Despite many recent advances in therapy, there is still no plateau in overall survival curves in multiple myeloma. Bispecific antibodies are a novel immunotherapeutic approach designed to bind antigens on malignant plasma cells and cytotoxic immune effector cells. Early-phase clinical trials targeting B-cell maturation antigen (BCMA), GPRC5D, and FcRH5 have demonstrated a favorable safety profile, with mainly low-grade cytokine release syndrome, cytopenias, and infections. Although dose escalation is ongoing in several studies, early efficacy data show response rates in the most active dose cohorts between 61% and 83% with many deep responses; however, durability remains to be established. Further clinical trial data are eagerly anticipated.

Significance:

Overall survival of triple-class refractory multiple myeloma remains poor. Bispecific antibodies are a novel immunotherapeutic modality with a favorable safety profile and impressive preliminary efficacy in heavily treated patients. Although more data are needed, bispecifics will likely become an integral part of the multiple myeloma treatment paradigm in the near future. Studies in earlier lines of therapy and in combination with other active anti–multiple myeloma agents will help further define the role of bispecifics in multiple myeloma.

Although there have been many advances in the treatment of multiple myeloma, there is still an unmet need for patients who are relapsed/refractory to currently available therapies. In particular, patients who are penta-refractory [refractory to two immunomodulatory drugs (IMiD), two proteasome inhibitors (PI), and an anti-CD38 monoclonal antibody] have been shown to have a median overall survival (OS) of less than 6 months (1). Immunotherapeutic approaches have been developed over recent years to harness the patient's immune system to destroy the malignant plasma cells. These developments include chimeric antigen receptor (CAR)-T cell therapy, antibody–drug conjugates (ADC), and, more recently, bispecific antibodies. Although each modality has its advantages and disadvantages, phase I trials of bispecific antibodies in multiple myeloma have shown early promise as a readily available off-the-shelf treatment with deep responses and limited incidence of grade ≥3 adverse events.

Bispecific antibodies are designed to bind both a target on the malignant plasma cells and on cytotoxic immune effector cells [T cells/natural killer (NK) cells] to create an immunologic synapse, leading to T/NK-cell activation and destruction of malignant plasma cells (Fig. 1; ref. 2). Bispecific antibodies have been developed with and without an Fc region. Although molecules lacking an Fc region have been shown to easily penetrate tumors due to their small size (3, 4), they require frequent or continuous infusion due to their short half-life. Bispecific antibodies with Fc regions have been shown to have an extended half-life, enabling less frequent dosing (5). For this reason, all of the bispecifics in ongoing phase I and II trials (with the exception of AMG420, which has been discontinued) have included an Fc region in their antibody structure. As of the writing of this article, there are currently at least 17 ongoing phase I/II trials (and two discontinued) with four different antigen targets (Table 1). All of these studies target CD3 on T cells; however, preclinical studies are also investigating NK-cell engagers as a novel mechanism of action, with early success (6–10). Additionally, trispecific antibodies, which are currently in preclinical investigation, attempt to add T-cell costimulatory proteins to decrease T-cell anergy (11, 12) or target dual myeloma antigens while engaging NK cells (7).

Figure 1.

Bispecific and trispecific antibody structure. Bispecific antibodies. A, Bispecific T-cell and NK-cell engagers bring immune effector cells in proximity to specific antigen-expressing myeloma cells to promote direct cell-mediated cytotoxicity. The Fc portion provides stability and a longer half-life in the circulation, allowing for intermittent rather than continuous dosing. B, Bispecific compounds lacking an Fc portion have a very short half-life and require continuous infusions. These are only representative schematics; there is significant variability in antibody structure across compounds, leading to differing pharmacokinetic and pharmacodynamic profiles. C, Trispecific antibody targeting an immune effector cell and two distinct myeloma antigens. D, Trispecific antibody with costimulation of the immune effector cell to enhance cytotoxicity.

Figure 1.

Bispecific and trispecific antibody structure. Bispecific antibodies. A, Bispecific T-cell and NK-cell engagers bring immune effector cells in proximity to specific antigen-expressing myeloma cells to promote direct cell-mediated cytotoxicity. The Fc portion provides stability and a longer half-life in the circulation, allowing for intermittent rather than continuous dosing. B, Bispecific compounds lacking an Fc portion have a very short half-life and require continuous infusions. These are only representative schematics; there is significant variability in antibody structure across compounds, leading to differing pharmacokinetic and pharmacodynamic profiles. C, Trispecific antibody targeting an immune effector cell and two distinct myeloma antigens. D, Trispecific antibody with costimulation of the immune effector cell to enhance cytotoxicity.

Close modal
Table 1.

Clinical trials for bispecific antibodies in multiple myeloma

AgentTargetsSponsorPhaseClinical trial IDStatus
AMG420 BCMAxCD3 Amgen NCT03836053 Completed 
AMG701 BCMAxCD3 Amgen I/II NCT03287908a Ongoing 
CC-93269 BCMAxCD3 Bristol Myers Squibb NCT03486067 Ongoing 
PF-06863135 BCMAxCD3 Pfizer I/II NCT04649359 Ongoing 
    NCT03269136a Ongoing 
REGN5458 BCMAxCD3 Regeneron I/II NCT03761108 Ongoing 
REGN5459 BCMAxCD3 Regeneron NCT04083534 Ongoing 
JNJ-64007957 (teclistamab) BCMAxCD3 Janssen NCT03145181 Ongoing 
   NCT04696809 Ongoing 
   I/II NCT04557098 Ongoing 
   Ib NCT04108195b Ongoing 
   Ib NCT04586426b Ongoing 
TNB-383B BCMAxCD3 TeneoBio NCT03933735 Ongoing 
GBR1342 CD38xCD3 Ichnos Sciences I/II NCT03309111 Ongoing 
AMG424 CD38xCD3 Amgen NCT03445663 Terminated 
JNJ-64407564 (talquetamab) GPRC5DxCD3 Janssen I/II NCT04634552 Ongoing 
   NCT03399799 Ongoing 
   Ib NCT04108195b Ongoing 
   Ib NCT04586426b Ongoing 
BFCR4350A (cevostamab) FcRH5xCD3 Genentech NCT03275103 Ongoing 
AgentTargetsSponsorPhaseClinical trial IDStatus
AMG420 BCMAxCD3 Amgen NCT03836053 Completed 
AMG701 BCMAxCD3 Amgen I/II NCT03287908a Ongoing 
CC-93269 BCMAxCD3 Bristol Myers Squibb NCT03486067 Ongoing 
PF-06863135 BCMAxCD3 Pfizer I/II NCT04649359 Ongoing 
    NCT03269136a Ongoing 
REGN5458 BCMAxCD3 Regeneron I/II NCT03761108 Ongoing 
REGN5459 BCMAxCD3 Regeneron NCT04083534 Ongoing 
JNJ-64007957 (teclistamab) BCMAxCD3 Janssen NCT03145181 Ongoing 
   NCT04696809 Ongoing 
   I/II NCT04557098 Ongoing 
   Ib NCT04108195b Ongoing 
   Ib NCT04586426b Ongoing 
TNB-383B BCMAxCD3 TeneoBio NCT03933735 Ongoing 
GBR1342 CD38xCD3 Ichnos Sciences I/II NCT03309111 Ongoing 
AMG424 CD38xCD3 Amgen NCT03445663 Terminated 
JNJ-64407564 (talquetamab) GPRC5DxCD3 Janssen I/II NCT04634552 Ongoing 
   NCT03399799 Ongoing 
   Ib NCT04108195b Ongoing 
   Ib NCT04586426b Ongoing 
BFCR4350A (cevostamab) FcRH5xCD3 Genentech NCT03275103 Ongoing 

aCombination study including non–bispecific antibody treatment.

bTrial includes JNJ-64007957 (teclistamab) and JNJ-64407564 (talquetamab).

In addition to the ongoing phase I/II trials, there are multiple preclinical bispecific agents under development (Table 2). These preclinical agents target multiple myeloma cells by way of BCMA, CD138, CD38, SLAMF7, and GPRC5D, and additionally target CD3, CD16a, NKp30, or NKG2D to recruit either T cells or NK cells. These studies have shown early preclinical success and suggest potential for future phase I trials in multiple myeloma (9, 12–26).

Table 2.

Preclinical bispecific and trispecific antibodies in multiple myeloma

TargetDrugSponsorNotes
BCMAxCD3 EM801 Bristol Myers Squibb Active also in high-risk patients and non–cross-resistant with previous lines of treatment (14) 
BCMAxCD3 AP163 Ampsource Biopharma Shanghai Inc. Induces less cytokine secretion than other bispecifics in vitro (16) 
BCMAxNKp30 CTX-8573 Compass Therapeutics Antitumor effect in mice even with low BCMA expression (9) 
BCMAxCD16a AFM26 Affimed Therapeutics NK-cell engager; may have superior safety profile over CD3 T-cell engagers (15) 
BCMAxCD16a RO7297089 Genentech Has a favorable safety profile and represents a novel MOA among other BCMA-targeting modalities (10) 
BCMAxMICA 2A9-MICA China Pharmaceutical University MICA binds NKG2D on NK cells to induce multiple myeloma cell death in vitro and in a mouse model (24) 
BCMAxCD200xCD16a Gantke et al. Affimed Therapeutics Results in increase in avidity leading to preferential lysis of antigen double-positive cells compared with antigen single-positive cells (7) 
CD138xCD3 STL001 Jiangsu, China Nanomolar-level affinity to recombinant human CD138 protein and shows more potent antitumor activity against RPMI-8226 cells than that of separate aCD3-ScFv-hIgFc and aCD138-ScFv-hIgFc, or the isotype mAb in vitro or in vivo (17) 
CD138xCD3 h-STL002, m-STL002 Jiangsu, China Shows potent cytotoxicity against multiple myeloma RPMI-8226 cell line through T-cell activation (18) 
CD38xCD3xCD28 Wu et al. Sanofi Demonstrates in vitro multiple myeloma cell killing 3–4 log higher than daratumumab (12) 
CD38xCD3 Sorrento CD38/CD3 Sorrento Therapeutics Demonstrates more potent tumor cell killing than daratumumab (19) 
CD38xCD3 Bi 38–3 Inserm Kills multiple myeloma cells in vitro and in a mouse model with no toxicity to B, T, and NK cells (23) 
SLAMF7xNKG2D SLAMF7-NKG2D Ohio State University In vivo, survival was significantly prolonged using SLAMF7-NKG2D biAb in a xenograft NOD-SCIDIL2γc−/− (NSG) mouse model engrafted with both human PBMCs and multiple myeloma cell lines (22) 
GPRC5DxCD3 GPRC5DxCD3 TRAB Chugai Pharmaceutical Suppresses tumor growth of GPRC5D-positive myeloma cells through the activation of T cellsin vitro and in vivo in xenograft models (20) 
NY-ESO-1xCD3 ImmTAC-NYE Immunocore Produces lysis of multiple myeloma cell line in vitro (25) 
A2/NY-ESO-1xCD3 Maruta et al. Ehime University, Japan Antimyeloma activity in vitro similar to CAR-T construct (26) 
TargetDrugSponsorNotes
BCMAxCD3 EM801 Bristol Myers Squibb Active also in high-risk patients and non–cross-resistant with previous lines of treatment (14) 
BCMAxCD3 AP163 Ampsource Biopharma Shanghai Inc. Induces less cytokine secretion than other bispecifics in vitro (16) 
BCMAxNKp30 CTX-8573 Compass Therapeutics Antitumor effect in mice even with low BCMA expression (9) 
BCMAxCD16a AFM26 Affimed Therapeutics NK-cell engager; may have superior safety profile over CD3 T-cell engagers (15) 
BCMAxCD16a RO7297089 Genentech Has a favorable safety profile and represents a novel MOA among other BCMA-targeting modalities (10) 
BCMAxMICA 2A9-MICA China Pharmaceutical University MICA binds NKG2D on NK cells to induce multiple myeloma cell death in vitro and in a mouse model (24) 
BCMAxCD200xCD16a Gantke et al. Affimed Therapeutics Results in increase in avidity leading to preferential lysis of antigen double-positive cells compared with antigen single-positive cells (7) 
CD138xCD3 STL001 Jiangsu, China Nanomolar-level affinity to recombinant human CD138 protein and shows more potent antitumor activity against RPMI-8226 cells than that of separate aCD3-ScFv-hIgFc and aCD138-ScFv-hIgFc, or the isotype mAb in vitro or in vivo (17) 
CD138xCD3 h-STL002, m-STL002 Jiangsu, China Shows potent cytotoxicity against multiple myeloma RPMI-8226 cell line through T-cell activation (18) 
CD38xCD3xCD28 Wu et al. Sanofi Demonstrates in vitro multiple myeloma cell killing 3–4 log higher than daratumumab (12) 
CD38xCD3 Sorrento CD38/CD3 Sorrento Therapeutics Demonstrates more potent tumor cell killing than daratumumab (19) 
CD38xCD3 Bi 38–3 Inserm Kills multiple myeloma cells in vitro and in a mouse model with no toxicity to B, T, and NK cells (23) 
SLAMF7xNKG2D SLAMF7-NKG2D Ohio State University In vivo, survival was significantly prolonged using SLAMF7-NKG2D biAb in a xenograft NOD-SCIDIL2γc−/− (NSG) mouse model engrafted with both human PBMCs and multiple myeloma cell lines (22) 
GPRC5DxCD3 GPRC5DxCD3 TRAB Chugai Pharmaceutical Suppresses tumor growth of GPRC5D-positive myeloma cells through the activation of T cellsin vitro and in vivo in xenograft models (20) 
NY-ESO-1xCD3 ImmTAC-NYE Immunocore Produces lysis of multiple myeloma cell line in vitro (25) 
A2/NY-ESO-1xCD3 Maruta et al. Ehime University, Japan Antimyeloma activity in vitro similar to CAR-T construct (26) 

Abbreviations: biAb, bispecific antibody; mAb, monoclonal antibody; MOA, mechanism of action; PBMC, peripheral blood mononuclear cell.

To maximize efficacy and minimize toxicity, bispecific antibodies should target an antigen that is unique and specific to multiple myeloma cells, with minimal expression in other healthy tissues. Although there are numerous potential myeloma cell targets (Fig. 2), four targets have been used in current phase I trials: BCMA, CD38, GPRC5D, and FcRH5.

Figure 2.

Bispecific and trispecific antibody targets in multiple myeloma. Filling color denotes functional class of the target antigen; filling intensity denotes stage of translational development: clinical (darker) or preclinical (lighter). NY-ESO-1 antigen is presented by MHC class I, which requires therapeutic antibody to recognize NY-ESO-1 in complex with patient-specific MHC-I allele. PD-1 has been targeted in combination with bispecific antibodies but is not itself a direct target of bispecifics.

Figure 2.

Bispecific and trispecific antibody targets in multiple myeloma. Filling color denotes functional class of the target antigen; filling intensity denotes stage of translational development: clinical (darker) or preclinical (lighter). NY-ESO-1 antigen is presented by MHC class I, which requires therapeutic antibody to recognize NY-ESO-1 in complex with patient-specific MHC-I allele. PD-1 has been targeted in combination with bispecific antibodies but is not itself a direct target of bispecifics.

Close modal

BCMA

B-cell maturation antigen (BCMA), or CD269, is an antigen that is expressed on both malignant and normal plasma cells at high levels (27, 28). It is a type III transmembrane glycoprotein in the tumor necrosis factor receptor superfamily, whose function is to regulate B-cell proliferation, maturation, survival, and differentiation to plasma cells through binding of its ligands, a proliferation-inducing ligand (APRIL) and B-cell–activating factor (BAFF; refs. 29–34). APRIL and BAFF signaling via BCMA are essential for humoral immunity, and blocking this interaction prevented antibody formation to the immunogenic carrier protein keyhole limpet hemocyanin and Pneumovax in mice (35). BCMA is almost exclusively expressed on mature B lymphocytes, plasmablasts, and plasma cells, while remaining undetectable in naïve B cells and hematopoietic stem cells. Although BCMA expression is also undetectable in most nonhematologic tissues, it has been found to have some expression in the testis, trachea, and gastrointestinal tract possibly due to the presence of plasma cells (36). BCMA has been shown to have increased expression on malignant plasma cells compared with normal plasma cells (27, 37) and is upregulated during disease progression from monoclonal gammopathy of undetermined significance to smoldering multiple myeloma to active multiple myeloma (38). Higher expression of soluble BCMA, thought to be from gamma-secretase–induced shedding, has been associated with worse outcomes in multiple myeloma (27). There are currently six bispecifics in clinical trials targeting BCMA.

CD38

CD38 is a surface glycoprotein that has been found to be an activation marker (39), an adhesion molecule (40), and an ectoenzyme involved in the metabolism of NAD+ and NADP (41, 42). CD38 is highly expressed on plasma cells, but is also found at lower levels on other hematologic cells including NK, lymphoid and myeloid cells, red blood cells, and platelets (43). CD38 expression has also been found at low levels on prostatic epithelial tissue, pancreatic islet cells, airway striated muscle cells, renal tubules, ganglia cells, and corneal cells and in the perikarya and dendrites of some neurons (44–47). This non–plasma cell expression of CD38 accounts for many of the side effects of the anti-CD38 antibodies daratumumab and isatuximab, and would presumably be a concern for bispecific antibodies with a CD38 target. Some examples of these side effects include bronchospasm during infusion, angle closure glaucoma, and myopic shift (48, 49). CD38 expression on plasma cells has also been shown to be downregulated after the first infusion of anti-CD38 antibodies, potentially generating resistance and inducing tumor escape over time (42, 50); however, clinical trials with daratumumab have shown durable responses in multiple myeloma (51–54). Two bispecifics targeting CD38 are in phase I trials. In addition, there is evidence that daratumumab causes depletion of CD38+ myeloid-derived suppressor cells and CD38+ regulatory T cells while inducing a clonal expansion of cytotoxic and helper T cells (55). This enhanced T-cell response can potentially be harnessed to increase T-cell engagement with bispecifics, and studies have begun adding daratumumab to non–CD38-targeting bispecifics (anti-BCMA and anti-GPRC5D) to test this hypothesis.

GPRC5D

The orphan G protein–coupled receptor class C group 5 member D (GPRC5D) is a relatively novel target for multiple myeloma immunotherapy. It is an orphan G protein–coupled receptor with unknown function that is highly expressed on malignant bone marrow plasma cells, as well as hard keratinized structures that include the hair shaft, nail, and central region of the tongue (56–58). High expression of GPRC5D has been associated with poor prognosis in multiple myeloma (59). GPRC5D has been used as a target in CAR-T therapy for multiple myeloma, with promising results in preclinical studies (56, 60), and is now a target for the bispecific antibody JNJ-64407564 (talquetamab) in four phase I trials.

FcRH5

The Fc receptor-homolog 5 (FcRH5) is a surface protein in the immunoglobulin superfamily that has been implicated in proliferation and isotype expression in the development of antigen-primed B cells (61–63). It is expressed only in the B-cell lineage with expression starting in pre–B cells and increasing expression through maturation to mature B cells and plasma cells (64). FcRH5 has also been shown to be more highly expressed in multiple myeloma plasma cells compared with normal plasma cells (61, 64, 65) and may be even more highly expressed in cell lines with 1q21 abnormalities (65, 66). Antibodies to FcRH5 were also found to internalize in preclinical models, making it a potential target for ADC, although a phase I trial was unsuccessful (67). Nonetheless, FcRH5, with its restricted expression to B-cell lineage and its upregulation in multiple myeloma, is the target of the bispecific BFCR4350Aa (cevostamab) in an ongoing phase I trial.

AMG420, a BCMAxCD3 bispecific T-cell engager, was the first to complete its phase I trial in multiple myeloma (68). As it did not have an Fc portion, it had a short half-life and was administered continuously through a pump for 4 weeks out of 6. Forty-two patients with at least two prior lines of therapy were enrolled, with 36% double refractory to an IMiD and PI. Dose-limiting toxicities (DLT) included grade 3 cytokine release syndrome (CRS) and grade 3 polyneuropathy. CRS occurred in 38% of patients, serious infections in 33% (including 12% central line infections), and grade 3 polyneuropathy in two (5%) patients. The polyneuropathies resolved after 2 to 3 months. At the maximum tolerated dose, 7/10 (70%) demonstrated a response. The development of this compound was subsequently discontinued in favor of a longer half-life bispecific antibody, AMG701. As a result, the following section focuses on compounds still in development, all of which have extended half-lives.

Safety

As of the writing of this article, there have been phase I data from eight additional trials, all reported from conference proceedings and not peer-reviewed publications (69–76). Two studies reached their recommended phase II dose (RP2D), with many studies reporting DLTs. These DLTs included acute kidney injury, elevated AST/ALT, delirium, confusion, thrombocytopenia, increased lipase, maculopapular rash, and pneumonia. These DLTs were generally manageable with supportive care. To date, there are no data available on study discontinuation due to adverse effects. The main adverse events are summarized below, with detailed data on individual trials presented in Table 3. As these are first-in-human phase I trials, there are no comparator arms to determine whether certain adverse effects occurred at a higher rate than expected for a heavily relapsed myeloma population.

Table 3.

Safety profile

AgentAMG701 (69)CC-93269 (75)PF-06863135 (76)REGN5458 (73)JNJ-64007957 (teclistamab) (72)TNB-383B (70)JNJ-64407564 (talquetamab) (74)BFCR4350A (cevostamab) (71)
Target BCMA BCMA BCMA BCMA BCMA BCMA GPRC5D FcRH5 
Patients (N) 65 19 30 49 149 (84 i.v., 65 s.c.) 58 157 (102 i.v., 55 s.c.) 53 
CRS (grade ≥3) 65% (9%) 90% (5%) 73.3% (0%) 39% (0%) 55% (0%) 45% (0%) 54% (3%) 76% (2%) 
Infection (grade ≥3) (17%) NR (26%) NR (30%) 47% (18%) 52% (15%) 21% (14%) 38% (8%) NR 
Anemia (grade ≥3) 42% NR (42%) 20% (16.7%) 37% (22%) 55% (32%) 21% (17%) 48% (27%) 28% (19%) 
Neutropenia (grade ≥3) 25% NR (53%) 33.3% (26.7%) 16% (14%) 57% (46%) 19% (16%) 47% (31%) 17% (15%) 
Lymphopenia (grade ≥3) NR NR 16% (16%) 18% (12%) NR NR 40% (36%) 15% (15%) 
Thrombocytopenia (grade ≥3) 21% NR (21%) 8% (5%) 18% (6%) 40% (22%) 17% (14%) 32% (13%) 32% (25%) 
Neurotoxicity (grade ≥3) NR NR 20% 12% (0%) 5% (1%) NR 6% (2%) 28% (0%) 
Other common SE (grade ≥3) Diarrhea 31%, hypophosphatemia 31% NR NR Fatigue 35% (6%), nausea 31% (0%), pyrexia 31% (2%), back pain 27% (4%) Pyrexia 30% (0%), diarrhea 23% (1%), nausea 22% (1%), fatigue 22% (1%), headache 22% (0%), cough 21% (2%) Fatigue 24% (2%), headache 22% (2%), nausea 21% (0%) Skin-related disorder 45%, dysgeusia 38%, fatigue 29% (1%), headache 27% (1%), pyrexia 27% (1%), diarrhea 25% (3%), nail disorders 17% Hypomagnesemia 28% (0%), diarrhea 28% (2%), hypokalemia 21% (4%) 
AgentAMG701 (69)CC-93269 (75)PF-06863135 (76)REGN5458 (73)JNJ-64007957 (teclistamab) (72)TNB-383B (70)JNJ-64407564 (talquetamab) (74)BFCR4350A (cevostamab) (71)
Target BCMA BCMA BCMA BCMA BCMA BCMA GPRC5D FcRH5 
Patients (N) 65 19 30 49 149 (84 i.v., 65 s.c.) 58 157 (102 i.v., 55 s.c.) 53 
CRS (grade ≥3) 65% (9%) 90% (5%) 73.3% (0%) 39% (0%) 55% (0%) 45% (0%) 54% (3%) 76% (2%) 
Infection (grade ≥3) (17%) NR (26%) NR (30%) 47% (18%) 52% (15%) 21% (14%) 38% (8%) NR 
Anemia (grade ≥3) 42% NR (42%) 20% (16.7%) 37% (22%) 55% (32%) 21% (17%) 48% (27%) 28% (19%) 
Neutropenia (grade ≥3) 25% NR (53%) 33.3% (26.7%) 16% (14%) 57% (46%) 19% (16%) 47% (31%) 17% (15%) 
Lymphopenia (grade ≥3) NR NR 16% (16%) 18% (12%) NR NR 40% (36%) 15% (15%) 
Thrombocytopenia (grade ≥3) 21% NR (21%) 8% (5%) 18% (6%) 40% (22%) 17% (14%) 32% (13%) 32% (25%) 
Neurotoxicity (grade ≥3) NR NR 20% 12% (0%) 5% (1%) NR 6% (2%) 28% (0%) 
Other common SE (grade ≥3) Diarrhea 31%, hypophosphatemia 31% NR NR Fatigue 35% (6%), nausea 31% (0%), pyrexia 31% (2%), back pain 27% (4%) Pyrexia 30% (0%), diarrhea 23% (1%), nausea 22% (1%), fatigue 22% (1%), headache 22% (0%), cough 21% (2%) Fatigue 24% (2%), headache 22% (2%), nausea 21% (0%) Skin-related disorder 45%, dysgeusia 38%, fatigue 29% (1%), headache 27% (1%), pyrexia 27% (1%), diarrhea 25% (3%), nail disorders 17% Hypomagnesemia 28% (0%), diarrhea 28% (2%), hypokalemia 21% (4%) 

Abbreviations: i.v., intravenous; NR, not reported; s.c., subcutaneous; SE, side effect.

Cytokine Release Syndrome

CRS is an acute systemic inflammatory syndrome caused by immunotherapies including CAR-T cells and bispecific antibodies. The pathophysiology involves activation of T cells and other immune effector cells with significant cytokine release. The severity of CRS is thought to result from a combination of disease burden, type of underlying malignancy, and type and dose of immunotherapy (77, 78). Fever is the only necessary symptom of CRS, but other possible clinical manifestations are wide-ranging and include hypotension, altered mental status, and hypoxia. There are different grading systems for CRS, making it difficult to compare CRS within and between immunotherapeutic modalities (79). In the six trials of anti-BCMA bispecific antibodies, the range of any-grade CRS was 39% to 95%. CRS of grade ≥3 ranged from 0% to 9%. In the anti-GPRC5D trial, CRS was found in 54% of patients with 3% grade ≥3. In the anti-FcRH5 trial, CRS was found in 76% of patients, with 2% grade ≥3. Across all studies, the use of tocilizumab and corticosteroids to treat CRS was 9% to 40% and 0% to 17%, respectively. Both the median time to onset and median duration of CRS ranged from 12 to 48 hours, with median time to onset generally quicker with intravenous administration (24 hours) compared with subcutaneous administration (48 hours). CRS was generally confined to the first cycle, and all patients recovered with no sequelae. Step-up dosing was used in most studies to attempt to mitigate the initial intensity of CRS.

Neurotoxicity

Neurotoxicity in these trials ranged from 5% to 28% at any grade and 0% to 2% at grade ≥3. Reported neurologic symptoms included headache, confusion, aphasia, cognitive disorder, and encephalopathy. Many of these symptoms were in the setting of CRS and resolved after CRS treatment. Of note, the AMG402 study, in the setting of continuous dosing (due to its short half-life), had a high rate of neuropathy. Neuropathy was not found to be a major side effect in all other reported trials based on presented data. Recently, the FDA placed a hold on PF-06863135 due to three cases of peripheral neuropathy.

Hematologic

Hematologic side effects across all trials that presented these data included anemia (any grade 21%–42%, grade ≥3 17%–42%), neutropenia (any grade 16%–57%, grade ≥3 14%–53%), lymphopenia (any grade 15%–40%, grade ≥3 12%–36%), leukopenia (any grade 23%–32%, grade ≥3 14%–16%), and thrombocytopenia (all 8%–40%, grade ≥3 6%–25%). As of this writing, there are no data on the timing and duration of these cytopenias. The dose–response relationship is also unknown from the early data. The mechanism of cytopenias is not currently understood as the targets of the bispecifics are not present on other cell lineages. Cytopenias can be seen even in patients attaining responses with these agents. Therefore, one hypothesis is a “bystander” effect from the release of cytokines and destruction of plasma cells in the bone marrow, though there is currently no evidence to support or refute this possibility.

Infection

In the six trials that reported, infection was found in 21% to 52% of patients, with grade ≥3 in 8% to 30%. The source and type of infection were not widely reported. The populations of patients in these phase I trials were all relapsed or refractory and were heavily pretreated. There is evidence of cumulative multifactorial immunosuppression with reduction of CD4+, CD45+, CD19+, and NK cells in relapsed/refractory patients with multiple lines of therapy (80). Treatment-emergent hypogammaglobulinemia has also been noted but has not been well characterized in studies to date. The relatively low rate of ≥ grade 3+ infections with anti-GPRC5D therapy and serious/unusual infections with the other agents raises the possibility that expression of target antigens on nonmyelomatous B cells could play a role. Preclinically, BCMA signaling is essential for generation of humoral immunity, which may predispose BCMA-treated patients to hypogammaglobulinemia, infectious complications, and lack of response to vaccines. The latter is particularly important in the era of COVID-19. At our institution, we found that multiple myeloma patients receiving BCMA-directed therapies had an odds ratio of 10.3 of not developing IgG antibodies to SARS-CoV-2 vaccination compared to non-BCMA–treated patients (81). Larger numbers of patients, longer follow-up, and ideally a control arm will be needed to truly elucidate the contribution of bispecific antibody treatment to infectious risk.

Other

Common nonhematologic side effects across all trials included fatigue (17%–35%), nausea (17%–31%), pyrexia (16%–31%), and diarrhea (12%–31%), while back pain, headache, and cough, and vomiting were also documented in some of the trials. Electrolyte abnormalities included hypophosphatemia, hypomagnesemia, and hypokalemia. The anti-GPRC5D antibody trial was unique in causing skin-related effects (45%), dysgeusia (38%), and nail disorders (17%), relating to the expression of GPRC5D on these hard keratinized structures. Although the majority of skin-related adverse events were grade 1/2, there were two DLTs (grade 3 maculopapular rashes). Data on tolerability and discontinuation are not yet publicly available. The anti-FcRH5 antibody trial was unique in causing AST increases in 15% of patients, though at this time, there is no detail on possible causes, including whether this occurred during CRS.

Efficacy

Triple-refractory and penta-refractory multiple myeloma patients have been shown to have a median OS of less than 1 year with an overall response rate (ORR) of 30% and 29%, respectively, to the next line of therapy, decreasing with each subsequent regimen (1). Monotherapy trials in this population that have led to FDA approval, including carfilzomib, pomalidomide, daratumumab, selinexor, belantamab mafodotin, and melflufen, have achieved an approximately 20% to 30% ORR and 3- to 4-month progression-free survival (PFS; refs. 82–87). The inclusion criteria in the bispecific phase I trials included relapsed/refractory multiple myeloma with ≥3 lines of therapy including an IMiD, a PI, and an anti-CD38 Ab; relapsed/refractory multiple myeloma for which no established therapy is available; or relapsed/refractory multiple myeloma and intolerant to other therapies. Triple-refractory patients comprised 62% to 100% of the population in the trials that reported these data, and the median number of prior lines of therapy was five to eight over a median of 5.6 to 7 years since diagnosis.

Although safety is the primary objective of these phase I dose-escalation trials, preliminary efficacy data are available for the most active dose cohorts, ranging from 6 to 22 patients (Table 4). Importantly, these trials are at differing stages of maturity, with only two having reached their RP2D, and as a result, ORRs are likely to change with more patients enrolled and longer follow-up. The ORR in these cohorts ranged from 61% to 83%, with a very good partial response (VGPR) rate of 39% to 78%. Two studies reported RP2D data. JNJ-64007957 (teclistamab) had 73% ORR at RP2D (55% ≥VGPR), with 70% ORR in triple- refractory and 75% ORR in penta-refractory patients. JNJ-64407564 (talquetamab) had a 69% ORR at RP2D (39% with ≥VGPR), with 67% ORR in triple-refractory (nine patients) and 100% ORR in penta-refractory (two patients); 17% of patients had received prior BCMA-directed therapy, but there are no results available for these patients at this time. The BFCR4350A (cevostamab) study had 21% with prior BCMA therapy; 5/8 (63%) of these patients had a response. Many of the studies reported minimal residual disease (MRD) negativity in evaluable patients.

Table 4.

Efficacy results

AgentAMG701 (69)CC-93269 (75)PF-06863135 (76)REGN5458 (73)JNJ-64007957 (teclistamab) (72)TNB-383B (70)JNJ-64407564 (talquetamab) (74)BFCR4350A (cevostamab) (71)
Target BCMA BCMA BCMA BCMA BCMA BCMA GPRC5D FcRH5 
Patients (N) 65 19 30 49 149 (84 i.v., 65 s.c.) 58 157 (102 i.v., 55 s.c.) 53 
Treatment Weekly i.v. Weekly i.v. Weekly s.c. Weekly i.v. Weekly i.v. or s.c. i.v. q3w Weekly i.v. or s.c. i.v. q3w 
Median prior lines 
Triple-class refractory 62% IMiD 84%, PI 90%, Dara 89% NR; 23% prior BCMA-directed 100% 81% 64% 82%; 17% prior BCMA directed 72%; 21% prior BCMA directed 
ORR at therapeutic dose (only two trials have reached RP2D) 26% all patients 10/12 (83%) 16/20 (80%) 5/8 (63%) 16/22 (73%) 12/15 (80%) 9/13 (69%) 11/18 (61%) 
 5/6 (83%) most recent cohort ≥6 mg i.v. 215–1,000 μg/kg s.c. 96 mg i.v. 1,500 μg/kg s.c. (RP2D) 40–60 mg i.v. 405 μg/kg s.c. (RP2D) 3.6/90 mg–3.6/132 mg i.v.5/8 (63%) for prior BCMA exposure 
Duration of response 17/21 (81%) ongoing at median 5.6 months NR NR 14/19 (74%) ongoing at median 6 months 32/25 (91%) s.c. ongoing at median 6.5 months 22/27 (81%) ongoing at median 4.5 months 17/17 (100%) ongoing at median 3.7 months at ≥ 405 μg/kg s.c. 11/18 (61%) ongoing at median 10.3 months at all doses 
AgentAMG701 (69)CC-93269 (75)PF-06863135 (76)REGN5458 (73)JNJ-64007957 (teclistamab) (72)TNB-383B (70)JNJ-64407564 (talquetamab) (74)BFCR4350A (cevostamab) (71)
Target BCMA BCMA BCMA BCMA BCMA BCMA GPRC5D FcRH5 
Patients (N) 65 19 30 49 149 (84 i.v., 65 s.c.) 58 157 (102 i.v., 55 s.c.) 53 
Treatment Weekly i.v. Weekly i.v. Weekly s.c. Weekly i.v. Weekly i.v. or s.c. i.v. q3w Weekly i.v. or s.c. i.v. q3w 
Median prior lines 
Triple-class refractory 62% IMiD 84%, PI 90%, Dara 89% NR; 23% prior BCMA-directed 100% 81% 64% 82%; 17% prior BCMA directed 72%; 21% prior BCMA directed 
ORR at therapeutic dose (only two trials have reached RP2D) 26% all patients 10/12 (83%) 16/20 (80%) 5/8 (63%) 16/22 (73%) 12/15 (80%) 9/13 (69%) 11/18 (61%) 
 5/6 (83%) most recent cohort ≥6 mg i.v. 215–1,000 μg/kg s.c. 96 mg i.v. 1,500 μg/kg s.c. (RP2D) 40–60 mg i.v. 405 μg/kg s.c. (RP2D) 3.6/90 mg–3.6/132 mg i.v.5/8 (63%) for prior BCMA exposure 
Duration of response 17/21 (81%) ongoing at median 5.6 months NR NR 14/19 (74%) ongoing at median 6 months 32/25 (91%) s.c. ongoing at median 6.5 months 22/27 (81%) ongoing at median 4.5 months 17/17 (100%) ongoing at median 3.7 months at ≥ 405 μg/kg s.c. 11/18 (61%) ongoing at median 10.3 months at all doses 

Abbreviations: Dara, daratumumab; i.v., intravenous; NR, not reported; q3w, every 3 weeks; s.c., subcutaneous.

Median duration of response (DOR) has not been reached, as most of these phase I trials are still ongoing and many patients are still responding and receiving treatment. Follow-up times vary across trials; at a median follow-up of 3.7 to 10.3 months, the majority of patients (61%–100%) across several trials have maintained their response. Median time to first response across all trials at all doses was 3 to 4 weeks.

Although further efficacy data are needed with larger numbers of patients and longer duration of follow-up, these preliminary response rates compare very favorably with recently approved drugs in multiple myeloma. The depth of response and likely DOR are impressive for a single agent in a heavily pretreated and refractory population. Additional phase I and II data are eagerly anticipated and will help inform the optimal use of bispecifics in multiple myeloma.

Despite the impressive preliminary efficacy of single-agent bispecifics, multiple myeloma remains a genetically heterogeneous disease (88), and relapses are still occurring. Multidrug therapy has long been standard of care for multiple myeloma, especially in relapsed and refractory patients, with several trials showing a benefit in PFS and sometimes OS with the addition of a third agent (52, 53, 89–96). Given the overall favorable toxicity profile of bispecifics, combination studies are the next logical step to improve further on efficacy.

At least four phase I/II trials are currently ongoing testing bispecific antibodies with/without one or more additional agents. The NCT03287908 trial is assessing a BCMAxCD3 antibody as monotherapy or in combination with pomalidomide with/without dexamethasone. The NCT03269136 trial is assessing a BCMAxCD3 bispecific antibody as monotherapy or in combination with either an anti–PD-1 monoclonal antibody, lenalidomide, or pomalidomide. The NCT04108195 trial has four treatment arms: BCMAxCD3 + daratumumab, GPRC5DxCD3 + daratumumab, BCMAxCD3 + daratumumab + pomalidomide, and GPRC5DxCD3 + daratumumab + pomalidomide. Lastly, the NCT04586426 trial combines BCMAxCD3 and GPRC5DxCD3 bispecific antibodies. These trials are ongoing, and no data have been reported yet.

Combination studies with anti-CD38 antibodies have a particularly strong preclinical rationale. Based on peripheral blood and bone marrow samples from patients on monotherapy trials, daratumumab was shown to decrease CD38+ myeloid-derived suppressor cells and CD38+ regulatory T cells, while causing a robust increase in cytotoxic and helper T cells in a majority of patients (47). In addition, there was a clonal expansion of T cells that correlated with myeloma response. Administering daratumumab alongside a bispecific antibody could potentially prime the patient's T cells and then optimally redirect them to the tumor cells. One preclinical study found that pretreatment with daratumumab increased efficacy of their BCMAxCD3 bispecific antibody in bone marrow samples (13).

Bispecific antibodies represent a promising class of therapeutics for multiple myeloma and will likely be integrated into the multiple myeloma treatment paradigm in the near future. However, there are still many unanswered questions and unexplored opportunities. Mechanisms of resistance are not well understood at this time and deserve further study to inform the rational sequencing/combinations of the various bispecifics. If immune checkpoints are overexpressed at the time of progression, it would provide a rationale for combination therapy with immune-checkpoint inhibitors, whereas if loss of efficacy is from decline in T-cell function, then perhaps anti-CD38 antibodies could be used to salvage these patients. Although some combination studies are in progress as described above, there are countless opportunities for combination therapy, particularly with agents that do not overlap with the main non-CRS toxicities of bispecifics, which include cytopenias and infections.

Although novel treatments in multiple myeloma are conventionally studied in patients who have exhausted most or all available therapies, there is significant potential for bispecifics to move into earlier lines of therapy. Many multiple myeloma patients will not be able to receive multiple lines of therapy, whether due to comorbidities, toxicities from treatment, or early mortality, and, additionally, each successive line of therapy brings shorter remissions (97). Importantly, T-cell function declines as a result of multiple myeloma, its therapies, and aging, and there is evidence of T-cell exhaustion as a distinguishing feature of relapse in multiple myeloma (98, 99). As such, there is a need to utilize T-cell–redirecting therapies and achieve deep and durable remissions early in the treatment course when patients can receive the most benefit. Given the high response rates, deep responses (including MRD negative), and potential durability of response, as well as limited toxicity and convenient dosing schedules, bispecifics may ultimately prove most useful in the upfront or early relapse setting. In addition, high-risk groups, including elderly/frail patients, high-risk cytogenetics, renal failure, and extramedullary disease, have not benefited from novel therapies to the same degree as standard-risk patients. Clinical trials of bispecifics in these patient populations are urgently needed.

At this time, the toxicities of bispecifics are not fully understood. Although low-grade CRS and neurotoxicity are common, it is not possible yet to predict who will be at highest risk and should be monitored in the hospital for the first dose versus those who could potentially be safely monitored at home. Further work is needed to better understand patient characteristics and biomarkers that may lead to higher risk of CRS and neurotoxicity in order to implement appropriate dosing (step-up or priming doses), monitoring strategies, and possible empiric use of tocilizumab/corticosteroids. Subcutaneous bispecifics have been shown to achieve similar plasma concentrations as intravenous formulations but with a slower time to maximal concentration and potentially less severe CRS, although this requires additional study. Cytopenias are not yet well characterized in terms of duration and need for supportive care, and further insight is needed into the mechanisms of this toxicity. Lastly, given that all of these bispecifics target normal plasma cells in addition to myeloma cells, the contribution of hypogammaglobulinemia to infectious risk needs to be elucidated, as this may inform prophylactic strategies, including the use of subcutaneous or intravenous immunoglobulin, as well as vaccination strategies, particularly regarding COVID-19.

Finally, with the recent approval of the first BCMA-directed CAR-T therapy in multiple myeloma, idecabtagene vicleucel (ide-cel), many questions linger about the utilization of bispecifics versus CAR-T in the future. As of now, there are no head-to-head trials comparing these modalities, and cross-trial comparisons of bispecific and CAR-T studies are not advisable given the small numbers of patients, differing patient populations, variable disease characteristics, multiple dosing schedules, and bridging chemotherapy for CAR-T patients. Until further trials are conducted, choosing between the two modalities will depend on a variety of practical considerations. Bispecifics are off-the-shelf products and induce rapid responses, which is ideal for rapidly progressing patients who may not be able to wait several weeks for the manufacture of CAR-T cells. Allogeneic CAR-Ts represent an opportunity for off-the-shelf CAR-Ts; however, these are still early in phase I trials. Additionally, CRS is typically milder with bispecifics (almost all grade 1/2), which may be easier to tolerate for elderly or frail patients. Bispecifics require ongoing treatment, in contrast to a one-time intervention with CAR-T, and may be less practical for patients who live far from academic medical centers or who otherwise are unable to maintain a consistent follow-up schedule. At this time, there are no data to inform the optimal duration of therapy with bispecifics. There is a theoretical risk of decreasing the efficacy of bispecifics if administered soon after CAR-T given the prolonged lymphodepletion from CAR-T conditioning therapy, which would favor sequencing bispecifics prior to CAR-T. Sequencing of targets is poorly understood at this time, and there will be a significant unmet need for post-BCMA therapies given the recent approval of ide-cel and the BCMA-targeted ADC belantamab mafodotin. Both bispecifics and CAR-T are expected to move into earlier lines of treatment given their high efficacy. However, as multiple myeloma remains a heterogeneous disease, combination therapies will be essential to prevent relapse. Bispecifics may be more easily incorporated with currently approved myeloma therapies given their dosing schedule and the ability to hold doses for toxicity, whereas there are currently several strategies to engineer CAR-T cells to overcome resistance, which are outside the scope of this review. Overall, both bispecifics and CAR-Ts have shown unprecedented response rates in heavily treated patients. As they usher in a new era of immunotherapy in multiple myeloma, it is heartening to have such powerful agents in our arsenal to personalize therapy for patients with multiple myeloma as we continue the search for a cure.

H.J. Cho reports other support from Multiple Myeloma Research Foundation and grants from Bristol Myers Squibb and Takeda outside the submitted work. S. Jagannath reports other from Bristol Myers Squibb, Janssen Pharmaceuticals, Karyopharm Therapeutics, Legend Biotech, Sanofi, and Takeda outside the submitted work. D. Madduri reports other from Amgen, Regeneron, and Allogene; personal fees and other from Janssen, Bristol Myers Squibb, and Celgene; and personal fees from GlaxoSmithKline, Kinevant, Sanofi, Legend, and Takeda outside the submitted work. J. Richter reports personal fees from Bristol Myers Squibb, Janssen, Takeda, Karyopharm, Adaptive Biotechnologies, and Oncopeptides outside the submitted work. A. Chari reports grants and personal fees from Janssen, Celgene, Novartis Pharmaceuticals, Amgen, Seattle Genetics, and Millenium/Takeda; personal fees from Bristol Myers Squibb, Karyopharm, Sanofi, Oncopeptides, Antengene, GlaxoSmithKline, and Secura Bio; and grants from Pharmacyclics outside the submitted work. No disclosures were reported by the other authors.

1.
Gandhi
UH
,
Cornell
RF
,
Lakshman
A
,
Gahvari
ZJ
,
McGehee
E
,
Jagosky
MH
, et al
Outcomes of patients with multiple myeloma refractory to CD38-targeted monoclonal antibody therapy
.
Leukemia
2019
;
33
:
2266
75
.
2.
Kontermann
RE
,
Brinkmann
U
. 
Bispecific antibodies
.
Drug Discov Today
2015
;
20
:
838
47
.
3.
Ellerman
D
. 
Bispecific T-cell engagers: towards understanding variables influencing the in vitro potency and tumor selectivity and their modulation to enhance their efficacy and safety
.
Methods
2019
;
154
:
102
17
.
4.
Adams
GP
,
Schier
R
,
McCall
AM
,
Simmons
HH
,
Horak
EM
,
Alpaugh
RK
, et al
High affinity restricts the localization and tumor penetration of single-chain fv antibody molecules
.
Cancer Res
2001
;
61
:
4750
5
.
5.
Saxena
A
,
Wu
D
. 
Advances in therapeutic Fc engineering–modulation of IgG-associated effector functions and serum half-life
.
Front Immunol
2016
;
7
:
580
.
6.
Ross
T
,
Reusch
U
,
Wingert
S
,
Haneke
T
,
Klausz
K
,
Otte
AK
, 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
2018
;
132
:
1927
.
7.
Gantke
T
,
Weichel
M
,
Herbrecht
C
,
Reusch
U
,
Ellwanger
K
,
Fucek
I
, et al
Trispecific antibodies for CD16A-directed NK cell engagement and dual-targeting of tumor cells
.
Protein Eng Des Sel
2017
;
30
:
673
84
.
8.
Draghi
M
,
Schafer
JL
,
Nelson
A
,
Frye
Z
,
Oliphant
A
,
Haserlat
S
, et al
Preclinical development of a first-in-class NKp30xBCMA NK cell engager for the treatment of multiple myeloma [abstract]
. In:
Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29–Apr 3; Atlanta, GA
.
Philadelphia (PA)
:
AACR
;
Cancer Res 2019;79(13 Suppl):Abstract nr 4972
.
9.
Watkins-Yoon
J
,
Guzman
W
,
Oliphant
A
,
Haserlat
S
,
Leung
A
,
Chottin
C
, et al
CTX-8573, an innate-cell engager targeting BCMA, is a highly potent multispecific antibody for the treatment of multiple myeloma
.
Blood
2019
;
134
(
Supplement_1
):
3182
.
10.
Kakiuchi-Kiyota
S
,
Schutten
MM
,
Adedeji
AO
,
Cai
H
,
Hendricks
R
,
Liu
L
, et al
Preclinical pharmacology and safety of RO7297089, a novel anti-BCMA/CD16a bispecific antibody for the treatment of multiple myeloma [abstract]
. In:
Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27–28 and Jun 22–24
.
Philadelphia (PA)
:
AACR
;
Cancer Res 2020;80(16 Suppl):Abstract nr 4556.
11.
Esensten
JH
,
Helou
YA
,
Chopra
G
,
Weiss
A
,
Bluestone
JA
. 
CD28 costimulation: from mechanism to therapy
.
Immunity
2016
;
44
:
973
88
.
12.
Wu
L
,
Seung
E
,
Xu
L
,
Rao
E
,
Lord
DM
,
Wei
RR
, et al
Trispecific antibodies enhance the therapeutic efficacy of tumor-directed T cells through T cell receptor co-stimulation
.
Nat Cancer
2020
;
1
:
86
98
.
13.
Frerichs
KA
,
Broekmans
ME
,
Soto
JAM
,
van Kessel
B
,
Heymans
MW
,
Holthof
LC
, et al
Preclinical activity of JNJ-7957, a novel BCMA × CD3 bispecific antibody for the treatment of multiple myeloma, is potentiated by daratumumab
.
Clin Cancer Res
2020
;
26
:
2203
15
.
14.
Seckinger
A
,
Delgado
JA
,
Moser
S
,
Moreno
L
,
Neuber
B
,
Grab
A
, et al
Target expression, generation, preclinical activity, and pharmacokinetics of the BCMA-T cell bispecific antibody EM801 for multiple myeloma treatment
.
Cancer Cell
2017
;
31
:
396
410
.
15.
Gantke
T
,
Reusch
U
,
Kellner
C
,
Ellwanger
K
,
Fucek
I
,
Weichel
M
, et al
AFM26 is a novel, highly potent BCMA/CD16A-directed bispecific antibody for high affinity NK-cell engagement in multiple myeloma
.
J Clin Oncol
2017
;
35
:
5
.
16.
Li
Z
,
Li
Q
,
Zhang
G
,
Ma
X
,
Hu
X
,
Ouyang
K
, et al
A novel bispecific BCMAxCD3 T cell-engaging antibody that treat multiple myeloma (MM) with minimal cytokine secretion
.
Ann Oncol
2019
;
30
:
v808
.
17.
Zou
J
,
Chen
D
,
Zong
Y
,
Ye
S
,
Tang
J
,
Meng
H
, et al
Immunotherapy based on bispecific T-cell engager with hIgG 1 Fc sequence as a new therapeutic strategy in multiple myeloma
.
Cancer Sci
2015
;
106
:
512
21
.
18.
Chen
D
,
Zou
J
,
Zong
Y
,
Meng
H
,
An
G
,
Yang
L
. 
Anti-human CD138 monoclonal antibodies and their bispecific formats: generation and characterization
.
Immunopharmacol Immunotoxicol
2016
;
38
:
175
83
.
19.
He
X
,
Zhang
Y
,
Lai
YW
,
Baguley
S
,
Li
Y
,
Cao
X
, et al
Preclinical characterization of an ANTI-CD38/CD3 T cell-redirecting bispecific antibody
.
Blood
2019
;
134
(
Supplement_1
):
4463
.
20.
Kodama
T
,
Kochi
Y
,
Nakai
W
,
Mizuno
H
,
Baba
T
,
Habu
K
, et al
Anti-GPRC5D/CD3 bispecific T-cell–redirecting antibody for the treatment of multiple myeloma
.
Mol Cancer Ther
2019
;
18
:
1555
64
.
21.
Verkleij
C
,
Broekmans
M
,
van Duin
M
,
Frerichs
K
,
Kuiper
R
,
Kaiser
M
, et al
Preclinical evaluation of the new GPRC5DxCD3 (JNJ-7564) bispecific antibody for the treatment of multiple myeloma
.
Clin Lymphoma Myeloma Leuk
2019
;
19
:
e122
3
.
22.
Chan
WK
,
Kang
S
,
Youssef
Y
,
Glankler
EN
,
Barrett
ER
,
Carter
AM
, et al
A CS1-NKG2D bispecific antibody collectively activates cytolytic immune cells against multiple myeloma
.
Cancer Immunol Res
2018
;
6
:
776
87
.
23.
Fayon
M
,
Martinez-Cingolani
C
,
Abecassis
A
,
Roders
N
,
Nelson
E
,
Choisy
C
, et al
Bi 38-3 is a novel CD38/CD3 bispecific T-cell engager with low toxicities for the treatment of multiple myeloma
.
Haematologica
2021
;
106
:
1193
7
.
24.
Wang
Y
,
Li
H
,
Xu
W
,
Pan
M
,
Qiao
C
,
Cai
J
, et al
BCMA-targeting bispecific antibody that simultaneously stimulates NKG2D-enhanced efficacy against multiple myeloma
.
J Immunother
2020
;
43
:
175
88
.
25.
McCormack
E
,
Adams
KJ
,
Hassan
NJ
,
Kotian
A
,
Lissin
NM
,
Sami
M
, et al
Bi-specific TCR-anti CD3 redirected T-cell targeting of NY-ESO-1- and LAGE-1-positive tumors
.
Cancer Immunol Immunother
2013
;
62
:
773
85
.
26.
Maruta
M
,
Ochi
T
,
Tanimoto
K
,
Asai
H
,
Saitou
T
,
Fujiwara
H
, et al
Direct comparison of target-reactivity and cross-reactivity induced by CAR- and BiTE-redirected T cells for the development of antibody-based T-cell therapy
.
Sci Rep
2019
;
9
:
13293
.
27.
Lee
L
,
Bounds
D
,
Paterson
J
,
Herledan
G
,
Sully
K
,
Seestaller-Wehr
LM
, et al
Evaluation of B cell maturation antigen as a target for antibody drug conjugate mediated cytotoxicity in multiple myeloma
.
Br J Haematol
2016
;
174
:
911
22
.
28.
Wei
J
,
Han
X
,
Bo
J
,
Han
W
. 
Target selection for CAR-T therapy
.
J Hematol Oncol
2019
;
12
:
1
9
.
29.
Madry
C
,
Laabi
Y
,
Callebaut
I
,
Roussel
J
,
Hatzoglou
A
,
Le Coniat
M
, et al
The characterization of murine BCMA gene defines it as a new member of the tumor necrosis factor receptor superfamily
.
Int Immunol
1998
;
10
:
1693
702
.
30.
Yu
B
,
Jiang
T
,
Liu
D
. 
BCMA-targeted immunotherapy for multiple myeloma
.
J Hematol Oncol
2020
;
13
:
1
24
.
31.
Coquery
CM
,
Erickson
LD
. 
Regulatory roles of the tumor necrosis factor receptor BCMA
.
Crit Rev Immunol
2012
;
32
:
287
305
.
32.
Thompson
JS
,
Schneider
P
,
Kalled
SL
,
Wang
L
,
Lefevre
EA
,
Cachero
TG
, et al
BAFF binds to the tumor necrosis factor receptor–like molecule B cell maturation antigen and is important for maintaining the peripheral B cell population
.
J Exp Med
2000
;
192
:
129
36
.
33.
Sasaki
Y
,
Casola
S
,
Kutok
JL
,
Rajewsky
K
,
Schmidt-Supprian
M
. 
TNF family member B cell-activating factor (BAFF) receptor-dependent and-independent roles for BAFF in B cell physiology
.
J Immunol
2004
;
173
:
2245
52
.
34.
Darce
JR
,
Arendt
BK
,
Chang
SK
,
Jelinek
DF
. 
Divergent effects of BAFF on human memory B cell differentiation into Ig-secreting cells
.
J Immunol
2007
;
178
:
5612
22
.
35.
Yu
G
,
Boone
T
,
Delaney
J
,
Hawkins
N
,
Kelley
M
,
Ramakrishnan
M
, et al
APRIL and TALL-1 and receptors BCMA and TACI: system for regulating humoral immunity
.
Nat Immunol
2000
;
1
:
252
6
.
36.
Carpenter
RO
,
Evbuomwan
MO
,
Pittaluga
S
,
Rose
JJ
,
Raffeld
M
,
Yang
S
, et al
B-cell maturation antigen is a promising target for adoptive T-cell therapy of multiple myeloma
.
Clin Cancer Res
2013
;
19
:
2048
60
.
37.
Eckhert
E
,
Hewitt
R
,
Liedtke
M
. 
B-cell maturation antigen directed monoclonal antibody therapies for multiple myeloma
.
Immunotherapy
2019
;
11
:
801
11
.
38.
Dispenzieri
A
,
Soof
CM
,
Rajkumar
SV
,
Gertz
MA
,
Kumar
S
,
Bujarski
S
, et al
Serum BCMA levels to predict outcomes for patients with MGUS and smoldering multiple myeloma (SMM)
.
J Clin Oncol 37
:
15s, 2019 (suppl; abstr 8020)
.
39.
Funaro
A
,
Spagnoli
G
,
Ausiello
C
,
Alessio
M
,
Roggero
S
,
Delia
D
, et al
Involvement of the multilineage CD38 molecule in a unique pathway of cell activation and proliferation
.
J Immunol
1990
;
145
:
2390
6
.
40.
Deaglio
S
,
Morra
M
,
Mallone
R
,
Ausiello
CM
,
Prager
E
,
Garbarino
G
, et al
Human CD38 (ADP-ribosyl cyclase) is a counter-receptor of CD31, an Ig superfamily member
.
J Immunol
1998
;
160
:
395
402
.
41.
Howard
M
,
Grimaldi
JC
,
Bazan
JF
,
Lund
FE
,
Santos-Argumedo
L
,
Parkhouse
R
, et al
Formation and hydrolysis of cyclic ADP-ribose catalyzed by lymphocyte antigen CD38
.
Science
1993
;
262
:
1056
9
.
42.
Morandi
F
,
Horenstein
AL
,
Costa
F
,
Giuliani
N
,
Pistoia
V
,
Malavasi
F
. 
CD38: a target for immunotherapeutic approaches in multiple myeloma
.
Front Immunol
2018
;
9
:
2722
.
43.
Deaglio
S
,
Mehta
K
,
Malavasi
F
. 
Human CD38: a (r) evolutionary story of enzymes and receptors
.
Leuk Res
2001
;
25
:
1
12
.
44.
Crowell
PD
,
Goldstein
AS
. 
Functional evidence that progenitor cells near sites of inflammation are precursors for aggressive prostate cancer
.
Mol Cell Oncol
2017
;
4
:
e1279723
.
45.
Kotlikoff
MI
,
Kannan
MS
,
Solway
J
,
Deng
K-Y
,
Deshpande
DA
,
Dowell
M
, et al
Methodologic advancements in the study of airway smooth muscle
.
J Allergy Clin Immunol
2004
;
114
:
S18
31
.
46.
Horenstein
AL
,
Sizzano
F
,
Lusso
R
,
Besso
FG
,
Ferrero
E
,
Deaglio
S
, et al
CD38 and CD157 ectoenzymes mark cell subsets in the human corneal limbus
.
Mol Med
2009
;
15
:
76
84
.
47.
van de Donk
NW
,
Richardson
PG
,
Malavasi
F
. 
CD38 antibodies in multiple myeloma: back to the future
.
Blood
2018
;
131
:
13
29
.
48.
Lee
AC
,
Greaves
G
,
Lee
R
,
Zdravkova
M
,
Ahmad
S
,
McGovern
K
, et al
Bilateral angle closure following the infusion of a monoclonal antibody to treat relapsing multiple myeloma
.
J Glaucoma
2018
;
27
:
e145
7
.
49.
Mavrommatis
MA
,
Jung
H
,
Chari
A
,
Barlogie
B
,
Chelnis
JG
. 
Daratumumab-induced transient myopic shift
.
Am J Ophthalmol Case Rep
2019
;
13
:
116
8
.
50.
Nijhof
IS
,
Casneuf
T
,
Van Velzen
J
,
van Kessel
B
,
Axel
AE
,
Syed
K
, et al
CD38 expression and complement inhibitors affect response and resistance to daratumumab therapy in myeloma
.
Blood
2016
;
128
:
959
70
.
51.
Lokhorst
HM
,
Plesner
T
,
Laubach
JP
,
Nahi
H
,
Gimsing
P
,
Hansson
M
, et al
Targeting CD38 with daratumumab monotherapy in multiple myeloma
.
N Engl J Med
2015
;
373
:
1207
19
.
52.
Palumbo
A
,
Chanan-Khan
A
,
Weisel
K
,
Nooka
AK
,
Masszi
T
,
Beksac
M
, et al
Daratumumab, bortezomib, and dexamethasone for multiple myeloma
.
N Engl J Med
2016
;
375
:
754
66
.
53.
Dimopoulos
MA
,
Oriol
A
,
Nahi
H
,
San-Miguel
J
,
Bahlis
NJ
,
Usmani
SZ
, et al
Daratumumab, lenalidomide, and dexamethasone for multiple myeloma
.
N Engl J Med
2016
;
375
:
1319
31
.
54.
Mateos
MV
,
Dimopoulos
MA
,
Cavo
M
,
Suzuki
K
,
Jakubowiak
A
,
Knop
S
, et al
Daratumumab plus bortezomib, melphalan, and prednisone for untreated myeloma
.
N Engl J Med
2018
;
378
:
518
28
.
55.
Krejcik
J
,
Casneuf
T
,
Nijhof
IS
,
Verbist
B
,
Bald
J
,
Plesner
T
, et al
Daratumumab depletes CD38+ immune regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma
.
Blood
2016
;
128
:
384
94
.
56.
Smith
EL
,
Harrington
K
,
Staehr
M
,
Masakayan
R
,
Jones
J
,
Long
TJ
, et al
GPRC5D is a target for the immunotherapy of multiple myeloma with rationally designed CAR T cells
.
Sci Transl Med
2019
;
11
:
eaau7746
.
57.
Pillarisetti
K
,
Edavettal
S
,
Mendonça
M
,
Li
Y
,
Tornetta
M
,
Babich
A
, et al
A T-cell–redirecting bispecific G-protein–coupled receptor class 5 member D x CD3 antibody to treat multiple myeloma
.
Blood
2020
;
135
:
1232
43
.
58.
Inoue
S
,
Nambu
T
,
Shimomura
T
. 
The RAIG family member, GPRC5D, is associated with hard-keratinized structures
.
J Invest Dermatol
2004
;
122
:
565
73
.
59.
Atamaniuk
J
,
Gleiss
A
,
Porpaczy
E
,
Kainz
B
,
Grunt
TW
,
Raderer
M
, et al
Overexpression of G protein-coupled receptor 5D in the bone marrow is associated with poor prognosis in patients with multiple myeloma
.
Eur J Clin Invest
2012
;
42
:
953
60
.
60.
de Larrea
CF
,
Staehr
M
,
Lopez
AV
,
Ng
KY
,
Chen
Y
,
Godfrey
WD
, et al
Defining an optimal dual-targeted CAR T-cell therapy approach simultaneously targeting BCMA and GPRC5D to prevent BCMA escape–driven relapse in multiple myeloma
.
Blood Cancer Discov
2020
;
1
:
146
.
61.
Elkins
K
,
Zheng
B
,
Go
M
,
Slaga
D
,
Du
C
,
Scales
SJ
, et al
FcRL5 as a target of antibody–drug conjugates for the treatment of multiple myeloma
.
Mol Cancer Ther
2012
;
11
:
2222
32
.
62.
Davis
RS
,
Ehrhardt
GR
,
Leu
CM
,
Hirano
M
,
Cooper
MD
. 
An extended family of Fc receptor relatives
.
Eur J Immunol
2005
;
35
:
674
80
.
63.
Dement-Brown
J
,
Newton
CS
,
Ise
T
,
Damdinsuren
B
,
Nagata
S
,
Tolnay
M
. 
Fc receptor-like 5 promotes B cell proliferation and drives the development of cells displaying switched isotypes
.
J Leukoc Biol
2012
;
91
:
59
67
.
64.
Polson
AG
,
Zheng
B
,
Elkins
K
,
Chang
W
,
Du
C
,
Dowd
P
, et al
Expression pattern of the human FcRH/IRTA receptors in normal tissue and in B-chronic lymphocytic leukemia
.
Int Immunol
2006
;
18
:
1363
73
.
65.
Li
J
,
Stagg
NJ
,
Johnston
J
,
Harris
MJ
,
Menzies
SA
,
DiCara
D
, et al
Membrane-proximal epitope facilitates efficient T cell synapse formation by anti-FcRH5/CD3 and is a requirement for myeloma cell killing
.
Cancer Cell
2017
;
31
:
383
95
.
66.
Inoue
J
,
Otsuki
T
,
Hirasawa
A
,
Imoto
I
,
Matsuo
Y
,
Shimizu
S
, et al
Overexpression of PDZK1 within the 1q12-q22 amplicon is likely to be associated with drug-resistance phenotype in multiple myeloma
.
Am J Pathol
2004
;
165
:
71
81
.
67.
Stewart
AK
,
Krishnan
AY
,
Singhal
S
,
Boccia
RV
,
Patel
MR
,
Niesvizky
R
, et al
Phase I study of the anti-FcRH5 antibody-drug conjugate DFRF4539A in relapsed or refractory multiple myeloma
.
Blood Cancer J
2019
;
9
:
1
12
.
68.
Topp
MS
,
Duell
J
,
Zugmaier
G
,
Attal
M
,
Moreau
P
,
Langer
C
, et al
Anti-B-cell maturation antigen BiTE molecule AMG 420 induces responses in multiple myeloma
.
J Clin Oncol
2020
;
38
:
775
83
.
69.
Harrison
SJ
,
Minnema
MC
,
Lee
HC
,
Spencer
A
,
Kapoor
P
,
Madduri
D
, et al
A phase 1 first in human (FIH) study of AMG 701, an anti-B-cell maturation antigen (BCMA) half-life extended (HLE) BiTE® (bispecific T-cell engager) molecule, in relapsed/refractory (RR) multiple myeloma (MM)
.
Blood
2020
;
136
:
28
9
.
70.
Rodriguez
C
,
D'Souza
A
,
Shah
N
,
Voorhees
PM
,
Buelow
B
,
Vij
R
, et al
Initial results of a phase I study of TNB-383B, a BCMA x CD3 bispecific T-cell redirecting antibody, in relapsed/refractory multiple myeloma
.
Blood
2020
;
136
:
43
4
.
71.
Cohen
AD
,
Harrison
SJ
,
Krishnan
A
,
Fonseca
R
,
Forsberg
PA
,
Spencer
A
, et al
Initial clinical activity and safety of BFCR4350A, a FcRH5/CD3 T-cell-engaging bispecific antibody, in relapsed/refractory multiple myeloma
.
Blood
2020
;
136
:
42
3
.
72.
Garfall
AL
,
Usmani
SZ
,
Mateos
MV
,
Nahi
H
,
van de Donk
NWCJ
,
San-Miguel
JF
, et al
Updated phase 1 results of teclistamab, a B-cell maturation antigen (BCMA) x CD3 bispecific antibody, in relapsed and/or refractory multiple myeloma (RRMM)
.
Blood
2020
;
136
:
27
.
73.
Madduri
D
,
Rosko
A
,
Brayer
J
,
Zonder
J
,
Bensinger
WI
,
Li
J
, et al
REGN5458, a BCMA x CD3 bispecific monoclonal antibody, induces deep and durable responses in patients with relapsed/refractory multiple myeloma (RRMM)
.
Blood
2020
;
136
:
41
2
.
74.
Chari
A
,
Berdeja
JG
,
Oriol
A
,
van de Donk
NWCJ
,
Rodriguez
P
,
Askari
E
, et al
A phase 1, first-in-human study of talquetamab, a G protein-coupled receptor family C group 5 member D (GPRC5D) x CD3 bispecific antibody, in patients with relapsed and/or refractory multiple myeloma (RRMM)
.
Blood
2020
;
136
:
40
1
.
75.
Costa
L
,
Wong
S
,
Bermudez
A
,
De la Rubia
J
,
Mateos
M
,
Ocio
E
, et al
Interim results from the first phase 1 clinical study of the B-cell maturation antigen (BCMA) 2+ 1 T-cell engager (TCE) CC-93269 in patients (pts) with relapsed/refractory multiple myeloma (RRMM)
.
EHA Library
2020
;
295025
:
S205
.
76.
Lesokhin
AM
,
Levy
MY
,
Dalovisio
AP
,
Bahlis
NJ
,
Solh
M
,
Sebag
M
, et al
Preliminary safety, efficacy, pharmacokinetics, and pharmacodynamics of subcutaneously (SC) administered PF-06863135, a B-cell maturation antigen (BCMA)-CD3 bispecific antibody, in patients with relapsed/refractory multiple myeloma (RRMM)
.
Blood
2020
;
136
:
8
9
.
77.
Lee
DW
,
Kochenderfer
JN
,
Stetler-Stevenson
M
,
Cui
YK
,
Delbrook
C
,
Feldman
SA
, et al
T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial
.
Lancet
2015
;
385
:
517
28
.
78.
Maude
SL
,
Teachey
DT
,
Porter
DL
,
Grupp
SA
. 
CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia
.
Blood
2015
;
125
:
4017
23
.
79.
Susanibar Adaniya
SP
,
Cohen
AD
,
Garfall
AL
. 
Chimeric antigen receptor T cell immunotherapy for multiple myeloma: a review of current data and potential clinical applications
.
Am J Hematol
2019
;
94
:
S28
33
.
80.
Schütt
P
,
Brandhorst
D
,
Stellberg
W
,
Poser
M
,
Ebeling
P
,
Müller
S
, et al
Immune parameters in multiple myeloma patients: influence of treatment and correlation with opportunistic infections
.
Leuk Lymphoma
2006
;
47
:
1570
82
.
81.
Van Oekelen
O
,
Gleason
CR
,
Agte
S
,
Srivastava
K
,
Beach
KF
,
Aleman
A
, et al
Highly variable SARS-CoV-2 spike antibody responses to two doses of COVID-19 RNA vaccination in patients with multiple myeloma
.
Cancer Cell
2021
;
39
:
1028
30
.
82.
Siegel
DS
,
Martin
T
,
Wang
M
,
Vij
R
,
Jakubowiak
AJ
,
Lonial
S
, et al
A phase 2 study of single-agent carfilzomib (PX-171-003-A1) in patients with relapsed and refractory multiple myeloma
.
Blood
2012
;
120
:
2817
25
.
83.
Richardson
PG
,
Siegel
DS
,
Vij
R
,
Hofmeister
CC
,
Baz
R
,
Jagannath
S
, et al
Pomalidomide alone or in combination with low-dose dexamethasone in relapsed and refractory multiple myeloma: a randomized phase 2 study
.
Blood
2014
;
123
:
1826
32
.
84.
Lonial
S
,
Weiss
BM
,
Usmani
SZ
,
Singhal
S
,
Chari
A
,
Bahlis
NJ
, et al
Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): an open-label, randomised, phase 2 trial
.
Lancet
2016
;
387
:
1551
60
.
85.
Chari
A
,
Vogl
DT
,
Gavriatopoulou
M
,
Nooka
AK
,
Yee
AJ
,
Huff
CA
, et al
Oral selinexor–dexamethasone for triple-class refractory multiple myeloma
.
N Engl J Med
2019
;
381
:
727
38
.
86.
Lonial
S
,
Lee
HC
,
Badros
A
,
Trudel
S
,
Nooka
AK
,
Chari
A
, et al
Belantamab mafodotin for relapsed or refractory multiple myeloma (DREAMM-2): a two-arm, randomised, open-label, phase 2 study
.
Lancet Oncol
2020
;
21
:
207
21
.
87.
Richardson
PG
,
Oriol
A
,
Larocca
A
,
Bladé
J
,
Cavo
M
,
Rodriguez-Otero
P
, et al
Melflufen and dexamethasone in heavily pretreated relapsed and refractory multiple myeloma
.
J Clin Oncol
2021
;
39
:
757
67
.
88.
Corre
J
,
Munshi
N
,
Avet-Loiseau
H
. 
Genetics of multiple myeloma: another heterogeneity level?
Blood
2015
;
125
:
1870
6
.
89.
Dimopoulos
M
,
Quach
H
,
Mateos
MV
,
Landgren
O
,
Leleu
X
,
Siegel
D
, et al
Carfilzomib, dexamethasone, and daratumumab versus carfilzomib and dexamethasone for patients with relapsed or refractory multiple myeloma (CANDOR): results from a randomised, multicentre, open-label, phase 3 study
.
Lancet
2020
;
396
:
186
97
.
90.
Dimopoulos
MA
,
Dytfeld
D
,
Grosicki
S
,
Moreau
P
,
Takezako
N
,
Hori
M
, et al
Elotuzumab plus pomalidomide and dexamethasone for multiple myeloma
.
N Engl J Med
2018
;
379
:
1811
22
.
91.
Moreau
P
,
Masszi
T
,
Grzasko
N
,
Bahlis
NJ
,
Hansson
M
,
Pour
L
, et al
Oral ixazomib, lenalidomide, and dexamethasone for multiple myeloma
.
N Engl J Med
2016
;
374
:
1621
34
.
92.
Lonial
S
,
Dimopoulos
M
,
Palumbo
A
,
White
D
,
Grosicki
S
,
Spicka
I
, et al
Elotuzumab therapy for relapsed or refractory multiple myeloma
.
N Engl J Med
2015
;
373
:
621
31
.
93.
Stewart
AK
,
Rajkumar
SV
,
Dimopoulos
MA
,
Masszi
T
,
Špička
I
,
Oriol
A
, et al
Carfilzomib, lenalidomide, and dexamethasone for relapsed multiple myeloma
.
N Engl J Med
2015
;
372
:
142
52
.
94.
Richardson
PG
,
Oriol
A
,
Beksac
M
,
Liberati
AM
,
Galli
M
,
Schjesvold
F
, et al
Pomalidomide, bortezomib, and dexamethasone for patients with relapsed or refractory multiple myeloma previously treated with lenalidomide (OPTIMISMM): a randomised, open-label, phase 3 trial
.
Lancet Oncol
2019
;
20
:
781
94
.
95.
San-Miguel
JF
,
Hungria
VT
,
Yoon
S-S
,
Beksac
M
,
Dimopoulos
MA
,
Elghandour
A
, et al
Panobinostat plus bortezomib and dexamethasone versus placebo plus bortezomib and dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma: a multicentre, randomised, double-blind phase 3 trial
.
Lancet Oncol
2014
;
15
:
1195
206
.
96.
Grosicki
S
,
Simonova
M
,
Spicka
I
,
Pour
L
,
Kriachok
I
,
Gavriatopoulou
M
, et al
Once-per-week selinexor, bortezomib, and dexamethasone versus twice-per-week bortezomib and dexamethasone in patients with multiple myeloma (BOSTON): a randomised, open-label, phase 3 trial
.
Lancet
2020
;
396
:
1563
73
.
97.
Yong
K
,
Delforge
M
,
Driessen
C
,
Fink
L
,
Flinois
A
,
Gonzalez-McQuire
S
, et al
Multiple myeloma: patient outcomes in real-world practice
.
Br J Haematol
2016
;
175
:
252
64
.
98.
Zelle-Rieser
C
,
Thangavadivel
S
,
Biedermann
R
,
Brunner
A
,
Stoitzner
P
,
Willenbacher
E
, et al
T cells in multiple myeloma display features of exhaustion and senescence at the tumor site
.
J Hematol Oncol
2016
;
9
:
1
12
.
99.
Chung
DJ
,
Pronschinske
KB
,
Shyer
JA
,
Sharma
S
,
Leung
S
,
Curran
SA
, et al
T-cell exhaustion in multiple myeloma relapse after autotransplant: optimal timing of immunotherapy
.
Cancer Immunol Res
2016
;
4
:
61
71
.