Progress in medical research has enhanced our understanding of tumor biology, delineated genetic and molecular mechanisms of tumor growth and survival, and defined the impact of the microenvironment in cancer pathogenesis. As a consequence of these advances, cancers deemed rapidly fatal only a few decades ago can now be treated effectively, with prolonged survival in an increasing proportion of patients. This is particularly true for multiple myeloma (MM), in which the introduction of drugs targeting the tumor in its microenvironment, such as the proteasome inhibitor bortezomib and the immunomodulatory drugs (IMiDs) thalidomide and lenalidomide, into initial, consolidation, maintenance, and salvage therapies has markedly improved patient outcome. In this perspective, we discuss the most promising therapies to even further improve MM treatment, with a focus on drugs inhibiting the ubiquitin-proteasome pathway; histone deacetylase (HDAC) inhibitors (HDACIs); immune therapies including IMiDs, monoclonal antibodies (mAbs), immune checkpoint inhibitors, agents targeting accessory plasmacytoid dendritic cells (pDCs), vaccines, and chimeric antigen receptor–engineered T (CAR-T) cells; drugs targeting tumor cell homing to, and exploiting hypoxia in, the bone marrow (BM) microenvironment; molecularly targeted therapies against kinesin spindle protein (KSP), v-akt murine thymoma viral oncogene homolog 1 (AKT), exportin 1 (XPO1), cyclin-dependent kinases (CDKs), bromodomain and extraterminal (BET) bromodomain 4, and serine/threonine kinase 4 (STK4); as well as delineating the impact of genomics on MM therapy. These advances in understanding the biology of MM will allow for earlier treatment of patients using rationally informed combination therapies with curative potential.

Melphalan plus prednisone treatment of MM was introduced in the 1960s and achieved median survival of 2 to 3 years.1  High-dose IV melphalan followed by autologous hematopoietic stem cell transplant (ASCT) was pioneered in the 1970s, with the first randomized trial of high-dose chemotherapy followed by ASCT vs conventional chemotherapy showing a 5-year overall survival (OS) rate of 52% vs 12%, respectively, in the 1990s.2  Remarkably, over the last decade, the introduction of novel agents targeting MM in the context of the BM microenvironment has transformed the MM treatment paradigm and markedly improved patient outcome.3  Landmark studies of the IMiDs thalidomide and lenalidomide and the proteasome inhibitor (PI) bortezomib provided the basis for rapid US Food and Drug Administration (FDA) approval of these treatments for patients with MM.4-6  Incorporation of combination novel agents into the ASCT algorithm as induction, consolidation, and maintenance therapy has resulted in unprecedented overall response rates (ORRs) and a threefold increase in OS.7  In this perspective, we focus on the targeted therapies that, in our view, hold the greatest potential to even further improve on this progress (Table 1 outlines investigational agents in advanced clinical development).

Table 1

Promising investigational agents in advanced clinical development in MM

Drug classDrug nameRelevant pharmacokinetic/ pharmacodynamic propertiesMechanisms of effectivenessMost advanced phase of developmentStudy design
PI Ixazomib (MLN9708) Orally bioavailable, reversible, boronate-based inhibitor of CT-L proteasome activity PolyUb protein accumulation Phase 3 Ixazomib/Len/Dex vs placebo/Len/Dex in RRMM 
Caspase-8– and caspase-9–mediated apoptosis Ixazomib/Len/Dex vs placebo/Len/Dex in transplant-ineligible NDMM 
p53 and p21 upregulation  
Terminal UPR induction  
miR33b upregulation  
PIM1 downregulation  
Marizomib (NPI-0052) Orally bioavailable, irreversible,
β-lactone-γ-lactam inhibitor of CT-L, T-L, and C-L proteasome activities 
PolyUb protein accumulation Phase 1 Marizomib/Pom/Dex in RRMM 
Caspase-8– and caspase-9–mediated apoptosis 
Oprozomib (ONX 0912, PR-047) Orally biovailable, irreversible epoxyketone inhibitor of CT-L proteasome activity PolyUb protein accumulation Phase 1b/2 Oprozomib/Dex in RRMM 
Caspase-8– and caspase-9–mediated apoptosis Oprozomib in RRMM 
HDACI Vorinostat (SAHA) Small molecule class I/II HDACI p21 and p53 upregulation Phase 3 Vorinostat/Bort vs Bort alone in RRMM 
Rb dephosphorylation 
BID cleavage 
Calpain activation 
Ricolinostat Orally bioavailable, small-molecule HDAC6 inhibitor Caspase-8– and caspase-9–mediated apoptosis Phase 1/2 Ricolinostat/Len/Dex in RR MM 
Terminal UPR induction Ricolinostat/Bort/Dex in RRMM 
PolyUb protein accumulation Ricolinostat/Pom/Dex in RRMM 
Aggresome disruption  
mAbs Elo Anti-SLAMF7 humanized mAb ADCC Phase 3 Elo/Len/Dex vs placebo/Len/Dex in NDMM 
Enhanced NK function 
Dara Anti-CD38 human mAb Cross-linking–mediated cytotoxicity Phase 3 Dara/Len/Dex vs placebo/Len/Dex in RRMM 
ADCC Dara/Bort/Dex vs placebo/Bort/Dex in RRMM 
CDC  
ADCP  
Sar Anti-CD38 humanized IgG1 mAb Homotypic aggregation–mediated cytotoxicity Phase 1 Sar/Len/Dex in RRMM 
ADCC Sar/Pom/Dex in RRMM 
CDC  
Indatuximab ravtansine (BT062) Chimeric, anti-CD138–conjugated maytansinoid DM4 Maytansinoid DM4 released intracellularly upon CD138-mAb internalization Phase 1/2 Indatuximab ravtansine/Len/Dex in RRMM 
Tubulin polymerization inhibition leading to cell cycle arrest and apoptosis 
J6M0-mcMMAF (GSK2857916) Humanized and afucosylated anti-BCMA–conjugated monomethyl auristatin F Monomethyl auristatin F released intracellularly upon BCMA-mAb internalization Phase 1 Dose escalation of J6M0-mcMMAF in RRMM 
Tubulin polymerization inhibition leading to cell cycle arrest and apoptosis 
Blockade of BAFF- and APRIL-induced NF-κB activation 
Immune therapies Pembrolizumab
(MK-3475) 
Humanized, anti-PD-1 mAb Increased anti-MM T, NK, and NK-T cell–mediated cytotoxicity Phase 1/2 Pembrolizumab + Len post-ASCT 
Pembrolizumab + Pom/Dex in RRMM 
Pidilizumab (CT-011) Humanized, anti-PD-1 mAb Increased anti-MM T, NK and NK-T-mediated cytotoxicity Phase 1/2 Pidilizumab + Len in RRMM 
Nivolumab (BMS936558) Human IgG4 anti-PD-1 mAb Increased T, NK, and NK-T antitumor cytotoxicity Phase 1 Nivolumab ± ipilimumab or lirilumab in R/RMM 
PVX-410 XBP-1–, CD138-, and SLAMF7-derived peptide vaccine targeting HLA2-restricted antigen-presenting cells Expansion and activation of MM-specific T cells Phase 1 PVX-410 alone or in combination with Len in SMM 
MM-DC vaccine Chemically fused autologous MMCs and DCs targeting CD4+ and CD8+ T cells Anti-MM humoral and cytotoxic response Phase 2 MM-DC vaccine alone or in combination with pidilizumab post-ASCT 
Anti-BCMA CAR-T cells Anti-BCMA CAR-T cells Specific, anti-MM T-cell response Phase 1 Anti-BCMA CAR-T cells in RRMM 
BM-targeting therapies TH-302 Hypoxia-activated DNA alkylator prodrug Genotoxic damage Phase 1/2 TH-302/Bort/Dex in RRMM 
NOX-A12 CXCL12 pegylated mirror-image
l-oligonucleotide 
CXCL12/CXCR4 signaling inhibition Phase 2a NOX-A12/Bort/Dex in RRMM 
Molecularly
targeted therapies 
Filanesib (ARRY-520) KSP inhibitor Mitotic arrest Phase 2 Filanesib alone or in combination with Carf/Dex in RRMM 
Apoptosis 
Selinexor (KPT-330) CRM-1 inhibitor Cell cycle arrest Phase 1/2 Selinexor/Dex 
Apoptosis Selinexor/liposomal Dox/Dex 
MYC inhibition Selinexor/Pom/Dex 
Mcl-1 inhibition Selinexor/Bort/Dex 
NF-κB inhibition Selinexor/Carf/Dex in RRMM 
GSK2141795 Afuresertib (GSK2110183) Akt inhibitors IL-6 pro-MM effect inhibition Phase 2 GSK2141795 + trametinib in RRMM 
Cell cycle arrest GSK2110183 in PI-refractory MM 
UPR induction  
Apoptosis  
Dinaciclib (SCH 727965) Inhibitor of CDK 1, 2, 5 and 9 Apoptosis Phase 2 Dinaciclib in RRMM 
Inhibition of nuclear localization of spliced XBP-1 
p53 accumulation 
MCL-1 downregulation 
GSK525762 CPI-0610 BET bromodomain small molecule inhibitor Myc downregulation Phase 1 GSK525762 in relapsed and refractory hematologic malignancies 
Cell cycle arrest CPI-0610 in RRMM 
Cell senescence  
Drug classDrug nameRelevant pharmacokinetic/ pharmacodynamic propertiesMechanisms of effectivenessMost advanced phase of developmentStudy design
PI Ixazomib (MLN9708) Orally bioavailable, reversible, boronate-based inhibitor of CT-L proteasome activity PolyUb protein accumulation Phase 3 Ixazomib/Len/Dex vs placebo/Len/Dex in RRMM 
Caspase-8– and caspase-9–mediated apoptosis Ixazomib/Len/Dex vs placebo/Len/Dex in transplant-ineligible NDMM 
p53 and p21 upregulation  
Terminal UPR induction  
miR33b upregulation  
PIM1 downregulation  
Marizomib (NPI-0052) Orally bioavailable, irreversible,
β-lactone-γ-lactam inhibitor of CT-L, T-L, and C-L proteasome activities 
PolyUb protein accumulation Phase 1 Marizomib/Pom/Dex in RRMM 
Caspase-8– and caspase-9–mediated apoptosis 
Oprozomib (ONX 0912, PR-047) Orally biovailable, irreversible epoxyketone inhibitor of CT-L proteasome activity PolyUb protein accumulation Phase 1b/2 Oprozomib/Dex in RRMM 
Caspase-8– and caspase-9–mediated apoptosis Oprozomib in RRMM 
HDACI Vorinostat (SAHA) Small molecule class I/II HDACI p21 and p53 upregulation Phase 3 Vorinostat/Bort vs Bort alone in RRMM 
Rb dephosphorylation 
BID cleavage 
Calpain activation 
Ricolinostat Orally bioavailable, small-molecule HDAC6 inhibitor Caspase-8– and caspase-9–mediated apoptosis Phase 1/2 Ricolinostat/Len/Dex in RR MM 
Terminal UPR induction Ricolinostat/Bort/Dex in RRMM 
PolyUb protein accumulation Ricolinostat/Pom/Dex in RRMM 
Aggresome disruption  
mAbs Elo Anti-SLAMF7 humanized mAb ADCC Phase 3 Elo/Len/Dex vs placebo/Len/Dex in NDMM 
Enhanced NK function 
Dara Anti-CD38 human mAb Cross-linking–mediated cytotoxicity Phase 3 Dara/Len/Dex vs placebo/Len/Dex in RRMM 
ADCC Dara/Bort/Dex vs placebo/Bort/Dex in RRMM 
CDC  
ADCP  
Sar Anti-CD38 humanized IgG1 mAb Homotypic aggregation–mediated cytotoxicity Phase 1 Sar/Len/Dex in RRMM 
ADCC Sar/Pom/Dex in RRMM 
CDC  
Indatuximab ravtansine (BT062) Chimeric, anti-CD138–conjugated maytansinoid DM4 Maytansinoid DM4 released intracellularly upon CD138-mAb internalization Phase 1/2 Indatuximab ravtansine/Len/Dex in RRMM 
Tubulin polymerization inhibition leading to cell cycle arrest and apoptosis 
J6M0-mcMMAF (GSK2857916) Humanized and afucosylated anti-BCMA–conjugated monomethyl auristatin F Monomethyl auristatin F released intracellularly upon BCMA-mAb internalization Phase 1 Dose escalation of J6M0-mcMMAF in RRMM 
Tubulin polymerization inhibition leading to cell cycle arrest and apoptosis 
Blockade of BAFF- and APRIL-induced NF-κB activation 
Immune therapies Pembrolizumab
(MK-3475) 
Humanized, anti-PD-1 mAb Increased anti-MM T, NK, and NK-T cell–mediated cytotoxicity Phase 1/2 Pembrolizumab + Len post-ASCT 
Pembrolizumab + Pom/Dex in RRMM 
Pidilizumab (CT-011) Humanized, anti-PD-1 mAb Increased anti-MM T, NK and NK-T-mediated cytotoxicity Phase 1/2 Pidilizumab + Len in RRMM 
Nivolumab (BMS936558) Human IgG4 anti-PD-1 mAb Increased T, NK, and NK-T antitumor cytotoxicity Phase 1 Nivolumab ± ipilimumab or lirilumab in R/RMM 
PVX-410 XBP-1–, CD138-, and SLAMF7-derived peptide vaccine targeting HLA2-restricted antigen-presenting cells Expansion and activation of MM-specific T cells Phase 1 PVX-410 alone or in combination with Len in SMM 
MM-DC vaccine Chemically fused autologous MMCs and DCs targeting CD4+ and CD8+ T cells Anti-MM humoral and cytotoxic response Phase 2 MM-DC vaccine alone or in combination with pidilizumab post-ASCT 
Anti-BCMA CAR-T cells Anti-BCMA CAR-T cells Specific, anti-MM T-cell response Phase 1 Anti-BCMA CAR-T cells in RRMM 
BM-targeting therapies TH-302 Hypoxia-activated DNA alkylator prodrug Genotoxic damage Phase 1/2 TH-302/Bort/Dex in RRMM 
NOX-A12 CXCL12 pegylated mirror-image
l-oligonucleotide 
CXCL12/CXCR4 signaling inhibition Phase 2a NOX-A12/Bort/Dex in RRMM 
Molecularly
targeted therapies 
Filanesib (ARRY-520) KSP inhibitor Mitotic arrest Phase 2 Filanesib alone or in combination with Carf/Dex in RRMM 
Apoptosis 
Selinexor (KPT-330) CRM-1 inhibitor Cell cycle arrest Phase 1/2 Selinexor/Dex 
Apoptosis Selinexor/liposomal Dox/Dex 
MYC inhibition Selinexor/Pom/Dex 
Mcl-1 inhibition Selinexor/Bort/Dex 
NF-κB inhibition Selinexor/Carf/Dex in RRMM 
GSK2141795 Afuresertib (GSK2110183) Akt inhibitors IL-6 pro-MM effect inhibition Phase 2 GSK2141795 + trametinib in RRMM 
Cell cycle arrest GSK2110183 in PI-refractory MM 
UPR induction  
Apoptosis  
Dinaciclib (SCH 727965) Inhibitor of CDK 1, 2, 5 and 9 Apoptosis Phase 2 Dinaciclib in RRMM 
Inhibition of nuclear localization of spliced XBP-1 
p53 accumulation 
MCL-1 downregulation 
GSK525762 CPI-0610 BET bromodomain small molecule inhibitor Myc downregulation Phase 1 GSK525762 in relapsed and refractory hematologic malignancies 
Cell cycle arrest CPI-0610 in RRMM 
Cell senescence  

The table summarizes the most salient properties of novel agents in advanced clinical development.

ADCC, antibody-dependent cellular cytotoxicity; ADCP, antibody-dependent cellular phagocytosis; APRIL, a proliferation-inducing ligand; BAFF, B-cell activating factor; BCMA, B-cell maturation antigen; Bort, bortezomib; Carf, carfilzomib; CDC, complement-dependent cytotoxicity; C-L, caspaselike; CRM-1, chromosome region maintenance 1; CT-L, chymotrypsin-like; CXCL, chemokine (CXC motif) ligand; Dara, daratumumab; DC, dendritic cell; Dex, dexamethasone; Dox, doxorubicin; Elo, elotuzumab; Ig, immunoglobulin; IL, interleukin; Len, lenalidomide; MCL-1, myeloid leukemia cell 1; miR, microRNA; MMC, MM cell; NDMM, newly diagnosed MM; NF, nuclear factor; NK, natural killer; PD-1, programmed cell death 1; PolyUb, polyubiquitinated; Pom, pomalidomide; RR, relapsed and refractory; R/R, relapsed or refractory; RRMM, relapsed and refractory MM; SAHA, suberoylanilide hydroxamic acid; Sar, SAR650984; SMM, smoldering MM; T-L, trypsinlike; UPR, unfolded protein response; XBP-1, X-box binding protein 1.

In preclinical studies, bortezomib, the first-in-class boronic acid inhibitor of the CT-L activity of the proteasome and immunoproteasome, inhibits cell cycle progression, growth, and DNA damage repair in MM cells (MMCs), as well as induces caspase-8– and caspase-9–mediated apoptosis, terminal UPR, proteotoxic stress, and heat shock protein response.8-10  In addition, it targets the BM microenvironment, evidenced by its antiosteoclast, antiangiogenesis, and proosteoblast activities.11,12  Preclinical studies moved rapidly to phase 1, 2, and 3 clinical trials that demonstrated durable responses to bortezomib and provided the basis for its FDA approval in all stages of MM management.5,13,14  Together with IMiDs and dexamethasone, bortezomib is now integrated as frontline therapy in the majority of MM patients, with ORRs as high as 100% with lenalidomide/bortezomib/dexamethasone, demonstrating the powerful synergy of using both PIs and IMiDs in combination.7,15  The relative inconvenience of parenteral administration, peripheral neuropathy attendant to IV (vs subcutaneous) bortezomib administration, and the emergence of resistance has since stimulated the development of second-generation PIs with improved pharmacodynamics and more potent and/or broader activity against proteasome catalytic subunits, as well as the potential for oral administration.

Carfilzomib, an epoxyketone irreversible inhibitor of the CT-L proteasome activity, was approved by the FDA for treatment of relapsed MM refractory to bortezomib and exposed to an IMiD, based on a 23.7% ORR and a median progression-free survival (PFS) of 3.7 months.16  In bortezomib-naïve patients, carfilzomib combined with low-dose dexamethasone achieved a 52.2% ORR in patients treated with the 27 mg/m2 dose, and median PFS was not reached at the time the trial was reported.17  When compared to the 41% ORR achieved with single-agent bortezomib in the Assessment of Proteasome Inhibition for Extending Remissions (APEX) trial, these data suggest that carfilzomib may be more effective than bortezomib.13,18  Indeed, interim analysis of the ENDEAVOR trial (NCT01568866, a randomized, open-label, phase 3 study of carfilzomib plus dexamethasone vs bortezomib plus dexamethasone in patients with relapsed multiple myeloma) showed that carfilzomib/dexamethasone achieved a PFS of 18.7 months vs 9.4 months for bortezomib/dexamethasone in largely bortezomib-pretreated patients. However, increased toxicities were also noted in this study, including renal and cardiopulmonary side effects at the higher dose of carfilzomib used (56 mg/m2). The recently completed phase 3 randomized ASPIRE trial compared carfilzomib/lenalidomide/dexamethasone (KRd) to lenalidomide/dexamethasone (Rd) in relapsed or in RRMM and showed a 26.3- vs 17.6-month PFS, respectively (P < .0001), which was associated with an increase in overall and extent of response to KRd. Importantly, there were no major differences in adverse events in either cohort, with the exception of hypertension, dyspnea, and deep venous thrombosis, all of which were higher with KRd. Encouragingly, as a frontline therapy, this triple combination achieved a 98% ORR, with a 62% near complete response (nCR) rate or better, and an estimated PFS rate at 2 years of 92% at a median follow-up of 13 months.19  Importantly, this combination can achieve molecular complete responses (CRs) without attendant neuropathy, but again, some caution is warranted because both venous thrombosis and significant shortness of breath (possibly due to diastolic dysfunction) were noted in some patients in this study.19  Randomized trials are now underway comparing KRd to lenalidomide/bortezomib/dexamethasone in newly diagnosed patients, and results of these studies are awaited with great interest.

Ixazomib (MLN9708) is a reversible, orally bioavailable boronic-acid based inhibitor of the CT-L activity of the 20S proteasome.20  It triggers both caspase-8– and caspase-9–mediated apoptosis, upregulates p53 and p21, induces terminal UPR, and can overcome bortezomib resistance in preclinical studies.20  It also induces tumor-suppressor microRNA 33b, with associated downregulation of the oncogene PIM-1.21  As a single agent, weekly oral ixazomib achieved an 18% ORR in RRMM, including bortezomib-resistant MM, and was also active when given twice weekly in more heavily pretreated patients.22,23  It is well tolerated, with low rates of peripheral neuropathy and treatment discontinuation. Remarkably, in a phase 1/2 study, the combination of ixazomib/Rd achieved a 90% ORR, with a 59% very good partial response rate or better in NDMM.24  Moreover, maintenance therapy with ixazomib, given as 1 tablet weekly, was well tolerated and further improved response.25  Ixazomib/Rd is being compared to Rd in 2 phase 3 clinical trials in RRMM (NCT01564537, TOURMALINE-MM1) and NDMM (NCT01850524, TOURMALINE-MM2). At the first interim analysis for patients enrolled in the TOURMALINE1 trial in RRMM, the primary end point of PFS extension with ixazomib/Rd vs Rd has been reached. This opportunity to use an all oral and well-tolerated regimen combining IMiDs and PIs both as salvage and especially as induction therapy is a major advance, as is the opportunity to use oral PI in maintenance treatment, which has particular promise in older patients in whom favorable tolerability is a premium.

Oprozomib (ONX 0912, PR-047), an orally bioavailable carfilzomib analog, is cytotoxic in preclinical MM models, including against bortezomib-resistant patient MMCs, and triggers synergistic cytotoxicity with lenalidomide and HDACIs.26  Similar to bortezomib and carfilzomib, it also has a bone anabolic effect in preclinical models.27  Oprozomib achieved a 33% to 37% ORR in RRMM, including bortezomib- and carfilzomib-refractory MM (NCT01416428).28  However, 20% of patients experienced severe (grade 3 or higher) gastrointestinal side effects, including 2 patients with fatal outcome. Preliminary results from a phase 1b/2 study of oprozomib/dexamethasone in RRMM showed a 42% ORR, with improved tolerability (NCT01832727), and phase 1/2 studies of oprozomib/dexamethasone plus IMiDs or cyclophosphamide in RRMM are ongoing.29 

To determine whether inhibition of all 3 proteolytic subunits of the proteasome can overcome bortezomib resistance, the panproteasome inhibitor marizomib (NPI-0052) is currently being evaluated in clinical trials.30  Twice-weekly marizomib in combination with dexamethasone achieved a 19% ORR, even in bortezomib-, carfilzomib-, lenalidomide-, and pomalidomide-refractory MM.31  Based on preclinical studies demonstrating synergistic cytotoxicity of marizomib and pomalidomide/dexamethasone, a phase 1 trial of this combination in RRMM is now ongoing (NCT02103335).32 

Access to the 20S proteolytic core of the proteasome requires the concerted activity of the 19S regulatory particles (RPs), which control gate opening and access to the core, along with deubiquitinating enzymes, which remove ubiquitin from target proteins prior to their degradation.33,34  The 19S ubiquitin receptor RPN13, as well as the deubiquitinating enzymes ubiquitin-specific peptidase (USP)7 and USP14/ubiquitin carboxyl-terminal hydrolase L5 (UCHL5), are upregulated in MM cell lines (MMCLs) and patient MMCs; conversely, knockdown of these targets decreases MM viability.35-37  RA190, P5091, and B-AP15 are small-molecule inhibitors of RPN13, USP7, and USP14/UCHL5, respectively. All are enzyme-specific inhibitors and are cytotoxic in vitro and in vivo against MMCLs and patient MMCs, including bortezomib-resistant MMCs.37  Culture of MMCs with BM stromal cells does not overcome the cytotoxic effect of these therapies, which trigger synergistic MM cytotoxicity when combined with IMiDs, bortezomib, and the HDACI vorinostat.35,38  Importantly, these agents block the ubiquitin-proteasome system (UPS) upstream of the proteasome, thereby causing accumulation of polyubiquitinated proteins without blocking the proteasome; they overcome PI resistance and trigger activation of intrinsic and extrinsic apoptotic pathways in a p53-independent fashion. The first USP14/UCHL5 inhibitor B-AP15 clinical trial for RRMM in humans is beginning soon, and will further validate the UPS as a therapeutic target in MM and assess whether targeting UPS upstream of the proteasome can overcome clinical PI resistance (Figure 1, section A).

Figure 1

Multimodality targeting of MM in the context of the BM microenvironment. In the center is the MMC (in light pink), with nuclear clumped chromatin and endoplasmic reticulum (ER). The surrounding 5 sections each represent a different modality of anti-MM therapy, with investigational agents outlined in red. In section A is the UPS, closely interacting with the aggresome pathway. Deubiquitinating enzymes USP7 and USP14/UCHL5 are symbolized by a scissors and the 19S ubiquitin (Ub) receptor RPN13 as a receptor associated with the proteasome cap. Section B contains the monoclonal antibodies mAbs) daratumumab (DARA) and SAR650984 (SAR), targeting CD38, as well as elotuzumab (ELO), targeting SLAMF-7, which mediate CDC, direct cytotoxicity from cross-linking, and ADCC. The antibody-drug conjugates (ADCs) indatuximab ravtansine (BT062) and J6M0-mcMMAF (J6M0) target CD138 and BCMA, respectively. Both toxins cause mitotic arrest and apoptosis after being released intracellularly upon internalization of the ADC–target complex and lysosomal lysis. Section C represents several strategies for modulation of cytotoxic immunity. In red is an anti-BCMA CAR-T cell; in orange is an MM-specific cytotoxic T cell activated via direct interaction with MM-DC vaccine or with autologous DC presenting peptides from the PVX-140 vaccine. Two different strategies to modulate epigenetic control of oncosuppressor and oncogene expression are outlined in section D. Nucleosomes are represented as spheres (histones) wrapped in a black thread (DNA). Open nucleosomes with acetylated (Ac) sites are green, whereas closed chromatin structure is pink. The BET bromodomain 4 protein (BRD4) is represented as a red trapezoid, binding to acetylated nucleosomes and inducing Myc transcription. Section E contains a representation of immune checkpoint blockade, with cytotoxic T and NK-T cells represented in green and blue, respectively. PD-1 and its ligand PD-L1 are represented as complementary transmembrane structures on effector cells and target cells (MMCs, myeloid-derived suppressor cells [MDSCs], and pDCs), respectively. Cytotoxic T-lymphocyte-associated protein 4 (CTLA4) and killer cell immunoglobulin-like receptor (KIR) are also pictured on effector cells. Outside the sections, TH-302 hypoxia–activated alkylating agent and NOX-A12 CXCL12 inhibitor are represented. In the 4 corners are key cellular and noncellular elements of the BM niche that contribute to MM pathogenesis: excess of osteoclasts (OC) compared to osteoblasts (OB; upper left corner); increased neoangiogenesis (upper right corner); tumor-tolerant immune system (lower right corner); and cancer-associated fibroblasts (CAF) responsible for the secretion of a pro-MM extracellular matrix (ECM), and MM-associated BM stromal cells (BMSC; lower left corner). Relevant cytokines in the BM milieu are represented as orange ovals. FcR, Fc receptor; HATs, histone acetylases; HGF, hepatocyte growth factor; MIP-1α, macrophage inflammatory protein 1α; PD-L1, programmed cell death ligand 1; RANKL, receptor activator of NF-κB ligand; RBC, red blood cell; TCR, T-cell receptor; TGFβ, transforming growth factor β; TH17, T helper 17 cells; TNFα, tumor necrosis factor α; Treg, regulatory T cell; VEGFA, vascular endothelial growth factor A.

Figure 1

Multimodality targeting of MM in the context of the BM microenvironment. In the center is the MMC (in light pink), with nuclear clumped chromatin and endoplasmic reticulum (ER). The surrounding 5 sections each represent a different modality of anti-MM therapy, with investigational agents outlined in red. In section A is the UPS, closely interacting with the aggresome pathway. Deubiquitinating enzymes USP7 and USP14/UCHL5 are symbolized by a scissors and the 19S ubiquitin (Ub) receptor RPN13 as a receptor associated with the proteasome cap. Section B contains the monoclonal antibodies mAbs) daratumumab (DARA) and SAR650984 (SAR), targeting CD38, as well as elotuzumab (ELO), targeting SLAMF-7, which mediate CDC, direct cytotoxicity from cross-linking, and ADCC. The antibody-drug conjugates (ADCs) indatuximab ravtansine (BT062) and J6M0-mcMMAF (J6M0) target CD138 and BCMA, respectively. Both toxins cause mitotic arrest and apoptosis after being released intracellularly upon internalization of the ADC–target complex and lysosomal lysis. Section C represents several strategies for modulation of cytotoxic immunity. In red is an anti-BCMA CAR-T cell; in orange is an MM-specific cytotoxic T cell activated via direct interaction with MM-DC vaccine or with autologous DC presenting peptides from the PVX-140 vaccine. Two different strategies to modulate epigenetic control of oncosuppressor and oncogene expression are outlined in section D. Nucleosomes are represented as spheres (histones) wrapped in a black thread (DNA). Open nucleosomes with acetylated (Ac) sites are green, whereas closed chromatin structure is pink. The BET bromodomain 4 protein (BRD4) is represented as a red trapezoid, binding to acetylated nucleosomes and inducing Myc transcription. Section E contains a representation of immune checkpoint blockade, with cytotoxic T and NK-T cells represented in green and blue, respectively. PD-1 and its ligand PD-L1 are represented as complementary transmembrane structures on effector cells and target cells (MMCs, myeloid-derived suppressor cells [MDSCs], and pDCs), respectively. Cytotoxic T-lymphocyte-associated protein 4 (CTLA4) and killer cell immunoglobulin-like receptor (KIR) are also pictured on effector cells. Outside the sections, TH-302 hypoxia–activated alkylating agent and NOX-A12 CXCL12 inhibitor are represented. In the 4 corners are key cellular and noncellular elements of the BM niche that contribute to MM pathogenesis: excess of osteoclasts (OC) compared to osteoblasts (OB; upper left corner); increased neoangiogenesis (upper right corner); tumor-tolerant immune system (lower right corner); and cancer-associated fibroblasts (CAF) responsible for the secretion of a pro-MM extracellular matrix (ECM), and MM-associated BM stromal cells (BMSC; lower left corner). Relevant cytokines in the BM milieu are represented as orange ovals. FcR, Fc receptor; HATs, histone acetylases; HGF, hepatocyte growth factor; MIP-1α, macrophage inflammatory protein 1α; PD-L1, programmed cell death ligand 1; RANKL, receptor activator of NF-κB ligand; RBC, red blood cell; TCR, T-cell receptor; TGFβ, transforming growth factor β; TH17, T helper 17 cells; TNFα, tumor necrosis factor α; Treg, regulatory T cell; VEGFA, vascular endothelial growth factor A.

Close modal

HDACs are multifunction enzymes with distinct structure and target specificities that mediate epigenetic silencing of gene expression, thereby modulating key cellular processes, including proliferation, migration, and survival (Figure 1, section D).39  HDACIs, therefore, represent a promising targeted therapy in oncology. In MM, a particular rationale for use of HDACIs is their role in disrupting aggresomal protein degradation. Combining bortezomib and HDACIs to simultaneously block the proteasome and aggresome, respectively, triggers synergistic cytotoxicity and overcomes bortezomib resistance in preclinical studies.40  Based on this data, the phase 3 Vantage 088 trial in RRMM compared bortezomib alone or in combination with the class I and IIb HDACI vorinostat.41  Although combination therapy achieved a 54% ORR vs a 41% ORR for bortezomib alone (P < .0001), there was only a modest prolongation in PFS (7.6 vs 6.8 months, respectively; P = .01), primarily due to diarrhea, fatigue, and thrombocytopenia leading to increased discontinuation of treatment in the combination arm.41  In contrast, a phase 1 trial of vorinostat in combination with Rd in RRMM achieved a 47% partial response (PR) rate and was better tolerated, but the therapeutic index remained narrow.42  A phase 3 clinical trial of bortezomib with or without the pan-HDACI panobinostat achieved a 4-month prolongation of PFS with combination vs bortezomib-alone treatment (12 vs 8 months; P < .0001); moreover, 28% vs 16% of patients achieved nCR or better (P = .00006). Based on these results, panobinostat in combination with bortezomib and dexamethasone was recently FDA approved as a third-line therapy in MM patients previously exposed to bortezomib and IMiDs. A 34.5% ORR to this combination was observed in a multicenter phase 2 trial in bortezomib-resistant RRMM patients, including patients with high-risk cytogenetics, which further supports its clinical activity in this setting.43,44  As with vorinostat, however, the side-effect profile observed in the phase 3 study led to discontinuation of treatment in 34% of combination-treated vs 17% of bortezomib-treated patients, highlighting the need for more selective HDACI approaches with potential for an improved therapeutic index.44 

In this context, HDAC6 plays a key role in aggresomal protein degradation because it binds to misfolded proteins on the one hand and to the dynein motility complex on the other, thereby shuttling polyubiquitinated proteins to the aggresome/lysosome for degradation.45  Ricolinostat (ACY-1215) is a specific orally bioavailable HDAC6 inhibitor that is cytotoxic against MMCs and synergizes with bortezomib and Rd in vitro.46  A phase 1b study of ricolinostat plus bortezomib/dexamethasone in RRMM showed a 45% ORR and a 29% ORR in bortezomib-refractory MM (NCT01323751).47  Importantly, preclinical studies showed that ricolinostat with IMiDs downregulates MYC and triggers synergistic cytotoxicity.48  A phase 1b trial of ricolinostat plus Rd in RRMM achieved a 64% ORR, including 85% in lenalidomide-sensitive and 50% in lenalidomide-refractory MM patients (NCT01583283).49  Importantly, there were no grade 3 or 4 adverse events when ricolinostat was combined with either PIs or IMiDs, and clinical trials of ricolinostat in combination with pomalidomide daily for 21 days in RRMM are ongoing. Lastly, preclinical studies are further evaluating other isoform-selective HDACIs targeting HDACs relevant for MM growth and proliferation. For example, HDAC3 knockdown triggers MM cytotoxicity and apoptosis, and HDAC3 selective inhibitor BG45 is active, alone or with bortezomib, in MM preclinical models.50  Thus, isoform-selective oral HDACIs may improve tolerability, allowing for their future clinical evaluation in combination with targeted and immune therapies.

IMiDs

The rationale for using thalidomide, the first-in-class IMiD, in MM was its antiangiogenic properties.4  However, the immunomodulatory effect of thalidomide and its more potent derivatives, lenalidomide and pomalidomide, was soon recognized as a major determinant of their anti-MM activity.51  IMiDs are now incorporated into therapies for RRMM and NDMM due to a series of FDA approvals based on their remarkable clinical activity.6,52,53  Moreover, maintenance therapy with lenalidomide has achieved improvement in PFS in both transplant-eligible and -ineligible patients, and incorporation of bortezomib into maintenance confers benefit even in high-risk disease.54-56  Importantly, both the FDA and the European Commission approved continuous Rd for treatment of transplant-ineligible NDMM patients based on the results of the FIRST trial, which compared continuous Rd vs Rd for 72 weeks vs melphalan/prednisone/thalidomide (MPT) for 72 weeks.53  Patients treated with continuous Rd achieved a PFS of 25.5 months vs 20.7 and 21.2 months for limited Rd and MPT, respectively (P < .001); the 4-year OS rate was 59% vs 56% and 51%, respectively. Importantly, there was a decreased incidence of hematologic and neurologic adverse events and of second primary hematologic cancers in patients treated with continuous Rd compared to MPT, further supporting continuous Rd as a first-line MM therapy in this setting.

Mechanisms of action of IMiDs include caspase-8–mediated apoptosis; abrogation of MMC binding to BM stromal cells; modulation of cytokine secretion; upregulation of T, NK, and NK-T cells; and downregulation of regulatory T cells.57  Most recently, the E3 ubiquitin ligase cereblon has been identified as the molecular target of lenalidomide.58,59  Binding of lenalidomide to cereblon causes proteasome-dependent degradation of Ikaros family zinc finger proteins 1 and 3, which in turn mediates both MM cytotoxic and immune effects of IMiDs. Ongoing research is focusing not only on identifying biomarkers predictive of lenalidomide sensitivity but also on designing novel IMiDs based on the IMiD binding site to cereblon.60  Moreover, efforts are attempting to delineate the mechanism of synergy underlying IMiD/PI combination therapy, because in vitro data indicate that an intact UPS is required for IMiD activity.59  PIs may only partially inhibit the proteasome at therapeutic concentrations, allowing degradation of certain substrates such as the Ikaros protein; alternatively, the polyubiquitinated Ikaros proteins could act as dominant negative, impairing the physiologic function of their nonubiquitinated counterpart.59  These efforts will allow for rational, next-generation combination therapies.

Importantly, predicated on their immunostimulatory effects, IMiDs enhance activities of immune therapies including mAbs, checkpoint inhibitors, and vaccines, providing the framework for combination clinical trials.61  Our group has long supported the theory that the malignant microenvironment is a necessary pathogenic element in MM, and we believe that disrupting this key interaction via immune therapy will prove vital to improving treatment outcome for MM patients (Figure 1).

mAbs

Elotuzumab is a fully humanized mAb directed against the glycoprotein SLAM family member 7 (SLAMF-7), which is highly expressed on the cell surface of MMCLs and patient MMCs.62  Elotuzumab triggers ADCC and enhances NK cell function against MMCs. As a single agent, it achieved stable disease in the setting of RRMM. Predicated on preclinical studies showing that lenalidomide augmented ADCC, a phase 2 study of elotuzumab with Rd in patients with RRMM, including patients with high-risk disease, achieved a 92% ORR and PFS of 32.5 months.63,64  Phase 3 studies of this combination vs Rd in patients with NDMM (NCT01335399, ELOQUENT-1) and RRMM (NCT01239797, ELOQUENT-2) are ongoing.

The target of the mAb daratumumab is CD38, a transmembrane protein expressed on MM and activated immune cells, hematopoietic progenitor cells, and endothelial cells. The anti-MM effect of daratumumab is both direct, due to inhibition of enzymatic activity and apoptotic signaling triggered via cross-linking on the MM cell surface; and immune-mediated via ADCC, complement-dependent cytotoxicity, and antibody-dependent cellular phagocytosis (Figure 1, section B).65,66  Preliminary studies showed very promising single-agent activity, with a 31% ORR in heavily pretreated RRMM, leading to designation of breakthrough status by the FDA and a strong rationale for combination approaches.67  Specifically, as with elotuzumab, the addition of Rd to daratumumab significantly increased depth of response to a 75% PR rate or better in RRMM.68  Phase 3 trials of daratumumab with Rd or bortezomib/dexamethasone in RRMM are currently accruing (NCT02076009 and NCT02136134, respectively); and a phase 3 study of daratumumab/Rd in NDMM is planned (NCT02252172). Similar results have been observed with another CD38 mAb, SAR650984, with responses even in carfilzomib- and pomalidomide-resistant MM.69 

Indatuximab ravtansine (BT062) is an antibody-drug conjugate comprising anti-CD138 mAb targeting syndecan1 (CD138) coupled to the potent maytansinoid DM4 toxin.70  Upon internalization of the CD138–antibody-drug conjugate complex and lysosome-mediated proteolysis, DM4 is released and inhibits tubulin polymerization, resulting in cell cycle arrest and apoptosis.71  In a phase 1/2 trial in combination with Rd, indatuximab ravtansine achieved a 78% ORR, including responses in bortezomib- and lenalidomide-refractory MM (NCT01638936).72  J6M0-mcMMAF (GSK2857916) is a humanized, afucosylated mAb directed against BCMA conjugated via a noncleavable linker to the antimitotic agent monomethyl auristatin F. The latter is released intracellularly via a mechanism similar to DM4 for BT062, and induces cell cycle arrest and apoptosis. Because BCMA is the receptor for B-cell activating factor and a proliferation-inducing ligand, J6M0-mcMMAF also blocks B-cell activating factor– and a proliferation-inducing ligand–induced nuclear factor κB activation.73  A phase I study of J6M0-mcMMAF in RRMM is now ongoing (NCT02064387).

Additional preclinical and clinical studies are evaluating mAbs directed against antigens expressed on MMCs such as CD40, CD54 (also known as intercellular adhesion molecule 1), CD56, and GM2 ganglioside; as well as the anti–vascular endothelial growth factor A (VEGF-A) mAb bevacizumab.

Immune checkpoint blockade

Cancer immune escape due to tumor-induced NK- and T-cell anergy/exhaustion has emerged as an important determinant of cancer progression and/or recurrence.74  In MM, the importance of host antitumor immunity is evidenced by long-term molecular CR observed post–allogeneic hematopoietic stem cell transplant due to ongoing graft-versus-MM effect.75  Most recently, mAbs to block the inhibitory interaction of PD-1 on T or NK cells with its ligand PD-L1 on tumor cells or tumor-promoting accessory cells have achieved remarkable responses in both solid tumors and hematologic malignancies.74,76 

In MM, tumor cells, pDCs, and myeloid-derived suppressor cells all express PD-L1, whereas BM-resident cytotoxic T, NK, and NK-T cells express PD-1.77,78  Our preclinical in vitro and ex vivo studies showed that blockade of PD-1/PD-L1 inhibits accessory cell (pDC or myeloid-derived suppressor cell)-induced MM proliferation and survival while triggering host T- and NK-cell anti-MM cytotoxicity. Moreover, these effects can be markedly enhanced by lenalidomide, suggesting the utility of combination immune therapies.

There are multiple checkpoint inhibitor clinical trials ongoing or planned in MM. A phase 2 study of the humanized anti–PD-1 mAb pembrolizumab (MK-3475) with lenalidomide post-ASCT (NCT02331368) and a phase 1/2 study of pembrolizumab plus pomalidomide/dexamethasone in RRMM (NCT02289222) are ongoing. The humanized anti–PD-1 mAb pidilizumab (CT-011) is being evaluated in combination with vaccination post-ASCT (NCT01067287), as well as with lenalidomide in RRMM patients (NCT02077959). The fully human immunoglobulin G4 anti–PD-1 antibody nivolumab (BMS936558), alone or in combination with the CTLA4-blocking antibody ipilimumab or the killer cell immunoglobulin-like receptor–blocking antibody lirilumab, is being evaluated in a phase 1 clinical trial in relapsed or refractory hematologic malignancies, including MM (NCT01592370) (Figure 1, section E). Future trials will combine checkpoint inhibitors, mAbs, vaccinations, and/or IMiDs in an attempt to further enhance autologous, selective anti-MM memory immunity and achieve durable clinical responses. Importantly, the potency, selectivity, and adaptability of the immune response may allow for effective host anti-MM immunity, even in the setting of ongoing genomic evolution, thereby preventing disease relapse.

Vaccines

Vaccination against cancer-specific antigens represents a promising strategy to modulate patient antitumor immune response, particularly in the settings of early-stage or minimal residual disease. For example, we are vaccinating patients with SMM with the goal of delaying their progression to active disease. The vaccine (PVX-410) consists of a cocktail of HLA-A2–specific peptides derived from X-box binding protein 1, CD138, and SLAMF-7 MM antigens, which can trigger HLA-restricted expansion and activation of MM-specific T cells. On going studies are combining PVX-410 with lenalidomide and with anti–PD-1 to further enhance MM-specific immune responses (NCT01718899).79 

An alternative approach involves vaccination of patients with their own tumor cells fused to autologous DCs (MM-DC fusion vaccine) (Figure 1, section C). In a phase 1 trial in RRMM, we have shown that MM-DC fusion vaccination triggers both humoral and cellular anti-MM responses, associated with 70% stable disease.80  Excitingly, MM-DC vaccination post-ASCT achieved a 78% very good partial response rate and a 47% CR or nCR rate, with responses improving from PR to CR/nCR after 100 days posttransplant in 24% of patients, suggesting its utility to treat minimal residual disease.81  A phase 2 randomized clinical trial of post-ASCT maintenance using lenalidomide with or without MM-DC vaccination is opening soon. Decreased regulatory–T cell function and minimal disease state posttransplant suggest that this setting is optimal for vaccination; again, the goal is to enhance vaccine-induced long-term autologous anti-MM memory immunity by combining vaccination with lenalidomide or anti-PD-1 antibodies, or both.

pDCs

pDCs are increased in MM BM and promote tumor cell proliferation, survival, and drug resistance; moreover, they also fail to trigger host antitumor immune response.82  Either cytosine-phosphate-guanosine (CpG) oligodeoxynucleotide A or C792 (a cytosine guanine dinucleotide oligodeoxynucleotide C acting as a Toll-like receptor 9 agonist) can mature MM pDCs, thereby restoring their immune-stimulatory ability while abrogating their pro-MM activities.83  Clinical trials of Toll-like receptor 7 agonist are planned to test the therapeutic benefit of solely targeting immune accessory cells, because these agents have no direct anti-MM activity. Importantly, because PD-L1 is expressed on pDCs, checkpoint inhibitor therapy can also abrogate the functional sequelae of pDCs in MM.78 

Modulation of cellular anti-MM immune surveillance

CD19-directed CAR-T cells have achieved remarkable responses in relapsed and refractory chronic lymphocytic leukemia, non-Hodgkin lymphoma, and acute lymphoblastic leukemia.84,85  CAR-T cells directed against CD38 and SLAMF-7 are in preclinical development in MM, whereas CAR-T cells against BCMA are already being evaluated in a phase 1 clinical trial (NCT02215967) (Figure 1, section C).86  The opportunity here is for dramatic tumor cell reduction, even in high-risk, refractory MM; moreover, use of lenalidomide and/or checkpoint inhibitors post–CAR-T cell therapy may allow for persistence of cancer immune surveillance by avoiding T-cell exhaustion.

Bispecific T-cell engagers (BiTEs) are composed of 2 single-chain variable fragments connected by a linker.87  BiTEs redirect anticancer immunity by binding to a T cell–specific antigen (typically CD3) with 1 fragment and to a cancer-specific epitope with the other fragment, thus juxtaposing the effector and cancer cells. The CD3-CD19 BiTE blinatumomab was granted FDA approval based on a 43% CR rate in relapsed or refractory B cell–precursor acute lymphoblastic leukemia.88  CD3-CD38 BiTEs are in preclinical development for MM.

MMCs establish a bidirectional prosurvival relationship with both cellular and noncellular elements of the BM milieu and can co-opt the function of BM accessory cells to create a permissive microenvironment for their growth and survival (Figure 1; 4 corners).89  Therefore, directly targeting the BM microenvironment represents a novel strategy to indirectly abrogate MM growth and survival.

CXCL12/CXCR4 axis inhibitors

CXCL12, also known as SDF-1α, mediates trafficking and homing of MMCs to the BM microenvironment.90  Plerixafor, an inhibitor of the CXCL12 ligand CXCR4, is used for mobilization of normal BM hematopoietic stem cells for ASCT and has been tested as an anti-MM therapy, predicated on mobilizing MMCs from their protective BM milieu.91,92  Inhibition of CXCL12 using NOX-A12 (olaptesed pegol), a high-affinity anti–SDF-1α pegylated mirror-image l-oligonucleotide, triggers MM cytotoxicity in preclinical studies.93  In a phase 2a trial in RRMM, combination NOX-A12/bortezomib/dexamethasone achieved a PFS of 6.5 months and a 73% ORR, even in high-risk and/or bortezomib-refractory patients.94  The regimen was well tolerated, and this combination is now entering phase 3 trials.

Exploiting hypoxia against MM

The BM microenvironment in MM is hypoxic, and hypoxia inducible factor 1α is upregulated in patient MMCs.95  Moreover, hypoxia is a driver of epithelial to mesenchymal transition in MMCs, thereby promoting their dissemination.96  TH-302 is a DNA alkylator prodrug selectively activated under hypoxic conditions, which triggers MM cytotoxicity, alone and with bortezomib, in preclinical models.97  A phase 1/2 study of TH-302 with dexamethasone/bortezomib showed no dose-limiting toxicity at the recommended phase 2 dose, with 29% and 50% ORRs in the phase 1 and 2 cohorts, respectively.98  Exploiting MMC vulnerability due to hypoxia may therefore allow for selective tumor cytotoxicity and a favorable therapeutic index.

Predicated on preclinical studies, a number of signaling molecules have been identified as potential molecular targets of MM therapy. Inhibitors of aurora kinase A and B, KSP XPO1 (also known as chromosome region maintenance 1 [CRM-1]) AKT, and CDKs are among those in promising early-phase clinical trials.99  The KSP inhibitor filanesib (ARRY-520) causes apoptosis in MM and is being evaluated in RRMM, alone (NCT02092922 and NCT00821249, phase II) or in combination with bortezomib/dexamethasone (NCT01248923, phase I) or carfilzomib/dexamethasone (NCT01372540 and NCT01989325, phase I and II, respectively).100  Because this agent is highly bound in serum to α1-acid glycoprotein, its activity is enhanced in patients with low α1-acid glycoprotein levels.101  Selinexor (KPT-330), an inhibitor of the nuclear export protein CRM1, functions by maintaining the cellular distribution of tumor suppressors in MMCs.102,103  Although single-agent activity was not observed in MM, phase 1/2 clinical trials in combination with dexamethasone, liposomal doxorubicin, pomalidomide/dexamethasone, or PIs/dexamethasone are ongoing, with promising interim results (NCT02336815, NCT02186834, NCT02343042, or NCT02199665, respectively).104  Multiple studies have shown that PIs trigger apoptotic signaling but also induce AKT.105  AKT inhibitors GSK2141795 and GSK2110183 are therefore being clinically evaluated in combination with bortezomib and other PIs. Lastly, a hallmark of MM is cyclin D dysregulation; and multiple preclinical studies have evaluated CDK inhibitors in MM.106,107  Based on promising preclinical studies, the CDK inhibitors dinaciclib (SCH 727965), and SNS-032 are now being evaluated in phase 1/2 trials in RRMM (NCT01096342, NCT00446342, and NCT01711528).108-110 

Among novel targeted therapies, BET bromodomain and STK4 inhibitors hold particular promise in MM and other hematologic malignancies.

BET bromodomains

Myc is an oncogene in solid tumors and hematologic malignancies including MM, and BET bromodomains have recently been shown to regulate Myc transcription in MM (Figure 1D).111,112  Importantly, inhibition of BET bromodomain 4 via small-molecule JQ1 downregulates Myc transcription and its downstream targets and is associated with decreased MMC growth in vitro and in vivo in murine models. Phase 1 clinical trials of BET bromodomain inhibitors GSK525762 (NCT01943851) and CPI-0610 (NCT02157636) are ongoing in RRMM. These studies will, for the first time, evaluate the therapeutic efficacy of targeting Myc and also inform combination approaches, such as with IMiDs.

YAP1/STK4

The Hippo coactivator Yes-associated protein 1 (YAP1) is essential for p53-independent, ABL1-induced apoptosis secondary to DNA damage.113  We have recently shown that YAP1 mRNA and protein levels are low in a subset of lymphoid and myeloid malignancies, including MM, which portends poor survival. In the setting of constitutive ongoing DNA damage, restoration of YAP1 levels and function in MMCs results in apoptosis via induction of p73 and downstream target genes. Importantly, STK4 regulates YAP1 phosphorylation and its degradation in MM; conversely, knockdown of STK4 results in upregulation of YAP1, associated with MM cytotoxicity both in vitro and in vivo in mouse xenograft models. Based on this synthetic lethality, our ongoing studies are developing clinical-grade STK4 inhibitors to upregulate YAP1 and induce p73-mediated apoptosis in MM and other hematologic malignancies characterized by low YAP1 expression.114  Importantly, STK4 inhibitors may restore YAP1 and p73 signaling even in high-risk, 17p-deleted MM lacking functional p53.

MM is characterized by complex genomic alterations, and no single predominant driver mutation has been identified.115  The majority of mutations detected in MM are already present at the stage of monoclonal gammopathy of undetermined significance and/or SMM, suggesting that genetic mutations per se are not sufficient for oncogenesis and clonal evolution.116  Whole-exon and -genome sequencing has been instrumental not only in identifying the genetic landscape of MM but also in delineating the mechanisms underlying progression and relapse. To date, genes identified to be mutated in MM are those implicated in protein homeostasis, nuclear factor κB signaling, and histone methylation, consistent with MM pathogenesis.117  Mutations have also been observed in genes not previously implicated in oncogenesis, such as FAM46C and SP140, which may therefore represent potential novel therapeutic targets in MM.117  Of note, B-RAF mutations have been described in 4% of MM, and vemurafenib has achieved responses in this setting.118  Our recent RNA sequencing study in patient-derived MMCs showed that only 27% of mutated alleles are expressed at the mRNA level and, therefore, have biological and clinical relevance.119  Importantly, clonal heterogeneity and clonal evolution is a hallmark in MM pathogenesis and progression.117,120 

This genomic heterogeneity and complexity in MM highlights the need to use combination therapies as early as possible to prevent genomic evolution and progression, as well as the need to define genomic signatures in patients at a particular time in their disease course to inform appropriate combination targeted therapies. In ongoing and future studies, it will be critical to identify those pathways to be targeted at a given point in time in order to inform combination targeted, epigenetic, and immune therapeutic approaches that will avoid genomic evolution underlying disease relapse.

Novel agents targeting MMC in the context of the BM microenvironment, with or without stem cell transplantation, have prolonged patient survival three- to fourfold. We believe that translational research focus in 3 areas will assure further progress. First, genomic, epigenomic, and proteomic profiling of MMCs will identify aberrant signaling pathways in the tumor cell and host tumor milieu to enhance our understanding of disease pathogenesis and to identify novel molecular targets.121  Second, given the genomic complexity of MM, immune therapies including mAbs, vaccines, immune checkpoint blockade, and CAR-T cells, likely in combination, will be integrated into the treatment paradigm to enhance autologous, anti-MM memory immunity. Lastly, utilization of effective, well-tolerated, and rational combination targeted and immune therapies early in the disease course, in SMM, or even in monoclonal gammopathy of undetermined significance, will delay and may ultimately avoid the development of MM.122-124 

The authors thank their colleagues in the MM research community and gratefully acknowledge the administrative assistance of Michelle Maglio in the preparation of this manuscript.

Contribution: G.B., P.G.R., and K.C.A. wrote the paper.

Conflict-of-interest disclosure: G.B. declares no competing financial interests. P.G.R. has served on advisory committees for Celgene, Millennium Pharmaceuticals, Johnson & Johnson, Bristol-Myers Squibb, Novartis, Genmab, Triphase Accelerator, and Onyx Pharmaceuticals. K.C.A. has served on advisory boards for Celgene, Millennium Pharmaceuticals, Bristol-Myers Squibb, Gilead Sciences, and Sanofi; and is a scientific founder of Acetylon Pharmaceuticals and Oncopep.

Correspondence: Kenneth C. Anderson, Dana-Farber Cancer Institute, Mayer 557, 450 Brookline Ave, Boston, MA, 02215; e-mail: kenneth_anderson@dfci.harvard.edu.

1
Kyle
 
RA
Rajkumar
 
SV
Multiple myeloma.
Blood
2008
, vol. 
111
 
6
(pg. 
2962
-
2972
)
2
Attal
 
M
Harousseau
 
JL
Stoppa
 
AM
, et al. 
A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. Intergroupe Français du Myélome.
N Engl J Med
1996
, vol. 
335
 
2
(pg. 
91
-
97
)
3
Kumar
 
SK
Rajkumar
 
SV
Dispenzieri
 
A
, et al. 
Improved survival in multiple myeloma and the impact of novel therapies.
Blood
2008
, vol. 
111
 
5
(pg. 
2516
-
2520
)
4
Singhal
 
S
Mehta
 
J
Desikan
 
R
, et al. 
Antitumor activity of thalidomide in refractory multiple myeloma.
N Engl J Med
1999
, vol. 
341
 
21
(pg. 
1565
-
1571
)
5
Richardson
 
PG
Barlogie
 
B
Berenson
 
J
, et al. 
A phase 2 study of bortezomib in relapsed, refractory myeloma.
N Engl J Med
2003
, vol. 
348
 
26
(pg. 
2609
-
2617
)
6
Richardson
 
PG
Blood
 
E
Mitsiades
 
CS
, et al. 
A randomized phase 2 study of lenalidomide therapy for patients with relapsed or relapsed and refractory multiple myeloma.
Blood
2006
, vol. 
108
 
10
(pg. 
3458
-
3464
)
7
Stewart
 
AK
Richardson
 
PG
San-Miguel
 
JF
How I treat multiple myeloma in younger patients.
Blood
2009
, vol. 
114
 
27
(pg. 
5436
-
5443
)
8
Hideshima
 
T
Richardson
 
P
Chauhan
 
D
, et al. 
The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells.
Cancer Res
2001
, vol. 
61
 
7
(pg. 
3071
-
3076
)
9
Obeng
 
EA
Carlson
 
LM
Gutman
 
DM
Harrington
 
WJ
Lee
 
KP
Boise
 
LH
Proteasome inhibitors induce a terminal unfolded protein response in multiple myeloma cells.
Blood
2006
, vol. 
107
 
12
(pg. 
4907
-
4916
)
10
Bianchi
 
G
Oliva
 
L
Cascio
 
P
, et al. 
The proteasome load versus capacity balance determines apoptotic sensitivity of multiple myeloma cells to proteasome inhibition.
Blood
2009
, vol. 
113
 
13
(pg. 
3040
-
3049
)
11
Mohty
 
M
Malard
 
F
Mohty
 
B
Savani
 
B
Moreau
 
P
Terpos
 
E
The effects of bortezomib on bone disease in patients with multiple myeloma.
Cancer
2014
, vol. 
120
 
5
(pg. 
618
-
623
)
12
Roccaro
 
AM
Hideshima
 
T
Raje
 
N
, et al. 
Bortezomib mediates antiangiogenesis in multiple myeloma via direct and indirect effects on endothelial cells.
Cancer Res
2006
, vol. 
66
 
1
(pg. 
184
-
191
)
13
Richardson
 
PG
Sonneveld
 
P
Schuster
 
MW
, et al. 
Assessment of Proteasome Inhibition for Extending Remissions (APEX) Investigators
Bortezomib or high-dose dexamethasone for relapsed multiple myeloma.
N Engl J Med
2005
, vol. 
352
 
24
(pg. 
2487
-
2498
)
14
San Miguel
 
JF
Schlag
 
R
Khuageva
 
NK
, et al. 
VISTA Trial Investigators
Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma.
N Engl J Med
2008
, vol. 
359
 
9
(pg. 
906
-
917
)
15
Richardson
 
PG
Weller
 
E
Lonial
 
S
, et al. 
Lenalidomide, bortezomib, and dexamethasone combination therapy in patients with newly diagnosed multiple myeloma.
Blood
2010
, vol. 
116
 
5
(pg. 
679
-
686
)
16
Siegel
 
DS
Martin
 
T
Wang
 
M
, et al. 
A phase 2 study of single-agent carfilzomib (PX-171-003-A1) in patients with relapsed and refractory multiple myeloma.
Blood
2012
, vol. 
120
 
14
(pg. 
2817
-
2825
)
17
Vij
 
R
Wang
 
M
Kaufman
 
JL
, et al. 
An open-label, single-arm, phase 2 (PX-171-004) study of single-agent carfilzomib in bortezomib-naive patients with relapsed and/or refractory multiple myeloma.
Blood
2012
, vol. 
119
 
24
(pg. 
5661
-
5670
)
18
Richardson
 
PG
Sonneveld
 
P
Schuster
 
M
, et al. 
Extended follow-up of a phase 3 trial in relapsed multiple myeloma: final time-to-event results of the APEX trial.
Blood
2007
, vol. 
110
 
10
(pg. 
3557
-
3560
)
19
Jakubowiak
 
AJ
Dytfeld
 
D
Griffith
 
KA
, et al. 
A phase 1/2 study of carfilzomib in combination with lenalidomide and low-dose dexamethasone as a frontline treatment for multiple myeloma.
Blood
2012
, vol. 
120
 
9
(pg. 
1801
-
1809
)
20
Chauhan
 
D
Tian
 
Z
Zhou
 
B
, et al. 
In vitro and in vivo selective antitumor activity of a novel orally bioavailable proteasome inhibitor MLN9708 against multiple myeloma cells.
Clin Cancer Res
2011
, vol. 
17
 
16
(pg. 
5311
-
5321
)
21
Tian
 
Z
Zhao
 
JJ
Tai
 
YT
, et al. 
Investigational agent MLN9708/2238 targets tumor-suppressor miR33b in MM cells.
Blood
2012
, vol. 
120
 
19
(pg. 
3958
-
3967
)
22
Kumar
 
SK
Bensinger
 
WI
Zimmerman
 
TM
, et al. 
Phase 1 study of weekly dosing with the investigational oral proteasome inhibitor ixazomib in relapsed/refractory multiple myeloma.
Blood
2014
, vol. 
124
 
7
(pg. 
1047
-
1055
)
23
Richardson
 
PG
Baz
 
R
Wang
 
M
, et al. 
Phase 1 study of twice-weekly ixazomib, an oral proteasome inhibitor, in relapsed/refractory multiple myeloma patients.
Blood
2014
, vol. 
124
 
7
(pg. 
1038
-
1046
)
24
Kumar
 
SK
Berdeja
 
JG
Niesvizky
 
R
, et al. 
Safety and tolerability of ixazomib, an oral proteasome inhibitor, in combination with lenalidomide and dexamethasone in patients with previously untreated multiple myeloma: an open-label phase 1/2 study.
Lancet Oncol
2014
, vol. 
15
 
13
(pg. 
1503
-
1512
)
25
Kumar
 
S
Berdeja
 
JG
Niesvizky
 
R
, et al. 
 
Long-term ixazomib maintenance is tolerable and improves depth of response following ixazomib-lenalidomide-dexamethasone induction in patients (Pts) with previously untreated multiple myeloma (MM): phase 2 study results [abstract]. Blood. 2014;124(21). Abstract 82
26
Chauhan
 
D
Singh
 
AV
Aujay
 
M
, et al. 
A novel orally active proteasome inhibitor ONX 0912 triggers in vitro and in vivo cytotoxicity in multiple myeloma.
Blood
2010
, vol. 
116
 
23
(pg. 
4906
-
4915
)
27
Hurchla
 
MA
Garcia-Gomez
 
A
Hornick
 
MC
, et al. 
The epoxyketone-based proteasome inhibitors carfilzomib and orally bioavailable oprozomib have anti-resorptive and bone-anabolic activity in addition to anti-myeloma effects.
Leukemia
2013
, vol. 
27
 
2
(pg. 
430
-
440
)
28
Vij
 
R
Savona
 
M
Siegel
 
DS
, et al. 
 
Clinical profile of single-agent oprozomib in patients (Pts) with multiple myeloma (MM): updated results from a multicenter, open-label, dose escalation phase 1b/2 study [abstract]. Blood. 2014;124(21). Abstract 34
29
Hari
 
PN
Shain
 
KH
Voorhees
 
PM
, et al. 
 
Oprozomib and dexamethasone in patients with relapsed and/or refractory multiple myeloma: initial results from the dose escalation portion of a phase 1b/2, multicenter, open-label study [abstract]. Blood. 2014;124(21). Abstract 3453
30
Chauhan
 
D
Catley
 
L
Li
 
G
, et al. 
A novel orally active proteasome inhibitor induces apoptosis in multiple myeloma cells with mechanisms distinct from Bortezomib.
Cancer Cell
2005
, vol. 
8
 
5
(pg. 
407
-
419
)
31
Richardson
 
PG
Spencer
 
A
Cannell
 
P
, et al. 
 
Phase 1 clinical evaluation of twice-weekly marizomib (NPI-0052), a novel proteasome inhibitor, in patients with relapsed/refractory multiple myeloma (MM) [abstract]. Blood. 2011;118(21). Abstract 302
32
Das
 
DS
Ravillah
 
D
Ray
 
A
, et al. 
 
Synergistic anti-myeloma activity of a proteasome inhibitor marizomib and IMiD immunomodulatory drug pomalidomide [abstract]. Blood. 2014;124(21). Abstract 2099
33
Peth
 
A
Besche
 
HC
Goldberg
 
AL
Ubiquitinated proteins activate the proteasome by binding to Usp14/Ubp6, which causes 20S gate opening.
Mol Cell
2009
, vol. 
36
 
5
(pg. 
794
-
804
)
34
Komander
 
D
Clague
 
MJ
Urbé
 
S
Breaking the chains: structure and function of the deubiquitinases.
Nat Rev Mol Cell Biol
2009
, vol. 
10
 
8
(pg. 
550
-
563
)
35
Tian
 
Z
D’Arcy
 
P
Wang
 
X
, et al. 
A novel small molecule inhibitor of deubiquitylating enzyme USP14 and UCHL5 induces apoptosis in multiple myeloma and overcomes bortezomib resistance.
Blood
2014
, vol. 
123
 
5
(pg. 
706
-
716
)
36
Song
 
Y
Das
 
DS
Ray
 
A
, et al. 
 
Targeting proteasome ubiquitin receptor RPN13/ADRM1 in multiple myeloma [abstract]. Blood. 2014;124(21). Abstract 419
37
Chauhan
 
D
Tian
 
Z
Nicholson
 
B
, et al. 
A small molecule inhibitor of ubiquitin-specific protease-7 induces apoptosis in multiple myeloma cells and overcomes bortezomib resistance.
Cancer Cell
2012
, vol. 
22
 
3
(pg. 
345
-
358
)
38
D’Arcy
 
P
Brnjic
 
S
Olofsson
 
MH
, et al. 
Inhibition of proteasome deubiquitinating activity as a new cancer therapy.
Nat Med
2011
, vol. 
17
 
12
(pg. 
1636
-
1640
)
39
Bolden
 
JE
Peart
 
MJ
Johnstone
 
RW
Anticancer activities of histone deacetylase inhibitors.
Nat Rev Drug Discov
2006
, vol. 
5
 
9
(pg. 
769
-
784
)
40
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 USA
2005
, vol. 
102
 
24
(pg. 
8567
-
8572
)
41
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.
Lancet Oncol
2013
, vol. 
14
 
11
(pg. 
1129
-
1140
)
42
Siegel
 
DS
Richardson
 
P
Dimopoulos
 
M
, et al. 
Vorinostat in combination with lenalidomide and dexamethasone in patients with relapsed or refractory multiple myeloma.
Blood Cancer J
2014
, vol. 
4
 pg. 
e182
 
43
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
, vol. 
122
 
14
(pg. 
2331
-
2337
)
44
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
, vol. 
15
 
11
(pg. 
1195
-
1206
)
45
Kawaguchi
 
Y
Kovacs
 
JJ
McLaurin
 
A
Vance
 
JM
Ito
 
A
Yao
 
TP
The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress.
Cell
2003
, vol. 
115
 
6
(pg. 
727
-
738
)
46
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
, vol. 
119
 
11
(pg. 
2579
-
2589
)
47
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]. Blood. 2014;124(21). Abstract 4764
48
Hideshima
 
T
Cottini
 
F
Ohguchi
 
H
, et al. 
Rational combination treatment with histone deacetylase inhibitors and immunomodulatory drugs in multiple myeloma.
Blood Cancer J
2015
, vol. 
5
 pg. 
e312
 
49
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 [abstract]. Blood. 2014;124(21). Abstract 4772
50
Minami
 
J
Suzuki
 
R
Mazitschek
 
R
, et al. 
Histone deacetylase 3 as a novel therapeutic target in multiple myeloma.
Leukemia
2014
, vol. 
28
 
3
(pg. 
680
-
689
)
51
Davies
 
FE
Raje
 
N
Hideshima
 
T
, et al. 
Thalidomide and immunomodulatory derivatives augment natural killer cell cytotoxicity in multiple myeloma.
Blood
2001
, vol. 
98
 
1
(pg. 
210
-
216
)
52
Richardson
 
PG
Siegel
 
DS
Vij
 
R
, et al. 
Pomalidomide alone or in combination with low-dose dexamethasone in relapsed and refractory multiple myeloma: a randomized phase 2 study.
Blood
2014
, vol. 
123
 
12
(pg. 
1826
-
1832
)
53
Benboubker
 
L
Dimopoulos
 
MA
Dispenzieri
 
A
, et al. 
FIRST Trial Team
Lenalidomide and dexamethasone in transplant-ineligible patients with myeloma.
N Engl J Med
2014
, vol. 
371
 
10
(pg. 
906
-
917
)
54
Attal
 
M
Lauwers
 
VC
Marit
 
G
, et al. 
Maintenance treatment with lenalidomide after transplantation for myeloma: final analysis of the IFM 2005-02 [abstract].
Blood
2010
, vol. 
116
 
21
 
Abstract 310
55
McCarthy
 
PL
Owzar
 
K
Hofmeister
 
CC
, et al. 
Lenalidomide after stem-cell transplantation for multiple myeloma.
N Engl J Med
2012
, vol. 
366
 
19
(pg. 
1770
-
1781
)
56
Bianchi
 
G
Richardson
 
PG
Anderson
 
KC
Best treatment strategies in high-risk multiple myeloma: navigating a gray area.
J Clin Oncol
2014
, vol. 
32
 
20
(pg. 
2125
-
2132
)
57
Davies
 
F
Baz
 
R
Lenalidomide mode of action: linking bench and clinical findings.
Blood Rev
2010
, vol. 
24
 
suppl 1
(pg. 
S13
-
S19
)
58
Krönke
 
J
Udeshi
 
ND
Narla
 
A
, et al. 
Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells.
Science
2014
, vol. 
343
 
6168
(pg. 
301
-
305
)
59
Lu
 
G
Middleton
 
RE
Sun
 
H
, et al. 
The myeloma drug lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins.
Science
2014
, vol. 
343
 
6168
(pg. 
305
-
309
)
60
Zhu
 
YX
Braggio
 
E
Shi
 
CX
, et al. 
Identification of cereblon-binding proteins and relationship with response and survival after IMiDs in multiple myeloma.
Blood
2014
, vol. 
124
 
4
(pg. 
536
-
545
)
61
Luptakova
 
K
Rosenblatt
 
J
Glotzbecker
 
B
, et al. 
Lenalidomide enhances anti-myeloma cellular immunity.
Cancer Immunol Immunother
2013
, vol. 
62
 
1
(pg. 
39
-
49
)
62
Tai
 
YT
Dillon
 
M
Song
 
W
, et al. 
Anti-CS1 humanized monoclonal antibody HuLuc63 inhibits myeloma cell adhesion and induces antibody-dependent cellular cytotoxicity in the bone marrow milieu.
Blood
2008
, vol. 
112
 
4
(pg. 
1329
-
1337
)
63
Lonial
 
S
Vij
 
R
Harousseau
 
JL
, et al. 
Elotuzumab in combination with lenalidomide and low-dose dexamethasone in relapsed or refractory multiple myeloma.
J Clin Oncol
2012
, vol. 
30
 
16
(pg. 
1953
-
1959
)
64
Richardson
 
PG
Jagannath
 
S
Moreau
 
P
, et al. 
 
Final results for the 1703 phase 1b/2 study of elotuzumab in combination with lenalidomide and dexamethasone in patients with relapsed/refractory multiple myeloma [abstract]. Blood. 2014;124(21). Abstract 302
65
de Weers
 
M
Tai
 
YT
van der Veer
 
MS
, et al. 
Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors.
J Immunol
2011
, vol. 
186
 
3
(pg. 
1840
-
1848
)
66
Laubach
 
JP
Tai
 
YT
Richardson
 
PG
Anderson
 
KC
Daratumumab granted breakthrough drug status.
Expert Opin Investig Drugs
2014
, vol. 
23
 
4
(pg. 
445
-
452
)
67
Laubach
 
JP
Richardson
 
PG
CD38-targeted immunochemotherapy in refractory multiple myeloma: a new horizon [published online ahead of print April 15, 2015].
Clin Cancer Res
68
Plesner
 
T
Arkenau
 
H-T
Lokhorst
 
HM
, et al. 
 
Safety and efficacy of daratumumab with lenalidomide and dexamethasone in relapsed or relapsed, refractory multiple myeloma [abstract]. Blood. 2014;124(21). Abstract 84
69
Martin
 
TG
Baz
 
R
Benson
 
DM
, et al. 
 
A phase Ib dose escalation trial of SAR650984 (anti-CD-38 mAb) in combination with lenalidomide and dexamethasone in relapsed/refractory multiple myeloma [abstract]. Blood. 2014;124(21). Abstract 83
70
Lopus
 
M
Oroudjev
 
E
Wilson
 
L
, et al. 
Maytansine and cellular metabolites of antibody-maytansinoid conjugates strongly suppress microtubule dynamics by binding to microtubules.
Mol Cancer Ther
2010
, vol. 
9
 
10
(pg. 
2689
-
2699
)
71
Ikeda
 
H
Hideshima
 
T
Fulciniti
 
M
, et al. 
The monoclonal antibody nBT062 conjugated to cytotoxic Maytansinoids has selective cytotoxicity against CD138-positive multiple myeloma cells in vitro and in vivo.
Clin Cancer Res
2009
, vol. 
15
 
12
(pg. 
4028
-
4037
)
72
Kelly
 
KR
Chanan-Khan
 
A
Heffner
 
LT
, et al. 
 
Indatuximab ravtansine (BT062) in combination with lenalidomide and low-dose dexamethasone in patients with relapsed and/or refractory multiple myeloma: clinical activity in patients already exposed to lenalidomide and bortezomib [abstract]. Blood. 2014;124(21). Abstract 4736
73
Tai
 
YT
Mayes
 
PA
Acharya
 
C
, et al. 
Novel anti-B-cell maturation antigen antibody-drug conjugate (GSK2857916) selectively induces killing of multiple myeloma.
Blood
2014
, vol. 
123
 
20
(pg. 
3128
-
3138
)
74
Pardoll
 
DM
The blockade of immune checkpoints in cancer immunotherapy.
Nat Rev Cancer
2012
, vol. 
12
 
4
(pg. 
252
-
264
)
75
Arnason
 
J
Avigan
 
D
Evolution of cellular immunotherapy: from allogeneic transplant to dendritic cell vaccination as treatment for multiple myeloma.
Immunotherapy
2012
, vol. 
4
 
10
(pg. 
1043
-
1051
)
76
Ansell
 
SM
Lesokhin
 
AM
Borrello
 
I
, et al. 
PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma.
N Engl J Med
2015
, vol. 
372
 
4
(pg. 
311
-
319
)
77
Gorgun
 
G
Samur
 
MK
Cowens
 
KB
, et al. 
Lenalidomide enhances immune checkpoint blockade induced immune response in multiple myeloma [published online ahead of print May 15, 2015].
Clin Cancer Res
78
Ray
 
A
Das
 
DS
Song
 
Y
, et al. 
Targeting PD1-PDL1 immune checkpoint in plasmacytoid dendritic cell interactions with T cells, natural killer cells and multiple myeloma cells [published online ahead of print January 30, 2015].
Leukemia
79
Bae
 
J
Prabhala
 
R
Voskertchian
 
A
, et al. 
A multiepitope of XBP1, CD138 and CS1 peptides induces myeloma-specific cytotoxic T lymphocytes in T cells of smoldering myeloma patients.
Leukemia
2015
, vol. 
29
 
1
(pg. 
218
-
229
)
80
Rosenblatt
 
J
Vasir
 
B
Uhl
 
L
, et al. 
Vaccination with dendritic cell/tumor fusion cells results in cellular and humoral antitumor immune responses in patients with multiple myeloma.
Blood
2011
, vol. 
117
 
2
(pg. 
393
-
402
)
81
Rosenblatt
 
J
Avivi
 
I
Vasir
 
B
, et al. 
Vaccination with dendritic cell/tumor fusions following autologous stem cell transplant induces immunologic and clinical responses in multiple myeloma patients.
Clin Cancer Res
2013
, vol. 
19
 
13
(pg. 
3640
-
3648
)
82
Chauhan
 
D
Singh
 
AV
Brahmandam
 
M
, et al. 
Functional interaction of plasmacytoid dendritic cells with multiple myeloma cells: a therapeutic target.
Cancer Cell
2009
, vol. 
16
 
4
(pg. 
309
-
323
)
83
Ray
 
A
Tian
 
Z
Das
 
DS
, et al. 
A novel TLR-9 agonist C792 inhibits plasmacytoid dendritic cell-induced myeloma cell growth and enhance cytotoxicity of bortezomib.
Leukemia
2014
, vol. 
28
 
8
(pg. 
1716
-
1724
)
84
Maude
 
SL
Frey
 
N
Shaw
 
PA
, et al. 
Chimeric antigen receptor T cells for sustained remissions in leukemia.
N Engl J Med
2014
, vol. 
371
 
16
(pg. 
1507
-
1517
)
85
Porter
 
DL
Levine
 
BL
Kalos
 
M
Bagg
 
A
June
 
CH
Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia.
N Engl J Med
2011
, vol. 
365
 
8
(pg. 
725
-
733
)
86
Maus
 
MV
June
 
CH
Zoom Zoom: racing CARs for multiple myeloma.
Clin Cancer Res
2013
, vol. 
19
 
8
(pg. 
1917
-
1919
)
87
Huehls
 
AM
Coupet
 
TA
Sentman
 
CL
Bispecific T-cell engagers for cancer immunotherapy.
Immunol Cell Biol
2015
, vol. 
93
 
3
(pg. 
290
-
296
)
88
Topp
 
MS
Gökbuget
 
N
Stein
 
AS
, et al. 
Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study.
Lancet Oncol
2015
, vol. 
16
 
1
(pg. 
57
-
66
)
89
Bianchi
 
G
Anderson
 
KC
Understanding biology to tackle the disease: Multiple myeloma from bench to bedside, and back.
CA Cancer J Clin
2014
, vol. 
64
 
6
(pg. 
422
-
444
)
90
Alsayed
 
Y
Ngo
 
H
Runnels
 
J
, et al. 
Mechanisms of regulation of CXCR4/SDF-1 (CXCL12)-dependent migration and homing in multiple myeloma.
Blood
2007
, vol. 
109
 
7
(pg. 
2708
-
2717
)
91
Azab
 
AK
Runnels
 
JM
Pitsillides
 
C
, et al. 
CXCR4 inhibitor AMD3100 disrupts the interaction of multiple myeloma cells with the bone marrow microenvironment and enhances their sensitivity to therapy.
Blood
2009
, vol. 
113
 
18
(pg. 
4341
-
4351
)
92
Flomenberg
 
N
Devine
 
SM
Dipersio
 
JF
, et al. 
The use of AMD3100 plus G-CSF for autologous hematopoietic progenitor cell mobilization is superior to G-CSF alone.
Blood
2005
, vol. 
106
 
5
(pg. 
1867
-
1874
)
93
Roccaro
 
AM
Sacco
 
A
Purschke
 
WG
, et al. 
SDF-1 inhibition targets the bone marrow niche for cancer therapy.
Cell Reports
2014
, vol. 
9
 
1
(pg. 
118
-
128
)
94
Ludwig
 
H
Weisel
 
K
Petrucci
 
MT
, et al. 
 
Final results from the phase IIa study of the anti-CXCL12 Spiegelmer olaptesed pegol (NOX-A12) in combination with bortezomib and dexamethasone in patients with multiple myeloma [abstract]. Blood. 2014;124(21). Abstract 2111
95
Colla
 
S
Storti
 
P
Donofrio
 
G
, et al. 
Low bone marrow oxygen tension and hypoxia-inducible factor-1α overexpression characterize patients with multiple myeloma: role on the transcriptional and proangiogenic profiles of CD138(+) cells.
Leukemia
2010
, vol. 
24
 
11
(pg. 
1967
-
1970
)
96
Azab
 
AK
Hu
 
J
Quang
 
P
, et al. 
Hypoxia promotes dissemination of multiple myeloma through acquisition of endothelial to mesenchymal transition-like features.
Blood
 
2012; 119(24):5782-5794
97
Hu
 
J
Van Valckenborgh
 
E
Xu
 
D
, et al. 
Synergistic induction of apoptosis in multiple myeloma cells by bortezomib and hypoxia-activated prodrug TH-302, in vivo and in vitro.
Mol Cancer Ther
2013
, vol. 
12
 
9
(pg. 
1763
-
1773
)
98
Laubach
 
JP
Raje
 
NS
Yee
 
AJ
, et al. 
 
A phase 1/2 trial of TH-302 and dexamethasone without or with bortezomib (TBorD) in patients with relapsed/refractory multiple myeloma [abstract]. Blood. 2014;124(21). Abstract 2142
99
Ocio
 
EM
Richardson
 
PG
Rajkumar
 
SV
, et al. 
New drugs and novel mechanisms of action in multiple myeloma in 2013: a report from the International Myeloma Working Group (IMWG).
Leukemia
2014
, vol. 
28
 
3
(pg. 
525
-
542
)
100
Tunquist
 
BJ
Woessner
 
RD
Walker
 
DH
Mcl-1 stability determines mitotic cell fate of human multiple myeloma tumor cells treated with the kinesin spindle protein inhibitor ARRY-520.
Mol Cancer Ther
2010
, vol. 
9
 
7
(pg. 
2046
-
2056
)
101
Lonial
 
S
Shah
 
JJ
Zonder
 
J
, et al. 
 
Prolonged survival and improved response rates with ARRY-520 in relapsed/refractory multiple myeloma (RRMM) patients with low α-1 acid glycoprotein (AAG) levels: results from a phase 2 study [abstract]. Blood. 2013;122(21). Abstract 285
102
Tai
 
YT
Landesman
 
Y
Acharya
 
C
, et al. 
CRM1 inhibition induces tumor cell cytotoxicity and impairs osteoclastogenesis in multiple myeloma: molecular mechanisms and therapeutic implications.
Leukemia
2014
, vol. 
28
 
1
(pg. 
155
-
165
)
103
Turner
 
JG
Dawson
 
J
Cubitt
 
CL
Baz
 
R
Sullivan
 
DM
Inhibition of CRM1-dependent nuclear export sensitizes malignant cells to cytotoxic and targeted agents.
Semin Cancer Biol
2014
, vol. 
27
 (pg. 
62
-
73
)
104
Chen
 
CI
Gutierrez
 
M
Siegel
 
DS
, et al. 
 
Selinexor demonstrates marked synergy with dexamethasone (Sel-Dex) in preclinical models and in patients with heavily pretreated refractory multiple myeloma (MM) [abstract]. Blood. 2014;124(21). Abstract 4773
105
Hideshima
 
T
Catley
 
L
Yasui
 
H
, et al. 
Perifosine, an oral bioactive novel alkylphospholipid, inhibits Akt and induces in vitro and in vivo cytotoxicity in human multiple myeloma cells.
Blood
2006
, vol. 
107
 
10
(pg. 
4053
-
4062
)
106
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
, vol. 
106
 
1
(pg. 
296
-
303
)
107
Semenov
 
I
Akyuz
 
C
Roginskaya
 
V
Chauhan
 
D
Corey
 
SJ
Growth inhibition and apoptosis of myeloma cells by the CDK inhibitor flavopiridol.
Leuk Res
2002
, vol. 
26
 
3
(pg. 
271
-
280
)
108
Kumar
 
SK
LaPlant
 
B
Chng
 
WJ
, et al. 
Mayo Phase 2 Consortium
Dinaciclib, a novel CDK inhibitor, demonstrates encouraging single-agent activity in patients with relapsed multiple myeloma.
Blood
2015
, vol. 
125
 
3
(pg. 
443
-
448
)
109
Raje
 
N
Kumar
 
S
Hideshima
 
T
, et al. 
Seliciclib (CYC202 or R-roscovitine), a small-molecule cyclin-dependent kinase inhibitor, mediates activity via down-regulation of Mcl-1 in multiple myeloma.
Blood
2005
, vol. 
106
 
3
(pg. 
1042
-
1047
)
110
Conroy
 
A
Stockett
 
DE
Walker
 
D
, et al. 
SNS-032 is a potent and selective CDK 2, 7 and 9 inhibitor that drives target modulation in patient samples.
Cancer Chemother Pharmacol
2009
, vol. 
64
 
4
(pg. 
723
-
732
)
111
Gabay
 
M
Li
 
Y
Felsher
 
DW
 
MYC activation is a hallmark of cancer initiation and maintenance. Cold Spring Harb Perspect Med. 2014;4(6)
112
Delmore
 
JE
Issa
 
GC
Lemieux
 
ME
, et al. 
BET bromodomain inhibition as a therapeutic strategy to target c-Myc.
Cell
2011
, vol. 
146
 
6
(pg. 
904
-
917
)
113
Cottini
 
F
Hideshima
 
T
Xu
 
C
, et al. 
Rescue of Hippo coactivator YAP1 triggers DNA damage-induced apoptosis in hematological cancers.
Nat Med
2014
, vol. 
20
 
6
(pg. 
599
-
606
)
114
Cottini
 
F
Hideshima
 
T
Buhrlage
 
S
, et al. 
 
Serine/threonine kinase STK4 is a novel target in myeloma [abstract]. Blood. 2014;124(21). Abstract 645
115
Neri
 
P
Bahlis
 
NJ
Genomic instability in multiple myeloma: mechanisms and therapeutic implications.
Expert Opin Biol Ther
2013
, vol. 
13
 
suppl 1
(pg. 
S69
-
S82
)
116
Fonseca
 
R
Bailey
 
RJ
Ahmann
 
GJ
, et al. 
Genomic abnormalities in monoclonal gammopathy of undetermined significance.
Blood
2002
, vol. 
100
 
4
(pg. 
1417
-
1424
)
117
Bolli
 
N
Avet-Loiseau
 
H
Wedge
 
DC
, et al. 
Heterogeneity of genomic evolution and mutational profiles in multiple myeloma.
Nat Commun
2014
, vol. 
5
 pg. 
2997
 
118
Andrulis
 
M
Lehners
 
N
Capper
 
D
, et al. 
Targeting the BRAF V600E mutation in multiple myeloma.
Cancer Discov
2013
, vol. 
3
 
8
(pg. 
862
-
869
)
119
Rashid
 
NU
Sperling
 
AS
Bolli
 
N
, et al. 
Differential and limited expression of mutant alleles in multiple myeloma.
Blood
2014
, vol. 
124
 
20
(pg. 
3110
-
3117
)
120
Nowell
 
PC
The clonal evolution of tumor cell populations.
Science
1976
, vol. 
194
 
4260
(pg. 
23
-
28
)
121
Bianchi
 
G
Munshi
 
NC
Pathogenesis beyond the cancer clone(s) in multiple myeloma.
Blood
2015
, vol. 
125
 
20
(pg. 
3049
-
3058
)
122
Laubach
 
JP
Voorhees
 
PM
Hassoun
 
H
Jakubowiak
 
A
Lonial
 
S
Richardson
 
PG
Current strategies for treatment of relapsed/refractory multiple myeloma.
Expert Rev Hematol
2014
, vol. 
7
 
1
(pg. 
97
-
111
)
123
Ocio
 
EM
Mitsiades
 
CS
Orlowski
 
RZ
Anderson
 
KC
Future agents and treatment directions in multiple myeloma.
Expert Rev Hematol
2014
, vol. 
7
 
1
(pg. 
127
-
141
)
124
Usmani
 
SZ
How long can we let the myeloma smolder?
Expert Rev Hematol
2014
, vol. 
7
 
1
(pg. 
17
-
19
)
Sign in via your Institution