1
|
Meng Z, Zheng H, Li Y, Bai J, Zhang L, Li L. Efficacy and Safety of triplet versus doublet regimens in patients with multiple myeloma: A systematic review and meta-analysis. Curr Probl Cancer 2025; 56:101202. [PMID: 40184871 DOI: 10.1016/j.currproblcancer.2025.101202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/08/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND The efficacy and safety of various therapies for multiple myeloma (MM) remain a subject of ongoing debate, with inconsistent findings. This meta-analysis aimed to compare the efficacy and safety of triplet versus doublet regimens in the management of MM. This study followed the guidelines delineated in the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) 2020 statement, with our protocol duly registered in PROSPERO (CRD42024527903). METHODS An exhaustive literature search was performed across four databases, PubMed, EMBASE, Web of Science, and Cochrane Library, from their commencement to March 5, 2024. Data on overall survival (OS), progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), adverse events (AEs), and grade ≥ 3 AEs were synthesized using either random-effects or fixed-effects models. RESULTS This analysis considered 29 studies, which cover approximately 11,230 MM patients in total. Triplet regimens were found to yield better PFS and OS for MM patients as compared to the doublet regimens. Although the incidence of serious AEs was higher under the triplet regimens, with pooled RRs of grade ≥ 3 AEs reaching 1.13. Besides, subgroup analysis demonstrated that patients with relapsed/refractory multiple myeloma (RRMM) tended to have better PFS and OS under triple therapy, in contrast to newly diagnosed multiple myeloma (NDMM) and older adults, who experienced little to no significant impact. CONCLUSIONS Triplet regimens demonstrate superior PFS and OS compared to doublet regimens in MM patients, but also have a higher likelihood of causing AEs of grade 3 or 4.
Collapse
Affiliation(s)
- Zilu Meng
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou Gansu 730030, China
| | - Hanxue Zheng
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou Gansu 730030, China
| | - Yanhong Li
- Key Laboratory of the Hematology of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou Gansu 730000, China
| | - Jun Bai
- Key Laboratory of the Hematology of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou Gansu 730000, China
| | - Liansheng Zhang
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou Gansu 730030, China; Key Laboratory of the Hematology of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou Gansu 730000, China.
| | - Lijuan Li
- Department of Hematology, The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou Gansu 730030, China; Key Laboratory of the Hematology of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou Gansu 730000, China.
| |
Collapse
|
2
|
Amin R, Dey BK, Darwin R, Cho WC, Sharifi-Rad J, Calina D. BCMA-targeted therapies in multiple myeloma: advances, challenges and future prospects. Med Oncol 2025; 42:204. [PMID: 40338452 DOI: 10.1007/s12032-025-02753-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 04/28/2025] [Indexed: 05/09/2025]
Abstract
Multiple myeloma (MM) is hematological cancer characterized by the aberrant proliferation of plasma cells. The treatment of MM has historically presented challenges, with a limited number of patients achieving sustained remission. Recent advancements in the therapeutic landscape have been marked by the development of B-cell maturation antigen (BCMA)-targeted therapies. BCMA, a plasma cell surface protein, is instrumental in the proliferation and survival of myeloma cells. This review aims to critically assess recent developments in BCMA-targeted therapies. The focus is on evaluating their efficacy and accessibility, as well as discussing potential future directions in this field. Emphasis is placed on chimeric antigen receptor (CAR) T-cell therapy and bispecific antibodies as emerging therapeutic strategies. An extensive review of current clinical trials and studies was conducted, centering on BCMA-targeted therapies. This encompassed an analysis of CAR T-cell therapies, which involve the genetic modification of patient T-cells to target BCMA, and bispecific antibodies that bind to both BCMA on myeloma cells and CD3 on T-cells. Clinical trials have demonstrated the efficacy of BCMA-targeted therapies in MM, with some patients achieving complete remission. However, these therapies are associated with adverse effects such as cytokine release syndrome and neurotoxicity. Research efforts are ongoing to reduce these side effects and enhance overall therapeutic effectiveness. BCMA-targeted therapies signify a notable advancement in MM treatment, offering prospects for prolonged remission and potentially curative outcomes. Despite existing challenges, these therapies represent a significant shift in MM management. The review highlights the necessity of ongoing research to optimize these therapies, improve patient outcomes, and increase treatment accessibility.
Collapse
Affiliation(s)
- Ruhul Amin
- Rahman Institute of Pharmaceutical Sciences and Research (RIPSR), Kamarkuchi, Kamrup (M), Tepesia, Assam, 782402, India
| | - Biplab Kumar Dey
- Dooars Institute of Pharmaceutical Sciences and Research (DIPSAR), Ghoksadanga, Cooch Behar, West Bengal, 736171, India
| | - Ronald Darwin
- School of Pharmaceutical Sciences, Vels Institute of Science Technology & Advanced Studies, Chennai, 600117, India
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China.
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, 092301, Ecuador.
- Centro de Estudios Tecnológicos y Universitarios del Golfo, Veracruz, Mexico.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| |
Collapse
|
3
|
Hu Y, Xie Y, Wang X, Yang L, Geng H, Yi Z, Zhang Y, Ma L, Chen F. Targeting BCMA in multiple myeloma: designs, challenges, and future directions. Cancer Immunol Immunother 2025; 74:77. [PMID: 39891674 PMCID: PMC11787132 DOI: 10.1007/s00262-024-03913-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/01/2024] [Indexed: 02/03/2025]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has emerged as a groundbreaking immunotherapy, demonstrating significant efficacy in treating B cell malignancies. In the context of multiple myeloma (MM), B cell maturation antigen (BCMA) has been identified as a critical target, driving the development of CAR T cell therapies designed to address this plasma cell cancer. Various CAR designs, utilizing different BCMA recognition domains, have yielded promising clinical results, leading to the approval of two BCMA-targeting CAR T cell therapies by the US Food and Drug Administration (FDA) for the treatment of MM. This review uniquely examines the BCMA CAR T cell landscape, emphasizing the design of recognition domains, clinical efficacy, and patient outcomes. It critically addresses emerging challenges such as antigen escape and toxicity profiles, which have surfaced alongside therapeutic advances. Moreover, the review spotlights cutting-edge developments, including dual-targeting CAR T strategies, advancements in CAR T cell manufacturing, and innovative allogeneic CAR T approaches utilizing healthy donor cells. By detailing both the breakthroughs and ongoing challenges in BCMA CAR T cell therapy, this review offers a comprehensive perspective on the current state and future possibilities of CAR T cell therapy for MM and its expanding role in treating hematologic malignancies and beyond.
Collapse
Affiliation(s)
- Yi Hu
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China
| | - Yuetao Xie
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China
| | - Xiaodong Wang
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China
| | - Lufeng Yang
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China
| | - He Geng
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China
| | - Zugang Yi
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China
| | - Yao Zhang
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China
| | - Lin Ma
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China
| | - Fang Chen
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China.
| |
Collapse
|
4
|
Ram M, Fraser MR, Vieira dos Santos J, Tasakis R, Islam A, Abo-Donia JU, Parekh S, Lagana A. The Genetic and Molecular Drivers of Multiple Myeloma: Current Insights, Clinical Implications, and the Path Forward. Pharmgenomics Pers Med 2024; 17:573-609. [PMID: 39723112 PMCID: PMC11669356 DOI: 10.2147/pgpm.s350238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024] Open
Abstract
Background Multiple myeloma (MM) is a hematological malignancy characterized by the clonal proliferation of malignant plasma cells within the bone marrow. The disease's complexity is underpinned by a variety of genetic and molecular abnormalities that drive its progression. Methods This review was conducted through a state-of-The-art literature search, primarily utilizing PubMed to gather peer-reviewed articles. We focused on the most comprehensive and cited studies to ensure a thorough understanding of the genetic and molecular landscapes of MM. Results We detail primary and secondary alterations such as translocations, hyperdiploidy, single nucleotide variants (SNVs), copy number alterations (CNAs), gene fusions, epigenetic modifications, non-coding RNAs, germline predisposing variants, and the influence of the tumor microenvironment (TME). Our analysis highlights the heterogeneity of MM and the challenges it poses in treatment and prognosis, emphasizing the distinction between driver mutations, which actively contribute to oncogenesis, and passenger mutations, which arise due to genomic instability and do not contribute to disease progression. Conclusion & Future Perspectives We report key controversies and challenges in defining the genetic drivers of MM, and examine their implications for future therapeutic strategies. We discuss the importance of systems biology approaches in understanding the dependencies and interactions among these alterations, particularly highlighting the impact of double and triple-hit scenarios on disease outcomes. By advancing our understanding of the molecular drivers and their interactions, this review sets the stage for novel therapeutic targets and strategies, ultimately aiming to improve clinical outcomes in MM patients.
Collapse
Affiliation(s)
- Meghana Ram
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Junia Vieira dos Santos
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rafail Tasakis
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ariana Islam
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jannah Usama Abo-Donia
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samir Parekh
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alessandro Lagana
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
5
|
Jeong J, Park J, Young Mo G, Shin J, Cho Y. Structural Basis for the Recognition of GPRC5D by Talquetamab, a Bispecific Antibody for Multiple Myeloma. J Mol Biol 2024; 436:168748. [PMID: 39181182 DOI: 10.1016/j.jmb.2024.168748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
Multiple myeloma (MM) is a complex hematological malignancy characterized by abnormal antibody production from plasma cells. Despite advances in the treatment, many patients experience disease relapse or become refractory to treatment. G-protein-coupled receptor class C group 5 member D (GPRC5D), an orphan GPCR predominantly expressed in MM cells, is emerging as a promising target for MM immunotherapy. Talquetamab, a Food and Drug Administration-approved T-cell-directing bispecific antibody developed for treatment of MM, targets GPRC5D. Here, we elucidate the structure of GPRC5D complexed with the Fab fragment of talquetamab, using cryo-electron microscopy, providing the basis for recognition of GPRC5D by the bispecific antibody. GPRC5D forms a symmetric homodimer with the interface between transmembrane helix (TM) 4 of one protomer and TM4/5 of the other protomer. A single talquetamab Fab interacts with the GPRC5D dimer with its orientation toward the dimer interface. All six complementarity-determining regions of talquetamab engage with extracellular loops and TM3/5/7. In particular, the side-chain of an arginine residue from the antibody penetrates into a shallow pocket on the extracellular surface of GPRC5D. The structure offers insights for optimizing antibody design against GPRC5D for relapsed or refractory MM therapy.
Collapse
Affiliation(s)
- Jihong Jeong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Kyungbook 37673, South Korea
| | - Junhyeon Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Kyungbook 37673, South Korea
| | - Geun Young Mo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Kyungbook 37673, South Korea
| | - Jinwoo Shin
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Kyungbook 37673, South Korea
| | - Yunje Cho
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Kyungbook 37673, South Korea; Institute of Convergence Science, Yonsei University, Seoul 166-20, South Korea.
| |
Collapse
|
6
|
Trudel S, Stewart AK. A Belantamab Mafodotin Revival in Multiple Myeloma Therapy. N Engl J Med 2024; 391:461-462. [PMID: 39083777 DOI: 10.1056/nejme2406799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Affiliation(s)
- Suzanne Trudel
- From the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto
| | - A Keith Stewart
- From the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto
| |
Collapse
|
7
|
Jiang M, Li Q, Xu B. Spotlight on ideal target antigens and resistance in antibody-drug conjugates: Strategies for competitive advancement. Drug Resist Updat 2024; 75:101086. [PMID: 38677200 DOI: 10.1016/j.drup.2024.101086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/09/2024] [Accepted: 04/18/2024] [Indexed: 04/29/2024]
Abstract
Antibody-drug conjugates (ADCs) represent a novel and promising approach in targeted therapy, uniting the specificity of antibodies that recognize specific antigens with payloads, all connected by the stable linker. These conjugates combine the best targeted and cytotoxic therapies, offering the killing effect of precisely targeting specific antigens and the potent cell-killing power of small molecule drugs. The targeted approach minimizes the off-target toxicities associated with the payloads and broadens the therapeutic window, enhancing the efficacy and safety profile of cancer treatments. Within precision oncology, ADCs have garnered significant attention as a cutting-edge research area and have been approved to treat a range of malignant tumors. Correspondingly, the issue of resistance to ADCs has gradually come to the fore. Any dysfunction in the steps leading to the ADCs' action within tumor cells can lead to the development of resistance. A deeper understanding of resistance mechanisms may be crucial for developing novel ADCs and exploring combination therapy strategies, which could further enhance the clinical efficacy of ADCs in cancer treatment. This review outlines the brief historical development and mechanism of ADCs and discusses the impact of their key components on the activity of ADCs. Furthermore, it provides a detailed account of the application of ADCs with various target antigens in cancer therapy, the categorization of potential resistance mechanisms, and the current state of combination therapies. Looking forward, breakthroughs in overcoming technical barriers, selecting differentiated target antigens, and enhancing resistance management and combination therapy strategies will broaden the therapeutic indications for ADCs. These progresses are anticipated to advance cancer treatment and yield benefits for patients.
Collapse
Affiliation(s)
- Mingxia Jiang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiao Li
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Binghe Xu
- Department of Medical Oncology, State Key Laboratory of Mocelular Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
8
|
Raje NS, Cohen AD, Patel KK, van de Donk NWCJ, Richter J, San-Miguel J. Practical Aspects of Immunotherapy: A Report from the 20th International Myeloma Society (IMS) Annual Meeting. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:350-357. [PMID: 38627181 DOI: 10.1016/j.clml.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 03/06/2024] [Indexed: 06/01/2024]
Abstract
Immunotherapeutic strategies, specifically T-cell-redirected therapies, have been transformative in the context of multiple myeloma (MM). With the approval of two chimeric antigen receptor T-cell (CAR-T) drug products and three bispecific antibodies/T-cell engagers (bsAbs/TCEs) in relapsed/refractory MM (RRMM), the 20th annual IMS meeting dedicated a session to the practical aspects of these therapies. Here, we highlight the discussion during this session, including the role of CAR-T and bsAb therapies in frontline MM treatment, management of acute toxicities, prevention and management of infections, and finally treatment sequencing of T-cell redirected therapies.
Collapse
Affiliation(s)
- Noopur S Raje
- Division of Hematology/Oncology, Massachusetts General Hospital Cancer Center, Boston, MA.
| | - Adam D Cohen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Krina K Patel
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Niels W C J van de Donk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Joshua Richter
- Tisch Cancer Institute Icahn School of Medicine at Mount Sinai New York, New York, NY
| | - Jesus San-Miguel
- Cancer Center Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona, Spain
| |
Collapse
|
9
|
Wang X, Zhao A, Zhu J, Niu T. Efficacy and safety of bispecific antibodies therapy for relapsed or refractory multiple myeloma: a systematic review and meta-analysis of prospective clinical trials. Front Immunol 2024; 15:1348955. [PMID: 38482019 PMCID: PMC10933024 DOI: 10.3389/fimmu.2024.1348955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 02/13/2024] [Indexed: 04/12/2024] Open
Abstract
Objective Bispecific antibody (BsAbs) therapy represents a promising immunotherapeutic approach with manageable toxicity and noteworthy preliminary efficacy in treating patients with relapsed or refractory multiple myeloma (RRMM). The objective of this systematic review and meta-analysis was to compare the efficacy and safety of B-cell maturation antigen (BCMA)-targeted BsAbs and non-BCMA-targeted BsAbs in the treatment of RRMM patients. Methods PubMed/MEDLINE, Web of Science, EMBASE, Cochrane Library and meeting libraries were searched from inception to August 16th, 2023. The efficacy evaluation included the complete objective response rate (ORR), complete response (CR) rate, stringent CR (sCR) rate, partial response (PR) rate, and very good PR (VGPR) rate. The efficacy evaluation included any grade adverse events (AEs) and grade ≥ 3 AEs. Results Fourteen studies with a total of 1473 RRMM patients were included. The pooled ORR of the entire cohort was 61%. The non-BCMA-targeted BsAbs group displayed a higher ORR than the BCMA-targeted BsAbs group (74% vs. 54%, P < 0.01). In terms of hematological AEs, BCMA-targeted BsAbs therapy exhibited higher risks of neutropenia (any grade: 48% vs. 18%, P < 0.01; grade ≥ 3: 43% vs. 15%, P < 0.01) and lymphopenia (any grade: 37% vs. 8%, P < 0.01; grade ≥ 3: 31% vs. 8%, P = 0.07). Regarding non-hematological AEs, there were no significant differences in the risks of cytokine release syndrome (CRS, any grade: 64% vs. 66%, P = 0.84; grade ≥ 3: 1% vs. 1%, P = 0.36) and infections (any grade: 47% vs. 49%, P = 0.86; grade ≥ 3: 24% vs. 20%, P = 0.06) between the two groups. However, non-BCMA-targeted BsAbs therapy was associated with a higher risk of immune effector cell-associated neurotoxicity syndrome (ICANS, any grade: 11% vs. 2%, P < 0.01) and lower risks of fatigue (any grade: 14% vs. 30%, P < 0.01) and pyrexia (any grade: 14% vs. 29%, P < 0.01). Conclusion This analysis suggest that non-BCMA-targeted BsAbs therapy may offer a more favorable treatment response and tolerability, while BCMA-targeted BsAbs therapy may be associated with diminished neurotoxic effects. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42018090768.
Collapse
Affiliation(s)
| | - Ailin Zhao
- *Correspondence: Ting Niu, ; Ailin Zhao,
| | | | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Rodriguez-Otero P, van de Donk NWCJ, Pillarisetti K, Cornax I, Vishwamitra D, Gray K, Hilder B, Tolbert J, Renaud T, Masterson T, Heuck C, Kane C, Verona R, Moreau P, Bahlis N, Chari A. GPRC5D as a novel target for the treatment of multiple myeloma: a narrative review. Blood Cancer J 2024; 14:24. [PMID: 38307865 PMCID: PMC10837198 DOI: 10.1038/s41408-023-00966-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/29/2023] [Accepted: 12/05/2023] [Indexed: 02/04/2024] Open
Abstract
Multiple myeloma is a genetically complex and heterogenous malignancy with a 5-year survival rate of approximately 60%. Despite advances in therapy, patients experience cycles of remission and relapse, with each successive line of therapy associated with poorer outcomes; therefore, therapies with different mechanisms of action against new myeloma antigens are needed. G protein-coupled receptor class C group 5 member D (GPRC5D) has emerged as a novel therapeutic target for the treatment of multiple myeloma. We review the biology and target validation of GPRC5D, and clinical data from early phase trials of GPRC5D-targeting bispecific antibodies, talquetamab and forimtamig, and chimeric antigen receptor T cell (CAR-T) therapies, MCARH109, OriCAR-017, and BMS-986393. In addition to adverse events (AEs) associated with T-cell-redirection therapies irrespective of target, a consistent pattern of dermatologic and oral AEs has been reported across several trials of GPRC5D-targeting bispecific antibodies, as well as rare cerebellar events with CAR-T therapy. Additional studies are needed to understand the underlying mechanisms involved in the development of skin- and oral-related toxicities. We review the strategies that have been used to manage these GPRC5D-related toxicities. Preliminary efficacy data showed overall response rates for GPRC5D-targeting T-cell-redirecting therapies were ≥64%; most responders achieved a very good partial response or better. Pharmacokinetics/pharmacodynamics showed that these therapies led to cytokine release and T-cell activation. In conclusion, results from early phase trials of GPRC5D-targeting T-cell-redirecting agents have shown promising efficacy and manageable safety profiles, including lower infection rates compared with B-cell maturation antigen- and Fc receptor-like protein 5-targeting bispecific antibodies. Further clinical trials, including those investigating GPRC5D-targeting T-cell-redirecting agents in combination with other anti-myeloma therapies and with different treatment modalities, may help to elucidate the future optimal treatment regimen and sequence for patients with multiple myeloma and improve survival outcomes. Video Summary.
Collapse
Affiliation(s)
| | - Niels W C J van de Donk
- Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | - Brandi Hilder
- Janssen Research & Development, Spring House, PA, USA
| | | | | | | | | | - Colleen Kane
- Janssen Research & Development, Spring House, PA, USA
| | - Raluca Verona
- Janssen Research & Development, Spring House, PA, USA
| | | | - Nizar Bahlis
- Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Ajai Chari
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
11
|
Wang Y, Jin S, Zhuang Q, Liu N, Chen R, Adam SA, Jin J, Sun J. Chimeric antigen receptor natural killer cells: a promising antitumor immunotherapy. MedComm (Beijing) 2023; 4:e422. [PMID: 38045827 PMCID: PMC10691297 DOI: 10.1002/mco2.422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 10/05/2023] [Accepted: 10/12/2023] [Indexed: 12/05/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cells have been successfully used in adoptive cell therapy for malignancies. However, some obstacles, including side effects such as graft-versus-host disease and cytokine release syndrome, therapy resistance, limited sources, as well as high cost, limited the application of CAR T cells. Recently, CAR natural killer (NK) cells have been pursued as the effector cells for adoptive immunotherapy for their attractive merits of strong intrinsic antitumor activity and relatively mild side effects. Additionally, CAR NK cells can be available from various sources and do not require strict human leukocyte antigen matching, which suggests them as promising "off-the-shelf" products for clinical application. Although the use of CAR NK cells is restrained by the limited proliferation and impaired efficiency within the immunosuppressive tumor microenvironment, further investigation in optimizing CAR structure and combination therapies will overcome these challenges. This review will summarize the advancement of CAR NK cells, CAR NK cell manufacture, the clinical outcomes of CAR NK therapy, the challenges in the field, and prospective solutions. Besides, we will discuss the emerging application of other immune cells for CAR engineering. Collectively, this comprehensive review will provide a valuable and informative summary of current progress and evaluate challenges and future opportunities of CAR NK cells in tumor treatment.
Collapse
Affiliation(s)
- Yan Wang
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
| | - Shengjie Jin
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
| | - Qiqi Zhuang
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
| | - Na Liu
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
- Department of OncologyAffiliated Hospital of Weifang Medical UniversitySchool of Clinical MedicineWeifang Medical UniversityWeifangShandongChina
| | - Ruyi Chen
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
| | - Sofia Abdulkadir Adam
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
| | - Jie Jin
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
- Zhejiang University Cancer CenterHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Hematological DisordersHangzhouZhejiangChina
| | - Jie Sun
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Hematological DisordersHangzhouZhejiangChina
| |
Collapse
|
12
|
Abramson HN. Immunotherapy of Multiple Myeloma: Current Status as Prologue to the Future. Int J Mol Sci 2023; 24:15674. [PMID: 37958658 PMCID: PMC10649824 DOI: 10.3390/ijms242115674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/22/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
The landscape of therapeutic measures to treat multiple myeloma has undergone a seismic shift since the dawn of the current century. This has been driven largely by the introduction of new classes of small molecules, such as proteasome blockers (e.g., bortezomib) and immunomodulators (e.g., lenalidomide), as well as by immunotherapeutic agents starting with the anti-CD38 monoclonal antibody daratumumab in 2015. Recently, other immunotherapies have been added to the armamentarium of drugs available to fight this malignancy. These include the bispecifics teclistamab, talquetamab, and elranatamab, and the chimeric antigen receptor (CAR) T-cell products idecabtagene vicleucel (ide-cel) and ciltacabtagene autoleucel (cilta-cel). While the accumulated benefits of these newer agents have resulted in a more than doubling of the disease's five-year survival rate to nearly 60% and improved quality of life, the disease remains incurable, as patients become refractory to the drugs and experience relapse. This review covers the current scope of antimyeloma immunotherapeutic agents, both those in clinical use and in development. Included in the discussion are additional monoclonal antibodies (mAbs), antibody-drug conjugates (ADCs), bi- and multitargeted mAbs, and CAR T-cells and emerging natural killer (NK) cells, including products intended for "off-the-shelf" (allogeneic) applications. Emphasis is placed on the benefits of each along with the challenges that need to be surmounted if MM is to be cured.
Collapse
Affiliation(s)
- Hanley N Abramson
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
13
|
Yassine F, Kharfan-Dabaja MA, Tsalantsanis A, Roy V, Zubair AC, Murthy HS, Ayala E, Iqbal M, Sher T, Ailawadhi S, Parrondo RD. Trends in utilization of stored cryopreserved autologous peripheral hematopoietic cells intended for a second (or beyond) autologous hematopoietic cell transplantation in patients with multiple myeloma: a single center experience. Bone Marrow Transplant 2023; 58:1130-1136. [PMID: 37479753 PMCID: PMC10555832 DOI: 10.1038/s41409-023-02035-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/04/2023] [Accepted: 06/21/2023] [Indexed: 07/23/2023]
Abstract
Due to the advent of effective novel therapies for multiple myeloma (MM), the use of cryopreserved autologous peripheral blood hematopoietic cells (APBHC) for a salvage autologous transplant (auto-HCT) is in decline. We evaluated utilization trends and costs associated with cryopreserved APBHC in patients with MM. We retrospectively evaluated the clinicopathologic data from 440 patients with MM who underwent APBHC mobilization and collection at Mayo Clinic Florida between 2010 and 2019. Based on institution-specific charges as of May 2021, the cost of 1 session of APBHC collection/apheresis was $4,680 and the cost of 1 year of APBHC cryopreservation was $4,790 per patient. Out of 347 patients who had APBHC in cryopreservation, 5 (1.4%) underwent a salvage auto-HCT and 61% of patients had ≥1 excess collection sessions for APBHC that ultimately went unused. The median cost of excess collection sessions was $4,680 per patient (range, $4,680-$32,760) and the median total cost for excess collection sessions plus costs for storage was $23,840 per patient (range, $4,680-$85,450). The sum of costs of excess collection sessions was $2,077,920 and the sum of costs of cryopreservation was $5,812,665. Institutional policies regarding universal APBHC collection and long-term storage should be reevaluated in the era of novel therapeutics.
Collapse
Affiliation(s)
- Farah Yassine
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Athanasios Tsalantsanis
- Program for Comparative Effectiveness Research, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Vivek Roy
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Abba C Zubair
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Hemant S Murthy
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Ernesto Ayala
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Madiha Iqbal
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Taimur Sher
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Sikander Ailawadhi
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Ricardo D Parrondo
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
14
|
The BAFF-APRIL System in Cancer. Cancers (Basel) 2023; 15:cancers15061791. [PMID: 36980677 PMCID: PMC10046288 DOI: 10.3390/cancers15061791] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/13/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
B cell-activating factor (BAFF; also known as CD257, TNFSF13B, BLyS) and a proliferation-inducing ligand (APRIL; also known as CD256, TNFSF13) belong to the tumor necrosis factor (TNF) family. BAFF was initially discovered as a B-cell survival factor, whereas APRIL was first identified as a protein highly expressed in various cancers. These discoveries were followed by over two decades of extensive research effort, which identified overlapping signaling cascades between BAFF and APRIL, controlling immune homeostasis in health and driving pathogenesis in autoimmunity and cancer, the latter being the focus of this review. High levels of BAFF, APRIL, and their receptors have been detected in different cancers and found to be associated with disease severity and treatment response. Here, we have summarized the role of the BAFF-APRIL system in immune cell differentiation and immune tolerance and detailed its pathogenic functions in hematological and solid cancers. We also highlight the emerging therapeutics targeting the BAFF-APRIL system in different cancer types.
Collapse
|
15
|
Matula Z, Uher F, Vályi-Nagy I, Mikala G. The Effect of Belantamab Mafodotin on Primary Myeloma–Stroma Co-Cultures: Asymmetrical Mitochondrial Transfer between Myeloma Cells and Autologous Bone Marrow Stromal Cells. Int J Mol Sci 2023; 24:ijms24065303. [PMID: 36982377 PMCID: PMC10048929 DOI: 10.3390/ijms24065303] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/25/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023] Open
Abstract
Belantamab mafodotin (belamaf) is an afucosylated monoclonal antibody conjugated to the microtubule disrupter monomethyl auristatin-F (MMAF) that targets B cell maturation antigen (BCMA) on the surface of malignant plasma cells. Belamaf can eliminate myeloma cells (MMs) through several mechanisms. On the one hand, in addition to inhibiting BCMA-receptor signaling and cell survival, intracellularly released MMAF disrupts tubulin polymerization and causes cell cycle arrest. On the other hand, belamaf induces effector cell-mediated tumor cell lysis via antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis. In our in vitro co-culture model, the consequences of the first mentioned mechanism can be investigated: belamaf binds to BCMA, reduces the proliferation and survival of MMs, and then enters the lysosomes of malignant cells, where MMAF is released. The MMAF payload causes a cell cycle arrest at the DNA damage checkpoint between the G2 and M phases, resulting in caspase-3-dependent apoptosis. Here, we show that primary MMs isolated from different patients can vary widely in terms of BCMA expression level, and inadequate expression is associated with extremely high resistance to belamaf according to our cytotoxicity assay. We also reveal that primary MMs respond to increasing concentrations of belamaf by enhancing the incorporation of mitochondria from autologous bone marrow stromal cells (BM-MSCs), and as a consequence, MMs become more resistant to belamaf in this way, which is similar to other medications we have analyzed previously in this regard, such as proteasome inhibitor carfilzomib or the BCL-2 inhibitor venetoclax. The remarkable resistance against belamaf observed in the case of certain primary myeloma cell cultures is a cause for concern and points towards the use of combination therapies to overcome the risk of antigen escape.
Collapse
Affiliation(s)
- Zsolt Matula
- Laboratory for Experimental Cell Therapy, Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, 1097 Budapest, Hungary;
- Correspondence:
| | - Ferenc Uher
- Laboratory for Experimental Cell Therapy, Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, 1097 Budapest, Hungary;
| | - István Vályi-Nagy
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, 1097 Budapest, Hungary; (I.V.-N.); (G.M.)
| | - Gábor Mikala
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, 1097 Budapest, Hungary; (I.V.-N.); (G.M.)
| |
Collapse
|