Abstract
The therapeutic landscape of multiple myeloma (MM) has dramatically changed in the last 15 years with the advent of Imids and proteasome inhibitors. However, majority of MM patients relapse and new therapies are needed. Various agents with diverse mechanisms of action and distinct targets, including cellular therapies, monoclonal antibodies, and small molecules are currently under investigation. In this review, we report novel drugs recently approved or under advanced investigation that will likely be incorporated in the near future as new standard for MM treatment, focusing on their mechanisms of action, cellular targets and stage of development.
The advent of Imids and proteasome inhibitors opened a new era in the therapy of multiple myeloma (MM) contributing to significantly prolongation of survival. In the last five years, the Food and Drug Administration approved six new drugs, pomalidomide (an analogue of lenalidomide), carfilzomib, ixazomib (two new proteasome inhibitors (PI)), panobinostat (a pan histone deacetylase) and two monoclonal antibodies, daratumumab (targeting CD38) and elotuzumab (targeting the signaling lymphocytic activation molecule F7 (SLAMF7)) (table 1). One of the new and most exciting challenges is now to identify the most efficient combinations of these new agents in induction and maintenance regimen, and to define the best therapeutic strategies according to risk stratification and patient characteristics1. Nevertheless, a subgroup of high-risk patients remain characterized by poor survival despite these most recent available treatment combinations and most patients continue to experience relapses, underlying the need for new active treatments to cure the disease2.
Table 1:
Drug | Class | Target |
---|---|---|
Melphalan | Alkylating agent | DNA |
BiCNU | Alkylating agent | DNA |
Cyclophosphamide | Alkylating agent | DNA |
Doxorubicin | Anthracyclin | DNA |
Thalidomide | IMid | Cereblon/E3 ubiquitin ligase complex |
Lenalidomide | IMid | Cereblon/E3 ubiquitin ligase complex |
Bendamustine | Alkylating agent Purine analog | DNA |
Bortezomib | Reversible proteasome Inhibitor | Proteasome 20s unit |
Ixazomib | Reversible proteasome Inhibitor | proteasome 20s unit |
Carfilzomib | Irreversible Proteasome Inhibitor | proteasome 20s subunit inhibition |
Daratumumab | Moncolnal antibody | CD38 |
Pomalidomide | IMid | Cereblon/E3 ubiquitin ligase complex |
Elotuzumab | Monoclonal antibody | CS1 |
Panobinostat | HDCA inhibitor | Pan-HDAC |
The study of MM genomics along with the discovery of new critical signaling pathways directing MM cell survival has allowed development of new and efficient therapeutic agents. Intracellular targets such as protein degradation, apoptosis pathway, transcriptomic regulation are essential mechanisms of MM cell survival and represent new therapeutic avenues3. In addition, the rise of immunotherapy with the development of monoclonal antibodies directly targeting tumor cells such as daratumumab or isatuximab (anti-CD38 mAb) or elotuzumab (targeting SLAMF7)4,5, monoclonal antibodies targeting immune checkpoints (such as pembrolizumab and nivolumab targeting PD1) and cellular therapies with Chimeric antigen receptors T cells (CAR T cells) are now under critical evaluation in myeloma6. In this review, we focus on the newest agents most recently approved or in advanced development in myeloma, excluding cellular immunotherapies and protein catabolism targeted therapies that are detailed elsewhere in this issue. For a matter of clarity, we present these new agents based on the their mechanisms of action or therapeutic target (table 2 and table 3).
Table 2:
Agent | Class | Target | ClinicalTrials.gov Identifier | Clinical Phase | Single/Combination |
---|---|---|---|---|---|
Anti-CD38 CAR-T | Cellular | CAR-T CD 38 | NCT03464916 | Phase 1 | Single |
bb21217 | Cellular | CAR-T BCMA | NCT03274219 | Phase 1 | Single |
KITE-585 | Cellular | CAR-T BCMA | NCT03318861 | Phase 1 | Single |
Kymriah | Cellular | CAR-T CD19 | NCT02529813 | Phase 1 | Single |
NAM-NK Cells | Cellular | NK Cells | NCT03019666 | Phase 1 | Single |
P-BCMA-101 | Cellular | CAR-T BCMA | NCT03288493 | Phase 1 | Single |
PNK-007 | cellular | Natural Killer Cell | NCT02955550 | Phase 1 | Single |
CYAD-01 | Cellular | CAR-T-NKG2D | NCT02203825 | Phase 1 | Single |
AUTO2 | Cellular | CAR-T BCMA | NCT03287804 | Phase1–2 | Single |
JCARH125 | Cellular | CAR-T BCMA | NCT03430011 | Phase1–2 | Single |
LCAR-B38M | Cellular | CAR-T BCMA | NCT03090659 | Phase1–2 | Single |
bb2121 | Cellular | CAR-T BCMA | NCT03361748, NCT03601078, NCT02658929 | Phase 2 | Single |
AMG 424 | Mab-bi | CD3-CD38 | NCT03445663 | Phase 1 | Single |
CJM112 | Mab | IL-17 | NCT03111992 | Phase 1 | Single and combination |
DKN-01 | Mab | Dickkopf-1 (DKK-1) | NCT01457417 | Phase 1 | Single |
JNJ-64007957 | Mab-bi | CD3-BCMA | NCT03145181 | Phase 1 | Single |
JNJ-64407564 | Mab-bi | CD3-GPRC5D | NCT03399799 | Phase 1 | Single |
Lirilumab | Mab | KIR | NCT02252263 | Phase 1 | Combination with Elotuzumab |
MEDI2228 | Mab | Antibody-drug Conjugate anti BCMA | NCT03489525 | Phase 1 | Single |
PF-06863135 | Mab-bi | CD3-BCMA | NCT03269136 | Phase 1 | Single |
SEA-BCMA | Mab | BCMA | NCT03582033 | Phase 1 | Single |
SGN-CD48A | Mab | Antibody-drug Conjugate anti CD48 | NCT03379584 | Phase 1 | Single |
STRO-001 | Mab | Antibody-drug Conjugate anti CD74 | NCT03424603 | Phase 1 | Single |
AMG 701 | Mab-bi | CD-3 BCMA | NCT03287908 | Phase 1 | Single |
CC-93269 | Mab-bi | CD-3 BCMA | NCT03486067 | Phase 1 | Single |
BION-1301 | Mab | APRIL | NCT03340883 | Phase1–2 | Single |
BT-062 | Mab | Antibody-drug Conjugate anti CD 138 | NCT01001442, NCT01638936 | Phase1–2 | Single and combination with Len or Pom and Dex |
MOR202 | Mab | CD38 | NCT01421186 | Phase1–2 | Combination with Dex and Len or Pom |
TAK-079 | Mab | CD38 | NCT03439280 | Phase 1–2 | Single |
TAK-573 | Mab | CD38 | NCT03215030 | Phase1–2 | Single |
BHQ880 | Mab | Dickkopf-1 (DKK-1) | NCT01302886, NCT01337752 | Phase 2 | Single |
GSK2857916 | Mab | Antibody-drug Conjugate anti BCMA | NCT03525678 | Phase 2 | Single |
ABBV-838 | Mab | Antibody-drug Conjugate anti anti CS1 | NCT02462525 | Phase 1 | Combination with Pom and Dex |
ImMucin | Peptide | Mucin 1 | NCT01232712 | Phase1–2 | Single |
PVX-410 | Peptide | Vaccine | NCT02886065 | Phase 1 | Combination with Citarinostat and lenalidomide |
Novel Immunotherapeutic agents in multiple myeloma and stage of ongoing clinical trials (Mab= monoclonal antibody, Mab-bi=monoclonal antibody bispecific, Dex=dexamethasone, Len= lenalidomide, Pom=pomalidomide)
Table 3:
Agent | Class | Target | ClinicalTrials.gov Identifier | Clinical Phase | Single/Combination |
---|---|---|---|---|---|
ALT-801 | Protein | p53 | NCT01670994 | Phase1–2 | Single |
TAS4464 | Small Molecule | NEDD8 | NCT02978235 | Phase1–2 | Single |
Vemurafenib* | Small molecule | V600E BRAF mutated | NCT02693535 | Phase 2 | In combination with Cobimetinib** |
Dabrafenib and/or Trametinib | Samll molecule | BRAF and MAPK | NCT03091257 | Phase 1 | Single agent or combined |
AMG 176 | Small Molecule | MCL-1 | NCT02675452 | Phase 1 | Single |
CB-5083 | Small Molecule | Cdc48p/p97 | NCT02223598 | Phase 1 | |
CC-122 | Small Molecule | E3 ubiquitin ligase | NCT01421524 | Phase 1 | Combination with Dex |
CPI-0610 | Small Molecule | Bromodomains | NCT02157636 | Phase 1 | Single |
CX-4945 | Small Molecule | Protein kinase CK2/Casein Kinase II | NCT00891280, NCT01199718 | Phase 1 | Single |
Jakafi | Small Molecule | JAK/STAT | NCT03110822 | Phase 1 | Combination with Len and methyprednisolone |
LGH447 | Small Molecule | PIM Kinase Family | NCT02160951 | Phase 1 | Single |
Venetoclax | Small Molecule | BCL-2 | NCT01794520 | Phase 2 | Combination with Dex |
MIK665 | Small Molecule | MCL-1 | NCT02992483 | Phase 1 | Single |
TG02 | Small Molecule | multi-kinase inhibitor | NCT01204164 | Phase 1 | Combination with Carfilzomib and Dex |
Selinexor | Small Molecule | Exportin-1/CRM1/XPO1 | NCT02389543 | Phase1–2 | Combination with Len and Dex |
Iberdomide (CC-220) | Small Molecule | Cereblon E3 ubiquitin ligase complex | NCT02773030 | Phase1–2 | Single or Combination with Dex, with Dara/Dex or Bortezomib/Dex |
CC-92480 | Small molecule | Cereblon E3 ubiquitin ligase complex | NCT03374085 | Phase 1 | Combination with Dex |
ONC201 | Small Molecule | Dopamine 2 (D2) Receptor | NCT02863991 | Phase1–2 | Single |
Oprozomib | Small Molecule | Proteasome | NCT01832727 | Phase1–2 | Combination with Dex |
PT-112 | Small Molecule | Apoptosis | NCT03288480 | Phase1–2 | Single |
Ricolinostat | Small Molecule | Histone Deacetylase | NCT01997840 | Phase1–2 | Combination with Pom/Dex |
EDO-S101 | Small Molecule | Histone Deacetylase DNA | NCT02576496 | Phase | Single |
LCL161 | Small Molecule | Apoptosis proteins) | NCT01955434 | Phase 2 | Single or in combination with cyclophosphamide |
CLR 131 | Small Molecule | Radio-pharmaceutical | NCT02952508 | Phase 2 | Single |
Filanesib | Small molecule | kinesin spindle protein | NCT01372540 | Phase 1 | Combination with carfilzomib |
Afuresertib | Small molecule | AKT | NCT02235740 | Phase 1 | Combination with carfilzomib |
Pelareorep | Viral | Oncolytic Virus | NCT00450814 | Phase1–2 | Single or in combination with cyclophosphamide |
Melflufen | Melphalanderivative | DNA | NCT02963493 | Phase 2 | Combination with Dex |
Novel agents in multiple myeloma and stage of ongoing clinical trials (Mab= monoclonal antibody, Mab-bi=monoclonal antibody bispecific, Dex=dexamethasone, Len= lenalidomide, Pom=pomalidomide, * this trial is also evaluating 14 other targeted therapies and Vemurafenib is combined with cobimetinib (MEK1 inhibitor))
1-. Antibodies targeting MM cells
Anti-myeloma monoclonal antibodies are humanized or chimeric antibodies targeting malignant plasma cells that can correspond to monoclonal antibodies, drug-conjugated monoclonal antibodies or bi-specific antibodies.
A-. Monoclonal antibodies targeting CS1
Elotuzumab is a humanized immunoglobulin G1 immunostimulatory monoclonal antibody targeted against signaling lymphocytic activation molecule F7 (SLAMF7, also called CS1 (cell-surface glycoprotein CD2 subset 1). SLAMF7 is a glycoprotein expressed on myeloma and natural killer cells7,8. SLAMF7 belongs to the SLAM family on chromosome 1q23 and is highly expressed in MM cells independently of cytogenetic abnormalities. The binding of elotuzumab to SLAMF7 activates NK cells by coupling with its adapter protein EAT-2 whereas it leads to antibody-dependent cell cytotoxicity (ADCC) in MM cells as EAT-2 is not expressed in MM cells9,10. Elotuzumab has modest impact as single agent in RRMM as shown in a phase 1 study in 35 relapse and refractory MM where 26.5% of the patients had stable disease but no overall response was observed11. However, considering the impact of Imids on the immune system, activating NK cells and inhibiting T cells regulators, several studies have combined elotuzumab with IMids and showed significant synergistic activity in combination. In two phase 1 studies in RRMM combining elotuzumab either with bortezomib/dexamethasone or with lenalidomide/dexamethasone, the ORR were 48 and 82% respectively12,13. In the phase 3 clinical trial ELOQUENT 214, elotuzumab was evaluated in addition to lenalidomide (Len) and dexamethasone (Dex) in relapse or refractory MM (RRMM). The overall response rate in the elotuzumab group was 79%, versus 66% in the control group (P<0.001) and the Median progression-free survival in the elotuzumab group was 19.4 months, versus 14.9 months in the control group (P<0.001). In another multicenter, randomized, open-label, phase 2 trial (ELOQUENT 3)15, MM patients refractory to lenalidomide and to a proteasome inhibitor, were treated with elotuzumab plus pomalidomide and dexamethasone or pomalidomide dexamethasone alone. The overall response rate was 53% in the elotuzumab group as compared with 26% in the control group (odds ratio, 3.25; 95% CI, 1.49 to 7.11). The median progression-free survival was 10.3 months in the elotuzumab group and 4.7 months in the control group. The risk of progression or death was significantly lower among those who received elotuzumab. Importantly the combination of elotuzumab with lenalidomide or pomalidomide and dexamethasone was not associated with an increased toxicity.
Conversely, combining elotuzumab with bortezomib did not show significant benefit in a phase 2 clinical trial although overall survival and PFS trended to be increased16. Elotuzumab was approved in combination with lenalidomide in RRMM in the US in 11/2015 and in Europe in 05/2016 and in combination with pomalidomide in 11/2018. An ongoing phase 3 clinical trial (ELOQUENT 1, NCT01335399) is evaluating elotuzumab combined with lenalidomde/dexamethasone versus lenalidomide/dexamethasone alone in newly diagnosed elderly or unfit patient and a similar combination is also evaluated in high risk smoldering MM patients ( NCT02279394).
B-. Monoclonal antibodies targeting CD38
CD38 is a 45-kD, transmembrane glycoprotein that links with cell-surface receptors in lipid rafts, regulates cytoplasmic Ca2+ flux, and mediates signal transduction in lymphoid and myeloid cells17. CD38 is highly expressed on myeloma cells while its expression is low on normal lymphoid and myeloid cells and in some tissues of nonhematopoietic origin18.
Daratumumab is a human IgG1κ monoclonal antibody that binds to a unique CD38 epitope19. Preclinical studies showed that activity of daratumumab is mediated by multiple mechanisms including complement-mediated and antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, apoptosis, and inhibition of the enzymatic activity of CD3820. In two phase 1/2 clinical trials, Daratumumab was tested as a single agent in RRMM21. In one study, The overall response rate was 36% in the cohort that received 16 mg per kilogram and 65% (95% CI, 28 to 86) of the patients who had a response did not have progression at 12 months21. In the second study, the overall responses were noted in 29% (95% CI 20.8–38.9), median duration of response was 7.4 months (95% CI 5.5-not estimable) and progression-free survival was 3.7 months (95% CI 2.8–4.6). The 12-month overall survival was 64.8% (95% CI 51.2–75.5) (SIRIUS trial)22.
Subsequently, daratumumab was used in combination with either bortezomib/Dexamethasone23 or Lenalidomide/Dexamethasone24 in two phase 3 clinical trials23,24. In the phase 3 clinical trial23 comparing velcade/dexamethasone to velcade/dexamethasone/daratumumab in 498 R/R MM patients (Castor trial) the 12-month rate of progression-free survival was 60.7% in the daratumumab group versus 26.9% in the control group. The rate of overall response was higher in the daratumumab25 group than in the control group as well (82.9% vs. 63.2%, P<0.001), as were the rates of complete response or better (19.2% vs. 9.0%, P=0.001). After a median follow-up period of 7.4 months, the median progression-free survival was not reached in the daratumumab group and was 7.2 months in the control group (hazard ratio for progression or death with daratumumab vs. control, 0.39; 95% confidence interval, 0.28 to 0.53; P<0.001). In the phase 3 clinical trial comparing Len/Dex to Dara/Len/Dex (Pollux trial) in 569 R/R MM patients, the progression-free survival at 12 months was 83.2% (95% CI, 78.3 to 87.2) in the daratumumab group, as compared with 60.1% (95% CI, 54.0 to 65.7) in the control group. A significantly higher rate of overall response was observed in the daratumumab group than in the control group (92.9% vs. 76.4%, P<0.001), as was a higher rate of complete response or better (43.1% vs. 19.2%, P<0.001). In the daratumumab group, 22.4% of the patients had results below the threshold (1 tumor cell per 105 white cells) for minimal residual disease (MRD), as compared with 4.6% of those in the control group (P<0.001); results below the threshold for MRD were associated with improved outcomes.
As upfront therapy, Daratumumab has been evaluated in patients ineligible for autotransplant in a phase 3 clinical trial comparing bortezomib, melphalan, and prednisone (MPV) either alone (control group) or with daratumumab (ALCYONE clinical trial)26. At a median follow-up of 16.5 months, the 18-month progression-free survival rate was 71.6% in the daratumumab group and 50.2% in the control group (hazard ratio for disease progression or death, 0.50; 95% CI, 0.38 to 0.65; P<0.001). The overall response rate was 90.9% in the daratumumab group, as compared with 73.9% in the control group (P<0.001), and the rate of complete response or better (including stringent complete response) was 42.6%, versus 24.4% (P<0.001). In the daratumumab group, 22.3% of the patients were negative for minimal residual disease (at a threshold of 1 tumor cell per 105 cells), as compared with 6.2% of those in the control group (P<0.001). Daratumumab is now approved by the FDA since 2016.
Isatuximab (SAR650984) is another chimeric IgG1 mAb made by variable domain resurfacing, targeting a different amino acid sequence epitope of CD38 than daratumumab. In addition, isatuximab has a direct toxic effect on MM cells as well27,28. Initial data suggest that isatuximab has clinical activity as a single agent, with an ORR of 32% obtained in patients with RRMM29. Isatuximab, in combination with lenalidomide/dexamethasone has been evaluated in a phase 1b clinical trial in 57 R/R MM patients. The overall response rate30 was 56% (29/52) and 52% in evaluable lenalidomide-refractory patients31. Isauximab is currently evaluated in RRMM as a single agent or with dexamethaosne ( NCT01084252) and in combination with RVD in newly diagnosed patients ( NCT02513186). Efficacy of Isatuximab inpatients relapsing after daratumumab will be interesting as both target slightly different epitopes on CD38 molecule.
A third anti-CD38 monoclonal antibody (MOR202)32 is also in early clinical trials and bi-specific antibodies targeting CD38 and CD3 (AMG 424)33, and a monoclonal antibody conjugated with α-emitter astatine-211 (211At) an α-particle emitting isotopes34,35, are also being evaluated in early phase clinical trial and pre-clinical models.
C-. Monoclonal antibodies targeting BCMA
B-cell maturation antigen (BCMA), a member of the tumor necrosis factor receptor superfamily (TNFRSF17), is selectively induced during plasma cell differentiation and nearly absent on naive and memory B cells and CD34 positive hematopoietic stem cells. BCMA is highly expressed in MM cells. B-cell activating factor (BAFF) and A proliferation-inducing ligand (APRIL)36 are 2 known ligands of BCMA that induce survival of long lived plasma cells. Significantly, soluble BCMA (sBCMA) levels are increased in MM patients in comparison to healthy individuals and correlate with disease status and prognosis. These data have highlighted BCMA as a very promising target in MM. Several monoclonal antibodies and cellular therapies targeting BCMA are under investigation including monoclonal antibodies, antibody-drug conjugate and bi-specific antibodies37.
GSK2857916 is humanized and afucosylated antagonistic anti-BCMA antibody-drug conjugate via a noncleavable linker which specifically blocks cell growth via G2/M arrest and induces caspase 3-dependent apoptosis38. In an international, multicentre, open-label, first-in-human phase 1/2 study, 73 patients: 38 patients with RRMM in the dose-escalation part 1 and 35 patients in the dose-expansion part 2 were treated. There were no dose-limiting toxicities and no maximum tolerated dose was identified in part 1. In part 2, 21 (60%; 95% CI 42·1–76·1) of 35 patients achieved an overall response suggesting a promising role of this conjugated monoclonal antibody. Of note, corneal events were common (53% of 38 patients in part 1 and 63% of 35 in part 2) but moderate in majority. The most common grade 3 or 4 events were thrombocytopenia (13 [34%] of 38 patients in part 1 and 12 [34%] of 35 in part 2) and anemia (6 [16%] in part 1 and 5 [14%] in part 2). There were 12 treatment-related serious adverse events and no treatment-related deaths. Other antibody-conjugated targeting BCMA are under investigation such as HDP-1, an antibody- amanitin conjugate or MeDi2228 a fully human antibody - specifically conjugated to a pyrrolobenzodiazepine dimer via a protease-cleavable linker39,40.
Bispecific monoclonal antibodies targeting BCMA are also being actively developed. BI 836909, a bispecific single-chain variable fragment (scFv) that simultaneously bind to CD3 and BCMA was the first reported. Pre-clinical evaluation was promising in mouse and monkeys and further investigations are on going although the short half life of the molecule might require frequent infusions41. Several other bi-specific antibodies are under development and include TNB383B, TNB-384B, Ab-957, EM801 and BCMA-TCB2, that are also IgG-based human bi-specific antibodies with two binding sites for BCMA and CD3 with significant toxicity on MM cells in pre-clinical models25,42,43. PF-3135 is a humanized immunoglobulin G (IgG2a) CD3 and BCMA bispecific monoclonal antibody that is now evaluated in an ongoing phase 1 clinical trial ( NCT03269136)44. AFM26 is a bispecific antibody, which targets BCMA and CD16A on NK cells with significant efficacy in pre-clinical models. Its impact on NK cells may suggest a potential for a synergistic activity with Imids45. Finally, tri-specific antibody-like molecules targeting BCMA are also under evaluation. An anti-CD16A/BCMA/CD200 antibody binding to CD16A on NK cells and to BCMA and CD200 on MM cells with potentially significant efficiency and increase selectivity of MM cells is under evaluation46.
D. Monoclonal antibodies targeting April
A Proliferation-inducing ligand36 is one of the 2 known ligands of BCMA and its binding to BCMA enhances plasma cell proliferation and survival. BION-1301 is a humanized anti-APRIL antibody blocking the binding of APRIL to BCMA and TACI that has significant impact in vitro and in co-culture models and currently evaluated in an early phase clinical trial in RRMM ( NCT03340883).
E. Monoclonal antibodies targeting CD138
Indatuximab ravtansine (BT062) is a monoclonal antibody-drug conjugate under development targeting CD138 (syndecan1) which is universally highly expressed on MM cells. The monoclonal antibody is coupled to the maytansinoid DM4 toxin. In a phase 1/2 clinical trial evaluating BT062 in combination with lenalidomide/dexamethasone or pomalidomide/dexamthasone in RRMM, promising preliminary results were observed with an ORR of 54% and 79% respectively in each arm47,48.
F. Monoclonal antibodies targeting immune checkpoints
The advent of immune checkpoint inhibitors is one of the most important progress in the last few years in oncology. The development of monoclonal antibodies targeting the immune checkpoints has considerably changed the treatment and prognosis of several cancer including melanoma, lung cancer and relapsed and refractory Hodgkin disease among others. In multiple myeloma, Programmed death 1 (PD-1) receptor is highly expressed suggesting that treatment targeting it or its ligand (PD-L1 or PD-L2) would be an effective strategy49. Several monoclonal antibodies targeting PD1 (pembrolizumab, Nivolumab) or PDL1 (durvolumab, atezolizumab) are approved for various other malignancies50. Based on promising phase 2 data with the combination of pembrolizumab with Pom/Dex and Len/Dex in RRMM, three phase 3 clinical trials- KEYNOTE-183 and KEYNOTE- 185 and checkmate 602- respectively evaluated pomalidomide/dexamathasone with or without pembrolizumab in RRMM, lenalidomide/Dexamethasone with and without pembrolizumab in newly diagnosed MM patients non eligible for auto-transplant and nivolumab plus pomalidomide–dexamethasone versus pomalidomide–dexamethasone alone or pomalidomamide/dexamethasone/elotuzumab/nivolumab in patients with RRMM. All studies were stopped prematurely because of an increased mortality in patients receiving pembrolizumab with a hazard ratio for death of 1.61 in KEYNOTE-183 and 2.06 in KEYNOTE-185, or nivolumab in checkmate 602 (hazard ratio for death was 1.19 (95% confidence interval, 0.64 to 2.20)). Furthermore the addition of pembrolizumab or nivolumab did not increase the ORR. Importantly, no specific cause of death was observed in all 3 trials and the pathogenesis behind the toxicity of the combination remains largely unknown as well as the absence of obvious efficacy of the monoclonal antibodies. These results have raised serious doubts regarding their utility in MM at least in combination with the immunomodulatory agents51. However, other immune checkpoints such as LAG3, TIM3 or TIGIT are under pre-clinical investigation52–55.
Various other cell surface molecules have been targeted using antibodies which have undergone promising preclinical evaluation (Table 2).
2. DNA damaging agents
Alkylating agents, remain a corner stone of MM treatment with high dose melphalan as the conditioning regimen of choice for auto-transplant. New alkylating agents have been developed in order to improve efficacy and decrease toxicity of DNA-damaging agents.
a. Melflufen
Melphalan flufenamide ethyl ester (melflufen) is a peptidase-potentiated alkylating agent which appears to be more efficient than melphalan56. Especially, in vitro evaluation showed that melflufen is active in melphalan resistant cell line by generating rapid and irreversible DNA damage. Three clinical trials are ongoing to confirm its efficacy in MM. The phase 2 clinical trial 012M1 evaluating Melflufen in RRMM with at least 2 prior lines of therapy including bortezomib and lenalidomide, showed promising results with an ORR of 31% in advanced RRMM patients57. The phase 2 clinical trial HORIZON evaluating melflufen in RRMM with at least 2 prior lines of treatment including Imids, PI, pomalidomide and daratumumab showed an ORR of 27%. A clinical phase 3 trial comparing melflufen versus pomalidomide in RRMM and a phase 1–2 trial combining melflufen with bortezomib and daratumumab in RRMM are ongoing and will likely confirm the role of this new alkylating agent in MM58,59.
b. Bendamustine
Bendamustine is an alkylating agent with a purine analog ring that has been developed several decades ago although its cautious evaluation in MM is more recent60. A phase III clinical trial compared bendamustine + prednisone with melphalan + prednisone in newly diagnosed patients ineligible for autotranplant showed a benefit especially in terms of TTP (14 vs. 10 months)61. Several phase 2 clinical studies combining Bendamustine with PI or Imids have been reported with relatively good results making bendamustine as an available additional therapeutic option.62–65
c. EDO-S101
Preclinical data suggested a synergism between alkylating agents and HDACi although their combination in patients was associated with important toxicity66–68. EDO-S101 is a first in class fusion molecule derived from bendamustine that has been linked to a class 1 and 2 HDACi. Now under pre-clinical evaluation69.
3. Inhibitors of BCL2 family proteins
Important efforts in research have been made to understand the mechanisms driving cancer cells survival. In the last two decades significant discoveries have shed light on the mechanisms regulating apoptosis in cancer cells. The BCL2 family proteins have been identified as critical regulators of apoptosis in cancer and healthy tissues. This family includes Bcl-2, Bcl-XL or Mcl-1, that are multi-domain anti-apoptotic proteins. Several groups have identified a tissue specific anti-apoptotic dependency and small molecules have been developed to target the BCL2 family proteins70. Interestingly, bcl2-dependency can be assessed functionally with a method using BH3 profiling, a flow-cytometry based method or simply by evaluating the levels of expression of BCL2, bcl-xl and MCL 171,72.
A. Venetoclax
Venetoclax (ABT-199) has been approved for several hematologic malignancies in the past few years including chronic lymphoid leukemia and mantle cell lymphoma. In MM, Venetoclax has been shown to be active in RRMM as a single agent in a phase 1 clinical trial73
Importantly, the presence of t(11 ;14) in MM confers a higher BCL2 dependency and prolonged, deep responses have been observed in the subgroup of t(11 ;14) MM which comprise ~20% of MM patients. In this trial, The ORR in patients with t(11;14) translocation was 40%, including 14% CR or better30. In the non t(11;14) group, two patients (6%) achieved at least a partial response. The phase 2 M13–367 study ( NCT01794520) is currently evaluating the efficacy of venetoclax (800 mg daily) in combination with dexamethasone in relapsed myeloma patients with t(11;14) translocation. Preliminary results revealed an ORR of 65%, including 35% VGPR in advanced relapsed MM patients (median of three prior therapies).
Venetoclax in combination with other agents has also shown efficacy in non-t(11 ;14) MM. Especially, bortezomib and other PIs have been shown to indirectly inhibit MCL-1 and potentially synergistic activity with venetoclax. Thus, in a phase 2 clinical trial evaluating Venetoclax/bortezomib/Dexamethasone in 66 RRMM patients, the overall response rate30 was 67% (44/66). ORR of 97% and ≥VGPR 73% were seen in patients not refractory to bortezomib who had 1 to 3 prior therapies. Patients with high BCL2 expression had a higher ORR (94%)74.
B. MCL1 inhibitors
With success of targeting BCL2, other anti-apoptotic molecules involved in MM cell survival are being evaluated as potential targets. MCL1 inhibitors are the second most advanced drugs in development. Currently, a phase 2 clinical trial is ongoing to evaluate safety and efficacy of MCL1-inhibitor in MM using MIK665 ( NCT02992483) and AMG 176 ( NCT02675452).
4. Epigenetic inhibitors
-Histone Deacetylase (HDAC)
Epigenetic mechanisms play a critical role in deregulating MM cells transcriptome and enhancing tumor progression75–77. HDACs include four classes of proteins according to their structure and function: class I (HDAC 1, 2, 3, and 8), class IIa (HDACs 4, 5, 7, and 9), class IIb (HDACs 6 and 10), class III (sirtuins), and class IV (HDAC11). Pan-HDACs inhibitors have been the first molecules tested and developed in MM. While a modest impact have been shown in monotherapy, the role of HDAC in disrupting aggresomal protein degradation led to evaluation of the efficiency of HDAC and PIs. Thus, panobinistat (a pan HDAC inhibitor) combined with bortezomib has been approved by the FDA as a third line of treatment in patients previously treated with Imids and PIs following a phase 3 clinical trial confirming efficacy in the setting of RRMM with an improvement of 4 months in EFS78. A challenge resulting from pan HDAC inhibition is the lack of specificity and the side effects observed that often lead to treatment discontinuation. For this reason, selective HDACs inhibitors are now under investigation. Particularly, HDAC6 inhibitor (rocilinostat) is now investigated in clinical trials and a phase 1b study showed that RRMM patients treated with rocilinostat plus bortezomib/dexamethasone had a 37% ORR in RRMM79. Another phase 1b trial showed a promising ORR of 55% when ricolinostat was combined with lenalidome/dexamethasone in RRMM with limited toxicity80.
-EZH2 inhibitors
Various epigenetic modifiers are altered in MM including overexpression of enhancer of zeste homologue 2 (EZH2) or MMSET overexpression and UTX/KDM6A mutations81,82. EZH2 is a histone -methyltransferase, component of polycomb repressive complex 2 (PRC2), which triggers H3K27me3 to repress gene transcriptome; MMSET is a histone methyl transferase which regulates gene expression through H3K36 methylation; and UTX regulates H3K27 acetylation. MMSET is overexpressed in t(4 ;14) which represents ~15% of MM and is associated with high risk disease. UTX/KDM6A mutation are observed in up to 5% of MM patients. UTX loss and MMSET overexpression are associated with increased transcriptomic control by EZH283. Recent advances include development of small molecule inhibitors of EZH2 which have shown significant efficacy against MM cells in vitro84–86. Tazemetostat, an oral EZH1/2 inhibitor is under pre-clinical evaluation in hematological malignancies. Alternatively, important efforts have been made to develop MMSET inhibitors to generate targeted therapy in this subgroup of patients but the improvements in bioavailability and specificity of the drugs are still in preclinical development.
6. MAPK pathway inhibitors
-Trametinib
Unlike other malignancies characterized by recurrent genomic driver such as MYD88 in Waldenström disease or BRAF V600E mutation in melanoma or hairy cell leukemia, MM is featured by a highly heterogeneous genetic background that includes various subtypes without clearly defined drivers. Only few recurrent mutations have been identified (NRAS, KRAS, TP53, DIS3, and FAM46C) with NFKB and MAPK pathways being the most recurrently affected pathways, in 43 % and 17 % of MM patients respectively87. For this reason, the MAPK pathway is an attractive target in MM. Vemurafenib as single agent or in combination with bortezomib has been reported as active in V600EBRAF mutated MM patients88,89 but this mutation is only present in approximately 2 to 5% of MM patients and is not systematically screened87. MEK inhibition using trametinib or in combination with dabrafenib could represent interesting therapeutic option. A first retrospective study reported potential benefit of trametinib as a single agent or in combination with other MM approved treatment90. However, patients were heavily treated and presented with very advanced disease and toxicity was a concern (24/58 patients discontinued therapy because of toxicities)90. However, this study did not select patients based on presence of perturbation in MAPK pathway, and thus its real role in possibly susceptible patient population will be determined by ongoing studies utilizing Trametinib and Dabrafenib in targeted patient population. Alone or in combination, these agents may become an important and efficient therapeutic strategy in MM.
-Afuresertib
Afuresertib is an oral AKT inhibitor. That has been evaluated in two phase 1 clinical trials in RRMM as a single agent and in combination with bortezomib and dexamethasone. As a single agent, Afuresertib showed limited activity but when combined with bortezomib and dexamethasone the ORR was up to 61%91,92. In another phase 1 study, Afuresertib has been combined to trametinib and evaluated in MM and solids tumors but the toxicity was high and the study discontinued93.
7. Inhibitors of nuclear cytoplasmic transport receptor: XPO1 inhibitor
The ubiquitous transport receptor chromosome maintenance protein 1 (CRM1, also known as XPO1) is a nuclear–cytoplasmic transport receptor that acts as a carrier molecule enhancing transportation into (importins) and out of (exportins) the nucleus. XPO1 is a member of the karyopherin family, which includes 19 members. XPO1 mediates the nuclear export of RNAs, and a large number of proteins carrying a canonical hydrophobic leucine-rich amino-acid sequence, some transcription factors involved in NFκB signaling and HDACs94. XPO1 expression is high in MM cells and correlate with survival in MM patients suggesting a critical role in MM biology.
Selinexor (KPT-330) is a first-in-class, orally bioavailable, selective inhibitor of XPO1-mediated nuclear export. A phase I study, evaluated selinexor alone or in combination with low-dose dexamethasone and showed broad activity and promising response in RRMM. Two phase 2 clinical trials recently published evaluated selinexor and dexamethasone, and selinexor in combination with bortezomib and dexamethasone in heavily pretreated RRMM patients. In the first study evaluating selinexor/dexamthasone, the ORR was 21% in all patients and 35% in high risk patients while the ORR was 63% for PI non refractory and 43% for PI-refractory patients in the study evaluating the combination of bortezomib/selinexor/dexamethasone. Both studies showed good response and selinexor is a promising target in MM with a limited increased toxicity95,96.
8. Imids
Approved immunomodulatory drugs (IMiDs) in MM include thalidomide, lenalidomide, and pomalidomide. These molecules share an antimyeloma activity that relates to its interaction with the intracellular complex Cereblon/E3 ubiquitin ligase/Cul4A/DDb1. This binding enhances the degradation of two key transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) through the proteasome and lead to intrupt MM cell proliferation and growth. In addition, IMids enhance cells activation of various immune cells including natural killer (NK), CD4 and CD8 T cells while inhibiting regulatory T cells97,98. Therefore, IMids have synergistic activity with other immunotherapies such has monoclonal antibodies or checkpoint inhibitors. Two new Imids CC220 (or Iberdomide) and CC-92480, harboring greater affinity for the Cereblon/Cul4a/E3ligase complex and degradation of its substrate, are now under investigation in various hematologic malignancies including MM99.
In addition, the understanding of the mechanism of action of the IMids has allowed for development of a new class of drug that are currently at in pre-clinical stage. These class of drugs called degraders use the properties of IMids to bind to the Cereblon/E3Ligase/Cul4a complex to degrade specific targets100.
9. kinesin spindle protein
Filanesib (ARRY-520) is a kinesin spindle protein inhibitor, with clinical activity as monotherapy in heavily pretreated MM patients. Kinesin spindle protein (EG5/KIF11) is a critical molecule in mitosis during cell division by contributing to the spindle apparatus and centrosome formation.101
In a phase 1 study, Arry-520 was tested in heavily pre-treated RRMM and was associated with an ORR of 16% with a tolerable safety102. Several ongoing phase 2 clinical trials are evaluating ARRY-520 in combination with pomalidomide/dexamethasone ( NCT02384083), bortezomib/dexamethasone ( NCT01248923) or carfilzomib/Dexamethasone ( NCT01372540) in RRMM and plasma cell leukemia.
The identification of new therapeutic targets in MM along with the rise of immunotherapy and small molecules inhibitors provide significant change in MM therapeutic landscape. Several new molecules have recently been approved and several other therapies referenced in this review will likely be soon validated too. Altogether, with the advent of cellular therapies discussed in a separate article in this issue will deeply modify the course of MM and eventually allow for a curative outcome in this disease. The detection of minimal residual disease by either next generation flow cytometry or next generation sequencing will likely be used as a new surrogate marker to monitor the disease and guide therapy while incorporating all these new efficient agents.
References
- 1.Schiffer CA & Zonder JA Transplantation for Myeloma - Now or Later? The New England journal of medicine 376, 1378–1379, doi: 10.1056/NEJMe1700453 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Kumar SK et al. Natural history of relapsed myeloma, refractory to immunomodulatory drugs and proteasome inhibitors: a multicenter IMWG study. Leukemia 31, 2443–2448, doi: 10.1038/leu.2017.138 (2017). [DOI] [PubMed] [Google Scholar]
- 3.Kumar S Emerging options in multiple myeloma: targeted, immune, and epigenetic therapies. Hematology. American Society of Hematology. Education Program 2017, 518–524, doi: 10.1182/asheducation-2017.1.518 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Brentuximab Vedotin with Chemotherapy for Stage III or IV Hodgkin’s Lymphoma. The New England journal of medicine 378, 878, doi: 10.1056/NEJMx180007 (2018). [DOI] [PubMed] [Google Scholar]
- 5.Younes A et al. Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas. The New England journal of medicine 363, 1812–1821, doi: 10.1056/NEJMoa1002965 (2010). [DOI] [PubMed] [Google Scholar]
- 6.Cohen AD CAR T Cells and Other Cellular Therapies for Multiple Myeloma: 2018 Update. American Society of Clinical Oncology educational book. American Society of Clinical Oncology. Annual Meeting, e6–e15, doi: 10.1200/EDBK_200889 (2018). [DOI] [PubMed] [Google Scholar]
- 7.Hsi ED et al. CS1, a potential new therapeutic antibody target for the treatment of multiple myeloma. Clinical cancer research : an official journal of the American Association for Cancer Research 14, 2775–2784, doi: 10.1158/1078-0432.CCR-07-4246 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Cannons JL, Tangye SG & Schwartzberg PL SLAM family receptors and SAP adaptors in immunity. Annual review of immunology 29, 665–705, doi: 10.1146/annurev-immunol-030409-101302 (2011). [DOI] [PubMed] [Google Scholar]
- 9.Tai YT et al. Anti-CS1 humanized monoclonal antibody HuLuc63 inhibits myeloma cell adhesion and induces antibody-dependent cellular cytotoxicity in the bone marrow milieu. Blood 112, 1329–1337, doi: 10.1182/blood-2007-08-107292 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Collins SM et al. Elotuzumab directly enhances NK cell cytotoxicity against myeloma via CS1 ligation: evidence for augmented NK cell function complementing ADCC. Cancer immunology, immunotherapy : CII 62, 1841–1849, doi: 10.1007/s00262-013-1493-8 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Zonder JA et al. A phase 1, multicenter, open-label, dose escalation study of elotuzumab in patients with advanced multiple myeloma. Blood 120, 552–559, doi: 10.1182/blood-2011-06-360552 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Jakubowiak AJ et al. Phase I trial of anti-CS1 monoclonal antibody elotuzumab in combination with bortezomib in the treatment of relapsed/refractory multiple myeloma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 30, 1960–1965, doi: 10.1200/JCO.2011.37.7069 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Lonial S et al. Elotuzumab in combination with lenalidomide and low-dose dexamethasone in relapsed or refractory multiple myeloma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 30, 1953–1959, doi: 10.1200/JCO.2011.37.2649 (2012). [DOI] [PubMed] [Google Scholar]
- 14.Lonial S et al. Elotuzumab Therapy for Relapsed or Refractory Multiple Myeloma. The New England journal of medicine 373, 621–631, doi: 10.1056/NEJMoa1505654 (2015). [DOI] [PubMed] [Google Scholar]
- 15.Dimopoulos MA et al. Elotuzumab plus Pomalidomide and Dexamethasone for Multiple Myeloma. The New England journal of medicine 379, 1811–1822, doi: 10.1056/NEJMoa1805762 (2018). [DOI] [PubMed] [Google Scholar]
- 16.Jakubowiak A et al. Randomized phase 2 study: elotuzumab plus bortezomib/dexamethasone vs bortezomib/dexamethasone for relapsed/refractory MM. Blood 127, 2833–2840, doi: 10.1182/blood-2016-01-694604 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Konopleva M, Estrov Z, Zhao S, Andreeff M & Mehta K Ligation of cell surface CD38 protein with agonistic monoclonal antibody induces a cell growth signal in myeloid leukemia cells. Journal of immunology 161, 4702–4708 (1998). [PubMed] [Google Scholar]
- 18.Lin P, Owens R, Tricot G & Wilson CS Flow cytometric immunophenotypic analysis of 306 cases of multiple myeloma. American journal of clinical pathology 121, 482–488, doi: 10.1309/74R4-TB90-BUWH-27JX (2004). [DOI] [PubMed] [Google Scholar]
- 19.de Weers M et al. Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. Journal of immunology 186, 1840–1848, doi: 10.4049/jimmunol.1003032 (2011). [DOI] [PubMed] [Google Scholar]
- 20.Overdijk MB et al. Antibody-mediated phagocytosis contributes to the anti-tumor activity of the therapeutic antibody daratumumab in lymphoma and multiple myeloma. mAbs 7, 311–321, doi: 10.1080/19420862.2015.1007813 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lokhorst HM et al. Targeting CD38 with Daratumumab Monotherapy in Multiple Myeloma. The New England journal of medicine 373, 1207–1219, doi: 10.1056/NEJMoa1506348 (2015). [DOI] [PubMed] [Google Scholar]
- 22.Lonial S et al. Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): an open-label, randomised, phase 2 trial. Lancet 387, 1551–1560, doi: 10.1016/S0140-6736(15)01120-4 (2016). [DOI] [PubMed] [Google Scholar]
- 23.Palumbo A et al. Daratumumab, Bortezomib, and Dexamethasone for Multiple Myeloma. The New England journal of medicine 375, 754–766, doi: 10.1056/NEJMoa1606038 (2016). [DOI] [PubMed] [Google Scholar]
- 24.Dimopoulos MA et al. Daratumumab, Lenalidomide, and Dexamethasone for Multiple Myeloma. The New England journal of medicine 375, 1319–1331, doi: 10.1056/NEJMoa1607751 (2016). [DOI] [PubMed] [Google Scholar]
- 25.Pillarisetti K et al. Development of a New BCMAxCD3 Duobody® Antibody for Multiple Myeloma. Blood 128, 2116–2116 (2016). [Google Scholar]
- 26.Mateos MV et al. Daratumumab plus Bortezomib, Melphalan, and Prednisone for Untreated Myeloma. The New England journal of medicine 378, 518–528, doi: 10.1056/NEJMoa1714678 (2018). [DOI] [PubMed] [Google Scholar]
- 27.Deckert J et al. SAR650984, a novel humanized CD38-targeting antibody, demonstrates potent antitumor activity in models of multiple myeloma and other CD38+ hematologic malignancies. Clinical cancer research : an official journal of the American Association for Cancer Research 20, 4574–4583, doi: 10.1158/1078-0432.CCR-14-0695 (2014). [DOI] [PubMed] [Google Scholar]
- 28.Jiang H et al. SAR650984 directly induces multiple myeloma cell death via lysosomal-associated and apoptotic pathways, which is further enhanced by pomalidomide. Leukemia 30, 399–408, doi: 10.1038/leu.2015.240 (2016). [DOI] [PubMed] [Google Scholar]
- 29.Martin T et al. A Dose Finding Phase II Trial of Isatuximab (SAR650984, Anti-CD38 mAb) As a Single Agent in Relapsed/Refractory Multiple Myeloma. Blood 126, 509–509 (2015). [Google Scholar]
- 30.Cook G et al. The effect of salvage autologous stem-cell transplantation on overall survival in patients with relapsed multiple myeloma (final results from BSBMT/UKMF Myeloma X Relapse [Intensive]): a randomised, open-label, phase 3 trial. The Lancet. Haematology 3, e340–351, doi: 10.1016/S2352-3026(16)30049-7 (2016). [DOI] [PubMed] [Google Scholar]
- 31.Martin T et al. A phase 1b study of isatuximab plus lenalidomide and dexamethasone for relapsed/refractory multiple myeloma. Blood 129, 3294–3303, doi: 10.1182/blood-2016-09-740787 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Boxhammer R, Weirather J, Steidl S & Endell J MOR202, a Human Anti-CD38 Monoclonal Antibody, Mediates Potent Tumoricidal Activity In Vivo and Shows Synergistic Efficacy in Combination with Different Antineoplastic Compounds. Blood 126, 3015–3015 (2015). [Google Scholar]
- 33.de Zafra C et al. Preclinical Characterization of AMG 424, a Novel Humanized T Cell-Recruiting Bispecific Anti-CD3/CD38 Antibody. Blood 130, 500–500 (2017). [Google Scholar]
- 34.Teiluf K et al. alpha-Radioimmunotherapy with (2)(1)(3)Bi-anti-CD38 immunoconjugates is effective in a mouse model of human multiple myeloma. Oncotarget 6, 4692–4703, doi: 10.18632/oncotarget.2986 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.O’Steen S et al. The Alpha Emitter Astatine-211 Targeted to CD38 Can Eradicate Multiple Myeloma in Minimal Residual Disease Models. Blood 132, 1941–1941, doi: 10.1182/blood-2018-99-119555 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kommoss S et al. Bevacizumab May Differentially Improve Ovarian Cancer Outcome in Patients with Proliferative and Mesenchymal Molecular Subtypes. Clinical cancer research : an official journal of the American Association for Cancer Research 23, 3794–3801, doi: 10.1158/1078-0432.CCR-16-2196 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Cho SF, Anderson KC & Tai YT Targeting B Cell Maturation Antigen (BCMA) in Multiple Myeloma: Potential Uses of BCMA-Based Immunotherapy. Frontiers in immunology 9, 1821, doi: 10.3389/fimmu.2018.01821 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Trudel S et al. Targeting B-cell maturation antigen with GSK2857916 antibody-drug conjugate in relapsed or refractory multiple myeloma (BMA117159): a dose escalation and expansion phase 1 trial. The Lancet. Oncology 19, 1641–1653, doi: 10.1016/S1470-2045(18)30576-X (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Singh RK et al. HDP101, a Novel B-Cell Maturation Antigen (BCMA)-Targeted Antibody Conjugated to α-Amanitin, Is Active Against Myeloma with Preferential Efficacy Against Pre-Clinical Models of Deletion 17p. Blood 132, 593–593, doi: 10.1182/blood-2018-99-118412 (2018). [DOI] [Google Scholar]
- 40.Kinneer K et al. Preclinical Evaluation of MEDI2228, a BCMA-Targeting Pyrrolobenzodiazepine-Linked Antibody Drug Conjugate for the Treatment of Multiple Myeloma. Blood 130, 3153–3153 (2017). [Google Scholar]
- 41.Hipp S et al. A novel BCMA/CD3 bispecific T-cell engager for the treatment of multiple myeloma induces selective lysis in vitro and in vivo. Leukemia 31, 2278, doi: 10.1038/leu.2017.219 (2017). [DOI] [PubMed] [Google Scholar]
- 42.Buelow B et al. T Cell Engagement without Cytokine Storm: A Novel Bcma x CD3 Antibody Killing Myeloma Cells with Minimal Cytokine Secretion. Blood 130, 501–501 (2017).28533307 [Google Scholar]
- 43.Seckinger A et al. Target Expression, Generation, Preclinical Activity, and Pharmacokinetics of the BCMA-T Cell Bispecific Antibody EM801 for Multiple Myeloma Treatment. Cancer cell 31, 396–410, doi: 10.1016/j.ccell.2017.02.002 (2017). [DOI] [PubMed] [Google Scholar]
- 44.Lesokhin AM et al. A Phase I, Open-Label Study to Evaluate the Safety, Pharmacokinetic, Pharmacodynamic, and Clinical Activity of PF-06863135, a B-Cell Maturation Antigen/CD3 Bispecific Antibody, in Patients with Relapsed/Refractory Advanced Multiple Myeloma. Blood 132, 3229–3229, doi: 10.1182/blood-2018-99-110427 (2018). [DOI] [Google Scholar]
- 45.Ross T et al. Preclinical Characterization of AFM26, a Novel B Cell Maturation Antigen (BCMA)-Directed Tetravalent Bispecific Antibody for High Affinity Retargeting of NK Cells Against Myeloma. Blood 132, 1927–1927, doi: 10.1182/blood-2018-99-118970 (2018). [DOI] [Google Scholar]
- 46.Gantke T et al. Trispecific Antibodies for Selective CD16A-Directed NK-Cell Engagement in Multiple Myeloma. Blood 128, 4513–4513 (2016). [Google Scholar]
- 47.Kelly KR 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. Blood 124, 4736–4736 (2014). [Google Scholar]
- 48.Kelly KR et al. Indatuximab Ravtansine (BT062) in Combination with Low-Dose Dexamethasone and Lenalidomide or Pomalidomide: Clinical Activity in Patients with Relapsed / Refractory Multiple Myeloma. Blood 128, 4486–4486 (2016). [Google Scholar]
- 49.Benson DM Jr. et al. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood 116, 2286–2294, doi: 10.1182/blood-2010-02-271874 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Lesokhin AM et al. Nivolumab in Patients With Relapsed or Refractory Hematologic Malignancy: Preliminary Results of a Phase Ib Study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 34, 2698–2704, doi: 10.1200/JCO.2015.65.9789 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Gormley NJ & Pazdur R Immunotherapy Combinations in Multiple Myeloma - Known Unknowns. The New England journal of medicine 379, 1791–1795, doi: 10.1056/NEJMp1803602 (2018). [DOI] [PubMed] [Google Scholar]
- 52.Yoon CJ et al. Genomic and Immune Profiles of Multiple Myeloma Revealed By Whole Genome and Transcriptome Sequencing. Blood 132, 4493–4493, doi: 10.1182/blood-2018-99-117546 (2018). [DOI] [Google Scholar]
- 53.Asimakopoulos F TIGIT checkpoint inhibition for myeloma. Blood 132, 1629–1630, doi: 10.1182/blood-2018-08-864231 (2018). [DOI] [PubMed] [Google Scholar]
- 54.Guillerey C et al. TIGIT immune checkpoint blockade restores CD8(+) T-cell immunity against multiple myeloma. Blood 132, 1689–1694, doi: 10.1182/blood-2018-01-825265 (2018). [DOI] [PubMed] [Google Scholar]
- 55.Neri P et al. Immunome Single Cell Profiling Reveals T Cell Exhaustion with Upregulation of Checkpoint Inhibitors LAG3 and Tigit on Marrow Infiltrating T Lymphocytes in Daratumumab and IMiDs Resistant Patients. Blood 132, 242–242, doi: 10.1182/blood-2018-99-117531 (2018). [DOI] [Google Scholar]
- 56.Wickstrom M et al. Melflufen - a peptidase-potentiated alkylating agent in clinical trials. Oncotarget 8, 66641–66655, doi: 10.18632/oncotarget.18420 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Richardson PG et al. First Report on Overall Survival (OS) and Improved Progression Free Survival (PFS) in a Completed Phase 2a Study of Melflufen in Advanced Relapsed Refractory Multiple Myeloma (RRMM). Blood 130, 3150–3150 (2017). [Google Scholar]
- 58.Pour L et al. The OP-104 Anchor Study: A Phase 1/2 Study of Safety and Efficacy of Melflufen and Dexamethasone in Combination with Either Bortezomib or Daratumumab in Patients with Rrmm; First Report on Phase 1 Data. Blood 132, 1967–1967, doi: 10.1182/blood-2018-99-112983 (2018). [DOI] [Google Scholar]
- 59.Richardson P et al. OP-106 Horizon — Melflufen Therapy for RRMM Patients Refractory to Daratumumab and/or Pomalidomide; Updated Results and First Report on PFS. Blood 132, 600–600, doi: 10.1182/blood-2018-99-113095 (2018). [DOI] [Google Scholar]
- 60.Anger G, Hesse P & Baufeld H [Treatment of multiple myeloma with a new cytostatic agent: gamma-l-methyl-5-bis-(beta-chlorethyl)-amino-benzimidazolyl-(2)-butyric acid hydrochloride]. Deutsche medizinische Wochenschrift 94, 2495–2500, doi: 10.1055/s-0028-1110470 (1969). [DOI] [PubMed] [Google Scholar]
- 61.Ponisch W et al. Treatment of bendamustine and prednisone in patients with newly diagnosed multiple myeloma results in superior complete response rate, prolonged time to treatment failure and improved quality of life compared to treatment with melphalan and prednisone--a randomized phase III study of the East German Study Group of Hematology and Oncology (OSHO). Journal of cancer research and clinical oncology 132, 205–212, doi: 10.1007/s00432-005-0074-4 (2006). [DOI] [PubMed] [Google Scholar]
- 62.Rodon P et al. Phase II study of bendamustine, bortezomib and dexamethasone as second-line treatment for elderly patients with multiple myeloma: the Intergroupe Francophone du Myelome 2009–01 trial. Haematologica 100, e56–59, doi: 10.3324/haematol.2014.110890 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Beck J et al. Lenalidomide in combination with bendamustine and prednisolone in relapsed/refractory multiple myeloma: results of a phase 2 clinical trial (OSHO-#077). Journal of cancer research and clinical oncology 143, 2545–2553, doi: 10.1007/s00432-017-2504-5 (2017). [DOI] [PubMed] [Google Scholar]
- 64.Ponisch W et al. Lenalidomide, bendamustine and prednisolone exhibits a favourable safety and efficacy profile in relapsed or refractory multiple myeloma: final results of a phase 1 clinical trial OSHO - #077. British journal of haematology 162, 202–209, doi: 10.1111/bjh.12361 (2013). [DOI] [PubMed] [Google Scholar]
- 65.Gomez-Bougie P et al. BH3-mimetic toolkit guides the respective use of BCL2 and MCL1 BH3-mimetics in myeloma treatment. Blood, doi: 10.1182/blood-2018-03-836718 (2018). [DOI] [PubMed] [Google Scholar]
- 66.Maiso P et al. The histone deacetylase inhibitor LBH589 is a potent antimyeloma agent that overcomes drug resistance. Cancer research 66, 5781–5789, doi: 10.1158/0008-5472.CAN-05-4186 (2006). [DOI] [PubMed] [Google Scholar]
- 67.Cai B et al. Combination of bendamustine and entinostat synergistically inhibits proliferation of multiple myeloma cells via induction of apoptosis and DNA damage response. Cancer letters 335, 343–350, doi: 10.1016/j.canlet.2013.02.046 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Berenson JR et al. A phase 1/2 study of oral panobinostat combined with melphalan for patients with relapsed or refractory multiple myeloma. Annals of hematology 93, 89–98, doi: 10.1007/s00277-013-1910-2 (2014). [DOI] [PubMed] [Google Scholar]
- 69.Besse L et al. The First in Class, Alkylator-Histone-Deacetylase-Inhibitor Fusion Molecule Edo-S101 in Combination with Proteasome Inhibitors Induces Highly Synergistic Pro-Apoptotic Signaling through UPR Activation and Suppression of c-Myc and BCL2 in Multiple Myeloma. Blood 128, 4466–4466 (2016). [Google Scholar]
- 70.Letai AG Diagnosing and exploiting cancer’s addiction to blocks in apoptosis. Nature reviews. Cancer 8, 121–132, doi: 10.1038/nrc2297 (2008). [DOI] [PubMed] [Google Scholar]
- 71.Valentin R, Grabow S & Davids MS The rise of apoptosis: targeting apoptosis in hematologic malignancies. Blood 132, 1248–1264, doi: 10.1182/blood-2018-02-791350 (2018). [DOI] [PubMed] [Google Scholar]
- 72.Touzeau C, Maciag P, Amiot M & Moreau P Targeting Bcl-2 for the treatment of multiple myeloma. Leukemia 32, 1899–1907, doi: 10.1038/s41375-018-0223-9 (2018). [DOI] [PubMed] [Google Scholar]
- 73.Kumar S et al. Efficacy of venetoclax as targeted therapy for relapsed/refractory t(11;14) multiple myeloma. Blood 130, 2401–2409, doi: 10.1182/blood-2017-06-788786 (2017). [DOI] [PubMed] [Google Scholar]
- 74.Moreau P et al. Promising efficacy and acceptable safety of venetoclax plus bortezomib and dexamethasone in relapsed/refractory MM. Blood 130, 2392–2400, doi: 10.1182/blood-2017-06-788323 (2017). [DOI] [PubMed] [Google Scholar]
- 75.Amodio N, D’Aquila P, Passarino G, Tassone P & Bellizzi D Epigenetic modifications in multiple myeloma: recent advances on the role of DNA and histone methylation. Expert opinion on therapeutic targets 21, 91–101, doi: 10.1080/14728222.2016.1266339 (2017). [DOI] [PubMed] [Google Scholar]
- 76.Dimopoulos K, Gimsing P & Gronbaek K The role of epigenetics in the biology of multiple myeloma. Blood cancer journal 4, e207, doi: 10.1038/bcj.2014.29 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Agirre X et al. Whole-epigenome analysis in multiple myeloma reveals DNA hypermethylation of B cell-specific enhancers. Genome research 25, 478–487, doi: 10.1101/gr.180240.114 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.San-Miguel JF 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 15, 1195–1206, doi: 10.1016/S1470-2045(14)70440-1 (2014). [DOI] [PubMed] [Google Scholar]
- 79.Vogl DT et al. Ricolinostat, the First Selective Histone Deacetylase 6 Inhibitor, in Combination with Bortezomib and Dexamethasone for Relapsed or Refractory Multiple Myeloma. Clinical cancer research : an official journal of the American Association for Cancer Research 23, 3307–3315, doi: 10.1158/1078-0432.CCR-16-2526 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Yee AJ et al. Ricolinostat plus lenalidomide, and dexamethasone in relapsed or refractory multiple myeloma: a multicentre phase 1b trial. The Lancet. Oncology 17, 1569–1578, doi: 10.1016/S1470-2045(16)30375-8 (2016). [DOI] [PubMed] [Google Scholar]
- 81.Dupere-Richer D & Licht JD Epigenetic regulatory mutations and epigenetic therapy for multiple myeloma. Curr. Opin. Hematol 24, 336–344, doi: 10.1097/MOH.0000000000000358 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Pawlyn C et al. The Spectrum and Clinical Impact of Epigenetic Modifier Mutations in Myeloma. Clin. Cancer Res 22, 5783–5794, doi: 10.1158/1078-0432.CCR-15-1790 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Licht JD Epigenetic Regulation and Therapeutic Targeting in Myeloma. Blood 132, SCI-37–SCI-37, doi: 10.1182/blood-2018-99-109522 (2018). [DOI] [Google Scholar]
- 84.Ezponda T et al. UTX/KDM6A Loss Enhances the Malignant Phenotype of Multiple Myeloma and Sensitizes Cells to EZH2 inhibition. Cell reports 21, 628–640, doi: 10.1016/j.celrep.2017.09.078 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Pawlyn C et al. Overexpression of EZH2 in multiple myeloma is associated with poor prognosis and dysregulation of cell cycle control. Blood cancer journal 7, e549, doi: 10.1038/bcj.2017.27 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Popovic R et al. Histone methyltransferase MMSET/NSD2 alters EZH2 binding and reprograms the myeloma epigenome through global and focal changes in H3K36 and H3K27 methylation. PLoS genetics 10, e1004566, doi: 10.1371/journal.pgen.1004566 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Walker BA et al. Identification of novel mutational drivers reveals oncogene dependencies in multiple myeloma. Blood 132, 587–597, doi: 10.1182/blood-2018-03-840132 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Raab MS et al. Spatially divergent clonal evolution in multiple myeloma: overcoming resistance to BRAF inhibition. Blood 127, 2155–2157, doi: 10.1182/blood-2015-12-686782 (2016). [DOI] [PubMed] [Google Scholar]
- 89.Andrulis M et al. Targeting the BRAF V600E mutation in multiple myeloma. Cancer Discov 3, 862–869, doi: 10.1158/2159-8290.CD-13-0014 (2013). [DOI] [PubMed] [Google Scholar]
- 90.Heuck CJ et al. Inhibiting MEK in MAPK pathway-activated myeloma. Leukemia 30, 976–980, doi: 10.1038/leu.2015.208 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Spencer A et al. The novel AKT inhibitor afuresertib shows favorable safety, pharmacokinetics, and clinical activity in multiple myeloma. Blood 124, 2190–2195, doi: 10.1182/blood-2014-03-559963 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Spencer A et al. Novel AKT Inhibitor Afuresertib In Combination With Bortezomib and Dexamethasone Demonstrates Favorable Safety Profile and Significant Clinical Activity In Patients With Relapsed/Refractory Multiple Myeloma. Blood 122, 283–283 (2013). [Google Scholar]
- 93.Tolcher AW et al. Phase I study of the MEK inhibitor trametinib in combination with the AKT inhibitor afuresertib in patients with solid tumors and multiple myeloma. Cancer Chemother. Pharmacol 75, 183–189, doi: 10.1007/s00280-014-2615-5 (2015). [DOI] [PubMed] [Google Scholar]
- 94.Tan DS, Bedard PL, Kuruvilla J, Siu LL & Razak AR Promising SINEs for embargoing nuclear-cytoplasmic export as an anticancer strategy. Cancer Discov 4, 527–537, doi: 10.1158/2159-8290.CD-13-1005 (2014). [DOI] [PubMed] [Google Scholar]
- 95.Vogl DT et al. Selective Inhibition of Nuclear Export With Oral Selinexor for Treatment of Relapsed or Refractory Multiple Myeloma. J. Clin. Oncol 36, 859–866, doi: 10.1200/JCO.2017.75.5207 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Bahlis NJ et al. Selinexor plus low-dose bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma. Blood, doi: 10.1182/blood-2018-06-858852 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Luptakova K et al. Lenalidomide enhances anti-myeloma cellular immunity. Cancer immunology, immunotherapy : CII 62, 39–49, doi: 10.1007/s00262-012-1308-3 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Kronke J et al. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science 343, 301–305, doi: 10.1126/science.1244851 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Amatangelo M et al. Iberdomide (CC-220) Has Synergistic Anti-Tumor and Immunostimulatory Activity Against Multiple Myeloma in Combination with Both Bortezomib and Dexamethasone, or in Combination with Daratumumab in Vitro. Blood 132, 1935–1935, doi: 10.1182/blood-2018-99-113383 (2018). [DOI] [Google Scholar]
- 100.Fisher SL & Phillips AJ Targeted protein degradation and the enzymology of degraders. Current opinion in chemical biology 44, 47–55, doi: 10.1016/j.cbpa.2018.05.004 (2018). [DOI] [PubMed] [Google Scholar]
- 101.Jackson JR, Patrick DR, Dar MM & Huang PS Targeted anti-mitotic therapies: can we improve on tubulin agents? Nature reviews. Cancer 7, 107–117, doi: 10.1038/nrc2049 (2007). [DOI] [PubMed] [Google Scholar]
- 102.Shah JJ et al. ARRY-520 Shows Durable Responses in Patients with Relapsed/Refractory Multiple Myeloma in a Phase 1 Dose-Escalation Study. Blood 118, 1860–1860 (2011). [Google Scholar]