1
|
De Novellis D, Derudas D, Vincelli D, Fontana R, Della Pepa R, Palmieri S, Accardi F, Rotondo F, Morelli E, Gigliotta E, Roccotelli D, Marano L, Barone ML, Cetani G, Esposito D, Lazzaro A, Delle Cave G, Serio B, Morini D, Porrazzo M, Urciuoli E, Masucci C, Fanelli F, Rizzo M, Arcamone M, Trastulli F, Rocco S, Leone A, Bianco R, Salvatore F, Idato A, Sicari M, Tosi P, Rascato MG, Di Perna M, Falcone AP, Morello L, Carlisi M, Svanera G, Annunziata M, Frigeri F, Califano C, Carella AM, Marcacci G, Pane F, Risitano AM, Giudice V, Botta C, Selleri C. Clinical Efficacy of Isatuximab Plus Carfilzomib and Dexamethasone in Relapsed/Refractory Multiple Myeloma Patients. Eur J Haematol 2025; 114:105-114. [PMID: 39370303 PMCID: PMC11613624 DOI: 10.1111/ejh.14314] [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: 07/23/2024] [Revised: 09/09/2024] [Accepted: 09/18/2024] [Indexed: 10/08/2024]
Abstract
Isatuximab, a novel anti-CD38 monoclonal antibody, is approved in combination with carfilzomib and dexamethasone (Isa-Kd) in relapsed/refractory multiple myeloma (RRMM) patients. Because of its recent introduction, real-world efficacy and safety are poorly reported. In this Italian multicenter real-life observational retrospective study, efficacy and safety of the Isa-Kd regimen were evaluated in a cohort of 103 RRMM patients. Overall response rate (ORR) was 85%, with stringent (sCR) or complete response (CR) in 18% of cases and very good partial response (VGPR) in 39%. Median PFS and OS were not reached within the study period, while 1-year PFS and OS were 72% and 77%, respectively. Hematological toxicities were observed in 42% of subjects, and cardiac toxicities occurred in 24% of cases. Moreover, we conducted a subanalysis on patients (N = 69) treated with Isa-Kd after one prior line of therapy, showing an ORR of 88%, with sCR + CR in 20% of subjects, VGPR in 46%, and PR in 22% of patients. In this group, median PFS and OS were not reached, while 1-year PFS and OS were 92% and 95%, respectively. In conclusions, our study confirmed Isa-Kd as an effective treatment option for RRMM with a manageable safety profile even in real-life settings.
Collapse
Affiliation(s)
- Danilo De Novellis
- Hematology and Transplant CenterUniversity Hospital "San Giovanni di Dio e Ruggi d'Aragona"SalernoItaly
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana"University of SalernoBaronissiItaly
| | - Daniele Derudas
- S.C. di Ematologia e C.T.M.O.Ospedale Oncologico di Riferimento Regionale "A. Businco"CagliariItaly
| | - Donatella Vincelli
- Division of HematologyAzienda Ospedaliera "Bianchi Melacrino Morelli"Reggio CalabriaItaly
| | - Raffaele Fontana
- Hematology and Transplant CenterUniversity Hospital "San Giovanni di Dio e Ruggi d'Aragona"SalernoItaly
| | - Roberta Della Pepa
- Hematology – Department of Clinical Medicine and SurgeryUniversity Hospital “Federico II”NaplesItaly
| | | | - Fabrizio Accardi
- U.O.C di OncoematologiaAzienda Ospedaliera Ospedali Riuniti Villa Sofia – CervelloPalermoItaly
| | | | - Emanuela Morelli
- Hematology and Transplant Center, Istituto Nazionale TumoriFondazione “G.Pascale”, IRRCSNaplesItaly
| | - Emilia Gigliotta
- Dipartimento di Promozione della Salute, Materno‐infantile, di medicina interna e specialistica d'eccellenza “G. D'alessandro”Università di PalermoPalermoItaly
| | - Daniela Roccotelli
- Division of Hematology and Stem Cell UnitIRCCS S. Giovanni Rotondo and Division of HematologySan Giovanni RotondoItaly
| | - Luana Marano
- Hematology, Hospital “San Giuseppe Moscati”AvellinoItaly
| | | | - Giusy Cetani
- Hematology, Hospital “Sant'Anna e San Sebastiano”CasertaItaly
| | | | - Antonio Lazzaro
- UO di Ematologia e Centro Trapianti Midollo OsseoDipartimento di Oncoematologia Azienda Usl di PiacenzaPiacenzaItaly
| | | | - Bianca Serio
- Hematology and Transplant CenterUniversity Hospital "San Giovanni di Dio e Ruggi d'Aragona"SalernoItaly
| | - Denise Morini
- Hematology and Transplant CenterUniversity Hospital "San Giovanni di Dio e Ruggi d'Aragona"SalernoItaly
| | - Marika Porrazzo
- U.O.C di OncoematologiaAzienda Ospedaliera Ospedali Riuniti Villa Sofia – CervelloPalermoItaly
| | | | - Chiara Masucci
- Hematology, Hospital “San Giuseppe Moscati”AvellinoItaly
| | - Fulvia Fanelli
- Hematology, Hospital “San Giuseppe Moscati”AvellinoItaly
| | - Michela Rizzo
- Hematology and Transplant CenterUniversity Hospital "San Giovanni di Dio e Ruggi d'Aragona"SalernoItaly
| | - Manuela Arcamone
- Hematology and Transplant Center, Istituto Nazionale TumoriFondazione “G.Pascale”, IRRCSNaplesItaly
| | | | - Stefano Rocco
- Hematology, Hospital “Antonio Cardarelli”NaplesItaly
| | - Aldo Leone
- Hematology – Department of Clinical Medicine and SurgeryUniversity Hospital “Federico II”NaplesItaly
| | - Rosario Bianco
- Hematology, Hospital “Sant'Anna e San Sebastiano”CasertaItaly
| | | | - Aurora Idato
- Division of HematologyAzienda Ospedaliera "Bianchi Melacrino Morelli"Reggio CalabriaItaly
| | - Maria Sicari
- Division of HematologyAzienda Ospedaliera "Bianchi Melacrino Morelli"Reggio CalabriaItaly
| | | | | | | | - Antonietta Pia Falcone
- Division of Hematology and Stem Cell UnitIRCCS S. Giovanni Rotondo and Division of HematologySan Giovanni RotondoItaly
| | - Lucia Morello
- UO di Ematologia e Centro Trapianti Midollo OsseoDipartimento di Oncoematologia Azienda Usl di PiacenzaPiacenzaItaly
| | - Melania Carlisi
- Dipartimento di Promozione della Salute, Materno‐infantile, di medicina interna e specialistica d'eccellenza “G. D'alessandro”Università di PalermoPalermoItaly
| | - Gino Svanera
- Hematology, Hospital “San Giuliano”GiuglianoItaly
| | | | | | | | - Angelo Michele Carella
- Division of Hematology and Stem Cell UnitIRCCS S. Giovanni Rotondo and Division of HematologySan Giovanni RotondoItaly
| | - Gianpaolo Marcacci
- Hematology and Transplant Center, Istituto Nazionale TumoriFondazione “G.Pascale”, IRRCSNaplesItaly
| | - Fabrizio Pane
- Hematology – Department of Clinical Medicine and SurgeryUniversity Hospital “Federico II”NaplesItaly
| | | | - Valentina Giudice
- Hematology and Transplant CenterUniversity Hospital "San Giovanni di Dio e Ruggi d'Aragona"SalernoItaly
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana"University of SalernoBaronissiItaly
| | - Ciro Botta
- Dipartimento di Promozione della Salute, Materno‐infantile, di medicina interna e specialistica d'eccellenza “G. D'alessandro”Università di PalermoPalermoItaly
| | - Carmine Selleri
- Hematology and Transplant CenterUniversity Hospital "San Giovanni di Dio e Ruggi d'Aragona"SalernoItaly
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana"University of SalernoBaronissiItaly
| |
Collapse
|
2
|
Pu J, Liu T, Sharma A, Jiang L, Wei F, Ren X, Schmidt-Wolf IGH, Hou J. Advances in adoptive cellular immunotherapy and therapeutic breakthroughs in multiple myeloma. Exp Hematol Oncol 2024; 13:105. [PMID: 39468695 PMCID: PMC11514856 DOI: 10.1186/s40164-024-00576-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/14/2024] [Indexed: 10/30/2024] Open
Abstract
The basic idea of modulating the immune system to better recognize and fight tumor cells has led to the successful introduction of adoptive cellular immunotherapy (ACT). ACT-based treatment regimens, in which the patient's own immune cells are isolated and subsequently expanded (ex vivo) and reinfused, have also contributed significantly to the development of a personalized treatment strategy. Complementing this, the unprecedented advances in ACTs as chimeric antigen receptor (CAR)-T cell therapies and their derivatives such as CAR-NK, CAR-macrophages, CAR-γδT and CAR-NKT have further maximized the therapeutic outcomes. Herein, we provide a comprehensive overview of the development of ACTs in multiple myeloma (MM) and outline how they have evolved from an experimental form to a mainstay of standard clinical settings. Besides, we provide insights into cytokine-induced killer cell (CIK) therapy, an alternative form of ACT that (as CIK or CAR-CIK) has enormous potential in the clinical spectrum of MM. We also summarize the results of the major preclinical and clinical studies of adoptive cell therapy in MM and address the current challenges (such as cytokine release syndrome (CRS) and neurotoxicity) that limit its complete success in the cancer landscape.
Collapse
Affiliation(s)
- Jingjing Pu
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Ting Liu
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, NRW, Germany
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany
| | - Liping Jiang
- Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China
| | - Feng Wei
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300070, China
| | - Xiubao Ren
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300070, China.
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany.
| | - Jian Hou
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
3
|
Zhang Y, Xie J, Wu H, Huang J, Zheng D, Wang S, Jia X, He Z, Gong Y, Ju L, Sun Q. NK cell based immunotherapy against oral squamous cell carcinoma. Front Immunol 2024; 15:1440764. [PMID: 39192980 PMCID: PMC11347299 DOI: 10.3389/fimmu.2024.1440764] [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: 05/30/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC), a major subtype of head and neck cancers, presents significant challenges due to its aggressive feature and limited therapeutic efficacy of conventional treatments. In response to these challenges, Natural Killer (NK) cells, a vital component of the innate immune system, are being explored for their therapeutic potential in OSCC due to their inherent ability to target and eliminate cancer cells without prior sensitization. This review uniquely focuses on the evolving role of NK cells specifically in OSCC, incorporating recent advancements in CAR-NK cell engineering and personalized therapy approaches that have not been comprehensively covered in previous reviews. The mechanisms through which NK cells exert cytotoxic effects on tumor cells include direct killing through the engagement of natural cytotoxic receptors and antibody-dependent cellular cytotoxicity (ADCC), making them promising agents in cancer immunotherapy. Additionally, the article explores recent advancements in engineering NK cells to enhance their antitumor activity, such as the modification with chimeric antigen receptors (CARs) to target specific tumor antigens. Clinical implications of NK cell-based therapies, including the challenges of integrating these treatments with existing protocols and the potential for personalized therapy, are examined. The review highlights the promise of NK cell therapies in improving outcomes for OSCC patients and outlines future directions for research in this dynamic field of oncological immunotherapy.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Stomatology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jianming Xie
- Department of Otolaryngology & Head and Neck Surgery, Anyuan People’s hospital, Ganzhou, China
| | - Haoran Wu
- Southern Medical University, Guangzhou, Guangdong, China
| | - Jinhui Huang
- Southern Medical University, Guangzhou, Guangdong, China
| | - Danna Zheng
- Southern Medical University, Guangzhou, Guangdong, China
| | - Shaotong Wang
- Southern Medical University, Guangzhou, Guangdong, China
| | - Xueqiang Jia
- Southern Medical University, Guangzhou, Guangdong, China
| | - Zongzhong He
- Department of Transfusion Medicine, General Hospital of Southern Theatre Command, Guangzhou, Guangdong, China
| | - Ying Gong
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Linling Ju
- Medical School of Nantong University, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Qiurong Sun
- Department of Stomatology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
4
|
Olejarz W, Basak G. Emerging Therapeutic Targets and Drug Resistance Mechanisms in Immunotherapy of Hematological Malignancies. Cancers (Basel) 2023; 15:5765. [PMID: 38136311 PMCID: PMC10741639 DOI: 10.3390/cancers15245765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/22/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
CAR-T cell therapy has revolutionized the treatment of hematological malignancies with high remission rates in the case of ALL and NHL. This therapy has some limitations such as long manufacturing periods, persistent restricted cell sources and high costs. Moreover, combination regimens increase the risk of immune-related adverse events, so the identification new therapeutic targets is important to minimize the risk of toxicities and to guide more effective approaches. Cancer cells employ several mechanisms to evade immunosurveillance, which causes resistance to immunotherapy; therefore, a very important therapeutic approach is to focus on the development of rational combinations of targeted therapies with non-overlapping toxicities. Recent progress in the development of new inhibitory clusters of differentiation (CDs), signaling pathway molecules, checkpoint inhibitors, and immunosuppressive cell subsets and factors in the tumor microenvironment (TME) has significantly improved anticancer responses. Novel strategies regarding combination immunotherapies with CAR-T cells are the most promising approach to cure cancer.
Collapse
Affiliation(s)
- Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
- Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Grzegorz Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland;
| |
Collapse
|
5
|
Fazio M, Del Fabro V, Parrinello NL, Allegra A, Markovic U, Botta C, Accardi F, Vincelli ID, Leotta S, Elia F, Esposito B, Garibaldi B, Sapuppo G, Orofino A, Romano A, Palumbo GA, Di Raimondo F, Conticello C. Multiple Myeloma in 2023 Ways: From Trials to Real Life. Curr Oncol 2023; 30:9710-9733. [PMID: 37999125 PMCID: PMC10670159 DOI: 10.3390/curroncol30110705] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/25/2023] Open
Abstract
Multiple myeloma is a chronic hematologic malignancy that obstinately tends to relapse. Basic research has made giant strides in better characterizing the molecular mechanisms of the disease. The results have led to the manufacturing of new, revolutionary drugs which have been widely tested in clinical trials. These drugs have been approved and are now part of the therapeutic armamentarium. As a consequence, it is essential to combine what we know from clinical trials with real-world data in order to improve therapeutic strategies. Starting with this premise, our review aims to describe the currently employed regimens in multiple myeloma and compare clinical trials with real-life experiences. We also intend to put a spotlight on promising therapies such as T-cell engagers and chimeric antigen receptor T-cells (CAR-T) which are proving to be effective in changing the course of advanced-stage disease.
Collapse
Affiliation(s)
- Manlio Fazio
- Post-Graduation School of Haematology, University of Catania, A.O.U. ‘Policlinico-San Marco’, 95123 Catania, Italy; (M.F.); (B.E.); (B.G.); (G.S.); (A.O.); (F.D.R.)
| | - Vittorio Del Fabro
- Division of Haematology and BMT, A.O.U. ‘Policlinico-San Marco’, 95123 Catania, Italy; (V.D.F.); (N.L.P.); (U.M.); (S.L.); (F.E.)
| | - Nunziatina Laura Parrinello
- Division of Haematology and BMT, A.O.U. ‘Policlinico-San Marco’, 95123 Catania, Italy; (V.D.F.); (N.L.P.); (U.M.); (S.L.); (F.E.)
| | - Alessandro Allegra
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Uroš Markovic
- Division of Haematology and BMT, A.O.U. ‘Policlinico-San Marco’, 95123 Catania, Italy; (V.D.F.); (N.L.P.); (U.M.); (S.L.); (F.E.)
| | - Cirino Botta
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy;
| | - Fabrizio Accardi
- Department of Hematology I, Azienda Ospedaliera Ospedali Riuniti Villa Sofia-Cervello, 90146 Palermo, Italy;
| | - Iolanda Donatella Vincelli
- Haematology Unit, Haemato-Oncology and Radiotherapy Department, Grande Ospedale Metropolitano “Bianchi-Melacrino-Morelli”, 89124 Reggio Calabria, Italy;
| | - Salvatore Leotta
- Division of Haematology and BMT, A.O.U. ‘Policlinico-San Marco’, 95123 Catania, Italy; (V.D.F.); (N.L.P.); (U.M.); (S.L.); (F.E.)
| | - Federica Elia
- Division of Haematology and BMT, A.O.U. ‘Policlinico-San Marco’, 95123 Catania, Italy; (V.D.F.); (N.L.P.); (U.M.); (S.L.); (F.E.)
| | - Benedetta Esposito
- Post-Graduation School of Haematology, University of Catania, A.O.U. ‘Policlinico-San Marco’, 95123 Catania, Italy; (M.F.); (B.E.); (B.G.); (G.S.); (A.O.); (F.D.R.)
| | - Bruno Garibaldi
- Post-Graduation School of Haematology, University of Catania, A.O.U. ‘Policlinico-San Marco’, 95123 Catania, Italy; (M.F.); (B.E.); (B.G.); (G.S.); (A.O.); (F.D.R.)
| | - Gabriele Sapuppo
- Post-Graduation School of Haematology, University of Catania, A.O.U. ‘Policlinico-San Marco’, 95123 Catania, Italy; (M.F.); (B.E.); (B.G.); (G.S.); (A.O.); (F.D.R.)
| | - Alessandra Orofino
- Post-Graduation School of Haematology, University of Catania, A.O.U. ‘Policlinico-San Marco’, 95123 Catania, Italy; (M.F.); (B.E.); (B.G.); (G.S.); (A.O.); (F.D.R.)
| | - Alessandra Romano
- Dipartimento di Specialità Medico-Chirurgiche, CHIRMED, Sezione di Ematologia, Università degli Studi di Catania, 95131 Catania, Italy;
| | - Giuseppe A. Palumbo
- Dipartimento di Scienze Mediche, Chirurgiche e Tecnologie Avanzate “G.F.Ingrassia”, University of Catania, 95131 Catania, Italy;
| | - Francesco Di Raimondo
- Post-Graduation School of Haematology, University of Catania, A.O.U. ‘Policlinico-San Marco’, 95123 Catania, Italy; (M.F.); (B.E.); (B.G.); (G.S.); (A.O.); (F.D.R.)
- Division of Haematology and BMT, A.O.U. ‘Policlinico-San Marco’, 95123 Catania, Italy; (V.D.F.); (N.L.P.); (U.M.); (S.L.); (F.E.)
- Dipartimento di Specialità Medico-Chirurgiche, CHIRMED, Sezione di Ematologia, Università degli Studi di Catania, 95131 Catania, Italy;
| | - Concetta Conticello
- Division of Haematology and BMT, A.O.U. ‘Policlinico-San Marco’, 95123 Catania, Italy; (V.D.F.); (N.L.P.); (U.M.); (S.L.); (F.E.)
| |
Collapse
|
6
|
Yang J, Zhou W, Li D, Niu T, Wang W. BCMA-targeting chimeric antigen receptor T-cell therapy for multiple myeloma. Cancer Lett 2023; 553:215949. [PMID: 36216149 DOI: 10.1016/j.canlet.2022.215949] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 11/24/2022]
Abstract
Multiple myeloma (MM) remains an incurable hematologic malignancy, despite the development of numerous innovative therapies during the past two decades. Immunotherapies are changing the treatment paradigm of MM and have improved the overall response and survival of patients with relapsed/refractory (RR) MM. B cell maturation antigen (BCMA), selectively expressed in normal and malignant plasma cells, has been targeted by several immunotherapeutic modalities. Chimeric antigen receptor (CAR) T cells, the breakthrough in cancer immunotherapy, have revolutionized the treatment of B cell malignancies and remarkably improved the prognosis of RRMM. BCMA-targeting CAR T cell therapy is the most developed CAR T cell therapy for MM, and the US Food and Drug Administration has already approved idecabtagene vicleucel (Ide-cel) and ciltacabtagene autoleucel (Cilta-cel) for MM. However, the development of novel BCMA-targeting CAR T cell therapies remains in progress. This review focuses on BCMA-targeting CAR T cell therapy, covering all stages of investigational progress, including the innovative preclinical studies, the initial phase I clinical trials, and the more developed phase II clinical trials. It also discusses possible measures to improve the efficacy and safety of this therapy.
Collapse
Affiliation(s)
- Jinrong Yang
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041, China
| | - Weilin Zhou
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041, China
| | - Dan Li
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041, China
| | - Ting Niu
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041, China.
| | - Wei Wang
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041, China.
| |
Collapse
|
7
|
De Novellis D, Fontana R, Giudice V, Serio B, Selleri C. Innovative Anti-CD38 and Anti-BCMA Targeted Therapies in Multiple Myeloma: Mechanisms of Action and Resistance. Int J Mol Sci 2022; 24:645. [PMID: 36614086 PMCID: PMC9820921 DOI: 10.3390/ijms24010645] [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: 12/05/2022] [Revised: 12/25/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
CD38 and B-cell maturation antigens (BCMAs) are prevalently expressed on neoplastic plasma cells in multiple myeloma (MM), making them ideal therapeutic targets. Anti-CD38 monoclonal antibodies, such as approved daratumumab and isatuximab, are currently the milestone in MM treatment because they induce plasma cell apoptosis and kill through several mechanisms, including antibody-dependent cellular cytotoxicity or phagocytosis. BCMA is considered an excellent target in MM, and three different therapeutic strategies are either already available in clinical practice or under investigation: antibody-drug conjugates, such as belantamab-mafodotin; bispecific T cell engagers; and chimeric antigen receptor-modified T cell therapies. Despite the impressive clinical efficacy of these new strategies in the treatment of newly diagnosed or multi-refractory MM patients, several mechanisms of resistance have already been described, including antigen downregulation, the impairment of antibody-dependent cell cytotoxicity and phagocytosis, T- and natural killer cell senescence, and exhaustion. In this review, we summarize the current knowledge on the mechanisms of action and resistance of anti-CD38 and anti-BCMA agents and their clinical efficacy and safety.
Collapse
Affiliation(s)
- Danilo De Novellis
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Raffaele Fontana
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Valentina Giudice
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Bianca Serio
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| |
Collapse
|
8
|
Wang Z, Chen C, Wang L, Jia Y, Qin Y. Chimeric antigen receptor T-cell therapy for multiple myeloma. Front Immunol 2022; 13:1050522. [PMID: 36618390 PMCID: PMC9814974 DOI: 10.3389/fimmu.2022.1050522] [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: 09/21/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Multiple myeloma (MM) is a malignant plasma cell disorder that remains incurable for most patients, as persistent clonal evolution drives new mutations which confer MM high-risk signatures and resistance to standard care. The past two decades have significantly refashioned the therapeutic options for MM, especially adoptive T cell therapy contributing to impressive response rate and clinical efficacy. Despite great promises achieved from chimeric antigen receptor T-cell (CAR-T) therapy, the poor durability and severe toxicity (cytokine release syndrome and neurotoxicity) are still huge challenges. Therefore, relapsed/refractory multiple myeloma (RRMM), characterized by the nature of clinicopathologic and molecular heterogeneity, is frequently associated with poor prognosis. B Cell Maturation Antigen (BCMA) is the most successful target for CAR-T therapy, and other potential targets either for single-target or dual-target CAR-T are actively being studied in numerous clinical trials. Moreover, mechanisms driving resistance or relapse after CAR-T therapy remain uncharacterized, which might refer to T-cell clearance, antigen escape, and immunosuppressive tumor microenvironment. Engineering CAR T-cell to improve both efficacy and safety continues to be a promising area for investigation. In this review, we aim to describe novel tumor-associated neoantigens for MM, summarize the data from current MM CAR-T clinical trials, introduce the mechanism of disease resistance/relapse after CAR-T infusion, highlight innovations capable of enhanced efficacy and reduced toxicity, and provide potential directions to optimize manufacturing processes.
Collapse
Affiliation(s)
| | | | | | - Yongxu Jia
- *Correspondence: Yongxu Jia, ; Yanru Qin,
| | - Yanru Qin
- *Correspondence: Yongxu Jia, ; Yanru Qin,
| |
Collapse
|
9
|
Gribkova IV. CAR NK-сells for the treatment of hematological malignancies: A review. JOURNAL OF MODERN ONCOLOGY 2022. [DOI: 10.26442/18151434.2022.3.201699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hematological malignant neoplasms include more than a hundred different subtypes and account for about 4.8% of all neoplastic diseases in Russia. Despite significant advances in diagnosis and treatment, many of them remain incurable. In recent years, cell-based therapy appears to be a promising approach to the treatment of these incurable hematologic malignancies, showing striking results in various clinical trials. The most studied and advanced cell therapy is the therapy with T-lymphocytes modified with chimeric antigen receptors (CAR). However, although the US Food and Drug Administration has approved CAR T cells for the treatment of B-cell lymphoma and acute lymphoblastic leukemia, significant problems remain in terms of production, cost, and serious side effects. An alternative to the use of T cells can be the use of innate immune cells, in particular natural killer cells (NK), which have a high antitumor potential. Recent studies have shown the antitumor efficacy of a therapy that uses genetically modified natural killer cells CAR NK cells. The purpose of this review was to describe and systematize the experience of using CAR NK cells for the treatment of hematological neoplasms. The review presents the advantages and disadvantages of this method, as well as the problems that still have to be solved for its widespread introduction into clinical practice.
Collapse
|
10
|
Tu Y, Yao Z, Yang W, Tao S, Li B, Wang Y, Su Z, Li S. Application of Nanoparticles in Tumour Targeted Drug Delivery and Vaccine. FRONTIERS IN NANOTECHNOLOGY 2022. [DOI: 10.3389/fnano.2022.948705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cancer is a major cause of death worldwide, and nearly 1 in 6 deaths each year is caused by cancer. Traditional cancer treatment strategies cannot completely solve cancer recurrence and metastasis. With the development of nanotechnology, the study of nanoparticles (NPs) has gradually become a hotspot of medical research. NPs have various advantages. NPs exploit the enhanced permeability and retention (EPR) of tumour cells to achieve targeted drug delivery and can be retained in tumours long-term. NPs can be used as a powerful design platform for vaccines as well as immunization enhancers. Liposomes, as organic nanomaterials, are widely used in the preparation of nanodrugs and vaccines. Currently, most of the anticancer drugs that have been approved and entered clinical practice are prepared from lipid materials. However, the current clinical conversion rate of NPs is still extremely low, and the transition of NPs from the laboratory to clinical practice is still a substantial challenge. In this paper, we review the in vivo targeted delivery methods, material characteristics of NPs and the application of NPs in vaccine preparation. The application of nanoliposomes is also emphasized. Furthermore, the challenges and limitations of NPs are briefly discussed.
Collapse
|
11
|
Yang P, Qu Y, Wang M, Chu B, Chen W, Zheng Y, Niu T, Qian Z. Pathogenesis and treatment of multiple myeloma. MedComm (Beijing) 2022; 3:e146. [PMID: 35665368 PMCID: PMC9162151 DOI: 10.1002/mco2.146] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 02/05/2023] Open
Abstract
Multiple myeloma (MM) is the second‐ranking malignancy in hematological tumors. The pathogenesis of MM is complex with high heterogeneity, and the development of the disease is a multistep process. Chromosomal translocations, aneuploidy, genetic mutations, and epigenetic aberrations are essential in disease initiation and progression. The correlation between MM cells and the bone marrow microenvironment is associated with the survival, progression, migration, and drug resistance of MM cells. In recent decades, there has been a significant change in the paradigm for the management of MM. With the development of proteasome inhibitors, immunomodulatory drugs, monoclonal antibodies, chimeric antigen receptor T‐cell therapies, and novel agents, the survival of MM patients has been significantly improved. In addition, nanotechnology acts as both a nanocarrier and a treatment tool for MM. The properties and responsive conditions of nanomedicine can be tailored to reach different goals. Nanomedicine with a precise targeting property has offered great potential for drug delivery and assisted in tumor immunotherapy. In this review, we summarize the pathogenesis and current treatment options of MM, then overview recent advances in nanomedicine‐based systems, aiming to provide more insights into the treatment of MM.
Collapse
Affiliation(s)
- Peipei Yang
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan China
| | - Ying Qu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan China
| | - Mengyao Wang
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan China
| | - Bingyang Chu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan China
| | - Wen Chen
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan China
| | - Yuhuan Zheng
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan China
| | - Ting Niu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan China
| | - Zhiyong Qian
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan China
| |
Collapse
|
12
|
Choi T, Kang Y. Chimeric antigen receptor (CAR) T-cell therapy for multiple myeloma. Pharmacol Ther 2022; 232:108007. [PMID: 34582835 PMCID: PMC8930424 DOI: 10.1016/j.pharmthera.2021.108007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/23/2022]
Abstract
Although treatment outcomes of multiple myeloma patients have improved significantly during the last two decades, myeloma is still an incurable disease. There are newly emerging immunotherapies to treat multiple myeloma including monoclonal antibodies, antibody-drug conjugate, bispecific antibodies, and chimeric antigen receptor (CAR) T cell therapy. Impressive response rate and clinical efficacy in heavily pretreated myeloma patients led to the FDA approval of the first myeloma CAR-T therapy in March 2021. Among many different targets for myeloma CAR-T therapies, B Cell Maturation Antigen (BCMA) has been the most successful target so far, but other targets which can be used either for single-target or dual-target CAR-T's are actively being explored. Clinical efficacy and safety of current myeloma CAR-T therapies will be presented here. Potential mechanisms leading to resistance include clearance of CAR-T cells, antigenic escape, and immunosuppressive tumor microenvironment. Novel strategies to enhance myeloma CAR-T will also be described. In this article, we provide a comprehensive review of the current data and the future directions of myeloma CAR-T therapies.
Collapse
Affiliation(s)
- Taewoong Choi
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC, USA
| | - Yubin Kang
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
13
|
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. [PMID: 35320942 PMCID: PMC8936073 DOI: 10.3389/fimmu.2022.839097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/07/2022] [Indexed: 11/24/2022] Open
Abstract
With the gradual improvement of treatment regimens, the survival time of multiple myeloma (MM) patients has been significantly prolonged. Even so, MM is still a nightmare with an inferior prognosis. B-cell maturation antigen (BCMA) is highly expressed on the surface of malignant myeloma cells. For the past few years, significant progress has been made in various BCMA-targeted immunotherapies for treating patients with RRMM, including anti-BCMA mAbs, antibody-drug conjugates, bispecific T-cell engagers, and BCMA-targeted adoptive cell therapy like chimeric antigen receptor (CAR)-T cell. The 63rd annual meeting of the American Society of Hematology updated some information about the application of BCMA in MM. This review summarizes part of the related points presented at this conference.
Collapse
Affiliation(s)
- Ruiting Guo
- First Center Clinic College of Tianjin Medical University, Tianjin, China
| | - Wenyi Lu
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Yi Zhang
- First Center Clinic College of Tianjin Medical University, Tianjin, China
| | - Xinping Cao
- First Center Clinic College of Tianjin Medical University, Tianjin, China
| | - Xin Jin
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
14
|
Baum N, Eggers M, Koenigsdorf J, Menzel S, Hambach J, Staehler T, Fliegert R, Kulow F, Adam G, Haag F, Bannas P, Koch-Nolte F. Mouse CD38-Specific Heavy Chain Antibodies Inhibit CD38 GDPR-Cyclase Activity and Mediate Cytotoxicity Against Tumor Cells. Front Immunol 2021; 12:703574. [PMID: 34539634 PMCID: PMC8446682 DOI: 10.3389/fimmu.2021.703574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/04/2021] [Indexed: 12/24/2022] Open
Abstract
CD38 is the major NAD+-hydrolyzing ecto-enzyme in most mammals. As a type II transmembrane protein, CD38 is also a promising target for the immunotherapy of multiple myeloma (MM). Nanobodies are single immunoglobulin variable domains from heavy chain antibodies that naturally occur in camelids. Using phage display technology, we isolated 13 mouse CD38-specific nanobodies from immunized llamas and produced these as recombinant chimeric mouse IgG2a heavy chain antibodies (hcAbs). Sequence analysis assigned these hcAbs to five distinct families that bind to three non-overlapping epitopes of CD38. Members of families 4 and 5 inhibit the GDPR-cyclase activity of CD38. Members of families 2, 4 and 5 effectively induce complement-dependent cytotoxicity against CD38-expressing tumor cell lines, while all families effectively induce antibody dependent cellular cytotoxicity. Our hcAbs present unique tools to assess cytotoxicity mechanisms of CD38-specific hcAbs in vivo against tumor cells and potential off-target effects on normal cells expressing CD38 in syngeneic mouse tumor models, i.e. in a fully immunocompetent background.
Collapse
Affiliation(s)
- Natalie Baum
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marie Eggers
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia Koenigsdorf
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Radiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephan Menzel
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia Hambach
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Radiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Staehler
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Fliegert
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frederike Kulow
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerhard Adam
- Department of Radiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Bannas
- Department of Radiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
15
|
Ye W, Wu X, Liu X, Zheng X, Deng J, Gong Y. Comparison of monoclonal antibodies targeting CD38, SLAMF7 and PD-1/PD-L1 in combination with Bortezomib/Immunomodulators plus dexamethasone/prednisone for the treatment of multiple myeloma: an indirect-comparison Meta-analysis of randomised controlled trials. BMC Cancer 2021; 21:994. [PMID: 34488679 PMCID: PMC8419924 DOI: 10.1186/s12885-021-08588-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 07/05/2021] [Indexed: 02/08/2023] Open
Abstract
Background Many clinical trials have assessed the effect and safety of monoclonal antibodies (MAbs) in combination with proteasome inhibitors or immunomodulators plus dexamethasone/prednisone for the treatment of multiple myeloma (MM). The treatment outcomes of comparing different MAbs in combination with the above-mentioned agents remained unclear. We performed the meta-analysis to indirectly compare the effect and safety of MAbs targeting CD38, SLAMF7, and PD-1/PD-L1 in combination with bortezomib/immunomodulators plus dexamethasone/prednisone for patients with MM. Methods We searched thoroughly in the databases for randomised controlled trials (RCTs) in which at least one of the three MAbs were included. We included eleven eligible RCTs with 5367 patients in the meta-analysis. Statistical analysis was carried out using StataMP14 and Indirect Treatment Comparisons software. Results We calculated hazard ratios (HRs) for overall survival (OS) and progression-free survival (PFS) and relative risk (RR) for overall response rate, complete response (CR) or better, very good partial response (VGPR) or better, VGPR, partial response, stable disease, and grade 3 or higher adverse events among the three groups. The HRs for PFS of the CD38 group vs SLAMF7 group, CD38 group vs PD-1/PD-L1 group, and SLAMF7 group vs PD-1/PD-L1 group were 0.662 (95%CI 0.543–0.806), 0.317 (95%CI 0.221–0.454), and 0.479 (95%CI 0.328–0.699), respectively. The HR for OS of the CD38 group vs SLAMF7 group was 0.812 (95%CI 0.584–1.127). The RR for CR or better in the CD38 group vs SLAMF7 group was 2.253 (95%CI 1.284–3.955). The RR for neutropenia of the CD38 group vs SLAMF7 group was 1.818 (95%CI 1.41–2.344). Conclusions Treatment with the CD38 group had longer PFS and better treatment response than that with the SLAMF7 or PD-1/PD-L1 group. In addition, the SLAMF7 group prolonged PFS compared with the PD-1/PD-L1 group and was associated with a lower incidence of grade 3 or higher neutropenia than the CD38 and PD-1/PD-L1 group. In conclusion, MAbs targeting CD38 are the best, followed by those targeting SLAMF7; MAbs targeting PD-1/PD-L1 are the worst when in combination with bortezomib/immunomodulators plus dexamethasone/prednisone for the treatment of MM. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08588-9.
Collapse
Affiliation(s)
- Wu Ye
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, No.37 GuoXue Xiang, Chengdu, 610041, Sichuan Province, China
| | - Xia Wu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, No.37 GuoXue Xiang, Chengdu, 610041, Sichuan Province, China
| | - Xiaoyan Liu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, No.37 GuoXue Xiang, Chengdu, 610041, Sichuan Province, China
| | - Xue Zheng
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, No.37 GuoXue Xiang, Chengdu, 610041, Sichuan Province, China
| | - Jili Deng
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, No.37 GuoXue Xiang, Chengdu, 610041, Sichuan Province, China
| | - Yuping Gong
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, No.37 GuoXue Xiang, Chengdu, 610041, Sichuan Province, China.
| |
Collapse
|
16
|
Feng Y, Liu X, Li X, Zhou Y, Song Z, Zhang J, Shi B, Wang J. Novel BCMA-OR-CD38 tandem-dual chimeric antigen receptor T cells robustly control multiple myeloma. Oncoimmunology 2021; 10:1959102. [PMID: 34434610 PMCID: PMC8381848 DOI: 10.1080/2162402x.2021.1959102] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BCMA-targeting chimeric antigen receptor (CAR)-T cell therapy has shown remarkable clinical efficacy against multiple myeloma, yet antigen escape and tumor relapse still occur after the use of these therapies. Designing CAR-T therapies that targets multiple antigens simultaneously seems a feasible way to avoid antigen escape, and it has been extensively studied elsewhere. Here, we report novel BCMA-OR-CD38 Tan CAR T cells that can trigger robust cytotoxicity against target cells expressing either BCMA or CD38. We demonstrate that, in in vitro studies, these BCMA-OR-CD38 Tan CAR T cells exhibit similar CAR expression, superior cytotoxicity and antigen-stimulated T cell proliferation as compared to single-targeted CAR T cells or CD38-OR-BCMA Tan CAR T cells. Importantly, these BCMA-OR-CD38 Tan CAR-T cells can achieve complete tumor clearance in myeloma-bearing mice with no relapse observed through the course of these experiments. Finally, this BCMA-OR-CD38 Tan CAR was fully compatible with existing clinical grade T cell manufacturing procedures and can be implemented using current clinical protocols. Taken together, our results present an effective solution to the challenge of antigen escape in BCMA CAR T-cell therapies.
Collapse
Affiliation(s)
- Yaru Feng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Xiuying Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaorui Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yating Zhou
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zhiru Song
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Bingjie Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Jianxun Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.,Department of Gene and Cellular Therapy, Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
17
|
Nabar NR, Heijjer CN, Shi CS, Hwang IY, Ganesan S, Karlsson MCI, Kehrl JH. LRRK2 is required for CD38-mediated NAADP-Ca 2+ signaling and the downstream activation of TFEB (transcription factor EB) in immune cells. Autophagy 2021; 18:204-222. [PMID: 34313548 PMCID: PMC8865229 DOI: 10.1080/15548627.2021.1954779] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
CD38 is a cell surface receptor capable of generating calcium-mobilizing second messengers. It has been implicated in host defense and cancer biology, but signaling mechanisms downstream of CD38 remain unclear. Mutations in LRRK2 (leucine-rich repeat kinase 2) are the most common genetic cause of Parkinson disease; it is also a risk factor for Crohn disease, leprosy, and certain types of cancers. The pathogenesis of these diseases involves inflammation and macroautophagy/autophagy, processes both CD38 and LRRK2 are implicated in. Here, we mechanistically and functionally link CD38 and LRRK2 as upstream activators of TFEB (transcription factor EB), a host defense transcription factor and the master transcriptional regulator of the autophagy/lysosome machinery. In B-lymphocytes and macrophages, we show that CD38 and LRRK2 exist in a complex on the plasma membrane. Ligation of CD38 with the monoclonal antibody clone 90 results in internalization of the CD38-LRRK2 complex and its targeting to the endolysosomal system. This generates an NAADP-dependent calcium signal, which requires LRRK2 kinase activity, and results in the downstream activation of TFEB. lrrk2 KO macrophages accordingly have TFEB activation defects following CD38 or LPS stimulation and fail to switch to glycolytic metabolism after LPS treatment. In overexpression models, the pathogenic LRRK2G2019S mutant promotes hyperactivation of TFEB even in the absence of CD38, both by stabilizing TFEB and promoting its nuclear translocation via aberrant calcium signaling. In sum, we have identified a physiological CD38-LRRK2-TFEB signaling axis in immune cells. The common pathogenic mutant, LRRK2G2019S, appears to hijack this pathway. Abbreviations:ADPR: ADP-ribose; AMPK: AMP-activated protein kinase; BMDM: bone marrow-derived macrophage; cADPR: cyclic-ADP-ribose; COR: C-terminal of ROC; CTSD: cathepsin D; ECAR: extracellular acidification rate; EDTA: ethylenediaminetetraacetic acid; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; GPN: Gly-Phe β-naphthylamide; GSK3B/GSK3β: glycogen synthase kinase 3 beta; GTP: guanosine triphosphate; KD: knockdown; LAMP1: lysosomal-associated membrane protein 1; LRR: leucine rich repeat; LRRK2: leucine rich repeat kinase 2; mAb: monoclonal antibody; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MAPK/ERK: mitogen-activated protein kinase; MCOLN1: mucolipin 1; MFI: mean fluorescence intensity; mRNA: messenger RNA; MTOR: mechanistic target of rapamycin kinase; NAADP: nicotinic acid adenine dinucleotide phosphate; NAD: nicotinamide adenine dinucleotide; NADP: nicotinamide adenine dinucleotide phosphate; PD: Parkinson disease; PPP3CB: protein phosphatase 3, catalytic subunit, beta isoform; q-RT-PCR: quantitative reverse transcription polymerase chain reaction; ROC: Ras of complex; siRNA: small interfering RNA; SQSTM1/p62: sequestome 1; TFEB: transcription factor EB; TPCN: two pore channel; TRPM2: transient receptor potential cation channel, subfamily M, member 2; ZKSCAN3: zinc finger with KRAB and SCAN domains 3
Collapse
Affiliation(s)
- Neel R Nabar
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christopher N Heijjer
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Chong-Shan Shi
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Il-Young Hwang
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sundar Ganesan
- Biological Imaging Section, Research Technologies Branch, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - John H Kehrl
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
18
|
Musto P, La Rocca F. Monoclonal antibodies in relapsed/refractory myeloma: updated evidence from clinical trials, real-life studies, and meta-analyses. Expert Rev Hematol 2021; 13:331-349. [PMID: 32153224 DOI: 10.1080/17474086.2020.1740084] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: In the last few years, monoclonal antibodies have rapidly modified the therapeutic strategies for treating patients with multiple myeloma.Areas covered: In this review, the most recent literature data regarding indications for which monoclonal antibodies are currently or will be shortly approved as salvage therapies in relapsed/refractory myeloma are discussed. In particular, updated results until March 22, 2020 of antibodies directed against CD38 (daratumumab and isatuximab), SLAMF7 (elotuzumab), BCMA (GSK2857916/belantamab mafodotin), and PD-1/PD-1 L axis (nivolumab and pembrolizumab) will be analyzed in detail.Expert opinion: Monoclonal antibodies represent a new, very effective approach that will open novel and dynamic treatment scenarios for myeloma patients in the coming years. Optimal positioning and selection of different antibodies that are or will be soon available, appropriate combinations and careful evaluation of possible new toxicities should be considered in the future management of these patients.
Collapse
Affiliation(s)
- Pellegrino Musto
- Chair of Hematology and Unit of Hematology and Stem Cell Transplantation, AOU Consorziale Policlinico, "Aldo Moro" University, School of Medicine, Bari, Italy
| | - Francesco La Rocca
- Laboratory of Clinical Research and Advanced Diagnostics, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (Pz), Italy
| |
Collapse
|
19
|
Lee HT, Kim Y, Park UB, Jeong TJ, Lee SH, Heo YS. Crystal structure of CD38 in complex with daratumumab, a first-in-class anti-CD38 antibody drug for treating multiple myeloma. Biochem Biophys Res Commun 2020; 536:26-31. [PMID: 33360095 DOI: 10.1016/j.bbrc.2020.12.048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 10/22/2022]
Abstract
Multiple myeloma is a blood cancer characterized by the plasma cell malignancy in the bone marrow, resulting in the destruction of bone tissue. Recently, the US FDA approved two antibody drugs for the treatment of multiple myeloma, daratumumab and isatuximab, targeting CD38, a type II transmembrane glycoprotein highly expressed in plasma cells and multiple myeloma cells. Here, we report the crystal structure of CD38 in complex with the Fab fragment of daratumumab, providing its exact epitope on CD38 and the structural insights into the mechanism of action of the antibody drug. Daratumumab binds to a specific discontinuous region on CD38 that includes residues located opposite to the active site of CD38. All the six complementarity determining regions of daratumumab are involved in the CD38 interaction. The epitopes of daratumumab and isatuximab do not overlap at all and their bindings to CD38 induce different structural changes within the CD38 protein. This structural study can facilitate the design of improved biologics or effective combination therapies for the treatment of multiple myeloma.
Collapse
Affiliation(s)
- Hyun Tae Lee
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Yujin Kim
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ui Beom Park
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Tae Jun Jeong
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Sang Hyung Lee
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Yong-Seok Heo
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
| |
Collapse
|
20
|
D’Agostino M, Innorcia S, Boccadoro M, Bringhen S. Monoclonal Antibodies to Treat Multiple Myeloma: A Dream Come True. Int J Mol Sci 2020; 21:E8192. [PMID: 33139668 PMCID: PMC7662679 DOI: 10.3390/ijms21218192] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 10/26/2020] [Accepted: 11/01/2020] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy is increasingly used in the treatment of multiple myeloma (MM). Monoclonal antibodies (mAbs) are safe and effective ways to elicit immunotherapeutic responses. In 2015, daratumumab has become the first mAb approved by the Food and Drug Administration for clinical use in MM and, in the last 5 years, a lot of clinical and preclinical research has been done to optimize the use of this drug class. Currently, mAbs have already become part of standard-of-care combinations for the treatment of relapsed/refractory MM and very soon they will also be used in the frontline setting. The success of simple mAbs ('naked mAbs') prompted the development of new types of molecules. Antibody-drug conjugates (ADCs) are tumor-targeting mAbs that release a cytotoxic payload into the tumor cells upon antigen binding in order to destroy them. Bispecific antibodies (BiAbs) are mAbs simultaneously targeting a tumor-associated antigen and an immune cell-associated antigen in order to redirect the immune cell cytotoxicity against the tumor cell. These different constructs produced solid preclinical data and promising clinical data in phase I/II trials. The aim of this review article is to summarize all the recent developments in the field, including data on naked mAbs, ADCs and BiAbs.
Collapse
Affiliation(s)
| | | | | | - Sara Bringhen
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (M.D.); (S.I.); (M.B.)
| |
Collapse
|
21
|
Bertamini L, Bonello F, Boccadoro M, Bringhen S. New drugs in early development for treating multiple myeloma: all that glitters is not gold. Expert Opin Investig Drugs 2020; 29:989-1004. [PMID: 32434394 DOI: 10.1080/13543784.2020.1772753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The last twenty years have introduced new therapeutic agents for multiple myeloma (MM); these include proteasome inhibitors (PIs), immunomodulatory drugs (IMDs) and monoclonal antibodies (mAbs). However, MM remains incurable, hence there is an unmet need for new agents for the treatment of advanced refractory disease. New agents could also be used in early lines to achieve improved, more sustained remission. AREAS COVERED We review the most promising agents investigated in early-phase trials for the treatment of MM and provide an emphasis on new agents directed against well-known targets (new PIs, IMDs and anti-CD38 mAbs). Drugs that work through distinct and numerous mechanisms of action (e.g. pro-apoptotic agents and tyrosine kinase inhibitors) and innovative immunotherapeutic approaches are also described. The paper culminates with our perspective on therapeutic approaches on the horizon for this disease. EXPERT OPINION IMD iberdomide and the export protein inhibitor selinexor demonstrated efficacy in heavily pretreated patients who had no other therapeutic options. We expect that immunotherapy with anti-BCMA BTEs and ADCs will revolutionize the approach to treating the early stages of the disease. Data on venetoclax in t(11;14)-positive patients may pave the way for personalized therapy. Not all new agents under early clinical evaluation will be investigated in regulatory phase III trials; one of the most important challenges is to identify those that could make a difference.
Collapse
Affiliation(s)
- Luca Bertamini
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino , Torino, Italy
| | - Francesca Bonello
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino , Torino, Italy
| | - Mario Boccadoro
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino , Torino, Italy
| | - Sara Bringhen
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino , Torino, Italy
| |
Collapse
|
22
|
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. [PMID: 32471019 DOI: 10.1111/sji.12910] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/23/2020] [Accepted: 05/19/2020] [Indexed: 12/14/2022]
Abstract
Multiple myeloma (MM) is a haematological malignancy caused by malignant proliferation of plasma cells in bone marrow. In recent years, MM patients are commonly treated with chemotherapy, autologous stem cell transplantation, protease inhibitors, immunomodulatory drugs and monoclonal antibodies, however most patients eventually relapse. Therefore, more effective therapies are highly needed. Anti-BCMA CAR-T therapy, a novel and efficacious method for treating MM and relapsed/refractory multiple myeloma (RRMM), has been designed and applied in clinics. The CAR-T can specifically recognize the targeted molecule B cell maturation antigen (BCMA) and kill MM cells expressing BCMA and several clinical trials have revealed high response rates in the therapy. Herein, we summarize the developments, the current design and clinical trials, the side effects of anti-BCMA CAR-T therapy and comparison of it with other CAR-T therapies.
Collapse
Affiliation(s)
- Deming Feng
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Jian Sun
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
23
|
Motais B, Charvátová S, Hrdinka M, Šimíček M, Jelínek T, Ševčíková T, Kořístek Z, Hájek R, Bagó JR. A Bird's-Eye View of Cell Sources for Cell-Based Therapies in Blood Cancers. Cancers (Basel) 2020; 12:E1333. [PMID: 32456165 PMCID: PMC7281611 DOI: 10.3390/cancers12051333] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/17/2020] [Accepted: 05/20/2020] [Indexed: 12/25/2022] Open
Abstract
: Hematological malignancies comprise over a hundred different types of cancers and account for around 6.5% of all cancers. Despite the significant improvements in diagnosis and treatment, many of those cancers remain incurable. In recent years, cancer cell-based therapy has become a promising approach to treat those incurable hematological malignancies with striking results in different clinical trials. The most investigated, and the one that has advanced the most, is the cell-based therapy with T lymphocytes modified with chimeric antigen receptors. Those promising initial results prepared the ground to explore other cell-based therapies to treat patients with blood cancer. In this review, we want to provide an overview of the different types of cell-based therapies in blood cancer, describing them according to the cell source.
Collapse
Affiliation(s)
- Benjamin Motais
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
| | - Sandra Charvátová
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
| | - Matouš Hrdinka
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Michal Šimíček
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Tomáš Jelínek
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Tereza Ševčíková
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Zdeněk Kořístek
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Roman Hájek
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Juli R. Bagó
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| |
Collapse
|
24
|
D'Agostino M, Mina R, Gay F. Anti-CD38 monoclonal antibodies in multiple myeloma: another cook in the kitchen? LANCET HAEMATOLOGY 2020; 7:e355-e357. [DOI: 10.1016/s2352-3026(19)30254-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 10/24/2022]
|
25
|
Musto P, La Rocca F. Monoclonal antibodies in newly diagnosed and smoldering multiple myeloma: an updated review of current clinical evidence. Expert Rev Hematol 2020; 13:501-517. [PMID: 32290723 DOI: 10.1080/17474086.2020.1753502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Monoclonal antibodies (MoAbs) are rapidly changing the therapeutic scenario of multiple myeloma. Most of the available data, however, come from studies performed in patients with relapsed or refractory disease.Area covered: Here, the most recent results from clinical trials that have investigated (or are investigating) efficacy and safety of MoAbs as front-line treatments in both transplant-eligible and not-eligible patients with newly diagnosed multiple myeloma, as well as in smoldering myeloma, are reviewed. PubMed reported articles before 28 March 2020, and abstracts presented at the last ASCO, ASH, EHA, and IMW meetings were considered. Among others, pertinent data regarding daratumumab, isatuximab, elotuzumab, and pembrolizumab will be analyzed.Expert opinion: Introduction of MoAbs as first-line therapy will likely provide a significant improvement in the clinical outcome of patients with multiple myeloma. This will also require an appropriate re-positioning of salvage therapies. The role of MoAbs in smoldering myeloma appears to be promising, but adequate follow-up is needed.
Collapse
Affiliation(s)
- Pellegrino Musto
- Chair of Hematology and Unit of Hematology and Stem Cell Transplantation, "Aldo Moro" University School of Medicine, AOU Consorziale Policlinico, Bari, Italy
| | - Francesco La Rocca
- Laboratory of Advanced Diagnostics and Clinical Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (Pz), Italy
| |
Collapse
|
26
|
Therapeutic Monoclonal Antibodies and Antibody Products: Current Practices and Development in Multiple Myeloma. Cancers (Basel) 2019; 12:cancers12010015. [PMID: 31861548 PMCID: PMC7017131 DOI: 10.3390/cancers12010015] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/13/2019] [Accepted: 12/16/2019] [Indexed: 12/26/2022] Open
Abstract
Immunotherapy is the latest innovation for the treatment of multiple myeloma (MM). Monoclonal antibodies (mAbs) entered the clinical practice and are under evaluation in clinical trials. MAbs can target highly selective and specific antigens on the cell surface of MM cells causing cell death (CD38 and CS1), convey specific cytotoxic drugs (antibody-drug conjugates), remove the breaks of the immune system (programmed death 1 (PD-1) and PD-ligand 1/2 (L1/L2) axis), or boost it against myeloma cells (bi-specific mAbs and T cell engagers). Two mAbs have been approved for the treatment of MM: the anti-CD38 daratumumab for newly-diagnosed and relapsed/refractory patients and the anti-CS1 elotuzumab in the relapse setting. These compounds are under investigation in clinical trials to explore their synergy with other anti-MM regimens, both in the front-line and relapse settings. Other antibodies targeting various antigens are under evaluation. B cell maturation antigens (BCMAs), selectively expressed on plasma cells, emerged as a promising target and several compounds targeting it have been developed. Encouraging results have been reported with antibody drug conjugates (e.g., GSK2857916) and bispecific T cell engagers (BiTEs®), including AMG420, which re-directs T cell-mediated cytotoxicity against MM cells. Here, we present an overview on mAbs currently approved for the treatment of MM and promising compounds under investigation.
Collapse
|
27
|
D'Agostino M, Raje N. Anti-BCMA CAR T-cell therapy in multiple myeloma: can we do better? Leukemia 2019; 34:21-34. [PMID: 31780814 DOI: 10.1038/s41375-019-0669-4] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/27/2019] [Accepted: 11/13/2019] [Indexed: 12/17/2022]
Abstract
Despite a substantial survival improvement and the availability of many new drugs in the last 2 decades, multiple myeloma (MM) remains largely incurable. Immunotherapeutic approaches are changing the current landscape in MM with B-cell maturation antigen (BCMA) as one of the most promising target antigens. Chimeric antigen receptor (CAR) T-cell therapy targeting BCMA produced unprecedented results in heavily pretreated relapsed and/or refractory MM. Data on more than 300 MM patients treated with anti-BCMA directed CAR T cells are available and these numbers are rapidly increasing. The response rate and the depth of responses induced by anti-BCMA CAR T cells are impressive; however, the majority of patients eventually relapse. Understanding the underlying mechanisms of response and resistance in treated MM patients will be critical to the rational development of this therapy. Moreover, the ideal place of this therapy in the treatment paradigm for MM is an important question that needs biological and clinical correlative data to help elucidate. T-cell-related, tumor-related and microenvironmental factors may play a role in the efficacy of anti-BCMA CAR T-cell therapy. In this review we summarize key clinical and correlative data on anti-BCMA CAR T-cell therapy. Based on available data we will try to highlight opportunities to further optimize this potential game-changing therapy for MM.
Collapse
Affiliation(s)
- Mattia D'Agostino
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy.,Center for Multiple Myeloma, Division of Hematology and Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Noopur Raje
- Center for Multiple Myeloma, Division of Hematology and Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
Gao Z, Tong C, Wang Y, Chen D, Wu Z, Han W. Blocking CD38-driven fratricide among T cells enables effective antitumor activity by CD38-specific chimeric antigen receptor T cells. J Genet Genomics 2019; 46:367-377. [PMID: 31466926 DOI: 10.1016/j.jgg.2019.06.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/09/2019] [Accepted: 06/17/2019] [Indexed: 12/15/2022]
Abstract
Chimeric antigen receptor T-cell (CAR T) therapy is a kind of effective cancer immunotherapy. However, designing CARs remains a challenge because many targetable antigens are shared by T cells and tumor cells. This shared expression of antigens can cause CAR T cell fratricide. CD38-targeting approaches (e.g., daratumumab) have been used in clinical therapy and have shown promising results. CD38 is a kind of surface glycoprotein present in a variety of cells, such as T lymphocytes and tumor cells. It was previously reported that CD38-based CAR T cells may undergo apoptosis or T cell-mediated killing (fratricide) during cell manufacturing. In this study, a CAR containing a sequence targeting human CD38 was designed to be functional. To avoid fratricide driven by CD38 and ensure the production of CAR T cells, two distinct strategies based on antibodies (clone MM12T or clone MM27) or proteins (H02H or H08H) were used to block CD38 or the CAR single-chain variable fragment (scFv) domain, respectively, on the T cell surface. The results indicated that the antibodies or proteins, especially the antibody MM27, could affect CAR T cells by inhibiting fratricide while promoting expansion and enrichment. Anti-CD38 CAR T cells exhibited robust and specific cytotoxicity to CD38+ cell lines and tumor cells. Furthermore, the levels of the proinflammatory factors TNF-α, IFN-γ and IL-2 were significantly upregulated in the supernatants of A549CD38+ cells. Finally, significant control of disease progression was demonstrated in xenograft mouse models. In conclusion, these findings will help to further enhance the expansion, persistence and function of anti-CD38 CAR T cells in subsequent clinical trials.
Collapse
Affiliation(s)
- Zhitao Gao
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Sciences, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100086, China
| | - Chuan Tong
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Sciences, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100086, China
| | - Yao Wang
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Sciences, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100086, China
| | - Deyun Chen
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Sciences, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100086, China
| | - Zhiqiang Wu
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Sciences, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100086, China.
| | - Weidong Han
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Sciences, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100086, China.
| |
Collapse
|