1
|
Renninger J, Kurz L, Stein H. Mitigation and Management of Common Toxicities Associated with the Administration of CAR-T Therapies in Oncology Patients. Drug Saf 2025; 48:719-737. [PMID: 40108072 DOI: 10.1007/s40264-025-01538-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2025] [Indexed: 03/22/2025]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapies are one of the main approaches among targeted cellular therapies. Despite the potential benefit and durable responses observed in some patients receiving CAR-T therapies, serious and potentially fatal toxicities remain a major challenge. The most common CAR-T-associated toxicities include cytokine release syndrome (CRS), neurotoxicity, cytopenias, and infections. While CRS and neurotoxicity are generally managed with tocilizumab and corticosteroids, respectively, high-grade toxicities can be life-threatening. Close postinfusion monitoring and assessment of clinical laboratory parameters, patient-related and clinical risk factors (e.g., age, tumor burden, comorbidities, baseline laboratory parameters, and underlying abnormalities), and therapy-related risk factors (e.g., CAR-T type, dose, and CAR-T-induced toxicity) are effective strategies to mitigate the toxicities. Clinical laboratory parameters, including various cytokines, have been identified for CRS (interleukin [IL]-1, IL-2, IL-5, IL-6, IL-8, IL-10, C-reactive protein [CRP], interferon [IFN]-γ, ferritin, granulocyte-macrophage colony-stimulating factor [GM-CSF], and monocyte chemoattractant protein-1), neurotoxicity (IL-1, IL-2, IL-6, IL-15, tumor necrosis factor [TNF]-α, GM-CSF, and IFN-γ), cytopenias (IL-2, IL-4, IL-6, IL-10, IFN-γ, ferritin, and CRP), and infections (IL-8, IL-1β, CRP, IFN-γ, and procalcitonin). CAR-T-associated toxicities can be monitored and treated to mitigate the risk to patients. Assessment of alterations in clinical laboratory parameter values that are correlated with CAR-T-associated toxicities may predict development and/or severity of a given toxicity, which can improve patient management strategies and ultimately enable the patients to better tolerate these therapies.
Collapse
Affiliation(s)
- Jonathan Renninger
- GSK Safety Evaluation and Risk Management, Global Safety, Philadelphia, PA, USA.
| | - Lisa Kurz
- GSK Safety Evaluation and Risk Management, Global Safety, Upper Providence, PA, USA
| | - Heather Stein
- GSK Safety Evaluation and Risk Management, Global Safety, Cambridge, MA, USA
| |
Collapse
|
2
|
Thomas M, Brabenec R, Gregor L, Andreu-Sanz D, Carlini E, Müller PJ, Gottschlich A, Simnica D, Kobold S, Marr C. The role of single cell transcriptomics for efficacy and toxicity profiling of chimeric antigen receptor (CAR) T cell therapies. Comput Biol Med 2025; 192:110332. [PMID: 40375426 DOI: 10.1016/j.compbiomed.2025.110332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 04/29/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025]
Abstract
CAR T cells are genetically modified T cells that target specific epitopes. CAR T cell therapy has proven effective in difficult-to-treat B cell cancers and is now expanding into hematology and solid tumors. To date, approved CAR therapies target only two specific epitopes on cancer cells. Identifying more suitable targets is challenged by the lack of truly cancer-specific structures and the potential for on-target off-tumor toxicity. We analyzed gene expression of potential targets in single-cell data from cancer and healthy tissues. Because safety and efficacy can ultimately only be defined clinically, we selected approved and investigational targets for which clinical trail data are available. We generated atlases using >300,000 cells from 48 patients with follicular lymphoma, multiple myeloma, and B-cell acute lymphoblastic leukemia, and integrated over 3 million cells from 35 healthy tissues, harmonizing datasets from over 300 donors. To contextualize findings, we compared target expression patterns with outcome data from clinical trials, linking target profiles to efficacy and toxicity, and ranked 15 investigational targets based on their similarity to approved ones. Target expression did not significantly correlate with reported clinical toxicities in patients undergoing therapy. This may be attributed to the intricate interplay of patient-specific variables, the limited amount of metadata, and the complexity underlying toxicity. Nevertheless, our study serves as a resource for retrospective and prospective target evaluation to improve the safety and efficacy of CAR therapies.
Collapse
Affiliation(s)
- Moritz Thomas
- Institute of AI for Health, Computational Health Center, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Ruben Brabenec
- Institute of AI for Health, Computational Health Center, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Lisa Gregor
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - David Andreu-Sanz
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Emanuele Carlini
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Philipp Jie Müller
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Adrian Gottschlich
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany; Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Donjete Simnica
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, a Partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Germany; Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Carsten Marr
- Institute of AI for Health, Computational Health Center, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
3
|
Chen DT, Goloubeva O, Rapoport AP, Dahiya S, Atanackovic D, Hardy N, Kocoglu M, Lutfi F, Alkhaldi H, Claiborne JP, Lee ST, Kline K, Law JY, Yared JA. CD19 CAR-T With Axicabtagene Ciloleucel in R/R Large B-Cell Lymphoma With/Without Prior Autologous Stem Cell Transplant. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:432-439. [PMID: 39865000 DOI: 10.1016/j.clml.2024.12.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/24/2024] [Accepted: 12/28/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND Anti-CD19 CAR-T therapy has been a breakthrough in treatment of primary refractory or relapsed large B-cell lymphoma (r/r LBCL) and is poised to supplant previous second line of high dose chemotherapy and autologous stem cell transplantation (HDT/ASCT). However, in clinical practice, high risk patients with chemoimmunotherapy sensitive disease continue to receive salvage chemoimmunotherapy or cannot access CAR-T in a timely manner and thus may still proceed to HDT/ASCT. Little is known about clinical outcomes of CAR-T in patients who receive HDT/ASCT compared to those who are transplant-naïve. DESIGN We conducted a retrospective study of patients with r/r LBCL who previously underwent HDT/ASCT or were transplant-naïve (n = 97) and received axicabtagene ciloleucel after at least 2 prior therapy lines between 1/1/2018 to 12/31/2021. Primary endpoint was progression-free survival (PFS). Secondary endpoints were overall survival (OS), nonrelapse mortality (NRM), and cumulative incidence of relapse/progression. RESULTS 82 (84.5%) patients were transplant-naïve and 15 (15.5%) previously received HDT/ASCT. No differences were found in the incidence of high-grade cytokine release syndrome or immune effector cell-associated neurotoxicity syndrome, length of hospital admission, or incidence of cytopenia at day 30. 90-day response, PFS, OS, cumulative incidence of relapse/progression, and NRM were not different. Factors that adversely affected outcomes were prior bridging therapy, elevated LDH or thrombocytopenia at time of lymphodepleting chemotherapy, and worse ECOG performance status. CONCLUSION Prior treatment with HDT/ASCT does not compromise the safety and efficacy of anti-CD19 CAR-T therapy, suggesting a continued role for HDT/ASCT in treatment of select patients with r/r DLBCL.
Collapse
MESH Headings
- Humans
- Male
- Female
- Middle Aged
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Immunotherapy, Adoptive/methods
- Retrospective Studies
- Antigens, CD19/immunology
- Antigens, CD19/therapeutic use
- Transplantation, Autologous/methods
- Aged
- Adult
- Hematopoietic Stem Cell Transplantation/methods
- Biological Products/therapeutic use
- Biological Products/pharmacology
Collapse
Affiliation(s)
- David T Chen
- Division of Hematology/Oncology, Department of Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD.
| | - Olga Goloubeva
- Department of Epidemiology and Public Health, Division of Biostatistics and Bioinformatics, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD
| | - Aaron P Rapoport
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Saurabh Dahiya
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Djordje Atanackovic
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Nancy Hardy
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Mehmet Kocoglu
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Forat Lutfi
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Hanan Alkhaldi
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - John Preston Claiborne
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Seung Tae Lee
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Kathryn Kline
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Jennie Y Law
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| | - Jean A Yared
- Transplantation & Cellular Therapy Program, Division of Hematology/Oncology, Department of Internal Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
4
|
Bröckelmann PJ, Cliff ERS, Iacoboni G, Simon F, Horowitz MM, Keating A, Mateos MV, Mohty M, Sidana S, Song Y, Wingard JR, Thanarajasingam G. Beyond maximum grade: tolerability of immunotherapies, cellular therapies, and targeted agents in haematological malignancies. Lancet Haematol 2025; 12:e470-e481. [PMID: 40447355 DOI: 10.1016/s2352-3026(25)00051-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/20/2024] [Accepted: 02/15/2025] [Indexed: 06/18/2025]
Abstract
The increasing use of immunotherapeutic approaches, cellular therapies, and targeted agents is rapidly and profoundly changing the treatment paradigms of haematological malignancies. These novel therapies are increasingly incorporated into earlier lines of treatment. Some are administered for a fixed duration, often with curative intent, whereas others are administered chronically for disease control. The associated acute, mid-term, and long-term toxic effects can differ markedly from conventional cytotoxic chemotherapy and radiotherapy. Accumulating clinical experience and data enable identification of class-specific effects and development of consensus-based guidelines for toxicity management. In this third paper in the Series on adverse event reporting, we build on our emerging understanding of toxicity profiles of novel treatments to propose an actionable framework for improved assessment, reporting, and critical appraisal of treatment tolerability. We discuss recent insights regarding second cancers and the relevance of infectious complications, explore tolerability aspects of time-limited treatments, and suggest approaches to address gaps in tolerability assessment.
Collapse
Affiliation(s)
- Paul J Bröckelmann
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany; German Hodgkin Study Group, Cologne, Germany; Max Planck Institute for Biology of Ageing, Cologne, Germany.
| | - Edward R Scheffer Cliff
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Gloria Iacoboni
- Department of Hematology, University Hospital Vall d'Hebron, Barcelona, Spain; Experimental Hematology, Vall d'Hebron Institute of Oncology, Barcelona, Spain; Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Florian Simon
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany; German CLL Study Group, Cologne, Germany
| | - Mary M Horowitz
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Armand Keating
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Maria-Victoria Mateos
- Hematology Department, Hospital Universitario de Salamanca-IBSAL, CIBERONC and Centro de Investigación del Cáncer, IBMCC, Salamanca, Spain
| | - Mohamad Mohty
- Department of Haematology, Saint Antoine Hospital, INSERM UMR 938, Sorbonne University, Paris, France
| | - Surbhi Sidana
- Stanford University School of Medicine, Stanford, CA, USA
| | - Yuqin Song
- Key Laboratory of Carcinogenesis and Translational Research, Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - John R Wingard
- Division of Hematology/Oncology, University of Florida College of Medicine, Gainesville, FL, USA
| | | |
Collapse
|
5
|
Gamboa L, Zamat AH, Thiveaud CA, Lee HJ, Kulaksizoglu E, Zha Z, Campbell NS, Chan CS, Fábrega S, Oliver SA, Su FY, Phuengkham H, Vanover D, Peck HE, Sivakumar A, Dahotre SN, Harris AM, Santangelo PJ, Kwong GA. Sensitizing solid tumors to CAR-mediated cytotoxicity by lipid nanoparticle delivery of synthetic antigens. NATURE CANCER 2025:10.1038/s43018-025-00968-5. [PMID: 40379831 DOI: 10.1038/s43018-025-00968-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/03/2025] [Indexed: 05/19/2025]
Abstract
Chimeric antigen receptor (CAR) T cell immunotherapy relies on CAR targeting of tumor-associated antigens; however, heterogenous antigen expression, interpatient variation and off-tumor expression by healthy cells remain barriers. Here we develop synthetic antigens to sensitize solid tumors for recognition and elimination by CAR T cells. Unlike tumor-associated antigens, we design synthetic antigens that are orthogonal to endogenous proteins to eliminate off-tumor targeting and that have a small genetic footprint to facilitate efficient tumor delivery to tumors by lipid nanoparticles. Using a camelid single-domain antibody (VHH) as a synthetic antigen, we show that adoptive transfer of anti-VHH CAR T cells to female mice bearing VHH-expressing tumors reduced tumor burden in multiple syngeneic and xenograft models of cancer, improved survival, induced epitope spread, protected against tumor rechallenge and mitigated antigen escape in heterogenous tumors. Our work supports the in situ delivery of synthetic antigens to treat antigen-low or antigen-negative tumors with CAR T cells.
Collapse
Affiliation(s)
- Lena Gamboa
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Ali H Zamat
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Chloé A Thiveaud
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Hee Jun Lee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Elif Kulaksizoglu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Zizhen Zha
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Noah S Campbell
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Ching Shen Chan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Sydney Fábrega
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - S Abbey Oliver
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Fang-Yi Su
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Hathaichanok Phuengkham
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Daryll Vanover
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Hannah E Peck
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Anirudh Sivakumar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Shreyas N Dahotre
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Adrian M Harris
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Philip J Santangelo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Gabriel A Kwong
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA.
- Parker H. Petit Institute of Bioengineering and Bioscience, Atlanta, GA, USA.
- Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, USA.
- The Georgia Immunoengineering Consortium, Emory University and Georgia Tech, Atlanta, GA, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
6
|
Zhang Z, Zheng J, Liang Y, Wu Q, Ding C, Ma L, Su L. Hematologic and lymphatic disorders associated with chimeric antigen receptor T-cell therapy: a pharmacovigilance analysis of the FDA adverse event reporting system (FAERS) database. BMC Cancer 2025; 25:846. [PMID: 40346502 PMCID: PMC12063233 DOI: 10.1186/s12885-025-14227-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 04/25/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND As the application of Chimeric Antigen Receptor T-cell (CAR-T) therapy in cancer treatment becomes increasingly widespread, associated hematologic and lymphatic system adverse events pose significant challenges to its clinical use. Therefore, we aim to comprehensively investigate and summarize the hematologic and lymphatic system AEs associated with CAR-T therapy. METHODS We extracted CAR-T-related adverse event reports from the FDA Adverse Event Reporting System (FAERS) database for the period from August 2017 to December 2023. Disproportionality analysis using the Reporting Odds Ratio (ROR) and Information Component (IC) was performed to identify CAR-T-associated hematologic and lymphatic system AEs. We employed LASSO regression analysis to identify hematologic and lymphatic system AEs associated with mortality. RESULTS In the FAERS database, we identified 1,600 individual case safety reports of hematologic and lymphatic system AEs related to CAR-T therapy. The median age of patients was 57 years (interquartile range [IQR] 32-67), with fatal outcomes in 15.3% of cases. We identified 25 significant adverse event signals associated with CAR-T therapy. B-cell aplasia (ROR025 = 1054.56, IC025 = 4.74), cytopenia (ROR025 = 17.27, IC025 = 3.81), hypofibrinogenemia (ROR025 = 100.18, IC025 = 2.46), anemia (ROR025 = 1.87, IC025 = 0.59), febrile bone marrow aplasia (ROR025 = 55.32, IC025 = 2.70), and pancytopenia (ROR025 = 7.18, IC025 = 1.42) were the most significant hematologic and lymphatic system AEs for tisa-cel, axi-cel, brexu-cel, liso-cel, ide-cel, and cilta-cel, respectively. Most hematologic and lymphatic system AEs occurred within 10 days post-CAR-T infusion. Hematologic and lymphatic system AEs were associated with a mortality rate of 15.3%. Our analysis revealed 15 hematologic and lymphatic system AEs closely associated with mortality in CAR-T-treated patients, including splenic hemorrhage, disseminated intravascular coagulation, and pancytopenia. CONCLUSIONS Our study found that hematologic and lymphatic system AEs were more closely associated with anti-CD19 CAR-T and CAR-T containing CD28. Splenic hemorrhage, disseminated intravascular coagulation, and pancytopenia were identified as hematologic and lymphatic system AEs that, while less frequently reported clinically, were highly associated with mortality.
Collapse
Affiliation(s)
- Zhenpo Zhang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Jingping Zheng
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Yankun Liang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Qimin Wu
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Chufeng Ding
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Lin Ma
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China.
- Medical Department, Guangdong Hospital of Traditional Chinese Medicine, Guangzhou, Guangdong, China.
| | - Ling Su
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
7
|
Canelo-Vilaseca M, Sabbah M, Di Blasi R, Cristinelli C, Sureda A, Caillat-Zucman S, Thieblemont C. Lymphodepletion chemotherapy in chimeric antigen receptor-engineered T (CAR-T) cell therapy in lymphoma. Bone Marrow Transplant 2025; 60:559-567. [PMID: 40148484 PMCID: PMC12061774 DOI: 10.1038/s41409-025-02539-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/29/2024] [Accepted: 02/17/2025] [Indexed: 03/29/2025]
Abstract
The development of chimeric antigen receptor (CAR) T-cells, engineered from peripheral T-lymphocytes of a patient with lymphoma, in order to specifically target tumor cells, has been a revolution in adoptive cell therapy (ACT). As outlined in this review, ACT was initiated by hematopoietic cell transplantation (HSCT) and re-injection of interleukin-boosted tumor-infiltrating lymphocytes (TIL). The innovative venture of genetically modifying autologous peripheral T-cells to target them to cell-surface tumoral antigens through an antibody-derived structure (i.e. independent of major histocompatibility antigen presentation, physiologically necessary for T-cell activation), and intracytoplasmic T-cell costimulatory peptides, via a novel membrane CAR, has been an outstanding breakthrough. Here, focusing on B-cell hematological malignancies and mostly non-Hodgkin lymphoma, attention is brought to the importance of providing an optimal microenvironment for such therapeutic cells to proliferate and positively develop anti-tumoral cytotoxicity. This, perhaps paradoxically, implies a pre-infusion step of deep lymphopenia and deregulation of immunosuppressive mechanisms enhanced by tumoral cells. Fludarabine and cyclophosphamide appear to be the most efficient lymphodepletive drugs in this context, dosage being of importance, as will be illustrated by a thorough literature review.
Collapse
Affiliation(s)
- Marta Canelo-Vilaseca
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Hémato-oncologie, Paris, France
| | - Mohamad Sabbah
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Hémato-oncologie, Paris, France
- Université Paris Cité, Paris, France
| | - Roberta Di Blasi
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Hémato-oncologie, Paris, France
| | - Caterina Cristinelli
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Hémato-oncologie, Paris, France
| | - Anna Sureda
- Clinical Hematology Department, Institut Català d'Oncologia-L'Hospitalet, IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | - Sophie Caillat-Zucman
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Laboratoire d'Immunologie, Paris, France
| | - Catherine Thieblemont
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Hémato-oncologie, Paris, France.
- Université Paris Cité, Paris, France.
- Inserm U1153, Hôpital Saint Louis, Paris, France.
| |
Collapse
|
8
|
Gambles MT, Kendell I, Li J, Spainhower K, Sborov D, Owen S, Stark A, Bearss D, Yang J, Kopeček J. Two-component T-cell immunotherapy enables antigen pre-targeting to reduce cytokine release without forfeiting efficacy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2025; 67:102825. [PMID: 40316226 DOI: 10.1016/j.nano.2025.102825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/16/2025] [Accepted: 04/22/2025] [Indexed: 05/04/2025]
Abstract
Contemporary T-cell immunotherapies, despite impressive targeting precision, are hindered by aberrant cytokine release and restrictive targeting stoichiometry. We introduce a two-component T-cell immunotherapy targeting B-cell malignancies: Multi-Antigen T-Cell Hybridizers (MATCH). This split antibody technology differs from current therapies by separating cancer cell-targeting components from T cell-engaging components. We demonstrate that this two-component structure facilitates tunable T-cell activation. αCD19 and αCD20 MATCH, administered in two steps, are both compared to the clinical standard bispecific antibody, blinatumomab. In vitro two-dimensional dose analysis and cytokine release data indicate MATCH improves cancer clearance with reduced cytokine release. Cytolytic mechanisms of action are evaluated. αCD20 MATCH anti-cancer efficacy is assayed using a human lymphoma murine model. Decreasing T-cell engager dose 10-fold yields comparable efficacy to non-reduced doses. Ultimately, this split-antibody paradigm may enhance antigen targeting while reducing cytokine release, with such safety and efficacy advantages augmented by the future possibility of multi-antigen targeting with MATCH.
Collapse
Affiliation(s)
- M Tommy Gambles
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA.
| | - Isaac Kendell
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Jiahui Li
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Kyle Spainhower
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Douglas Sborov
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Shawn Owen
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Alex Stark
- U2TAH Therapeutics Accelerator, University of Utah, Salt Lake City, UT 84112, USA
| | - David Bearss
- U2TAH Therapeutics Accelerator, University of Utah, Salt Lake City, UT 84112, USA
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA.
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
9
|
Hirano D, Fujimaru T, Sako M, Tanaka S, Inaba A, Uchimura T, Kamei K, Kubota T, Ohta T, Okamoto T, Tanaka H, Hamada R, Ito S. Recovery from rituximab-associated persistent hypogammaglobulinaemia in children with nephrotic syndrome. Nephrol Dial Transplant 2025; 40:967-977. [PMID: 39419779 DOI: 10.1093/ndt/gfae228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND There are limited data on the long-term outcomes and risk factors for non-recovery after development of rituximab (RTX)-associated persistent hypogammaglobulinaemia among children with idiopathic nephrotic syndrome (NS). METHODS A nationwide Japanese survey was conducted to determine the prognosis of patients with childhood-onset idiopathic NS who developed persistent hypogammaglobulinaemia after RTX administration. Specifically, predictors of IgG level recovery and risk factors for serious infection were examined. RESULTS The cohort comprised 118 patients (66.1% boys; median age at initial RTX administration, 7.5 years). Among the 121 patients diagnosed with persistent hypogammaglobulinaemia, only 31 (26.3%) recovered within a median observation period of 2.8 years; approximately 70% of patients continued to exhibit persistent hypogammaglobulinaemia. Among the patients who recovered from hypogammaglobulinaemia, the median time to recovery was 14.1 months. Patients with a history of steroid-resistant NS were less likely to recover from persistent hypogammaglobulinaemia (hazard ratio 0.28; 95% confidence interval 0.09-0.87). In addition, of the 118 eligible patients, 18 (15.3%) developed serious infections requiring hospitalization, and the main risk factor for infection during hypogammaglobulinaemia was agranulocytosis (a well-known adverse effect of RTX in children). CONCLUSIONS A significant portion of patients with RTX-associated persistent hypogammaglobulinaemia did not exhibit recovery even after 1 year. Moreover, the data indicate that patients with a history of steroid-resistant NS have a significantly lower probability of recovering from this condition. Agranulocytosis under hypogammaglobulinaemia was significantly associated with an elevated risk of serious infections.
Collapse
Affiliation(s)
- Daishi Hirano
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Takuya Fujimaru
- Department of Nephrology, St Luke's International Hospital, Tokyo, Japan
| | - Mayumi Sako
- Department of Clinical Research Promotion, Clinical Research Center, National Center for Child Health and Development, Tokyo, Japan
| | - Seiji Tanaka
- Department of Pediatrics and Child Health, Kurume University Medical Center, Kurume, Japan
| | - Aya Inaba
- Department of Pediatrics, Yokohama City University Medical Center, Yokohama, Japan
| | - Toru Uchimura
- Department of Pediatrics, Yokohama City University Medical Center, Yokohama, Japan
| | - Koichi Kamei
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan
| | - Takuo Kubota
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Toshiyuki Ohta
- Department of Pediatric Nephrology, Hiroshima Prefectural Hospital, Hiroshima, Japan
| | - Takayuki Okamoto
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hiroshi Tanaka
- Department of Pediatrics, Hirosaki University Hospital, Hirosaki, Japan
- Department of School Health Science, Hirosaki University Faculty of Education and Department of Pediatrics, Hirosaki University Hospital, Hirosaki, Japan
| | - Riku Hamada
- Department of Nephrology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Shuichi Ito
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University Medical Center, Yokohama, Japan
| |
Collapse
|
10
|
Lussana F, Magnani CF, Galimberti S, Gritti G, Gaipa G, Belotti D, Cabiati B, Napolitano S, Ferrari S, Moretti A, Buracchi C, Borleri GM, Rambaldi B, Rizzuto G, Grassi A, Paganessi M, Meli C, Tettamanti S, Risca G, Pais G, Spinozzi G, Benedicenti F, Cazzaniga G, Capelli C, Gotti E, Introna M, Golay J, Montini E, Balduzzi A, Valsecchi MG, Dastoli G, Rambaldi A, Biondi A. Donor-derived CARCIK-CD19 cells engineered with Sleeping Beauty transposon in acute lymphoblastic leukemia relapsed after allogeneic transplantation. Blood Cancer J 2025; 15:54. [PMID: 40180925 PMCID: PMC11968829 DOI: 10.1038/s41408-025-01260-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 04/05/2025] Open
Abstract
Non-viral engineering can ease CAR-T cell production and reduce regulatory and cost requirements. We utilized Sleeping Beauty transposon to engineer donor-derived anti-CD19.CD28.OX40.CD3zeta T cells differentiated in cytokine-induced killer (CARCIK-CD19) for B-cell precursor acute lymphoblastic leukemia (BCP-ALL) patients relapsed after allogeneic hematopoietic stem cell transplantation (alloHSCT). We report the results of CARCIK-CD19 observed in 36 patients (4 children and 32 adults) treated according to the final recommended dose. Cytokine release syndrome of grade 2 or lower occurred in 15 patients, ICANS grade 2 in 1 patient, and late-onset peripheral neurotoxicity of grade 3 in 2 patients. GVHD never occurred after treatment with allogeneic CARCIK-CD19. Complete remission was achieved by 30 out of 36 patients (83.3%), with MRD negativity in 89% of responders. With a median follow-up of 2.2 years, the 1-year overall survival was 57.0%, and event-free survival was 32.0%. The median duration of response at 1 year was 38.6%. CAR-T cells expanded rapidly after infusion and remained detectable for over 2 years. Integration site analysis after infusion showed a high clonal diversity. These data demonstrated that SB-engineered CAR-T cells are safe and induce durable remission in heavily pretreated patients with BCP-ALL relapsed after alloHSCT. Trial registration: The phase 1/2 and phase II trials are registered at www.clinicaltrials.gov as NCT#03389035 and NCT#05252403.
Collapse
Affiliation(s)
- Federico Lussana
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - Chiara F Magnani
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Stefania Galimberti
- Department of Medicine and Surgery, Bicocca Bioinformatics, Biostatistics and Bioimaging Centre B4, University of Milano-Bicocca, Milan, Italy
- Biostatistics and Clinical Epidemiology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Giuseppe Gritti
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Giuseppe Gaipa
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Laboratorio di Terapia Cellulare e Genica Stefano Verri, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Daniela Belotti
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Laboratorio di Terapia Cellulare e Genica Stefano Verri, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Benedetta Cabiati
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Laboratorio di Terapia Cellulare e Genica Stefano Verri, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Sara Napolitano
- Department of Pediatrics, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Silvia Ferrari
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Alex Moretti
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Pediatrics, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Chiara Buracchi
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Gian Maria Borleri
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Benedetta Rambaldi
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Giuliana Rizzuto
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - Anna Grassi
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Muriel Paganessi
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Cristian Meli
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Sarah Tettamanti
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Giulia Risca
- Biostatistics and Clinical Epidemiology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Giulia Pais
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulio Spinozzi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabrizio Benedicenti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giovanni Cazzaniga
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Chiara Capelli
- USS Centro di Terapia Cellulare "G. Lanzani", Bergamo, Italy
| | - Elisa Gotti
- USS Centro di Terapia Cellulare "G. Lanzani", Bergamo, Italy
| | - Martino Introna
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
- USS Centro di Terapia Cellulare "G. Lanzani", Bergamo, Italy
| | - Josée Golay
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
- USS Centro di Terapia Cellulare "G. Lanzani", Bergamo, Italy
| | - Eugenio Montini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Adriana Balduzzi
- Department of Pediatrics, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Maria Grazia Valsecchi
- Department of Medicine and Surgery, Bicocca Bioinformatics, Biostatistics and Bioimaging Centre B4, University of Milano-Bicocca, Milan, Italy
- Biostatistics and Clinical Epidemiology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Giuseppe Dastoli
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Alessandro Rambaldi
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - Andrea Biondi
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy.
- Department of Pediatrics, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy.
| |
Collapse
|
11
|
Mousavi S, Khazaee-Nasirabadi MH, Seyedmehdi MS, Bazi A, Mirzaee Khalilabadi R. Natural killer cells: a new promising source for developing chimeric antigen receptor anti-cancer cells in hematological malignancies. Leuk Lymphoma 2025; 66:594-616. [PMID: 39656564 DOI: 10.1080/10428194.2024.2438802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 11/18/2024] [Accepted: 12/01/2024] [Indexed: 12/17/2024]
Abstract
In recent times, the application of CAR-T cell treatment has significantly progressed, showing auspicious treatment outcomes in hematologic malignancies. However, along with these advances, certain limitations and challenges hurdle the widespread utilization of this technology. Recently, CAR-NK cells have gained attention in cancer treatment, as this approach has an important advantage over CART therapy (i.e. no need for HLA matching) for targeting foreign cells. This review aims to explore the benefits of CAR NK cell therapy, and generation strategies, as well as the challenges and limitations hindering the application of CAR NK cells in experimental studies and trials on hematologic malignancies.
Collapse
Affiliation(s)
- Shahrzad Mousavi
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Maryam Sadat Seyedmehdi
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Technology, Shahid Beheshti University, Tehran, Islamic Republic of Iran
| | - Ali Bazi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Faculty of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Roohollah Mirzaee Khalilabadi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
12
|
Waddell D, Collins J, Sadrameli S. Utility of Thrombopoietin Receptor Agonists for Prolonged Thrombocytopenia After Chimeric Antigen Receptor T-cell Therapy. Transplant Cell Ther 2025; 31:238.e1-238.e12. [PMID: 39880098 DOI: 10.1016/j.jtct.2025.01.887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 01/15/2025] [Accepted: 01/19/2025] [Indexed: 01/31/2025]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has revolutionized the treatment landscape for various hematological malignancies. However, it is associated with a range of hematologic complications, including severe and often prolonged thrombocytopenia. Currently, there are no known effective preventative or management measures against CAR-T-induced thrombocytopenia. At the University of Chicago Medical Center, thrombopoietin receptor agonists (TPO-RAs) eltrombopag and romiplostim have been utilized intermittently, per attending preference, in patients post CAR-T treatment presenting with prolonged thrombocytopenia (platelets <50 × 103 cells/μL for at least 14 days). However, whether these treatments yield positive outcomes in this context remains uncertain. This study aims to evaluate the efficacy and safety of TPO-RAs in patients with CAR-T-induced thrombocytopenia. The primary objective is to compare the incidence of platelet recovery (defined as two consecutive platelet counts of ≥50 × 103 cells/μL) in patients who received TPO-RAs versus those who did not for CAR-T-associated prolonged thrombocytopenia between January 1, 2018, and June 30, 2023. The secondary objectives include time to platelet recovery, incidence of clinically relevant bleed, hospital length of stay, incidence of adverse effects associated with TPO-RA administration, overall survival, and financial toxicity. This is a single-center, retrospective study conducted at the University of Chicago Medical Center. Eighty-five patients with prolonged, CAR-T-induced thrombocytopenia were enrolled in the study; 12 of these patients were managed with TPO-RA therapy while the remaining 73 received supportive care. Statistical analysis was performed using STATA, incorporating the Chi-squared test for nominal data and the Wilcoxon Rank-sum test for continuous data. A P value of <.05 was used to determine statistical significance. The incidence of platelet recovery was similar between the two groups; in the supportive care group, 53 patients (73%) experienced resolution of thrombocytopenia, compared to 9 patients (75%) in the TPO-RA treated group (P = 1.0). The median time to thrombocytopenia resolution was 56 days in the TPO-RA-treated group and 41 days in those not managed with TPO-RAs (P = .14). The median time to TPO-RA initiation postinfusion was 45 days. There were no statistically significant differences in incidence of clinically relevant bleed or readmission within 1 year of CAR-T infusion between the two groups, but 25% of patients receiving TPO-RA therapy experienced associated arthralgia requiring treatment modification. Additionally, the median cost of a course of eltrombopag was estimated at $86,921.52 per patient at the reported average wholesale price. While TPO-RAs represent a theoretical therapeutic option for CAR-T patients based on their role in chemotherapy-induced thrombocytopenia, our study showed that their use did not provide significant clinical benefit compared to the supportive care approaches. Therefore, without larger, randomized, prospective trials, we are unable to recommend TPO-RA use in this setting, given the current lack of demonstrated efficacy, potential adverse effects, and concerns regarding financial impact.
Collapse
Affiliation(s)
- Donald Waddell
- The University of Chicago Medical Center, Chicago, Illinois.
| | | | | |
Collapse
|
13
|
Guo S, Liu J, Wang B, Zhang X, Zhao Y, Xu J, Cao X, Zhao M, Xiao X, Zhao M. A viral infection prediction model for patients with r/r B-cell malignancies after CAR-T therapy: a retrospective analysis. Front Oncol 2025; 15:1549809. [PMID: 40190552 PMCID: PMC11968754 DOI: 10.3389/fonc.2025.1549809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
Background Chimeric antigen receptor T cell (CAR-T) therapy for relapsed/refractory (r/r) B cell acute lymphoblastic leukemia (B-ALL) and B cell non-Hodgkin lymphoma (B-NHL) patients has shown promising effects, but side effects such as viral infections have been observed. Methods A total of 45 patients with r/r B-ALL and r/r B-NHL were included in this retrospective study. Patient demographics were recorded, with the primary endpoint being viral infection within 3 months post CAR-T treatment. Univariate and multivariate logistic regression analyses and least absolute shrinkage and selection operator (LASSO) regression analysis were used to analyze independent factors. The patients were divided into a training cohort of 28 and a validation cohort of 17 to construct a prediction model based on determined independent factors. The model's discrimination and calibration were assessed using the receiver operating characteristic curve (ROC), calibration plot, and decision curve analysis (DCA curve). Results The univariate and multivariate logistic regression analyses of the 43 patients showed that low baseline lymphocyte ratio was an independent risk factor and using granulocyte colony-stimulating factor (G-CSF) early was a protective factor for viral infection after CAR-T therapy in patients with B-ALL and B-NHL. Based on that, the area under the ROC curve (AUC) of the training cohort and validation cohort was 0.935 (95% CI 0.837-1.000) and 0.869 (95%CI 0.696-1.000), respectively, showing excellent predictive value. Conclusions We established a nomogram to predict the factors' influence on viral infection after CAR-T therapy and found that the ratio of baseline lymphocytes and using G-CSF early or lately were able to predict viral infection after CAR-T therapy in r/r B-ALL and B-NHL.
Collapse
Affiliation(s)
- Shujing Guo
- First Center Clinical College, Tianjin Medical University, Tianjin, China
| | - Jile Liu
- First Center Clinical College, Tianjin Medical University, Tianjin, China
| | - Bing Wang
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Xiaomei Zhang
- School of Medicine, Nankai University, Tianjin, China
| | - Yifan Zhao
- First Center Clinical College, Tianjin Medical University, Tianjin, China
| | - Jianmei Xu
- Department of Hematology, Hebei University Affiliated Hospital, Baoding, Hebei, China
| | - Xinping Cao
- First Center Clinical College, Tianjin Medical University, Tianjin, China
| | - Mohan Zhao
- First Center Clinical College, Tianjin Medical University, Tianjin, China
| | - Xia Xiao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
14
|
Dickinson M, O'Leary N, Hamad N, Cochrane T, Ho PJ, Cheah CY, Bishop D, Gregory GP, Butler J, Barraclough A. Establishing best practice in the Australian haematology setting for the use of chimeric antigen receptor T-cell therapy for relapsed and refractory lymphoma. Intern Med J 2025; 55 Suppl 2:4-27. [PMID: 40171767 DOI: 10.1111/imj.16544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 09/24/2024] [Indexed: 04/04/2025]
Abstract
Autologous CD19 chimeric antigen receptor T (CAR T)-cell therapies have significantly improved clinical outcomes for many patients with relapsed and refractory (R/R) lymphoma. However, the process of delivering CAR T-cell therapy is specialised and complex, in part due to specific post-infusion toxicities. Several CAR T-cell products are now available in Australia, although not all states have treatment centres. In this review, we aim to define best practice for the referral and treatment of patients with R/R B-cell lymphoma with CAR T-cell therapy in Australia. We outline the processes for referral, optimal patient selection and best practice in the management of patients receiving CAR T cells.
Collapse
Affiliation(s)
- Michael Dickinson
- Peter MacCallum Cancer Centre, Royal Melbourne Hospital and the Sir Peter MacCallum Department of Oncology at the University of Melbourne, Sydney, New South Wales, Australia
| | - Nicole O'Leary
- Peter MacCallum Cancer Centre, Royal Melbourne Hospital and the Sir Peter MacCallum Department of Oncology at the University of Melbourne, Sydney, New South Wales, Australia
| | - Nada Hamad
- Department of Haematology, St Vincent's Hospital, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of NSW, Sydney, New South Wales, Australia
- School of Medicine, University of Notre Dame, Sydney, New South Wales, Australia
| | - Tara Cochrane
- Department of Haematology, Gold Coast University Hospital, Southport, Queensland, Australia
- School of Medicine and Dentistry, Griffith University, Southport, Queensland, Australia
| | - P Joy Ho
- Department of Haematology, Royal Prince Alfred Hospital and University of Sydney, Sydney, New South Wales, Australia
| | - Chan Y Cheah
- Department of Haematology, Sir Charles Gairdner Hospital, Nedlands, and University of Western Australia, Perth, Western Australia, Australia
| | - David Bishop
- Department of Haematology, Westmead Hospital, Sydney, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Gareth P Gregory
- School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Queensland, Australia
| | - Jason Butler
- Department of Bone Marrow Transplantation, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Allison Barraclough
- Department of Haematology, Fiona Stanley Hospital, Perth, Western Australia, Australia
| |
Collapse
|
15
|
Umair MM, Lai X, Xue Y, Yao H. Influence of CAR T-cell therapy associated complications. Front Oncol 2025; 15:1494986. [PMID: 40052127 PMCID: PMC11882432 DOI: 10.3389/fonc.2025.1494986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/21/2025] [Indexed: 03/09/2025] Open
Abstract
Since the introduction of chimeric antigen receptor (CAR) T-cell therapy, it has elicited an immense response in both targeted and residual cancers. Its clinical efficacy is often accompanied by a group of side effects that may become serious because of factors such as tumor burden, the extent of lymphodepletion, and the type of co-stimulus. It is also crucial to know the common toxicities associated with CAR T-cell therapy, including cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), cardiotoxicity, metabolic disorders, pulmonary toxicity, macrophage activation syndrome (MAS), prolonged cytopenia, coagulation disorders, and potential off-target effects on various organs. If not well managed, these can be fatal. However, knowledge about molecular pathways, calcineurin inhibitors, IL-6 receptor antagonists, steroids, suppression of nitric oxide synthase, various therapeutic approaches, and other recent advances have been developed to mitigate the fatal results of various short-term and chronic adverse events related to CAR T-cell therapy. This study provides a comprehensive perspective on contemporary management strategies and presumed causative processes of CAR T-cell-related adverse effects, albeit with several limitations. When CAR T-cell complications, costs, and challenges of toxicity management are properly considered, the CAR T-cell therapy of the future will include a number of toxicity-escaping options.
Collapse
Affiliation(s)
- Mohammad Mussab Umair
- Cancer Biotherapy Center & Cancer Research Institute, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xun Lai
- Department of Hematology, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - YuanBo Xue
- Cancer Biotherapy Center, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hong Yao
- Cancer Biotherapy Center & Cancer Research Institute, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
16
|
Ward AC. Secondary Neutropenias. Biomedicines 2025; 13:497. [PMID: 40002910 PMCID: PMC11853056 DOI: 10.3390/biomedicines13020497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/07/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025] Open
Abstract
Neutrophils are a critical component of immunity, particularly against bacteria and other pathogens, but also in inflammation and tissue repair. As a consequence, individuals with neutropenia, defined by a reduction in absolute neutrophil counts, exhibit a strong propensity to severe infections that typically present with muted symptoms. Neutropenias encompass a heterogeneous set of disorders, comprising primary neutropenias, in which specific genes are mutated, and the more common secondary neutropenias, which have diverse non-genetic causes. These include hematological and other cancers, involving both direct effects of the cancer itself and indirect impacts via the chemotherapeutic, biological agents and cell-based approaches used for treatment. Other significant causes of secondary neutropenias are non-chemotherapeutic drugs, autoimmune and other immune diseases, infections and nutrient deficiencies. These collectively act by impacting neutrophil production in the bone marrow and/or destruction throughout the body. This review describes the biological and clinical manifestations of secondary neutropenias, detailing their underlying causes and management, with a discussion of alternative and emerging therapeutic approaches.
Collapse
Affiliation(s)
- Alister C. Ward
- School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia;
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Waurn Ponds, VIC 3216, Australia
| |
Collapse
|
17
|
Watanabe K, Shimada N, Kanzaki M, Fukuoka K, Asano K. Late-onset Neutropenia after Rituximab Treatment for MPO-ANCA-associated Vasculitis. Intern Med 2025; 64:581-584. [PMID: 39019601 PMCID: PMC11904456 DOI: 10.2169/internalmedicine.3357-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 06/02/2024] [Indexed: 07/19/2024] Open
Abstract
An underestimated side effect of rituximab is late-onset neutropenia (R-LON), which often resolves spontaneously and rarely results in a severe infection. We herein report a case of febrile neutropenia due to R-LON in a 91-year-old woman with renal failure who was treated with rituximab to induce remission of myeloperoxidase antineutrophil cytoplasmic antibodies-associated vasculitis. Fifty-four days after the last rituximab administration, the patient was hospitalized for febrile neutropenia due to R-LON, which improved with granulocyte colony-stimulating factor and antibiotics. Although R-LON may resolve spontaneously and remain unnoticed, it can cause severe infections in the elderly and patients with renal failure.
Collapse
Affiliation(s)
| | | | - Motoko Kanzaki
- Department of Nephrology, Kurashiki Central Hospital, Japan
| | - Kosuke Fukuoka
- Department of Nephrology, Kurashiki Central Hospital, Japan
| | | |
Collapse
|
18
|
Liang EC, Huang JJ, Portuguese AJ, Ortiz-Maldonado V, Albittar A, Wuliji N, Basom R, Jeon Y, Wu Q, Torkelson A, Kirchmeier D, Chutnik A, Pender B, Sorror M, Hill JA, Kopmar NE, Banerjee R, Cowan AJ, Green D, Gopal AK, Poh C, Shadman M, Hirayama AV, Till BG, Kimble EL, Iovino L, Chapuis AG, Otegbeye F, Cassaday RD, Milano F, Turtle CJ, Maloney DG, Gauthier J. Development and validation of predictive models of early immune effector cell-associated hematotoxicity. Blood Adv 2025; 9:606-616. [PMID: 39626349 PMCID: PMC11847049 DOI: 10.1182/bloodadvances.2024014455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/28/2024] [Accepted: 11/10/2024] [Indexed: 02/03/2025] Open
Abstract
ABSTRACT Immune effector cell-associated hematotoxicity (ICAHT) is associated with morbidity and mortality after chimeric antigen receptor (CAR) T-cell therapy. To date, the factors associated with ICAHT are poorly characterized, and there is no validated predictive model of ICAHT as defined by current consensus criteria. Therefore, we performed comprehensive univariate analyses to identify factors associated with severe (grade 3-4) early ICAHT (eICAHT) in 691 patients who received commercial or investigational CAR T-cell therapy for hematologic malignancies. In univariate logistic regression, preinfusion factors associated with severe eICAHT included disease type (acute lymphoblastic leukemia), prelymphodepletion (pre-LD) blood counts including absolute neutrophil count (ANC), lactate dehydrogenase (LDH), and inflammatory (C-reactive protein [CRP], ferritin, and interleukin-6 [IL-6]) and coagulopathy biomarkers (D-dimer). Postinfusion laboratory markers associated with severe eICAHT included early and peak levels of inflammatory biomarkers (CRP, ferritin, and IL-6), coagulopathy biomarkers (D-dimer), peak cytokine release syndrome grade, and peak neurotoxicity grade. We trained (n = 483) and validated (n = 208) 2 eICAHT prediction models (eIPMs): eIPMPre including preinfusion factors only (disease type and pre-LD ANC, platelet count, LDH, and ferritin) and eIPMPost containing both preinfusion (disease type and pre-LD ANC, platelet count, and LDH) and early postinfusion (day +3 ferritin) factors. Both models generated calibrated predictions and high discrimination (area under the receiver operating characteristic curve in test set, 0.87 for eIPMPre and 0.88 for eIPMPost), with higher net benefit in decision curve analysis for eIPMPost. Individualized predictions of severe eICAHT can be generated from both eIPMs using our online tool (available at https://eipm.fredhutch.org).
Collapse
Affiliation(s)
- Emily C. Liang
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Jennifer J. Huang
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Andrew J. Portuguese
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Valentín Ortiz-Maldonado
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Hematology, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Aya Albittar
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Natalie Wuliji
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Ryan Basom
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Yein Jeon
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Qian Wu
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Aiko Torkelson
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Abigail Chutnik
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Barbara Pender
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Mohamed Sorror
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Joshua A. Hill
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Noam E. Kopmar
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Rahul Banerjee
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Andrew J. Cowan
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Damian Green
- Division of Transplantation and Cellular Therapy, Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Ajay K. Gopal
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Christina Poh
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Mazyar Shadman
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Alexandre V. Hirayama
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Brian G. Till
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Erik L. Kimble
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Lorenzo Iovino
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Aude G. Chapuis
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Folashade Otegbeye
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Ryan D. Cassaday
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Filippo Milano
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Cameron J. Turtle
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - David G. Maloney
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Jordan Gauthier
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
19
|
Evangelidis P, Tragiannidis K, Vyzantiadis A, Evangelidis N, Kalmoukos P, Vyzantiadis TA, Tragiannidis A, Kourti M, Gavriilaki E. Invasive Fungal Disease After Chimeric Antigen Receptor-T Immunotherapy in Adult and Pediatric Patients. Pathogens 2025; 14:170. [PMID: 40005545 PMCID: PMC11858289 DOI: 10.3390/pathogens14020170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 01/25/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Invasive fungal diseases (IFDs) have been documented among the causes of post-chimeric antigen receptor-T (CAR-T) cell immunotherapy complications, with the incidence of IFDs in CAR-T cell therapy recipients being measured between 0% and 10%, globally. IFDs are notorious for their potentially life-threatening nature and challenging diagnosis and treatment. In this review, we searched the recent literature aiming to examine the risk factors and epidemiology of IFDs post-CAR-T infusion. Moreover, the role of antifungal prophylaxis is investigated. CAR-T cell therapy recipients are especially vulnerable to IFDs due to several risk factors that contribute to the patient's immunosuppression. Those include the underlying hematological malignancies, the lymphodepleting chemotherapy administered before the treatment, existing leukopenia and hypogammaglobinemia, and the use of high-dose corticosteroids and interleukin-6 blockers as countermeasures for immune effector cell-associated neurotoxicity syndrome and cytokine release syndrome, respectively. IFDs mostly occur within the first 60 days following the infusion of the T cells, but cases even a year after the infusion have been described. Aspergillus spp., Candida spp., and Pneumocystis jirovecii are the main cause of these infections following CAR-T cell therapy. More real-world data regarding the epidemiology of IFDs and the role of antifungal prophylaxis in this population are essential.
Collapse
Affiliation(s)
- Paschalis Evangelidis
- 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (P.E.); (N.E.); (P.K.)
| | - Konstantinos Tragiannidis
- Children & Adolescent Hematology-Oncology Unit, Second Department of Pediatrics, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.T.); (A.T.); (M.K.)
| | - Athanasios Vyzantiadis
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.V.); (T.-A.V.)
| | - Nikolaos Evangelidis
- 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (P.E.); (N.E.); (P.K.)
| | - Panagiotis Kalmoukos
- 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (P.E.); (N.E.); (P.K.)
| | - Timoleon-Achilleas Vyzantiadis
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.V.); (T.-A.V.)
| | - Athanasios Tragiannidis
- Children & Adolescent Hematology-Oncology Unit, Second Department of Pediatrics, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.T.); (A.T.); (M.K.)
| | - Maria Kourti
- Children & Adolescent Hematology-Oncology Unit, Second Department of Pediatrics, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.T.); (A.T.); (M.K.)
| | - Eleni Gavriilaki
- 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (P.E.); (N.E.); (P.K.)
- Hematology Department and Bone Marrow Transplant (BMT) Unit, G. Papanicolaou Hospital, 57010 Thessaloniki, Greece
| |
Collapse
|
20
|
Molinos-Quintana Á, Martínez-Cibrian N, Alonso-Saladrigues A, Galán-Gómez V, Bailén R, Buendía-López S, Fuentes-Socorro C, Kwon M, González-Vincent M, Pérez de Soto C, González-Martínez B, Rives S, Pérez-Hurtado JM, Ortiz-Maldonado V, Pérez-Simón JA. Successful allogeneic CD34 + hematopoietic stem cell boost for prolonged cytopenias following CAR T-cell therapy in B-cell acute lymphoblastic leukemia. On behalf of the Spanish Group for Hematopoietic Transplantation and Cellular Therapy (GETH-TC). Bone Marrow Transplant 2025; 60:250-253. [PMID: 39551836 DOI: 10.1038/s41409-024-02473-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024]
Affiliation(s)
- Águeda Molinos-Quintana
- Pediatric Unit, Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS) / CSIC, Universidad de Sevilla, Seville, Spain.
| | | | - Anna Alonso-Saladrigues
- CAR T-cell Unit, Leukemia and Lymphoma Department, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Medicine and Surgery School, University of Barcelona, Barcelona, Spain
| | - Víctor Galán-Gómez
- Pediatric Hemato-Oncology Department, University Hospital La Paz, Institute for Health Research (IdiPAZ), Universidad Autónoma de Madrid, Madrid, Spain
| | - Rebeca Bailén
- Haematology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Susana Buendía-López
- Pediatric Hemato-Oncology Department, Peditric University Hospital del Niño Jesús, Madrid, Spain
| | | | - Mi Kwon
- Haematology Department, Hospital General Universitario Gregorio Marañón. Instituto de Investigación Sanitaria Gregorio Marañón. Universidad Complutense de Madrid, Madrid, Spain
| | - Marta González-Vincent
- Department of Stem Cell Transplantation, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Concepción Pérez de Soto
- Pediatric Unit, Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS) / CSIC, Universidad de Sevilla, Seville, Spain
| | - Berta González-Martínez
- Pediatric Hemato-Oncology Department, University Hospital La Paz, Institute for Health Research (IdiPAZ), Universidad Autónoma de Madrid, Madrid, Spain
| | - Susana Rives
- Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Sant Joan de Déu, Leukemia and Pediatric Hematology Disorders, Developmental Tumors Biology Group, Barcelona, Spain
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red De Enfermedades Raras (CIBERER), Madrid, Spain
| | - José María Pérez-Hurtado
- Pediatric Unit, Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS) / CSIC, Universidad de Sevilla, Seville, Spain
| | | | - José Antonio Pérez-Simón
- Department of Hematology, University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS)/CSIC, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
21
|
van den Berg J, Läubli H, Khanna N, Jeker LT, Holbro A. Basic Concepts and Indications of CAR T Cells. Hamostaseologie 2025; 45:14-23. [PMID: 39970899 DOI: 10.1055/a-2491-3652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has revolutionized cancer immunotherapy, particularly for hematological malignancies. This personalized approach is based on genetically engineering T cells derived from the patient to target antigens expressed-among others-on malignant cells. Nowadays they offer new hope where conventional therapies, such as chemotherapy and radiation, have often failed. Since the first FDA approval in 2017, CAR T cell therapy has rapidly expanded, proving highly effective against previously refractory diseases with otherwise a dismal outcome. Despite its promise, CAR T cell therapy continues to face significant challenges, including complex manufacturing, the management of toxicities, resistance mechanisms that impact long-term efficacy, and limited access as well as high costs, which continue to shape ongoing research and clinical applications. This review aims to provide an overview of CAR T cell therapy, including its fundamental concepts, clinical applications, current challenges, and future directions in hematological malignancies.
Collapse
Affiliation(s)
- Jana van den Berg
- Division of Hematology, University Hospital Basel, Basel, Switzerland
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
| | - Heinz Läubli
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Nina Khanna
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| | - Lukas T Jeker
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | - Andreas Holbro
- Division of Hematology, University Hospital Basel, Basel, Switzerland
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Regional Blood Transfusion Service, Swiss Red Cross, Basel, Switzerland
| |
Collapse
|
22
|
Alfaro-Quinde C, Krstanovic KE, Vásquez PA, Kathrein KL. STOCHASTIC MODELING OF HEMATOPOIETIC STEM CELL DYNAMICS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635091. [PMID: 39974985 PMCID: PMC11838373 DOI: 10.1101/2025.01.27.635091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The study of hematopoietic stem cell (HSCs) maintenance and differentiation to supply the hematopoietic system presents unique challenges, given the complex regulation of the process and the difficulty in observing cellular interactions in the stem cell niche. Quantitative methods and tools have emerged as valuable mechanisms to address this issue; however, the stochasticity of HSCs presents significant challenges for mathematical modeling, especially when bridging the gap between theoretical models and experimental validation. In this work, we have built a flexible and user-friendly stochastic dynamical and spatial model for long-term HSCs (LT-HSCs) and short-term HSCs (ST-HSCs) that captures experimentally observed cellular variability and heterogeneity. Our model implements the behavior of LT-HSCs and ST-HSCs and predicts their homeostatic dynamics. Furthermore, our model can be modified to explore various biological scenarios, such as stress-induced perturbations mediated by apoptosis, and successfully implement these conditions. Finally, the model incorporates spatial dynamics, simulating cell behavior in a 2D environment by combining Brownian motion with spatially graded parameters.
Collapse
Affiliation(s)
| | | | - Paula A. Vásquez
- Department of Mathematics, University of South Carolina, Columbia, SC
| | - Katie L. Kathrein
- Department of Biological Sciences, University of South Carolina, Columbia, SC
| |
Collapse
|
23
|
Zandaki D, Selukar S, Bi Y, Li Y, Zinsky M, Bonifant CL, Epperly R, Keerthi D, Triplett BM, Gottschalk S, Naik S, Talleur AC. EASIX and m-EASIX predict CRS and ICANS in pediatric and AYA patients after CD19-CAR T-cell therapy. Blood Adv 2025; 9:270-279. [PMID: 39325974 PMCID: PMC11782822 DOI: 10.1182/bloodadvances.2024014027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/30/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
ABSTRACT Cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) are complications of CD19-directed chimeric antigen receptor (CD19-CAR) T-cell therapy. The Endothelial Activation and Stress Index (EASIX) and modified EASIX (m-EASIX) scores have been retrospectively proven to be predictive of CRS and ICANS in adult CAR T-cell recipients. However, these scores have not been evaluated in pediatric cohorts. We retrospectively report on 76 pediatric and adolescent and young adult (AYA) patients with relapsed/refractory B-cell acute lymphoblastic leukemia treated with CD19-CAR T cells at St. Jude Children's Research Hospital or Johns Hopkins Hospital. Data included patient, disease, and treatment characteristics. EASIX and m-EASIX scores were calculated at days -5 before, 0, and +3 after CAR T-cell infusion. CRS and ICANS occurred in 47 and 17 patients, respectively. At all evaluated time points, the median EASIX scores were higher for patients who developed severe CRS and any grade ICANS, and the median m-EASIX scores were higher in patients who developed severe CRS and severe ICANS than those with no/mild CRS/ICANS. Receiver operating characteristic curve analysis showed that both scores were strong predictors of CRS, especially severe CRS, at all time points. Any grade and severe ICANS were best predicted by both scores at day +3. m-EASIX uniformly outperformed EASIX, except for predicting any grade ICANS. Our results validate the potential utility of EASIX and m-EASIX scores for predicting CAR T-cell-related complications for pediatric and AYA patients.
Collapse
Affiliation(s)
- Dua’a Zandaki
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| | - Subodh Selukar
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Yu Bi
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Ying Li
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| | - Megan Zinsky
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Challice L. Bonifant
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Rebecca Epperly
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| | - Dinesh Keerthi
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| | - Brandon M. Triplett
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| | - Swati Naik
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| | - Aimee C. Talleur
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
24
|
Bangolo A, Amoozgar B, Mansour C, Zhang L, Gill S, Ip A, Cho C. Comprehensive Review of Early and Late Toxicities in CAR T-Cell Therapy and Bispecific Antibody Treatments for Hematologic Malignancies. Cancers (Basel) 2025; 17:282. [DOI: 1.bangolo a, amoozgar b, mansour c, zhang l, gill s, ip a, cho c.comprehensive review of early and late toxicities in car t-cell therapy and bispecific antibody treatments for hematologic malignancies.cancers (basel).2025 jan 17;17(2):282.doi: 10.3390/cancers17020282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2025] Open
Abstract
Chimeric antigen receptor T-cell (or CAR-T) therapy and bispecific antibodies (BsAbs) have revolutionized the treatment of hematologic malignancies, offering new options for relapsed or refractory cases. However, these therapies carry risks of early complications, such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), and delayed issues like graft-versus-host disease (GVHD), infections, and secondary cancers. Effective management requires early diagnosis using advanced biomarkers and imaging, along with prompt interventions involving immunosuppressants, corticosteroids, and cytokine inhibitors. A multidisciplinary approach is essential, integrating hematologists, oncologists, and infectious disease specialists, with emerging strategies like targeted biologics and personalized medicine showing promise in balancing efficacy with toxicity management. Ongoing research is critical to refine diagnostics and treatments, ensuring that these therapies not only extend survival but also improve patients’ quality of life. This review provides critical insights for healthcare professionals to quickly recognize and treat complications of CAR-T and BsAbs therapies. By focusing on early detection through biomarkers and imaging and outlining timely therapeutic interventions, it aims to equip the multidisciplinary care team with the knowledge necessary to manage the challenges of these advanced treatments effectively, ultimately optimizing patient outcomes.
Collapse
Affiliation(s)
- Ayrton Bangolo
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Behzad Amoozgar
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | | | - Lili Zhang
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Sarvarinder Gill
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Andrew Ip
- Division of Lymphoma, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Christina Cho
- Division of Stem Cell Transplant and Cellular Therapy, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| |
Collapse
|
25
|
Bangolo A, Amoozgar B, Mansour C, Zhang L, Gill S, Ip A, Cho C. Comprehensive Review of Early and Late Toxicities in CAR T-Cell Therapy and Bispecific Antibody Treatments for Hematologic Malignancies. Cancers (Basel) 2025; 17:282. [PMID: 39858064 PMCID: PMC11764151 DOI: 10.3390/cancers17020282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Chimeric antigen receptor T-cell (or CAR-T) therapy and bispecific antibodies (BsAbs) have revolutionized the treatment of hematologic malignancies, offering new options for relapsed or refractory cases. However, these therapies carry risks of early complications, such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), and delayed issues like graft-versus-host disease (GVHD), infections, and secondary cancers. Effective management requires early diagnosis using advanced biomarkers and imaging, along with prompt interventions involving immunosuppressants, corticosteroids, and cytokine inhibitors. A multidisciplinary approach is essential, integrating hematologists, oncologists, and infectious disease specialists, with emerging strategies like targeted biologics and personalized medicine showing promise in balancing efficacy with toxicity management. Ongoing research is critical to refine diagnostics and treatments, ensuring that these therapies not only extend survival but also improve patients' quality of life. This review provides critical insights for healthcare professionals to quickly recognize and treat complications of CAR-T and BsAbs therapies. By focusing on early detection through biomarkers and imaging and outlining timely therapeutic interventions, it aims to equip the multidisciplinary care team with the knowledge necessary to manage the challenges of these advanced treatments effectively, ultimately optimizing patient outcomes.
Collapse
Affiliation(s)
- Ayrton Bangolo
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA; (B.A.); (L.Z.); (S.G.)
| | - Behzad Amoozgar
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA; (B.A.); (L.Z.); (S.G.)
| | | | - Lili Zhang
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA; (B.A.); (L.Z.); (S.G.)
| | - Sarvarinder Gill
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA; (B.A.); (L.Z.); (S.G.)
| | - Andrew Ip
- Division of Lymphoma, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA;
| | - Christina Cho
- Division of Stem Cell Transplant and Cellular Therapy, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA;
| |
Collapse
|
26
|
Alsuliman T, Aubrun C, Bay JO, Beguin Y, Bigenwald C, Brissot E, Chalandon Y, Chevallier P, Pagliuca S, Magro L, Srour M. [Hematological toxicities post-CAR-T cells: Recommendations of the Francophone Society of Bone Marrow Transplantation and Cellular Therapy (SFGM-TC)]. Bull Cancer 2025; 112:S103-S110. [PMID: 38631984 DOI: 10.1016/j.bulcan.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/12/2024] [Accepted: 02/19/2024] [Indexed: 04/19/2024]
Abstract
Chimeric antigen receptor T cell (CAR-T cell) therapy has become a standard-of-care for several hematological and a promising treatment for solid malignancies or for selected non-malignant autoimmune disorders. Hematological complications following this treatment are very common with the majority of patients experiencing at least one cytopenia after CAR-T cell injections. The management of these adverse events is not standardized and represents an area of active research and unmet clinical needs. This harmonization workshop, gathering a group of experts who analyzed this topic, has been conceived for the optimization of the management of patients presenting with post-CAR-T cell hematological toxicities. Based on the data present in the literature, these practical recommendations were made to harmonize the practices of Francophone centers involved in the management of these patients.
Collapse
Affiliation(s)
- Tamim Alsuliman
- Service d'hématologie et de thérapie cellulaire, hôpital Saint-Antoine, AP-HP Sorbonne université, 184, Faubourg-Saint-Antoine, 75012 Paris, France.
| | - Clotilde Aubrun
- Coordination greffe-hémato, CHU Ambroise-Paré, 2, boulevard Kennedy, 7000 Mons, Belgique.
| | - Jacques Olivier Bay
- Service de thérapie cellulaire et d'hématologie clinique adulte, CHU de Clermont-Ferrand, Clermont-Ferrand, France.
| | - Yves Beguin
- Department of Hematology and GIGA Laboratory of Hematology, University Hospital of Liège and ULiège, Liège, Belgique.
| | - Camille Bigenwald
- Département d'hématologie, Gustave-Roussy, université Paris Saclay, Villejuif, France.
| | - Eolia Brissot
- Service d'hématologie et de thérapie cellulaire, hôpital Saint-Antoine, AP-HP Sorbonne université, 184, Faubourg-Saint-Antoine, 75012 Paris, France.
| | - Yves Chalandon
- Service d'hématologie, département d'oncologie, hôpitaux universitaire Genève (HUG) et faculté de médecine, université de Genève, Genève, Suisse.
| | | | - Simona Pagliuca
- Service d'hématologie, UMR 7365, IMoPA, CNRS, campus Brabois Santé, hôpitaux de Brabois, CHRU de Nancy, université de Lorraine, Vandœuvre-lès-Nancy, France.
| | - Léonardo Magro
- Maladies du sang, hôpital Huriez, CHRU de Lille, rue Michel-Polonowski, 59000 Lille, France.
| | - Micha Srour
- Maladies du sang, hôpital Huriez, CHRU de Lille, rue Michel-Polonowski, 59000 Lille, France.
| |
Collapse
|
27
|
Stock S, Bücklein VL, Blumenberg V, Magno G, Emhardt A, Holzem AME, Cordas dos Santos DM, Schmidt C, Grießhammer S, Frölich L, Kobold S, von Bergwelt‐Baildon M, Rejeski K, Subklewe M. Prognostic significance of immune reconstitution following CD19 CAR T-cell therapy for relapsed/refractory B-cell lymphoma. Hemasphere 2025; 9:e70062. [PMID: 39807276 PMCID: PMC11726691 DOI: 10.1002/hem3.70062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/26/2024] [Accepted: 10/18/2024] [Indexed: 01/16/2025] Open
Abstract
Immune deficits after CD19 chimeric antigen receptor (CAR) T-cell therapy can be long-lasting, predisposing patients to infections and non-relapse mortality. In B-cell non-Hodgkin lymphoma (B-NHL), the prognostic impact of immune reconstitution (IR) remains ill-defined, and detailed cross-product comparisons have not been performed to date. In this retrospective observational study, we longitudinally characterized lymphocyte subsets and immunoglobulin levels in 105 B-NHL patients to assess patterns of immune recovery arising after CD19 CAR-T. Three key IR criteria were defined as CD4+ T helper (TH) cells > 200/µL, any detectable B cells, and serum immunoglobulin G (IgG) levels >4 g/L. After a median follow-up of 24.6 months, 38% of patients displayed TH cells, 11% showed any B cells, and 41% had IgG recovery. Notable product-specific differences emerged, including deeper TH cell aplasia with CD28z- versus longer B-cell aplasia with 41BBz-based products. Patients with any IR recovery experienced extended progression-free survival (PFS) (median 20.8 vs. 1.7 months, p < 0.0001) and overall survival (OS) (34.9 vs. 4.0 months, p < 0.0001). While landmark analysis at 90 days confirmed improved PFS in patients with any recovery (34.9 vs. 8.6 months, p = 0.005), no significant OS difference was noted. Notably, 72% of patients with refractory disease never displayed recovery of any IR criteria. Early progressors showed diminished IR at the time of progression/relapse compared to patients with late progression/recurrence (after Day 90). Our results highlight the profound immune deficits observed after CD19 CAR-T and shed light on the intersection of IR and efficacy in B-NHL. Importantly, IR was impaired considerably postprogression, carrying significant implications for subsequent T-cell-engaging therapies and treatment sequencing.
Collapse
Affiliation(s)
- Sophia Stock
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- Division of Clinical Pharmacology, Department of Medicine IVLMU University Hospital, LMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
| | - Veit L. Bücklein
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- Laboratory for Translational Cancer Immunology, LMU Gene CenterMunichGermany
| | - Viktoria Blumenberg
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- Laboratory for Translational Cancer Immunology, LMU Gene CenterMunichGermany
- Cellular Immunotherapy ProgramMassachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown and Broad Institute of Harvard University and Massachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Giulia Magno
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
| | | | | | - David M. Cordas dos Santos
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
- Dana‐Farber Cancer InstituteBostonMassachusettsUSA
| | - Christian Schmidt
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
| | | | - Lisa Frölich
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IVLMU University Hospital, LMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU)NeuherbergGermany
| | - Michael von Bergwelt‐Baildon
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
| | - Kai Rejeski
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
- Adult BMT and Cellular Therapy Service, Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Marion Subklewe
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
- Laboratory for Translational Cancer Immunology, LMU Gene CenterMunichGermany
| |
Collapse
|
28
|
Melica G, Luna de Abia A, Shah GL, Devlin S, Corona M, Fein J, Dahi PB, Giralt SA, Lin RJ, Palomba ML, Parascondola A, Park J, Salles G, Saldia A, Scordo M, Shouval R, Perales MA, Seo SK. Shift from Widespread to Tailored Antifungal Prophylaxis in Lymphoma Patients Treated with CD19 CAR T Cell Therapy: Results from a Large Retrospective Cohort. Transplant Cell Ther 2025; 31:36-44. [PMID: 39448032 PMCID: PMC11780678 DOI: 10.1016/j.jtct.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/19/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024]
Abstract
Patients undergoing CD19 chimeric antigen receptor (CAR)-T cell therapy exhibit multiple immune deficits that may increase their susceptibility to infections. Invasive fungal infections (IFIs) are life-threatening events in the setting of hematologic diseases. However, there is ongoing debate regarding the optimal role and duration of antifungal prophylaxis in this specific patient population. The objective of this study was to provide a comprehensive overview of the evolution of IFI prophylactic strategies over time and to assess IFI incidence rates in a cohort of patients with relapsed or refractory (R/R) lymphoma treated with CAR-T cell therapy. A single-center retrospective study was conducted on a cohort of patients with R/R B cell lymphoma treated with CD19 CAR-T cell therapy between April 2016 and March 2023. Group A (April 2016-August 2020) consisted of patients primarily treated with fluconazole, irrespective of their individual IFI risk profile. In Group B (September 2020-March 2023) antifungal prophylaxis was recommended only for high-risk patients. Overall, 330 patients were included. Antifungal prophylaxis was prescribed to 119/142 (84%) patients in Group A and 58/188 (31%) in Group B (P < .001). Anti-mold azoles were prescribed to 8 (5.6%) patients in Group A and 21 (11.2%) patients in Group B. In Group A, 42 (29%) patients were switched to another antifungal, 9 (21%) because of toxicity, with 6 cases of transaminitis and 3 cases of prolonged QTc. In Group B, 21 (11.2%) patients were switched to the antifungal drug, mainly from fluconazole or micafungin to a mold-active agent following revised guidelines. No difference was found in liver toxicity between the two groups at infusion, day 10, and day 30. No significant differences were observed between the groups. IFIs following CAR-T cell therapy were rare, with 1 case of cryptococcal meningoencephalitis in group A (.7%) and 1 case of invasive aspergillosis in Group B (.5%), both occurring in patients on micafungin prophylaxis. In this large single-center cohort of patients with R/R lymphoma treated with CAR-T cells, we show that individualized prophylaxis, alongside careful management of CAR-T cell-related toxicities such as CRS, was associated with a very low IFI rate, avoiding the risk of unnecessary toxicities, drug-drug interactions, and high costs.
Collapse
Affiliation(s)
- Giovanna Melica
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Infectious Diseases and Clinical Immunology, Henri Mondor Hospital, APHP, Creteil, France
| | - Alejandro Luna de Abia
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Adult Bone Marrow Transplantation, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Gunjan L Shah
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Sean Devlin
- Department of Medicine, Weill Cornell Medical College, New York, New York; Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Magdalena Corona
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joshua Fein
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Parastoo B Dahi
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Sergio A Giralt
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Richard J Lin
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - M Lia Palomba
- Department of Medicine, Weill Cornell Medical College, New York, New York; Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Allison Parascondola
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jae Park
- Department of Medicine, Weill Cornell Medical College, New York, New York; Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gilles Salles
- Department of Medicine, Weill Cornell Medical College, New York, New York; Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Amethyst Saldia
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael Scordo
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Roni Shouval
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Susan K Seo
- Department of Medicine, Weill Cornell Medical College, New York, New York; Department of Medicine, Infectious Disease Service, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
29
|
Cheng H, Shao L, Wang D, Chen Y, Sun Y, Chen Z, Liu J, Wang X, Chen W, Sang W, Qi K, Li Z, Sun C, Shi M, Qiao J, Wu Q, Zeng L, Zheng J, Li L, Xu K, Cao J. Comprehensive characterization of cytopenia after chimeric antigen receptor-T cell infusion in patients with relapsed or refractory multiple myeloma. Cytotherapy 2025; 27:16-24. [PMID: 39283287 DOI: 10.1016/j.jcyt.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/01/2024] [Accepted: 08/14/2024] [Indexed: 12/25/2024]
Abstract
BACKGROUND Many studies have demonstrated the effectiveness of chimeric antigen receptor-T (CAR-T) cell therapy for relapsed or refractory multiple myeloma (RRMM), but the hematologic toxicity has not been well characterized. METHODS A total of 111 adults with RRMM who received BCMA CAR-T cells, BCMA + CD19 CAR-T cells or tandem BCMA/CD19 dual-target (BC19) CAR-T cells infusion were enrolled. We characterized cytopenia and hematologic recovery at different time points after CAR-T-cell therapy, analyzed the effect of cytopenia on prognosis and identified the risk factors. RESULTS Patients had a high probability of cytopenia, with anemia, neutropenia and thrombocytopenia occurring in 92%, 95% and 73%, respectively. There were 60 (54%) patients had prolonged hematologic toxicity (PHT) after D28. The median hemoglobin and platelet count were significantly lower at D28 post-CAR-T cell therapy than at baseline. Hemoglobin increased to above baseline at D90. The median absolute neutrophil count was lower than baseline at D0 and D28, and it recovered to baseline at D180. The baseline level of lactate dehydrogenase was associated with thrombocytopenia. Extramedullary involvement was associated with hemoglobin recovery, while the baseline level of albumin and types of CAR-T were related to platelet recovery. Patients with anemia at baseline and at D0, D180 and D360 had shorter progression-free survival (PFS), while anemia at D0, D60, D180 and D360 was associated with shorter overall survival (OS). Neutropenia at D0 was associated with shorter PFS and patients with neutropenia at D90 or D180 had shorter OS. Patients with thrombocytopenia at any time had shorter PFS and OS. Compared to patients without PHT, patients with PHT had shorter PFS and OS. CONCLUSIONS The majority of RRMM patients treated with CAR-T cells experienced cytopenia. Cytopenia occurred at specific time points was associated with a poorer prognosis.
Collapse
Affiliation(s)
- Hai Cheng
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Lingyan Shao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dandan Wang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yegan Chen
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yingjun Sun
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zihan Chen
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jiaying Liu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xue Wang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wei Chen
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wei Sang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Kunming Qi
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhenyu Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Cai Sun
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ming Shi
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Jianlin Qiao
- Jiangsu Bone Marrow Stem Cell Institute, Xuzhou, China
| | - Qingyun Wu
- Jiangsu Bone Marrow Stem Cell Institute, Xuzhou, China
| | - Lingyu Zeng
- Jiangsu Bone Marrow Stem Cell Institute, Xuzhou, China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Li Li
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| | - Jiang Cao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
30
|
Guo Q, Li J, Wang J, Li L, Wei J, Zhang L. The advent of chimeric antigen receptor T Cell therapy in recalibrating immune balance for rheumatic autoimmune disease treatment. Front Pharmacol 2024; 15:1502298. [PMID: 39734406 PMCID: PMC11672202 DOI: 10.3389/fphar.2024.1502298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
CAR-T cell therapy, a cutting-edge cellular immunotherapy with demonstrated efficacy in treating hematologic malignancies, also exhibits significant promise for addressing autoimmune diseases. This innovative therapeutic approach holds promise for achieving long-term remission in autoimmune diseases, potentially offering significant benefits to affected patients. Current targets under investigation for the treatment of these conditions include CD19, CD20, and BCMA, among others. However, CAR-T therapy faces difficulties such as time-consuming cell manufacturing, complex and expensive process, and the possibility of severe adverse reactions complicating the treatment, etc. This article examines CAR-T therapy across various rheumatic autoimmune diseases, including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), Sjögren's syndrome (SS), systemic sclerosis (SSc), antisynthetase syndrome (ASS), and ANCA-associated vasculitis (AAV), highlighting both therapeutic advancements and ongoing challenges.
Collapse
Affiliation(s)
- Qianyu Guo
- Department of Rheumatology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Jie Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Juanjuan Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Linxin Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Jia Wei
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
31
|
Tatake IJ, Arnason JE. CARs for lymphoma. Best Pract Res Clin Haematol 2024; 37:101601. [PMID: 40074511 DOI: 10.1016/j.beha.2025.101601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/17/2025] [Indexed: 03/14/2025]
Abstract
Chimeric Antigen Receptor (CAR)-T cell therapy has revolutionized treatment options for B-cell Non-Hodgkin Lymphoma (NHL). CD19-targeting CAR-T cell therapy is approved for treatment in Diffuse Large B Cell Lymphoma, Follicular Lymphoma, Mantle Cell Lymphoma, and Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma. CAR-T cells demonstrate robust and durable responses even in heavily pretreated patients. Clinicians should monitor for Cytokine Release Syndrome (CRS) and Immune Effector Cell Neurotoxicity Syndrome (ICANS), as well as cytopenias, infection, and secondary malignancies. Ongoing questions remain in improving manufacturing efficacy, sequencing CAR-T cells amongst other therapies including bi-specific antibodies (BiTEs), and predicting optimal responders. In addition, novel CARs are being developed with alternative targets or that secrete activating cytokines (i.e. "armored CARs"). CAR-T cells represent an effective lymphoma therapy and should be considered for eligible patients.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/therapeutic use
- Receptors, Chimeric Antigen/genetics
- Lymphoma, Follicular/therapy
- Lymphoma, Follicular/immunology
- Lymphoma, Follicular/pathology
- Lymphoma, Mantle-Cell/therapy
- Lymphoma, Mantle-Cell/immunology
- Lymphoma, Mantle-Cell/pathology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/pathology
- Cytokine Release Syndrome/immunology
- Antigens, CD19/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
Collapse
Affiliation(s)
- Ishan J Tatake
- Department of Medicine, Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Jon E Arnason
- Department of Medicine, Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
32
|
Tix T, Subklewe M, von Bergwelt-Baildon M, Rejeski K. Survivorship in Chimeric Antigen Receptor T-Cell Therapy Recipients: Infections, Secondary Malignancies, and Non-Relapse Mortality. Oncol Res Treat 2024; 48:212-219. [PMID: 39561735 DOI: 10.1159/000542631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T-cell therapy has significantly advanced the treatment of hematologic malignancies, offering curative potential for patients with relapsed or refractory disease. However, the long-term survivorship of these patients is marked by unique challenges, particularly immune deficits and infectious complications, second primary malignancies (SPMs), and non-relapse mortality (NRM). Understanding and addressing these risks is paramount to improving patient outcomes and quality of life. SUMMARY This review explores the incidence and risk factors for NRM and long-term complications following CAR T-cell therapy. Infections are the leading cause of NRM, accounting for over 50% of cases, driven by neutropenia, hypogammaglobulinemia, and impaired cellular immunity. SPMs, including secondary myeloid and T-cell malignancies, are increasingly recognized, prompting the FDA to issue a black box warning, although their direct link to CAR T cells remains disputed. While CAR T-cell-specific toxicities like cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome contribute to morbidity, they represent only a minority of NRM cases. The management of these complications is critical as CAR T-cell therapy is being evaluated for broader use, including in earlier treatment lines and for non-malignant conditions like autoimmune diseases. KEY MESSAGES CAR T-cell therapy has revolutionized cancer treatment, but survivorship is complicated by infections, SPMs, and ultimately endangered by NRM. Prophylactic strategies, close monitoring, and toxicity management strategies are key to improving long-term outcomes.
Collapse
Affiliation(s)
- Tobias Tix
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany,
| | - Marion Subklewe
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany
| | - Kai Rejeski
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
33
|
Palacios-Berraquero ML, Rodriguez-Marquez P, Calleja-Cervantes ME, Berastegui N, Zabaleta A, Burgos L, Alignani D, San Martin-Uriz P, Vilas-Zornoza A, Rodriguez-Diaz S, Inoges S, Lopez-Diaz de Cerio A, Huerga S, Tamariz E, Rifon J, Alfonso-Pierola A, Lasarte JJ, Paiva B, Hernaez M, Rodriguez-Otero P, San-Miguel J, Ezponda T, Rodriguez-Madoz JR, Prosper F. Molecular mechanisms promoting long-term cytopenia after BCMA CAR-T therapy in multiple myeloma. Blood Adv 2024; 8:5479-5492. [PMID: 39058976 PMCID: PMC11532743 DOI: 10.1182/bloodadvances.2023012522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 06/12/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
ABSTRACT Hematologic toxicity is a common side effect of chimeric antigen receptor T-cell (CAR-T) therapies, being particularly severe among patients with relapsed or refractory multiple myeloma (MM). In this study, we characterized 48 patients treated with B-cell maturation antigen (BCMA) CAR-T cells to understand kinetics of cytopenia, identify predictive factors, and determine potential mechanisms underlying these toxicities. We observed that overall incidence of cytopenia was 95.7%, and grade >3 thrombocytopenia and neutropenia, 1 month after infusion, was observed in 57% and 53% of the patients, respectively, being still present after 1 year in 4 and 3 patients, respectively. Baseline cytopenia and high peak inflammatory markers were highly correlated with cytopenia that persisted up to 3 months. To determine potential mechanisms underlying cytopenias, we evaluated the paracrine effect of BCMA CAR-T cells on hematopoietic stem and progenitor cell (HSPC) differentiation using an ex vivo myeloid differentiation model. Phenotypic analysis showed that supernatants from activated CAR-T cells (spCAR) halted HSPC differentiation, promoting more immature phenotypes, which could be prevented with a combination of interferon γ, tumor necrosis factor α/β, transforming growth factor β, interleukin-6 (IL-6) and IL-17 inhibitors. Single-cell RNA sequencing demonstrated upregulation of transcription factors associated with early stages of hematopoietic differentiation in the presence of spCAR (GATA2, RUNX1, CEBPA) and a decrease in the activity of key regulons involved in neutrophil and monocytic maturation (ID2 and MAFB). These results suggest that CAR-T activation induces HSPC maturation arrest through paracrine effects and provides potential treatments to mitigate the severity of this toxicity.
Collapse
Affiliation(s)
- Maria Luisa Palacios-Berraquero
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Paula Rodriguez-Marquez
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Maria Erendira Calleja-Cervantes
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Computational Biology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Nerea Berastegui
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Aintzane Zabaleta
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Leire Burgos
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Diego Alignani
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Patxi San Martin-Uriz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Amaia Vilas-Zornoza
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Saray Rodriguez-Diaz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Susana Inoges
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Immunology and Immunotherapy Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Ascensión Lopez-Diaz de Cerio
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Immunology and Immunotherapy Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Sofia Huerga
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Esteban Tamariz
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Jose Rifon
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Ana Alfonso-Pierola
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Juan Jose Lasarte
- Immunology and Immunotherapy Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Bruno Paiva
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Mikel Hernaez
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Computational Biology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Data Science and Artificial Intelligence Institute, Universidad de Navarra, Pamplona, Spain
| | - Paula Rodriguez-Otero
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Jesus San-Miguel
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Teresa Ezponda
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Juan Roberto Rodriguez-Madoz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Felipe Prosper
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| |
Collapse
|
34
|
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: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar 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
|
35
|
Deng L, Yu X, Song X, Guan R, Li W, Hou Y, Shao Y, Zhao Y, Wang J, Liu Y, Xiao Q, Xin B, Zhou F. Efficacy and risk of donor-derived CAR-T treatment of relapsed B-cell acute lymphoblastic leukemia after hematopoietic stem cell transplantation. Cytotherapy 2024; 26:1301-1307. [PMID: 38888526 DOI: 10.1016/j.jcyt.2024.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024]
Abstract
The one-year survival rate for patients experiencing a relapse of B-cell acute lymphocytic leukemia (B-ALL) following hematopoietic stem cell transplantation (HSCT) is approximately 30%. Patients experiencing a relapse after allogeneic HSCT frequently encounter difficulties in obtaining autologous CAR-T products. We conducted a study involving 14 patients who received donor-derived CAR-T therapy for relapsed B-ALL following HSCT between August 2019 and May 2023 in our center. The results revealed a CR/CRi rate of 78.6% (11/14), a GVHD rate of 21.4% (3/14), and a 1-year overall survival (OS) rate of 56%. Decreased bone marrow donor cell chimerism in 9 patients recovered after CAR-T therapy. The main causes of death were disease progression and infection. Further analysis showed that GVHD (HR 7.224, 95% CI 1.42-36.82, P = 0.017) and platelet recovery at 30 days (HR 6.807, 95% CI 1.61-28.83, P = 0.009) are significantly associated with OS after CAR-T therapy. Based on the findings, we conclude that donor-derived CAR-T cells are effective in treating relapsed B-ALL patients following HSCT. Additionally, GVHD and poor platelet recovery impact OS, but further verification with a larger sample size is needed.
Collapse
Affiliation(s)
- Lei Deng
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Xiaolin Yu
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Xiaocheng Song
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Rui Guan
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Wenjun Li
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Yixi Hou
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Yan Shao
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Yuerong Zhao
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Jing Wang
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Yue Liu
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Qianqian Xiao
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Bo Xin
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Fang Zhou
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China.
| |
Collapse
|
36
|
Shahid Z, Jain T, Dioverti V, Pennisi M, Mikkilineni L, Thiruvengadam SK, Shah NN, Dadwal S, Papanicolaou G, Hamadani M, Carpenter PA, Alfaro GM, Seo SK, Hill JA. Best Practice Considerations by The American Society of Transplant and Cellular Therapy: Infection Prevention and Management After Chimeric Antigen Receptor T Cell Therapy for Hematological Malignancies. Transplant Cell Ther 2024; 30:955-969. [PMID: 39084261 DOI: 10.1016/j.jtct.2024.07.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is rapidly advancing, offering promising treatments for patients with hematological malignancy. However, associated infectious complications remain a significant concern because of their contribution to patient morbidity and non-relapse mortality. Recent epidemiological insights shed light on risk factors for infections after CAR T-cell therapy. However, the available evidence is predominantly retrospective, highlighting a need for further prospective studies. Institutions are challenged with managing infections after CAR T-cell therapy but variations in the approaches taken underscore the importance of standardizing infection prevention and management protocols across different healthcare settings. Therefore, the Infectious Diseases Special Interest Group of the American Society of Transplantation and Cellular Therapy assembled an expert panel to develop best practice considerations. The aim was to guide healthcare professionals in optimizing infection prevention and management for CAR T-cell therapy recipients and advocates for early consultation of Infectious Diseases during treatment planning phases given the complexities involved. By synthesizing current evidence and expert opinion these best practice considerations provide the basis for understanding infection risk after CAR T-cell therapies and propose risk-mitigating strategies in children, adolescents, and adults. Continued research and collaboration will be essential to refining and effectively implementing these recommendations.
Collapse
Affiliation(s)
- Zainab Shahid
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Tania Jain
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Veronica Dioverti
- Division of Infectious Disease, Department of Medicine, John Hopkins School of Medicine, Baltimore, Maryland
| | - Martini Pennisi
- Division of Hematology and Stem Cell Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Lekha Mikkilineni
- Division of Bone and Marrow Transplant & Cellular Therapies, Stanford School of Medicine, Palo Alto, California
| | - Swetha Kambhampati Thiruvengadam
- Division of Lymphoma, Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sanjeet Dadwal
- Division of Infectious Disease, Department of Medicine, City of Hope National Medical Center, Duarte, California
| | - Genovefa Papanicolaou
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mehdi Hamadani
- Bone Marrow Transplant & Cellular Therapy Program, Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Milwaukee, Wisconsin
| | - Paul A Carpenter
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Gabriela Maron Alfaro
- Department of Infectious Diseases, St. Jude Children's Research Hospital and Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Susan K Seo
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joshua A Hill
- Vaccine and Infectious Disease Division, Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| |
Collapse
|
37
|
Woo JS, Nguyen K, Liu L, Krishnan A, Siddiqi T, Borogovac A. Mobilizing CARs: Benefits, drawbacks, and directions for outpatient CAR T-cell therapy. Semin Hematol 2024; 61:273-283. [PMID: 39327109 DOI: 10.1053/j.seminhematol.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/16/2024] [Accepted: 08/19/2024] [Indexed: 09/28/2024]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has heralded a new era in the treatment of various hematological malignancies, increasingly being utilized in earlier lines of therapy. Moreover, cellular therapies are currently under investigation for their potential in treating solid malignancies and autoimmune disorders. As the scope of indications for CAR-T therapy continues to expand, along with the associated reductions in costs and hospital admissions, many medical centers are transitioning towards outpatient CAR-T models. Moreover, ongoing efforts to mitigate complications such as cytokine release syndrome (CRS) or neurotoxicity include the development of premedication strategies, prompt management of adverse events, and the advancement of newer, safer CAR-T cell therapies. However, despite these advancements, the inherent risk of these life-threatening complications remains a critical concern in CAR-T therapy. Institutions must diligently anticipate and effectively manage these complications to ensure the safety and well-being of patients undergoing CAR-T therapy. This includes establishing robust protocols for timely identification and intervention of adverse events, and seamless pathways for transitioning patients to a higher level of care if necessary. This review provides an overview of the current landscape of outpatient CAR-T therapy and offers essential insights into the key clinical and operational considerations needed to implement a successful outpatient CAR-T program.
Collapse
Affiliation(s)
- Jennifer S Woo
- City of Hope Lennar Foundation Cancer Center, City of Hope National Medical Center, Irvine, CA
| | - Kim Nguyen
- City of Hope Lennar Foundation Cancer Center, City of Hope National Medical Center, Irvine, CA
| | - Lawrence Liu
- City of Hope Comprehensive Cancer Center, City of Hope National Medical Center, Duarte, CA
| | - Amrita Krishnan
- City of Hope Lennar Foundation Cancer Center, City of Hope National Medical Center, Irvine, CA
| | - Tanya Siddiqi
- City of Hope Lennar Foundation Cancer Center, City of Hope National Medical Center, Irvine, CA
| | - Azra Borogovac
- City of Hope Lennar Foundation Cancer Center, City of Hope National Medical Center, Irvine, CA.
| |
Collapse
|
38
|
Xia Y, Wang L, Shen X, Xu Y, Xu W, Li J, Fan L, Chen L. Mesenchymal stem cell infusion for enhancing hematopoietic recovery and addressing cytopenias in CAR-T cell therapy. Stem Cell Res Ther 2024; 15:333. [PMID: 39334276 PMCID: PMC11437967 DOI: 10.1186/s13287-024-03941-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-T therapy has emerged as a promising treatment for hematologic malignancies. However, cytopenia remains one of the most frequent and challenging adverse effects of this therapy. METHODS We conducted a retrospective analysis of 26 patients with relapsed/refractory aggressive B-cell lymphoma who received CAR-T therapy at our center. Subsequently, to investigate measures to address cytopenias following CAR-T therapy, we isolated and generated murine CAR-T cells and bone marrow-derived mesenchymal stem cells (MSCs), establishing a murine syngeneic CAR-T therapy model. We assessed the impact of MSC infusion on hematopoietic recovery post-CAR-T therapy by evaluating complete blood count, bone marrow hematopoietic stem cells and their subpopulations, bone marrow histomorphology, and hematopoiesis-related genes. RESULTS All patients experienced cytopenias to varying degrees, with complete lineage involvement in half of the patients. Grade ≥ 3 cytopenias were observed in 88.46% of the patients. CAR-T therapy was associated with a higher incidence of biphasic, late-onset, or prolonged cytopenias. Survival analysis indicated that neutropenia and lymphopenia tended to be associated with better prognosis, whereas thrombocytopenia tended to be related to poorer outcomes. Through animal experiments, we discovered that MSCs infusion boosted HSCs and their long-term subpopulations, enhancing hematopoietic recovery, particularly in the megakaryocyte lineage, and mitigating bone marrow damage. Importantly, both in vitro and in vivo experiments demonstrated that MSCs did not compromise the activity or antitumor efficacy of CAR-T cells. CONCLUSIONS Our findings propose MSCs infusion as a promising strategy to address cytopenias, particularly thrombocytopenia, after CAR-T therapy. This approach could help overcome certain limitations of cellular immunotherapy by enhancing hematopoietic recovery without compromising the efficacy of CAR-T cells. HIGHLIGHTS 1 Cytopenia is a frequently observed adverse effect following CAR-T therapy, and it is often characterized by biphasic and prolonged patterns. 2 MSCs play a critical role in promoting hematopoietic recovery and mitigating bone marrow damage in a murine model of CAR-T therapy 3 The activity and antitumor efficacy of CAR-T cells were not impaired by MSCs.
Collapse
Affiliation(s)
- Yuan Xia
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Department of Hematology, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, Taizhou, 225300, Jiangsu, China
| | - Li Wang
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xuxing Shen
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Ying Xu
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Wei Xu
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jianyong Li
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Lei Fan
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Lijuan Chen
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
39
|
Stella F, Chiappella A, Casadei B, Bramanti S, Ljevar S, Chiusolo P, Di Rocco A, Tisi MC, Carrabba MG, Cutini I, Martino M, Dodero A, Bonifazi F, Santoro A, Sorà F, Botto B, Barbui AM, Russo D, Musso M, Grillo G, Krampera M, Olivieri J, Ladetto M, Cavallo F, Massaia M, Arcaini L, Pennisi M, Zinzani PL, Miceli R, Corradini P. A Multicenter Real-life Prospective Study of Axicabtagene Ciloleucel versus Tisagenlecleucel Toxicity and Outcomes in Large B-cell Lymphomas. Blood Cancer Discov 2024; 5:318-330. [PMID: 38953781 PMCID: PMC11369587 DOI: 10.1158/2643-3230.bcd-24-0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/29/2024] [Accepted: 05/30/2024] [Indexed: 07/04/2024] Open
Abstract
This real-world prospective observational study across 21 Italian centers (CART-SIE) compares axicabtagene ciloleucel (axi-cel) and tisagenlecleucel (tisa-cel) outcomes in 485 patients with relapsed/refractory large B-cell lymphoma with baseline characteristics matched by stabilized inverse propensity score weighting. Axi-cel versus tisa-cel had higher all-grade cytokine release syndrome (78.6% vs. 89.3%, P = 0.0017) and neurotoxicity (9.9% vs. 32.2%, P < 0.0001) but also superior progression-free survival (PFS) at 1 year (46.5% vs. 34.1%, P = 0.0009). Even among patients who failed bridging therapy, axi-cel PFS was superior to tisa-cel (37.5% vs. 22.7%, P = 0.0059). Differences in overall survival and high-grade immune toxicities were not significant. The CAR-HEMATOTOX score not only predicted hematologic toxicity but also 1-year survival outcomes (51.5% in CAR-HEMATOTOX high vs. 77.2% in CAR-HEMATOTOX low, P < 0.0001). Twenty patients developed second primary malignancies, including two cases of T-cell neoplasms. These findings enable more informed selection of anti-CD19 CAR T-cell therapy, balancing bridging, safety, and efficacy considerations for individual patients. Significance: The findings of this study on 485 patients with relapsed/refractory large B-cell lymphoma treated with commercial axi-cel and tisa-cel indicate axi-cel's superior PFS after propensity score weighting. The predictive utility of CAR-HEMATOTOX in assessing not only toxicity but also outcomes across both CAR T-cell products may guide future risk-stratified management strategies.
Collapse
MESH Headings
- Humans
- Male
- Female
- Middle Aged
- Prospective Studies
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/mortality
- Aged
- Biological Products/adverse effects
- Biological Products/therapeutic use
- Biological Products/administration & dosage
- Adult
- Antigens, CD19/immunology
- Antigens, CD19/therapeutic use
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Treatment Outcome
- Receptors, Antigen, T-Cell/therapeutic use
- Receptors, Antigen, T-Cell/immunology
- Aged, 80 and over
Collapse
Affiliation(s)
| | - Annalisa Chiappella
- Division of Hematology and Stem Cell Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| | - Beatrice Casadei
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy.
| | - Stefania Bramanti
- IRCCS Humanitas Research Hospital, Transplantation Unit Department of Oncology and Haematology, Milan, Italy.
| | - Silva Ljevar
- Unit of Biostatistics for Clinical Research, Department of Data Science, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| | - Patrizia Chiusolo
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Alice Di Rocco
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.
| | - Maria C. Tisi
- Hematology Unit, San Bortolo Hospital, Vicenza, Italy.
| | | | - Ilaria Cutini
- SOD Terapie Cellulari e Medicina Trasfusionale, AAD Trapianto di midollo osseo, Ospedale Careggi, Firenze, Italy.
| | - Massimo Martino
- Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano “Bianchi-Melacrino-Morelli”, Reggio Calabria, Italy.
| | - Anna Dodero
- Division of Hematology and Stem Cell Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| | - Francesca Bonifazi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy.
| | - Armando Santoro
- IRCCS Humanitas Research Hospital, Transplantation Unit Department of Oncology and Haematology, Milan, Italy.
| | - Federica Sorà
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy.
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Barbara Botto
- SC Ematologia AOU Città della Salute e della Scienza, Torino, Italy.
| | - Anna M. Barbui
- Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy.
| | - Domenico Russo
- Unit of Blood Disease and Bone Marrow Transplantation, and Unit of Hematology, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy.
| | - Maurizio Musso
- UOC di Oncoematologia e TMO, Dipartimento Oncologico “La Maddalena”, Palermo, Italy.
| | - Giovanni Grillo
- Dipartimento di Ematologia e trapianto di midollo, ASST Grande Ospedale Metropolitano Niguarda, Milano, Italy.
| | - Mauro Krampera
- Hematology and Bone Marrow Transplant Unit, Section of Biomedicine of Innovation, Department of Engineering for Innovative Medicine (DIMI), University of Verona, Verona, Italy.
| | - Jacopo Olivieri
- Clinica Ematologica, Centro Trapianti e Terapie Cellulari “Carlo Melzi”, Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy.
| | - Marco Ladetto
- Dipartimento di Medicina Traslazionale, Università del Piemonte Orientale ed SCDU Ematologia AOU SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy.
| | - Federica Cavallo
- Division of Hematology, University Hospital A.O.U. “Città della Salute e della Scienza”, Turin, Italy.
- Division of Hematology, Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy.
| | - Massimo Massaia
- Division of Hematology–AO S. Croce e Carle, Cuneo and Laboratory of Blood Tumor Immunology, Molecular Biotechnology Center “Guido Tarone”, University of Torino, Torino, Italy.
| | - Luca Arcaini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Martina Pennisi
- Division of Hematology and Stem Cell Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| | - Pier L. Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy.
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy.
| | - Rosalba Miceli
- Unit of Biostatistics for Clinical Research, Department of Data Science, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| | - Paolo Corradini
- Chair of Hematology, University of Milan, Milano, Italy.
- Division of Hematology and Stem Cell Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| |
Collapse
|
40
|
Schroeder T, Martens T, Fransecky L, Valerius T, Schub N, Pott C, Baldus C, Stölzel F. Management of chimeric antigen receptor T (CAR-T) cell-associated toxicities. Intensive Care Med 2024; 50:1459-1469. [PMID: 39172238 PMCID: PMC11377606 DOI: 10.1007/s00134-024-07576-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/21/2024] [Indexed: 08/23/2024]
Abstract
The use of chimeric antigen receptor T (CAR-T) cells is a significant therapeutic improvement increasing the prognosis for patients with a variety of hematological malignancies. However, this therapy has also sometimes life-threatening, complications. Therefore, knowledge of the treatment and management of these complications, especially in treatment centers and intensive care units, respectively, is of outstanding importance. This review provides recommendations for the diagnosis, management, and treatment of CAR-T cell-associated complications such as cytokine release syndrome, immune effector cell associated neurotoxicity syndrome, hematotoxicity, hypogammaglobulinemia, and CAR-T cell-induced pseudo-progression amongst others for physicians treating patients with CAR-T cell-associated complications and intensivists.
Collapse
Affiliation(s)
- Torsten Schroeder
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Tjark Martens
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Lars Fransecky
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Thomas Valerius
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Natalie Schub
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Christiane Pott
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Claudia Baldus
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Friedrich Stölzel
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany.
| |
Collapse
|
41
|
Miller A, Daum R, Wang T, Wu M, Tat C, Pfeiffer T, Navai S, Heczey A, Hegde M, Ahmed N, Whittle SB, Hill L, Martinez C, Krance R, Ramos CA, Rouce RH, Lulla P, Heslop HE, Omer B, Shekar M. Prolonged cytopenias after immune effector cell therapy and lymphodepletion in patients with leukemia, lymphoma and solid tumors. Cytotherapy 2024; 26:1026-1032. [PMID: 38819365 PMCID: PMC11344664 DOI: 10.1016/j.jcyt.2024.04.075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 04/10/2024] [Accepted: 04/29/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND AIMS The success of chimeric antigen receptor (CAR) T-cell therapy in treating B-cell malignancies has led to the evaluation of CAR T-cells targeting a variety of other malignancies. Although the efficacy of CAR T-cells is enhanced when administered post-lymphodepleting chemotherapy, this can trigger bone marrow suppression and sustained cytopenia after CD19.CAR T-cell therapy. Additionally, systemic inflammation associated with CAR T-cell activity may contribute to myelosuppression. Cytopenias, such as neutropenia and thrombocytopenia, elevate the risk of severe infections and bleeding, respectively. However, data on the incidence of prolonged cytopenias after immune effector therapy in the solid tumor context remain limited. OBJECTIVE We compared the incidence of prolonged cytopenias after immune effector therapy including genetically modified T-cells, virus-specific T-cells (VSTs) and NKT-cells, as well non-gene-modified VSTs for leukemia, lymphoma, and solid tumors (ST) to identify associated risk factors. METHODS A retrospective analysis was conducted of 112 pediatric and adult patients with relapsed and/or refractory cancers who received lymphodepleting chemotherapy followed by immune effector therapy. Patients treated with 13 distinct immune effector cell therapies through 11 single-center clinical trials and 2 commercial products over a 6-year period were categorized into 3 types of malignancies: leukemia, lymphoma and ST. We obtained baseline patient characteristics and adverse events data for each participant, and tracked neutrophil and platelet counts following lymphodepletion. RESULTS Of 112 patients, 104 (92.9%) experienced cytopenias and 88 (79%) experienced severe cytopenias. Patients with leukemia experienced significantly longer durations of severe neutropenia (median duration of 14 days) compared with patients with lymphoma (7 days) or ST (11 days) (P = 0.002). Patients with leukemia also had a higher incidence of severe thrombocytopenia (74.1%), compared with lymphoma (46%, P = 0.03) and ST (14.3%, P < 0.0001). Prolonged cytopenias were significantly associated with disease type (63% of patients with leukemia, 44% of patients with lymphoma, and 22.9% of patients with ST, P = 0.006), prior hematopoietic stem cell transplant (HSCT) (66.7% with prior HSCT versus 38.3% without prior HSCT, P = 0.039), and development of immune effector cell-associated neurotoxicity syndrome (ICANS) (75% with ICANS versus 38% without ICANS, P = 0.027). There was no significant association between prolonged cytopenias and cytokine release syndrome. CONCLUSIONS Immune effector recipients often experience significant cytopenias due to marrow suppression following lymphodepletion regardless of disease, but prolonged severe cytopenias are significantly less common after treatment of patients with lymphoma and solid tumors.
Collapse
Affiliation(s)
- Anne Miller
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Rachel Daum
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - Tao Wang
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Mengfen Wu
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Candise Tat
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Thomas Pfeiffer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA; Department of Pediatrics, Division of Hematology/Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shoba Navai
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Andras Heczey
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Meenakshi Hegde
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Nabil Ahmed
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Sarah B Whittle
- Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - LaQuisa Hill
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Caridad Martinez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Robert Krance
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Carlos A Ramos
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Rayne H Rouce
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Premal Lulla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Bilal Omer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Meghan Shekar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
42
|
Mingot-Castellano ME, Reguera-Ortega JL, Zafra Torres D, Hernani R, Lopez-Godino O, Guerreiro M, Herrero B, López-Corral L, Luna A, Gonzalez-Pinedo L, Chinea-Rodriguez A, Africa-Martín A, Bailen R, Martinez-Cibrian N, Balsalobre P, Filaferro S, Alonso-Saladrigues A, Barba P, Perez-Martinez A, Calbacho M, Perez-Simón JA, Sánchez-Pina JM, On Behalf Of The Spanish Group Of Hematopoietic Transplant And Cell Therapy Geth-Tc. Use of Eltrombopag to Improve Thrombocytopenia and Tranfusion Requirement in Anti-CD19 CAR-T Cell-Treated Patients. J Clin Med 2024; 13:5117. [PMID: 39274330 PMCID: PMC11396136 DOI: 10.3390/jcm13175117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/16/2024] Open
Abstract
Background/Objectives: Immune effector cell-associated hematotoxicity (ICAHT) is a frequent adverse event after chimeric antigen receptor (CAR)-T cell therapy. Grade ≥ 3 thrombocytopenia occurs in around one-third of patients, and many of them become platelet transfusion-dependent. Eltrombopag is a thrombopoietin receptor agonist (TPO-RA) able to accelerate megakaryopoiesis, which has been used successfully in patients with bone marrow failure and immune thrombocytopenia (ITP). Its role in managing thrombocytopenia and other cytopenias in CAR-T cell-treated patients has been scarcely addressed. Our aim was to report the safety and efficacy of this approach in patients included in the Spanish Group for Hematopoietic Transplantation and Cellular Therapy (GETH-TC) registry. Methods: This is a retrospective, multicenter, observational study. Patients who developed platelet transfusion dependence subsequently to CAR-T cells and received eltrombopag to improve platelet counts were recruited in 10 Spanish hospitals. Results: Thirty-eight patients were enrolled and followed up for a median (interquartile range [IQR]) of 175 (99, 489) days since CAR-T cell infusion. At the moment eltrombopag was indicated, 18 patients had thrombocytopenia and another severe cytopenia, while 8 patients had severe pancytopenia. After 32 (14, 38) days on eltrombopag, 29 (76.3%) patients recovered platelet transfusion independence. The number of platelet units transfused correlated with the time needed to restore platelet counts higher than 20 × 109/L (Rho = 0.639, p < 0.001). Non-responders to eltrombopag required more platelet units (58 [29, 69] vs. 12 [6, 26] in responders, p = 0.002). Nineteen out of twenty-three (82.6%) patients recovered from severe neutropenia after 22 (11, 31) days on eltrombopag. Twenty-nine out of thirty-five (82.9%) patients recovered red blood cell (RBC) transfusion independence after 29 (17, 44) days. Seven patients recovered all cell lineages while on treatment. No thromboembolic events were reported. Only two transient toxicities (cholestasis, hyperbilirubinemia) were reported during eltrombopag treatment, none of which compelled permanent drug withdrawal. Conclusions: Eltrombopag could be safely used to manage thrombocytopenia and accelerate transfusion independence in CAR-T cell-treated patients.
Collapse
Affiliation(s)
- Maria-Eva Mingot-Castellano
- Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, IBiS/CSIC, Universidad de Sevilla, 41004 Sevilla, Spain
| | - Juan Luis Reguera-Ortega
- Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, IBiS/CSIC, Universidad de Sevilla, 41004 Sevilla, Spain
| | | | - Rafael Hernani
- INCLIVA Health Research Institute, Hospital Clínico Universitario, 46010 Valencia, Spain
| | - Oriana Lopez-Godino
- Hematology Department, Centro Regional de Hemodonación, IMIB-Pascual Parrilla, Hospital Universitario Morales-Meseguer, 30008 Murcia, Spain
| | - Manuel Guerreiro
- Servicio de Hematología, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Blanca Herrero
- Hospital Infantil Universitario del Niño Jesús, 28009 Madrid, Spain
| | - Lucia López-Corral
- IBSAL, CIBERONC, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), Hospital Universitario de Salamanca (Spain), 37007 Salamanca, Spain
| | - Alejandro Luna
- Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Lesli Gonzalez-Pinedo
- Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Spain
| | | | - Ana Africa-Martín
- IBSAL, CIBERONC, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), Hospital Universitario de Salamanca (Spain), 37007 Salamanca, Spain
| | - Rebeca Bailen
- Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, 28009 Madrid, Spain
| | - Nuria Martinez-Cibrian
- Hospital Clinic Barcelona, Institut de Recerca Sant Joan de Déu, Barcelona Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | | | | | | | - Pere Barba
- Grupo Español de Trasplante Hematopoyético y Terapia Celular, 28040 Madrid, Spain
| | - Antonio Perez-Martinez
- Pediatric Hematology-Oncology Department, La Paz University Hospital, 28034 Madrid, Spain
- Pediatric Department, Autonomous University of Madrid, 28034 Madrid, Spain
- CIBERER-ISCIII, IdiPAZ-CNIO Pediatric OncoHematology Clinical Research Unit, 28034 Madrid, Spain
| | - María Calbacho
- Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Jose Antonio Perez-Simón
- Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, IBiS/CSIC, Universidad de Sevilla, 41004 Sevilla, Spain
| | | | | |
Collapse
|
43
|
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] [Scholar 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
|
44
|
Canichella M, de Fabritiis P. Cell-Based Treatment in Acute Myeloid Leukemia Relapsed after Allogeneic Stem Cell Transplantation. Biomedicines 2024; 12:1721. [PMID: 39200186 PMCID: PMC11351713 DOI: 10.3390/biomedicines12081721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 09/02/2024] Open
Abstract
Allogeneic stem cell transplant (ASCT) remains the only treatment option for patients with high-risk acute myeloid leukemia (AML). Recurrence of leukemic cells after ASCT represents a dramatic event associated with a dismal outcome, with a 2-year survival rate of around 20%. Adoptive cell therapy (ACT) is a form of cell-based strategy that has emerged as an effective therapy to treat and prevent post-ASCT recurrence. Lymphocytes are the principal cells used in this therapy and can be derived from a hematopoietic stem cell donor, the patient themselves, or healthy donors, after being engineered to express the chimeric antigen receptor (CAR-T and UniCAR-T). In this review, we discuss recent advances in the established strategy of donor lymphocyte infusion (DLI) and the progress and challenges of CAR-T cells.
Collapse
Affiliation(s)
| | - Paolo de Fabritiis
- Hematology Unit, St. Eugenio Hospital, ASL Roma2, 00144 Rome, Italy;
- Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| |
Collapse
|
45
|
Soff GA, Al‐Samkari H, Leader A, Eisen M, Saad H. Romiplostim in chemotherapy-induced thrombocytopenia: A review of the literature. Cancer Med 2024; 13:e7429. [PMID: 39135303 PMCID: PMC11319220 DOI: 10.1002/cam4.7429] [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] [Scholar Register] [Received: 11/24/2023] [Revised: 06/04/2024] [Accepted: 06/13/2024] [Indexed: 08/16/2024] Open
Abstract
Chemotherapy-induced thrombocytopenia (CIT) is a common challenge of cancer therapy and can lead to chemotherapy dose reduction, delay, and/or discontinuation, affecting relative dose intensity, and possibly adversely impacting cancer care. Besides changing anticancer regimens, standard management of CIT has been limited to platelet transfusions and supportive care. Use of the thrombopoietin receptor agonist romiplostim, already approved for use in immune thrombocytopenia, has shown promising signs of efficacy in CIT. In a phase 2 prospective randomized study of solid tumor patients with platelet counts <100 × 109/L for ≥4 weeks due to CIT, weekly romiplostim corrected the platelet count to >100 × 109/L in 93% (14/15) of patients within 3 weeks versus 12.5% (1/8) of untreated patients (p < 0.001). Including patients treated with romiplostim in an additional single-arm cohort, 85% (44/52) of all romiplostim-treated patients responded with platelet count correction within 3 weeks. Several retrospective studies of CIT have also shown responses to weekly romiplostim, with the largest study finding that poor response to romiplostim was predicted by tumor invasion of the bone marrow (odds ratio, 0.029; 95% CI: 0.0046-0.18; p < 0.001), prior pelvic irradiation (odds ratio, 0.078; 95% CI: 0.0062-0.98; p = 0.048), and prior temozolomide treatment (odds ratio 0.24; 95% CI: 0.061-0.96; p = 0.043). Elsewhere, lower baseline TPO levels were predictive of romiplostim response (p = 0.036). No new safety signals have emerged from romiplostim CIT studies. Recent treatment guidelines, including those from the National Comprehensive Cancer Network, now support consideration of romiplostim use in CIT. Data are expected from two ongoing phase 3 romiplostim CIT trials.
Collapse
Affiliation(s)
- Gerald A. Soff
- University of Miami Health System/Sylvester Comprehensive Cancer CenterMiamiFloridaUSA
| | - Hanny Al‐Samkari
- Center for Hematology, Massachusetts General Hospital Cancer CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Avi Leader
- Section of Hematology, Memorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | | | | |
Collapse
|
46
|
Brudno JN, Kochenderfer JN. Current understanding and management of CAR T cell-associated toxicities. Nat Rev Clin Oncol 2024; 21:501-521. [PMID: 38769449 PMCID: PMC11529341 DOI: 10.1038/s41571-024-00903-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2024] [Indexed: 05/22/2024]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has revolutionized the treatment of several haematological malignancies and is being investigated in patients with various solid tumours. Characteristic CAR T cell-associated toxicities such as cytokine-release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) are now well-recognized, and improved supportive care and management with immunosuppressive agents has made CAR T cell therapy safer and more feasible than it was when the first regulatory approvals of such treatments were granted in 2017. The increasing clinical experience with these therapies has also improved recognition of previously less well-defined toxicities, including movement disorders, immune effector cell-associated haematotoxicity (ICAHT) and immune effector cell-associated haemophagocytic lymphohistiocytosis-like syndrome (IEC-HS), as well as the substantial risk of infection in patients with persistent CAR T cell-induced B cell aplasia and hypogammaglobulinaemia. A more diverse selection of immunosuppressive and supportive-care pharmacotherapies is now being utilized for toxicity management, yet no universal algorithm for their application exists. As CAR T cell products targeting new antigens are developed, additional toxicities involving damage to non-malignant tissues expressing the target antigen are a potential hurdle. Continued prospective evaluation of toxicity management strategies and the design of less-toxic CAR T cell products are both crucial for ongoing success in this field. In this Review, we discuss the evolving understanding and clinical management of CAR T cell-associated toxicities.
Collapse
Affiliation(s)
- Jennifer N Brudno
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - James N Kochenderfer
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
47
|
Hu B, Korsos V, Palomba ML. Chimeric antigen receptor T-cell therapy for aggressive B-cell lymphomas. Front Oncol 2024; 14:1394057. [PMID: 39011476 PMCID: PMC11246842 DOI: 10.3389/fonc.2024.1394057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/21/2024] [Indexed: 07/17/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a revolutionary approach in the treatment of lymphoma. This review article provides an overview of the four FDA-approved CAR T-cell products for aggressive B-cell lymphoma, including diffuse large B-cell lymphoma and mantle cell lymphoma, highlighting their efficacy and toxicity as well as discussing future directions.
Collapse
Affiliation(s)
- Bei Hu
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute/Wake Forest School of Medicine, Charlotte, NC, United States
| | - Victoria Korsos
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - M. Lia Palomba
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
48
|
Kitamura W, Asada N, Ikegawa S, Fujiwara H, Kamoi C, Ennishi D, Nishimori H, Fujii K, Fujii N, Matsuoka KI, Maeda Y. Activated CD4 + T Cell Proportion in the Peripheral Blood Correlates with the Duration of Cytokine Release Syndrome and Predicts Clinical Outcome after Chimeric Antigen Receptor T Cell Therapy. Intern Med 2024; 63:1863-1872. [PMID: 38945932 PMCID: PMC11272506 DOI: 10.2169/internalmedicine.2556-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 10/16/2023] [Indexed: 07/02/2024] Open
Abstract
Objective Chimeric antigen receptor (CAR) T cell therapy is an emerging and effective therapy for relapsed or refractory diffuse large B cell lymphoma (R/R DLBCL). The characteristic toxicities of CAR T cell therapy include cytokine release syndrome (CRS) and prolonged cytopenia. We investigated the factors associated with these complications after CAR T cell therapy by analyzing lymphocyte subsets following CAR T cell infusion. Methods We retrospectively analyzed peripheral blood samples on days 7, 14, and 28 after tisagenlecleucel (tisa-cel) infusion by flow cytometry at our institution between June 2020 and September 2022. Patients Thirty-five patients with R/R DLBCL who received tisa-cel therapy were included. Results A flow cytometry-based analysis of blood samples from these patients revealed that the proportion of CD4+CD25+CD127+ T cells (hereafter referred to as "activated CD4+ T cells" ) among the total CD4+ T cells on day 7 after tisa-cel infusion correlated with the duration of CRS (r=0.79, p<0.01). In addition, a prognostic analysis of the overall survival (OS) using time-dependent receiver operating characteristic curves indicated a significantly more favorable OS and progression-free survival of patients with a proportion of activated CD4+ T cells among the total CD4+ T cells <0.73 (p=0.01, and p<0.01, respectively). Conclusion These results suggest that the proportion of activated CD4+ T cells on day 7 after tisa-cel infusion correlates with the CRS duration and predicts clinical outcomes after CAR T cell therapy. Further studies with a larger number of patients are required to validate these observations.
Collapse
MESH Headings
- Humans
- Male
- Female
- Cytokine Release Syndrome/blood
- Cytokine Release Syndrome/etiology
- Cytokine Release Syndrome/therapy
- Cytokine Release Syndrome/immunology
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Middle Aged
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/blood
- Lymphoma, Large B-Cell, Diffuse/immunology
- Aged
- Retrospective Studies
- CD4-Positive T-Lymphocytes/immunology
- Adult
- Treatment Outcome
- Receptors, Chimeric Antigen/immunology
- Prognosis
- Receptors, Antigen, T-Cell
Collapse
Affiliation(s)
- Wataru Kitamura
- Department of Hematology and Oncology, Okayama University Hospital, Japan
| | - Noboru Asada
- Department of Hematology and Oncology, Okayama University Hospital, Japan
| | - Shuntaro Ikegawa
- Department of Hematology and Oncology, Okayama University Hospital, Japan
- Division of Blood Transfusion, Okayama University Hospital, Japan
| | - Hideaki Fujiwara
- Department of Hematology and Oncology, Okayama University Hospital, Japan
| | - Chihiro Kamoi
- Department of Hematology and Oncology, Okayama University Hospital, Japan
- Division of Blood Transfusion, Okayama University Hospital, Japan
| | - Daisuke Ennishi
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Japan
| | - Hisakazu Nishimori
- Department of Hematology and Oncology, Okayama University Hospital, Japan
| | - Keiko Fujii
- Division of Clinical Laboratory, Okayama University Hospital, Japan
| | - Nobuharu Fujii
- Division of Blood Transfusion, Okayama University Hospital, Japan
| | - Ken-Ichi Matsuoka
- Department of Hematology and Oncology, Okayama University Hospital, Japan
| | - Yoshinobu Maeda
- Department of Hematology and Oncology, Okayama University Hospital, Japan
| |
Collapse
|
49
|
Liang EC, Rejeski K, Fei T, Albittar A, Huang JJ, Portuguese AJ, Wu Q, Raj S, Subklewe M, Shouval R, Gauthier J. Development and validation of an automated computational approach to grade immune effector cell-associated hematotoxicity. Bone Marrow Transplant 2024; 59:910-917. [PMID: 38627450 PMCID: PMC11380036 DOI: 10.1038/s41409-024-02278-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/19/2024] [Accepted: 03/27/2024] [Indexed: 05/24/2024]
Abstract
Hematologic toxicity frequently complicates chimeric antigen receptor (CAR) T-cell therapy, resulting in significant morbidity and mortality. In an effort to standardize reporting, the European Hematology Association (EHA) and European Society of Blood and Marrow Transplantation (EBMT) devised the immune effector cell-associated hematotoxicity (ICAHT) grading system, distinguishing between early (day 0-30) and late (after day +30) events based on neutropenia depth and duration. However, manual implementation of ICAHT grading criteria is time-consuming and susceptible to subjectivity and error. To address these challenges, we introduce a novel computational approach, utilizing the R programming language, to automate early and late ICAHT grading. Given the complexities of early ICAHT grading, we benchmarked our approach both manually and computationally in two independent cohorts totaling 1251 patients. Our computational approach offers significant implications by streamlining grading processes, reducing manual time and effort, and promoting standardization across varied clinical settings. We provide this tool to the scientific community alongside a comprehensive implementation guide, fostering its widespread adoption and enhancing reporting consistency for ICAHT.
Collapse
Affiliation(s)
- Emily C Liang
- Fred Hutchinson Cancer Center, Seattle, WA, USA.
- University of Washington, Seattle, WA, USA.
| | - Kai Rejeski
- Adult Bone Marrow Transplantation and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Teng Fei
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Jennifer J Huang
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- University of Washington, Seattle, WA, USA
| | - Andrew J Portuguese
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- University of Washington, Seattle, WA, USA
| | - Qian Wu
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Sandeep Raj
- Adult Bone Marrow Transplantation and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marion Subklewe
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Roni Shouval
- Adult Bone Marrow Transplantation and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jordan Gauthier
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- University of Washington, Seattle, WA, USA
| |
Collapse
|
50
|
Bindal P, Patell R, Chiasakul T, Lauw MN, Ko A, Wang TF, Zwicker JI. A meta-analysis to assess the risk of bleeding and thrombosis following chimeric antigen receptor T-cell therapy: Communication from the ISTH SSC Subcommittee on Hemostasis and Malignancy. J Thromb Haemost 2024; 22:2071-2080. [PMID: 38574863 PMCID: PMC11437522 DOI: 10.1016/j.jtha.2024.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 02/26/2024] [Accepted: 03/17/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Chimeric antigen receptor T-cell (CAR T-cell) therapy is increasingly utilized for treatment of hematologic malignancies. Hematologic toxicities including thrombosis and bleeding complications have been reported. Accurate estimates for thrombotic and bleeding outcomes are lacking. OBJECTIVES We performed a systematic review and meta-analysis in patients who received CAR T-cell therapy for an underlying hematologic malignancy with the objective to: a) assess the thrombosis and bleeding risk associated with CAR T-cell therapy, b) assess the impact of CRS and ICANS on the risks of thrombosis and bleeding, and c) assess the safety of anticoagulant or antiplatelet use in the period following treatment with CAR T-cell therapy. METHODS We searched MEDLINE, EMBASE, and Cochrane CENTRAL up to February 2022 for studies reporting thrombotic or bleeding outcomes in patients receiving CAR T-cell therapy. Pooled event rates were calculated using a random-effects model. We performed subgroup analyses stratified by follow-up duration, CAR T-cell target antigen, and underlying hematologic malignancy. RESULTS We included 47 studies with a total of 7040 patients. High heterogeneity between studies precluded reporting of overall pooled rates of thrombotic and bleeding events. In studies with follow-up duration of ≤6 months, the pooled incidence of venous thrombotic events was 2.4% (95% CI, 1.4%-3.4%; I2 = 0%) per patient-month, whereas the rate was 0.1% (95% CI, 0%-0.1%; I2 = 0%) per patient-month for studies with longer follow-up periods (>6 months). The pooled incidences of any bleeding events per patient-month in studies with follow-up duration of ≤6 months and >6 months were 1.9% (95% CI, 0.6%-3.1%; I2 = 78%) and 0.3% (95% CI: 0%-0.8%, I2 = 40%), respectively. Secondary analyses by CAR T-cell target antigen, underlying malignancy, and primary outcome of the studies did not reveal significant differences in the rates of thromboembolism, any bleeding events, or major bleeding events. CONCLUSION The risk of both thrombosis and bleeding following CAR T-cell therapy appears to be highest in the initial months following infusion.
Collapse
Affiliation(s)
- Poorva Bindal
- Division of Hematologic Malignancies and Cellular Therapies, University of Massachusetts, Worcester, Massachusetts, USA
| | - Rushad Patell
- Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA. https://twitter.com/rushadpatell
| | - Thita Chiasakul
- Center of Excellence in Translational Hematology, Division of Hematology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Mandy N Lauw
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Amica Ko
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Tzu-Fei Wang
- Department of Medicine, University of Ottawa at The Ottawa Hospital and Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Jeffrey I Zwicker
- Department of Medicine, Hematology Service, Memorial Sloan Kettering Cancer Center, New York City, New York, USA; Weill Cornell Medical School, New York City, New York, USA.
| |
Collapse
|