Bispecific antibodies are monoclonal antibodies targeting both a surface molecule on the malignant plasma cells and CD3 on T cells, leading to tumor cell death by activated T cells. Bispecific antibodies targeting B-cell maturation antigen, GPRC5D or FcRH5, demonstrated promising efficacy with favorable safety profile in patients with triple-class refractory multiple myeloma. This novel immunotherapeutic modality will likely change the treatment paradigm in the coming years.

The outcome of patients with relapsed/refractory multiple myeloma (RRMM) remains challenging. Patients with triple-class exposed RRRM (prior exposure to immunomodulatory drugs, proteasome inhibitor, and anti-CD38 monoclonal antibodies) have a median overall survival (OS) of nearly 1 year.1 Effective novel therapies are therefore needed. The most promising developments include novel immunotherapeutic approaches such as chimeric antigen receptor (CAR) T-cell therapy and bispecific antibodies (BispAbs).2,3 The CAR T-cell therapy idecabtagene vicleucel (ide-cel) has been approved for triple-class exposed patients with MM refractory to their last therapy.4 Across all target doses, the median progression-free survival (PFS) was 8.6 months, and the median OS was 24.8 months.5 Ciltacabtagene autoleucel (cilta-cel), a CAR T-cell therapy with 2 B-cell maturation antigen (BCMA)–targeting single-domain antibodies showing a 97.9% response rate and 18-month PFS and OS rates of 66.0% and 80.9%, respectively,6 is also approved for triple-class–exposed patients. One important challenge with CAR T-cell therapy is the lengthy (6-8 weeks) and individualized manufacturing process, which might not be feasible for patients with aggressive disease course.4,5 On the opposite side, BispAbs are readily available off-the-shelf products. Ongoing phase 1/2 clinical trials using different constructs, with different targets on myeloma cells, are showing a favorable safety profile with high response rates in heavily pretreated patients.

A BispAb is able to bind to an antigen on the tumor cell and to another antigen on T-cell lymphocyte to redirect these immune cells toward malignant cells.2,7-18 This immunologic synapse formation is followed by T-cell activation and degranulation and the release of granzymes and perforins, leading to tumor cell lysis. BispAb constructs promote sustained T-cell activation, leading to polyclonal expansion of memory T cells.10 BispAbs act independently of major histocompatibility complex or T-cell receptor specificity.2,7-18 Different formats of bispecific T-cell–redirecting antibodies have been developed. Bispecific T-cell engager (BiTE) consists of a single-chain fragment variable for binding the target antigen on the malignant cell and another single-chain fragment variable for binding a T cell, the 2 being connected by a linker peptide.2,7-18 BispAbs lacking an Fc region more easily penetrate tumors due to their small size but are associated with short half-life requiring frequent or continuous infusion.7-18 BispAbs with Fc domain (full size) creating an IgG-like molecule have an extended half-life, allowing intermittent dosing, Fc-mediated effector functions, and potential subcutaneous administration.7-18 Additionally, it is possible to build IgG-like TrispAbs with additional binding sites targeting T-cell and 2 distinct plasma cell antigens or a single myeloma antigen and 2 distinct antigens of the immune effector cell.19-21 

The optimal target is an antigen constantly expressed by myeloma cells with minimal expression by healthy tissues to minimize toxicity. Numerous potential myeloma cell targets are under evaluation for clinical development of BispAbs, such as CD38, CD138, CD200, and SLAMF7, but as of the writing of this article, clinical data are available for BCMA, GPRC5D, and FcRH5.2,7-9,14,18,22

BCMA, or CD269, is currently the major target for BispAbs, with at least 8 different compounds in preclinical/clinical development to date.2,7-9,14,18,22 BCMA, a type III transmembrane glycoprotein belonging to the tumor necrosis factor receptor superfamily, regulates B-cell proliferation, maturation, survival, and differentiation to plasma cells.23 BCMA is expressed at high levels on both malignant and normal plasma cells yet remains undetectable in hematopoietic stem cells and most nonhematologic tissues.23-25 GPRC5D is a transmembrane orphan receptor of the G protein-coupled receptor family class C group 5 member D whose functions are poorly characterized.26-29 It is highly expressed on myeloma cells (no expression on other hematopoietic cells), as well as keratinized structures, including hair shaft, nail, and central region of the tongue.26-29 GPRC5D overexpression has been correlated with adverse prognosis in MM.30 Of note, plasma cells of patients with MM exhibit differential expression of both BCMA and GPRC5D, consistent with data suggesting that BCMA and GPRC5D may be independently regulated in MM cells.31 GPRC5D represents a novel and attractive target for MM, and 2 BispAbs are currently being evaluated in preclinical27 or clinical studies.28 

The Fc receptor-homolog 5 (FcRH5), also designated as CD307, FcRL5, and IRTA2, is a membrane protein from the immunoglobulin superfamily implicated in proliferation and isotype expression in the development of antigen-primed B cells.32 Its expression is restricted to the B-cell lineage with increased expression in mature B cells and plasma cells.33 FcRH5 expression has also been shown to be higher in myeloma cells in comparison with normal plasma cells.32,33 Interestingly, the FcRH5 gene is located in the chromosome region 1q21.34 Some data suggest that the 1q21 gain can lead to FcRH5 overexpression in patients with high-risk MM.34-36 Therefore, FcRH5 is an interesting target for the BispAb cevostamab.2,3,7-15,18,22

Only 2 phase 1 trials have been reported in peer-reviewed publications to date, the first investigating anti-BCMA BiTE molecule AMG 42037 (whose development has been discontinued due to inadequate PK), and the second investigating teclistamab, a BCMA × CD3 T-cell–redirecting BispAb, in heavily pretreated patients with RRMM.38 However, data from 8 early phase trials have been already reported in conference proceedings that have enrolled 485 patients treated with BCMA-BispAb,39-44 95 treated with GPRC5D-BispAb,45 and 160 treated with FcRH5-BispAB,46 respectively.

The most common adverse events (AEs), similar across trials, are summarized (Table 1) from 740 heavily pretreated patients with a median age of 64 years.39-46 

Table 1.

Safety and efficacy of BispAbs

CC-9326938 
n = 30
AMG70139 
N = 75
Teclistamab39 
n = 159
Elranatamab40 
n = 50
RENG545841 
n = 68
Tnb-383B42 
n = 103
Talquetamab43 
n = 95
Cevostamab44 
n = 160
Target BCMA BCMA BCMA BCMA BCMA BCMA GPRC5D FcRH5 
Phase 1/2 1/2 
Administration IV* IV weekly SC weekly SC Q2W IV weekly, then Q2W IV Q3W SC weekly or Q2W IV Q3W 
Median number of prior regimens 5 (3-13) 6 (1-25) 5 (2-15) 5 (2-17) 5 (1-15) NR 6 (2-18) 
Median age, y 64 (42-78) 63 64 (33-84) 64 (32-86) 64 (41-81) 67 (35-88) 61 (46-84) 64 (33-82) 
Triple refractory (%) NR 68 77 98 100 62 81 85 
RP2D Not reached Not reported 1.5 mg/kg weekly 1 mg/kg NR 60 mg Q3W 405 μg/kg weekly or 800 μg/kg Q2W NR 
CRS, grade ≥3 (%) 77, 4 61, 7 67, 1 83, 0 38, 0 52, 3 73, 3 at 405 μg/kg weekly
78, 0 at 800 μg/kg Q2W 
80, 1 
Median duration of CRS (d) NR 2 (1-9) NR NR NR 
Median time to onset of CRS (d) NR NR 2 (1-6) NR NR NR 
Neurotoxicity, grade ≥3 (%) NR 8, 0 2.5, 0 NR NR, 0 NR NR 13.1, 3.8 
Neutropenia, grade ≥3 (%) NR, 43 23, NR 53, 45 64, 60 16, 13 17, NR 67, 60 at 405 μg/kg weekly
44, 35 at 800 μg/kg Q2W 
15, 13.8 
Anemia, grade ≥3 (%) NR, 37 43, NR 41, 27 55, 38 NR 9, NR NR 31.9, 21.9 
Thrombocytopenia, grade ≥3 (%) NR, 17 20, NR 33, 18 52, 31 NR 14, NR NR NR 
Infections, grade ≥3 (%) NR, 30 NR, 17 45, 22.537  NR NR 28, NR 37, NR at 405 μg/kg weekly
13, NR at 800 μg/kg Q2W 
42.5, 18.8 
ORR across all dose levels (%) 43 23.6 NR NR NR NR NR NR 
ORR at RP2D or efficacious dose range (%) 89
(9/10 at 10 mg) 
83
(5/6 at 9 mg) 
65 70 73.3 at 96 mg + 200 mg dose levels 64 for cohorts ≥ 40 mg 70 at 405 μg/kg weekly
71 at 800 μg/kg Q2W 
36.7 at 90 mg
54.5 at 160 mg 
Median time to response (d) 29 29 29 22 NR NR NR 29 
DOR 5.3 to 40.6 wk 3.8 mo (1.9, 7.4) 6 mo: 90% 95% CI at 6 mo: 92.3% (56.7-98.9) DOR ≥ 8 mo: 92.1% NR 6 mo: 67% at 405 μg/kg weekly Median: 15.6 mo (95% CI: 6.4-21.6) 
CC-9326938 
n = 30
AMG70139 
N = 75
Teclistamab39 
n = 159
Elranatamab40 
n = 50
RENG545841 
n = 68
Tnb-383B42 
n = 103
Talquetamab43 
n = 95
Cevostamab44 
n = 160
Target BCMA BCMA BCMA BCMA BCMA BCMA GPRC5D FcRH5 
Phase 1/2 1/2 
Administration IV* IV weekly SC weekly SC Q2W IV weekly, then Q2W IV Q3W SC weekly or Q2W IV Q3W 
Median number of prior regimens 5 (3-13) 6 (1-25) 5 (2-15) 5 (2-17) 5 (1-15) NR 6 (2-18) 
Median age, y 64 (42-78) 63 64 (33-84) 64 (32-86) 64 (41-81) 67 (35-88) 61 (46-84) 64 (33-82) 
Triple refractory (%) NR 68 77 98 100 62 81 85 
RP2D Not reached Not reported 1.5 mg/kg weekly 1 mg/kg NR 60 mg Q3W 405 μg/kg weekly or 800 μg/kg Q2W NR 
CRS, grade ≥3 (%) 77, 4 61, 7 67, 1 83, 0 38, 0 52, 3 73, 3 at 405 μg/kg weekly
78, 0 at 800 μg/kg Q2W 
80, 1 
Median duration of CRS (d) NR 2 (1-9) NR NR NR 
Median time to onset of CRS (d) NR NR 2 (1-6) NR NR NR 
Neurotoxicity, grade ≥3 (%) NR 8, 0 2.5, 0 NR NR, 0 NR NR 13.1, 3.8 
Neutropenia, grade ≥3 (%) NR, 43 23, NR 53, 45 64, 60 16, 13 17, NR 67, 60 at 405 μg/kg weekly
44, 35 at 800 μg/kg Q2W 
15, 13.8 
Anemia, grade ≥3 (%) NR, 37 43, NR 41, 27 55, 38 NR 9, NR NR 31.9, 21.9 
Thrombocytopenia, grade ≥3 (%) NR, 17 20, NR 33, 18 52, 31 NR 14, NR NR NR 
Infections, grade ≥3 (%) NR, 30 NR, 17 45, 22.537  NR NR 28, NR 37, NR at 405 μg/kg weekly
13, NR at 800 μg/kg Q2W 
42.5, 18.8 
ORR across all dose levels (%) 43 23.6 NR NR NR NR NR NR 
ORR at RP2D or efficacious dose range (%) 89
(9/10 at 10 mg) 
83
(5/6 at 9 mg) 
65 70 73.3 at 96 mg + 200 mg dose levels 64 for cohorts ≥ 40 mg 70 at 405 μg/kg weekly
71 at 800 μg/kg Q2W 
36.7 at 90 mg
54.5 at 160 mg 
Median time to response (d) 29 29 29 22 NR NR NR 29 
DOR 5.3 to 40.6 wk 3.8 mo (1.9, 7.4) 6 mo: 90% 95% CI at 6 mo: 92.3% (56.7-98.9) DOR ≥ 8 mo: 92.1% NR 6 mo: 67% at 405 μg/kg weekly Median: 15.6 mo (95% CI: 6.4-21.6) 

DOR, duration of response; NR, not reported; ORR, overall response rate; Q2W, every 2 weeks; Q3W, every 3 weeks; RP2D, recommended phase 2 dose; SC, subcutaneous.

*

On days D1, 8, 15, and 22 for cycles (C) 1-3; D1 and 15 for C4-6; and on D1 for C7 and beyond, all in 28-day cycles for up to 2 years.

Cytokine release syndrome

Cytokine release syndrome (CRS) is an acute systemic inflammatory syndrome characterized by the activation of T cells and other immune effector cells leading to significant cytokine release, whose severity is related to disease burden and dose of immunotherapy.47 Step-up dosing with progressively increasing doses administered during the week prior to first full dose was used in most studies to mitigate CRS intensity. Incidence of CRS ranged from 38% to 83%, including only 0% to 4% grade ≥3 CRS. Across all studies, the use of tocilizumab (which should be considered as first treatment) and corticosteroids (which should be added in case of limited improvement following a repeated dose of anti–interleukin-6 therapy) to treat CRS was approximately 40% and 20%, respectively. Median time to CRS ranged from 12 to 48 hours, with earlier event (24 hours) with intravenously administered BispAbsin compared with subcutaneously administered (2 days) BispAbs. The median CRS duration ranged from 12 to 48 hours. CRS events are generally restricted to the first cycle, and all patients but one39 recovered with no sequelae.

Neurotoxicity

Any-grade neurotoxicity ranged from 2% to 13%, including 0% to 4% grade ≥3 events. Symptoms included headache, confusion, aphasia, cognitive disorder, and encephalopathy. Neurologic symptoms were commonly concomitant of CRS and fully resolved after CRS therapy.

Hematologic effects

Hematologic side effects across all trials included anemia (any grade 9% to 41%; grade ≥3 21.9% to 38%), neutropenia (any grade 15% to 67%; grade ≥3 13% to 60%), and thrombocytopenia (any grade 14% to 33%; grade ≥3 17% to 31%). The origin of cytopenias is not yet well understood considering the mechanism of action of BispAb. Therefore, 1 hypothesis involves a “bystander” effect from the cytokine release and destruction of plasma cells in the bone marrow.

Infections

Infection was observed in 13% to 52% of patients, with grade ≥3 in 18% to 30%.38-46 In this heavily pretreated population with severe immunosuppression and profound hypogammaglobulinemia, the source, type, and time to onset of infection were not widely reported. Nevertheless, a recent series on 36 patients treated with BispAbs suggests a higher rate of infections than CAR-T due to continuous therapy.48 An important issue in the COVID-19 era is the lack of response to vaccines. A recent study from the Mount Sinai NY group showed, in a small number of cases, that patients receiving BCMA-targeted therapy, including BispAbs, had impaired T-cell response and lower severe acute respiratory syndrome coronavirus 2 spike-binding IgG antibody levels after 2 doses of vaccine compared with patients receiving other antimyeloma therapy.49 More data with longer follow-up are needed before making appropriate recommendations for screening, monitoring, and prophylaxis of infections in patients receiving BispAbs.50 

Other specific toxicity

The anti-GPRC5D talquetamab antibody was unique in causing dysgeusia (60%; grade 2: 29%), skin-related AEs (77%; all grade 1/2) and nail disorders (30%)45 in conjunction with the GPRC5D expression on hard keratinized structures. The dermatologic and oral events are mostly low grade and rarely require dose modifications. AEs have been manageable with early and consistent supportive care.51 

Although safety and dose identification are the primary objectives of these phase I trials,38-46 preliminary efficacy data are available for the most active dose cohorts (Table 1). Responses were rapid, with a median time to first response of 4 weeks. For these heavily pretreated (median number of prior therapies: 6; triple-class refractory: 81%) patients treated across the efficacious dose range, the overall response rates ranged from 36.7% to 89%, with a dose-dependent increase in clinical efficacy. Minimal disease negativity data were immature. Because the median follow-up periods from these early phase studies are still short, the median duration of response has not been reached. Although efficacy data need to be confirmed in larger numbers of patients, these preliminary response rates compare favorably with recently approved drugs in MM for triple-class exposed/refractory patients, such as selinexor52 or belantamab-mafodotin.53 No data are available yet in specific populations of interest (ie, elderly/frail patients and patients with high-risk cytogenetics, renal failure, or extramedullary disease).

Resistance may be related to tumor-related features, T-cell characteristics, and immunosuppressive microenvironment. T-cell exhaustion is a feature of MM that may be potentialized by BispAbs therapy.36,54,55 Dysregulation of regulatory T cells, tumor-associated macrophages, and myeloid-derived suppressor cells have also been described as resistance mechanisms.8,9,56-58 Recently, a case of a biallelic BCMA gene deletion was described as a cause of acquired-resistance to a BCMA-specific BispAb.59 Furthermore, by using whole-genome sequencing data, the authors demonstrated that genomic alterations in genes encoding immunotherapy targets were preexisting in some patients before immunotherapy and could be used as biomarker of response.59 

Two strategies are under evaluation to improve efficacy further on: combination therapies and earlier use of BispAbs (Table 2). There is a strong rationale for combining BispAbs with CD38 antibodies. Daratumumab impacts immune cell populations (ie, increasing helper and cytotoxic T cells and decreasing suppressive CD38+ immunoregulatory cells).60 Preclinical studies showed that addition of daratumumab enhanced teclistamab56- and talquetamab61-mediated lysis of MM cells, suggesting the combination may also increase clinical activity in patients with RRMM. Preliminary data from the phase 1b multicohort study TRIMM-2 (combination of talquetamab62 or teclistamab63 with daratumumab) have been recently reported and showed that the 2 combinations were well tolerated and demonstrated promising efficacy in patients with RRMM. Because of their ability to promote T-cell activity, there is also a strong rationale to combine BispAbs with immunomodulatory drugs. Due to the high mutational burden of MM and the multiclonal nature of the tumor, a logical approach could be to combine BispAbs targeting different antigens. An ongoing study (#NCT04586426) is already evaluating teclistamab plus talquetamab in advanced patients. T-cell exhaustion as a potential mechanism favoring progression in MM supports the use of BispAbs earlier in the course of the disease.64,65 Moreover, the frequency of deletions and mutations in genes encoding immunotherapy targets is higher in patients with very advanced disease.59 Ongoing trials are already testing BispAbs in combination with pomalidomide or carfilzomib at earlier lines of therapy, and other trials are planning to use these agents as part of frontline therapy.

Table 2.

Ongoing studies evaluating BispAbs in multiple myeloma

StudyPopulationPhaseTreatment
CD3 × BCMA    
 NCT04557098 (MajesTEC-1) RRMM 1-2 Teclistamab 
 NCT04722146 (MajesTEC-2) RRMM and NDMM 1b Teclistamab in combination with
  • - dara pom

  • - dara len

  • - dara btz len

  • - len

  • - nirogacestat

 
 NCT05083169 (MajesTEC-3) RRMM 3 randomized Teclistamab dara
vs DPd/DVd 
 NCT04108195 (TRIMM-2) RRMM 1b Teclistamab in combination with
  • - dara

  • - dara pom

 
 NCT04586426 RRMM 1b Teclistamab in combination with
  • - talquetamab

  • - talquetamab dara

 
 NCT05243797 (MajesTEC-4) NDMM
(maintenance) 
3 randomized Teclistamab len
vs len 
 NCT05231629 (Master-2) NDMM 3 randomized Teclistamab dara 
 NCT03269136
 (MAGNETISMM-1) 
RRMM Elranatamab 
 NCT04649359  (MAGNETISMM-3) RRMM Elranatamab 
 NCT05090566
 (MAGNETISMM-4) 
RRMM 1b-2 Elranatamab + nirogacestat 
 NCT05020236  (MAGNETISMM-5) RRMM Elranatamab dara
vs DPd 
 NCT05137054 RRMM 1b REGN5458 in combination with
  • - carfilzomib

  • - len dex

  • - btz dex

  • - dara dex

 
 NCT03933735 RRMM TNB-383B 
 NCT04184050 RRMM 1-2 HPN217 (trispecific BCMA × CD3 × albumin) 
 NCT04735575 RRMM 1-2 EMB-06 
GPRC5D × CD3    
 NCT03399799  (MONUMENTAL-1) RRMM Talquetamab 
 TRIMM-2
 NCT04108195 
RRMM 1b Talquetamab dara Talquetamab dara pom 
 NCT05050097
 (MONUMENTAL-2) 
RRMM 1b Talquetamab in combination with
  • - carfilzomib

  • - carfilzomib dara

  • - len

  • - len dara

  • - pom

 
FCRH5 × CD3    
 NCT03275103 (GRACE) RRMM Cevostamab 
CD38 × CD3    
 NCT03309111 RRMM ISB 1342 
 NCT05011097 RRMM Y150 
StudyPopulationPhaseTreatment
CD3 × BCMA    
 NCT04557098 (MajesTEC-1) RRMM 1-2 Teclistamab 
 NCT04722146 (MajesTEC-2) RRMM and NDMM 1b Teclistamab in combination with
  • - dara pom

  • - dara len

  • - dara btz len

  • - len

  • - nirogacestat

 
 NCT05083169 (MajesTEC-3) RRMM 3 randomized Teclistamab dara
vs DPd/DVd 
 NCT04108195 (TRIMM-2) RRMM 1b Teclistamab in combination with
  • - dara

  • - dara pom

 
 NCT04586426 RRMM 1b Teclistamab in combination with
  • - talquetamab

  • - talquetamab dara

 
 NCT05243797 (MajesTEC-4) NDMM
(maintenance) 
3 randomized Teclistamab len
vs len 
 NCT05231629 (Master-2) NDMM 3 randomized Teclistamab dara 
 NCT03269136
 (MAGNETISMM-1) 
RRMM Elranatamab 
 NCT04649359  (MAGNETISMM-3) RRMM Elranatamab 
 NCT05090566
 (MAGNETISMM-4) 
RRMM 1b-2 Elranatamab + nirogacestat 
 NCT05020236  (MAGNETISMM-5) RRMM Elranatamab dara
vs DPd 
 NCT05137054 RRMM 1b REGN5458 in combination with
  • - carfilzomib

  • - len dex

  • - btz dex

  • - dara dex

 
 NCT03933735 RRMM TNB-383B 
 NCT04184050 RRMM 1-2 HPN217 (trispecific BCMA × CD3 × albumin) 
 NCT04735575 RRMM 1-2 EMB-06 
GPRC5D × CD3    
 NCT03399799  (MONUMENTAL-1) RRMM Talquetamab 
 TRIMM-2
 NCT04108195 
RRMM 1b Talquetamab dara Talquetamab dara pom 
 NCT05050097
 (MONUMENTAL-2) 
RRMM 1b Talquetamab in combination with
  • - carfilzomib

  • - carfilzomib dara

  • - len

  • - len dara

  • - pom

 
FCRH5 × CD3    
 NCT03275103 (GRACE) RRMM Cevostamab 
CD38 × CD3    
 NCT03309111 RRMM ISB 1342 
 NCT05011097 RRMM Y150 

btz, bortezomib; dara, daratumumab; DPd, daratumumab-pomalidomide-dexamethasone; DVd, daratumumab-bortezomib-dexamethasone; len, lenalidomide; NDMM, newly diagnosed multiple myeloma; pom, pomalidomide.

BispAbs represent a promising new class of agents for MM and will probably become a standard of care in the near future. Their safety profile is favorable, allowing ambulatory use. Additional data are needed to further evaluate long-term toxicities and the risk of infection more specifically. BispAbs are off-the-shelf products and allow rapid responses, which is crucial for patients with rapidly progressive disease. More data for PFS and OS are requested, but their efficacy is highly promising. With the recent approval of BCMA-directed CAR-T therapy in MM, many issues are arising about the use of BispAbs versus CAR-T. The choice between the two modalities depends on various practical considerations: efficacy, disease status, age, comorbidities, product availability, distance from an academic center, and optimal sequencing (prior exposure to BCMA therapy). Finally, BispAbs might be easily incorporated with currently approved myeloma therapies in earlier lines of treatment to increase efficacy as we continue the quest for cure.

Contribution: P.M. and C.T. wrote the manuscript.

Conflict-of-interest disclosure: P.M. received honoraria and is on the advisory board for Celgene, Amgen, Janssen, AbbVie, and Sanofi. C.T. received honoraria and is on the advisory board for Celgene, Amgen, Janssen, AbbVie, and Sanofi.

Correspondence: Philippe Moreau, Department of Clinical Hematology, University Hospital Hotel-Dieu, Place Ricordeau, 44093 Nantes, France; e-mail: philippe.moreau@chu-nantes.fr.

1.
Mateos
MV
,
Weisel
K
,
De Stefano
V
, et al
.
LocoMMotion: a prospective, non-interventional, multinational study of real-life current standards of care in patients with relapsed and/or refractory multiple myeloma
.
Leukemia.
2022
;
36
:
1371
-
1376
.
2.
Lakshman
A
,
Kumar
SK
.
Chimeric antigen receptor T-cells, bispecific antibodies, and antibody-drug conjugates for multiple myeloma: an update
.
Am J Hematol.
2022
;
97
(
1
):
99
-
118
.
3.
Rasche
L
,
Wäsch
R
,
Munder
M
,
Goldschmidt
H
,
Raab
MS
.
Novel immunotherapies in multiple myeloma: chances and challenges
.
Haematologica.
2021
;
106
(
10
):
2555
-
2565
.
4.
Munshi
NC
,
Anderson
LD
Jr
,
Shah
N
, et al
.
Idecabtagene icleucel in relapsed and refractory multiple myeloma
.
N Engl J Med.
2021
;
384
(
8
):
705
-
716
.
5.
Anderson
LD
,
Munshi
NC
,
Shah
N
, et al
.
Idecabtagene vicleucel (ide-cel; bb2121), a BCMA-directed CAR T cell therapy, in relapsed and refractory multiple myeloma: updated KarMMa results
.
J Clin Oncol.
2021
;
39
(
suppl 15
):
8016
.
6.
Martin
T
,
Usmani
SZ
,
Berdeja
GJ
, et al
.
Updated results from CARTITUDE-1: phase 1b/2 study of ciltacabtagene autoleucel, a B-cell maturation antigen–directed chimeric antigen receptor T cell therapy, in patients with relapsed/refractory multiple myeloma [abstract]
.
Blood.
2021
;
138
(
suppl 1
). Abstract
549
.
7.
Lancman
G
,
Sastow
DL
,
Cho
HJ
, et al
.
Bispecific antibodies in multiple myeloma: present and future
.
Blood Cancer Discov.
2021
;
2
(
5
):
423
-
433
.
8.
Hosny
M
,
Verkleij
CPM
,
van der Schans
J
, et al
.
Current state of the art and prospects of T cell-redirecting bispecific antibodies in multiple myeloma
.
J Clin Med.
2021
;
10
(
19
):
4593
.
9.
Alhallak
K
,
Sun
J
,
Jeske
A
, et al
.
Bispecific T cell engagers for the treatment of multiple myeloma: achievements and challenges
.
Cancers (Basel).
2021
;
13
(
12
):
2853
.
10.
Cohen
AD
,
Raje
N
,
Fowler
JA
,
Mezzi
K
,
Scott
EC
,
Dhodapkar
MV
.
How to train your T cells: overcoming immune dysfunction in multiple myeloma
.
Clin Cancer Res.
2020
;
26
(
7
):
1541
-
1554
.
11.
Bargou
R
,
Leo
E
,
Zugmaier
G
, et al
.
Tumor regression in cancer patients by very low doses of a T cell-engaging antibody
.
Science.
2008
;
321
(
5891
):
974
-
977
.
12.
Velasquez
MP
,
Bonifant
CL
,
Gottschalk
S
.
Redirecting T cells to hematological malignancies with bispecific antibodies
.
Blood.
2018
;
131
(
1
):
30
-
38
.
13.
Fan
G
,
Wang
Z
,
Hao
M
,
Li
J
.
Bispecific antibodies and their applications
.
J Hematol Oncol.
2015
;
8
:
130
.
14.
Mohan
M
,
Maatman
TC
,
Schinke
C
.
The role of monoclonal antibodies in the era of bi-specifics antibodies and CAR T cell therapy in multiple myeloma
.
Cancers (Basel).
2021
;
13
(
19
):
4909
.
15.
Verkleij
CPM
,
Frerichs
KA
,
Broekmans
M
, et al
.
T-cell redirecting bispecific antibodies targeting BCMA for the treatment of multiple myeloma
.
Oncotarget.
2020
;
11
(
45
):
4076
-
4081
.
16.
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
-
117
.
17.
Saxena
A
,
Wu
D
.
Advances in therapeutic Fc engineering–modulation of IgG-associated effector functions and serum half-life
.
Front Immunol.
2016
;
7
:
580
.
18.
Caraccio
C
,
Krishna
S
,
Phillips
DJ
,
Schürch
CM
.
Bispecific antibodies for multiple myeloma: a review of targets, drugs, clinical trials, and future directions
.
Front Immunol.
2020
;
11
:
501
.
19.
Gantke
T
,
Weichel
M
,
Herbrecht
C
, et al
.
Trispecific antibodies for CD16A-directed NK cell engagement and dual-targeting of tumor cells
.
Protein Eng Des Sel.
2017
;
30
(
9
):
673
-
684
.
20.
Esensten
JH
,
Helou
YA
,
Chopra
G
,
Weiss
A
,
Bluestone
JA
.
CD28 costimulation: from mechanism to therapy
.
Immunity.
2016
;
44
(
5
):
973
-
988
.
21.
Wu
L
,
Seung
E
,
Xu
L
, et al
.
Trispecific antibodies enhance the therapeutic efficacy of tumor-directed T cells through T cell receptor co-stimulation
.
Nat Can.
2020
;
1
(
1
):
86
-
98
.
22.
Podar
K
,
Leleu
X
.
Relapsed/refractory multiple myeloma in 2020/2021 and beyond
.
Cancers.
2021
;
14
(
20
):
5154
.
23.
Madry
C
,
Laabi
Y
,
Callebaut
I
, 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
(
11
):
1693
-
1702
.
24.
Cho
SF
,
Anderson
KC
,
Tai
YT
.
Cell maturation antigen (BCMA) in multiple myeloma: potential uses of BCMA-based immunotherapy
.
Front Immunol.
2018
;
9
:
1821
.
25.
Eckhert
E
,
Hewitt
R
,
Liedtke
M
.
B-cell maturation antigen directed monoclonal antibody therapies for multiple myeloma
.
Immunotherapy.
2019
;
11
(
9
):
801
-
811
.
26.
Frigyesi
I
,
Adolfsson
J
,
Ali
M
, et al
.
Robust isolation of malignant plasma cells in multiple myeloma
.
Blood.
2014
;
123
(
9
):
1336
-
1340
.
27.
Kodama
T
,
Kochi
Y
,
Nakai
W
, et al
.
Anti-GPRC5D/CD3 bispecific T-cell-redirecting antibody for the treatment of multiple myeloma
.
Mol Cancer Ther.
2019
;
18
(
9
):
1555
-
1564
.
28.
Pillarisetti
K
,
Edavettal
S
,
Mendonça
M
, 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
(
15
):
1232
-
1243
.
29.
Inoue
S
,
Nambu
T
,
Shimomura
T
.
The RAIG family member, GPRC5D, is associated with hard-keratinized structures
.
J Invest Dermatol.
2004
;
122
(
3
):
565
-
573
.
30.
Atamaniuk
J
,
Gleiss
A
,
Porpaczy
E
, 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
(
9
):
953
-
960
.
31.
Smith
EL
,
Harrington
K
,
Staehr
M
, et al
.
GPRC5D is a target for the immunotherapy of multiple myeloma with rationally designed CAR T cells
.
Sci Transl Med.
2019
;
11
(
485
):
eaau7746
.
32.
Elkins
K
,
Zheng
B
,
Go
M
, et al
.
FcRL5 as a target of antibody-drug conjugates for the treatment of multiple myeloma
.
Mol Cancer Ther.
2012
;
11
(
10
):
2222
-
2232
.
33.
Polson
AG
,
Zheng
B
,
Elkins
K
, et al
.
Expression pattern of the human FcRH/IRTA receptors in normal tissue and in B-chronic lymphocytic leukemia
.
Int Immunol.
2006
;
18
(
9
):
1363
-
1373
.
34.
Sawyer
JR
,
Tricot
G
,
Mattox
S
,
Jagannath
S
,
Barlogie
B
.
Jumping translocations of chromosome 1q in multiple myeloma: evidence for a mechanism involving decondensation of pericentromeric heterochromatin
.
Blood.
1998
;
91
(
5
):
1732
-
1741
.
35.
Hatzivassiliou
G
,
Miller
I
,
Takizawa
J
, et al
.
IRTA1 and IRTA2, novel immunoglobulin superfamily receptors expressed in B cells and involved in chromosome 1q21 abnormalities in B cell malignancy
.
Immunity.
2001
;
14
(
3
):
277
-
289
.
36.
Li
J
,
Stagg
NJ
,
Johnston
J
, 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
(
3
):
383
-
395
.
37.
Topp
MS
,
Duell
J
,
Zugmaier
G
, et al
.
Anti-B-cell maturation antigen BiTE molecule AMG 420 induces responses in multiple myeloma
.
J Clin Oncol.
2020
;
38
(
8
):
775
-
783
.
38.
Usmani
SZ
,
Garfall
AL
,
van de Donk
NWCJ
, et al
.
Teclistamab, a B-cell maturation antigen × CD3 bispecific antibody, in patients with relapsed or refractory multiple myeloma (MajesTEC-1): a multicentre, open-label, single-arm, phase 1 study
.
Lancet.
2021
;
398
(
10301
):
665
-
674
.
39.
Costa
L
,
Wong
S
,
Bermudez
A
, et al
.
Interim results from the first phase 1 clinical study of the B-cell maturation antigen (BCMA) 2+ 1 T-cell engager CC-93269 in patients with relapsed/refractory multiple myeloma
.
EHA Library.
2020
;
295025
:
S205
.
40.
Harrison
SJ
,
Minnema
MC
,
Lee
HC
, et al
.
A phase 1 first in human study of AMG 701, an anti-B-cell maturation antigen (BCMA) half-life extended BiTE (bispecific T-cell engager) molecule, in relapsed/refractory multiple myeloma [abstract]
.
Blood.
2021
;
136
(
suppl 1
). Abstract
181
.
41.
Moreau
P
,
Usmani
SZ
,
Garfall
AL
, et al
.
Updated results from MajesTEC-1: phase 1/2 study of teclistamab, a B-cell maturation antigen x CD3 bispecific antibody, in relapsed/refractory multiple myeloma [abstract]
.
Blood.
2021
;
138
(
suppl 1
). Abstract
896
.
42.
Sebag
M
,
Raje
NS
,
Bahlis
NJ
, et al
.
Elranatamab (PF-06863135), a B-cell maturation antigen targeted CD3-engaging bispecific molecule, for patients with relapsed or refractory multiple myeloma: results from Magnetismm-1 [abstract]
.
Blood.
2021
;
138
(
suppl 1
). Abstract
895
.
43.
Zonder
JA
,
Richter
J
,
Bumma
N
, et al
.
Early, deep, and durable responses, and low rates of cytokine release syndrome with REGN5458, a BCMAxCD3 bispecific monoclonal antibody, in a Phase 1/2 first-in-human study in patients with relapsed/refractory multiple myeloma [abstract]
.
Blood.
2021
;
138
(
suppl 1
). Abstract
160
.
44.
Kumar
S
,
D’Souza
A
,
Shah
N
, et al
.
A phase 1 first-in-human study of Tnb-383B, a BCMA x CD3 bispecific T-cell redirecting antibody, in patients with relapsed/refractory multiple myeloma [abstract]
.
Blood.
2021
;
138
(
suppl 1
). Abstract
900
.
45.
Krishnan
AY
,
Minnema
MC
,
Berdeja
JG
, et al
.
Updated phase 1 results from MonumenTAL-1: first-in-human study of talquetamab, a G protein-coupled receptor family C group 5 member D x CD3 bispecific antibody, in patients with relapsed/refractory multiple myeloma [abstract]
.
Blood.
2021
;
138
(
suppl 1
). Abstract
158
.
46.
Trudel
S
,
Cohen
AD
,
Krishnan
AY
, et al
.
Cevostamab monotherapy continues to show clinically meaningful activity and manageable safety in patients with heavily pre-treated relapsed/refractory multiple myeloma: updated results from an ongoing phase I study [abstract]
.
Blood.
2021
;
138
(
suppl 1
). Abstract
157
.
47.
Lee
DW
,
Santomasso
BD
,
Locke
FL
, et al
.
ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells
.
Biol Blood Marrow Transplant.
2019
;
25
(
4
):
625
-
638
.
48.
Mohan
M
,
Nagavally
S
,
Dhakal
B
, et al
.
Risk of infections with B cell maturation antigen (BCMA) directed immunotherapy in multiple myeloma
.
Blood Adv.
2022;
6
(
8
):
2466
-
2470
.
49.
Van Oekelen
O
,
Gleason
CR
,
Agte
S
, et al;
PVI/Seronet team
.
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
(
8
):
1028
-
1030
.
50.
Los-Arcos
I
,
Iacoboni
G
,
Aguilar-Guisado
M
, et al
.
Recommendations for screening, monitoring, prevention, and prophylaxis of infections in adult and pediatric patients receiving CAR T-cell therapy: a position paper
.
Infection.
2021
;
49
(
2
):
215
-
231
.
51.
Mancia
SS
,
Farrell
A
,
Louw
K
, et al
.
Characterization and management of oral and dermatological toxicities in patients receiving the CD3 X GPRC5D bispecific antibody talquetamab (JNJ-64407564) for the treatment of relapsed and/or refractory multiple myeloma [abstract]
.
Blood.
2021
;
138
(
suppl 1
). Abstract
1658
.
52.
Chari
A
,
Vogl
DT
,
Gavriatopoulou
M
, et al
.
Oral selinexor-dexamethasone for triple-class refractory multiple myeloma
.
N Engl J Med.
2019
;
381
(
8
):
727
-
738
.
53.
Lonial
S
,
Lee
HC
,
Badros
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
(
2
):
207
-
221
.
54.
Suen
H
,
Brown
R
,
Yang
S
, et al
.
Multiple myeloma causes clonal T-cell immunosenescence: identification of potential novel targets for promoting tumour immunity and implications for checkpoint blockade
.
Leukemia.
2016
;
30
(
8
):
1716
-
1724
.
55.
Köhnke
T
,
Krupka
C
,
Tischer
J
,
Knösel
T
,
Subklewe
M
.
Increase of PD-L1 expressing B-precursor ALL cells in a patient resistant to the CD19/CD3-bispecific T cell engager antibody blinatumomab
.
J Hematol Oncol.
2015
;
8
:
111
.
56.
Frerichs
KA
,
Broekmans
MEC
,
Marin Soto
JA
, et al
.
Preclinical activity of JNJ-7957, a novel BCMAxCD3 bispecific antibody for the treatment of multiple myeloma, is potentiated by daratumumab
.
Clin Cancer Res.
2020
;
26
(
9
):
2203
-
2215
.
57.
Romano
A
,
Storti
P
,
Marchica
V
, et al
.
Mechanisms of action of the new antibodies in use in multiple myeloma
.
Front Oncol.
2021
;
8
:
684561
.
58.
Leone
P
,
Solimando
AG
,
Malerba
E
, et al
.
Actors on the scene: immune cells in the myeloma niche
.
Front Oncol.
2020
;
10
:
599098
.
59.
Truger
MS
,
Duell
J
,
Zhou
X
, et al
.
Single- and double-hit events in genes encoding immune targets before and after T cell-engaging antibody therapy in MM
.
Blood Adv.
2021
;
5
(
19
):
3794
-
3798
.
60.
van de Donk
NWCJ
,
Richardson
PG
,
Malavasi
F
.
CD38 antibodies in multiple myeloma: back to the future
.
Blood.
2018
;
131
(
1
):
13
-
29
.
61.
Verkleij
CPM
,
Broekmans
MEC
,
van Duin
M
, et al
.
Preclinical activity and determinants of response of the GPRC5DxCD3 bispecific antibody talquetamab in multiple myeloma
.
Blood Adv.
2021
;
5
(
8
):
2196
-
2215
.
62.
Chari
A
,
Hari
P
,
Bahlis
NJ
, et al
.
Phase 1b results for subcutaneous talquetamab plus daratumumab in patients with relapsed/refractory multiple myeloma [abstract]
.
Blood.
2021
;
138
(
suppl 1
). Abstract
161
.
63.
Rodriguez-Otero
P
,
Dholaria
B
,
Askari
E
, et al
.
Subcutaneous teclistamab in combination with daratumumab for the treatment of patients with relapsed/refractory multiple myeloma: results from a phase 1b multicohort study [abstract]
.
Blood.
2021
;
1647
(
suppl 1
). Abstract
161
.
64.
Zelle-Rieser
C
,
Thangavadivel
S
,
Biedermann
R
, et al
.
T cells in multiple myeloma display features of exhaustion and senescence at the tumor site
.
J Hematol Oncol.
2016
;
9
(
1
):
116
.
65.
Chung
DJ
,
Pronschinske
KB
,
Shyer
JA
, et al
.
T-cell exhaustion in multiple myeloma relapse after autotransplant: optimal timing of immunotherapy
.
Cancer Immunol Res.
2016
;
4
(
1
):
61
-
71
.
Sign in via your Institution