed only modest clinical activity in cases of relapsed or refractory MM.6668 HDAC inhibitors have been clinically evaluated in combination with other agents, especially with proteasome inhibitors. Clinical efficacy of vorinostat has been studied in combination with bortezomib in phase I trials,69,70 and subsequently in phase II and III trials (Vorinostat Clinical Trials in Hematologic and Solid Malignancies or VANTAGE trials).71,72 In the phase II VANTAGE 095 trial, 17 % ORR and 31% clinical benefit rate (CBR) were observed. The phase III VANTAGE 088 trial showed only a modest statistically significant difference in median progression free survival (PFS). The combination of panobinostat with bortezomib has been studied in RRMM patients in a phase Ib study73 and later in phase II and III.74,75 In phase II PANORAMA trial, the ORR was 34.5 % and the CBR was 52.7 %. In the randomized, double-blind phase III PANORAMA trial, the combination of panobinostat with bortezomib improved median PFS. The results of the phase III PANORAMA trial resulted in the FDA approval of panobinostat in combination with bortezomib and dexamethasone. A HDAC6 selective inhibitor ricolinostat has also been studied as monotherapy and in combination with bortezomib and dexamethasone.76,77 This combination showed ORR of 29% and CBR of 39% while in bortezomib refractory patients, ORR was 14% in all combination doses.77

B). Emerging Targets: Pathways for which only preclinical data exist, but there is a potential to develop future targeted therapies.

JAK-STAT inhibition

JAK2 (Janus kinase 2) is a tyrosine kinase that binds to cytokine receptors. It is thought that autocrine and paracrine IL-6 leads to upregulation of the JAK2 pathway.78 Upon binding of cytokine IL-6 to the receptor, the receptor will dimerize and allow JAK2 to cross phosphorylate the receptors. Once phosphorylated these act as docking site for STAT3 to bind and influence gene expression as a transcription factor.79 Inhibition of the IL-6 receptor and STAT3 pathway has been reported to induce apoptosis in various myeloma cell lines.80,81 JAK2 pathway activation in MM leads to increased BCL-XL expression, which protects cells from chemotherapy-induced cell death.82 Interestingly, the JAK2 V617F mutation which is commonly found in myeloproliferative diseases was not found in 93 MM cases.83 JAK pathway inhibitors, including ruxolitinib, act by binding to the cytokine receptor and preventing JAK2 association, thereby decreasing activity.84 Zhang et al identified a JAK2/STAT3 inhibitor, SC99, that displays potent activity against MM cells by decreasing their proliferation and increasing MM cell apoptosis in both in vitro and MM xenograft models in mice.85

Proteasome inhibition of NF-κB pathway

The NF-kB pathway is primarily involved in DNA transcription, production of cytokines, cell proliferation and ultimately cell survival. NF-kB is thought to play an important role in the pathogenesis of MM, as the pathway becomes constitutively active, a potential mechanism by which tumor cells function independently of the marrow microenvironment.86 Dysregulation of the NF-kB pathway has led to the enhanced proliferation of cancerous cells and an increased likelihood of tumor development. Additionally, NF-kB pathway activation leads to MM lytic bone disease by activating osteoclasts.87,88 NF-kB also regulates cyclin D1expression.89,90

Proteasome inhibitors91, which partially act by inhibiting NF-kB, have shown great clinical success in NDMM as well as RRMM.92 DANFIN (N,N’-bis-(2,4-dimethyl-phenyl)-ethane-1,2-diamine) has also been shown to inhibit NF-κB activation and signaling. In preclinical studies, Uematsu et al. treated a MM xenograft mouse model with DANFIN plus bortezomib; this combination decreased tumor weight by more than 60% and enhanced the apoptosis of MM cells.93

MDM2 inhibition

TP53 is a tumor suppressor gene that promotes normal cellular proliferation, differentiation and apoptosis. Murine double minute (MDM) 2 is a pleiotropic protein that functions as an E3 ubiquitin ligase that limits the accumulation and function of TP53.94 It does so by helping ubiquitination of TP53 and thus facilitating its proteasome-mediated degradation. It also binds to TP53 amino acids 15–29, thereby limiting TP53 transcription.94 MDM2 is a rational target for TP53 mutated MM.95 Over-expression of MDM2 is seen in 0-20% of MM patient population 9698 and its overexpression is associated with increased proliferation and survival of myeloma cells, partly due to down-regulation of cyclin-dependent kinase inhibitor p21.99 Nutlins are potent and selective small molecule antagonists of MDM2. They function by binding to MDM2 in the p53 binding pocket, releasing p53 from negative control. Preclinical data have demonstrated nutlin-induced apoptosis in MM cells, supporting further investigation of this therapeutic intervention in MM100.

c-MYC/WNT

Pathologic activation of c-MYC has a key role in cancer development by upregulating the transcriptional program of the cell, enhancing cell proliferation.101,102 Amplification, translocation or rearrangement of MYC is among the most common genetic alterations observed in cancer genomes, including MM103,104 where activation is found in more than 60% of patient-derived MM cells.105 Increased activity of MYC upregulates ribosomal biogenesis and translation.

MYC inhibition

With a high-throughput screen, Manier et al. determined rocaglate derivatives are active against MM. Rocaglates are plant-derived cytotoxic compounds known to inhibit protein synthesis and can repress translation of specific messenger RNAs. Rocaglates inhibit a specific translation oncogenic program related to high expression of MYC, with potent in vitro and in vivo activity in preclinical studies. Due to their specificity, targeting dysregulated translation initiation with rocaglates rather than targeting the elongation machinery with other translation inhibitors might be less toxic to normal tissues.106

WNT inhibition

Deletion or mutation of the tumor suppressor gene encoding the deubiquitinating enzyme CYLD is a common genomic aberration in MM. CYLD acts as a negative regulator of NF-κB and WNTβ-catenin signaling and loss of CYLD sensitizes MM cells to NF-κB-stimuli and WNT ligands. In NDMM, low CYLD expression is strongly correlated with proliferation and a WNT signaling-gene expression signature.107 In a preclinical study, Schmmel et al demonstrated that targeting WNT signaling with ethacrynic acid and ciclopirox olamine with piceatannol had a synergistic effect, increasing cytotoxicity in myeloma cells with minimal effect on healthy cells.108 Given these early preclinical results, targeting the WNT pathway is a potential therapeutic strategy in MM with loss of CYLD activity.

Histone methyltransferase inhibition

MMSET, also known as Wolf-Hirschhorn syndrome candidate 1 (WHSC1) or nuclear receptor-binding SET domain 2 (NSD2), is a member of the nuclear receptor binding SET domain (NSD) histone methyltransferase (HMT) family, which also includes NSD1 and NSD3.109111 NSDs are histone modifiers that maintain chromatin, methylating histone H3 lysine 36 (H3K36) and histone H4 lysine 20 (H4K20). Multiple myeloma SET domain (MMSET) is specifically over-expressed in 15% of MM cases due to chromosomal translocation of t (4;14); putting the MMSET locus under the control of the Ig heavy chain promoter.112 As a lysine histone methyltransferase, MMSET regulates the methylation of histones, thereby causing global changes to the chromatin state and subsequent gene expression in t(4;14) MM cells.

Min et al. showed that MMSET stimulates cell growth and enhances the expression of cMYC protein in t(4;14) cells.112 Various studies have shown that when MMSET expression is inhibited, there is reduction of proliferation, induction of apoptosis and alteration of cell adhesion.113115 Due to alternative splicing and differential promoter usage, the MMSET locus produces several different transcripts and is also overexpressed in many other cancers including: pediatric acute lymphoblastic leukemia, lung, prostate, bladder and other solid tumors.116

Di Luccio et al. discovered a small molecule, LEM-06, which inhibits the H3K36 methylation action of MMSET by putatively blocking the histone binding pocket.117 Tisi et al. demonstrated that N-alkyl sinefungin derivatives also assert low level inhibition of MMSET. 116 Though these compounds have not yet progressed to clinical trial, preclinical research suggests the feasibility of these products or their derivatives to inhibit epigenetic remodeling mechanisms driving MM progression.

BET inhibition

The bromodomain and extra terminal (BET) family of proteins can bind to acetylated chromatin and regulate gene transcription. These molecules were first studied in myeloma and were found to repress c-MYC expression which resulted in significant downregulation of c-MYC regulated genes. A bromodomain (BRD) is an approximately 110 amino acid protein domain that recognizes acetylated lysine residues, such as those on the N-terminal tails of histones and is responsible for transducing the signal carried by acetylated lysine residues and recruiting different molecular partners, including chromatin factors and transcriptional machinery.118 Histone acetylation is a critical process of chromatin remodeling that underlies the open chromatin architecture dynamics and activation of transcription.119,120 Histone acetyltransferases (HATs) and histone deacetylases (HDACs) catalyze the addition and removal of acetyl groups on lysine residues of histones and other regulatory proteins, then BRDs recognize the relaxed chromatin segments and bind to the acetylated nucleosomes, transcription factors and co-activators.121,122,123

Given this, inhibitors of BRDs have demonstrated positive effects in both solid and hematologic malignancies.124 The selective small molecule bromodomain inhibitor, JQ-1 has shown to selectively inhibit the interaction between BET proteins and acetylated chromatin, resulting in dissociation. It was demonstrated to inhibit proliferation when tested against a panel of 25 MM cell lines and primary MM samples. The molecule was similarly tested in a xenograft mouse model of MM where treatment with JQ-1 resulted in decreased tumor burden, reduced immunoglobulin expression and increased overall survival.125 Jake et al. showed that BET inhibition by JQ-1 downregulates MYC and inhibits MM cell proliferation including MM cell lines that are resistant to FDA approved drugs like dexamethasone and melphalan.125 Another BET inhibitor, CPI-0610, is currently in phase I testing and preliminary results show that human MM cell lines are sensitive to BET inhibition. CPI-0610 induces apoptosis, G1 cell cycle arrest and caspase-dependent cell death associated with inhibition of MYC, IKZF1 and IRF4.126,127 Boi et al. demonstrated the antiproliferative activity of BET inhibitor OTX015, in a preclinical study using multiple myeloma cells.128 OTX015 acts via binding to BRD motifs and is currently in phase I trials.129,130

BET inhibition by CPI203 increases lenalidomide/dexamethasone efficacy in MM cell lines. Anti-myeloma activity of CPI203 is independent of cell sensitivity to lenalidomide and involves MYC and IKZF1 downregulation.131 There is a synergistic effect of CPI-203 on cell death induction which is followed by the reduced expression of MYC and IRF4.132 CPI-203 and bortezomib have been shown to have synergistic effects in drug resistant myeloma.133 In MM cells, a novel inhibitor CG13250 is capable of suppressing the MYC transcription by impeding BRD4 binding to the MYC promoter.134

Epigenetic Remodeling

The dysregulation of epigenetics is increasingly recognized in the pathogenesis of hematological malignancies and recently a number of epigenetic therapies targeting DNA methylation, acetylation, and posttranslational histone modification have been introduced in clinical practice.135

Conclusion

Recent advancements in preclinical and clinical research have identified novel aberrant molecular pathways in MM and by targeting these pathways, we could develop treatment modalities for achieving better results with less side effects. Targets for further clinical development include BCL-2, MDM2, KRAS, NRAS, BRAF MEK, JAK-STAT, NF-kB, c-MYC/Wnt, PI3K/Akt/mTOR, Cyclin D, MMSET and BET. Cobimetinib (MEK) and vorinostat (Histone deacetylase inhibitor) are currently in phase 3 drug testing. Phase 2 trials of Selumetinib (MEK1/2 inhibitor) and palbociclib (CDK4/6 inhibitor) are underway while venetoclax (a BCL-2 inhibitor) is in phase 1 testing. In various preclinical studies, vemurafenib (BRAF inhibitor), SC99 (JAK2/STAT3 inhibitor), DANFIN (NF-κB), BEZ235 (P13K and mTOR inhibitor), and clioquinol (mTOR inhibitor) displayed potent anti-myeloma activity. Monoclonal antibodies targeting FGFR also have potential in the treatment of MM. Development of targeted therapies against these pathways offer promise for the future of precision and personalized treatment of MM.

Biography:

FA is an MD and Associate Professor of Medicine at the Taussig Cancer Center, Department of Hematology, Medical Oncology, Stem Cell Transplantation and Multiple Myeloma Program Cleveland Clinic, Cleveland, Ohio.

KMG completed his MS in Molecular and Cellular Biology. He is pharmacy student at University of Arizona.

AI is an MD and currently in PGY1 at the University of Arizona Internal Medicine Residency Program Tucson.

MB is an MD, currently working as hospitalist in Internal Medicine at Summit Media.

ADR has an MD/PhD in Genetics and is currently in PGY2 at the University of Arizona Internal Medicine Residency Program Tucson.

SS is a graduate student at the University of Arizona College of Public Health. She is also working as a scribe at Urological Associates of Southern Arizona and a research specialist at BUMC trauma surgery center.

MAZ completed his MBBS from Rawalpindi Medical College Pakistan. He is currently a research assistant at the University of Arizona Cancer Center.

FR completed her MBBS degree from Foundation University Medical College Pakistan, she is currently working in hospital medicine.

JC has a PhD and is working as Co-Director of Hematologic Malignancy Disease Team at The University of Arizona Cancer Center.

SN has a PhD and is Research Director of Medical Oncology at The University of Arizona Cancer Center.

HAK is an Associate Professor of Pathology. He is the Director of Laboratory for Genetics and Genomics (LGG), Division of Human Genetics at Children’s Hospital Cincinnati.

NNC is an MD and currently in PGY2 at the University of Arizona Internal Medicine Residency Program Tucson.

AM has his PharmD and MS and is the Clinical Coordinator of Hematology/Oncology UACC.

JV is an MD and works at the Taussig Cancer Center, Department of Hematology, Medical Oncology, Cleveland Clinic, Cleveland, Ohio.

CS is a DO and works at the Taussig Cancer Center, Department of Hematology, Medical Oncology, Cleveland Clinic, Cleveland, Ohio.

Acknowledgements:

We acknowledge Ms. Judy Burdett for her assistance in writing and final editing of manuscript.

Financial disclosure statement: This work was supported by grant P30 CA023074 from the National Cancer Institute, National Institutes of Health, Bethesda, MD.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of interest statement: Dr. Anwer is on the speaker bureau of Incyte and received honoraria from Seattle genetics for advisory board participation. Other authors declare that there is no conflict of interest with this manuscript. The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties. No writing assistance was utilized in the production of this manuscript.

References:

  • 1.Van Wier S, Braggio E, Baker A, et al. Hypodiploid multiple myeloma is characterized by more aggressive molecular markers than non-hyperdiploid multiple myeloma. Haematologica. 2013;98(10):1586–1592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Debes-Marun CS, Dewald GW, Bryant S, et al. Chromosome abnormalities clustering and its implications for pathogenesis and prognosis in myeloma. Leukemia. 2003;17(2):427–436. [DOI] [PubMed] [Google Scholar]
  • 3.Smadja NV, Bastard C, Brigaudeau C, Leroux D, Fruchart C. Hypodiploidy is a major prognostic factor in multiple myeloma. Blood. 2001;98(7):2229–2238. [DOI] [PubMed] [Google Scholar]
  • 4.Rajkumar SV. Multiple Myeloma: 2016 update on Diagnosis, Risk-stratification and Management. American journal of hematology. 2016;91(7):719–734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Mikhael JR, Dingli D, Roy V, et al. Management of newly diagnosed symptomatic multiple myeloma: updated Mayo Stratification of Myeloma and Risk-Adapted Therapy (mSMART) consensus guidelines 2013. Paper presented at: Mayo Clinic Proceedings2013. [DOI] [PubMed] [Google Scholar]
  • 6.Palumbo A, Avet-Loiseau H, Oliva S, et al. Revised international staging system for multiple myeloma: a report from International Myeloma Working Group. Journal of Clinical Oncology. 2015;33(26):2863–2869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Cory S, Adams JM. Killing cancer cells by flipping the Bcl-2/Bax switch. Cancer cell. 2005;8(1):5–6. [DOI] [PubMed] [Google Scholar]
  • 8.Moreau P, Chanan-Khan A, Roberts AW, et al. Promising efficacy and acceptable safety of venetoclax plus bortezomib and dexamethasone in relapsed/refractory MM. Blood. 2017;130(22):2392–2400. [DOI] [PubMed] [Google Scholar]
  • 9.Kumar S, Kaufman JL, Gasparetto C, et al. Efficacy of venetoclax as targeted therapy for relapsed/refractory t (11; 14) multiple myeloma. Blood. 2017;130(22):2401–2409. [DOI] [PubMed] [Google Scholar]
  • 10.Walker BA, Boyle EM, Wardell CP, et al. Mutational spectrum, copy number changes, and outcome: results of a sequencing study of patients with newly diagnosed myeloma. Journal of clinical oncology. 2015;33(33):3911–3920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Chng W, Gonzalez-Paz N, Price-Troska T, et al. Clinical and biological significance of RAS mutations in multiple myeloma. Leukemia. 2008;22(12):2280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Rasmussen T, Kuehl M, Lodahl M, Johnsen HE, Dahl IMS. Possible roles for activating RAS mutations in the MGUS to MM transition and in the intramedullary to extramedullary transition in some plasma cell tumors. Blood. 2005;105(1):317–323. [DOI] [PubMed] [Google Scholar]
  • 13.Chesi M, Brents LA, Ely SA, et al. Activated fibroblast growth factor receptor 3 is an oncogene that contributes to tumor progression in multiple myeloma. Blood. 2001;97(3):729–736. [DOI] [PubMed] [Google Scholar]
  • 14.Ogata A, Chauhan D, Teoh G, et al. IL-6 triggers cell growth via the Ras-dependent mitogen-activated protein kinase cascade. The Journal of Immunology. 1997;159(5):2212–2221. [PubMed] [Google Scholar]
  • 15.Xu J, Pfarr N, Endris V, et al. Molecular signaling in multiple myeloma: association of RAS/RAF mutations and MEK/ERK pathway activation. Oncogenesis. 2017;6(5):e337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Mulligan G, Lichter DI, Di Bacco A, et al. Mutation of NRAS but not KRAS significantly reduces myeloma sensitivity to single-agent bortezomib therapy. Blood. 2014;123(5):632–639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Annunziata CM, Hernandez L, Davis RE, et al. A mechanistic rationale for MEK inhibitor therapy in myeloma based on blockade of MAF oncogene expression. Blood. 2011;117(8):2396–2404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Popovic R, Licht JD. MEK and MAF in myeloma therapy. Blood. 2011;117(8):2300–2302. [DOI] [PubMed] [Google Scholar]
  • 19.Ocio E, Fernández-Lázaro D, San-Segundo L, et al. In vivo murine model of acquired resistance in myeloma reveals differential mechanisms for lenalidomide and pomalidomide in combination with dexamethasone. Leukemia. 2015;29(3):705. [DOI] [PubMed] [Google Scholar]
  • 20.Holkova B, Zingone A, Kmieciak M, et al. A Phase 2 Trial of AZD6244 (Selumetinib, ARRY-142886), an Oral MEK1/2 Inhibitor, in Relapsed/Refractory Multiple Myeloma. Clinical cancer research : an official journal of the American Association for Cancer Research. 2016;22(5):1067–1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Srkalovic G, Hussein MA, Hoering A, et al. A phase II trial of BAY 43-9006 (sorafenib) (NSC-724772) in patients with relapsing and resistant multiple myeloma: SWOG S0434. Cancer Med. 2014;3(5):1275–1283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Chapman MA, Lawrence MS, Keats JJ, et al. Initial genome sequencing and analysis of multiple myeloma. Nature. 2011;471(7339):467–472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Walker BA, Wardell CP, Melchor L, et al. Intraclonal heterogeneity and distinct molecular mechanisms characterize the development of t(4;14) and t(11;14) myeloma. Blood. 2012;120(5):1077–1086. [DOI] [PubMed] [Google Scholar]
  • 24.Andrulis M, Lehners N, Capper D, et al. Targeting the BRAF V600E mutation in multiple myeloma. Cancer Discov. 2013;3(8):862–869. [DOI] [PubMed] [Google Scholar]
  • 25.Samuel J, Macip S, Dyer MJ. Efficacy of vemurafenib in hairy-cell leukemia. New England Journal of Medicine. 2014;370(3):286–288. [DOI] [PubMed] [Google Scholar]
  • 26.Chapman PB, Robert C, Larkin J, et al. Vemurafenib in patients with BRAFV600 mutation-positive metastatic melanoma: final overall survival results of the randomized BRIM-3 study. Annals of Oncology. 2017;28(10):2581–2587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Mey UJ, Renner C, von Moos R. Vemurafenib in combination with cobimetinib in relapsed and refractory extramedullary multiple myeloma harboring the BRAF V600E mutation. Hematol Oncol. 2016. [DOI] [PubMed] [Google Scholar]
  • 28.Larkin J, Ascierto PA, Dreno B, et al. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. New England Journal of Medicine. 2014;371(20):1867–1876. [DOI] [PubMed] [Google Scholar]
  • 29.Richardson P, Roy A, Acharyya S, et al. Treatment-free interval as a metric of patient experience and a health outcome of value for advanced multiple myeloma: the case for the histone deacetylase inhibitor panobinostat, a next-generation novel agent. Expert Review of Hematology. 2017;10(10):933–939. [DOI] [PubMed] [Google Scholar]
  • 30.Chesi M, Nardini E, Brents LA, et al. Frequent translocation t(4;14)(p16.3;q32.3) in multiple myeloma is associated with increased expression and activating mutations of fibroblast growth factor receptor 3. Nat Genet. 1997;16(3):260–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Trudel S, Stewart AK, Rom E, et al. The inhibitory anti-FGFR3 antibody, PRO-001, is cytotoxic to t(4;14) multiple myeloma cells. Blood. 2006;107(10):4039–4046. [DOI] [PubMed] [Google Scholar]
  • 32.Qing J, Du X, Chen Y, et al. Antibody-based targeting of FGFR3 in bladder carcinoma and t(4;14)-positive multiple myeloma in mice. J Clin Invest. 2009;119(5):1216–1229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Trudel S, Bergsagel PL, Singhal S, et al. A Phase I Study of the Safety and Pharmacokinetics of Escalating Doses of MFGR1877S, a Fibroblast Growth Factor Receptor 3 (FGFR3) Antibody, in Patients with Relapsed or Refractory t(4;14)-Positive Multiple Myeloma. Blood. 2012;120(21):4029–4029. [Google Scholar]
  • 34.Trudel S, Li ZH, Wei E, et al. CHIR-258, a novel, multitargeted tyrosine kinase inhibitor for the potential treatment of t(4;14) multiple myeloma. Blood. 2005;105(7):2941–2948. [DOI] [PubMed] [Google Scholar]
  • 35.Scheid C, Reece D, Beksac M, et al. Phase 2 study of dovitinib in patients with relapsed or refractory multiple myeloma with or without t(4;14) translocation. Eur J Haematol. 2015;95(4):316–324. [DOI] [PubMed] [Google Scholar]
  • 36.Majumder MM, Silvennoinen R, Anttila P, et al. Identification of precision treatment strategies for relapsed/refractory multiple myeloma by functional drug sensitivity testing. Oncotarget. 2017;8(34):56338–56350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gavine PR, Mooney L, Kilgour E, et al. AZD4547: an orally bioavailable, potent, and selective inhibitor of the fibroblast growth factor receptor tyrosine kinase family. Cancer Res. 2012;72(8):2045–2056. [DOI] [PubMed] [Google Scholar]
  • 38.Xie L, Su X, Zhang L, et al. FGFR2 gene amplification in gastric cancer predicts sensitivity to the selective FGFR inhibitor AZD4547. Clin Cancer Res. 2013;19(9):2572–2583. [DOI] [PubMed] [Google Scholar]
  • 39.Velasquez MP, Bonifant CL, Gottschalk S. Redirecting T cells to hematological malignancies with bispecific antibodies. Blood. 2018;131(1):30–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Terpos E, Gobbi M, Potamianou A, Lahaye M, Couturier C, Cavo M. Retreatment and prolonged therapy with subcutaneous bortezomib in patients with relapsed multiple myeloma: A randomized, controlled, phase III study. European Journal of Haematology. 2018;100(1):10–19. [DOI] [PubMed] [Google Scholar]
  • 41.Bergsagel PL, Kuehl WM, Zhan F, Sawyer J, Barlogie B, Shaughnessy J. Cyclin D dysregulation: an early and unifying pathogenic event in multiple myeloma. Blood. 2005;106(1):296–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Misiewicz-Krzeminska I, Sarasquete ME, Vicente-Dueñas C, et al. Post-transcriptional modifications contribute to the upregulation of cyclin D2 in multiple myeloma. Clinical Cancer Research. 2016;22(1):207–217. [DOI] [PubMed] [Google Scholar]
  • 43.Barille-Nion S, Barlogie B, Bataille R, et al. Advances in biology and therapy of multiple myeloma. Hematology Am Soc Hematol Educ Program. 2003:248–278. [PubMed] [Google Scholar]
  • 44.Specht K, Haralambieva E, Bink K, et al. Different mechanisms of cyclin D1 overexpression in multiple myeloma revealed by fluorescence in situ hybridization and quantitative analysis of mRNA levels. Blood. 2004;104(4):1120–1126. [DOI] [PubMed] [Google Scholar]
  • 45.Fonseca R, Witzig TE, Gertz MA, et al. Multiple myeloma and the translocation t (11; 14)(q13; q32): a report on 13 cases. British journal of haematology. 1998;101(2):296–301. [DOI] [PubMed] [Google Scholar]
  • 46.Laï JL, Michaux L, Dastugue N, et al. Cytogenetics in multiple myeloma: a multicenter study of 24 patients with t (11; 14)(q13; q32) or its variant. Cancer genetics and cytogenetics. 1998;104(2):133–138. [DOI] [PubMed] [Google Scholar]
  • 47.Mateo G, Mateos M, Rosinol L, Montalban M, Lopez-Berges C, Bladé J. Prognostic influence of antigenic markers in 587 multiple myeloma patients uniformly treated with high dose therapy. Haematologica. 2005;90(s1):103. [Google Scholar]
  • 48.Cook JR, Hsi ED, Worley S, Tubbs RR, Hussein M. Immunohistochemical analysis identifies two cyclin D1+ subsets of plasma cell myeloma, each associated with favorable survival. American journal of clinical pathology. 2006;125(4):615–624. [DOI] [PubMed] [Google Scholar]
  • 49.Dawson MA, Opat SS, Taouk Y, et al. Clinical and immunohistochemical features associated with a response to bortezomib in patients with multiple myeloma. Clinical Cancer Research. 2009;15(2):714–722. [DOI] [PubMed] [Google Scholar]
  • 50.Ngo BT, Felthaus J, Hein M, et al. Monitoring bortezomib therapy in multiple myeloma: screening of cyclin D1, D2, and D3 via reliable real-time polymerase chain reaction and association with clinico-pathological features and outcome. Leuk Lymphoma. 2010;51(9):1632–1642. [DOI] [PubMed] [Google Scholar]
  • 51.Niesvizky R, Badros AZ, Costa LJ, et al. Phase 1/2 study of cyclin-dependent kinase (CDK)4/6 inhibitor palbociclib (PD-0332991) with bortezomib and dexamethasone in relapsed/refractory multiple myeloma. Leuk Lymphoma. 2015;56(12):3320–3328. [DOI] [PubMed] [Google Scholar]
  • 52.Gan ZY, Fitter S, Vandyke K, To LB, Zannettino AC, Martin SK. The effect of the dual PI3K and mTOR inhibitor BEZ235 on tumour growth and osteolytic bone disease in multiple myeloma. Eur J Haematol. 2015;94(4):343–354. [DOI] [PubMed] [Google Scholar]
  • 53.Tu Y, Gardner A, Lichtenstein A. The phosphatidylinositol 3-kinase/AKT kinase pathway in multiple myeloma plasma cells: roles in cytokine-dependent survival and proliferative responses. Cancer Research. 2000;60(23):6763–6770. [PubMed] [Google Scholar]
  • 54.Hideshima T, Nakamura N, Chauhan D, Anderson KC. Biologic sequelae of interleukin-6 induced PI3-K/Akt signaling in multiple myeloma. Oncogene. 2001;20(42):5991. [DOI] [PubMed] [Google Scholar]
  • 55.Hsu J-h, Shi Y, Hu L, Fisher M, Franke TF, Lichtenstein A. Role of the AKT kinase in expansion of multiple myeloma clones: effects on cytokine-dependent proliferative and survival responses. Oncogene. 2002;21(9):1391. [DOI] [PubMed] [Google Scholar]
  • 56.Han K, Xu X, Xu Z, et al. SC06, a novel small molecule compound, displays preclinical activity against multiple myeloma by disrupting the mTOR signaling pathway. Scientific reports. 2015;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Li J, Zhu J, Cao B, Mao X. The mTOR signaling pathway is an emerging therapeutic target in multiple myeloma. Current pharmaceutical design. 2014;20(1):125–135. [DOI] [PubMed] [Google Scholar]
  • 58.Feng N, Luo J, Guo X. Silybin suppresses cell proliferation and induces apoptosis of multiple myeloma cells via the PI3K/Akt/mTOR signaling pathway. Mol Med Rep. 2016;13(4):3243–3248. [DOI] [PubMed] [Google Scholar]
  • 59.Zhu J, Wang M, Yu Y, et al. A novel PI3K inhibitor PIK-C98 displays potent preclinical activity against multiple myeloma. Oncotarget. 2015;6(1):185–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Tang J, Zhu J, Yu Y, et al. A virtual screen identified C96 as a novel inhibitor of phosphatidylinositol 3-kinase that displays potent preclinical activity against multiple myeloma in vitro and in vivo. Oncotarget. 2014;5(11):3836–3848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Cao B, Li J, Zhou X, et al. Clioquinol induces pro-death autophagy in leukemia and myeloma cells by disrupting the mTOR signaling pathway. Scientific reports. 2014;4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Todoerti K, Ronchetti D, Agnelli L, et al. Transcription repression activity is associated with the type I isoform of the MMSET gene involved in t(4;14) in multiple myeloma. Br J Haematol. 2005;131(2):214–218. [DOI] [PubMed] [Google Scholar]
  • 63.Kim JY, Kee HJ, Choe NW, et al. Multiple-myeloma-related WHSC1/MMSET isoform RE-IIBP is a histone methyltransferase with transcriptional repression activity. Mol Cell Biol. 2008;28(6):2023–2034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Marango J, Shimoyama M, Nishio H, et al. The MMSET protein is a histone methyltransferase with characteristics of a transcriptional corepressor. Blood. 2008;111(6):3145–3154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Smith EM, Boyd K, Davies FE. The potential role of epigenetic therapy in multiple myeloma. Br J Haematol. 2010;148(5):702–713. [DOI] [PubMed] [Google Scholar]
  • 66.Wolf JL, Siegel D, Goldschmidt H, et al. Phase II trial of the pan-deacetylase inhibitor panobinostat as a single agent in advanced relapsed/refractory multiple myeloma. Leukemia & lymphoma. 2012;53(9):1820–1823. [DOI] [PubMed] [Google Scholar]
  • 67.Richardson P, Mitsiades C, Colson K, et al. Phase I trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) in patients with advanced multiple myeloma. Leukemia & lymphoma. 2008;49(3):502–507. [DOI] [PubMed] [Google Scholar]
  • 68.Niesvizky R, Ely S, Mark T, et al. Phase 2 trial of the histone deacetylase inhibitor romidepsin for the treatment of refractory multiple myeloma. Cancer. 2011;117(2):336–342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Weber DM, Graef T, Hussein M, et al. Phase I trial of vorinostat combined with bortezomib for the treatment of relapsing and/or refractory multiple myeloma. Clinical lymphoma, myeloma & leukemia. 2012;12(5):319–324. [DOI] [PubMed] [Google Scholar]
  • 70.Badros A, Burger AM, Philip S, et al. Phase I study of vorinostat in combination with bortezomib for relapsed and refractory multiple myeloma. Clin Cancer Res. 2009;15(16):5250–5257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.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, double-blind study. The Lancet Oncology. 2013;14(11):1129–1140. [DOI] [PubMed] [Google Scholar]
  • 72.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. Clinical lymphoma, myeloma & leukemia. 2016;16(6):329–334 e321. [DOI] [PubMed] [Google Scholar]
  • 73.San-Miguel JF, Richardson PG, Gunther A, et al. Phase Ib study of panobinostat and bortezomib in relapsed or relapsed and refractory multiple myeloma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2013;31(29):3696–3703. [DOI] [PubMed] [Google Scholar]
  • 74.Richardson PG, Schlossman RL, Alsina M, et al. PANORAMA 2: panobinostat in combination with bortezomib and dexamethasone in patients with relapsed and bortezomib-refractory myeloma. Blood. 2013;122(14):2331–2337. [DOI] [PubMed] [Google Scholar]
  • 75.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. The Lancet Oncology. 2014;15(11):1195–1206. [DOI] [PubMed] [Google Scholar]
  • 76.Vogl DT, Hari PN, Jagannath S, et al. ACY-1215, a Selective Histone Deacetylase (HDAC) 6 Inhibitor: Interim Results Of Combination Therapy With Bortezomib In Patients With Multiple Myeloma (MM). Blood. 2013;122(21):759–759.23616623 [Google Scholar]
  • 77.Vogl DT, Raje N, Jagannath S, et al. Ricolinostat, the first selective histone deacetylase 6 inhibitor, in combination with bortezomib and dexamethasone for relapsed or refractory multiple myeloma. Clin Cancer Res. 2017;23(13):3307–3315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Kawano M, Hirano T, Matsuda T, et al. Autocrine generation and requirement of BSF-2/IL-6 for human multiple myelomas. Nature. 1988;332(6159):83–85. [DOI] [PubMed] [Google Scholar]
  • 79.Shuai K The STAT family of proteins in cytokine signaling. Progress in biophysics and molecular biology. 1999;71(3):405–422. [DOI] [PubMed] [Google Scholar]
  • 80.Chatterjee M, Stühmer T, Herrmann P, Bommert K, Dörken B, Bargou RC. Combined disruption of both the MEK/ERK and the IL-6R/STAT3 pathways is required to induce apoptosis of multiple myeloma cells in the presence of bone marrow stromal cells. Blood. 2004;104(12):3712–3721. [DOI] [PubMed] [Google Scholar]
  • 81.Monaghan K, Khong T, Burns C, Spencer A. The novel JAK inhibitor CYT387 suppresses multiple signalling pathways, prevents proliferation and induces apoptosis in phenotypically diverse myeloma cells. Leukemia. 2011;25(12):1891. [DOI] [PubMed] [Google Scholar]
  • 82.Alas S, Bonavida B. Inhibition of constitutive STAT3 activity sensitizes resistant non-Hodgkin’s lymphoma and multiple myeloma to chemotherapeutic drug-mediated apoptosis. Clinical cancer research. 2003;9(1):316–326. [PubMed] [Google Scholar]
  • 83.Fiorini A, Farina G, Reddiconto G, et al. Screening of JAK2 V617F mutation in multiple myeloma. Leukemia. 2006;20(10):1912–1912. [DOI] [PubMed] [Google Scholar]
  • 84.Harrison C, Kiladjian J-J, Al-Ali HK, et al. JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. New England Journal of Medicine. 2012;366(9):787–798. [DOI] [PubMed] [Google Scholar]
  • 85.Zhang Z, Mao H, Du X, et al. A novel small molecule agent displays potent anti-myeloma activity by inhibiting the JAK2-STAT3 signaling pathway. Oncotarget. 2016;7(8):9296–9308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Godwin P, Baird A-M, Heavey S, Barr M, O’Byrne K, Gately K. Targeting Nuclear Factor-Kappa B to Overcome Resistance to Chemotherapy. Frontiers in Oncology. 2013;3(120). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ang E, Pavlos NJ, Rea SL, et al. Proteasome inhibitors impair RANKL-induced NF-κB activity in osteoclast-like cells via disruption of p62, TRAF6, CYLD, and IKBa signaling cascades. Journal of cellular physiology. 2009;220(2):450–459. [DOI] [PubMed] [Google Scholar]
  • 88.Hongming H, Jian H. Bortezomib inhibits maturation and function of osteoclasts from PBMCs of patients with multiple myeloma by downregulating TRAF6. Leukemia research. 2009;33(1):115–122. [DOI] [PubMed] [Google Scholar]
  • 89.Guttridge DC, Albanese C, Reuther JY, Pestell RG, Baldwin AS. NF-κB controls cell growth and differentiation through transcriptional regulation of cyclin D1. Molecular and cellular biology. 1999;19(8):5785–5799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Hinz M, Krappmann D, Eichten A, Heder A, Scheidereit C, Strauss M. NF-B function in growth control: regulation of cyclin D1 expression and G0/G1-to-S-phase transition. Molecular and cellular biology. 1999;19(4):2690–2698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Richardson PG, Barlogie B, Berenson J, et al. A phase 2 study of bortezomib in relapsed, refractory myeloma. New England Journal of Medicine. 2003;348(26):2609–2617. [DOI] [PubMed] [Google Scholar]
  • 92.Rajkumar SV, Hayman SR, Lacy MQ, et al. Combination therapy with lenalidomide plus dexamethasone (Rev/Dex) for newly diagnosed myeloma. Blood. 2005;106(13):4050–4053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Uematsu A, Kido K, Manabe E, et al. DANFIN functions as an inhibitor of transcription factor NF-kappaB and potentiates the antitumor effect of bortezomib in multiple myeloma. Biochem Biophys Res Commun. 2018;495(3):2289–2295. [DOI] [PubMed] [Google Scholar]
  • 94.Fahraeus R, Olivares-Illana V. MDM2’s social network. Oncogene. 2014;33(35):4365–4376. [DOI] [PubMed] [Google Scholar]
  • 95.Zhang B, Golding BT, Hardcastle IR. Small-molecule MDM2-p53 inhibitors: recent advances. Future Med Chem. 2015;7(5):631–645. [DOI] [PubMed] [Google Scholar]
  • 96.Quesnel B, Preudhomme C, Fournier J, Fenaux P, Peyrat JP. MDM2 gene amplification in human breast cancer. Eur J Cancer. 1994;30A(7):982–984. [DOI] [PubMed] [Google Scholar]
  • 97.Elnenaei MO, Gruszka-Westwood AM, A’Hernt R, et al. Gene abnormalities in multiple myeloma; the relevance of TP53, MDM2, and CDKN2A. Haematologica. 2003;88(5):529–537. [PubMed] [Google Scholar]
  • 98.Fonseca R, Barlogie B, Bataille R, et al. Genetics and cytogenetics of multiple myeloma: a workshop report. AACR; 2004. [DOI] [PubMed] [Google Scholar]
  • 99.Teoh G, Urashima M, Ogata A, et al. MDM2 protein overexpression promotes proliferation and survival of multiple myeloma cells. Blood. 1997;90(5):1982–1992. [PubMed] [Google Scholar]
  • 100.Saha MN, Jiang H, Jayakar J, Reece D, Branch DR, Chang H. MDM2 antagonist nutlin plus proteasome inhibitor velcade combination displays a synergistic anti-myeloma activity. Cancer Biol Ther. 2010;9(11):936–944. [DOI] [PubMed] [Google Scholar]
  • 101.Dang CV. MYC, microRNAs and glutamine addiction in cancers. Taylor & Francis; 2009. [DOI] [PubMed] [Google Scholar]
  • 102.Kim J, Chu J, Shen X, Wang J, Orkin SH. An extended transcriptional network for pluripotency of embryonic stem cells. Cell. 2008;132(6):1049–1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Beroukhim R, Mermel CH, Porter D, et al. The landscape of somatic copy-number alteration across human cancers. Nature. 2010;463(7283):899–905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Shou Y, Martelli ML, Gabrea A, et al. Diverse karyotypic abnormalities of the c-myc locus associated with c-myc dysregulation and tumor progression in multiple myeloma. Proceedings of the National Academy of Sciences. 2000;97(1):228–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Chng W, Huang G, Chung T, et al. Clinical and biological implications of MYC activation: a common difference between MGUS and newly diagnosed multiple myeloma. Leukemia. 2011;25(6):1026–1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Manier S, Salem KZ, Park J, Landau DA, Getz G, Ghobrial IM. Genomic complexity of multiple myeloma and its clinical implications. Nature Reviews Clinical Oncology. 2017;14(2):100–113. [DOI] [PubMed] [Google Scholar]
  • 107.van Andel H, Kocemba K, de Haan-Kramer A, et al. Loss of CYLD expression unleashes Wnt signaling in multiple myeloma and is associated with aggressive disease. Oncogene. 2017;36(15):2105–2115. [DOI] [PubMed] [Google Scholar]
  • 108.Schmeel LC, Schmeel FC, Kim Y, Endo T, Lu D, Schmidt-Wolf IG. Targeting the Wnt/beta-catenin pathway in multiple myeloma. Anticancer research. 2013;33(11):4719–4726. [PubMed] [Google Scholar]
  • 109.Huang N, vom Baur E, Garnier JM, et al. Two distinct nuclear receptor interaction domains in NSD1, a novel SET protein that exhibits characteristics of both corepressors and coactivators. EMBO J. 1998;17(12):3398–3412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Stec I, Wright TJ, van Ommen GJ, et al. WHSC1, a 90 kb SET domain-containing gene, expressed in early development and homologous to a Drosophila dysmorphy gene maps in the Wolf-Hirschhorn syndrome critical region and is fused to IgH in t(4;14) multiple myeloma. Hum Mol Genet. 1998;7(7):1071–1082. [DOI] [PubMed] [Google Scholar]
  • 111.Angrand PO, Apiou F, Stewart AF, Dutrillaux B, Losson R, Chambon P. NSD3, a new SET domain-containing gene, maps to 8p12 and is amplified in human breast cancer cell lines. Genomics. 2001;74(1):79–88. [DOI] [PubMed] [Google Scholar]
  • 112.Min D, Ezponda T, Kim M, et al. MMSET stimulates myeloma cell growth through microRNA-mediated modulation of c-MYC. Leukemia. 2013;27(3):686–694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.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(2):856–864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.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(1):211–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.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(1):78–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Tisi D, Chiarparin E, Tamanini E, et al. Structure of the Epigenetic Oncogene MMSET and Inhibition by N-Alkyl Sinefungin Derivatives. ACS Chemical Biology. 2016;11(11):3093–3105. [DOI] [PubMed] [Google Scholar]
  • 117.di Luccio E Inhibition of nuclear receptor binding SET domain 2/multiple myeloma SET domain by LEM-06 implication for epigenetic cancer therapies. Journal of cancer prevention. 2015;20(2):113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Ntranos A, Casaccia P. Bromodomains: Translating the words of lysine acetylation into myelin injury and repair. Neuroscience letters. 2016;625:4–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Bayarsaihan D Deciphering the Epigenetic Code in Embryonic and Dental Pulp Stem Cells. The Yale journal of biology and medicine. 2016;89(4):539–563. [PMC free article] [PubMed] [Google Scholar]
  • 120.Dutta A, Abmayr SM, Workman JL. Diverse Activities of Histone Acylations Connect Metabolism to Chromatin Function. Mol Cell. 2016;63(4):547–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Fu LL, Tian M, Li X, et al. Inhibition of BET bromodomains as a therapeutic strategy for cancer drug discovery. Oncotarget. 2015;6(8):5501–5516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Ferri E, Petosa C, McKenna CE. Bromodomains: Structure, function and pharmacology of inhibition. Biochemical pharmacology. 2016;106:1–18. [DOI] [PubMed] [Google Scholar]
  • 123.Taniguchi Y The Bromodomain and Extra-Terminal Domain (BET) Family: Functional Anatomy of BET Paralogous Proteins. International journal of molecular sciences. 2016;17(11). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Wadhwa E, Nicolaides T. Bromodomain Inhibitor Review: Bromodomain and Extra-terminal Family Protein Inhibitors as a Potential New Therapy in Central Nervous System Tumors. Cureus. 2016;8(5):e620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Delmore JE, Issa GC, Lemieux ME, et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell. 2011;146(6):904–917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Siu KT, Eda H, Santo L, et al. Effect of the BET inhibitor, Cpi-0610, alone and in combination with lenalidomide in multiple myeloma. Am Soc Hematology; 2015. [Google Scholar]
  • 127.Siu KT, Ramachandran J, Yee AJ, et al. Preclinical activity of CPI-0610, a novel small-molecule bromodomain and extra-terminal protein inhibitor in the therapy of multiple myeloma. Leukemia. 2017;31(8):1760–1769. [DOI] [PubMed] [Google Scholar]
  • 128.Boi M, Gaudio E, Bonetti P, et al. The BET Bromodomain inhibitor OTX015 affects pathogenetic pathways in pre-clinical B-cell tumor models and synergizes with targeted drugs. Clinical Cancer Research. 2015:clincanres. 1561.2014. [DOI] [PubMed] [Google Scholar]
  • 129.Noel JK, Iwata K, Ooike S, Sugahara K, Nakamura H, Daibata M. Abstract C244: Development of the BET bromodomain inhibitor OTX015. AACR; 2013. [Google Scholar]
  • 130.Amorim S, Stathis A, Gleeson M, et al. Bromodomain inhibitor OTX015 in patients with lymphoma or multiple myeloma: a dose-escalation, open-label, pharmacokinetic, phase 1 study. The Lancet Haematology. 2016;3(4):e196–e204. [DOI] [PubMed] [Google Scholar]
  • 131.Díaz T, Rodríguez V, Lozano E, et al. The BET bromodomain inhibitor CPI203 improves lenalidomide and dexamethasone activity in in vitro and in vivo models of multiple myeloma by blockade of Ikaros and MYC signaling. haematologica. 2017;102(10):1776–1784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Moros A, Rodriguez V, Saborit-Villarroya I, et al. Synergistic antitumor activity of lenalidomide with the BET bromodomain inhibitor CPI203 in bortezomib-resistant mantle cell lymphoma. Leukemia. 2014;28(10):2049–2059. [DOI] [PubMed] [Google Scholar]
  • 133.Siegel MB, Liu SQ, Davare MA, et al. Small molecule inhibitor screen identifies synergistic activity of the bromodomain inhibitor CPI203 and bortezomib in drug resistant myeloma. Oncotarget. 2015;6(22):18921–18932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Imayoshi N, Yoshioka M, Chauhan J, et al. CG13250, a novel bromodomain inhibitor, suppresses proliferation of multiple myeloma cells in an orthotopic mouse model. Biochem Biophys Res Commun. 2017;484(2):262–268. [DOI] [PubMed] [Google Scholar]
  • 135.Campbell RM, Tummino PJ. Cancer epigenetics drug discovery and development: the challenge of hitting the mark. J Clin Invest. 2014;124(1):64–69. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES