Download PDF
Review  |  Open Access  |  21 Mar 2023

Anti-BCMA novel therapies for multiple myeloma

Views: 1152 |  Downloads: 1013 |  Cited:   2
Cancer Drug Resist 2023;6:169-81.
10.20517/cdr.2022.138 |  © The Author(s) 2023.
Author Information
Article Notes
Cite This Article

Abstract

Recent advances in multiple myeloma therapy have increased the depth of response and ultimately survivals; however, the prognosis remains poor. The BCMA antigen is highly expressed in myeloma cells, thus representing a target for novel therapies. Several agents that target BCMA through different mechanisms, including bispecific T cell engagers drug conjugated to antibody and CAR-T cells, are now available or under development. Immunotherapies targeting BCMA have shown good results in efficacy and safety in multiple myeloma patients previously treated with several lines of therapy. This review will discuss the recent development of anti-BCMA targeted treatments in myeloma, with a special focus on currently available agents.

Keywords

Multiple myeloma, BCMA, belantamab, teclistamab, CART

INTRODUCTION

Multiple myeloma (MM) is a clonal plasma cell (PC) disorder accounting for 10% of hematologic neoplasms[1]. Novel therapies such as proteasome inhibitors (PI), immunomodulatory drugs (IMiDs), and anti-CD38 monoclonal antibodies (mAbs), together with autologous stem cell transplant (ASCT), have significantly improved treatment outcomes of newly diagnosed MM patients with a continuous increase of the overall survival (OS) that today reaches a median of 10 years[2-9]. However, MM patients still do relapse and MM is considered an incurable disease[10,11]. In particular, triple-class refractory (refractory to PI, IMiDs, and anti-CD38 antibody) and penta-refractory (first and second-generation PIs, two generations of IMiDs, anti-CD38 antibody) patients have a median OS of 5.6 months, especially in the presence of high-risk cytogenetics (HR)[12-20] or positive minimal residual disease[21-26]. Therefore, novel therapies, especially for relapsed/refractory myeloma patients (RRMM), are necessary[25-26]. BCMA is a B-cell maturation antigen highly expressed in myeloma cells, thus offering an encouraging potential target for novel treatments[27-28].

BCMA in multiple myeloma

BCMA, i.e., CD269 or TNFRSF17, is a TNF receptor superfamily 17 member, expressed on differentiated plasma cells and plasmablasts under physiological conditions and nearly on all MM tumor cells[29-31]. BCMA ligands include APRIL (A Proliferations-Inducing Ligand) and BAFF (B-cell activating factor) which are involved in the maturation and differentiation of PCs[32]. APRIL can bind to BCMA more avidly than to BAFF, and both can induce BCMA downstream signals to PI3K-PKB/Akt (i.e., phosphoinositide-3-kinase-protein kinase B/Akt), to RAS/MAPK (i.e., rat sarcoma/mitogen-activated protein kinase), and also to NF-κB (i.e., nuclear factor kappa-B), inducing increased plasma cells proliferation and survival[31,33-37]. Interestingly, PCs long-term survival in BCMA-/- mice is defective, suggesting BCMA is crucial for a sustained humoral immune response[38-39].

BCMA is overexpressed in myeloma PCs compared to normal ones, and its expression levels are elevated regardless of the stage of MGUS (monoclonal gammopathy of undetermined significance), SMM (smoldering multiple myeloma), and symptomatic MM[40-41]. Moreover, compared to healthy controls, APRIL and BAFF serum levels are 5-fold higher in myeloma patients. Recent studies showed that osteoclasts could be stimulated to produce more APRIL by MM cells, thus producing an immunosuppressive microenvironment[31,35,42] Interestingly, MM cell proliferation can be reduced, in a mouse xenograft model, by a moAb directed against APRIL. Anti-BCMA immunotherapies, together with APRIL inhibition, can defeat MM-induced immunosuppressive microenvironment and intensify the ADCC (antibody-dependent cell-mediated cytotoxicity) against myeloma cells[31,35,43].

sBCMA is the soluble form of BCMA, and it is produced by a γ-secretase acting on membrane BCMA[44]. sBCMA levels have been related to plasma cell infiltration in the bone marrow and may predict MM patients’ outcome. Indeed, some studies have shown that after MM treatment, the responsive patients resulted in lower sBCMA levels compared to patients with progressive disease[45-47]. Moreover, MGUS and SMM patients with higher levels of sBCMA showed a higher risk of progression to MM[48-49]. Thus, sBCMA might be used as a biomarker for disease progression and treatment response, allowing appropriate therapeutic management in case of drug resistance[10,50]. Additionally, one preliminary study in patients with non-secretory myeloma, for whom bone marrow aspirate and PET-CT scan are the only methods for disease monitoring, has shown that sBMCA levels correlate with the bone marrow PC infiltration, although this need to be confirmed[45-46]. Further studies are needed to validate sBCMA as a novel biomarker for MM and no approved diagnostic tool for measuring serum levels of sBCMA is available yet[10].

Finally, sBCMA reduces BCMA expression on PCs’ surface, thus resulting in reduced efficacy of BCMA-targeted therapies and MM cells’ immune escape[27]. Additionally, authors showed that sBCMA at high levels might interfere with anti-BCMA therapy, thus reducing effective binding to MM cells[51]. Preclinical studies of γ-secretase inhibitor (GSI) have shown that it may decrease sBCMA levels and increase MM cells expressing surface BCMA, thereby improving response to BCMA chimeric antigen receptor T cell (CAR-T) therapy. Hence, the association of a GSI and BCMA-targeting therapy in MM patients is being evaluated in early-phase clinical trials[2,52].

BCMA-TARGETED TREATMENT IN MULTIPLE MYELOMA

The evidence that BCMA could be a suitable target for effective antitumor activity in preclinical studies led to the development of drugs targeting BCMA with several mechanisms [Figure 1]. Presently, BCMA-targeted therapies available are represented by: antibody-drug conjugates (ADCs), bispecific T cell engager (BiTEs), and chimeric antigen receptor (CAR)-T cells [Table 1][53,54].

Anti-BCMA novel therapies for multiple myeloma

Figure 1. CAR: Chimeric antigen receptor.

Table 1

Characteristics of currently approved BCMA targeted agents

DrugMechanism of actionRegimen of administrationAdverse effectsORR/CR (%)*PFS (months)*
Belantamab
(ADCs)
Monoclonal antibody conjugated with a cytotoxic agentIntravenous (every 21 days)Corneal toxicities
Thrombocytopenia
31/32.8
Teclistamab (BITEs)Fully humanized IgG4 bispecific antibody redirecting, CD3-positive T-cells to BCMASubcutaneous (weekly)CRS
ICANS
Hematological toxicities
63/3911.3
Idecabtagene Vicleucel (CAR-T)BCMA targeted CAR-T incorporating anti-BCMA antibody costimulation domain, CD3ζ signaling domainSingle intravenous infusionCRS
ICANS
Hematological toxicities
73/338.8
Ciltacabtagene Autoleucel (CAR-T)BCMA-targeted CAR-T-cell product with two single anti-BCMA domain antibodies, CD3-ζ signaling domain costimulatory domainSingle intravenous infusionCRS
ICANS
Hematological toxicities
97/67Not reached

BCMA antibody drug conjugates

Antibody-drug conjugate (ADC) consists of a monoclonal antibody directed against a tumor- antigen and a cytotoxic agent inducing cell death (payload). ADC is internalized after binding to the related antigen on the tumor cell’s surface, then the linker is hydrolyzed inside of the lysosomes or endosomes and the payloads are released to cause cell death. ADCs can selectively target malignant cells with great efficiency on tumor cells and limited toxicities. Auristatin is a tubulin polymerase inhibitor used as a payload for MM[55-60].

Belantamab Mafodotin (GSK2857916)

Belantamab mafodotin (Bel) is a humanized IgG1 ADC, first-in-class, originally approved by the FDA (US Food and Drug Administration) as monotherapy in relapsed myeloma patients treated with four prior therapies including a proteasome inhibitor, anti-CD38 monoclonal antibody, and an immunomodulatory agent[61]. Bel is formed by an antibody directed to BCMA and covalently linked to MMAF (the microtubule inhibitor monomethyl auristatin F)[62]. After binding to BCMA on MM plasma cell, Bel is internalized and MMAF is released, provoking cell-cycle arrest and apoptosis[63]. Other effects that seem to be mediated by Bel-binding BCMA are ADCC and antibody-dependent cellular phagocytosis (ADCP)[64,65].

The multicenter phase I trial (DREAMM1) enrolled 73 RRMM patients. An ORR of 60% and PFS of 12 months were reported with acceptable toxicities. Corneal toxicity resulted in the most common non-hematologic side effect[66,67]. Subsequently, the phase II registrational study DREAMM2 enrolled 196 MM patients. The recommended dose was intravenous 2.5 mg/kg, Q3W. Reported ORR was 31%, with toxicities confirmed as manageable. A program was established to evaluate possible Keratopathy (Risk Evaluation and Mitigation Strategy, REMS) prior to drug administration[68-71]. Bel is currently being studied in different combination regimens in MM patients. The randomized, phase II study DREAMM4 is investigating Bel with pembrolizumab in patients with MM refractory to multiple lines of therapy. The DREAMM5 is testing Bel with other mAbs, such as isatuximab. The DREAMM-6 trial is exploring the combination of Bel, bortezomib, and dexamethasone vs. Bel, lenalidomide, and dexamethasone, while the DREAMM-7 and the DREAMM-8 studies are comparing Bel, bortezomib and dexamethasone vs. daratumumab, bortezomib and dexamethasone and Bel, pomalidomide and dexamethasone vs. pomalidomide, bortezomib and dexamethasone, respectively. Finally, the DREAMM-9 is testing Bel in the induction therapy in NDMM patients[2,10,72,73]. However, in November 2022, the FDA withdrew belantamab’s US marketing authorization as the DREAMM-3 trial (Bel vs. pomalidomide in combination with low-dose dexamethasone in RRMM) did not meet its primary endpoint of PFS (11.2 vs. 7 months, HR 1.03; 95%CI: 0.72-1.47). Sustainability could be a reason. Other Bel studies are ongoing, and results are awaited. Other studies including different anti-BCMA mAbs as well as ADCs targeting BCMA are ongoing[74-76].

BCMA bispecifics

BITEs are bispecific T cell engagers and represent a different modality of immunotherapy targeting BCMA. These agents are engineered proteins with two different antigen-binding fragments that bind to MM cells and T cells, thus creating an immunological synapse with direct plasma cell killing by T-cells[77-79]. The two common antigens involved are CD3 and CD16, and BCMA is the target of MM plasma cells. Many studies with BITEs utilizing BCMA showed great efficacy with moderate toxicity, such as CRS (cytokine release syndrome) and associated neurotoxicity syndrome (ICANS)[80-82].

Teclistamab (JNJ-64007957)

In the MajesTEC-1 clinical trial, Teclistamab (Tec) was tested as an IgG4 bispecific antibody targeting CD3 on T-cells and BCMA in RRMM. Included patients were heavily pretreated, with two-thirds of them triple-class refractory and 30% penta-refractory. Tec was initially administered intravenously or subcutaneously in different cohorts, and safety was particularly improved, particularly in terms of reduced CRS, for the latter formulation. The recommended dose was 1500 μg/kg weekly subcutaneously, after two escalating doses of 60 and 300 μg/kg. The ORR was 63% (median PFS 11.3 months). CRS was observed in 72% of the patients (only 1 patient with a grade 3) and Il-6 inhibitor tocilizumab was needed in 37% of patients. The most common neurotoxicity reported was headache in 8% of the patients[83-86]. Those results were followed by Tec authorization for marketing as monotherapy in MM patients who showed disease progression during the last of three prior therapies, including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 antibody[87].

Others BITEs currently studied are Elranatamab (PF-06863135), ABBV-383, and alnuctamab (CC-93269). In addition, novel tri-specific agents that target BCMA are under preclinical evaluation and are demonstrating high clinical potential[88-97].

BCMA CAR-Ts

CART (Chimeric antigen receptor T) cell therapy act as cell-mediated immunotherapy. Briefly, after an in vitro gene transfer strategy, the patient’s T cells acquire the ability to can recognize tumor antigens (mostly used is BCMA) on MM plasma cells and thus destroy them. The CARs are formed by a receptor with an extracellular portion that binds to the antigen and an intracellular signaling domain. Moreover, the extracellular portion is formed by a single-chain variable fragment, i.e., scFv, connected to a transmembrane domain. CD28 is used as a costimulatory molecule. The final product results in a combination effect of mAbs and T cells cytotoxicit[98-103]. Leukapheresis of the patient’s T cells is the first step of CART generation. Thereafter, the scFv and costimulatory domains are introduced with a viral vector. Before reinfusion, patients usually receive a conditioning regimen of fludarabine and cyclophosphamide (a chemotherapy regimen used to achieve lymphodepletion) to decrease autologous T cells and permit CARTs proliferation[104,105].

BCMA represents an ideal target for CAR-T therapy, and to date, two autologous BCMA-targeting CAR-Ts have been approved by the FDA, but several are being investigated in clinical trials[106-108]. BCMA is also being tested combined with CD19 for CAR-Ts multiple targeting[109-111]. A good efficacy has been demonstrated in early-phase clinical trials with bispecific CAR-Ts that target BCMA, CD19, or CD38[112]. Future alternative approaches could be represented by allogenic BCMA CAR-T cells or CAR-NK (CAR-natural killer), which are now investigated in early clinical trials[113-121].

Ide-Cel, idecabtagene vicleucel ( bb2121)

Ide-Cel is a CAR-T of the second generation that targets BCMA. Ide-Cel includes a CD3ζ signaling domain and an scFv, a costimulating domain. A great efficacy has been shown in preclinical experiments against MM plasma cells. It is independent of levels of BCMA expression or sBCMA levels[107]. Ide-Cel showed an ORR of 85% and a median PFS of 11.8 months in heavily pretreated MM patients in a phase I study. Toxicities such as CRS and ICANS (mostly grade 1-2) were observed in 76% and 42% of patients, respectively[122]. The KarMMa phase II study was conducted in 128 MM patients who had previously received three or more lines of therapy, including a PI, an IMiD, and an anti-CD38 mAb. CAR-Ts infusion produced an ORR of 73%. Also, MRD negativity at 10-5 was seen in 26% of the patients (median PFS 8.8 months, 20.2 months when CR was achieved). Of note, when CAR-T was employed in high-risk disease patients (i.e., penta-refractory disease, extramedullary disease, or high-risk cytogenetic), results were confirmed. Toxicities were acceptable (CRS 84%, but only 7 patients (5%) with ≥ grade 3; ICANS 18%, with ≥ grade 3 in 4 (3%) MM patients[123]. Ide-Cel was approved by the US FDA thereafter for MM patients treated with four lines of therapy (comprehensive of a PI, an IMiD, and an anti-CD38 mAb)[124]. Ide-Cel is now being used in several trials to explore its efficacy in various scenario, including the use in first-line therapy or at early relapse[125-127].

Cilta-cel ciltacabtagene autoleucel LCAR-B38M/JNJ-4528; Carvykti

Cilta-cel is a CAR-T-cell targeting BCMA with two antibodies to increase the binding avidity, a CD3-ζ signaling domain and a 4-1BB costimulatory domain[109]. In a recent phase I clinical trial, responses were high (ORR 88%) in RRMM patients after three or more prior lines of therapy (median PFS of 15 months). Toxicities were mostly grade 1-2 (CRS 90%, ICANS in 1 case)[128]. Subsequently, Cilta-cel was tested in 97 MM patients previously treated with multiple lines of therapy, with 40% of them being penta-refractory (CARTITUDE-1 trial). Interestingly, the response rate was quite high (> VGPR in 95%, MRD undetectable at 10-5 was achieved in 92%). Reported CRS and ICANS were similar to the previous study, but hematologic toxicities occurred more frequently (grade 3-4)[129]. Cilta-cel was then approved by FDA, in February 2022, for RRMM patients treated with > 4 prior lines of therapy including an anti-CD38 mAb, an IMiD, and a PI[130]. Recent ongoing phase III trials are CARTITUDE-2, evaluating cilta-cel efficacy and safety in different clinical settings in RRMM[131]; CARTITUDE-4, comparing Cilta-cel vs. pomalidomide, bortezomib and dexamethasone (PVd) vs. daratumumab, pomalidomide and dexamethasone (DPd) in RRMM; CARTITUDE-5, comparing bortezomib, lenalidomide, and dexamethasone (VRd) and Cilta-cel vs. VRd followed by lenalidomide and dexamethasone (Rd) therapy in transplant-ineligible patients MM at diagnosis[132].

BCMA, DRUG RESISTANCE, AND MM

While the efficacy and safety of BCMA-targeting agents have been demonstrated, data regarding drug resistance are also emerging, though the exact mechanisms of resistance towards these agents have not been fully understood[133]. Bone disease could be a reservoir for disease recurrence and a mechanism of resistance. Imaging is an important tool to detect residual disease outside the bone marrow or in extramedullary disease, although it is not known how BCMA antigen could be expressed on plasma cells outside the bone marrow. PET-CT is the gold standard technique to detect active disease and translated from lymphomas to MM[134,135]. In addition, whole body-MRI studies showed equal sensibility vs. PET-CT and can be used[136]. Downregulation of BCMA on PCs surface could be associated with resistance in a similar way as it has been described for CD19 and CD20 target therapies. Multi-targeted immunotherapies or the combination of BCMA targeting agents with γ-secretase inhibitors could overcome BCMA loss and both strategies are under investigation in clinical trials[52].

Humoral and cellular immune responses could limit the persistence of BCMA CAR-T, leading to loss of efficacy and disease relapse. Alternative manufacturing processes, such as the application of human scFVs or the removal of the light-chain domain from the CAR antigen-binding domain, have been demonstrated to reduce CAR-T immunogenicity. In addition, BCMA CAR-T persistency could be increased by the addition of a phosphoinositide 3 kinase inhibitor during ex vivo culture to augment the memory-like T cells of the final product. Besides, allogenic CAR-T could overcome resistance related to T cell exhaustion which may be present in RRMM patients[137-139].

Eventually, the tumor microenvironment is now considered to play a central role in promoting MM cell growth and has also been associated with drug resistance. Combination of BCMA targeting drugs with immunomodulatory agents could overcome this intrinsic mechanism of resistance, while trials are evaluating the next generation of armored CAR-T cells engineered to secret immunostimulatory cytokines or antibodies against inhibitory immune checkpoint receptors such as PD-1 and PD-L1.

DISCUSSION

Despite novel therapeutic advantages in recent years, MM remains incurable. BCMA immunotherapies are a novel anti-MM therapeutic approach that holds promise to improve MM survival in the future. ADCs, BITEs, and CAR-T cells are the newest therapeutic options targeting BCMA. Early clinical trials showed great efficacy and safety even IN MM patients treated with > 4 prior therapy lines. Since comparative studies of anti-BCMA targeted therapies are still lacking, it is not yet known whether one of these classes of agents is superior to another; however, they all have unique toxicities and logistical challenges. ADC is an interesting and efficacious therapy, but corneal toxicities need further understanding. Bispecific antibodies are therapies that can be used with excellent clinical activity. Disadvantages of bispecific antibodies could be their short lifetime and the need to start treatment in a hospital setting, as severe CRS/ICANS side effects usually appear at the beginning of therapy.

CAR-T cells are also a great option, as clinical trials reported high response rates in heavily pretreated MM patients. The main drawbacks of CAR-T cells include manufacturing time and expenses, leukapheresis necessity, and use of chemotherapy and infusion in a hospital setting for toxicities management. In addition, a relevant mechanism of resistance could be represented by the limited CAR-T growth and contact with the adverse plasma cell myeloma microenvironment, thus resulting in limited therapeutic effects after one year[140]. To overcome these problems, new strategies are currently under investigation utilizing combos of drug agents with CAR-T, maintenance therapies after CAR-T, novel methods to extend CAR-T’s duration, and implementing CAR-T production[141]. Additionally, the combination of a checkpoint inhibitor with CAR-Ts is being tested as it may offer an advantage of reducing T cell downfall[142].

The appropriate timing when to utilize a BCMA-targeted therapy is presently under investigation, with trials evaluating its role in earlier lines of therapy, including frontline. In fact, T cell-stimulating agents, such as CAR-T cells and BITEs, could probably produce deeper and longer responses if used at diagnosis or after only one or two lines of therapy, when MM patients are not heavily treated and may be at lower risk for T cell exhaustion.

In conclusion, therapies that target BCMA will play an important role in MM therapy, with the ambitious purpose of improving the cure rate; however, further investigations are still necessary to better define their real impact in clinical practice.

DECLARATIONS

Authors’ contributions

Made substantial contributions to the conception and design of the study: Sammartano V, Gozzetti A

Wrote the manuscript: Sammartano V, Franceschini M, Fredducci S, Caroni F, Ciofini S, Pacelli P

Supervised the manuscript: Gozzetti A, Bocchia M

Availability of data and materials

Not applicable.

Financial support and sponsorship

None.

Conflicts of interest

All authors declared that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2023.

REFERENCES

1. Rajkumar SV. Multiple myeloma: 2022 update on diagnosis, risk stratification, and management. Am J Hematol 2022;97:1086-107.

2. Paul B, Rodriguez C, Usmani SZ. BCMA-targeted biologic therapies: the next standard of care in multiple myeloma therapy. Drugs 2022;82:613-31.

3. Fonseca R, Abouzaid S, Bonafede M, et al. Trends in overall survival and costs of multiple myeloma, 2000-2014. Leukemia 2017;31:1915-21.

4. Usmani SZ, Hoering A, Cavo M, et al. Clinical predictors of long-term survival in newly diagnosed transplant eligible multiple myeloma - an IMWG research project. Blood Cancer J 2018;8:123.

5. Nishimura KK, Barlogie B, van Rhee F, et al. Long-term outcomes after autologous stem cell transplantation for multiple myeloma. Blood Adv 2020;4:422-31.

6. Joseph NS, Kaufman JL, Dhodapkar MV, et al. Long-term follow-up results of lenalidomide, bortezomib, and dexamethasone induction therapy and risk-adapted maintenance approach in newly diagnosed multiple myeloma. J Clin Oncol 2020;38:1928-37.

7. Gozzetti A, Candi V, Papini G, Bocchia M. Therapeutic advancements in multiple myeloma. Front Oncol 2014;4:241.

8. Palumbo A, Falco P, Falcone A, et al. Melphalan, prednisone, and lenalidomide for newly diagnosed myeloma: kinetics of neutropenia and thrombocytopenia and time-to-event results. Clin Lymphoma Myeloma 2009;9:145-50.

9. Cavo M, Benni M, Ronconi S, et al. Melphalan-prednisone versus alternating combination VAD/MP or VND/MP as primary therapy for multiple myeloma: final analysis of a randomized clinical study. Haematologica 2002;87:934-42.

10. Yu B, Jiang T, Liu D. BCMA-targeted immunotherapy for multiple myeloma. J Hematol Oncol 2020;13:125.

11. Usmani S, Ahmadi T, Ng Y, et al. Analysis of real-world data on overall survival in multiple myeloma patients with ≥ 3 prior lines of therapy including a proteasome inhibitor (PI) and an immunomodulatory drug (IMiD), or double refractory to a PI and an ImiD. Oncologist 2016;21:1355-61.

12. Kleber M, Ntanasis-Stathopoulos I, Terpos E. BCMA in multiple myeloma-A promising key to therapy. J Clin Med 2021;10:4088.

13. Gozzetti A, Ciofini S, Simoncelli M, et al. Anti CD38 monoclonal antibodies for multiple myeloma treatment. Hum Vaccin Immunother 2022;18:2052658.

14. Kumar SK, Dimopoulos MA, Kastritis E, et al. Natural history of relapsed myeloma, refractory to immunomodulatory drugs and proteasome inhibitors: a multicenter IMWG study. Leukemia 2017;31:2443-8.

15. Gozzetti A, Cerase A, Lotti F, et al. Extramedullary intracranial localization of multiple myeloma and treatment with novel agents: a retrospective survey of 50 patients. Cancer 2012;118:1574-84.

16. Castillo JJ, Jurczyszyn A, Brozova L, et al. IgM myeloma: a multicenter retrospective study of 134 patients. Am J Hematol 2017;92:746-51.

17. Jurczyszyn A, Radocha J, Davila J, et al. Prognostic indicators in primary plasma cell leukemia: a multicentre retrospective study of 117 patients. Br J Haematol 2018;180:831-9.

18. Pick M, Vainstein V, Goldschmidt N, et al. Daratumumab resistance is frequent in advanced-stage multiple myeloma patients irrespective of CD38 expression and is related to dismal prognosis. Eur J Haematol 2018;100:494-501.

19. Gandhi UH, Cornell RF, Lakshman A, et al. Outcomes of patients with multiple myeloma refractory to CD38-targeted monoclonal antibody therapy. Leukemia 2019;33:2266-75.

20. Costa LJ, Hari P, Kumar SK, et al. Overall survival of triple class refractory, penta-exposed multiple myeloma (MM) patients treated with selinexor plus dexamethasone or conventional care: a combined analysis of the STORM and mammoth studies. Blood 2019;134:3125.

21. Gozzetti A, Cerase A. Novel agents in CNS myeloma treatment. Cent Nerv Syst Agents Med Chem 2014;14:23-7.

22. Munshi NC, Avet-Loiseau H, Rawstron AC, et al. Association of minimal residual disease with superior survival outcomes in patients with multiple myeloma: a meta-analysis. JAMA Oncol 2017;3:28-35.

23. Kastritis E, Roussou M, Eleutherakis-Papaiakovou E, et al. Early relapse after autologous transplant is associated with very poor survival and identifies an ultra-high-risk group of patients with myeloma. Clin Lymphoma Myeloma Leuk 2020;20:445-52.

24. Gozzetti A, Raspadori D, Bacchiarri F, et al. Minimal residual disease in multiple myeloma: state of the art and applications in clinical practice. J Pers Med 2020;10:120.

25. Avigan D, Rosenblatt J. Current treatment for multiple myeloma. N Engl J Med 2014;371:961-2.

26. Zamagni E, Barbato S, Cavo M. How I treat high-risk multiple myeloma. Blood 2022;139:2889-903.

27. Guo R, Lu W, Zhang Y, Cao X, Jin X, Zhao M. Targeting BCMA to treat multiple myeloma: updates from the 2021 ASH annual meeting. Front Immunol 2022;13:839097.

28. Feng D, Sun J. Overview of anti-BCMA CAR-T immunotherapy for multiple myeloma and relapsed/refractory multiple myeloma. Scand J Immunol 2020;92:e12910.

29. Avery DT, Kalled SL, Ellyard JI, et al. BAFF selectively enhances the survival of plasmablasts generated from human memory B cells. J Clin Invest 2003;112:286-97.

30. 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;10:1693-702.

31. Tai YT, Acharya C, An G, et al. APRIL and BCMA promote human multiple myeloma growth and immunosuppression in the bone marrow microenvironment. Blood 2016;127:3225-36.

32. Coquery CM, Erickson LD. Regulatory roles of the tumor necrosis factor receptor BCMA. Crit Rev Immunol 2012;32:287-305.

33. Rennert P, Schneider P, Cachero TG, et al. A soluble form of B cell maturation antigen, a receptor for the tumor necrosis factor family member APRIL, inhibits tumor cell growth. J Exp Med 2000;192:1677-84.

34. Demchenko YN, Glebov OK, Zingone A, Keats JJ, Bergsagel PL, Kuehl WM. Classical and/or alternative NF-kappaB pathway activation in multiple myeloma. Blood 2010;115:3541-52.

35. Moreaux J, Legouffe E, Jourdan E, et al. BAFF and APRIL protect myeloma cells from apoptosis induced by interleukin 6 deprivation and dexamethasone. Blood 2004;103:3148-57.

36. Shen X, Guo Y, Qi J, Shi W, Wu X, Ju S. Binding of B-cell maturation antigen to B-cell activating factor induces survival of multiple myeloma cells by activating Akt and JNK signaling pathways. Cell Biochem Funct 2016;34:104-10.

37. Hatzoglou A, Roussel J, Bourgeade MF, et al. TNF receptor family member BCMA (B cell maturation) associates with TNF receptor-associated factor (TRAF) 1, TRAF2, and TRAF3 and activates NF-kappa B, elk-1, c-Jun N-terminal kinase, and p38 mitogen-activated protein kinase. J Immunol 2000;165:1322-30.

38. O'Connor BP, Raman VS, Erickson LD, et al. BCMA is essential for the survival of long-lived bone marrow plasma cells. J Exp Med 2004;199:91-8.

39. Xu S, Lam KP. B-cell maturation protein, which binds the tumor necrosis factor family members BAFF and APRIL, is dispensable for humoral immune responses. Mol Cell Biol 2001;21:4067-74.

40. Carpenter RO, Evbuomwan MO, Pittaluga 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.

41. Sanchez E, Li M, Kitto A, et al. Serum B-cell maturation antigen is elevated in multiple myeloma and correlates with disease status and survival. Br J Haematol 2012;158:727-38.

42. Yaccoby S, Pennisi A, Li X, et al. Atacicept (TACI-Ig) inhibits growth of TACI(high) primary myeloma cells in SCID-hu mice and in coculture with osteoclasts. Leukemia 2008;22:406-13.

43. Lin L, Cho SF, Wen K, et al. Impacts of a proliferation-inducing ligand on current therapeutic monoclonal antibody-induced cytotoxicity against human multiple myeloma cells. Blood 2019;134:3105.

44. Laurent SA, Hoffmann FS, Kuhn PH, et al. γ-Secretase directly sheds the survival receptor BCMA from plasma cells. Nat Commun 2015;6:7333.

45. Ghermezi M, Li M, Vardanyan S, et al. Serum B-cell maturation antigen: a novel biomarker to predict outcomes for multiple myeloma patients. Haematologica 2017;102:785-95.

46. Gavriatopoulou M, Ntanasis-Stathopoulos I, Dimopoulos MA, Terpos E. Anti-BCMA antibodies in the future management of multiple myeloma. Expert Rev Anticancer Ther 2019;19:319-26.

47. Ali SA, Shi V, Maric I, et al. T cells expressing an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma. Blood 2016;128:1688-700.

48. Martino M, Paviglianiti A. An update on B-cell maturation antigen-targeted therapies in multiple myeloma. Expert Opin Biol Ther 2021;21:1025-34.

49. Visram A, Soof C, Rajkumar SV, et al. Serum BCMA levels predict outcomes in MGUS and smoldering myeloma patients. Blood Cancer J 2021;11:120.

50. Sanchez E, Gillespie A, Tang G, et al. Soluble B-Cell maturation antigen mediates tumor-induced immune deficiency in multiple myeloma. Clin Cancer Res 2016;22:3383-97.

51. Chen H, Li M, Xu N, et al. Serum B-cell maturation antigen (BCMA) reduces binding of anti-BCMA antibody to multiple myeloma cells. Leuk Res 2019;81:62-6.

52. Pont MJ, Hill T, Cole GO, et al. γ-Secretase inhibition increases efficacy of BCMA-specific chimeric antigen receptor T cells in multiple myeloma. Blood 2019;134:1585-97.

53. Sherbenou DW, Behrens CR, Su Y, Wolf JL, Martin TG 3rd, Liu B. The development of potential antibody-based therapies for myeloma. Blood Rev 2015;29:81-91.

54. Shah N, Chari A, Scott E, Mezzi K, Usmani SZ. B-cell maturation antigen (BCMA) in multiple myeloma: rationale for targeting and current therapeutic approaches. Leukemia 2020;34:985-1005.

55. Bruins WSC, Zweegman S, Mutis T, van de Donk NWCJ. Targeted therapy with immunoconjugates for multiple myeloma. Front Immunol 2020;11:1155.

56. Pahl A, Lutz C, Hechler T. Amanitins and their development as a payload for antibody-drug conjugates. Drug Discov Today Technol 2018;30:85-9.

57. Bera TK. Anti-BCMA immunotoxins: design, production, and preclinical evaluation. Biomolecules 2020;10:1387.

58. McCombs JR, Owen SC. Antibody drug conjugates: design and selection of linker, payload and conjugation chemistry. AAPS J 2015;17:339-51.

59. Yu B, Liu D. Antibody-drug conjugates in clinical trials for lymphoid malignancies and multiple myeloma. J Hematol Oncol 2019;12:94.

60. Abdollahpour-Alitappeh M, Lotfinia M, Gharibi T, et al. Antibody-drug conjugates (ADCs) for cancer therapy: strategies, challenges, and successes. J Cell Physiol 2019;234:5628-42.

61. Tai YT, Mayes PA, Acharya C, et al. Novel anti-B-cell maturation antigen antibody-drug conjugate (GSK2857916) selectively induces killing of multiple myeloma. Blood 2014;123:3128-38.

62. Sheikh S, Lebel E, Trudel S. Belantamab mafodotin in the treatment of relapsed or refractory multiple myeloma. Future Oncol 2020;16:2783-98.

63. Markham A. Belantamab mafodotin: first approval. Drugs 2020;80:1607-13.

64. Eastman S, Shelton C, Gupta I, Krueger J, Blackwell C, Bojczuk PM. Synergistic activity of belantamab mafodotin (anti-BCMA immuno-conjugate) with PF-03084014 (gamma-secretase inhibitor) in Bcma-expressing cancer cell lines. Blood 2019;134:4401.

65. Montes de Oca R, Alavi AS, Vitali N, et al. Belantamab mafodotin (GSK2857916) drives immunogenic cell death and immune-mediated antitumor responses in vivo. Mol Cancer Ther 2021;20:1941-55.

66. Trudel S, Lendvai N, Popat R, et al. Targeting B-cell maturation antigen with GSK2857916 antibody-drug conjugate in relapsed or refractory multiple myeloma (BMA117159): a dose escalation and expansion phase 1 trial. Lancet Oncol 2018;19:1641-53.

67. Trudel S, Lendvai N, Popat R, et al. Antibody-drug conjugate, GSK2857916, in relapsed/refractory multiple myeloma: an update on safety and efficacy from dose expansion phase I study. Blood Cancer J 2019;9:37.

68. Lonial S, Lee HC, Badros A, et al. Longer term outcomes with single-agent belantamab mafodotin in patients with relapsed or refractory multiple myeloma: 13-month follow-up from the pivotal DREAMM-2 study. Cancer 2021;127:4198-212.

69. 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;21:207-21.

70. Richardson PG, Lee HC, Abdallah AO, et al. Single-agent belantamab mafodotin for relapsed/refractory multiple myeloma: analysis of the lyophilised presentation cohort from the pivotal DREAMM-2 study. Blood Cancer J 2020;10:106.

71. Wahab A, Rafae A, Mushtaq K, et al. Ocular toxicity of belantamab mafodotin, an oncological perspective of management in relapsed and refractory multiple myeloma. Front Oncol 2021;11:678634.

72. Offidani M, Corvatta L, Morè S, Olivieri A. Belantamab mafodotin for the treatment of multiple myeloma: an overview of the clinical efficacy and safety. Drug Des Devel Ther 2021;15:2401-15.

73. Nooka AK, Weisel K, van de Donk NW, et al. Belantamab mafodotin in combination with novel agents in relapsed/refractory multiple myeloma: DREAMM-5 study design. Future Oncol 2021;17:1987-2003.

74. O’Donnell EK, Raje NS. New monoclonal antibodies on the horizon in multiple myeloma. Ther Adv Hematol 2017;8:41-53.

75. Lee HC, Raje NS, Landgren O, et al. Phase 1 study of the anti-BCMA antibody-drug conjugate AMG 224 in patients with relapsed/refractory multiple myeloma. Leukemia 2021;35:255-8.

76. Figueroa-Vazquez V, Ko J, Breunig C, et al. HDP-101, an anti-BCMA antibody-drug conjugate, safely delivers amanitin to induce cell death in proliferating and resting multiple myeloma cells. Mol Cancer Ther 2021;20:367-78.

77. Suurs FV, Lub-de Hooge MN, de Vries EGE, de Groot DJA. A review of bispecific antibodies and antibody constructs in oncology and clinical challenges. Pharmacol Ther 2019;201:103-19.

78. Offner S, Hofmeister R, Romaniuk A, Kufer P, Baeuerle PA. Induction of regular cytolytic T cell synapses by bispecific single-chain antibody constructs on MHC class I-negative tumor cells. Mol Immunol 2006;43:763-71.

79. Fan G, Wang Z, Hao M, Li J. Bispecific antibodies and their applications. J Hematol Oncol 2015;8:130.

80. Ravi G, Costa LJ. Bispecific T-cell engagers for treatment of multiple myeloma. Am J Hematol 2023;98:S13-S21.

81. Shimabukuro-Vornhagen A, Gödel P, Subklewe M, et al. Cytokine release syndrome. J Immunother Cancer 2018;6:56.

82. Lee DW, Gardner R, Porter DL, et al. Current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014;124:188-95.

83. Pillarisetti K, Powers G, Luistro L, et al. Teclistamab is an active T cell-redirecting bispecific antibody against B-cell maturation antigen for multiple myeloma. Blood Adv 2020;4:4538-49.

84. 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;398:665-74.

85. Moreau P, Garfall AL, van de Donk NWCJ, et al. Teclistamab in relapsed or refractory multiple myeloma. N Engl J Med 2022;387:495-505.

86. Girgis S, Lin SXW, Pillarisetti K, et al. Translational modeling predicts efficacious therapeutic dosing range of teclistamab for multiple myeloma. Target Oncol 2022;17:433-9.

87. Mullard A. BCMA-targeted bispecific gets first green light, in the EU. Nat Rev Drug Discov 2022;21:626.

88. 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.

89. DiLillo DJ, Olson K, Mohrs K, et al. A BCMAxCD3 bispecific T cell-engaging antibody demonstrates robust antitumor efficacy similar to that of anti-BCMA CAR T cells. Blood Adv 2021;5:1291-304.

90. Clarke SC, Ma B, Trinklein ND, et al. Multispecific antibody development platform based on human heavy chain antibodies. Front Immunol 2019;9:3037.

91. Sebag M, Raje NS, Bahlis NJ, et al. Elranatamab (PF-06863135), a B-cell maturation antigen (BCMA) targeted CD3-Engaging bispecific molecule, for patients with relapsed or refractory multiple myeloma: results from Magnetismm-1. Blood 2021;138:895.

92. Harris KE, Aldred SF, Davison LM, et al. Sequence-based discovery demonstrates that fixed light chain human transgenic rats produce a diverse repertoire of antigen-specific antibodies. Front Immunol 2018;9:889.

93. Buelow B, D'Souza A, Rodriguez C, et al. TNB383B. 0001: a multicenter, phase 1, open-label, dose-escalation andexpansion study of TNB-383B, a bispecific antibodytargeting BCMA in subjects with relapsed or refractorymultiple myeloma. Blood 2019;134:1874.

94. 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. Blood 2021;138:900.

95. Harrison SJ, Minnema MC, Lee HC, 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.

96. Costa LJ, Wong SW, Bermúdez A, et al. First 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): interim results of a phase 1 multicenter trial. Blood 2019;134:143.

97. Vrohlings M, Müller J, Jungmichel S, et al. Preclinical assessment of CDR101-a BCMAxCD3xPD-L1 trispecific antibody with superior anti-tumor efficacy. Blood 2021;138:1583.

98. Jayaraman J, Mellody MP, Hou AJ, et al. CAR-T design: elements and their synergistic function. EBioMedicine 2020;58:102931.

99. Sadelain M, Brentjens R, Rivière I. The basic principles of chimeric antigen receptor design. Cancer Discov 2013;3:388-98.

100. de Donk NWCJ, Usmani SZ, Yong K. CAR T-cell therapy for multiple myeloma: state of the art and prospects. Lancet Haematol 2021;8:e446-61.

101. Song DG, Ye Q, Poussin M, Harms GM, Figini M, Powell DJJr. CD27 costimulation augments the survival and antitumor activity of redirected human T cells in vivo. Blood 2012;119:696-706.

102. Rafiq S, Hackett CS, Brentjens RJ. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat Rev Clin Oncol 2020;17:147-67.

103. Mikkilineni L, Kochenderfer JN. Chimeric antigen receptor T-cell therapies for multiple myeloma. Blood 2017;130:2594-602.

104. Suryadevara CM, Desai R, Farber SH, et al. Preventing Lck activation in CAR T cells confers treg resistance but requires 4-1BB signaling for them to persist and treat solid tumors in nonlymphodepleted hosts. Clin Cancer Res 2019;25:358-68.

105. Gattinoni L, Finkelstein SE, Klebanoff CA, et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med 2005;202:907-12.

106. Nobari ST, Nojadeh JN, Talebi M. B-cell maturation antigen targeting strategies in multiple myeloma treatment, advantages and disadvantages. J Transl Med 2022;20:82.

107. Friedman KM, Garrett TE, Evans JW, et al. Effective targeting of multiple B-cell maturation antigen-expressing hematological malignances by anti-B-cell maturation antigen chimeric antigen receptor T cells. Hum Gene Ther 2018;29:585-601.

108. Bu DX, Singh R, Choi EE, et al. Pre-clinical validation of B cell maturation antigen (BCMA) as a target for T cell immunotherapy of multiple myeloma. Oncotarget 2018;9:25764-80.

109. Xu J, Chen LJ, Yang SS, et al. Exploratory trial of a biepitopic CAR T-targeting B cell maturation antigen in relapsed/refractory multiple myeloma. Proc Natl Acad Sci USA 2019;116:9543-51.

110. Kang L, Zhang J, Li M, et al. Characterization of novel dual tandem CD19/BCMA chimeric antigen receptor T cells to potentially treat multiple myeloma. Biomark Res 2020;8:14.

111. Zhang H, Gao L, Liu L, et al. A Bcma and CD19 bispecific CAR-T for relapsed and refractory multiple myeloma. Blood ;134:3147.

112. Mei H, Li C, Jiang H, et al. A bispecific CAR-T cell therapy targeting BCMA and CD38 in relapsed or refractory multiple myeloma. J Hematol Oncol 2021;14:1-7.

113. Manier S, Ingegnere T, Escure G, et al. Current state and next-generation CAR-T cells in multiple myeloma. Blood Rev 2022;54:100929.

114. Roex G, Campillo-Davo D, Flumens D, et al. Two for one: targeting BCMA and CD19 in B-cell malignancies with off-the-shelf dual-CAR NK-92 cells. J Transl Med 2022;20:124.

115. Depil S, Duchateau P, Grupp SA, Mufti G, Poirot L. “Off-the-shelf” allogeneic CAR T cells: development and challenges. Nat Rev Drug Discov 2020;19:185-99.

116. Graham C, Jozwik A, Pepper A, Benjamin R. Allogeneic CAR-T cells: more than ease of access? Cells 2018;7:155.

117. Sommer C, Boldajipour B, Kuo TC, et al. Preclinical evaluation of allogeneic CAR T cells targeting BCMA for the treatment of multiple myeloma. Mol Ther 2019;27:1126-38.

118. Mailankody S, Matous JV, Liedtke M, et al. Universal: an allogeneic first-in-human study of the anti-BCMA ALLO-715 and the anti-CD52 ALLO-647 in relapsed/refractory multiple myeloma. Blood 2020;136:24-5.

119. Gong Y, Klein Wolterink RGJ, Wang J, Bos GMJ, Germeraad WTV. Chimeric antigen receptor natural killer (CAR-NK) cell design and engineering for cancer therapy. J Hematol Oncol 2021;14:73.

120. Williams RL, Cooley S, Bachanova V, et al. Recipient T cell exhaustion and successful adoptive transfer of haploidentical natural killer cells. Biol Blood Marrow Transplant 2018;24:618-22.

121. Martín EM, Encinas J, García-Ortiz A, et al. Exploring NKG2D and BCMA-CAR NK-92 for adoptive cellular therapy to multiple myeloma. Clin Lymphoma Myeloma Leuk 2019;19:e24-5.

122. Raje N, Berdeja J, Lin Y, et al. Anti-BCMA CAR T-cell therapy bb2121 in relapsed or refractory multiple myeloma. N Engl J Med 2019;380:1726-37.

123. Munshi NC, Anderson LD Jr, Shah N, et al. Idecabtagene vicleucel in relapsed and refractory multiple myeloma. N Engl J Med 2021;384:705-16.

124. Anderson LD Jr. Idecabtagene vicleucel (ide-cel) CAR T-cell therapy for relapsed and refractory multiple myeloma. Future Oncol 2022;18:277-89.

125. Oriol A, Abril L, Torrent A, Ibarra G, Ribera JM. The role of idecabtagene vicleucel in patients with heavily pretreated refractory multiple myeloma. Ther Adv Hematol 2021;12:20406207211019622.

126. Delforge M, Baz RC, Cavo M, et al. KarMMa-3: a phase 3 study of idecabtagene vicleucel (ide-cel, bb2121), a BCMA-directed CAR T cell therapy vs standard regimens in relapsed and refractory multiple myeloma. Blood 2020;136:24-5.

127. Usmani SZ, Berdeja JG, Truppel-Hartmann A, et al. KarMMa-4: idecabtagene vicleucel (ide-cel, bb2121), a BCMA-directed CAR T-cell therapy, in high-risk newly diagnosed multiple myeloma. Blood 2020;136:18-9.

128. Zhao WH, Liu J, Wang BY, et al. A phase 1, open-label study of LCAR-B38M, a chimeric antigen receptor T cell therapy directed against B cell maturation antigen, in patients with relapsed or refractory multiple myeloma. J Hematol Oncol 2018;11:141.

129. Berdeja JG, Madduri D, Usmani SZ, et al. Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): a phase 1b/2 open-label study. Lancet 2021;398:314-24.

130. Martin T, Usmani SZ, Berdeja JG, et al. Ciltacabtagene autoleucel, an anti-B-cell maturation antigen chimeric antigen receptor T-cell therapy, for relapsed/refractory multiple myeloma: CARTITUDE-1 2-year follow-up. J Clin Oncol 2022;41:JCO.2200842.

131. Cohen YC, Cohen AD, Delforge M, et al. Efficacy and safety of ciltacabtagene autoleucel (cilta-cel), a B-cell maturation antigen (BCMA)-directed chimeric antigen receptor (CAR) T-cell therapy, in lenalidomide-refractory patients with progressive multiple myeloma after 1-3 prior lines of therapy: updated results from CARTITUDE-2. Blood 2021;138:3866.

132. Dytfeld D, Dhakal B, Agha M, et al. Bortezomib, lenalidomide and dexamethasone (VRd) followed by ciltacabtagene autoleucel versus Vrd followed by lenalidomide and dexamethasone (Rd) maintenance in patients with newly diagnosed multiple myeloma not intended for transplant: a randomized, phase 3 study (CARTITUDE-5). Blood 2021;138:1835.

133. Gozzetti A, Ciofini S, Sicuranza A, et al. Drug resistance and minimal residual disease in multiple myeloma. Cancer Drug Resist 2022;5:171-83.

134. Zamagni E, Nanni C, Gay F et al. MRD evaluation by PET/CT according to deauville criteria combined with multiparameter flow cytometry in newly diagnosed transplant eligible multiple myeloma (MM) patients enrolled in the phase II randomized forte trial. Blood 2019;134:4321.

135. Zinzani P. L, Zompatori M, Bendandi M, et al. Monitoring bulky mediastinal disease with gallium-67, CT-scan and magnetic resonance imaging in Hodgkin’s disease and high-grade non-Hodgkin’slymphoma. Leuk Lymphoma 1996;22:131-5.

136. Hillengass J, Usmani S, Rajkumar SV et al. International myeloma working group consensus recommendations on imaging in monoclonal plasma cell disorders. Lancet Oncol 2019;20:e346.

137. Cencini E, Fabbri A, Sicuranza A, Gozzetti A, Bocchia M. The role of tumor-associated macrophages in hematologic malignancies. Cancers 2021;13:3597.

138. Majzner RG, Mackall CL. Tumor antigen escape from CAR T-cell therapy. Cancer Discov 2018;8:1219-26.

139. de Donk NWCJ, Themeli M, Usmani SZ. Determinants of response and mechanisms of resistance of CAR T-cell therapy in multiple myeloma. Blood Cancer Discov 2021;2:302-18.

140. Shah NN, Fry TJ. Mechanisms of resistance to CAR T cell therapy. Nat Rev Clin Oncol 2019;16:372-85.

141. Atilla PA, Atilla E. Resistance against anti-CD19 and anti-BCMA CAR T cells: recent advances and coping strategies. Transl Oncol 2022;22:101459.

142. Rafiq S, Yeku OO, Jackson HJ, et al. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat Biotechnol 2018;36:847-56.

Cite This Article

Export citation file: BibTeX | RIS

OAE Style

Sammartano V, Franceschini M, Fredducci S, Caroni F, Ciofini S, Pacelli P, Bocchia M, Gozzetti A. Anti-BCMA novel therapies for multiple myeloma. Cancer Drug Resist 2023;6:169-81. http://dx.doi.org/10.20517/cdr.2022.138

AMA Style

Sammartano V, Franceschini M, Fredducci S, Caroni F, Ciofini S, Pacelli P, Bocchia M, Gozzetti A. Anti-BCMA novel therapies for multiple myeloma. Cancer Drug Resistance. 2023; 6(1): 169-81. http://dx.doi.org/10.20517/cdr.2022.138

Chicago/Turabian Style

Sammartano, Vincenzo, Marta Franceschini, Sara Fredducci, Federico Caroni, Sara Ciofini, Paola Pacelli, Monica Bocchia, Alessandro Gozzetti. 2023. "Anti-BCMA novel therapies for multiple myeloma" Cancer Drug Resistance. 6, no.1: 169-81. http://dx.doi.org/10.20517/cdr.2022.138

ACS Style

Sammartano, V.; Franceschini M.; Fredducci S.; Caroni F.; Ciofini S.; Pacelli P.; Bocchia M.; Gozzetti A. Anti-BCMA novel therapies for multiple myeloma. Cancer Drug Resist. 2023, 6, 169-81. http://dx.doi.org/10.20517/cdr.2022.138

About This Article

© The Author(s) 2023. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
1152
Downloads
1013
Citations
2
Comments
0
15

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Cite This Article 11 clicks
Like This Article 15 likes
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Cancer Drug Resistance
ISSN 2578-532X (Online)

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/