1
|
Cai W, Lu Y, He H, Li J, Liu S, Geng H, Yang Q, Zeng L, Wu D, Li C. Efficacy of emapalumab in the management of anti‑CD19 chimeric antigen receptor T‑cell therapy‑associated cytokine release syndrome: A report of two cases. Oncol Lett 2025; 29:71. [PMID: 39628826 PMCID: PMC11612719 DOI: 10.3892/ol.2024.14817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/09/2024] [Accepted: 10/01/2024] [Indexed: 12/06/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is an effective treatment for diffuse large B-cell lymphoma (DLBCL). However, it may activate the systemic immune system of the patient, resulting in cytokine release syndrome (CRS). Emapalumab is a human monoclonal antibody targeting interferon-γ, inhibiting its interaction with cell surface receptors and the subsequent activation of inflammatory pathways. The present report describes the cases of 2 patients with relapsed DLBCL treated with CAR T-cell therapy, in which the severe CRS associated with CAR T-cell therapy was attenuated without compromising antitumor efficacy after receiving emapalumab. Further prospective clinical trials are warranted to determine the role of emapalumab in CAR T-cell therapy.
Collapse
Affiliation(s)
- Wenzhi Cai
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Institute of Blood and Marrow Transplantation, Suzhou University Medical College, Suzhou, Jiangsu 215006, P.R. China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yutong Lu
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Institute of Blood and Marrow Transplantation, Suzhou University Medical College, Suzhou, Jiangsu 215006, P.R. China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Haiju He
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Institute of Blood and Marrow Transplantation, Suzhou University Medical College, Suzhou, Jiangsu 215006, P.R. China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jiaqi Li
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Institute of Blood and Marrow Transplantation, Suzhou University Medical College, Suzhou, Jiangsu 215006, P.R. China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Shuangzhu Liu
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Institute of Blood and Marrow Transplantation, Suzhou University Medical College, Suzhou, Jiangsu 215006, P.R. China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Hongzhi Geng
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Qin Yang
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Liangyu Zeng
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Depei Wu
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Institute of Blood and Marrow Transplantation, Suzhou University Medical College, Suzhou, Jiangsu 215006, P.R. China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Caixia Li
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
2
|
Chen S, Zhang C, Luo J, Lin Z, Chang T, Dong L, Chen D, Tang ZH. Macrophage activation syndrome in Sepsis: from pathogenesis to clinical management. Inflamm Res 2024; 73:2179-2197. [PMID: 39404874 DOI: 10.1007/s00011-024-01957-7] [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] [Academic Contribution Register] [Received: 03/20/2024] [Revised: 08/01/2024] [Accepted: 10/01/2024] [Indexed: 12/11/2024] Open
Abstract
BACKGROUND Sepsis represents a significant global health and hygiene challenge. Excessive activation of macrophages in sepsis can result in certain patients displaying characteristics akin to those observed in Macrophage Activation Syndrome (MAS). MAS represents a grave immune system disorder characterized by persistent and severe inflammation within the body. In the context of sepsis, MAS presents atypically, leading some researchers to refer to it as Macrophage Activation-Like Syndrome (MALS). However, there are currently no effective treatment measures for this situation. The purpose of this article is to explore potential treatment methods for sepsis-associated MALS. OBJECTIVE The objective of this review is to synthesize the specific pathophysiological mechanisms and treatment strategies of MAS to investigate potential therapeutic approaches for sepsis-associated MALS. METHOD We searched major databases (including PubMed, Web of Science, and Google Scholar etc.) for literature encompassing macrophage activation syndrome and sepsis up to Mar 2024 and combined with studies found in the reference lists of the included studies. CONCLUSION We have synthesized the underlying pathophysiological mechanism of MALS in sepsis, and then summarized the diagnostic criteria and the effects of various treatment modalities utilized in patients with MAS or MALS. In both scenarios, heterogeneous treatment responses resulting from identical treatment approaches were observed. The determination of whether the patient is genuinely experiencing MALS significantly impacts the ultimate outcomes of therapeutic efficacy. In order to tackle this concern, additional clinical trials and research endeavors are imperative.
Collapse
Affiliation(s)
- Shunyao Chen
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Cong Zhang
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jialiu Luo
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhiqiang Lin
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Teding Chang
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Liming Dong
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Deng Chen
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Zhao-Hui Tang
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
3
|
Yang Y, Peng H, Wang J, Li F. New insights into CAR T-cell hematological toxicities: manifestations, mechanisms, and effective management strategies. Exp Hematol Oncol 2024; 13:110. [PMID: 39521987 PMCID: PMC11549815 DOI: 10.1186/s40164-024-00573-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/19/2024] [Accepted: 10/12/2024] [Indexed: 11/16/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy represents a highly efficacious treatment modality demonstrated to enhance outcomes in patients afflicted with malignancies, particularly those enduring relapsed or refractory hematological malignancies. However, the escalating adoption of CAR T-cell therapy has unveiled several life-threatening toxicities, notably cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), infections, and hematological toxicities (HTs), thereby hindering the broad implementation of CAR T-cell therapy. HTs encompass a spectrum of adverse effects, including cytopenias, hemophagocytic lymphohistiocytosis (HLH), coagulopathies, and B-cell aplasia. While our comprehension of the underlying mechanisms governing CRS and ICANS is advancing, the intricate pathophysiology of HTs remains inadequately elucidated. Such knowledge gaps may precipitate suboptimal therapeutic decisions, potentially culminating in substantial medical resource depletion and detriment to patients' quality of life. In this comprehensive review, based on recent updated findings, we delineate various mechanisms contributing to HTs subsequent to CAR T-cell therapy, explicate manifestations of HTs, and proffer strategic interventions to mitigate this relevant clinical challenge.
Collapse
Affiliation(s)
- Yuanyuan Yang
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hongwei Peng
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, National Clinical Research Center for Blood Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Fei Li
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
- Jiangxi Clinical Research Center for Hematologic Disease, Nanchang, Jiangxi, China.
- Institute of Lymphoma and Myeloma, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
4
|
Zhang SH, Peng LL, Chen YF, Xu Y, Moradi V. Focusing on exosomes to overcome the existing bottlenecks of CAR-T cell therapy. Inflamm Regen 2024; 44:45. [PMID: 39490997 PMCID: PMC11533312 DOI: 10.1186/s41232-024-00358-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/15/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Since chimeric antigen receptor T (CAR-T) cells were introduced three decades ago, the treatment using these cells has led to outstanding outcomes, and at the moment, CAR-T cell therapy is a well-established mainstay for treating CD19 + malignancies and multiple myeloma. Despite the astonishing results of CAR-T cell therapy in B-cell-derived malignancies, several bottlenecks must be overcome to promote its safety and efficacy and broaden its applicability. These bottlenecks include cumbersome production process, safety concerns of viral vectors, poor efficacy in treating solid tumors, life-threatening side effects, and dysfunctionality of infused CAR-T cells over time. Exosomes are nano-sized vesicles that are secreted by all living cells and play an essential role in cellular crosstalk by bridging between cells. In this review, we discuss how the existing bottlenecks of CAR-T cell therapy can be overcome by focusing on exosomes. First, we delve into the effect of tumor-derived exosomes on the CAR-T cell function and discuss how inhibiting their secretion can enhance the efficacy of CAR-T cell therapy. Afterward, the application of exosomes to the manufacturing of CAR-T cells in a non-viral approach is discussed. We also review the latest advancements in ex vivo activation and cultivation of CAR-T cells using exosomes, as well as the potential of engineered exosomes to in vivo induction or boost the in vivo proliferation of CAR-T cells. Finally, we discuss how CAR-engineered exosomes can be used as a versatile tool for the direct killing of tumor cells or delivering intended therapeutic payloads in a targeted manner.
Collapse
Affiliation(s)
- Si-Heng Zhang
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, 310000, China
| | - Ling-Long Peng
- Wuhu Hospital, East China Normal University (The Second People's Hospital of Wuhu), Wuhu, 241000, China
| | - Yi-Fei Chen
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China
| | - Yan Xu
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, 310000, China.
| | - Vahid Moradi
- Hematology and Bood Transfusion Science Department, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Scala JJ, Eckrich MJ, Lipak K, Yates B, Yuan C, Wang HW, Dahiya S, Henter JI, Huo JS, Frank MJ, Shah NN. Treatment strategies for progressive immune effector cell-associated hemophagocytic lymphohistiocytosis-like syndrome: case series. Haematologica 2024; 109:3439-3445. [PMID: 38813719 PMCID: PMC11443363 DOI: 10.3324/haematol.2023.284784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/12/2024] [Accepted: 05/17/2024] [Indexed: 05/31/2024] Open
Affiliation(s)
- Jack J Scala
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford Cancer Institute, Stanford University, School of Medicine, Stanford, CA
| | - Michael J Eckrich
- Pediatric Stem Cell Transplant and Cellular Therapy, Atrium Health Levine Children's Hospital, Charlotte, NC
| | - Keagan Lipak
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Bonnie Yates
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Constance Yuan
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Hao-Wei Wang
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Saurabh Dahiya
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford Cancer Institute, Stanford University, School of Medicine, Stanford, CA
| | - Jan-Inge Henter
- Division of Pediatric Oncology and Surgery, Department of Women's and Children's Health, Karolinska Institute, and Department of Paediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Jeffrey S Huo
- Pediatric Stem Cell Transplant and Cellular Therapy, Atrium Health Levine Children's Hospital, Charlotte, NC
| | - Matthew J Frank
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford Cancer Institute, Stanford University, School of Medicine, Stanford, CA.
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
6
|
Ong SY, Baird JH. A Primer on Chimeric Antigen Receptor T-cell Therapy-related Toxicities for the Intensivist. J Intensive Care Med 2024; 39:929-938. [PMID: 37899577 DOI: 10.1177/08850666231205264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/31/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is an innovative treatment approach that has shown remarkable efficacy against several hematologic malignancies. However, its use can be associated with unique and sometimes severe toxicities that require admission to intensive care unit in 30% of patients, and intensivists should be aware of immune-mediated toxicities of CAR T-cell therapy and management of adverse events. We will review available literature on current diagnostic criteria and therapeutic strategies for mitigating these most common toxicities associated with CAR T-cell therapy including cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) in the post-infusion period. The authors will also review other toxicities associated with CAR T-cell therapy including cytopenias, acquired immunocompromised states, and infections, and discuss the available literature on best supportive care and prophylaxis recommendations. Critical care medicine specialists play a crucial role in the management of patients undergoing CAR T-cell therapies. With the expanding use of these products in increasing numbers of treating centers, intensivists' roles as part of the multidisciplinary team caring for these patients will have an outsized impact on the continued success of these promising therapies.
Collapse
Affiliation(s)
- Shin Yeu Ong
- Division of Lymphoma, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - John H Baird
- Division of Lymphoma, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
7
|
Johnson WT, Epstein-Peterson ZD, Ganesan N, Pak T, Chang T, Dao P, Moskowitz AJ, Stuver RN, Ghione P, Galasso N, Khan N, Palomba ML, Caron PC, Kumar A, Tamari R, Lue JK, Noy A, Falchi L, Intlekofer AM, Gyurkocza B, Perales MA, Scordo M, Herskovits AZ, Salles G, Vardhana SA, Horwitz SM. Emapalumab as salvage therapy for adults with malignancy-associated hemophagocytic lymphohistiocytosis. Haematologica 2024; 109:2998-3003. [PMID: 38752279 PMCID: PMC11367207 DOI: 10.3324/haematol.2023.284179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/25/2023] [Accepted: 05/07/2024] [Indexed: 09/03/2024] Open
Abstract
Not available.
Collapse
Affiliation(s)
- William T Johnson
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY.
| | - Zachary D Epstein-Peterson
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY
| | - Nivetha Ganesan
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Timothy Pak
- Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital-Weill Cornell Medical College, New York, NY
| | - Tiffany Chang
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Phuong Dao
- Department of Pharmacy, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alison J Moskowitz
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY
| | - Robert N Stuver
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY
| | - Paola Ghione
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY
| | - Natasha Galasso
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Niloufer Khan
- Department of Hematology and Hematopoietic Cell Transplant, Lymphoma Division, City of Hope Comprehensive Cancer, Duarte, CA
| | - M Lia Palomba
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY
| | - Philip C Caron
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY
| | - Anita Kumar
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY
| | - Roni Tamari
- Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA; Department of Medicine, Adult Bone Marrow Transplant and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jennifer K Lue
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY
| | - Ariela Noy
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY
| | - Lorenzo Falchi
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY
| | - Andrew M Intlekofer
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Boglarka Gyurkocza
- Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA; Department of Medicine, Adult Bone Marrow Transplant and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Miguel-Angel Perales
- Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA; Department of Medicine, Adult Bone Marrow Transplant and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael Scordo
- Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA; Department of Medicine, Adult Bone Marrow Transplant and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - A Zara Herskovits
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gilles Salles
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY
| | - Santosha A Vardhana
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Steven M Horwitz
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY
| |
Collapse
|
8
|
Li J, Chen H, Xu C, Hu M, Li J, Chang W. Systemic toxicity of CAR-T therapy and potential monitoring indicators for toxicity prevention. Front Immunol 2024; 15:1422591. [PMID: 39253080 PMCID: PMC11381299 DOI: 10.3389/fimmu.2024.1422591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/24/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024] Open
Abstract
Malignant tumors of the hematologic system have a high degree of malignancy and high mortality rates. Chimeric antigen receptor T cell (CAR-T) therapy has become an important option for patients with relapsed/refractory tumors, showing astonishing therapeutic effects and thus, it has brought new hope to the treatment of malignant tumors of the hematologic system. Despite the significant therapeutic effects of CAR-T, its toxic reactions, such as Cytokine Release Syndrome (CRS) and Immune Effector Cell-Associated Neurotoxicity Syndrome (ICANS), cannot be ignored since they can cause damage to multiple systems, including the cardiovascular system. We summarize biomarkers related to prediction, diagnosis, therapeutic efficacy, and prognosis, further exploring potential monitoring indicators for toxicity prevention. This review aims to summarize the effects of CAR-T therapy on the cardiovascular, hematologic, and nervous systems, as well as potential biomarkers, and to explore potential monitoring indicators for preventing toxicity, thereby providing references for clinical regulation and assessment of therapeutic effects.
Collapse
Affiliation(s)
- Jingxian Li
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Huiguang Chen
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Chaoping Xu
- Department of Hematology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Mengci Hu
- Department of Hematology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Jiangping Li
- Department of Blood Transfusion, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Wei Chang
- Department of Hematology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Lin Y, Qiu L, Usmani S, Joo CW, Costa L, Derman B, Du J, Einsele H, Fernandez de Larrea C, Hajek R, Ho PJ, Kastritis E, Martinez-Lopez J, Mateos MV, Mikhael J, Moreau P, Nagarajan C, Nooka A, O'Dwyer M, Schjesvold F, Sidana S, van de Donk NW, Weisel K, Zweegman S, Raje N, Otero PR, Anderson LD, Kumar S, Martin T. Consensus guidelines and recommendations for the management and response assessment of chimeric antigen receptor T-cell therapy in clinical practice for relapsed and refractory multiple myeloma: a report from the International Myeloma Working Group Immunotherapy Committee. Lancet Oncol 2024; 25:e374-e387. [PMID: 38821074 DOI: 10.1016/s1470-2045(24)00094-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/18/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 06/02/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has shown promise in patients with late-line refractory multiple myeloma, with response rates ranging from 73 to 98%. To date, three products have been approved: Idecabtagene vicleucel (ide-cel) and ciltacabtagene autoleucel (cilta-cel), which are approved by the US Food and Drug Administration, the European Medicines Agency, Health Canada (ide-cel only), and Brazil ANVISA (cilta-cel only); and equecabtagene autoleucel (eque-cel), which was approved by the Chinese National Medical Products Administration. CAR T-cell therapy is different from previous anti-myeloma therapeutics with unique toxic effects that require distinct mitigation strategies. Thus, a panel of experts from the International Myeloma Working Group was assembled to provide guidance for clinical use of CAR T-cell therapy in myeloma. This consensus opinion is from experts in the field of haematopoietic cell transplantation, cell therapy, and multiple myeloma therapeutics.
Collapse
Affiliation(s)
- Yi Lin
- Department of Hematology, Mayo Clinic, Rochester, MN, USA.
| | - Lugui Qiu
- National Clinical Research Center for Blood Diseases and State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Pekin Union Medical College, Tianjin, China
| | - Saad Usmani
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chng Wee Joo
- Department of Medical Oncology, National University Cancer Institute, Singapore
| | - Luciano Costa
- Department of Hematology Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Benjamin Derman
- Department of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Juan Du
- Department of Hematology, Myeloma and Lymphoma Center, Shanghai Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital, Wurzburg, Germany
| | - Carlos Fernandez de Larrea
- Department of Hematology, Hospital Clinic de Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Roman Hajek
- Department of Hematooncology, University Hospital Ostrava, Ostrava, Czech Republic; Department of Hematooncology, Faculty of Medicine, University Ostrava, Ostrava, Czech Republic
| | - P Joy Ho
- Institute of Hematology, Royal Prince Alfred Hospital, University of Sydney, Sydney, NSW, Australia
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Joaquin Martinez-Lopez
- Hospital Universitario 12 de Octubre, Department of Medicine, Complutense University, CNIO, Madrid, Spain
| | | | - Joseph Mikhael
- Translational Genomics Research Institute, City of Hope Cancer Center, Phoenix, AZ, USA
| | - Philippe Moreau
- Department of Hematology, University Hospital of Nantes, Nantes, France
| | | | - Ajay Nooka
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Michael O'Dwyer
- Department of Medicine and Department of Haematology, National University of Ireland, Galway, Ireland
| | | | - Surbhi Sidana
- School of Medicine, Stanford University, San Francisco, CA, USA
| | - Niels Wcj van de Donk
- Department of Hematology, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Katja Weisel
- Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sonja Zweegman
- Department of Hematology, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Noopur Raje
- Department of Medicine, Harvard University, Cambridge, MA, USA
| | - Paula Rodriguez Otero
- Department of Hematology, Cancer Center Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Larry D Anderson
- Simmons Comprehensive Cancer Center, Dallas, TX, USA; University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shaji Kumar
- Department of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Tom Martin
- Department of Hematology, University of California, San Francisco, CA, USA
| |
Collapse
|
10
|
Hughes AD, Teachey DT, Diorio C. Riding the storm: managing cytokine-related toxicities in CAR-T cell therapy. Semin Immunopathol 2024; 46:5. [PMID: 39012374 PMCID: PMC11252192 DOI: 10.1007/s00281-024-01013-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/11/2023] [Accepted: 03/18/2024] [Indexed: 07/17/2024]
Abstract
The advent of chimeric antigen receptor T cells (CAR-T) has been a paradigm shift in cancer immunotherapeutics, with remarkable outcomes reported for a growing catalog of malignancies. While CAR-T are highly effective in multiple diseases, salvaging patients who were considered incurable, they have unique toxicities which can be life-threatening. Understanding the biology and risk factors for these toxicities has led to targeted treatment approaches which can mitigate them successfully. The three toxicities of particular interest are cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and immune effector cell-associated hemophagocytic lymphohistiocytosis (HLH)-like syndrome (IEC-HS). Each of these is characterized by cytokine storm and hyperinflammation; however, they differ mechanistically with regard to the cytokines and immune cells that drive the pathophysiology. We summarize the current state of the field of CAR-T-associated toxicities, focusing on underlying biology and how this informs toxicity management and prevention. We also highlight several emerging agents showing promise in preclinical models and the clinic. Many of these established and emerging agents do not appear to impact the anti-tumor function of CAR-T, opening the door to additional and wider CAR-T applications.
Collapse
Affiliation(s)
- Andrew D Hughes
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David T Teachey
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Caroline Diorio
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Casey M, Lee C, Hoyte SM, Johnston RL, Kwok WY, Law SC, Gandhi MK, Harrison SJ, Nakamura K. Harnessing the cytotoxic granule exocytosis to augment the efficacy of T-cell-engaging bispecific antibody therapy. Haematologica 2024; 109:2131-2143. [PMID: 38268493 PMCID: PMC11215359 DOI: 10.3324/haematol.2023.284435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/30/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024] Open
Abstract
T-cell-engaging bispecific antibody (T-BsAb, also known as BiTE) therapy has emerged as a powerful therapeutic modality against multiple myeloma. Given that T-BsAb therapy redirects endogenous T cells to eliminate tumor cells, reinvigorating dysfunctional T cells may be a potential approach to improve the efficacy of T-BsAb. While various immunostimulatory cytokines can potentiate effector T-cell functions, the optimal cytokine treatment for T-BsAb therapy is yet to be established, partly due to a concern of cytokine release syndrome driven by aberrant interferon (IFN)-γ production. Here, we functionally screen immunostimulatory cytokines to determine an ideal combination partner for T-BsAb therapy. This approach reveals interleukin (IL)-21 as a potential immunostimulatory cytokine with the ability to augment T-BsAb-mediated release of granzyme B and perforin, without increasing IFN-γ release. Transcriptome profiling and functional characterization strongly support that IL-21 selectively targets the cytotoxic granule exocytosis pathway, but not pro-inflammatory responses. Notably, IL-21 modulates multiple steps of cytotoxic effector functions including upregulation of co-activating CD226 receptor, increasing cytotoxic granules, and promoting cytotoxic granule delivery at the immunological synapse. Indeed, T-BsAb-mediated myeloma killing is cytotoxic granule-dependent, and IL-21 priming significantly augments cytotoxic activities. Furthermore, in vivo IL-21 treatment induces cytotoxic effector reprogramming in bone marrow T cells, showing synergistic anti-myeloma effects in combination with T-BsAb therapy. Together, harnessing the cytotoxic granule exocytosis pathway by IL-21 may be a potential approach to achieve better responses by T-BsAb therapy.
Collapse
Affiliation(s)
- Mika Casey
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD
| | - Carol Lee
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD
| | - Sharon M Hoyte
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD
| | - Rebecca L Johnston
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD
| | - Wing Yu Kwok
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD
| | - Soi Cheng Law
- Mater Research, University of Queensland, Brisbane, QLD
| | | | - Simon J Harrison
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital Melbourne VIC Australia; Sir Peter MacCallum, Department of Oncology, University of Melbourne, Parkville
| | - Kyohei Nakamura
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD.
| |
Collapse
|
12
|
Qureshi Z, Altaf F, Jamil A, Siddique R. Optimization Strategies in CAR T-cell Therapy: A Comprehensive Evaluation of Cytopenia, HLH/MAS, and Other Adverse Events. Am J Clin Oncol 2024:00000421-990000000-00204. [PMID: 38907604 DOI: 10.1097/coc.0000000000001124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 06/24/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has emerged as a transformative treatment for various hematological malignancies. Still, its remarkable efficacy is accompanied by unique adverse events that must be carefully managed. This comprehensive literature review evaluates the safety profile of CAR T-cell therapy, focusing on cytopenia, hemophagocytic lymphohistiocytosis (HLH)/macrophage activation syndrome (MAS), and other potential complications. Cytopenia, characterized by reduced blood cell counts, affects a significant proportion of patients, with rates of anemia, neutropenia, and thrombocytopenia reaching up to 60%, 70%, and 80%, respectively. Risk factors include high tumor burden, prior chemotherapy, and bone marrow involvement. Cytokine release syndrome (CRS) occurs in 13% to 77% of patients and is linked to the cytokine storm induced by CAR T cells, target antigen expression, and preexisting immune dysregulation. Other notable adverse events discussed are cytokine release syndrome, neurotoxicity, and infections. Understanding the mechanisms, risk factors, and management strategies for these adverse events is crucial for optimizing patient outcomes and unlocking the full potential of this revolutionary therapy. The review highlights the need for continued research, interdisciplinary collaboration, and evidence-based approaches to enhance the safety and efficacy of CAR T-cell therapy.
Collapse
Affiliation(s)
- Zaheer Qureshi
- Department of Medicine, The Frank H. Netter MD School of Medicine at Quinnipiac University, Bridgeport, CT
| | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York
| | - Abdur Jamil
- Department of Medicine, Samaritan Medical Centre
| | | |
Collapse
|
13
|
Shah K, Leandro M, Cragg M, Kollert F, Schuler F, Klein C, Reddy V. Disrupting B and T-cell collaboration in autoimmune disease: T-cell engagers versus CAR T-cell therapy? Clin Exp Immunol 2024; 217:15-30. [PMID: 38642912 PMCID: PMC11188544 DOI: 10.1093/cei/uxae031] [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] [Academic Contribution Register] [Received: 10/07/2023] [Revised: 02/07/2024] [Accepted: 04/18/2024] [Indexed: 04/22/2024] Open
Abstract
B and T cells collaborate to drive autoimmune disease (AID). Historically, B- and T-cell (B-T cell) co-interaction was targeted through different pathways such as alemtuzumab, abatacept, and dapirolizumab with variable impact on B-cell depletion (BCD), whereas the majority of patients with AID including rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, and organ transplantation benefit from targeted BCD with anti-CD20 monoclonal antibodies such as rituximab, ocrelizumab, or ofatumumab. Refractory AID is a significant problem for patients with incomplete BCD with a greater frequency of IgD-CD27+ switched memory B cells, CD19+CD20- B cells, and plasma cells that are not directly targeted by anti-CD20 antibodies, whereas most lymphoid tissue plasma cells express CD19. Furthermore, B-T-cell collaboration is predominant in lymphoid tissues and at sites of inflammation such as the joint and kidney, where BCD may be inefficient, due to limited access to key effector cells. In the treatment of cancer, chimeric antigen receptor (CAR) T-cell therapy and T-cell engagers (TCE) that recruit T cells to induce B-cell cytotoxicity have delivered promising results for anti-CD19 CAR T-cell therapies, the CD19 TCE blinatumomab and CD20 TCE such as mosunetuzumab, glofitamab, or epcoritamab. Limited evidence suggests that anti-CD19 CAR T-cell therapy may be effective in managing refractory AID whereas we await evaluation of TCE for use in non-oncological indications. Therefore, here, we discuss the potential mechanistic advantages of novel therapies that rely on T cells as effector cells to disrupt B-T-cell collaboration toward overcoming rituximab-resistant AID.
Collapse
Affiliation(s)
| | - Maria Leandro
- Centre for Rheumatology, UCLH, London,UK
- Department of Rheumatology, University College London Hospital, London, UK
| | - Mark Cragg
- University of Southampton Faculty of Medicine, Antibody and Vaccine Group, Centre for Cancer Immunology, University of Southampton, Southampton, UK
| | - Florian Kollert
- Roche Innovation Center Basel, Early Development Immunology, Infectious Diseases & Ophthalmology, Basel, Switzerland
| | - Franz Schuler
- Roche Innovation Center Basel, Roche Pharma Research and Early Development, Schlieren, Switzerland
| | - Christian Klein
- Roche Innovation Center Zurich, Cancer Immunotherapy Discovery, Oncology Discovery & Translational Area, Schlieren, Switzerland
| | - Venkat Reddy
- Centre for Rheumatology, UCLH, London,UK
- Department of Rheumatology, University College London Hospital, London, UK
| |
Collapse
|
14
|
Pullarkat S, Black G, Bleakley M, Buenrostro D, Chapuis AG, Hirayama AV, Jaeger-Ruckstuhl CA, Kimble EL, Lee BM, Maloney DG, Radich J, Seaton BW, Specht JM, Turtle CJ, Woolston DW, Wright JH, Yeung CCS. qPCR assay for detection of Woodchuck Hepatitis Virus Post-Transcriptional Regulatory Elements from CAR-T and TCR-T cells in fresh and formalin-fixed tissue. PLoS One 2024; 19:e0303057. [PMID: 38843256 PMCID: PMC11156344 DOI: 10.1371/journal.pone.0303057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/29/2024] [Accepted: 04/19/2024] [Indexed: 06/09/2024] Open
Abstract
As adoptive cellular therapies become more commonplace in cancer care, there is a growing need to monitor site-specific localization of engineered cells-such as chimeric antigen receptor T (CAR-T) cells and T-cell receptor T (TCR-T) cells-in patients' tissues to understand treatment effectiveness as well as associated adverse events. Manufacturing CAR-T and TCR-T cells involves transduction with viral vectors commonly containing the WPRE gene sequence to enhance gene expression, providing a viable assay target unique to these engineered cells. Quantitative PCR (qPCR) is currently used clinically in fresh patient tissue samples and blood with target sequences specific to each immunotherapy product. Herein, we developed a WPRE-targeted qPCR assay that is broadly applicable for detection of engineered cell products in both fresh and archival formalin-fixed paraffin embedded (FFPE) tissues. Using both traditional PCR and SYBR Green PCR protocols, we demonstrate the use of this WPRE-targeted assay to successfully detect two CAR-T cell and two TCR-T cell products in FFPE tissue. Standard curve analysis reported a reproducible limit of detection at 100 WPRE copies per 20μL PCR reaction. This novel and inexpensive technique could provide better understanding of tissue abundance of engineered therapeutic T cells in both tumor and second-site toxicity tissues and provide quantitative assessment of immune effector cell trafficking in archival tissue.
Collapse
MESH Headings
- Humans
- Formaldehyde
- Hepatitis B Virus, Woodchuck/genetics
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tissue Fixation/methods
- Immunotherapy, Adoptive/methods
- Real-Time Polymerase Chain Reaction/methods
Collapse
Affiliation(s)
- Shalini Pullarkat
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Graeme Black
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Marie Bleakley
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
- Program in Immunology, University of Washington, Seattle, Washington, United States of America
| | - Denise Buenrostro
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Program in Immunology, University of Washington, Seattle, Washington, United States of America
| | - Aude G. Chapuis
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Program in Immunology, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, Washington, United States of America
| | - Alexandre V. Hirayama
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, Washington, United States of America
| | - Carla A. Jaeger-Ruckstuhl
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Erik L. Kimble
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, Washington, United States of America
| | - Bo M. Lee
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - David G. Maloney
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, Washington, United States of America
| | - Jerald Radich
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Brandon W. Seaton
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Jennifer M. Specht
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, Washington, United States of America
| | - Cameron J. Turtle
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, Washington, United States of America
- Integrated Immunotherapy Research Center, Fred Hutchinson Cancer Center, Seattle, WA, United States of America
| | - David W. Woolston
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Jocelyn H. Wright
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Program in Immunology, University of Washington, Seattle, Washington, United States of America
| | - Cecilia C. S. Yeung
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| |
Collapse
|
15
|
Ferreri CJ, Bhutani M. Mechanisms and management of CAR T toxicity. Front Oncol 2024; 14:1396490. [PMID: 38835382 PMCID: PMC11148294 DOI: 10.3389/fonc.2024.1396490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/05/2024] [Accepted: 05/07/2024] [Indexed: 06/06/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapies have dramatically improved treatment outcomes for patients with relapsed or refractory B-cell acute lymphoblastic leukemia, large B-cell lymphoma, follicular lymphoma, mantle cell lymphoma, and multiple myeloma. Despite unprecedented efficacy, treatment with CAR T cell therapies can cause a multitude of adverse effects which require monitoring and management at specialized centers and contribute to morbidity and non-relapse mortality. Such toxicities include cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, neurotoxicity distinct from ICANS, immune effector cell-associated hemophagocytic lymphohistiocytosis-like syndrome, and immune effector cell-associated hematotoxicity that can lead to prolonged cytopenias and infectious complications. This review will discuss the current understanding of the underlying pathophysiologic mechanisms and provide guidelines for the grading and management of such toxicities.
Collapse
Affiliation(s)
- Christopher J Ferreri
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health Wake Forest University School of Medicine, Charlotte, NC, United States
| | - Manisha Bhutani
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health Wake Forest University School of Medicine, Charlotte, NC, United States
| |
Collapse
|
16
|
Jallouk AP, Kui N, Sun R, Westin JR, Steiner RE, Nair R, Nastoupil LJ, Fayad LE, Zaki AA, Hawkins M, Adkins S, Noorani M, Das K, Henderson J, Shpall EJ, Kebriaei P, Ramdial J, Flowers CR, Neelapu SS, Ahmed S, Strati P. Effect of delayed cell infusion in patients with large B-cell lymphoma treated with chimeric antigen receptor T-cell therapy. Haematologica 2024; 109:1460-1468. [PMID: 38031807 PMCID: PMC11063866 DOI: 10.3324/haematol.2023.284453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/12/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023] Open
Abstract
Complications occurring after lymphodepleting chemotherapy (LDC) may delay chimeric antigen receptor (CAR) T-cell infusion. The effect of these delays on clinical outcomes is unclear. We performed a retrospective analysis of 240 patients with relapsed/refractory large B-cell lymphoma treated with standard-of-care axicabtagene ciloleucel (axi-cel) and identified 40 patients (16.7%) who had delay in axi-cel infusion. Of these, 85% had delay due to infection. At time of LDC initiation, patients with delayed infusion had lower absolute neutrophil count (P=0.006), lower platelets (P=0.004), lower hemoglobin (P<0.001) and higher C-reactive protein (P=0.001) than those with on-time infusion. Patients with delayed infusion had lower day 30 overall response rates (59.0% vs. 79.4%; P=0.008) and shorter median progression-free survival (PFS) (3.5 vs. 8.2 months; P=0.002) and overall survival (7.8 vs. 26.4 months; P=0.046) than those with on-time infusion. The association with PFS was maintained on multivariate analysis. There was also an association between extent of delay and survival, with shorter median PFS in patients who had delays of 2-5 days (1.8 vs. 8.2 months; P=0.001) and >5 days (4.6 vs. 8.2 months; P=0.036), but not 1 day (5.7 vs. 8.2 months; P=0.238). Following propensity score matching, patients with delayed infusion continued to have shorter median PFS (3.5 vs. 6.0 months; P=0.015). Levels of pro-inflammatory cytokines on day of infusion were significantly higher in patients with delayed infusion. Together, these findings suggest that delays in CAR T-cell administration after initiation of LDC are associated with inferior outcomes. Further studies are needed to guide strategies to improve efficacy in such patients.
Collapse
Affiliation(s)
- Andrew P Jallouk
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN
| | - Naishu Kui
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ryan Sun
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jason R Westin
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Raphael E Steiner
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ranjit Nair
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Loretta J Nastoupil
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Luis E Fayad
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ajlan Al Zaki
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Misha Hawkins
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sherry Adkins
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mansoor Noorani
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kaberi Das
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jared Henderson
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jeremy Ramdial
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Christopher R Flowers
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sattva S Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sairah Ahmed
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX.
| | - Paolo Strati
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
17
|
Kim SJ, Yoon SE, Kim WS. Current Challenges in Chimeric Antigen Receptor T-cell Therapy in Patients With B-cell Lymphoid Malignancies. Ann Lab Med 2024; 44:210-221. [PMID: 38205527 PMCID: PMC10813822 DOI: 10.3343/alm.2023.0388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/30/2023] [Revised: 11/18/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a promising immunotherapy based on genetically engineered T cells derived from patients. The introduction of CAR T-cell therapy has changed the treatment paradigm of patients with B-cell lymphoid malignancies. However, challenging issues including managing life-threatening toxicities related to CAR T-cell infusion and resistance to CAR T-cell therapy, leading to progression or relapse, remain. This review summarizes the issues with currently approved CAR T-cell therapies for patients with relapsed or refractory B-cell lymphoid malignancies, including lymphoma and myeloma. We focus on unique toxicities after CAR T-cell therapy, such as cytokine-related events and hematological toxicities, and the mechanisms underlying post-CAR T-cell failure.
Collapse
Affiliation(s)
- Seok Jin Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
- CAR T-cell Therapy Center, Samsung Comprehensive Cancer Center, Seoul, Korea
| | - Sang Eun Yoon
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- CAR T-cell Therapy Center, Samsung Comprehensive Cancer Center, Seoul, Korea
| | - Won Seog Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
- CAR T-cell Therapy Center, Samsung Comprehensive Cancer Center, Seoul, Korea
| |
Collapse
|
18
|
Shumnalieva R, Velikova T, Monov S. Expanding the role of CAR T-cell therapy: From B-cell hematological malignancies to autoimmune rheumatic diseases. Int J Rheum Dis 2024; 27:e15182. [PMID: 38742463 DOI: 10.1111/1756-185x.15182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/23/2023] [Revised: 04/04/2024] [Accepted: 04/28/2024] [Indexed: 05/16/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a form of immunotherapy where the lymphocytes, mostly T-cells, are redirected to specifically recognize and eliminate a target antigen by coupling them with CARs. The binding of CAR and target cell surface antigens leads to vigorous T cell activation and robust anti-tumor immune responses. Areas of implication of CAR T-cell therapies include mainly hematological malignancies (i.e., advanced B-cell cancers); however, recent studies have proven the unprecedented success of the new immunotherapy also in autoimmune rheumatic diseases. We aim to review the recent advances in CAR T-cell therapies in rheumatology but also to address the limitations of their use in the real clinical practice based on the data on their efficacy and safety.
Collapse
Affiliation(s)
- Russka Shumnalieva
- Department of Rheumatology, Clinic of Rheumatology, Medical University-Sofia, Faculty of Medicine, Sofia, Bulgaria
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University "St. Kliment Ohridski"- Sofia, Sofia, Bulgaria
| | - Simeon Monov
- Department of Rheumatology, Clinic of Rheumatology, Medical University-Sofia, Faculty of Medicine, Sofia, Bulgaria
| |
Collapse
|
19
|
Ruggeri Barbaro N, Drashansky T, Tess K, Djedaini M, Hariri R, He S, van der Touw W, Karasiewicz K. Placental circulating T cells: a novel, allogeneic CAR-T cell platform with preserved T-cell stemness, more favorable cytokine profile, and durable efficacy compared to adult PBMC-derived CAR-T. J Immunother Cancer 2024; 12:e008656. [PMID: 38684370 PMCID: PMC11107807 DOI: 10.1136/jitc-2023-008656] [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] [Academic Contribution Register] [Accepted: 04/12/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-T cell quality and stemness are associated with responsiveness, durability, and memory formation, which benefit clinical responses. Autologous T cell starting material across patients with cancer is variable and CAR-T expansion or potency can fail during manufacture. Thus, strategies to develop allogeneic CAR-T platforms including the identification and expansion of T cell subpopulations that correspond with CAR-T potency are an active area of investigation. Here, we compared CAR-T cells generated from healthy adult peripheral blood T cells versus placental circulating T (P-T) cells. METHODS CAR-T cells from healthy adult peripheral blood mononuclear cells (PBMCs) and P-T cells were generated using the same protocol. CAR-T cells were characterized in detail by a combination of multiparameter flow cytometry, functional assays, and RNA sequencing. In vivo antitumor efficacy and persistence of CAR-T cells were evaluated in a Daudi lymphoma xenograft model. RESULTS P-T cells possess stemness advantages compared with T cells from adult PBMCs. P-T cells are uniformly naïve prior to culture initiation, maintain longer telomeres, resist immune checkpoint upregulation, and resist further differentiation compared with PBMC T cells during CD19 CAR-T manufacture. P-T CD19 CAR-T cells are equally cytotoxic as PBMC-CD19 CAR-T cells but produce less interferon gamma in response to lymphoma. Transcriptome analysis shows P-T CD19 CAR-T cells retain a stem-like gene signature, strongly associate with naïve T cells, an early memory phenotype, and a unique CD4 T cell signature compared with PBMC-CD19 CAR-T cells, which enrich for exhaustion and stimulated memory T cell signatures. Consistent with functional data, P-T CD19 CAR-T cells exhibit attenuated inflammatory cytokine and chemokine gene signatures. In a murine in vivo model, P-T CD19 CAR-T cells eliminate lymphoma beyond 90 days. PBMC-CD19 CAR-T cells provide a non-durable benefit, which only delays disease onset. CONCLUSION We identified characteristics of T cell stemness enriched in P-T CD19 CAR-T which are deficient in PBMC-derived products and translate into response durability in vivo. Our findings demonstrate that placental circulating T cells are a valuable cell source for allogeneic CAR-T products. Stemness advantages inherent to P-T cells translate to in vivo persistence advantages and long-term durable activity.
Collapse
Affiliation(s)
| | | | | | | | | | - Shuyang He
- Celularity Inc, Florham Park, New Jersey, USA
| | | | | |
Collapse
|
20
|
Varsha KK, Yang X, Cannon AS, Zhong Y, Nagarkatti M, Nagarkatti P. Identification of miRNAs that target Fcγ receptor-mediated phagocytosis during macrophage activation syndrome. Front Immunol 2024; 15:1355315. [PMID: 38558807 PMCID: PMC10981272 DOI: 10.3389/fimmu.2024.1355315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/13/2023] [Accepted: 02/27/2024] [Indexed: 04/04/2024] Open
Abstract
Macrophage activation syndrome (MAS) is a life-threatening complication of systemic juvenile arthritis, accompanied by cytokine storm and hemophagocytosis. In addition, COVID-19-related hyperinflammation shares clinical features of MAS. Mechanisms that activate macrophages in MAS remain unclear. Here, we identify the role of miRNA in increased phagocytosis and interleukin-12 (IL-12) production by macrophages in a murine model of MAS. MAS significantly increased F4/80+ macrophages and phagocytosis in the mouse liver. Gene expression profile revealed the induction of Fcγ receptor-mediated phagocytosis (FGRP) and IL-12 production in the liver. Phagocytosis pathways such as High-affinity IgE receptor is known as Fc epsilon RI -signaling and pattern recognition receptors involved in the recognition of bacteria and viruses and phagosome formation were also significantly upregulated. In MAS, miR-136-5p and miR-501-3p targeted and caused increased expression of Fcgr3, Fcgr4, and Fcgr1 genes in FGRP pathway and consequent increase in phagocytosis by macrophages, whereas miR-129-1-3p and miR-150-3p targeted and induced Il-12. Transcriptome analysis of patients with MAS revealed the upregulation of FGRP and FCGR gene expression. A target analysis of gene expression data from a patient with MAS discovered that miR-136-5p targets FCGR2A and FCGR3A/3B, the human orthologs of mouse Fcgr3 and Fcgr4, and miR-501-3p targets FCGR1A, the human ortholog of mouse Fcgr1. Together, we demonstrate the novel role of miRNAs during MAS pathogenesis, thereby suggesting miRNA mimic-based therapy to control the hyperactivation of macrophages in patients with MAS as well as use overexpression of FCGR genes as a marker for MAS classification.
Collapse
Affiliation(s)
| | | | | | | | | | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina School of Medicine, Columbia, SC, United States
| |
Collapse
|
21
|
Walton ZE, Frigault MJ, Maus MV. Current and emerging pharmacotherapies for cytokine release syndrome, neurotoxicity, and hemophagocytic lymphohistiocytosis-like syndrome due to CAR T cell therapy. Expert Opin Pharmacother 2024; 25:263-279. [PMID: 38588525 DOI: 10.1080/14656566.2024.2340738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/12/2023] [Accepted: 03/01/2024] [Indexed: 04/10/2024]
Abstract
INTRODUCTION Chimeric antigen receptor (CAR) T cells have revolutionized the treatment of multiple hematologic malignancies. Engineered cellular therapies now offer similar hope to transform the management of solid tumors and autoimmune diseases. However, toxicities can be serious and often require hospitalization. AREAS COVERED We review the two chief toxicities of CAR T therapy, cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), and the rarer immune effector cell-associated hemophagocytic lymphohistiocytosis-like syndrome. We discuss treatment paradigms and promising future pharmacologic strategies. Literature and therapies reviewed were identified by PubMed search, cited references therein, and review of registered trials. EXPERT OPINION Management of CRS and ICANS has improved, aided by consensus definitions and guidelines that facilitate recognition and timely intervention. Further data will define optimal timing of tocilizumab and corticosteroids, current foundations of management. Pathophysiologic understanding has inspired off-label use of IL-1 receptor antagonism, IFNγ and IL-6 neutralizing antibodies, and janus kinase inhibitors, with data emerging from ongoing clinical trials. Further strategies to reduce toxicities include novel pharmacologic targets and safety features engineered into CAR T cells themselves. As these potentially curative therapies are used earlier in oncologic therapy and even in non-oncologic indications, effective accessible strategies to manage toxicities are critical.
Collapse
Affiliation(s)
- Zandra E Walton
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Division of Rheumatology, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Matthew J Frigault
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
22
|
McNerney KO, Hsieh EM, Shalabi H, Epperly R, Wolters PL, Hill JA, Gardner R, Talleur AC, Shah NN, Rossoff J. INSPIRED Symposium Part 3: Prevention and Management of Pediatric Chimeric Antigen Receptor T Cell-Associated Emergent Toxicities. Transplant Cell Ther 2024; 30:38-55. [PMID: 37821079 PMCID: PMC10842156 DOI: 10.1016/j.jtct.2023.10.006] [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] [Academic Contribution Register] [Received: 09/07/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023]
Abstract
Chimeric antigen receptor (CAR) T cell (CAR-T) therapy has emerged as a revolutionary cancer treatment modality, particularly in children and young adults with B cell malignancies. Through clinical trials and real-world experience, much has been learned about the unique toxicity profile of CAR-T therapy. The past decade brought advances in identifying risk factors for severe inflammatory toxicities, investigating preventive measures to mitigate these toxicities, and exploring novel strategies to manage refractory and newly described toxicities, infectious risks, and delayed effects, such as cytopenias. Although much progress has been made, areas needing further improvements remain. Limited guidance exists regarding initial administration of tocilizumab with or without steroids and the management of inflammatory toxicities refractory to these treatments. There has not been widespread adoption of preventive strategies to mitigate inflammation in patients at high risk of severe toxicities, particularly children. Additionally, the majority of research related to CAR-T toxicity prevention and management has focused on adult populations, with only a few pediatric-specific studies published to date. Given that children and young adults undergoing CAR-T therapy represent a unique population with different underlying disease processes, physiology, and tolerance of toxicities than adults, it is important that studies be conducted to evaluate acute, delayed, and long-term toxicities following CAR-T therapy in this younger age group. In this pediatric-focused review, we summarize key findings on CAR-T therapy-related toxicities over the past decade, highlight emergent CAR-T toxicities, and identify areas of greatest need for ongoing research.
Collapse
Affiliation(s)
- Kevin O McNerney
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.
| | - Emily M Hsieh
- Pediatric Hematology/Oncology, Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, California
| | - Haneen Shalabi
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Rebecca Epperly
- Department of Bone Marrow Transplant, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Pamela L Wolters
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Joshua A Hill
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Rebecca Gardner
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Aimee C Talleur
- Department of Bone Marrow Transplant, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jenna Rossoff
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| |
Collapse
|
23
|
Fugere T, Baltz A, Mukherjee A, Gaddam M, Varma A, Veeraputhiran M, Gentille Sanchez CG. Immune Effector Cell-Associated HLH-like Syndrome: A Review of the Literature of an Increasingly Recognized Entity. Cancers (Basel) 2023; 15:5149. [PMID: 37958323 PMCID: PMC10647774 DOI: 10.3390/cancers15215149] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/23/2023] [Revised: 10/21/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Since CAR-T cell therapy was initially approved in 2017, its use has become more prevalent and so have its side effects. CAR-T-related HLH, also named immune effector cell-associated HLH-like syndrome (IEC-HS), is a rare but fatal toxicity if not recognized promptly. We conducted a review of the literature in order to understand the prevalence of IEC-HS as well as clarify the evolution of the diagnostic criteria and treatment recommendations. IEC-HS occurrence varies between CAR-T cell products and the type of malignancy treated. Diagnosis can be challenging as there are no standardized diagnostic criteria, and its clinical features can overlap with cytokine release syndrome and active hematological disease. Suggested treatment strategies have been extrapolated from prior experience in HLH and include anakinra, corticosteroids and ruxolitinib. IEC-HS is a potentially fatal toxicity associated with CAR-T cell therapy. Early recognition with reliable diagnostic criteria and prompt implementation of treatment specific to IEC-HS is imperative for improving patient outcomes.
Collapse
Affiliation(s)
- Tyler Fugere
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.B.); (A.M.); (M.G.); (A.V.); (M.V.); (C.G.G.S.)
| | | | | | | | | | | | | |
Collapse
|
24
|
Schuelke MR, Bassiri H, Behrens EM, Canna S, Croy C, DiNofia A, Gollomp K, Grupp S, Lambert M, Lambrix A, Maude SL, Myers R, Newman H, Petrosa W, Seif A, Sullivan KE, Teachey DT, Diorio C. Emapalumab for the treatment of refractory cytokine release syndrome in pediatric patients. Blood Adv 2023; 7:5603-5607. [PMID: 37721859 PMCID: PMC10514204 DOI: 10.1182/bloodadvances.2023010712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/11/2023] [Accepted: 07/27/2023] [Indexed: 09/20/2023] Open
Affiliation(s)
- Matthew R. Schuelke
- Pediatrics Residency Program, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Hamid Bassiri
- Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Edward M. Behrens
- Division of Rheumatology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Scott Canna
- Division of Rheumatology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Colleen Croy
- Department of Pharmacy, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Amanda DiNofia
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Kandace Gollomp
- Division of Hematology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Stephan Grupp
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Michele Lambert
- Division of Hematology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Arathi Lambrix
- Department of Pharmacy, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Shannon L. Maude
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Regina Myers
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Haley Newman
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Whitney Petrosa
- Immune Dysregulation Program, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Alix Seif
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Kathleen E. Sullivan
- Division of Immunology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - David T. Teachey
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Caroline Diorio
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
25
|
Rejeski K, Subklewe M, Aljurf M, Bachy E, Balduzzi A, Barba P, Bruno B, Benjamin R, Carrabba MG, Chabannon C, Ciceri F, Corradini P, Delgado J, Di Blasi R, Greco R, Houot R, Iacoboni G, Jäger U, Kersten MJ, Mielke S, Nagler A, Onida F, Peric Z, Roddie C, Ruggeri A, Sánchez-Guijo F, Sánchez-Ortega I, Schneidawind D, Schubert ML, Snowden JA, Thieblemont C, Topp M, Zinzani PL, Gribben JG, Bonini C, Sureda A, Yakoub-Agha I. Immune effector cell-associated hematotoxicity: EHA/EBMT consensus grading and best practice recommendations. Blood 2023; 142:865-877. [PMID: 37300386 DOI: 10.1182/blood.2023020578] [Citation(s) in RCA: 110] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/27/2023] [Revised: 05/15/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Hematological toxicity is the most common adverse event after chimeric antigen receptor (CAR) T-cell therapy. Cytopenias can be profound and long-lasting and can predispose for severe infectious complications. In a recent worldwide survey, we demonstrated that there remains considerable heterogeneity in regard to current practice patterns. Here, we sought to build consensus on the grading and management of immune effector cell-associated hematotoxicity (ICAHT) after CAR T-cell therapy. For this purpose, a joint effort between the European Society for Blood and Marrow Transplantation (EBMT) and the European Hematology Association (EHA) involved an international panel of 36 CAR T-cell experts who met in a series of virtual conferences, culminating in a 2-day meeting in Lille, France. On the basis of these deliberations, best practice recommendations were developed. For the grading of ICAHT, a classification system based on depth and duration of neutropenia was developed for early (day 0-30) and late (after day +30) cytopenia. Detailed recommendations on risk factors, available preinfusion scoring systems (eg, CAR-HEMATOTOX score), and diagnostic workup are provided. A further section focuses on identifying hemophagocytosis in the context of severe hematotoxicity. Finally, we review current evidence and provide consensus recommendations for the management of ICAHT, including growth factor support, anti-infectious prophylaxis, transfusions, autologous hematopoietic stem cell boost, and allogeneic hematopoietic cell transplantation. In conclusion, we propose ICAHT as a novel toxicity category after immune effector cell therapy, provide a framework for its grading, review literature on risk factors, and outline expert recommendations for the diagnostic workup and short- and long-term management.
Collapse
Affiliation(s)
- Kai Rejeski
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Marion Subklewe
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Mahmoud Aljurf
- Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Emmanuel Bachy
- Department of Hematology, Hospices Civils de Lyon and Université Claude Bernard Lyon 1, Lyon, France
| | - Adriana Balduzzi
- Pediatric Transplantation Unit, Department of Medicine and Surgery, University of Milan-Bicocca-Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Pere Barba
- Department of Hematology, Vall d'Hebron University Hospital, Experimental Hematology, Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Benedetto Bruno
- Division of Hematology and Cell Therapy Unit, Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Reuben Benjamin
- School of Cancer & Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Matteo G Carrabba
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Milan, Italy
| | - Christian Chabannon
- Institut Paoli-Calmettes Comprehensive Cancer Centre and Module Biothérapies du Centre d'Investigations Cliniques de Marseille, INSERM-Aix-Marseille Université-AP-HM-IPC, CBT-1409, Marseille, France
| | - Fabio Ciceri
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Milan, Italy
| | - Paolo Corradini
- Division of Hematology and Stem Cell Transplantation, Fondazione IRCCS Instituto Nazionale dei Tumori, University of Milan, Milan, Italy
| | - Julio Delgado
- Oncoimmunotherapy Unit, Department of Hematology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Roberta Di Blasi
- Université de Paris, Assistance Publique-Hopitaux de Paris, Service d'hémato-oncologie, Paris, France
| | - Raffaella Greco
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Milan, Italy
| | - Roch Houot
- Department of Hematology, CHU Rennes, University of Rennes, INSERM U1236, Rennes, France
| | - Gloria Iacoboni
- Department of Hematology, Vall d'Hebron University Hospital, Experimental Hematology, Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Ulrich Jäger
- Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Marie José Kersten
- Department of Hematology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Stephan Mielke
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Department of Laboratory Medicine and Medicine Huddinge, Karolinska University Hospital and Institute, Stockholm, Sweden
| | - Arnon Nagler
- Division of Hematology, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | - Francesco Onida
- Hematology and Bone Marrow Transplantation Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Zinaida Peric
- Department of Hematology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Claire Roddie
- Department of Hematology, University College London Hospital, London, United Kingdom
| | - Annalisa Ruggeri
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Milan, Italy
| | - Fermín Sánchez-Guijo
- University of Salamanca, IBSAL-University Hospital of Salamanca, Salamanca, Spain
| | - Isabel Sánchez-Ortega
- Executive Office, European Society for Blood and Marrow Transplantation, Barcelona, Spain
| | - Dominik Schneidawind
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | | | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Catherine Thieblemont
- Université de Paris, Assistance Publique-Hopitaux de Paris, Service d'hémato-oncologie, Paris, France
| | - Max Topp
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli," Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - John G Gribben
- Barts Cancer Institute, Queen Mary, University of London, London, United Kingdom
| | - Chiara Bonini
- Division of Immunology, Transplantation and Infectious Disease, Experimental Hematology Unit, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Anna Sureda
- Clinical Hematology Department, Institut Català d'Oncologia-L'Hospitalet, Barcelona, Spain
| | | |
Collapse
|
26
|
Chen S, Zhang C, Chen D, Dong L, Chang T, Tang ZH. Advances in attractive therapeutic approach for macrophage activation syndrome in COVID-19. Front Immunol 2023; 14:1200289. [PMID: 37483597 PMCID: PMC10358730 DOI: 10.3389/fimmu.2023.1200289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/04/2023] [Accepted: 06/12/2023] [Indexed: 07/25/2023] Open
Abstract
Nowadays, people have relaxed their vigilance against COVID-19 due to its declining infection numbers and attenuated virulence. However, COVID-19 still needs to be concern due to its emerging variants, the relaxation of restrictions as well as breakthrough infections. During the period of the COVID-19 infection, the imbalanced and hyper-responsive immune system plays a critical role in its pathogenesis. Macrophage Activation Syndrome (MAS) is a fatal complication of immune system disease, which is caused by the excessive activation and proliferation of macrophages and cytotoxic T cells (CTL). COVID-19-related hyperinflammation shares common clinical features with the above MAS symptoms, such as hypercytokinemia, hyperferritinemia, and coagulopathy. In MAS, immune exhaustion or defective anti-viral responses leads to the inadequate cytolytic capacity of CTL which contributes to prolonged interaction between CTL, APCs and macrophages. It is possible that the same process also occurred in COVID-19 patients, and further led to a cytokine storm confined to the lungs. It is associated with the poor prognosis of severe patients such as multiple organ failure and even death. The main difference of cytokine storm is that in COVID-19 pneumonia is mainly the specific damage of the lung, while in MAS is easy to develop into a systemic. The attractive therapeutic approach to prevent MAS in COVID-19 mainly includes antiviral, antibiotics, convalescent plasma (CP) therapy and hemadsorption, extensive immunosuppressive agents, and cytokine-targeted therapies. Here, we discuss the role of the therapeutic approaches mentioned above in the two diseases. And we found that the treatment effect of the same therapeutic approach is different.
Collapse
Affiliation(s)
- Shunyao Chen
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Wuhan, China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong Zhang
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Wuhan, China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Deng Chen
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Wuhan, China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liming Dong
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Wuhan, China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Teding Chang
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Wuhan, China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao-Hui Tang
- Department of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Wuhan, China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|