51
|
Wang Y, Lv L, Song Y, Wei X, Zhou H, Liu Q, Xu K, Yan D, Zhang C, Liu S, Jin J, Mei H, Niu T, Liang A, Gu R, Ren J, Feng Y, Jin W, Zhou Y, Deng Y, Wang J. Inaticabtagene autoleucel in adult relapsed or refractory B-cell acute lymphoblastic leukemia. Blood Adv 2025; 9:836-843. [PMID: 39626300 PMCID: PMC11872425 DOI: 10.1182/bloodadvances.2024014182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/06/2024] [Indexed: 03/05/2025] Open
Abstract
ABSTRACT Before November 2023, CD19 chimeric antigen receptor (CAR) T-cell therapies had not been approved in China for patients with relapsed or refractory B-cell acute lymphoblastic leukemia (R/R B-ALL), leaving a significant unmet need. In response, inaticabtagene autoleucel (Inati-cel), a novel CD19 CAR T-cell therapy with a distinct single-chain variable fragment (HI19α), was developed and showed promising efficacy in preliminary clinical research. We conducted a phase 2, single-arm, multicenter study of Inati-cel in adult CD19+ R/R B-ALL in China. The primary end point was the overall remission rate (ORR) at the end of month 3. Forty-eight patients who underwent Inati-cel infusion were evaluated for both efficacy and safety. Among them, 34 patients achieved and maintained remission beyond 3 months, with a 3-month ORR of 70.8% (95% confidence interval [CI], 55.9-83.1). The best ORR was 85.4%, with all responders reaching minimal residual disease negativity. With a median follow-up of 23.7 months, the median duration of remission was 20.7 months (95% CI, 6.4 to not reached), and the median overall survival was not reached (95% CI, 13.0 months to not reached). Additionally, grade ≥3 cytokine release syndrome and neurologic events occurred in 12.5% and 6.2% of patients, respectively. The 2-year follow-up data suggest that Inati-cel demonstrates encouraging and durable responses with manageable safety profiles in R/R B-ALL. Based on the data from this pivotal trial, Inati-cel was approved as the first CAR T-cell therapy for adult R/R B-ALL in China and underscores its potential therapeutic benefits for this patient population. This trial was registered at www.ClinicalTrials.gov as #NCT04684147.
Collapse
Affiliation(s)
- Ying Wang
- Leukemia Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center of Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Lulu Lv
- Juventas Cell Therapy Ltd, Tianjin, China
| | - Yongping Song
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xudong Wei
- Department of Hematology, Henan Cancer Hospital, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongsheng Zhou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dongmei Yan
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Cheng Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
| | - Shuangyou Liu
- Department of Hematology, Beijing Gobroad Boren Hospital, Beijing, China
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Heng Mei
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Aibin Liang
- Department of Hematology, Tongji Hospital of Tongji University, Shanghai, China
| | - Runxia Gu
- Leukemia Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center of Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jienan Ren
- Juventas Cell Therapy Ltd, Tianjin, China
| | - Yi Feng
- Juventas Cell Therapy Ltd, Tianjin, China
| | - Wei Jin
- Juventas Cell Therapy Ltd, Tianjin, China
| | - Yan Zhou
- Juventas Cell Therapy Ltd, Tianjin, China
| | | | - Jianxiang Wang
- Leukemia Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center of Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
52
|
Schulenburg A, Rüsing L, Bumberger A, Mitterbauer M, Cserna J, Petrasch C, Oesterreicher S, Worel N, Rabitsch W. Retrospective Analysis: S100 as Marker for Immune Effector Cell-Associated Neurotoxicity Syndrome. Oncology 2025:1-4. [PMID: 39999826 DOI: 10.1159/000543949] [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: 11/28/2024] [Accepted: 01/22/2025] [Indexed: 02/27/2025]
Abstract
INTRODUCTION Chimeric antigen receptor (CAR) T-cell therapy has emerged as a promising treatment for hematologic malignancies, offering significant therapeutic benefits. However, this therapy is also associated with adverse effects such as cytokine release syndrome and Immune Effector Cell-Associated Neurotoxicity Syndrome (ICANS), which can lead to severe neurological symptoms. The pathophysiology of ICANS remains unclear but is believed to involve immune-mediated inflammation in the brain. This study investigates the potential of S100, a protein marker associated with blood-brain barrier integrity, as an early indicator of ICANS. METHODS We retrospectively analyzed daily blood samples for S100 levels in patients undergoing CAR T-cell therapy, correlating these levels with the onset and severity of ICANS. RESULTS The results show that S100 levels significantly increased in patients who developed ICANS, with a positive correlation between the duration of elevation and the severity of the neurological symptoms. CONCLUSION These findings suggest that S100 may serve as a useful biomarker for early detection of ICANS and could potentially guide therapeutic interventions. However, further studies are needed to fully understand its prognostic value in this context.
Collapse
Affiliation(s)
- Axel Schulenburg
- Bone Marrow Transplantation Unit, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Lina Rüsing
- Bone Marrow Transplantation Unit, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Armin Bumberger
- Bone Marrow Transplantation Unit, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Margit Mitterbauer
- Bone Marrow Transplantation Unit, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Julia Cserna
- Bone Marrow Transplantation Unit, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Clemens Petrasch
- Bone Marrow Transplantation Unit, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Sophia Oesterreicher
- Bone Marrow Transplantation Unit, Department of Transfusion Medicine and Cell Therapy, Medical University of Vienna, Vienna, Austria
| | - Nina Worel
- Bone Marrow Transplantation Unit, Department of Transfusion Medicine and Cell Therapy, Medical University of Vienna, Vienna, Austria
| | - Werner Rabitsch
- Bone Marrow Transplantation Unit, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
53
|
McNerney KO, Schultz LM. Tisagenlecleucel in Practice: Real-World Lessons in Pediatric and Young Adult B-ALL. Transplant Cell Ther 2025:S2666-6367(25)01050-4. [PMID: 39993597 DOI: 10.1016/j.jtct.2025.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/12/2025] [Accepted: 02/15/2025] [Indexed: 02/26/2025]
Abstract
The global multi-institutional registration trial (ELIANA) of CD19.41BB.zeta chimeric antigen receptor (CAR) T cell therapy forged the path to the first FDA-approved CAR T product, tisagenlecleucel. Since its approval, extensive post-market experience with CAR T cells in children and young adults has amassed, allowing several multi-institutional efforts to leverage real-world data. Real-world data has validated clinical trial findings and provided insights into CAR T-cell use in patient groups not included in early clinical trials, such as children <3 years, patients with active CNS and isolated extramedullary disease, and patients treated in first relapse. Data from multi-centered consortia has also identified cohorts who experienced inferior outcomes post-tisagenlecleucel, informing high-risk groups for whom further treatment optimization is needed, and delineating treatment variables, such as CAR T cell dose and lymphodepleting chemotherapy pharmacokinetics, that impact outcomes. In this early stage of CAR T-cell therapies, real-world experience provides an increasingly rich data reservoir and an invaluable resource to investigate and address clinical gaps for CAR T recipients. This review highlights key insights gained from post-market studies that have informed clinical use of CAR T-cell therapy for children and young adults with B-ALL.
Collapse
Affiliation(s)
- Kevin O McNerney
- Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital, Chicago, Illinois
| | - Liora M Schultz
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, Palo Alto, California.
| |
Collapse
|
54
|
Umair MM, Lai X, Xue Y, Yao H. Influence of CAR T-cell therapy associated complications. Front Oncol 2025; 15:1494986. [PMID: 40052127 PMCID: PMC11882432 DOI: 10.3389/fonc.2025.1494986] [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: 09/11/2024] [Accepted: 01/21/2025] [Indexed: 03/09/2025] Open
Abstract
Since the introduction of chimeric antigen receptor (CAR) T-cell therapy, it has elicited an immense response in both targeted and residual cancers. Its clinical efficacy is often accompanied by a group of side effects that may become serious because of factors such as tumor burden, the extent of lymphodepletion, and the type of co-stimulus. It is also crucial to know the common toxicities associated with CAR T-cell therapy, including cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), cardiotoxicity, metabolic disorders, pulmonary toxicity, macrophage activation syndrome (MAS), prolonged cytopenia, coagulation disorders, and potential off-target effects on various organs. If not well managed, these can be fatal. However, knowledge about molecular pathways, calcineurin inhibitors, IL-6 receptor antagonists, steroids, suppression of nitric oxide synthase, various therapeutic approaches, and other recent advances have been developed to mitigate the fatal results of various short-term and chronic adverse events related to CAR T-cell therapy. This study provides a comprehensive perspective on contemporary management strategies and presumed causative processes of CAR T-cell-related adverse effects, albeit with several limitations. When CAR T-cell complications, costs, and challenges of toxicity management are properly considered, the CAR T-cell therapy of the future will include a number of toxicity-escaping options.
Collapse
Affiliation(s)
- Mohammad Mussab Umair
- Cancer Biotherapy Center & Cancer Research Institute, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xun Lai
- Department of Hematology, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - YuanBo Xue
- Cancer Biotherapy Center, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hong Yao
- Cancer Biotherapy Center & Cancer Research Institute, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
55
|
Song Y, Wang Y, Man J, Xu Y, Zhou G, Shen W, Chao Y, Yang K, Pei P, Hu L. Chimeric Antigen Receptor Cells Solid Tumor Immunotherapy Assisted by Biomaterials Tools. ACS APPLIED MATERIALS & INTERFACES 2025; 17:10246-10264. [PMID: 39903799 DOI: 10.1021/acsami.4c20275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Chimeric antigen receptor (CAR) immune cell therapies have revolutionized oncology, particularly in hematological malignancies, yet their efficacy against solid tumors remains limited due to challenges such as dense stromal barriers and immunosuppressive microenvironments. With advancements in nanobiotechnology, researchers have developed various strategies and methods to enhance the CAR cell efficacy in solid tumor treatment. In this Review, we first outline the structure and mechanism of CAR-T (T, T cell), CAR-NK (NK, natural killer), and CAR-M (M, macrophage) cell therapies and deeply analyze the potential of these cells in the treatment of solid tumors and the challenges they face. Next, we explore how biomaterials can optimize these treatments by improving the tumor microenvironment, controlling CAR cell release, promoting cell infiltration, and enhancing efficacy. Finally, we summarize the current challenges and potential solutions, emphasize the effective combination of biomaterials and CAR cell therapy, and look forward to its future clinical application and treatment strategies. This Review provides important theoretical perspectives and practical guidance for the future development of more effective solid tumor treatment strategies.
Collapse
Affiliation(s)
- Yujie Song
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yifan Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jianping Man
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yihua Xu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Wenhao Shen
- Department of Oncology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, Jiangsu 225300, China
| | - Yu Chao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Pei Pei
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Lin Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
56
|
Lonez C, Bolsée J, Huberty F, Nguyen T, Jacques-Hespel C, Anguille S, Flament A, Breman E. Clinical Proof-of-Concept of a Non-Gene Editing Technology Using miRNA-Based shRNA to Engineer Allogeneic CAR T-Cells. Int J Mol Sci 2025; 26:1658. [PMID: 40004122 PMCID: PMC11855736 DOI: 10.3390/ijms26041658] [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: 01/08/2025] [Revised: 01/31/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
With the success of chimeric antigen receptor (CAR) T-cell therapy in B-cell malignancies, efforts are being made to extend this therapy to other malignancies and broader patient populations. However, limitations associated with the time-consuming and highly personalized manufacturing of autologous CAR T-cells remain. Allogeneic CAR T-cell approaches may overcome these challenges but require further engineering to reduce their alloreactivity. As a means to prevent graft-versus-host disease (GvHD) of allogeneic CAR T-cells, we have selected a micro RNA (miRNA)-based short hairpin RNA (shRNA) targeting CD3ζ which efficiently downregulates the expression of the T-cell receptor (TCR) below detection level. We generated allogeneic anti-B-cell maturation antigen CAR T-cells (CYAD-211) that co-express an anti-CD3ζ miRNA-based shRNA within the CAR construct which efficiently inhibited TCR-mediated signaling in vitro and GvHD in vivo. CYAD-211 was subsequently evaluated in a Phase-I clinical trial (NCT04613557), in patients with relapsed or refractory multiple myeloma. No signs of GvHD were observed despite evidence of engraftment, demonstrating efficient downregulation of the TCR. Our data provide proof of concept that a non-gene-edited technology can generate fully functional allogeneic CAR T-cells, without any signs of GvHD. However, further engineering of the CAR T-cells is needed to improve their persistence and long-term activity.
Collapse
MESH Headings
- MicroRNAs/genetics
- Humans
- Graft vs Host Disease/prevention & control
- Graft vs Host Disease/genetics
- Graft vs Host Disease/immunology
- Animals
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- RNA, Small Interfering/genetics
- Mice
- Immunotherapy, Adoptive/methods
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- CD3 Complex/genetics
- CD3 Complex/immunology
- Multiple Myeloma/therapy
- Multiple Myeloma/genetics
- Multiple Myeloma/immunology
- Female
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Transplantation, Homologous
- Male
- Proof of Concept Study
- Gene Editing/methods
Collapse
Affiliation(s)
- Caroline Lonez
- Celyad Oncology, 1435 Mont-Saint-Guibert, Belgium (E.B.)
| | | | - Fanny Huberty
- Celyad Oncology, 1435 Mont-Saint-Guibert, Belgium (E.B.)
| | - Thuy Nguyen
- Celyad Oncology, 1435 Mont-Saint-Guibert, Belgium (E.B.)
| | | | - Sebastien Anguille
- Division of Hematology, University Hospital Antwerp (UZ Antwerp), 2650 Antwerp, Belgium
| | - Anne Flament
- Celyad Oncology, 1435 Mont-Saint-Guibert, Belgium (E.B.)
| | - Eytan Breman
- Celyad Oncology, 1435 Mont-Saint-Guibert, Belgium (E.B.)
| |
Collapse
|
57
|
Nair AG, Ehrhardt GRA, Grunebaum E. Variable Lymphocyte Receptor B Technologies - Are They Ready for Prime Time? Immunol Invest 2025:1-21. [PMID: 39936604 DOI: 10.1080/08820139.2025.2462536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
OBJECTIVE To review the current and the potential research and clinical use of VLRBs. METHODS A literature search was conducted for English studies published in the past 20 years using the terms "Variable Lymphocyte Receptor," "VLR," "VLRB" or "Repebody." Only primary reports were included. RESULTS VLRB-based technologies are currently being investigated for diagnosis, imaging, and treatment of diverse conditions including solid organ and hematological malignancies, infectious diseases, autoimmunity, and degenerative and metabolic disorders. VLRB mAbs can be used to directly recognize disease biomarkers, such as B cells from chronic lymphocytic leukemia, or to deliver drugs to the brain or cancer cells. The VLRB C-terminal multimerization domain has been utilized to create vaccines while VLR-based chimeric antigen receptor (CAR) T cell constructs are being investigated for cancer therapies. CONCLUSIONS The extensive knowledge gained with VLRB mAbs in diverse in vitro and in vivo models emphasizes their promise for translation into clinical applications and readiness for prime time.
Collapse
Affiliation(s)
- Arundhati G Nair
- Developmental and Stem Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Götz R A Ehrhardt
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Eyal Grunebaum
- Developmental and Stem Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
58
|
Crespo E, Loureiro LR, Stammberger A, Hoffmann L, Berndt N, Hoffmann A, Dagostino C, Soto KEG, Rupp L, Arndt C, Schneider M, Ball CR, Bachmann M, Schmitz M, Feldmann A. RevCAR-mediated T-cell response against PD-L1-expressing cells turns suppression into activation. NPJ Precis Oncol 2025; 9:42. [PMID: 39924591 PMCID: PMC11808103 DOI: 10.1038/s41698-025-00828-6] [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: 08/21/2024] [Accepted: 01/28/2025] [Indexed: 02/11/2025] Open
Abstract
Applying CAR T-cell therapy to treat solid tumors is especially challenging due to the immunosuppressive tumor microenvironment (TME). While our modular RevCAR system enhances the safety and controllability of CAR T-cell therapy, effectively targeting solid tumors remains difficult. Since PD-L1 is an immune checkpoint frequently upregulated by cancer cells and their microenvironment, it is a relevant target for solid tumors. Here, we introduce a novel PD-L1 RevTM capable of redirecting RevCAR T-cells to specifically target and kill PD-L1-expressing tumor cells, becoming activated and secreting pro-inflammatory cytokines. This is shown in vitro with monolayer and 3D models, including patient-derived cultures, and in vivo. Furthermore, we demonstrate in vitro and in vivo an AND-gated targeting of cells simultaneously expressing PD-L1 and another tumor-associated antigen by the Dual RevCAR system. Our findings suggest that RevCAR-mediated targeting of PD-L1 could be a promising therapeutic approach for modulating the TME and improving solid tumor treatment.
Collapse
Affiliation(s)
- Eugenia Crespo
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Liliana R Loureiro
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Antonia Stammberger
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Lydia Hoffmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Nicole Berndt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Anja Hoffmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Claudia Dagostino
- Department for Translational Medical Oncology, National Center for Tumor Diseases Dresden (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Karla E G Soto
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Luise Rupp
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Claudia Arndt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Claudia R Ball
- Department for Translational Medical Oncology, National Center for Tumor Diseases Dresden (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Faculty of Biology, TUD Dresden University of Technology, Dresden, Germany
- German Cancer Consortium (DKTK), partner site Dresden, Dresden, Germany
| | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
- German Cancer Consortium (DKTK), partner site Dresden, Dresden, Germany.
- National Center for Tumor Diseases (NCT), partner site Dresden, Dresden, Germany.
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- German Cancer Consortium (DKTK), partner site Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), partner site Dresden, Dresden, Germany
| | - Anja Feldmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
- German Cancer Consortium (DKTK), partner site Dresden, Dresden, Germany.
- National Center for Tumor Diseases (NCT), partner site Dresden, Dresden, Germany.
| |
Collapse
|
59
|
Mulvey A, Trueb L, Coukos G, Arber C. Novel strategies to manage CAR-T cell toxicity. Nat Rev Drug Discov 2025:10.1038/s41573-024-01100-5. [PMID: 39901030 DOI: 10.1038/s41573-024-01100-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2024] [Indexed: 02/05/2025]
Abstract
The immune-related adverse events associated with chimeric antigen receptor (CAR)-T cell therapy result in substantial morbidity as well as considerable cost to the health-care system, and can limit the use of these treatments. Current therapeutic strategies to manage immune-related adverse events include interleukin-6 receptor (IL-6R) blockade and corticosteroids. However, because these interventions do not always address the side effects, nor prevent progression to higher grades of adverse events, new approaches are needed. A deeper understanding of the cell types involved, and their associated signalling pathways, cellular metabolism and differentiation states, should provide the basis for alternative strategies. To preserve treatment efficacy, cytokine-mediated toxicity needs to be uncoupled from CAR-T cell function, expansion, long-term persistence and memory formation. This may be achieved by targeting CAR or independent cytokine signalling axes transiently, and through novel T cell engineering strategies, such as low-affinity CAR-T cells, reversible on-off switches and versatile adaptor systems. We summarize the current management of cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome, and review T cell- and myeloid cell-intrinsic druggable targets and cellular engineering strategies to develop safer CAR-T cells.
Collapse
Affiliation(s)
- Arthur Mulvey
- Department of Oncology UNIL-CHUV, Service of Immuno-Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Lionel Trueb
- Department of Oncology UNIL-CHUV, Service of Immuno-Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - George Coukos
- Department of Oncology UNIL-CHUV, Service of Immuno-Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Caroline Arber
- Department of Oncology UNIL-CHUV, Service of Immuno-Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland.
- Departments of Oncology UNIL-CHUV and Laboratory Medicine and Pathology, Service and Central Laboratory of Hematology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
| |
Collapse
|
60
|
Sun J, Luo P, Guo Y, He Y, Wang C. Clinical Features, Treatment, and Outcomes of Nivolumab-Induced Hemophagocytic Lymphohistiocytosis. J Immunother 2025; 48:58-62. [PMID: 39183530 DOI: 10.1097/cji.0000000000000540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/06/2024] [Indexed: 08/27/2024]
Abstract
Haemophagocytic lymphohistiocytosis (HLH) is a rare and fatal immune-related event of nivolumab. The clinical features of nivolumab-induced HLH are unclear. The aim of this study was to investigate the clinical features, treatment, and outcome of nivolumab-induced HLH to provide information for prevention and treatment. We collected nivolumab-induced HLH-related case reports for retrospective analysis by searching the Chinese and English databases from inception to March 31, 2024. HLH developed in 24 patients, with a median age of 57 years (range: 26, 86). The onset of HLH symptoms ranged from 3 days to 68 weeks after administration, with a median time of 5.5 weeks. Fever (87.5%) was the most common symptom and could be accompanied by splenomegaly (66.7%) and hepatomegaly (20.8%). Laboratory tests revealed hemocytopenia, hypertriglyceridemia, hypofibrinogenemia, hyperferritinemia, increased sCD25, and decreased natural killer cell activity. Bone marrow biopsy showed hemophagocytosis (62.5%). After discontinuing nivolumab, HLH patients receiving systemic steroids, tocilizumab, and anakinra showed positive results. As a rare adverse reaction of nivolumab, HLH requires rapid diagnosis and appropriate treatment based on clinical symptoms and laboratory tests. Tocilizumab and anakinra can be used as an effective treatment against the steroid HLH.
Collapse
Affiliation(s)
- Jichun Sun
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Panpan Luo
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yuge Guo
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yang He
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chunjiang Wang
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
61
|
Chodup P, Samodelov SL, Visentin M, Kullak‐Ublick GA. Drug-Induced Liver Injury Associated With Emerging Cancer Therapies. Liver Int 2025; 45:e70002. [PMID: 39853863 PMCID: PMC11760653 DOI: 10.1111/liv.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/26/2025]
Abstract
Targeted therapies and immunotherapies have shown great promise as best-in-class treatments for several cancers with respect to efficacy and safety. While liver test abnormalities are rather common in patients treated with kinase inhibitors or immunotherapy, events of severe hepatotoxicity in these patients are rare in comparison with those associated with chemotherapeutics. The underlying mechanisms and risk factors for severe hepatotoxicity with novel oncology therapies are not well understood, complicating the drug-induced liver injury (DILI) risk assessment in the preclinical and clinical phases of drug development. The epidemiological and clinical characteristics, as well as mechanisms of liver toxicity, are described here to the current state of knowledge. Tools to study and assess the risk of DILI during drug development are concisely summarised, focusing on caveats thereof for novel oncology treatments. Emerging tools to optimise safety assessments and gather additional mechanistic insights into DILI are introduced. Particularly in oncology, where standard liver signals during drug development are tolerated to a marginally higher degree than in other indications due to the life-saving, life-extending and quality-of-life improvements for patients with severe or advanced cancers versus previous standard-of-care therapeutics, safety assessments must be tailored to the drug and indication. Trends in patient safety-centred drug development programmes and regulatory approval processes must continually be revisited and streamlined via obtaining an overall greater understanding of DILI and the tools available to assess mechanisms of injury, frequency, severity and prognosis.
Collapse
Affiliation(s)
- Piotr Chodup
- Department of Clinical Pharmacology and ToxicologyUniversity Hospital Zürich, University of ZürichZürichSwitzerland
| | - Sophia L. Samodelov
- Department of Clinical Pharmacology and ToxicologyUniversity Hospital Zürich, University of ZürichZürichSwitzerland
| | - Michele Visentin
- Department of Clinical Pharmacology and ToxicologyUniversity Hospital Zürich, University of ZürichZürichSwitzerland
| | - Gerd A. Kullak‐Ublick
- Department of Clinical Pharmacology and ToxicologyUniversity Hospital Zürich, University of ZürichZürichSwitzerland
- Mechanistic Safety, Patient Safety & Pharmacovigilance, Novartis DevelopmentBaselSwitzerland
| |
Collapse
|
62
|
Walmsley CS, Schoepflin Z, De Brabandt C, Rangachari D, Berwick S, Patell R. Hemophagocytic lymphohistiocytosis associated with immune checkpoint inhibitor use: A review of the current knowledge and future directions. Blood Cells Mol Dis 2025; 110:102896. [PMID: 39366077 DOI: 10.1016/j.bcmd.2024.102896] [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: 08/10/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/06/2024]
Abstract
Hemophagocytic lymphohistiocytosis (HLH) is a severe and often lethal inflammatory syndrome characterized by excessive immune activation leading to fever, cytopenias, and multiorgan involvement. Immune checkpoint inhibitors (ICIs) are central to many contemporary cancer regimens, but their use is associated with immune-related adverse events. Here, we report a case of ICI-induced HLH successfully treated with single agent dexamethasone and provide a scoping review of the literature for cases of ICI-induced HLH with a focus on treatment strategies and outcomes. Using the Medline database, we searched for cases of ICI-associated HLH, with a total of 51 cases reported between 2017 and 2023. Our results underscore the severe nature of this disease, with a 13.7 % mortality rate across 51 case reports. Treatment strategies for ICI-induced HLH were variable: steroids alone (56.9 %), steroids with etoposide (17.6 %), steroids with tociluzumab (11.8 %), among other combinations. Our literature review indicates that steroids alone may be sufficient treatment in some cases of ICI-HLH, with comparable mortality with steroids alone (n = 29) (13.8 %) to that of cases treated with both steroids and immunomodulators (n = 15, 13.3 %). Moreover, all patients treated with steroids and tocilizumab survived (n = 6), suggesting that tocilizumab may be a reasonable next line of therapy when steroid monotherapy proves inadequate. We propose an outline for investigation and treatment of this rare complication of ICI use. Finally, we discuss possible future approaches to develop evidence-based strategies for the diagnosis and management of ICI-induced HLH including the importance of integrating the role of patient community involvement.
Collapse
Affiliation(s)
- Charlotte S Walmsley
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| | - Zachary Schoepflin
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Charlotte De Brabandt
- Breast Cancer Center, Beth Israel Deaconess Medical Center, Boston, MA, United States of America; Patient Ambassador Program, American Cancer Society, Boston, MA, United States of America
| | - Deepa Rangachari
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Shana Berwick
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Rushad Patell
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States of America.
| |
Collapse
|
63
|
Li YR, Lyu Z, Shen X, Fang Y, Yang L. Boosting CAR-T cell therapy through vaccine synergy. Trends Pharmacol Sci 2025; 46:180-199. [PMID: 39755457 DOI: 10.1016/j.tips.2024.12.004] [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: 11/08/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 01/06/2025]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has transformed the treatment landscape for hematological cancers. However, achieving comparable success in solid tumors remains challenging. Factors contributing to these limitations include the scarcity of tumor-specific antigens (TSAs), insufficient CAR-T cell infiltration, and the immunosuppressive tumor microenvironment (TME). Vaccine-based strategies are emerging as potential approaches to address these challenges, enhancing CAR-T cell expansion, persistence, and antitumor efficacy. In this review, we explore diverse vaccine modalities, including mRNA, peptide, viral vector, and dendritic cell (DC)-based vaccines, and their roles in augmenting CAR-T cell responses. Special focus is given to recent clinical advancements combining mRNA-based vaccines with CAR-T therapy for the treatment of genitourinary cancers. In addition, we discuss crucial considerations for optimizing vaccine dosing, scheduling, and delivery to maximize CAR-T synergy, aiming to refine this combination strategy to improve treatment efficacy and safety.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Zibai Lyu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xinyuan Shen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ying Fang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
64
|
Rejeski HA, Hartz A, Rackl E, Li L, Schwepcke C, Rejeski K, Schmid C, Rank A, Schmohl J, Kraemer D, Bojko P, Schmetzer HM. Concentration-dependent effects of immunomodulatory cocktails on the generation of leukemia-derived dendritic cells, DC leu mediated T-cell activation and on-target/off-tumor toxicity. Front Immunol 2025; 15:1527961. [PMID: 39949718 PMCID: PMC11821930 DOI: 10.3389/fimmu.2024.1527961] [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: 11/14/2024] [Accepted: 12/19/2024] [Indexed: 02/16/2025] Open
Abstract
Acute myeloid leukemia (AML) remains a devastating diagnosis in clear need of therapeutic advances. Both targeted dendritic cells (DC) and particularly leukemia-derived dendritic cells (DCleu) can exert potent anti-leukemic activity. By converting AML blasts into immune activating and leukemia-antigen presenting cells, DC/DCleu-generating protocols can induce immune responses against AML blasts. Such protocols combine approved response modifiers (i.e., GM-CSF and PGE1/OK-432/PGE2) that synergistically improve the conversion of AML blasts into (mature) DC/DCleu. To guide potential clinical application of these response modifiers, we analyzed three different DC-generating protocols that combine a constant GM-CSF dose with varying concentrations of PGE1 (Kit-M), OK-432 (Kit-I), and PGE2 (Kit-K). Here, we specifically aimed to assess how different response modifier concentrations impact DC/DCleu generation, immune cell activation and leukemic blast lysis. We found that all immunomodulatory kits were effective in generating mature and leukemia-derived DCs from healthy and leukemic whole blood. For Kit-M, we noted optimal generation of DC-subsets at intermediary concentration ranges of PGE1 (0.25-4.0 µg/mL), which facilitated upregulation of activated and memory T-cells upon mixed lymphocyte culture, and efficient anti-leukemic activity in cytotoxicity assays. For Kit-I, we observed DC/DCleu generation and enhanced T- and immune cell activation across a broader range of OK-432 concentrations (5-40 µg/mL), which also facilitated improved leukemic blast killing. In conclusion, our results highlight that Kit-mediated DC/DCleu generation, immune cell activation and blast lysis are dependent on the concentration of response modifiers, which will guide future clinical development. Overall, DCleu-based immunotherapy represents a promising treatment strategy for AML patients.
Collapse
Affiliation(s)
- Hazal Aslan Rejeski
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich Site, Munich, Germany
| | - Anne Hartz
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich Site, Munich, Germany
| | - Elias Rackl
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich Site, Munich, Germany
| | - Lin Li
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich Site, Munich, Germany
| | - Christoph Schwepcke
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich Site, Munich, Germany
| | - Kai Rejeski
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich Site, Munich, Germany
| | - Christoph Schmid
- Department of Hematology and Oncology, University Hospital of Augsburg, Augsburg, Germany
- Department of Hematology and Oncology, Diakonieklinikum Stuttgart, Stuttgart, Germany
| | - Andreas Rank
- Bavarian Cancer Research Center (BZKF), Munich Site, Munich, Germany
- Department of Hematology and Oncology, University Hospital of Augsburg, Augsburg, Germany
| | - Jörg Schmohl
- Department of Hematology and Oncology, Diakonieklinikum Stuttgart, Stuttgart, Germany
| | - Doris Kraemer
- Department of Hematology and Oncology, St.-Josefs-Hospital, Hagen, Germany
| | - Peter Bojko
- Department of Hematology and Oncology, Rotkreuzklinikum Munich, Munich, Germany
| | - Helga Maria Schmetzer
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich Site, Munich, Germany
| |
Collapse
|
65
|
Liao S, Li X, Lu Y, Luo K. Nanomedicine in Immunotherapy for Non-Small Cell Lung Cancer: Applications and Perspectives. SMALL METHODS 2025:e2401783. [PMID: 39871783 DOI: 10.1002/smtd.202401783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/17/2025] [Indexed: 01/29/2025]
Abstract
Non-small cell lung cancer (NSCLC) has a strikingly high incidence rate globally. Although immunotherapy brings a great breakthrough in its clinical treatment of NSCLC, significant challenges still need to be overcome. The development of novel multi-functional nanomedicines in the realm of tumor immunotherapy offers promising opportunities for NSCLC patients, as nanomedicines exhibit significant advantages, including specific targeting of tumor cells, improved drug bioavailability, reduced systemic toxicity, and overcoming of immune resistance. In this review, the core features and current clinical status of strategies for NSCLC immunotherapy including immune checkpoint blockade, antibody-drug conjugates, cell engagers, adoptive cells, and cancer vaccines, are surveyed. Particular emphasis is placed on the recent development of nanomedicines that boost these strategies. Nanomedicine can provide novel perspectives for NSCLC immunotherapy.
Collapse
Affiliation(s)
- Shuangsi Liao
- Division of Thoracic Tumor Multimodality Treatment, Department of Radiation Oncology, Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Cancer Center, Breast Center, Institute of Breast Health Medicine, Laboratory of Clinical Cell Therapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoling Li
- Division of Thoracic Tumor Multimodality Treatment, Department of Radiation Oncology, Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Cancer Center, Breast Center, Institute of Breast Health Medicine, Laboratory of Clinical Cell Therapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - You Lu
- Division of Thoracic Tumor Multimodality Treatment, Department of Radiation Oncology, Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Cancer Center, Breast Center, Institute of Breast Health Medicine, Laboratory of Clinical Cell Therapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kui Luo
- Division of Thoracic Tumor Multimodality Treatment, Department of Radiation Oncology, Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Cancer Center, Breast Center, Institute of Breast Health Medicine, Laboratory of Clinical Cell Therapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
66
|
Chen Z, Zeng C, Yang L, Che Y, Chen M, Sau L, Wang B, Zhou K, Chen Y, Qing Y, Shen C, Zhang T, Wunderlich M, Wu D, Li W, Wang K, Leung K, Sun M, Tang T, He X, Zhang L, Swaminathan S, Mulloy JC, Müschen M, Huang H, Weng H, Xiao G, Deng X, Chen J. YTHDF2 promotes ATP synthesis and immune evasion in B cell malignancies. Cell 2025; 188:331-351.e30. [PMID: 39694037 DOI: 10.1016/j.cell.2024.11.007] [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: 11/18/2023] [Revised: 04/21/2024] [Accepted: 11/08/2024] [Indexed: 12/20/2024]
Abstract
Long-term durable remission in patients with B cell malignancies following chimeric antigen receptor (CAR)-T cell immunotherapy remains unsatisfactory, often due to antigen escape. Malignant B cell transformation and oncogenic growth relies on efficient ATP synthesis, although the underlying mechanisms remain unclear. Here, we report that YTHDF2 facilitates energy supply and antigen escape in B cell malignancies, and its overexpression alone is sufficient to cause B cell transformation and tumorigenesis. Mechanistically, YTHDF2 functions as a dual reader where it stabilizes mRNAs as a 5-methylcytosine (m5C) reader via recruiting PABPC1, thereby enhancing their expression and ATP synthesis. Concomitantly, YTHDF2 also promotes immune evasion by destabilizing other mRNAs as an N6-methyladenosine (m6A) reader. Small-molecule-mediated targeting of YTHDF2 suppresses aggressive B cell malignancies and sensitizes them to CAR-T cell therapy.
Collapse
Affiliation(s)
- Zhenhua Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Center for RNA Biology and Therapeutics, City of Hope Beckman Research Institute, Duarte, CA 91010, USA.
| | - Chengwu Zeng
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Center for RNA Biology and Therapeutics, City of Hope Beckman Research Institute, Duarte, CA 91010, USA; Jinan University Institute of Hematology, and Department of Hematology, The Fifth Affiliated Hospital Guangzhou Medical University, Guangzhou 510700, China
| | - Lu Yang
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Center for RNA Biology and Therapeutics, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| | - Yuan Che
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Center for RNA Biology and Therapeutics, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| | - Meiling Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Center for RNA Biology and Therapeutics, City of Hope Beckman Research Institute, Duarte, CA 91010, USA; Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| | - Lillian Sau
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Center for RNA Biology and Therapeutics, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| | - Bintao Wang
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Center for RNA Biology and Therapeutics, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| | - Keren Zhou
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Center for RNA Biology and Therapeutics, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| | - Yu Chen
- Molecular Instrumentation Center, University of California, Los Angeles, CA 90095, USA
| | - Ying Qing
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Center for RNA Biology and Therapeutics, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| | - Chao Shen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Center for RNA Biology and Therapeutics, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| | - Tingjian Zhang
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang 110122, China
| | - Mark Wunderlich
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Dong Wu
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Center for RNA Biology and Therapeutics, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| | - Wei Li
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Center for RNA Biology and Therapeutics, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| | - Kitty Wang
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Center for RNA Biology and Therapeutics, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| | - Keith Leung
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Center for RNA Biology and Therapeutics, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| | - Miao Sun
- Keck School of Medicine, University of Southern California, and Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Tingting Tang
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Center for RNA Biology and Therapeutics, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| | - Xin He
- Department of Hematological Malignancies Translational Science, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Lianjun Zhang
- Department of Hematological Malignancies Translational Science, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Srividya Swaminathan
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - James C Mulloy
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Markus Müschen
- Center of Molecular and Cellular Oncology, and Department of Immunobiology, Yale University, New Haven, CT 06511, USA
| | - Huilin Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Hengyou Weng
- Guangzhou Laboratory, Guangzhou, Guangdong 510005, China
| | - Gang Xiao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Xiaolan Deng
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Center for RNA Biology and Therapeutics, City of Hope Beckman Research Institute, Duarte, CA 91010, USA.
| | - Jianjun Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Center for RNA Biology and Therapeutics, City of Hope Beckman Research Institute, Duarte, CA 91010, USA.
| |
Collapse
|
67
|
O'Shea PJ, Johnson PC, El-Jawahri A, Leblanc TW. Unmet needs and lived experience of patients receiving CAR T-cell therapy. Leuk Lymphoma 2025:1-11. [PMID: 39838695 DOI: 10.1080/10428194.2025.2455488] [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: 09/03/2024] [Revised: 12/16/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025]
Abstract
Chimeric Antigen Receptor T-Cell (CAR-T) therapy is an effective therapy and promising frontier in the treatment of hematologic malignancies. However, this revolutionary treatment has led to new challenges for patients, caregivers, and the healthcare system. In this review article, we discuss the various difficulties patients face both in the acute and long-term period following CAR-T infusion. We highlight the various ways these difficulties are addressed, as well as further areas of research and support needed to improve patient experience. Additionally, we consider the difficulties and burdens placed on caregivers and healthcare systems, as well as barriers to accessing CAR-T therapy. Finally, we address future directions of research and intervention development to meet patient and caregiver needs and improve equitable access. We pose early integration of specialty palliative care for individuals and their caregivers undergoing CAR-T therapy as one promising strategy to help improve patient experience and meet their needs.
Collapse
Affiliation(s)
- Patrick J O'Shea
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Patrick Connor Johnson
- Division of Hematology & Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Areej El-Jawahri
- Division of Hematology & Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Thomas W Leblanc
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Cancer Institute, Durham, NC, USA
| |
Collapse
|
68
|
Li YR, Zhou K, Lee D, Zhu Y, Halladay T, Yu J, Zhou Y, Lyu Z, Fang Y, Chen Y, Semaan S, Yang L. Generating allogeneic CAR-NKT cells for off-the-shelf cancer immunotherapy with genetically engineered HSP cells and feeder-free differentiation culture. Nat Protoc 2025:10.1038/s41596-024-01077-w. [PMID: 39825143 DOI: 10.1038/s41596-024-01077-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/20/2024] [Indexed: 01/20/2025]
Abstract
The clinical potential of current chimeric antigen receptor-engineered T (CAR-T) cell therapy is hampered by its autologous nature that poses considerable challenges in manufacturing, costs and patient selection. This spurs demand for off-the-shelf therapies. Here we introduce an ex vivo feeder-free culture method to differentiate gene-engineered hematopoietic stem and progenitor (HSP) cells into allogeneic invariant natural killer T (AlloNKT) cells and their CAR-armed derivatives (AlloCAR-NKT cells). We include detailed information on lentivirus generation and titration, as well as the five stages of ex vivo culture required to generate AlloCAR-NKT cells, including HSP cell engineering, HSP cell expansion, NKT cell differentiation, NKT cell deep differentiation and NKT cell expansion. In addition, we describe procedures for evaluating the pharmacology, antitumor efficacy and mechanism of action of AlloCAR-NKT cells. It takes ~2 weeks to generate and titrate lentiviruses and ~6 weeks to generate mature AlloCAR-NKT cells. Competence with human stem cell and T cell culture, gene engineering and flow cytometry is required for optimal results.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kuangyi Zhou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Derek Lee
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tyler Halladay
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jiaji Yu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yang Zhou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zibai Lyu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ying Fang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yuning Chen
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sasha Semaan
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lili Yang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Centre of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
69
|
Bangolo A, Amoozgar B, Mansour C, Zhang L, Gill S, Ip A, Cho C. Comprehensive Review of Early and Late Toxicities in CAR T-Cell Therapy and Bispecific Antibody Treatments for Hematologic Malignancies. Cancers (Basel) 2025; 17:282. [PMID: 39858064 PMCID: PMC11764151 DOI: 10.3390/cancers17020282] [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: 12/24/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Chimeric antigen receptor T-cell (or CAR-T) therapy and bispecific antibodies (BsAbs) have revolutionized the treatment of hematologic malignancies, offering new options for relapsed or refractory cases. However, these therapies carry risks of early complications, such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), and delayed issues like graft-versus-host disease (GVHD), infections, and secondary cancers. Effective management requires early diagnosis using advanced biomarkers and imaging, along with prompt interventions involving immunosuppressants, corticosteroids, and cytokine inhibitors. A multidisciplinary approach is essential, integrating hematologists, oncologists, and infectious disease specialists, with emerging strategies like targeted biologics and personalized medicine showing promise in balancing efficacy with toxicity management. Ongoing research is critical to refine diagnostics and treatments, ensuring that these therapies not only extend survival but also improve patients' quality of life. This review provides critical insights for healthcare professionals to quickly recognize and treat complications of CAR-T and BsAbs therapies. By focusing on early detection through biomarkers and imaging and outlining timely therapeutic interventions, it aims to equip the multidisciplinary care team with the knowledge necessary to manage the challenges of these advanced treatments effectively, ultimately optimizing patient outcomes.
Collapse
Affiliation(s)
- Ayrton Bangolo
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA; (B.A.); (L.Z.); (S.G.)
| | - Behzad Amoozgar
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA; (B.A.); (L.Z.); (S.G.)
| | | | - Lili Zhang
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA; (B.A.); (L.Z.); (S.G.)
| | - Sarvarinder Gill
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA; (B.A.); (L.Z.); (S.G.)
| | - Andrew Ip
- Division of Lymphoma, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA;
| | - Christina Cho
- Division of Stem Cell Transplant and Cellular Therapy, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA;
| |
Collapse
|
70
|
Mandavkar AA, Padakanti SSN, Gupta S, Akram S, Jaffar N, Chauhan J, Allu LR, Saini P, Nasrallah J, Omar MA, Mugibel MA, Syed S, Ravindran KO, Dwivedi A, Dhingra GS, Dhingra A, Kakadiya J, Kotaich J, Beniwal SS. Emerging therapies in Multiple Myeloma: Leveraging immune checkpoint inhibitors for improved outcomes. Hum Antibodies 2025:10932607241301699. [PMID: 39973812 DOI: 10.1177/10932607241301699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
BACKGROUND: Multiple Myeloma is a hematological malignancy characterized by the proliferation of clonal plasma cells and associated with severe clinical manifestations. Despite advancements in diagnosis and management, Multiple Myeloma remains incurable, necessitating further research into more effective therapies. AIM: The primary objective of this review is to provide an informative and critical summary of the Multiple Myeloma microenvironment, and emerging revolutionary therapeutic approaches with potential combination therapy to improve the quality of life for Multiple Myeloma patients. EMERGING APPROACHES: Recent advancements in immunotherapy, particularly immune checkpoint inhibitors (ICIs), have shown improvements in immune response against Multiple Myeloma. ICIs target inhibitory pathways such as PD-1/PD-L1 and CTLA-4, potentially overcoming tumor-induced immunosuppression. Combination therapies integrate ICIs with proteasome inhibitors, immunomodulators, and monoclonal antibodies to enhance the anti-tumor immune response. Additionally, Chimeric Antigen Receptor T-cell (CAR-T) therapy has demonstrated effectiveness against Multiple Myeloma, particularly when coupled with ICIs to decrease resistance and relapse. CHALLENGES: Although the efficacy of ICIs in treating Multiple Myeloma has been hindered by the complexity of the tumor microenvironment and immune evasion mechanisms, this challenge has led to the exploration of combination therapies. Potential side effects are still a big challenge for newly recognized ICIs and combination treatment. FUTURE DIRECTIONS: Investigations of new immune checkpoints and the development of targeted therapies against these markers are in progress, creating possibilities for more personalized and effective treatment strategies. Continuous research and robust clinical trials are needed to comprehend the complex dynamics of the Multiple Myeloma microenvironment to develop revolutionary therapeutic targets.
Collapse
Affiliation(s)
| | | | - Srajan Gupta
- SV Medical College, Tirupati, Andhra Pradesh, India
| | - Samiyah Akram
- Shadan Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Nida Jaffar
- Mid and South Essex NHS Foundation Trust, Southend University Hospital, Southend-on-Sea Essex, England
| | - Jugalkishor Chauhan
- Dr. N D Desai Faculty of Medical Science and Research, Nadiad, Gujarat, India
| | | | - Pulkit Saini
- Sri Devaraj URS Medical College, Kolar, Karnataka, India
| | - Jamil Nasrallah
- Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | | | - Muna Ali Mugibel
- College of Medicine and Health Sciences, Hadhramout University, Mukalla, Yemen
| | - Saif Syed
- Royal College of Surgeons, Dublin, Ireland
| | | | - Ayush Dwivedi
- Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | | | - Avleen Dhingra
- Dayanand Medical College and Hospital, Ludhiana, Punjab, India
| | | | - Jana Kotaich
- Faculty of Medical Sciences, Lebanese University, Lebanon
| | | |
Collapse
|
71
|
Duell J, Westin J. The future of immunotherapy for diffuse large B-cell lymphoma. Int J Cancer 2025; 156:251-261. [PMID: 39319495 PMCID: PMC11578085 DOI: 10.1002/ijc.35156] [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: 08/22/2023] [Revised: 06/21/2024] [Accepted: 07/15/2024] [Indexed: 09/26/2024]
Abstract
With the introduction of anti-CD19 chimeric antigen receptor (CAR) T-cell (CAR T) therapies, bispecific CD3/CD20 antibodies and anti-CD19 antibodies, immunotherapy continues to transform the treatment of diffuse large B-cell lymphoma (DLBCL). A number of novel immunotherapeutic strategies are under investigation to build upon current clinical benefit and offer further options to those patients who cannot tolerate conventional intensive therapies due to their age and/or state of health. Alongside immunotherapies that leverage the adaptive immune response, natural killer (NK) cell and myeloid cell-engaging therapies can utilize the innate immune system. Monoclonal antibodies engineered for greater recognition by the patient's immune system can enhance antitumor cytotoxic mechanisms mediated by NK cells and macrophages. In addition, CAR technology is extending into NK cells and macrophages and investigational immune checkpoint inhibitors targeting macrophage/myeloid cell checkpoints via the CD47/SIRPα axis are in development. Regimens that engage both innate and adaptive immune responses may help to overcome resistance to current immunotherapies. Furthermore, combinations of immunotherapy and oncogenic pathway inhibitors to reprogram the immunosuppressive tumor microenvironment of DLBCL may also potentiate antitumor responses. As immunotherapy treatment options continue to expand, both in the first-line setting and further lines of therapy, understanding how to harness these immunotherapies and the potential for combination approaches will be important for the development of future DLBCL treatment approaches.
Collapse
Affiliation(s)
- Johannes Duell
- Department of Internal Medicine 2University Hospital of WürzburgWürzburgGermany
| | - Jason Westin
- Department of Lymphoma and MyelomaMD Anderson Cancer CenterHoustonTexasUSA
| |
Collapse
|
72
|
Zhou H, Zhu W, Ma Q, Liu N, Jin M, Feng Y, Zhao L, Sun R, Li R, Li H, Shi Y, Wang J, Liu L, Guo Z. Case report: The case of T-cell acute lymphoblastic leukemia treated with chemotherapy followed by anti-CD7 CAR-T cells using retroviral vector. Front Immunol 2025; 15:1519055. [PMID: 39877371 PMCID: PMC11772494 DOI: 10.3389/fimmu.2024.1519055] [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: 10/29/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
CD7-targeted chimeric antigen receptor-T (CAR-T) cell therapy has shown great promise in the treatment of relapsed/refractory T-cell acute lymphoblastic leukemia (T-ALL). In this study, we reported a case of a 34-year-old male patient with T-ALL who finally developed multi-line drug resistance and refractoriness after multiple lines of high-intensity chemotherapy. After physician evaluation, this patient received allogeneic hematopoietic stem cell transplantation (allo-HSCT). Then, The patient remained in complete remission (CR) for four months before a relapse with 26.64% chimerism rate, so he was treated with allogeneic anti-CD7 CAR-T cells after chemotherapy reducing the tumor burden. The CAR-T product was a novel anti-CD7 CAR-T based on retroviral vectors (RV). After infusion, the patient achieved CR within 1 month after anti-CD7 CAR-T infusion and the remission has been ongoing for 9 months to date. Cytokine release syndrome (CRS) 1 was experienced while no immune effector cell-associated neurotoxicity syndrome (ICANS) was found. In addition, CAR copy number peaked at 350, 758 copies/μg on day 6. This case report of clinical treatment of T-ALL with anti-CD7 CAR-T cells prepared using a retroviral vector without gene editing and combined with chemotherapy, which demonstrated that the RV-based anti-CD7 CAR-T cells had good therapeutic effect and high safety in triple-refractory T-ALL patients.
Collapse
Affiliation(s)
- Huanhuan Zhou
- Department of Hematology, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Wenxiang Zhu
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, China
| | - Qihong Ma
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, China
| | - Ning Liu
- Department of Hematology, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Mengdi Jin
- Department of Hematology, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Yaru Feng
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, China
| | - Lijun Zhao
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, China
| | - Rui Sun
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, China
| | - Rongyou Li
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, China
| | - Huaxiu Li
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, China
| | - Yuanyuan Shi
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, China
| | - Jianxun Wang
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, China
| | - Liqiong Liu
- Department of Hematology, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Zhi Guo
- Department of Hematology, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang, China
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
73
|
Bajwa A, Zhao Q, Geer M, Lin C, Westholder J, Maakaron J, Ghosh M, Frame D, Galal A, Tossey J, Ahmed N, Bezerra E, Denlinger N, de Lima M, Epperla N, Caimi P, Voorhees T. Siltuximab for chimeric antigen receptor T-cell therapy-related CRS and ICANS: a multicenter retrospective analysis. Blood Adv 2025; 9:170-175. [PMID: 39437770 PMCID: PMC11788129 DOI: 10.1182/bloodadvances.2024013688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/08/2024] [Accepted: 09/08/2024] [Indexed: 10/25/2024] Open
Abstract
ABSTRACT Chimeric antigen receptor T-cell (CAR-T) therapies are effective in many hematologic malignancies; however, adverse events including cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) can affect a significant number of patients. Those who develop refractory CRS or ICANS have few treatment options. Siltuximab, a monoclonal antibody binding circulating interleukin-6, has been proposed to have clinical activity in both CRS and ICANS. We conducted a multicenter retrospective analysis of siltuximab treatment for CRS and ICANS after CAR-T therapy in a real-world cohort from 6 academic centers. Fifty-four patients were evaluated. Sixteen patients had CRS previously treated with tocilizumab and 17 patients had ICANS previously treated with steroids. Of the patients with CRS at the time of siltuximab, 75% had improvement in CRS grade. Of the patients with ICANS at the time of siltuximab, 60% had improvement in ICANS grade. To our knowledge, this is the largest cohort of patients treated with siltuximab for CRS and/or ICANS after CAR-T therapies. Siltuximab appeared to be effective for both CRS and ICANS, including previously treated toxicities. These data support the use of siltuximab in CRS and ICANS as well as provide rationale for future prospective studies.
Collapse
Affiliation(s)
- Amneet Bajwa
- The James Comprehensive Cancer Center, Division of Hematology, The Ohio State University, Columbus, OH
| | - Qiuhong Zhao
- The James Comprehensive Cancer Center, Division of Hematology, The Ohio State University, Columbus, OH
| | - Marcus Geer
- Rogel Cancer Center, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI
| | - Chenyu Lin
- Duke Cancer Center, Division of Hematology, Duke University, Durham, NC
| | - James Westholder
- Masonic Cancer Center, Division of Hematology and Oncology, University of Minnesota, Minneapolis, MN
| | - Joseph Maakaron
- Masonic Cancer Center, Division of Hematology and Oncology, University of Minnesota, Minneapolis, MN
| | - Monalisa Ghosh
- Rogel Cancer Center, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI
| | - David Frame
- Rogel Cancer Center, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI
| | - Ahmed Galal
- Duke Cancer Center, Division of Hematology, Duke University, Durham, NC
| | - Justin Tossey
- The James Comprehensive Cancer Center, Division of Hematology, The Ohio State University, Columbus, OH
| | - Nausheen Ahmed
- University of Kansas Cancer Center, Division of Hematologic Malignancies and Cellular Therapeutics, Overland Park, KS
| | - Evandro Bezerra
- The James Comprehensive Cancer Center, Division of Hematology, The Ohio State University, Columbus, OH
| | - Nathan Denlinger
- The James Comprehensive Cancer Center, Division of Hematology, The Ohio State University, Columbus, OH
| | - Marcos de Lima
- The James Comprehensive Cancer Center, Division of Hematology, The Ohio State University, Columbus, OH
| | - Narendranath Epperla
- The James Comprehensive Cancer Center, Division of Hematology, The Ohio State University, Columbus, OH
| | - Paolo Caimi
- Taussig Cancer Institute, Division of Hematology and Medical Oncology, Cleveland Clinic Foundation, Cleveland, OH
| | - Timothy Voorhees
- The James Comprehensive Cancer Center, Division of Hematology, The Ohio State University, Columbus, OH
| |
Collapse
|
74
|
Zong J, Li YR. iPSC Technology Revolutionizes CAR-T Cell Therapy for Cancer Treatment. Bioengineering (Basel) 2025; 12:60. [PMID: 39851334 PMCID: PMC11763340 DOI: 10.3390/bioengineering12010060] [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: 11/15/2024] [Revised: 01/03/2025] [Accepted: 01/10/2025] [Indexed: 01/26/2025] Open
Abstract
Chimeric Antigen Receptor (CAR)-engineered T (CAR-T) cell therapy represents a highly promising modality within the domain of cancer treatment. CAR-T cell therapy has demonstrated notable efficacy in the treatment of hematological malignancies, solid tumors, and various infectious diseases. However, current CAR-T cell therapy is autologous, which presents challenges related to high costs, time-consuming manufacturing processes, and the necessity for careful patient selection. A potential resolution to this restriction could be found by synergizing CAR-T technology with the induced pluripotent stem cell (iPSC) technology. iPSC technology has the inherent capability to furnish an inexhaustible reservoir of T cell resources. Experimental evidence has demonstrated the successful generation of various human CAR-T cells using iPSC technology, showcasing high yield, purity, robustness, and promising tumor-killing efficacy. Importantly, this technology enables the production of clinical-grade CAR-T cells, significantly reducing manufacturing costs and time, and facilitating their use as allogeneic cell therapies to treat multiple cancer patients simultaneously. In this review, we aim to elucidate essential facets of current cancer therapy, delineate its utility, enumerate its advantages and drawbacks, and offer an in-depth evaluation of a novel and pragmatic approach to cancer treatment.
Collapse
Affiliation(s)
| | - Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
75
|
Cai L, Wen X, Qiu Z, Fu A, Guo D, Zhu M. Characteristics, risk factors and a risk prediction model of tocilizumab-induced hypofibrinogenemia: a retrospective real-world study of inpatients. BMC Pharmacol Toxicol 2025; 26:5. [PMID: 39789607 PMCID: PMC11721054 DOI: 10.1186/s40360-024-00827-6] [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: 08/16/2024] [Accepted: 12/17/2024] [Indexed: 01/12/2025] Open
Abstract
OBJECTIVE The occurrence of hypofibrinogenemia after tocilizumab treatment has attracted increasing attention, which may cause bleeding and even life-threatening. This study aims to explore the risk factors for tocilizumab-induced hypofibrinogenemia (T-HFIB) and construct a risk prediction model. METHODS A total of 221 inpatients that received tocilizumab from 2015 to 2023 were retrospectively collected and divided into T-HFIB group or control group. The risk factors for T-HFIB were obtained by logistic regression equation and used to establish the nomogram. RESULTS T-HFIB was observed in 121 of 221 patients (54.75%). Multifactorial logistic regression analysis revealed that infection (OR = 2.002, 95%CI:1.018 ~ 3.935), COVID-19 (OR = 3.752, 95%CI:1.264 ~ 11.139), CAR-T therapy (OR = 4.409, 95%CI:2.017 ~ 0.894), and concomitant glucocorticoids (OR = 5.303, 95%CI:0.227 ~ 0.894) were identified as independent risk factors for T-HFIB, while high baseline fibrinogen level (OR = 0.813, 95%CI:0.670 ~ 0.988) and concomitant antirheumatic drugs (OR = 0.451, 95%CI:0.227 ~ 0.894) were identified as protective factors. A nomogram was established, and area under the curve (AUC) of prediction model was 0.772 (95%CI:0.709 ~ 0.836). Calibration curve showed a good prediction accuracy for the occurrence of T-HFIB. CONCLUSION The infection, COVID-19, CAR-T therapy, and concomitant glucocorticoids were independent risk factors for T-HFIB, while high baseline fibrinogen and concomitant antirheumatic drugs were protective factors. This nomogram can help early identify the patients at potential high risk of developing T-HFIB.
Collapse
Affiliation(s)
- Le Cai
- Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, 28 Fu Xing Road, Beijing, 100853, China
| | - Xiao Wen
- Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, 28 Fu Xing Road, Beijing, 100853, China
| | - Zihan Qiu
- School of Pharmacy, Tian Jin Medical University, Tianjin, 300070, China
| | - An Fu
- Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, 28 Fu Xing Road, Beijing, 100853, China
| | - Daihong Guo
- Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, 28 Fu Xing Road, Beijing, 100853, China
| | - Man Zhu
- Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, 28 Fu Xing Road, Beijing, 100853, China.
| |
Collapse
|
76
|
Carter S, Wigmore T. Immunotherapy on ICU: a narrative review. Anaesthesia 2025. [PMID: 39776055 DOI: 10.1111/anae.16453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Patients with cancer account for 15% of all admissions to critical care and so an understanding of the pathophysiology and anticipated complications of specialist treatment is essential for the intensive care clinician. The development of chimeric antigen receptor T-cell therapy for haematological malignancies and immune checkpoint inhibitors for solid organ tumours has led to significant improvements in the prognosis of those patients whose tumours respond. This review is intended to provide the non-specialist with an understanding of the current concepts in pathophysiology, diagnosis and management of complications due to chimeric antigen receptor T-cell therapy and immune checkpoint inhibitors for malignant disease. METHODS We performed searches of electronic databases to identify relevant peer-reviewed publications in the literature. Basic science; clinical trials; cohort studies; systematic reviews; meta-analyses; and guidelines were eligible for inclusion. Abstracts were screened to identify publications relevant to immune effector cell toxicities of chimeric antigen receptor T-cell therapy and immune-related adverse events of immune checkpoint inhibitors. RESULTS While the pathophysiology for toxicities due to chimeric antigen receptor T-cells and immune checkpoint inhibitors remains incompletely understood, targeted drug therapies have been successfully implemented for toxicities such as cytokine release syndrome. Corticosteroids remain an important component of pharmacological management. The diagnosis of toxicities remains largely clinical, and a high index of suspicion should remain for infective complications. Management of toxicities should be undertaken in conjunction with the patient's primary oncologist. CONCLUSION Despite significant advances in the development of targeted immunotherapy, the mechanism of action for the resultant toxicities remains poorly understood and limits the development of predictive models, diagnostic biomarkers and highly effective treatment options. Further research is needed to identify treatment regimens which minimise the use of corticosteroids in chimeric antigen receptor T-cell and immune checkpoint inhibitor-associated toxicities.
Collapse
Affiliation(s)
- Sef Carter
- East of England School of Anaesthesia, Basildon, UK
| | - Timothy Wigmore
- Department of Critical Care, The Royal Marsden Hospital, London, UK
| |
Collapse
|
77
|
Matthews AJ, Starkie FE, Staniaszek LE, Kane NM. The Role of Electroencephalography Following CAR-T Cell Therapy in Clinical Practice. Clin EEG Neurosci 2025:15500594241312451. [PMID: 39773224 DOI: 10.1177/15500594241312451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Objectives:Neurotoxicity, encephalopathy, and seizures can occur following chimeric antigen receptor (CAR)-T cell therapy. Our aim was to assess what value electroencephalography (EEG) offers for people undergoing CAR-T treatment in clinical practice, including possible diagnostic, management, and prognostic roles. Methods: All patients developing CAR-T related neurotoxicity referred for EEG were eligible for inclusion. Reasons for EEG referral and qualitative EEG findings were analysed and reported. The relationship between objective quantitative EEG (QEEG) encephalopathy grade and clinical neurotoxicity (immune effector cell-associated neurotoxicity syndrome; ICANS) grade was determined. The prognostic ability of QEEG grade was assessed for survival and functional status. Results: Twenty-eight patients with 53 EEG recordings were included. Common reasons given on EEG referrals were possible seizure diagnosis (n = 38), reduced consciousness (n = 8), and superimposed cerebral infection (n = 4). Four focal seizures were detected on three (3/53; 5.7%) EEGs. There was a moderately positive correlation between QEEG grade and ICANS grade (r = + 0.41, p = .030). QEEG grade could not predict survival at 3 months (Area Under Curve; AUC = 0.673) or 6 months (AUC = 0.578), nor could it predict functional status at 1 month (r = + 0.40; p = .080), 3 months (r = + 0.19; p = .439), or time to return to baseline (r = + 0.32; p = .156). Conclusions: EEG was useful in seizure diagnosis. QEEG has a possible role as a specific biomarker of encephalopathy/neurotoxicity. EEG generated no tangible changes in patient management. QEEG was unable to prognosticate survival or functional status.
Collapse
Affiliation(s)
- Alexander J Matthews
- Neurophysiology Department, University Hospitals Bristol and Weston NHS Foundation Trust, Upper Maudlin Street, Bristol, BS2 8BJ, UK
| | - Fiona E Starkie
- Bristol Haematology and Oncology Centre, University Hospitals Bristol and Weston NHS Foundation Trust, Horfield Road, Bristol, BS2 8ED, UK
| | - Lydia E Staniaszek
- Neurophysiology Department, University Hospitals Bristol and Weston NHS Foundation Trust, Upper Maudlin Street, Bristol, BS2 8BJ, UK
| | - Nicholas M Kane
- Neurophysiology Department, University Hospitals Bristol and Weston NHS Foundation Trust, Upper Maudlin Street, Bristol, BS2 8BJ, UK
| |
Collapse
|
78
|
Jambon S, Sun J, Barman S, Muthugounder S, Bito XR, Shadfar A, Kovach AE, Wood BL, Thoppey Manoharan V, Morrissy AS, Bhojwani D, Wayne AS, Pulsipher MA, Kim YM, Asgharzadeh S, Parekh C, Moghimi B. CD33-CD123 IF-THEN Gating Reduces Toxicity while Enhancing the Specificity and Memory Phenotype of AML-Targeting CAR-T Cells. Blood Cancer Discov 2025; 6:55-72. [PMID: 39624992 PMCID: PMC11707512 DOI: 10.1158/2643-3230.bcd-23-0258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 06/11/2024] [Accepted: 11/08/2024] [Indexed: 01/11/2025] Open
Abstract
SIGNIFICANCE Our study demonstrates the use of "IF-THEN" SynNotch-gated CAR-T cells targeting CD33 and CD123 in AML reduces off-tumor toxicity. This strategy enhances T-cell phenotype, improves expansion, preserves HSPCs, and mitigates cytokine release syndrome-addressing critical limitations of existing AML CAR-T therapies.
Collapse
MESH Headings
- Humans
- Sialic Acid Binding Ig-like Lectin 3/metabolism
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/therapy
- Interleukin-3 Receptor alpha Subunit/metabolism
- Immunotherapy, Adoptive/methods
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Phenotype
- Animals
Collapse
Affiliation(s)
- Samy Jambon
- Division of Hematology and Oncology, Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Jianping Sun
- Division of Hematology and Oncology, Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Shawn Barman
- Division of Hematology and Oncology, Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Sakunthala Muthugounder
- Division of Hematology and Oncology, Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Xue Rachel Bito
- Division of Hematology and Oncology, Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Armita Shadfar
- Division of Hematology and Oncology, Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Alexandra E. Kovach
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Brent L. Wood
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | | | - A. Sorana Morrissy
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
| | - Deepa Bhojwani
- Division of Hematology and Oncology, Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Alan S. Wayne
- Division of Hematology and Oncology, Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Michael A. Pulsipher
- Division of Hematology and Oncology, Primary Children’s Hospital, Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine at the University of Utah, Salt Lake City, Utah
| | - Yong-Mi Kim
- Division of Hematology and Oncology, Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Shahab Asgharzadeh
- Division of Hematology and Oncology, Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Chintan Parekh
- Division of Hematology and Oncology, Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Babak Moghimi
- Division of Hematology and Oncology, Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
79
|
Pinto E, Lione L, Compagnone M, Paccagnella M, Salvatori E, Greco M, Frezza V, Marra E, Aurisicchio L, Roscilli G, Conforti A. From ex vivo to in vivo chimeric antigen T cells manufacturing: new horizons for CAR T-cell based therapy. J Transl Med 2025; 23:10. [PMID: 39755643 PMCID: PMC11700462 DOI: 10.1186/s12967-024-06052-3] [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: 09/19/2024] [Accepted: 12/24/2024] [Indexed: 01/06/2025] Open
Abstract
In the past decades, Chimeric Antigen Receptor (CAR)-T cell therapy has achieved remarkable success, leading to the approval of six therapeutic products for haematological malignancies. Recently, the therapeutic potential of this therapy has also been demonstrated in non-tumoral diseases. Currently, the manufacturing process to produce clinical-grade CAR-T cells is complex, time-consuming, and highly expensive. It involves multiple steps, including the collection of T cells from patients or healthy donors, in vitro engineering and expansion, and finally reinfusion into patients. Therefore, despite the impressive clinical outcomes, ex vivo manufacturing process makes CAR-T cells out of reach for many cancer patients. Direct in vivo engineering of T cells could be a more rapid solution able to circumvent both the complexity and the costs associated with ex vivo manufactured CAR-T cells. This novel approach allows to completely eliminate ex vivo cell manipulation and expansion while producing therapeutic cell populations directly in vivo. To date, several studies have demonstrated the feasibility of in vivo T cell reprogramming, by employing injectable viral- or nanocarrier-based delivery platforms in tumour animal models. Additionally, in vivo production of CAR-T cells might reduce the incidence, or at least the severity, of systemic toxicities frequently occurring with ex vivo produced CAR-T cells, such as cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. In this review, we highlight the challenges associated with the current ex vivo manufacturing protocols and review the latest progresses in the emerging field of in vivo CAR-T therapy, by comparing the various platforms so far investigated. Moreover, we offer an overview of the advantages deriving from in vivo reprogramming of other immune cell types, such as Natural Killer and macrophages, with CAR constructs.
Collapse
Affiliation(s)
- E Pinto
- Evvivax Biotech, Via Castel Romano 100, 00128, Rome, Italy
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - L Lione
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - M Compagnone
- Evvivax Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - M Paccagnella
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - E Salvatori
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - M Greco
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - V Frezza
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - E Marra
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - L Aurisicchio
- Evvivax Biotech, Via Castel Romano 100, 00128, Rome, Italy
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - G Roscilli
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy
| | - A Conforti
- Evvivax Biotech, Via Castel Romano 100, 00128, Rome, Italy.
- Takis Biotech, Via Castel Romano 100, 00128, Rome, Italy.
| |
Collapse
|
80
|
He X, Hu B, Zhang Y, Liu F, Li Q, Zheng C, Shen J, Yang Z, Wang J, Ma D, Qian C, Lu M, Mao J. Treatment of two pediatric patients with refractory systemic lupus erythematosus using CD19-targeted CAR T-cells. Autoimmun Rev 2025; 24:103692. [PMID: 39561867 DOI: 10.1016/j.autrev.2024.103692] [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: 10/27/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has emerged as a promising strategy for treating autoimmune diseases, particularly in systemic lupus erythematosus (SLE). However, CAR T-cell therapy for pediatric patients with SLE remains unexplored. Herein, we present a compelling investigation of two pediatric patients with refractory SLE who underwent infusion of CD19-targeted CAR T-cells. Patients 1 and 2 tolerated the CAR T-cell therapy well, which demonstrated remarkable efficacy after five and four months, respectively.
Collapse
Affiliation(s)
- Xue He
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National clinical research center for child health, Hangzhou 310052, China
| | - Bin Hu
- Department of Allergy Immunology and Rheumatology, Children's Hospital, Zhejiang University School of Medicine, National clinical research center for child health, Hangzhou 310052, China
| | - Yingzi Zhang
- Chongqing Precision Biotech Co., Ltd., Chongqing 400039, China
| | - Fei Liu
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National clinical research center for child health, Hangzhou 310052, China
| | - Qiuyu Li
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National clinical research center for child health, Hangzhou 310052, China
| | - Chen Zheng
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National clinical research center for child health, Hangzhou 310052, China
| | - Junjie Shen
- Chongqing Precision Biotech Co., Ltd., Chongqing 400039, China
| | - Zhi Yang
- Chongqing Precision Biotech Co., Ltd., Chongqing 400039, China
| | - Jingjing Wang
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National clinical research center for child health, Hangzhou 310052, China
| | - Daqing Ma
- Perioperative and Systems Medicine Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China; Division of Anesthetics, Pain Medicine & Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom
| | - Cheng Qian
- Chongqing Precision Biotech Co., Ltd., Chongqing 400039, China.
| | - Meiping Lu
- Department of Allergy Immunology and Rheumatology, Children's Hospital, Zhejiang University School of Medicine, National clinical research center for child health, Hangzhou 310052, China.
| | - Jianhua Mao
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National clinical research center for child health, Hangzhou 310052, China.
| |
Collapse
|
81
|
Chhabra L, Pandey RK, Kumar R, Sundar S, Mehrotra S. Navigating the Roadblocks: Progress and Challenges in Cell-Based Therapies for Human Immunodeficiency Virus. J Cell Biochem 2025; 126:e30669. [PMID: 39485037 DOI: 10.1002/jcb.30669] [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: 07/30/2024] [Revised: 09/26/2024] [Accepted: 10/11/2024] [Indexed: 11/03/2024]
Abstract
Cell-based therapies represent a major advancement in the treatment and management of HIV/AIDS, with a goal to overcome the limitations of traditional antiretroviral therapy (ART). These innovative approaches not only promise a functional cure by reconstructing the immune landscape but also address the persistent viral reservoirs. For example, stem cell therapies have emerged from the foundational success of allogeneic hematopoietic stem cell transplantation in curing HIV infection in a limited number of cases. B cell therapies make use of genetically modified B cells constitutively expressing broadly neutralizing antibodies (bNAbs) against target viral particles and infected cells. Adoptive cell transfer (ACT), including TCR-T therapy, CAR-T cells, NK-CAR cells, and DC-based therapy, is adapted from cancer immunotherapy and repurposed for HIV eradication. In this review, we summarize the mechanisms through which these engineered cells recognize and destroy HIV-infected cells, the modification strategies, and their role in sustaining remission in the absence of ART. The review also addresses the challenges to cell-based therapies against HIV and discusses the recent advancements aimed at overcoming them.
Collapse
Affiliation(s)
- Lakshay Chhabra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | | | - Rajiv Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| |
Collapse
|
82
|
Kanbay M, Mizrak B, Alper EN, Copur S, Ortiz A. Acute kidney injury following CAR-T cell therapy: a nephrologist's perspective. Clin Kidney J 2025; 18:sfae359. [PMID: 39781479 PMCID: PMC11704793 DOI: 10.1093/ckj/sfae359] [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: 09/28/2024] [Indexed: 01/12/2025] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy, an emerging personalized immunotherapy for various haematologic malignancies, autoimmune diseases and other conditions, involves the modification of patients' T cells to express a chimeric antigen receptor that recognizes tumour or autoimmune cell antigens, allowing CAR-T cells to destroy cancerous and other target cells selectively. Despite remarkable clinical improvements in patients, multiple adverse effects have been associated with CAR-T cell therapy. Among the most recognized adverse effects are cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome and tumour lysis syndrome. Even though less recognized, the incidence of acute kidney injury (AKI) ranges from 5 to 33%. The wide range of reported AKI incidence rates might depend on patient population characteristics and comorbidities and specific CAR-T cell therapy features. Even though the exact pathophysiology remains unknown, several key mechanisms, including cytokine release syndrome, tumour lysis syndrome and other factors such as direct renal toxicity of CAR-T cell therapy, conditioning regimens or other medications (e.g. antibiotics), and infectious complications (e.g. sepsis) have been proposed. Risk factors for CAR-T-related AKI include lower baseline glomerular filtration rate, higher rates of allopurinol or rasburicase use, intravenous contrast material exposure, elevated baseline lactate dehydrogenase and grade 3 or higher cytokine release syndrome. Future prospective studies with larger patient populations are needed to gain insights into the pathophysiology of CAR-T-related AKI and, more importantly, to be able to prevent as well as to develop novel and more efficient treatment modalities. In this narrative review, we discuss the underlying pathophysiology, risk factors, potential interventions and future directions related to AKI following CAR-T cell therapy.
Collapse
Affiliation(s)
- Mehmet Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey
| | - Berk Mizrak
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Ezgi N Alper
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Sidar Copur
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Alberto Ortiz
- Department of Medicine, Universidad Autonoma de Madrid and IIS-Fundacion Jimenez Diaz, Madrid, Spain
| |
Collapse
|
83
|
Cwynarski K, Iacoboni G, Tholouli E, Menne T, Irvine DA, Balasubramaniam N, Wood L, Shang J, Xue E, Zhang Y, Basilico S, Neves M, Raymond M, Scott I, El-Kholy M, Jha R, Dainton-Smith H, Hussain R, Day W, Ferrari M, Thomas S, Lilova K, Brugger W, Marafioti T, Lao-Sirieix P, Maciocia P, Pule M. TRBC1-CAR T cell therapy in peripheral T cell lymphoma: a phase 1/2 trial. Nat Med 2025; 31:137-143. [PMID: 39528665 PMCID: PMC11750712 DOI: 10.1038/s41591-024-03326-7] [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: 01/18/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024]
Abstract
Relapsed/refractory peripheral T cell lymphomas (PTCLs) are aggressive tumors with a poor prognosis. Unlike B cell lymphomas, treatment of PTCL has not benefited from advances in immunotherapy. This is largely due to a lack of suitable target antigens that discriminate malignant from normal T cells, thus avoiding severe immunosuppression consequent to depletion of the entire T cell compartment. We recently described a targeting strategy based on the mutually exclusive expression of T cell antigen receptor beta-chain constant domain (TRBC) 1 and 2. Selective targeting of the T cell antigen receptor beta-chain expressed by the (clonal) malignancy spares normal T cells expressing the other chain. The LibraT1 study is an ongoing, multicenter, international, single-arm phase 1/2 study of TRBC1-directed autologous chimeric antigen receptor (CAR) T cells (AUTO4) in relapsed/refractory TRBC1-positive PTCL. Primary objectives were assessment of safety and tolerability of AUTO4 infusion. Key secondary endpoints included efficacy, CAR T cell expansion and persistence. Here we describe the findings from dose escalation in LibraT1 in the first ten patients, in a non-prespecified interim analysis. AUTO4 resulted in low frequency of severe immunotoxicity, with one of ten patients developing grade 3 cytokine release syndrome. Complete metabolic response was observed in four of ten evaluable patients, with remissions being durable beyond 1 year in two patients. While an absence of circulating CAR T cells was observed, CAR T cells were readily detected in lymph node biopsy samples from sites of original disease suggesting homing to tumor sites. These results support the continuing exploration of TRBC1 targeting in PTCL. ClinicalTrials.gov registration: NCT03590574 .
Collapse
MESH Headings
- Humans
- Lymphoma, T-Cell, Peripheral/therapy
- Lymphoma, T-Cell, Peripheral/immunology
- Lymphoma, T-Cell, Peripheral/pathology
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Male
- Female
- Middle Aged
- Adult
- Aged
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Kate Cwynarski
- Department of Haematology, University College London, London, UK
- University College London Hospital, London, UK
| | - Gloria Iacoboni
- Department of Haematology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Eleni Tholouli
- Department of Haematology, Manchester Royal Infirmary, Manchester, UK
| | - Tobias Menne
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | | | - Leigh Wood
- University College London Hospital, London, UK
| | | | - Eric Xue
- Autolus Therapeutics, London, UK
| | | | | | | | | | | | | | - Ram Jha
- Autolus Therapeutics, London, UK
| | | | | | | | | | | | | | | | | | | | - Paul Maciocia
- Department of Haematology, University College London, London, UK
- University College London Hospital, London, UK
| | - Martin Pule
- Department of Haematology, University College London, London, UK.
- Autolus Therapeutics, London, UK.
| |
Collapse
|
84
|
Melica G, Luna de Abia A, Shah GL, Devlin S, Corona M, Fein J, Dahi PB, Giralt SA, Lin RJ, Palomba ML, Parascondola A, Park J, Salles G, Saldia A, Scordo M, Shouval R, Perales MA, Seo SK. Shift from Widespread to Tailored Antifungal Prophylaxis in Lymphoma Patients Treated with CD19 CAR T Cell Therapy: Results from a Large Retrospective Cohort. Transplant Cell Ther 2025; 31:36-44. [PMID: 39448032 PMCID: PMC11780678 DOI: 10.1016/j.jtct.2024.10.010] [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: 05/10/2024] [Revised: 09/19/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024]
Abstract
Patients undergoing CD19 chimeric antigen receptor (CAR)-T cell therapy exhibit multiple immune deficits that may increase their susceptibility to infections. Invasive fungal infections (IFIs) are life-threatening events in the setting of hematologic diseases. However, there is ongoing debate regarding the optimal role and duration of antifungal prophylaxis in this specific patient population. The objective of this study was to provide a comprehensive overview of the evolution of IFI prophylactic strategies over time and to assess IFI incidence rates in a cohort of patients with relapsed or refractory (R/R) lymphoma treated with CAR-T cell therapy. A single-center retrospective study was conducted on a cohort of patients with R/R B cell lymphoma treated with CD19 CAR-T cell therapy between April 2016 and March 2023. Group A (April 2016-August 2020) consisted of patients primarily treated with fluconazole, irrespective of their individual IFI risk profile. In Group B (September 2020-March 2023) antifungal prophylaxis was recommended only for high-risk patients. Overall, 330 patients were included. Antifungal prophylaxis was prescribed to 119/142 (84%) patients in Group A and 58/188 (31%) in Group B (P < .001). Anti-mold azoles were prescribed to 8 (5.6%) patients in Group A and 21 (11.2%) patients in Group B. In Group A, 42 (29%) patients were switched to another antifungal, 9 (21%) because of toxicity, with 6 cases of transaminitis and 3 cases of prolonged QTc. In Group B, 21 (11.2%) patients were switched to the antifungal drug, mainly from fluconazole or micafungin to a mold-active agent following revised guidelines. No difference was found in liver toxicity between the two groups at infusion, day 10, and day 30. No significant differences were observed between the groups. IFIs following CAR-T cell therapy were rare, with 1 case of cryptococcal meningoencephalitis in group A (.7%) and 1 case of invasive aspergillosis in Group B (.5%), both occurring in patients on micafungin prophylaxis. In this large single-center cohort of patients with R/R lymphoma treated with CAR-T cells, we show that individualized prophylaxis, alongside careful management of CAR-T cell-related toxicities such as CRS, was associated with a very low IFI rate, avoiding the risk of unnecessary toxicities, drug-drug interactions, and high costs.
Collapse
Affiliation(s)
- Giovanna Melica
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Infectious Diseases and Clinical Immunology, Henri Mondor Hospital, APHP, Creteil, France
| | - Alejandro Luna de Abia
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Adult Bone Marrow Transplantation, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Gunjan L Shah
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Sean Devlin
- Department of Medicine, Weill Cornell Medical College, New York, New York; Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Magdalena Corona
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joshua Fein
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Parastoo B Dahi
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Sergio A Giralt
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Richard J Lin
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - M Lia Palomba
- Department of Medicine, Weill Cornell Medical College, New York, New York; Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Allison Parascondola
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jae Park
- Department of Medicine, Weill Cornell Medical College, New York, New York; Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gilles Salles
- Department of Medicine, Weill Cornell Medical College, New York, New York; Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Amethyst Saldia
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael Scordo
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Roni Shouval
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Susan K Seo
- Department of Medicine, Weill Cornell Medical College, New York, New York; Department of Medicine, Infectious Disease Service, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
85
|
Tomarchio V, Crescenzi A, Tafuri M, Verri M, Di Cecca M, Rigacci L, Annibali O. The past, the present and the future of immune checkpoints inhibitors in multiple myeloma. Expert Rev Hematol 2025; 18:201-214. [PMID: 39987500 DOI: 10.1080/17474086.2025.2469720] [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: 10/29/2024] [Revised: 01/20/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Abstract
INTRODUCTION Myeloma genesis is a very complex mechanism in which the interaction between plasma cells and microenvironments with immune cells, cytokines and chemokines have a central role. In the last years, the improved knowledge of immune checkpoint models led to the development of new drugs (anti-PD1/PD-L1 axis or anti-TIGIT) that now have a crucial role in the treatment of many hematological malignancies. AREAS COVERED In this review, the current significant literature was discussed. In the past, initial trials combining immune checkpoint inhibitors (ICIs) with immunomodulatory drugs or proteasome inhibitors demonstrated suboptimal results in terms of efficacy and safety. On the other hand, recent trials based on the combination of ICIs with immunotherapies, such as CAR-T cells or bispecific antibodies, are a particularly promising area of investigation. EXPERT OPINION Our idea after the evaluation of scientific literature is that despite the past, ICIs may represent a promising therapeutic approach for myeloma, particularly when combined with CAR-T cells or bispecific antibodies. By targeting immune evasion mechanisms, ICIs may enhance the efficacy of these treatments and provide new hope for patients with resistant disease. Future research will be crucial to further elucidate their optimal use in myeloma and to develop personalized treatment strategies.
Collapse
Affiliation(s)
- Valeria Tomarchio
- Operative Research Unit of Hematology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Anna Crescenzi
- Operative Research Unit of Unit of Pathological Anatomy, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Mariantonietta Tafuri
- Operative Research Unit of Hematology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Martina Verri
- Operative Research Unit of Unit of Pathological Anatomy, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Monica Di Cecca
- Operative Research Unit of Hematology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Luigi Rigacci
- Operative Research Unit of Hematology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Ombretta Annibali
- Operative Research Unit of Hematology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| |
Collapse
|
86
|
Munir M, Sayed A, Addison D, Epperla N. Cardiovascular toxicities associated with novel cellular immune therapies. Blood Adv 2024; 8:6282-6296. [PMID: 39418640 PMCID: PMC11698921 DOI: 10.1182/bloodadvances.2024013849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/26/2024] [Accepted: 10/13/2024] [Indexed: 10/19/2024] Open
Abstract
ABSTRACT Over the past decade, T-cell-directed therapies, including chimeric antigen receptor T-cell (CAR-T) and bispecific T-cell engager (BTE) therapies, have reshaped the treatment of an expanding number of hematologic malignancies, whereas tumor-infiltrating lymphocytes, a recently approved cellular therapy, targets solid tumor malignancies. Emerging data suggest that these therapies may be associated with a high incidence of serious cardiovascular toxicities, including atrial fibrillation, heart failure, ventricular arrhythmias, and other cardiovascular toxicities. The development of these events is a major limitation to long-term survival after these treatments. This review examines the current state of evidence, including reported incidence rates, risk factors, mechanisms, and management strategies of cardiovascular toxicities after treatment with these novel therapies. We specifically focus on CAR-T and BTE therapies and their relation to arrhythmia, heart failure, myocarditis, bleeding, and other major cardiovascular events. Beyond the relationship between cytokine release syndrome and cardiotoxicity, we describe other potential mechanisms and highlight key unanswered questions and future directions of research.
Collapse
Affiliation(s)
- Malak Munir
- Department of Medicine, Ain Shams University Faculty of Medicine, Cairo, Egypt
| | - Ahmed Sayed
- Department of Medicine, Ain Shams University Faculty of Medicine, Cairo, Egypt
- Department of Cardiology, Houston Methodist DeBakey Heart & Vascular Center, Houston, TX
| | - Daniel Addison
- Division of Cancer Prevention and Control, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH
- Division of Epidemiology, College of Public Health, The Ohio State University, Columbus, OH
| | - Narendranath Epperla
- Division of Hematology and Hematologic Malignancies, Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| |
Collapse
|
87
|
Fleischer A, Kurth S, Duell J, Topp M, Strunz PP, Mersi J, Rasche L, Sanges C, Hudecek M, Einsele H, Maatouk I. Neuropsychiatric manifestations following chimeric antigen receptor T cell therapy for cancer: a systematic review of clinical outcomes and management strategies. J Immunother Cancer 2024; 12:e009174. [PMID: 39794934 PMCID: PMC11667355 DOI: 10.1136/jitc-2024-009174] [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: 03/20/2024] [Accepted: 11/15/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-T cell therapy has emerged as a transformative modality in the treatment of patients with cancer. However, it is increasingly evident that this therapeutic approach is not without its challenges. The unique nature of CAR-T cells as living drugs introduces a distinct set of side effects. As the application of CAR-T cell therapy expands to treat a broader range of diseases, it becomes increasingly important to devise effective strategies for handling the associated toxicities. Challenges in treating patients with CAR-T cells include addressing complications such as cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and cytopenias. This comprehensive review seeks to systematically identify, categorize and elucidate all previously described neurological and psychological side effects associated with CAR-T cell therapy, shedding light on the pertinent laboratory findings that underscore these phenomena. METHODS PubMed, Springer Link, and ScienceDirect were systematically searched for empirical studies on adult patients with cancer receiving CAR-T cell therapy for hemato-oncological malignancies. Quality assessment was conducted using Version 2 of the Cochrane risk-of-bias tool (RoB 2) for randomized trials and adherence to the STROBE (Strengthening the Reporting of Observational Studies in Epidemiology) checklist for observational studies. The synthesis of findings was conducted via a narrative approach, consolidating the diverse array of data into a coherent framework. RESULTS From an initial pool of 2,276 citations, 546 studies met the inclusion criteria, exhibiting a rich tapestry of heterogeneity in terms of study characteristics and patient samples. The incidence of neuropsychological symptoms varied notably across different CAR-T cell products and hematological malignancies. Among the most frequently reported neuropsychological symptoms were aphasia, attention deficits, impaired consciousness, and disorientation, alongside a constellation of other symptoms including confusion, cognitive impairment, memory loss, writing difficulties, fatigue, headache, agitation, tremor, seizures, and psychomotor retardation. Early intervention strategies, including corticosteroids and tocilizumab, have shown the potential to reduce the intensity of neuropsychological symptoms and prevent their progression to critical complications. CONCLUSION These insights underscore the imperative of extending neuropsychological assessments beyond the conventional Immune Effector Cell-Associated Encephalopathy score framework.
Collapse
Affiliation(s)
- Anna Fleischer
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Sophia Kurth
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Johannes Duell
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Max Topp
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | | | - Julia Mersi
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Leo Rasche
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Carmen Sanges
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Michael Hudecek
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Imad Maatouk
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| |
Collapse
|
88
|
Daryanani AE, Abbasi MA, Gomez Ardila MF, Tellez-Garcia E, Garzon-Dangond JM, Lin Y, Paludo J, Herrmann J, Ansell SM, Rosenthal AC, Villarraga HR. Baseline echocardiographic variables as predictors of hemodynamically significant cytokine release syndrome in adults treated with CD19 CAR T-cell therapy for hematological malignancies. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:91. [PMID: 39709512 DOI: 10.1186/s40959-024-00290-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 11/14/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND CD19 CAR T-cell therapy is a novel anti-cancer treatment that has produced remarkable responses in relapsed or refractory B-cell hematological malignancies. Cytokine Release Syndrome (CRS) is a dysregulated immune response that frequently occurs after CAR T-cell infusion. It can cause cardiac dysfunction and circulatory collapse negatively impacting outcomes and survival. To endure the insults of CRS, patients are typically screened for adequate cardiac reserve before treatment. The relationship between baseline cardiac function by echocardiography and the development of moderate to severe presentations of CRS is unclear. METHODS This study aimed to identify baseline echocardiographic variables that can predict the development of hemodynamically significant CRS (CRS ≥ 2), evaluate their behavior at follow-up, and investigate the incidence of cancer therapy-related cardiac dysfunction (CTRCD). An observational retrospective cohort study of patients treated with CD19 CAR T-cell therapy with a baseline echocardiogram was performed. Demographic, clinical and echocardiographic variables were abstracted from the electronic health record. Patients were grouped and compared by the occurrence of CRS < 2 and ≥ 2. Adjusted logistic regression analysis was used to evaluate the association between echocardiographic variables and the development of CRS ≥ 2. RESULTS 291 patients were included in the study. Median age was 60 (IQR: 51, 67 years), 73% were male, and 71% had diffuse large B-cell lymphoma. Logistic regression analysis did not reveal any significant baseline echocardiographic predictors of CRS ≥ 2, including left ventricular ejection fraction and global longitudinal strain. Systolic and diastolic echocardiographic variables remained within normal limits at follow-up overall and in both CRS groups. The incidence of CTRCD was 4.5% and occurred mostly in the setting of CRS ≥ 2. CONCLUSION No specific echocardiographic variables predicted the development of CRS ≥ 2, and therefore the mechanism leading to hemodynamic decompensation and producing worsening hypoxia and hypotension could be multifactorial and not directly cardiac mediated.
Collapse
Affiliation(s)
- Andres E Daryanani
- Department of Cardiovascular Medicine, Mayo Clinic, 200 1 St SW, Rochester, MN, 55905, USA
| | - Muhannad A Abbasi
- Department of Cardiovascular Medicine, Mayo Clinic, 200 1 St SW, Rochester, MN, 55905, USA
| | - Maria F Gomez Ardila
- Department of Cardiovascular Medicine, Mayo Clinic, 200 1 St SW, Rochester, MN, 55905, USA
| | - Eduardo Tellez-Garcia
- Department of Cardiovascular Medicine, Mayo Clinic, 200 1 St SW, Rochester, MN, 55905, USA
| | - Juan M Garzon-Dangond
- Department of Cardiovascular Medicine, Mayo Clinic, 200 1 St SW, Rochester, MN, 55905, USA
| | - Yi Lin
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jonas Paludo
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, 200 1 St SW, Rochester, MN, 55905, USA
| | - Stephen M Ansell
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Allison C Rosenthal
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Phoenix, ARZ, USA
| | - Hector R Villarraga
- Department of Cardiovascular Medicine, Mayo Clinic, 200 1 St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
89
|
Rankin AW, Duncan BB, Allen C, Silbert SK, Shah NN. Evolving strategies for addressing CAR T-cell toxicities. Cancer Metastasis Rev 2024; 44:17. [PMID: 39674824 PMCID: PMC11646216 DOI: 10.1007/s10555-024-10227-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 10/02/2024] [Indexed: 12/16/2024]
Abstract
The field of chimeric antigen receptor (CAR) T-cell therapy has grown from a fully experimental concept to now boasting a multitude of treatments including six FDA-approved products targeting various hematologic malignancies. Yet, along with their efficacy, these therapies come with side effects requiring timely and thoughtful interventions. In this review, we discuss the most common toxicities associated with CAR T-cells to date, highlighting risk factors, prognostication, implications for critical care management, patient experience optimization, and ongoing work in the field of toxicity mitigation. Understanding the current state of the field and standards of practice is critical in order to improve and manage potential toxicities of both current and novel CAR T-cell therapies as they are applied in the clinic.
Collapse
Affiliation(s)
- Alexander W Rankin
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Brynn B Duncan
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Cecily Allen
- Division of Hematology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Critical Care Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Sara K Silbert
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
90
|
Tabbara N, Dioverti-Prono MV, Jain T. Mitigating and managing infection risk in adults treated with CAR T-cell therapy. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2024; 2024:116-125. [PMID: 39644015 PMCID: PMC11706248 DOI: 10.1182/hematology.2024000535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
Chimeric antigen receptor T-cell therapy (CAR-T) has transformed the treatment paradigm of relapsed/refractory B-cell malignancies. Yet, this therapy is not without toxicities. While the early inflammation-mediated toxicities are now better understood, delayed hematopoietic recovery and infections result in morbidity and mortality risks that persist for months following CAR-T. The predisposition to infections is a consequence of immunosuppression from the underlying disease, prior therapies, lymphodepletion chemotherapy, delayed hematopoietic recovery, B-cell aplasia, and delayed T-cell immune reconstitution. These risks and epidemiology can vary over a post-CAR-T timeline of early (<30 days), prolonged (30-90 days), or late (>90 days) follow-up. Antibacterial, antiviral, and antifungal prophylaxis; growth factors and stem cell boost to expedite count recovery; immunoglobulin replacement therapy; and possibly revaccination programs are important prevention strategies to consider for infection mitigation. Assessment of risk factors, evaluation, and treatment for pathogen(s) prevalent in a particular time frame post-CAR-T are important clinical considerations in patients presenting with clinical features suggestive of infectious pathology. As more data emerge on the topic, personalized risk assessments to inform the type and duration of prophylaxis use and planning interventions will continue to emerge. Herein, we review our current approach toward infection mitigation while recognizing that this continues to evolve and that there are differences among practices stemming from data availability limitations.
Collapse
Affiliation(s)
- Nadeem Tabbara
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - M Veronica Dioverti-Prono
- Division of Infectious Disease, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD
| | - Tania Jain
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
91
|
Wang XS, Srour SA. Patient-reported outcomes after CAR T-cell therapy in patients with hematological malignancies. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2024; 2024:102-108. [PMID: 39644035 DOI: 10.1182/hematology.2024000536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
The remarkable improvement in survival among individuals with hematological malignancies receiving chimeric antigen receptor (CAR) T-cell therapy has highlighted the growing unmet need to incorporate patient-centered assessments in management guidelines for these patients. That CAR T-cell therapy is associated with unique toxicities and relatively high symptom burden in the first few weeks after cell infusion is well known. Magnifying the patient's voice by using patient-reported outcomes (PROs) might support personalized intervention in the acute-care setting, optimize the use of medical resources, improve satisfaction with therapy, and enhance survival benefit. However, various factors impede PRO use in routine patient care: (1) the feasibility of PRO assessment during the acute phase of treatment, especially in patients experiencing neurological toxicities, is not well established; (2) although PROs are widely used in drug- development trials, the assessment tools used in clinical trials primarily inform quality-of-life or safety comparisons among study arms and are rarely the proper tools for assessing and capturing clinically meaningful adverse events that should be monitored in routine patient care; (3) PRO data that could guide how best to monitor and capture the delayed effects of CAR T-cell therapy in long-term survivors are limited. There is a pressing need to overcome these barriers to integrating evidence-based PROs into standard-of-care guidelines for patients receiving CAR T-cell therapy. In this review, we present the current state of PRO utilization in CAR T-cell therapy. We also discuss practical approaches and future directions for successful implementation of PROs in the care of patients receiving CAR T-cell therapy.
Collapse
Affiliation(s)
| | - Samer A Srour
- University of Texas M.D. Anderson Cancer Center, Houston, TX
| |
Collapse
|
92
|
Takahashi H, Kojima D, Watanabe M. Therapeutic potential of trained immunity for malignant disease. Immunol Med 2024:1-12. [PMID: 39639550 DOI: 10.1080/25785826.2024.2438426] [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: 06/06/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
Trained immunity (TI) is functional memory displayed by innate immune cells (IICs). TI facilitates rapid, non-specific responses to pathogens upon secondary challenge. It is driven by immunological signaling and metabolic rewriting via epigenetic alteration, triggered by recognition of certain stimuli. Recently, immune checkpoint inhibitors have come into common use in clinical oncology settings, and genetically engineered cytotoxic T cells comprise a potent cancer treatment strategy. However, the contributions of TI in the tumor microenvironment (TME) are only beginning to be uncovered. Accumulating evidence that various microorganisms and vaccines convey tumoricidal ability suggest that TI may become a useful anti-cancer tool. The expected roles of TI in tumor therapy are the 1) promotion of proinflammatory cytokine section, 2) enhancement of phagocytosis, 3) quick expansion and recruitment of cancer-specific cytotoxic T cells to the TME through neoantigen presentation, 4) reversal of immunosuppression in the TME, and 5) removal of pathogens associated with carcinogenesis or tumor development. Medium- to long-term TI durability may reduce the risk of tumor development. Recent findings on TI usher in new aspirations for cancer treatment.
Collapse
Affiliation(s)
- Hiroyuki Takahashi
- Department of Surgery, Fukuoka University Chikushi Hospital, Chikushino, Fukuoka, Japan
| | - Daibo Kojima
- Department of Surgery, Fukuoka University Chikushi Hospital, Chikushino, Fukuoka, Japan
| | - Masato Watanabe
- Department of Surgery, Fukuoka University Chikushi Hospital, Chikushino, Fukuoka, Japan
| |
Collapse
|
93
|
Yao P, Liu YG, Huang G, Hao L, Wang R. The development and application of chimeric antigen receptor natural killer (CAR-NK) cells for cancer therapy: current state, challenges and emerging therapeutic advances. Exp Hematol Oncol 2024; 13:118. [PMID: 39633491 PMCID: PMC11616395 DOI: 10.1186/s40164-024-00583-7] [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: 05/16/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Abstract
Immunotherapy has transformed the landscape of cancer treatment, with chimeric antigen receptor (CAR)-engineered T (CAR-T) cell therapy emerging as a front runner in addressing some hematological malignancies. Despite its considerable efficacy, the occurrence of severe adverse effects associated with CAR-T cell therapy has limited their scope and prompted the exploration of alternative therapeutic strategies. Natural killer (NK) cells, characterized by both their innate cytotoxicity and ability to lyse target cells without the constraint of peptide specificity conferred by a major histocompatibility complex (MHC), have similarly garnered attention as a viable immunotherapy. As such, another therapeutic approach has recently emerged that seeks to combine the continued success of CAR-T cell therapy with the flexibility of NK cells. Clinical trials involving CAR-engineered NK (CAR-NK) cell therapy have exhibited promising efficacy with fewer deleterious side effects. This review aims to provide a concise overview of the cellular and molecular basis of NK cell biology, facilitating a better understanding of advancements in CAR design and manufacturing. The focus is on current approaches and strategies employed in CAR-NK cell development, exploring at both preclinical and clinical settings. We will reflect upon the achievements, advantages, and challenges intrinsic to CAR-NK cell therapy. Anticipating the maturation of CAR-NK cell therapy technology, we foresee its encouraging prospects for a broader range of cancer patients and other conditions. It is our belief that this CAR-NK progress will bring us closer to making significant strides in the treatment of refractory and recurrent cancers, as well as other immune-mediated disorders.
Collapse
Affiliation(s)
- Pin Yao
- Department of Health Management, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Ya-Guang Liu
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Gang Huang
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Liangchun Hao
- Department of Pediatrics, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Runan Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
94
|
Damiani D, Tiribelli M. Advancing Chimeric Antigen Receptor T-Cell Therapy for Acute Myeloid Leukemia: Current Limitations and Emerging Strategies. Pharmaceuticals (Basel) 2024; 17:1629. [PMID: 39770471 PMCID: PMC11728840 DOI: 10.3390/ph17121629] [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: 07/06/2024] [Revised: 11/27/2024] [Accepted: 11/30/2024] [Indexed: 01/16/2025] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy represents one of the most impressive advances in anticancer therapy of the last decade. While CAR T-cells are gaining ground in various B cell malignancies, their use in acute myeloid leukemia (AML) remains limited, and no CAR-T product has yet received approval for AML. The main limitation of CAR-T therapy in AML is the lack of specific antigens that are expressed in leukemic cells but not in their healthy counterparts, such as hematopoietic stem cells (HSCs), as their targeting would result in an on-target/off-tumor toxicity. Moreover, the heterogeneity of AML and the tendency of blasts to modify surface antigens' expression in the course of the disease make identification of suitable targets even more challenging. Lastly, AML's immunosuppressive microenvironment dampens CAR-T therapeutic activities. In this review, we focus on the actual pitfalls of CAR T-cell therapy in AML, and we discuss promising approaches to overcome them.
Collapse
Affiliation(s)
- Daniela Damiani
- Division of Hematology and Stem Cell Transplantation, University Hospital, 33100 Udine, Italy;
- Department of Medicine (DMED), University of Udine, 33100 Udine, Italy
| | - Mario Tiribelli
- Division of Hematology and Stem Cell Transplantation, University Hospital, 33100 Udine, Italy;
- Department of Medicine (DMED), University of Udine, 33100 Udine, Italy
| |
Collapse
|
95
|
Reddy ST, Hosoya H, Mikkilineni L. CAR T-cell therapy to treat multiple myeloma: current state and future directions. Cancer Metastasis Rev 2024; 44:14. [PMID: 39625587 DOI: 10.1007/s10555-024-10219-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 11/07/2024] [Indexed: 12/17/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy represents a transformative advancement in treating relapsed or refractory multiple myeloma (MM) in both early- and late-line settings. MM, a plasma cell malignancy, traditionally requires ongoing complex drug regimens, posing significant burdens on patients. In contrast, CAR T-cell therapy offers a one-time treatment option without the need for continuous maintenance therapy. CAR T-cell therapy leverages engineered T-cells to target specific antigens on tumor cells, leading to their elimination. Current approved therapies target B-cell maturation antigen (BCMA); new targets are under investigation, such as G-protein-coupled receptor class C group 5 member D (GPRC5D). Despite its efficacy, CAR T-cell therapy is associated with serious toxicities such as cytokine release syndrome (CRS) and immune-effector cell-associated neurotoxicity syndrome (ICANS), necessitating careful management. The review will provide an overview of the design and manufacturing of CAR T-cells and current FDA indications, as well as challenges and future directions of CAR-T therapy for MM treatment.
Collapse
Affiliation(s)
- Siddhartha Thammineni Reddy
- Division of Bone and Marrow Transplantation & Cellular Therapies, Stanford University, 870 Welch Road, Palo Alto, CA, 94304, USA
- Hackensack University Medical Center, NJ, USA
| | - Hitomi Hosoya
- Division of Bone and Marrow Transplantation & Cellular Therapies, Stanford University, 870 Welch Road, Palo Alto, CA, 94304, USA
| | - Lekha Mikkilineni
- Division of Bone and Marrow Transplantation & Cellular Therapies, Stanford University, 870 Welch Road, Palo Alto, CA, 94304, USA.
| |
Collapse
|
96
|
Cordas Dos Santos DM, Toenges R, Bertamini L, Alberge JB, Ghobrial IM. New horizons in our understanding of precursor multiple myeloma and early interception. Nat Rev Cancer 2024; 24:867-886. [PMID: 39414947 DOI: 10.1038/s41568-024-00755-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/06/2024] [Indexed: 10/18/2024]
Abstract
Multiple myeloma is an incurable plasma cell malignancy that evolves over decades through the selection and malignant transformation of monoclonal plasma cells. The evolution from precursor states to symptomatic disease is characterized by an increasing complexity of genomic alterations within the plasma cells and a remodelling of the microenvironment towards an immunosuppressive state. Notably, in patients with advanced disease, similar mechanisms of tumour escape and immune dysfunction mediate resistance to modern T cell-based therapies, such as T cell-engaging bispecific antibodies and chimeric antigen receptor (CAR)-T cells. Thus, an increasing number of clinical trials are assessing the efficiency and safety of these therapies in individuals with newly diagnosed multiple myeloma and high-risk smoldering multiple myeloma. In this Review, we summarize the current knowledge about tumour intrinsic and extrinsic processes underlying progression from precursor states to symptomatic myeloma and discuss the rationale for early interception including the use of T cell-redirecting therapies.
Collapse
Affiliation(s)
- David M Cordas Dos Santos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Rosa Toenges
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Luca Bertamini
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Hematology, Erasmus MC Cancer Institute Rotterdam, Rotterdam, The Netherlands
| | - Jean-Baptiste Alberge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Irene M Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
97
|
Tatake IJ, Arnason JE. CARs for lymphoma. Best Pract Res Clin Haematol 2024; 37:101601. [PMID: 40074511 DOI: 10.1016/j.beha.2025.101601] [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: 10/03/2024] [Accepted: 02/17/2025] [Indexed: 03/14/2025]
Abstract
Chimeric Antigen Receptor (CAR)-T cell therapy has revolutionized treatment options for B-cell Non-Hodgkin Lymphoma (NHL). CD19-targeting CAR-T cell therapy is approved for treatment in Diffuse Large B Cell Lymphoma, Follicular Lymphoma, Mantle Cell Lymphoma, and Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma. CAR-T cells demonstrate robust and durable responses even in heavily pretreated patients. Clinicians should monitor for Cytokine Release Syndrome (CRS) and Immune Effector Cell Neurotoxicity Syndrome (ICANS), as well as cytopenias, infection, and secondary malignancies. Ongoing questions remain in improving manufacturing efficacy, sequencing CAR-T cells amongst other therapies including bi-specific antibodies (BiTEs), and predicting optimal responders. In addition, novel CARs are being developed with alternative targets or that secrete activating cytokines (i.e. "armored CARs"). CAR-T cells represent an effective lymphoma therapy and should be considered for eligible patients.
Collapse
MESH Headings
- Humans
- Receptors, Chimeric Antigen/therapeutic use
- Receptors, Chimeric Antigen/immunology
- Immunotherapy, Adoptive
- Cytokine Release Syndrome/etiology
- Cytokine Release Syndrome/therapy
- Cytokine Release Syndrome/immunology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/pathology
- Antigens, CD19/immunology
- Antigens, CD19/therapeutic use
- Neurotoxicity Syndromes/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Lymphoma, B-Cell/therapy
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/pathology
- Lymphoma/therapy
- Lymphoma/immunology
- Lymphoma, Mantle-Cell/therapy
- Lymphoma, Mantle-Cell/immunology
- Lymphoma, Mantle-Cell/pathology
Collapse
Affiliation(s)
- Ishan J Tatake
- Department of Medicine, Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Jon E Arnason
- Department of Medicine, Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
98
|
Hoogland AI, Barata A, Li X, Irizarry-Arroyo N, Jain MD, Welniak T, Rodriguez Y, Oswald LB, Gudenkauf LM, Chavez JC, Khimani F, Lazaryan A, Liu HD, Nishihori T, Pinilla-Ibarz J, Shah BD, Crowder SL, Parker NH, Carson TL, Vinci CE, Pidala JA, Logue J, Locke FL, Jim HSL. Prospective Assessment of Quality of Life and Patient-Reported Toxicities Over the First Year After Chimeric Antigen Receptor T-Cell Therapy. Transplant Cell Ther 2024; 30:1219.e1-1219.e11. [PMID: 39306278 PMCID: PMC11635818 DOI: 10.1016/j.jtct.2024.09.013] [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: 06/03/2024] [Revised: 09/05/2024] [Accepted: 09/14/2024] [Indexed: 09/26/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has transformed survival outcomes in patients with relapsed and refractory large B-cell lymphoma (LBCL), but it is associated with a variety of side effects. This study examined changes in patient-reported quality of life (QoL) and toxicities, as well as risk factors for worse QoL and toxicities, in the first year after treatment. Patients with LBCL completed questionnaires assessing QoL and toxicity severity before infusion, and 90, 180, and 360 days after infusion. Mixed models were used to examine changes in QoL and toxicities over time, and clinical moderators of change in QoL and toxicities. Patients reported improvements in physical functioning and fatigue in the year after treatment (P values <.01), but there were no changes in pain, anxiety, or depression over time. Patients with active disease at day 90 reported more physical dysfunction at all postinfusion timepoints (Ps ≤ .01) compared to patients who responded to treatment. Similarly, patients with active disease at day 90 reported worsening depression over time, such that at day 360, depressive symptoms were worse for patients with active disease than patients without active disease (P = .02). Patients treated with 4+ lines of prior therapy reported worsening pain and anxiety over time, such that at day 360, both pain and anxiety were significantly worse for patients previously treated with 4 of more lines of therapy than patients treated with fewer lines of therapy (Ps ≤ .01). Regarding toxicities, patients reported decreasing overall toxicity burden up to day 180, with subsequent worsening at day 360 (P = .02). Most patients reported at least one or two grade 2 toxicities at each timepoint. Patients demonstrated unchanging or improved QoL after treatment with CAR T-cell therapy, but active disease and greater prior lines of therapy were associated with worse QoL outcomes over time. Toxicity severity also improved during the first 6 months post-treatment, but worsened thereafter, particularly among patients with active disease after treatment.
Collapse
Affiliation(s)
- Aasha I Hoogland
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida.
| | - Anna Barata
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Xiaoyin Li
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida
| | | | - Michael D Jain
- Department of Blood and Marrow Transplantation and Cellular Therapy, Moffitt Cancer Center, Tampa, Florida
| | - Taylor Welniak
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida
| | - Yvelise Rodriguez
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida
| | - Laura B Oswald
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida
| | - Lisa M Gudenkauf
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida
| | - Julio C Chavez
- Department of Blood and Marrow Transplantation and Cellular Therapy, Moffitt Cancer Center, Tampa, Florida
| | - Farhad Khimani
- Department of Blood and Marrow Transplantation and Cellular Therapy, Moffitt Cancer Center, Tampa, Florida
| | - Aleksandr Lazaryan
- Department of Blood and Marrow Transplantation and Cellular Therapy, Moffitt Cancer Center, Tampa, Florida
| | - Hien D Liu
- Department of Blood and Marrow Transplantation and Cellular Therapy, Moffitt Cancer Center, Tampa, Florida
| | - Taiga Nishihori
- Department of Blood and Marrow Transplantation and Cellular Therapy, Moffitt Cancer Center, Tampa, Florida
| | - Javier Pinilla-Ibarz
- Department of Blood and Marrow Transplantation and Cellular Therapy, Moffitt Cancer Center, Tampa, Florida
| | - Bijal D Shah
- Department of Blood and Marrow Transplantation and Cellular Therapy, Moffitt Cancer Center, Tampa, Florida
| | - Sylvia L Crowder
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida
| | - Nathan H Parker
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida
| | - Tiffany L Carson
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida
| | - Christine E Vinci
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida
| | - Joseph A Pidala
- Department of Blood and Marrow Transplantation and Cellular Therapy, Moffitt Cancer Center, Tampa, Florida
| | - Jennifer Logue
- Moffitt Malignant Hematology & Cellular Therapy, Memorial Healthcare System, Pembroke Pines, Florida
| | - Frederick L Locke
- Department of Blood and Marrow Transplantation and Cellular Therapy, Moffitt Cancer Center, Tampa, Florida
| | - Heather S L Jim
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
99
|
Pan D, Richter J. Management of Toxicities Associated with BCMA, GPRC5D, and FcRH5-Targeting Bispecific Antibodies in Multiple Myeloma. Curr Hematol Malig Rep 2024; 19:237-245. [PMID: 39145912 DOI: 10.1007/s11899-024-00740-z] [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] [Accepted: 07/31/2024] [Indexed: 08/16/2024]
Abstract
PURPOSE OF REVIEW The introduction of bispecific antibodies is one of the most significant recent advances in the treatment of relapsed/refractory multiple myeloma. This review will summarize the management of the toxicities associated with newly approved T cell-engaging bispecific antibodies and those which may be approved in the near future. RECENT FINDINGS Numerous trials have shown that bispecific antibodies can be both effective and tolerable when adverse events are properly managed. Cytokine release syndrome and increased infections are observed across all bispecific antibodies. Additional adverse events are target-specific, such as the more severe hypogammaglobulinemia and infections of BCMA bispecific antibodies and the dysgeusia, nail dystrophy, and skin changes of GPRC5D bispecific antibodies. Bispecific antibodies will surely become a mainstay of multiple myeloma therapy given their efficacy and accessibility. Their unique toxicities must be carefully considered and managed to ensure they are utilized safely.
Collapse
Affiliation(s)
- Darren Pan
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Joshua Richter
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA.
| |
Collapse
|
100
|
Yang M, Lin W, Huang J, Mannucci A, Luo H. Novel immunotherapeutic approaches in gastric cancer. PRECISION CLINICAL MEDICINE 2024; 7:pbae020. [PMID: 39397869 PMCID: PMC11467695 DOI: 10.1093/pcmedi/pbae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/08/2024] [Accepted: 09/08/2024] [Indexed: 10/15/2024] Open
Abstract
Gastric cancer is a malignant tumor that ranks third in cancer-related deaths worldwide. Early-stage gastric cancer can often be effectively managed through surgical resection. However, the majority of cases are diagnosed in advanced stages, where outcomes with conventional radiotherapy and chemotherapy remain unsatisfactory. Immunotherapy offers a novel approach to treating molecularly heterogeneous gastric cancer by modifying the immunosuppressive tumor microenvironment. Immune checkpoint inhibitors and adoptive cell therapy are regarded as promising modalities in cancer immunotherapy. Food and Drug Administration-approved programmed death-receptor inhibitors, such as pembrolizumab, in combination with chemotherapy, have significantly extended overall survival in gastric cancer patients and is recommended as a first-line treatment. Despite challenges in solid tumor applications, adoptive cell therapy has demonstrated efficacy against various targets in gastric cancer treatment. Among these approaches, chimeric antigen receptor-T cell therapy research is the most widely explored and chimeric antigen receptor-T cell therapy targeting claudin18.2 has shown acceptable safety and robust anti-tumor capabilities. However, these advancements primarily remain in preclinical stages and further investigation should be made to promote their clinical application. This review summarizes the latest research on immune checkpoint inhibitors and adoptive cell therapy and their limitations, as well as the role of nanoparticles in enhancing immunotherapy.
Collapse
Affiliation(s)
- Meng Yang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Wuhao Lin
- Department of Molecular Diagnostics, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jiaqian Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Alessandro Mannucci
- Gastroenterology and Gastrointestinal Emndoscopy Unit, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan 20132, Italy
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope; Monrovia, CA 91016, USA
| | - Huiyan Luo
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| |
Collapse
|