1
|
Qi S, Li J, Gu X, Zhang Y, Zhou W, Wang F, Wang W. Impacts of ageing on the efficacy of CAR-T cell therapy. Ageing Res Rev 2025; 107:102715. [PMID: 40058461 DOI: 10.1016/j.arr.2025.102715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/19/2025] [Accepted: 03/02/2025] [Indexed: 03/17/2025]
Abstract
Chimeric antigen receptor T cells recognizing CD19 (19CAR-T) cell therapy has achieved robust clinical efficacy when treating some hematological malignancies, but which patient subgroups benefit mostly remains elusive. Here we summarized the data of 541 patients from 30 clinical trials who underwent 19 CAR-T therapy and analyzed the different clinical responses between young (<44 years), middle-aged (45-59 years) and elderly (>60 years) patients and found that the young patients showed a higher level of complete response (CR) rate. Therefore, we then summarize the advances of studies focusing on the effects of age on anti-tumor efficacy of CAR-T therapy and analyze the reasons for the low CR rate after CAR-T cell therapy in elderly patients with tumors, aiming to provide hints for oncologists to select the most suitable candidate for this cancer immunotherapy.
Collapse
Affiliation(s)
- Shimao Qi
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Jiaqian Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Xinyu Gu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Yalan Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Weilin Zhou
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Fengling Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Wei Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
2
|
Nie J, Zhou L, Tian W, Liu X, Yang L, Yang X, Zhang Y, Wei S, Wang DW, Wei J. Deep insight into cytokine storm: from pathogenesis to treatment. Signal Transduct Target Ther 2025; 10:112. [PMID: 40234407 PMCID: PMC12000524 DOI: 10.1038/s41392-025-02178-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/22/2024] [Accepted: 02/12/2025] [Indexed: 04/17/2025] Open
Abstract
Cytokine storm (CS) is a severe systemic inflammatory syndrome characterized by the excessive activation of immune cells and a significant increase in circulating levels of cytokines. This pathological process is implicated in the development of life-threatening conditions such as fulminant myocarditis (FM), acute respiratory distress syndrome (ARDS), primary or secondary hemophagocytic lymphohistiocytosis (HLH), cytokine release syndrome (CRS) associated with chimeric antigen receptor-modified T (CAR-T) therapy, and grade III to IV acute graft-versus-host disease following allogeneic hematopoietic stem cell transplantation. The significant involvement of the JAK-STAT pathway, Toll-like receptors, neutrophil extracellular traps, NLRP3 inflammasome, and other signaling pathways has been recognized in the pathogenesis of CS. Therapies targeting these pathways have been developed or are currently being investigated. While novel drugs have demonstrated promising therapeutic efficacy in mitigating CS, the overall mortality rate of CS resulting from underlying diseases remains high. In the clinical setting, the management of CS typically necessitates a multidisciplinary team strategy encompassing the removal of abnormal inflammatory or immune system activation, the preservation of vital organ function, the treatment of the underlying disease, and the provision of life supportive therapy. This review provides a comprehensive overview of the key signaling pathways and associated cytokines implicated in CS, elucidates the impact of dysregulated immune cell activation, and delineates the resultant organ injury associated with CS. In addition, we offer insights and current literature on the management of CS in cases of FM, ARDS, systemic inflammatory response syndrome, treatment-induced CRS, HLH, and other related conditions.
Collapse
Grants
- 82070217, 81873427 National Natural Science Foundation of China (National Science Foundation of China)
- 82100401 National Natural Science Foundation of China (National Science Foundation of China)
- 81772477, 81201848, 82473220 National Natural Science Foundation of China (National Science Foundation of China)
- 82330010,81630010,81790624 National Natural Science Foundation of China (National Science Foundation of China)
- National High Technology Research and Development Program of China, Grant number: 2021YFA1101500.
- The Hubei Provincial Natural Science Foundation (No.2024AFB050)
- Project of Shanxi Bethune Hospital, Grant Numbber: 2023xg02); Fundamental Research Program of Shanxi Province, Grant Numbber: 202303021211224
- The Key Scientific Research Project of COVID-19 Infection Emergency Treatment of Shanxi Bethune Hospital (2023xg01), 2023 COVID-19 Research Project of Shanxi Provincial Health Commission (No.2023XG001, No. 2023XG005), Four “Batches” Innovation Project of Invigorating Medical through Science and Technology of Shanxi Province (2023XM003), Cancer special Fund research project of Shanxi Bethune Hospital (No. 2020-ZL04), and External Expert Workshop Fund Program of Shanxi Provincial Health Commission(Proteomics Shanxi studio for Huanghe professor)
- Fundamental Research Program of Shanxi Province(No.202303021221192); 2023 COVID-19 Emergency Project of Shanxi Health Commission (Nos.2023XG001,2023XG005)
Collapse
Affiliation(s)
- Jiali Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Ling Zhou
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China
| | - Weiwei Tian
- 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
| | - Xiansheng Liu
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China
- 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
| | - Liping Yang
- 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
| | - Xingcheng Yang
- 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
| | - Yicheng Zhang
- 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
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China.
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China.
| | - Jia Wei
- 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.
| |
Collapse
|
3
|
Kannan A, Jeffrey K, Misbah S, Ramasamy K. Practical guidance on the prevention and management of infection in multiple myeloma patients: A case-based approach. Blood Rev 2025:101287. [PMID: 40240231 DOI: 10.1016/j.blre.2025.101287] [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: 02/10/2025] [Revised: 03/25/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025]
Abstract
The risk of infection in multiple myeloma patients is significant, due to immune dysfunction secondary to myeloma, immunosenescence and age-related comorbidities, given the elderly myeloma patient demographic. Newer treatments, despite providing unprecedented improvements in disease-control, have further elevated infection risk. This risk is so substantial that we are approaching a period where a subset of older myeloma patients may be more likely to die secondary to infectious complications imposed by redirected T-cell therapy rather than from myeloma. As a result, it is essential to provide myeloma patients with the appropriate prophylaxis and monitoring against infection. In this review, we discuss disease-related, patient-related and treatment-related reasons for the increased infection risk in myeloma patients, and how to both prevent and manage this risk through creating a dynamic, infection prevention plan that is personalised to the individual patient.
Collapse
Affiliation(s)
- A Kannan
- Medical Sciences Division, Medical Sciences Division, Academic Centre, John Radcliffe Hospital, University of Oxford, Headington OX3 9DU, United Kingdom.
| | - K Jeffrey
- Oxford University Hospitals, John Radcliffe Hospital, NHS Foundation Trust, Oxford OX3 9DU, UK; Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford OX3 7BN, UK.
| | - S Misbah
- Oxford University Hospitals, John Radcliffe Hospital, NHS Foundation Trust, Oxford OX3 9DU, UK.
| | - K Ramasamy
- Oxford University Hospitals, John Radcliffe Hospital, NHS Foundation Trust, Oxford OX3 9DU, UK; Oxford Translational Myeloma Centre, NDORMS, University of Oxford, Oxford OX3 7LD, UK.
| |
Collapse
|
4
|
Pagano L, Maschmeyer G, Lamoth F, Blennow O, Xhaard A, Spadea M, Busca A, Cordonnier C, Maertens J. Primary antifungal prophylaxis in hematological malignancies. Updated clinical practice guidelines by the European Conference on Infections in Leukemia (ECIL). Leukemia 2025:10.1038/s41375-025-02586-7. [PMID: 40200079 DOI: 10.1038/s41375-025-02586-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 02/27/2025] [Accepted: 03/24/2025] [Indexed: 04/10/2025]
Abstract
At the 10th European Conference on Infections in Leukaemia (ECIL), the guidelines for antifungal prophylaxis in pediatric and adult patients with hematological malignancies (HM) were updated and some changes introduced. Regarding acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) patients undergoing remission induction chemotherapy, a B-II grading has been assigned to isavuconazole, micafungin, and caspofungin, based on non-randomized studies that have shown efficacy in preventing invasive fungal diseases (IFD). Regarding high-risk MDS patients treated with azacytidine, prophylaxis with posaconazole during the first four cycles of treatment is supported in the literature. Prophylaxis is not indicated in patients treated for myeloproliferative neoplasms (NPM), acute lymphoid leukemia (ALL), and Hodgkin lymphoma (HL). For patients with chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL), prophylaxis is not generally indicated. For patients with multiple myeloma (MM), prophylaxis is not indicated and the limited epidemiological data available do not support the use of prophylaxis in subjects treated with bispecific antibodies. For patients undergoing allogeneic hematopoietic stem cell transplantation (HSCT), no substantial changes were made, apart from the addition of isavuconazole with grading B-II in the post-engraftment period. In patients undergoing auto-HSCT, antifungal prophylaxis is not indicated. Previous ECIL guidelines did not include CAR-T cells. The expert panel proposes to endorse the use of anti-mold prophylaxis in high-risk patients during pre-infusion and post-infusion, while in low-risk patients, anti-yeast prophylaxis can be recommended (B-II). For pediatric hematology patients, based on newly published data, caspofungin received a B-I grading as mold-active prophylaxis. Moreover, patients with ALL with insufficient treatment response during induction therapy, and children older than 12 y.o are now considered at high risk for IFD and are recommended to receive antifungal prophylaxis.
Collapse
Affiliation(s)
- Livio Pagano
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Università Cattolica del Sacro Cuore, Roma, Italia.
- Divisione di Ematologia Geriatrica ed Emopatie Rare, Fondazione Policlinico Universitario Agostino Gemelli - IRCCS, Roma, Italia.
| | | | - Frederic Lamoth
- Infectious Diseases Service and Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ola Blennow
- Department of Infectious Diseases, Karolinska Universitetssjukhuset, Stockholm, Sweden
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Alienor Xhaard
- Hematologie greffe, hopital Saint-Louis, APHP, Universite Paris Cite, Paris, France
| | - Manuela Spadea
- Department of Pediatric and Publich Health Sciences, University of Turin, Turin, Italy
- Regina Margherita Children's Hospital, Turin, Italy
| | - Alessandro Busca
- Trapianto Cellule Staminali, AOU Citta' della Salute e della Scienza, Turin, Italy
| | | | - Johan Maertens
- Department of Haematology, Department of Microbiology, Immunology and Transplantation, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
5
|
Vonberg FW, Malik I, O'Reilly M, Hyare H, Carr AS, Roddie C. Neurotoxic complications of chimeric antigen receptor (CAR) T-cell therapy. J Neurol Neurosurg Psychiatry 2025:jnnp-2024-333924. [PMID: 40185628 DOI: 10.1136/jnnp-2024-333924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/25/2025] [Indexed: 04/07/2025]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has revolutionised the treatment of haematological malignancies and has demonstrated efficacy in early trials for solid tumours, neurological and rheumatological autoimmune diseases. However, CAR-T is complicated in some patients by neurotoxicity syndromes including immune-effector cell-associated neurotoxicity syndrome, and the more recently described movement and neurocognitive treatment-emergent adverse events, and tumour inflammation-associated neurotoxicity. These neurotoxic syndromes remain poorly understood and are associated with significant morbidity and mortality. A multidisciplinary approach, including neurologists, haematologists and oncologists, is critical for the diagnosis and management of CAR-T neurotoxicity. This approach will be of increasing importance as the use of CAR-T expands, its applications increase and as novel neurotoxic syndromes emerge.
Collapse
Affiliation(s)
- Frederick W Vonberg
- National Hospital for Neurology and Neurosurgery, London, UK
- UCL Queen Square Institute of Neurology, London, UK
| | - Imran Malik
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Maeve O'Reilly
- Haematology, University College London Hospitals NHS Foundation Trust, London, UK
- UCL Cancer Institute, London, UK
| | - Harpreet Hyare
- UCL Queen Square Institute of Neurology, London, UK
- Neuroradiology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Aisling S Carr
- UCL Queen Square Institute of Neurology, London, UK
- Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Claire Roddie
- Haematology, University College London Hospitals NHS Foundation Trust, London, UK
- UCL Cancer Institute, London, UK
| |
Collapse
|
6
|
Passweg JR, Baldomero H, Atlija M, Kleovoulou I, Witaszek A, Alexander T, Angelucci E, Averbuch D, Bazarbachi A, Ciceri F, Greco R, Hazenberg MD, Kalwak K, McLornan DP, Neven B, Perić Z, Risitano AM, Ruggeri A, Sánchez-Ortega I, Snowden JA, Sureda A. The 2023 EBMT report on hematopoietic cell transplantation and cellular therapies. Increased use of allogeneic HCT for myeloid malignancies and of CAR-T at the expense of autologous HCT. Bone Marrow Transplant 2025; 60:519-528. [PMID: 39939433 PMCID: PMC11971038 DOI: 10.1038/s41409-025-02524-2] [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: 12/16/2024] [Revised: 01/13/2025] [Accepted: 01/30/2025] [Indexed: 02/14/2025]
Abstract
In 2023, 47,731 HCT (20,485 (42.9%) allogeneic and 27,246 (57.1%) autologous) in 43,902 patients were reported by 696 European centers. 6042 patients received advanced cellular therapies, 4888 of which were CAR-T. Compared to the previous year there was an increase in CAR-T (+52.5%), in allogeneic HCT (+7.8%) but none in autologous HCT (+0.4%). Main indications for allogeneic HCT were myeloid (11,748; 60.7%), lymphoid malignancies (4,850; 25.0%), and non-malignant disorders (2558; 13.2%). Use of allogeneic HCT increased for AML (+12.1%) and for NHL (+11.0%), particularly in T-NHL (+25.6%). Main indications for autologous HCT were lymphomas (7890; 32.2%), PCD (14,271; 58.2%), and solid tumors (1608; 6.6%) with recovering numbers for autoimmune diseases. In patients with allogeneic HCT, the use of sibling donors increased by +1.0%, haploidentical donors by +11.7%, and unrelated donors by +11.1%. Cord blood HCT decreased again by -5.4%. Pediatric HCT activity increased slightly (5455; +0.1%) with differences between allogeneic (4111; -0.5%) and autologous HCT (1344: +1.7%). Use of CAR-T increased to a cumulative total of 13,927 patients including patients treated for autoimmune diseases. Overall, numbers show a complete recovery from the pandemic dip with increased cellular therapy at the expense of autologous HCT. Allogeneic HCT activity focuses on myeloid malignancies.
Collapse
Affiliation(s)
- Jakob R Passweg
- EBMT Activity Survey Office Basel, Hematology Division, University Hospital, Basel, Switzerland
| | - Helen Baldomero
- EBMT Activity Survey Office Basel, Hematology Division, University Hospital, Basel, Switzerland.
| | - Marina Atlija
- EBMT Activity Survey Office Leiden, Leiden, The Netherlands
| | | | | | - Tobias Alexander
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Emanuele Angelucci
- Hematology and Cellular Therapy Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Dina Averbuch
- Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ali Bazarbachi
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Fabio Ciceri
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Raffaella Greco
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Mette D Hazenberg
- Department of Hematology, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands
- Department of Hematopoiesis, Sanquin Research, Amsterdam, The Netherlands
| | - Krzysztof Kalwak
- Clinical Department of Pediatric BMT, Hematology and Oncology, Wroclaw Medical University, Wroclaw, Poland
| | - Donal P McLornan
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Bénédicte Neven
- Pediatric Immune-Hematology Unit, Necker Children Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | | | - Antonio M Risitano
- Hematology and Hematopoietic Transplant Unit, Azienda Ospedaliera di Rilievo Nazionale San Giuseppe Moscati" (A.O.R.N. Giuseppe Moscati), Avellino, Italy
| | - Annalisa Ruggeri
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | | | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Anna Sureda
- Clinical Hematology Department, Institut Català d'Oncologia-Hospitalet, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), University of Barcelona, Barcelona, Spain
| |
Collapse
|
7
|
Gaoual Y, Mahyaoui A, Yachi L, Bouatia M, Aliat Z, Rahali Y. Advancements and challenges in CAR T cell therapy for pediatric brain tumors: A review. J Oncol Pharm Pract 2025:10781552251331609. [PMID: 40156311 DOI: 10.1177/10781552251331609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
Chimeric Antigen Receptor (CAR) T cell therapy represents a groundbreaking advancement in immunotherapy, initially gaining FDA approval for treating hematological malignancies. This therapy has shown promising results in solid tumors, particularly in pediatric brain tumors, which are the leading cause of cancer-related death in children. CAR T cells are engineered to target specific antigens on tumor cells, thereby reducing off-target effects and increasing the cytotoxic impact on cancer cells. Over the years, CAR T cell technology has evolved through five generations, each enhancing the structure, functionality, and safety of these cells. Despite these advancements, the application of CAR T cells in solid tumors, especially within the central nervous system (CNS), faces significant challenges. These include the physical barrier posed by the blood-brain barrier (BBB), the immunosuppressive tumor microenvironment (TME), and the heterogeneity of tumor antigens. The review discusses several promising antigenic targets for CAR T cells in pediatric brain tumors, such as HER2, EphA2, IL-13Rα2, and Survivin, which have been explored in recent clinical trials. These trials have shown early promise in improving patient outcomes, though the risks of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) remain concerns. The future of CAR T cell therapy lies in overcoming these barriers through innovative approaches like "Armored CARs" or TRUCKs, designed to modulate the TME and improve CAR T cell efficacy in solid tumors. Additionally, combination therapies and safety switches in next-generation CAR T cells are being explored to enhance therapeutic potential while minimizing adverse effects.
Collapse
Affiliation(s)
- Yasmina Gaoual
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Specialties Hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
| | - Adam Mahyaoui
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Specialties Hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
| | - Lamyae Yachi
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Children's hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
- Team of analytical chemistry and bromatology, Faculty of Medicine and Pharmacy, Mohammed V University- Rabat, 10 170 Rabat, Morocco
| | - Mustapha Bouatia
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Children's hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
- Team of analytical chemistry and bromatology, Faculty of Medicine and Pharmacy, Mohammed V University- Rabat, 10 170 Rabat, Morocco
- Ibn Sina University Hospital Center, 10 170 Rabat, Morocco
| | - Zineb Aliat
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Specialties Hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
- Team of Formulation and Quality Control of Health Products, Faculty of Medicine and Pharmacy, Mohammed V University- Rabat, 10 170 Rabat, Morocco
| | - Younes Rahali
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Specialties Hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
- Ibn Sina University Hospital Center, 10 170 Rabat, Morocco
- Team of Formulation and Quality Control of Health Products, Faculty of Medicine and Pharmacy, Mohammed V University- Rabat, 10 170 Rabat, Morocco
| |
Collapse
|
8
|
Dahunsi D, Eleanya C, Akintunde A, Oluwole O. Prolonged Cytopenia with CAR-T Cell Therapy and Management Recommendations. Clin Hematol Int 2025; 7:47-54. [PMID: 40161263 PMCID: PMC11952652 DOI: 10.46989/001c.126463] [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: 06/24/2024] [Accepted: 08/27/2024] [Indexed: 04/02/2025] Open
Abstract
Chimeric antigen receptor T-cell (CAR T-cell) therapy has revolutionized the treatment of lymphoid malignancies. Prolonged cytopenias, though poorly understood, have emerged as important considerations in the treatment process. In this review, we classified cytopenias into early (< 30 days post CAR T infusion), and late-occurring (after day 30 post infusion). We identified previous chemotherapy and lymphodepletion chemotherapy as the major risk factors contributing to early cytopenia. Product characteristics, such as costimulatory domains, and side effects of therapy such as cytokine release syndrome (CRS) and immune effector cell associated neurotoxicity syndrome (ICANS) were identified as contributing factors to prolonged cytopenias occurring more than 30 days post CAR-T infusion. We recommend close monitoring with frequent checks, enhanced care with granulocyte colony stimulating factor (GCSF) support for grade 3-4 neutropenia, blood transfusion for severe anemia (Hb < 7g/dL), platelets for severe thrombocytopenia (< 10,000/µL) and thrombopoietin (TPO) mimetics such as eltrombopag or romiplostim for prolonged severe thrombocytopenia in patients at high-risk of hemorrhagic complications.
Collapse
|
9
|
Luan D, DeWolf S, Fei T, Raj S, Shah GL, Lareau CA, Alhomoud M, Salles G, Rivas-Delgado A, Rejeski K, Park JH, Luttwak E, Luna de Abia A, Corona M, Ntrivalas E, Cassanello G, Gomez-Llobell M, Parascondola A, Scordo M, Hsu KC, Palomba ML, Perales MA, Shouval R. Dynamics of Immune Reconstitution and Impact on Outcomes across CAR-T Cell Products in Large B-cell Lymphoma. Blood Cancer Discov 2025; 6:119-130. [PMID: 39666878 PMCID: PMC11876948 DOI: 10.1158/2643-3230.bcd-24-0163] [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: 10/09/2024] [Accepted: 12/11/2024] [Indexed: 12/14/2024] Open
Abstract
SIGNIFICANCE This study reveals differences in IR patterns after CAR-T therapy in patients with large B-cell lymphoma, with early NK cell recovery emerging as a key predictor of survival. These findings provide potential future avenues of research for improving patient outcomes and tailoring post-therapy management strategies to mitigate relapse risk.
Collapse
Affiliation(s)
- Danny Luan
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Susan DeWolf
- Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Teng Fei
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sandeep Raj
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gunjan L. Shah
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Caleb A. Lareau
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mohammad Alhomoud
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gilles Salles
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Kai Rejeski
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine III – Hematology/Oncology, LMU University Hospital, Munich, Germany
| | - Jae H. Park
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Efrat Luttwak
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alejandro Luna de Abia
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Adult Bone Marrow Transplantation Service, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Magdalena Corona
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Evangelos Ntrivalas
- Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Giulio Cassanello
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Marina Gomez-Llobell
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Allison Parascondola
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael Scordo
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Katharine C. Hsu
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - M. Lia Palomba
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Miguel-Angel Perales
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Roni Shouval
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
10
|
Zhou X, Kortuem KM, Rasche L, Einsele H. Bispecific antibody and chimeric antigen receptor (CAR) modified T-cell in the treatment of multiple myeloma: Where do we stand today? Presse Med 2025; 54:104265. [PMID: 39662761 DOI: 10.1016/j.lpm.2024.104265] [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: 09/27/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024] Open
Abstract
Although the prognosis of patients with multiple myeloma (MM) has been significantly improved by the introduction of proteasome inhibitors, immunomodulatory drugs and monoclonal antibodies, MM is still considered an incurable disease in the vast majority of the patients. In recent years, T-cell based immunotherapy represents a novel treatment strategy for relapsed/refractory (RR) MM. So far, chimeric antigen receptor (CAR) modified T-cells and bispecific T-cell engaging antibodies (bsAb) have shown promising anti-MM efficacy and manageable safety profile within clinical trials, and B-cell maturation antigen (BCMA) is the most commonly used immune target for T-cell based immunotherapies in MM. To date, several CAR T-cell and bsAb products have already been approved for the treatment of RRMM, leading to a paradigm shift in the MM therapy and providing a potential curative option. In this review, we provide a summary of mechanisms of action, immune targets, selected clinical data, resistance mechanisms and therapy sequencing of CAR T-cell and bsAb in MM.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - K Martin Kortuem
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Leo Rasche
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany.
| |
Collapse
|
11
|
Kharfan-Dabaja MA, Mohty R, Easwar N, Johnston P, Iqbal M, Epperla N, Yared J, Ahmed N, Hamadani M, Beitinjaneh A, Narkhede M, Jagadeesh D, Ramakrishnan Geethakumari P, Dholaria B, Gergis U, Munoz J, Sandoval-Sus J, Locke FL, Fein J, Khurana A, Ayala E, Annunzio K, Rapoport AP, Lutfi F, Akhtar OS, Lekakis L, Mehta A, Oluwole OO, Logue J, Jain MD, Shore T, Durani U, Alhaj Moustafa M, McGuirk J, Lin Y, Yamshon S, Chavez JC. Chimeric antigen receptor T cell therapy in octogenarians with B cell lymphoma: a real-world US multicenter collaborative study. Bone Marrow Transplant 2025:10.1038/s41409-025-02541-1. [PMID: 40025178 DOI: 10.1038/s41409-025-02541-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/12/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
Older patients with lymphoma are typically underrepresented in clinical trials with chimeric antigen receptor T cell (CAR T) therapy. In this multicenter, observational study we aimed to assess the safety and efficacy of standard CD19 CAR T in patients 80 years of age or older. At total of 88 patients, median age 82 (range, 80-89) years, were included. Diffuse large B cell lymphoma (DLBCL) (N = 60, 68.2%) represented the most common histology. Patients were treated mostly with axicabtagene ciloleucel (N = 41, 46.6%) followed by lisocabtagene maraleucel (N = 25, 28.4%). Cytokine release syndrome (CRS) (any grade) was seen in 68 (77.3%) and 51 (58%) developed immune effector cell-associated neurotoxicity syndrome (ICANS). Incidence of grade 3-4 CRS and ICANS were 7.4% and 31.4%, respectively. For patients with DLBCL/tFL, the 1-year NRM, relapse, PFS, and OS were 11.6%, 40.8%, 47.6%, and 61.2%, respectively. We conclude that CAR T is feasible and effective in patients 80 years or older with B cell lymphomas. These patients must be provided the opportunity to be evaluated for this curative approach.
Collapse
Affiliation(s)
- Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, FL, USA
| | | | - Neela Easwar
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | | | - Madiha Iqbal
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, FL, USA
| | | | - Jean Yared
- Transplant and Cellular Therapy Program, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Nausheen Ahmed
- Hematologic Malignancies and Cellular Therapeutics, Department of Medicine, University of Kansas Cancer Center, Westwood, KS, USA
| | - Mehdi Hamadani
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Amer Beitinjaneh
- Division of Transplantation and Cellular Therapy, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mayur Narkhede
- Division of Hematology/Oncology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Praveen Ramakrishnan Geethakumari
- Section of Hematologic Malignancies and Cellular Therapy, Division of Hematology and Oncology, at Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bhagirathbhai Dholaria
- Division of Hematology Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Usama Gergis
- Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Jose Sandoval-Sus
- Moffitt Cancer Center at Memorial Health Care system, Pembroke Pines, FL, USA
| | | | - Joshua Fein
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | | | - Ernesto Ayala
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, FL, USA
| | - Kaitlin Annunzio
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Aaron P Rapoport
- Transplant and Cellular Therapy Program, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Forat Lutfi
- Hematologic Malignancies and Cellular Therapeutics, Department of Medicine, University of Kansas Cancer Center, Westwood, KS, USA
| | - Othman Salim Akhtar
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Lazaros Lekakis
- Division of Transplantation and Cellular Therapy, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Amitkumar Mehta
- Division of Hematology/Oncology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Olalekan O Oluwole
- Division of Hematology Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jennifer Logue
- Moffitt Cancer Center at Memorial Health Care system, Pembroke Pines, FL, USA
| | | | - Tsiporah Shore
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | | | - Muhamad Alhaj Moustafa
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, FL, USA
| | - Joseph McGuirk
- Hematologic Malignancies and Cellular Therapeutics, Department of Medicine, University of Kansas Cancer Center, Westwood, KS, USA
| | - Yi Lin
- Mayo Clinic, Rochester, MN, USA
| | - Samuel Yamshon
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | | |
Collapse
|
12
|
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
|
13
|
Mulvey A, Trueb L, Coukos G, Arber C. Novel strategies to manage CAR-T cell toxicity. Nat Rev Drug Discov 2025:10.1038/s41573-024-01100-5. [PMID: 39901030 DOI: 10.1038/s41573-024-01100-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2024] [Indexed: 02/05/2025]
Abstract
The immune-related adverse events associated with chimeric antigen receptor (CAR)-T cell therapy result in substantial morbidity as well as considerable cost to the health-care system, and can limit the use of these treatments. Current therapeutic strategies to manage immune-related adverse events include interleukin-6 receptor (IL-6R) blockade and corticosteroids. However, because these interventions do not always address the side effects, nor prevent progression to higher grades of adverse events, new approaches are needed. A deeper understanding of the cell types involved, and their associated signalling pathways, cellular metabolism and differentiation states, should provide the basis for alternative strategies. To preserve treatment efficacy, cytokine-mediated toxicity needs to be uncoupled from CAR-T cell function, expansion, long-term persistence and memory formation. This may be achieved by targeting CAR or independent cytokine signalling axes transiently, and through novel T cell engineering strategies, such as low-affinity CAR-T cells, reversible on-off switches and versatile adaptor systems. We summarize the current management of cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome, and review T cell- and myeloid cell-intrinsic druggable targets and cellular engineering strategies to develop safer CAR-T cells.
Collapse
Affiliation(s)
- Arthur Mulvey
- Department of Oncology UNIL-CHUV, Service of Immuno-Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Lionel Trueb
- Department of Oncology UNIL-CHUV, Service of Immuno-Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - George Coukos
- Department of Oncology UNIL-CHUV, Service of Immuno-Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Caroline Arber
- Department of Oncology UNIL-CHUV, Service of Immuno-Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland.
- Departments of Oncology UNIL-CHUV and Laboratory Medicine and Pathology, Service and Central Laboratory of Hematology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
| |
Collapse
|
14
|
Mougiakakos D, Sengupta R, Gold R, Schroers R, Haghikia A, Lorente M, Pendleton M, Register A, Heesen C, Kröger N, Schett G, Mackensen A, Podoll A, Gutman J, Furie R, Bayer R, Distler JHW, Dietrich S, Krönke G, Bullinger L, Walker K. Successful generation of fully human, second generation, anti-CD19 CAR T cells for clinical use in patients with diverse autoimmune disorders. Cytotherapy 2025; 27:236-246. [PMID: 39530971 DOI: 10.1016/j.jcyt.2024.09.008] [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: 06/27/2024] [Revised: 09/19/2024] [Accepted: 09/29/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND B-cell targeting chimeric antigen receptor (CAR) T-cell therapies, which lead to profound B-cell depletion, have been well-established in hematology-oncology. This deep B-cell depletion mechanism has prompted the exploration of their use in B-cell driven autoimmune diseases. We herein report on the manufacturing of KYV-101, a fully human anti-CD19 CAR T-cell therapy, derived from patients who were treated across a spectrum of autoimmune diseases. METHODS KYV-101 was manufactured from peripheral blood-derived mononuclear cells of 20 patients across seven autoimmune disease types (neurological autoimmune diseases, n = 13; rheumatological autoimmune diseases, n = 7). Patients ranged from 18 to 75 years of age. Duration of disease ranged from <1 to 23 years since diagnosis. Patients were heavily pretreated, and most were refractory to prior immunosuppressive treatments. Apheresis was collected across nine sites, cryopreserved, and shipped to the manufacturing facility. Healthy donor apheresis samples were collected for manufacturing comparison. Manufacturing was performed using the CliniMACS Prodigy system. Cells were enriched for CD4+/CD8+ T cells, transduced with a third generation lentiviral vector encoding the CAR, expanded in vitro, and harvested. Percent cell viability, T-cell purity, cellular expansion, and transduction efficiency were assessed. Activity was assessed using cytokine release assays for KYV-101 CAR T cells co-cultured with different CD19+/- target cell lines. RESULTS KYV-101 was successfully manufactured for 100% of patients. Transduced cell populations were highly viable, with expansion ranging from 11 to 66 fold at Day 8, and were comparable across disease types. Healthy donor-derived controls displayed similar expansion ranges. High CAR expression and transduction rates were observed, ranging between 37 and 77% with low variation in transgene copy number (two to four per cell). Cell viability of the final KYV-101 drug product ranged from 87 to 97%. KYV-101 displayed robust CD19-dependent and effector dose-related release of the pro-inflammatory cytokine IFN-γ. CONCLUSIONS KYV-101 manufacturing yielded a CAR T-cell product with high viability and consistent composition and functionality, regardless of disease indication, pre-treatment, and heterogeneity of the incoming material. Cryopreservation of the apheresis and final drug product enabled widespread distribution. These results support the robustness of the manufacturing process for the fully human KYV-101 anti-CD19 CAR T-cell therapy drug product for patients across diverse autoimmune disease types.
Collapse
Affiliation(s)
- Dimitrios Mougiakakos
- Department of Hematology, Oncology, and Cell Therapy, Otto-von-Guericke University, Magdeburg, Germany
| | | | - Ralf Gold
- Department of Neurology, Ruhr University Bochum, Bochum, Germany
| | - Roland Schroers
- Department of Hematology and Oncology, Ruhr University Bochum, Bochum, Germany
| | - Aiden Haghikia
- Department of Neurology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Mario Lorente
- Kyverna Therapeutics, Inc, Emeryville, California, USA
| | | | - Ames Register
- Kyverna Therapeutics, Inc, Emeryville, California, USA
| | - Christoph Heesen
- Institute of Neuroimmunology and Multiple Sclerosis and Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicolaus Kröger
- Department for Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5 - Hematology and Oncology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Amber Podoll
- Division of Renal Diseases and Hypertension, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jonathan Gutman
- Division of Hematology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Richard Furie
- Division of Rheumatology, Northwell Health, Great Neck, New York, USA
| | - Ruthee Bayer
- Department of Hematology/Oncology, Northwell Cancer Institute, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, New York, USA
| | - Jörg H W Distler
- Department of Rheumatology and Hiller Research Center, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sascha Dietrich
- Department of Hematology, Oncology and Immunology, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Gerhard Krönke
- Department of Rheumatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Karen Walker
- Kyverna Therapeutics, Inc, Emeryville, California, USA
| |
Collapse
|
15
|
Bloom MW, Vo JB, Rodgers JE, Ferrari AM, Nohria A, Deswal A, Cheng RK, Kittleson MM, Upshaw JN, Palaskas N, Blaes A, Brown SA, Ky B, Lenihan D, Maurer MS, Fadol A, Skurka K, Cambareri C, Chauhan C, Barac A. Cardio-Oncology and Heart Failure: a Scientific Statement From the Heart Failure Society of America. J Card Fail 2025; 31:415-455. [PMID: 39419165 DOI: 10.1016/j.cardfail.2024.08.045] [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: 04/16/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 10/19/2024]
Abstract
Heart failure and cancer remain 2 of the leading causes of morbidity and mortality, and the 2 disease entities are linked in a complex manner. Patients with cancer are at increased risk of cardiovascular complications related to the cancer therapies. The presence of cardiomyopathy or heart failure in a patient with new cancer diagnosis portends a high risk for adverse oncology and cardiovascular outcomes. With the rapid growth of cancer therapies, many of which interfere with cardiovascular homeostasis, heart failure practitioners need to be familiar with prevention, risk stratification, diagnosis, and management strategies in cardio-oncology. This Heart Failure Society of America statement addresses the complexities of heart failure care among patients with active cancer diagnoses and cancer survivors. Risk stratification, monitoring and management of cardiotoxicity are presented across stages A through D heart failure, with focused discussion on heart failure with preserved ejection fraction and special populations, such as survivors of childhood and young-adulthood cancers. We provide an overview of the shared risk factors between cancer and heart failure, highlighting heart failure as a form of cardiotoxicity associated with many different cancer therapeutics. Finally, we discuss disparities in the care of patients with cancer and cardiac disease and present a framework for a multidisciplinary-team approach and critical collaboration among heart failure, oncology, palliative care, pharmacy, and nursing teams in the management of these complex patients.
Collapse
Affiliation(s)
| | - Jacqueline B Vo
- Radiation Epidemiology Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, Bethesda, MD
| | - Jo E Rodgers
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, NC
| | - Alana M Ferrari
- Division of Hematology/ Oncology, University of Virginia Health, Charlottesville, VA
| | - Anju Nohria
- Cardio-Oncology Program, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Richard K Cheng
- Division of Cardiology, University of Washington, Seattle, WA
| | - Michelle M Kittleson
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | - Nicolas Palaskas
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anne Blaes
- Division of Hematology/Oncology/Transplantation, University of Minnesota, Minneapolis, MN
| | - Sherry-Ann Brown
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI; Research Collaborator, Mayo Clinic, Rochester, MN
| | - Bonnie Ky
- Division of Cardiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Thalheimer Center for Cardio-Oncology, Abramson Cancer Center and Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Daniel Lenihan
- Saint Francis Healthcare, Cape Girardeau, MO and the International Cardio-Oncology Society, Tampa, FL
| | - Mathew S Maurer
- Division of Cardiology, Columbia University Irving Medical Center, New York, NY
| | | | | | - Christine Cambareri
- Clinical Oncology Pharmacist, Hospital of the University of Pennsylvania, Abramson Cancer Center, Philadelphia, PA
| | | | - Ana Barac
- Department of Cardiology, Inova Schar Heart and Vascular, Inova Schar Cancer, Falls Church, VA
| |
Collapse
|
16
|
Sutherland NM, Zhou B, Zhang L, Ong MS, Hong JS, Pak A, Liu KJ, Frigault MJ, Maus MV, Hill JA, Reynolds K, Walter JE, Camargo CA, Barmettler S. Association of CD19 +-targeted chimeric antigen receptor (CAR) T-cell therapy with hypogammaglobulinemia, infection, and mortality. J Allergy Clin Immunol 2025; 155:605-615. [PMID: 39505278 PMCID: PMC11805655 DOI: 10.1016/j.jaci.2024.10.021] [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: 06/03/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND CD19-targeted chimeric antigen receptor T-cell therapy (CAR-T therapy) has revolutionized the treatment of hematologic malignancies. As these cells target CD19+ receptors on B cells, there is the potential for B-cell aplasia and hypogammaglobulinemia. Data on the degree and clinical significance of hypogammaglobulinemia are sparse. OBJECTIVES We sought to evaluate hypogammaglobulinemia after CD19-targeted CAR-T therapy and risk factors for hypogammaglobulinemia, infections, and mortality. METHODS We performed a retrospective evaluation of 579 patients receiving CD19-directed CAR-T therapy and evaluated demographics, hypogammaglobulinemia (IgG ≤600 mg/dL), infections prior to and after CAR-T therapy, and risk factors for hypogammaglobulinemia, infection, hospitalizations, and mortality. RESULTS Patients had a mean age of 64 years and 64% were male. Prior to CAR-T therapy, 60% of patients had hypogammaglobulinemia, which increased to 91% post-CAR-T therapy. Mean IgG levels decreased from pre- to post-CAR-T therapy levels (587 to 362 mg/dL; P < .0001). Thirty-seven percent of patients developed a serious infection post-CAR-T therapy. Hypogammaglobulinemia prior to CAR-T therapy was associated with worsening hypogammaglobulinemia after CAR-T therapy. Hypogammaglobulinemia post CAR-T therapy was associated with an increased risk of serious infection following CAR-T therapy (incidence rate ratio: 2.7; 95% CI: 1.5-5.2; P = .002). Risk factors for mortality included mild hypogammaglobulinemia (400 mg/dL < IgG ≤ 600 mg/dL), infections ≤100 days post-CAR-T therapy, and hospitalizations for infections. Immunoglobulin replacement was associated with a decreased risk of mortality. CONCLUSIONS We identified ∼90% of patients with hypogammaglobulinemia after CAR-T therapy. Hypogammaglobulinemia before CAR-T therapy was strongly predictive of worsening hypogammaglobulinemia after CAR-T therapy, which was associated with an increased risk of serious infection and mortality post CAR-T therapy. Increased immunological monitoring is needed to identify high-risk patients who may benefit from interventions to decrease morbidity and mortality.
Collapse
Affiliation(s)
| | - Baijun Zhou
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass
| | - Lingxiao Zhang
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass
| | - Mei-Sing Ong
- Department of Population Medicine, Harvard Medical School, Boston, Mass; Harvard Pilgrim Health Care Institute, Boston, Mass
| | - Joseph S Hong
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass
| | - Andrew Pak
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass
| | - Katherine J Liu
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass
| | - Matthew J Frigault
- Harvard Medical School, Boston, Mass; Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Boston, Mass
| | - Marcela V Maus
- Harvard Medical School, Boston, Mass; Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, Mass
| | - Joshua A Hill
- Department of Medicine, University of Washington, Seattle, Wash; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Wash; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Wash; Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, Wash
| | - Kerry Reynolds
- Harvard Medical School, Boston, Mass; Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Boston, Mass
| | - Jolan E Walter
- Department of Pediatrics, University of South Florida, St Petersburg, Fla; Department of Medicine, University of South Florida, St Petersburg, Fla; Division of Pediatric Allergy/Immunology, Johns Hopkins-All Children's Hospital, St Petersburg, Fla; Division of Pediatric Allergy Immunology, Massachusetts General Hospital, Boston, Mass
| | - Carlos A Camargo
- Harvard Medical School, Boston, Mass; Department of Emergency Medicine, Massachusetts General Hospital, Boston, Mass
| | - Sara Barmettler
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass; Harvard Medical School, Boston, Mass.
| |
Collapse
|
17
|
Passweg JR, Baldomero H, Alexander T, Angelucci E, Averbuch D, Bazarbachi A, Ciceri F, Raffaella G, Hazenberg MD, Kalwak K, McLornan DP, Risitano AM, Ruggeri A, Snowden JA, Sureda A. Utilization of hematopoietic cell transplantation and cellular therapy technology in Europe and associated Countries. Using the 2022 activity survey data to correlate with economic and demographic factors. A report from the EBMT. Bone Marrow Transplant 2025; 60:227-236. [PMID: 39578528 PMCID: PMC11810786 DOI: 10.1038/s41409-024-02459-0] [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] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/24/2024]
Abstract
We looked at treatment rates and center density across countries for patients treated in 2022; 46,143 HCTs (19,011 (41.2%) allogeneic, 27,132 (58.8%) autologous) reported by 689 centers. 4329 patients received advanced cellular therapies, 3205 were CAR-T. We found considerable differences in utilization of autologous, allogeneic HCT and more so for CAR-T. Differences in procedure type and for allogeneic HCT in donor use and disease indication are highlighted. For instance, countries with the highest use of unrelated donors per 10 million inhabitants were Germany (297) and the Netherlands (230), for identical sibling HCT it was Israel (148) and Lebanon (113), for haploidentical it was Israel (94) and Italy (94) and for cord blood it was the Netherlands (24) and the United Kingdom (15). We looked at HCT use for specific indications in allogeneic HCT (AML CR1, MDS, MPN and BMF). We correlated treatment rates with GNI and with demographic age structure and show correlations in HCT and CAR-T use and center density, highest in Italy for allogeneic and autologous HCT and in Switzerland for CAR-T. Resource restricted countries tend to concentrate HCT use in a limited number of centers. These data are useful for comparisons across countries.
Collapse
Affiliation(s)
- Jakob R Passweg
- Hematology Division, EBMT Activity Survey Office, University Hospital, Basel, Switzerland
| | - Helen Baldomero
- Hematology Division, EBMT Activity Survey Office, University Hospital, Basel, Switzerland.
| | - Tobias Alexander
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Emanuele Angelucci
- Hematology and Cellular Therapy Unit. IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Dina Averbuch
- Faculty of Medicine, Hebrew University of Jerusalem; Hadassah Medical Center, Jerusalem, Israel
| | - Ali Bazarbachi
- Department of Internal Medicine, Bone Marrow Transplantation Program, American University of Beirut, Beirut, Lebanon
| | - Fabio Ciceri
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Greco Raffaella
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Mette D Hazenberg
- Department of Hematology, Amsterdam University Medical Centres, University of Amsterdam, and Department of Hematopoiesis, Sanquin Research, Amsterdam, The Netherlands
| | - Krzysztof Kalwak
- Clinical Department of Pediatric BMT, Hematology and Oncology, Wroclaw Medical University, Wroclaw, Poland
| | - Donal P McLornan
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Antonio M Risitano
- Hematology and Hematopoietic Transplant Unit, Azienda Ospedaliera di Rilievo Nazionale "San Giuseppe Moscati" (A.O.R.N. Giuseppe Moscati), Avellino, Italy
| | - Annalisa Ruggeri
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Anna Sureda
- Clinical Hematology Department, Institut Català d'Oncologia-Hospitalet, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), University of Barcelona, Barcelona, Spain
| |
Collapse
|
18
|
Ow KV. CAR T-Cell Therapy Unveiled: Navigating Beyond CRS and ICANS to Address Delayed Complications and Optimize Management Strategies. J Adv Pract Oncol 2025; 16:1-15. [PMID: 39990042 PMCID: PMC11840332 DOI: 10.6004/jadpro.2025.16.7.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has ushered in a transformative era in the management of relapsed/refractory hematologic malignancies. The extensive phase II trials targeting relapsed/refractory non-Hodgkin lymphoma, including diverse subtypes such as diffuse large B-cell lymphoma, follicular lymphoma, and mantle cell lymphoma, along with multiple myeloma and B-cell acute lymphoblastic leukemia, have culminated in the endorsement of various CAR T-cell products for these specific indications by the US Food and Drug Administration. Although CAR T-cell therapy has achieved remarkable success, it is important to recognize that this innovative approach often gives rise to notable toxicities and is frequently associated with a distinctive pattern of adverse effects. Advanced practice providers, including advanced practice nurses and physician associates, involved in the care of these patients should be able to recognize these toxicities and be versed in treatment strategies to mitigate their impact.
Collapse
Affiliation(s)
- Karla V Ow
- From The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
19
|
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
|
20
|
Fleischer A, Kurth S, Duell J, Topp M, Strunz PP, Mersi J, Rasche L, Sanges C, Hudecek M, Einsele H, Maatouk I. Neuropsychiatric manifestations following chimeric antigen receptor T cell therapy for cancer: a systematic review of clinical outcomes and management strategies. J Immunother Cancer 2024; 12:e009174. [PMID: 39794934 PMCID: PMC11667355 DOI: 10.1136/jitc-2024-009174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 11/15/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-T cell therapy has emerged as a transformative modality in the treatment of patients with cancer. However, it is increasingly evident that this therapeutic approach is not without its challenges. The unique nature of CAR-T cells as living drugs introduces a distinct set of side effects. As the application of CAR-T cell therapy expands to treat a broader range of diseases, it becomes increasingly important to devise effective strategies for handling the associated toxicities. Challenges in treating patients with CAR-T cells include addressing complications such as cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and cytopenias. This comprehensive review seeks to systematically identify, categorize and elucidate all previously described neurological and psychological side effects associated with CAR-T cell therapy, shedding light on the pertinent laboratory findings that underscore these phenomena. METHODS PubMed, Springer Link, and ScienceDirect were systematically searched for empirical studies on adult patients with cancer receiving CAR-T cell therapy for hemato-oncological malignancies. Quality assessment was conducted using Version 2 of the Cochrane risk-of-bias tool (RoB 2) for randomized trials and adherence to the STROBE (Strengthening the Reporting of Observational Studies in Epidemiology) checklist for observational studies. The synthesis of findings was conducted via a narrative approach, consolidating the diverse array of data into a coherent framework. RESULTS From an initial pool of 2,276 citations, 546 studies met the inclusion criteria, exhibiting a rich tapestry of heterogeneity in terms of study characteristics and patient samples. The incidence of neuropsychological symptoms varied notably across different CAR-T cell products and hematological malignancies. Among the most frequently reported neuropsychological symptoms were aphasia, attention deficits, impaired consciousness, and disorientation, alongside a constellation of other symptoms including confusion, cognitive impairment, memory loss, writing difficulties, fatigue, headache, agitation, tremor, seizures, and psychomotor retardation. Early intervention strategies, including corticosteroids and tocilizumab, have shown the potential to reduce the intensity of neuropsychological symptoms and prevent their progression to critical complications. CONCLUSION These insights underscore the imperative of extending neuropsychological assessments beyond the conventional Immune Effector Cell-Associated Encephalopathy score framework.
Collapse
Affiliation(s)
- Anna Fleischer
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Sophia Kurth
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Johannes Duell
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Max Topp
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | | | - Julia Mersi
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Leo Rasche
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Carmen Sanges
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Michael Hudecek
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Imad Maatouk
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| |
Collapse
|
21
|
Luo Y, Gadd ME, Qie Y, Otamendi-Lopez A, Sanchez-Garavito JE, Brooks MM, Ulloa Navas MJ, Hundal T, Li S, Jones VK, Lou Y, Patel T, Dronca R, Kharfan-Dabaja MA, Dong H, Quinones-Hinojosa A, Qin H. Solid cancer-directed CAR T cell therapy that attacks both tumor and immunosuppressive cells via targeting PD-L1. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200891. [PMID: 39498357 PMCID: PMC11532918 DOI: 10.1016/j.omton.2024.200891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/31/2024] [Accepted: 10/03/2024] [Indexed: 11/07/2024]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has encountered limited success in solid tumors. The lack of dependable antigens and the immunosuppressive tumor microenvironment (TME) are major challenges. Within the TME, tumor cells along with immunosuppressive cells employ an immune-evasion mechanism that upregulates programmed death ligand 1 (PD-L1) to deactivate effector T cells; this makes PD-L1 a reliable, universal target for solid tumors. We developed a novel PD-L1 CAR (MC9999) using our humanized anti-PD-L1 monoclonal antibody, designed to simultaneously target tumor and immunosuppressive cells. The antigen-specific antitumor effects of MC9999 CAR T cells were observed consistently across four solid tumor models: breast cancer, lung cancer, melanoma, and glioblastoma multiforme (GBM). Notably, intravenous administration of MC9999 CAR T cells eradicated intracranially established LN229 GBM tumors, suggesting penetration of the blood-brain barrier. The proof-of-concept data demonstrate the cytolytic effect of MC9999 CAR T cells against immunosuppressive cells, including microglia HMC3 cells and M2 macrophages. Furthermore, MC9999 CAR T cells elicited cytotoxicity against primary tumor-associated macrophages within GBM tumors. The concept of targeting both tumor and immunosuppressive cells with MC9999 was further validated using CAR T cells derived from cancer patients. These findings establish MC9999 as a foundation for the development of effective CAR T cell therapies against solid tumors.
Collapse
Affiliation(s)
- Yan Luo
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Martha E. Gadd
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
| | - Yaqing Qie
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Mieu M. Brooks
- The Neurosurgery Department, Mayo Clinic, Jacksonville, FL, USA
| | | | - Tanya Hundal
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
| | - Shuhua Li
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
| | | | - Yanyan Lou
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Tushar Patel
- Hepatology & Liver Transplantation, Mayo Clinic, Jacksonville, FL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Roxana Dronca
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Mohamed A. Kharfan-Dabaja
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
- Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, FL, USA
| | - Haidong Dong
- Department of Urology, Mayo Clinic, Rochester, MN, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Alfredo Quinones-Hinojosa
- The Neurosurgery Department, Mayo Clinic, Jacksonville, FL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Hong Qin
- Regenerative Immunotherapy and CAR-T Translational Research Program, Mayo Clinic, Jacksonville, FL, USA
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
22
|
Bates SM, Evans KV, Delsing L, Wong R, Cornish G, Bahjat M. Immune safety challenges facing the preclinical assessment and clinical progression of cell therapies. Drug Discov Today 2024; 29:104239. [PMID: 39521331 DOI: 10.1016/j.drudis.2024.104239] [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: 04/08/2024] [Revised: 10/15/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
The promise of curative outcomes for life-limiting diseases using cell therapies is starting to become a reality, not only for patients with end-stage cancer, but also increasingly for regenerative therapies, including dentistry, ocular, neurodegenerative, and cardiac diseases. The introduction of often genetically modified cells into a patient can come with an extensive range of safety considerations. From an immune perspective, cell-based therapies carry inherent consequences and consideration of factors, such as the cell source (donor-derived autologous cells versus allogeneic cells), the intrinsic cellular nature of the therapy, and engineering/manufacturing methods, all of which influence the likelihood of inducing unwanted immune responses. Here, we provide an overview of the potential immune safety risks associated with cell therapies and explore possible mitigation approaches.
Collapse
Affiliation(s)
- Stephanie M Bates
- Safety Innovation, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Kelly V Evans
- Safety Innovation, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Louise Delsing
- Cell and Gene Therapy Safety, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ryan Wong
- Cell and Gene Therapy Safety, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Georgina Cornish
- Oncology Safety, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Mahnoush Bahjat
- Safety Innovation, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
23
|
Greco R, Alexander T, Yakoub-Agha I, Farge D. In response to the letter by Howard JF and colleagues (eclinm-D-24-00922). EClinicalMedicine 2024; 78:102917. [PMID: 39640931 PMCID: PMC11617733 DOI: 10.1016/j.eclinm.2024.102917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/18/2024] [Indexed: 12/07/2024] Open
Affiliation(s)
- Raffaella Greco
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Tobias Alexander
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Berlin, Germany
| | - Ibrahim Yakoub-Agha
- CHU de Lille, Univ Lille, INSERM U1286, Infinite, 59000, Lille, France
- Chair of the Practice Harmonization and Guidelines Committee of EBMT, Barcelona, Spain
| | - Dominique Farge
- Internal Medicine Unit (04): CRMR MATHEC, Maladies Auto-immunes et Thérapie Cellulaire, Centre de Référence des Maladies auto-immunes Systémiques Rares d’Ile-de-France, AP-HP, St-Louis Hospital Paris-Cite University France and Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
24
|
Bennett R, Dickinson M. SOHO State of the Art Updates and Next Questions | Current Evidence and Future Directions for Bispecific Antibodies in Large B-Cell Lymphoma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:809-820. [PMID: 38871556 DOI: 10.1016/j.clml.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 06/15/2024]
Abstract
The CD20xCD3 bispecific antibodies (bsAb) are "off-the-shelf" T-cell re-directing therapies that demonstrate remarkable single-agent clinical activity in B-cell lymphomas. Two agents, epcoritamab (epcor) and glofitamab (glofit) have recent global approvals for patients with relapsed/refractory DLBCL (RR DLBCL) following 2 prior treatment lines. Both agents demonstrate activity in patients with prior exposure to chimeric antigen receptor T-cell (CAR-T) treatment. As multiyear follow-up data become available, it is clear that the majority of patients achieving complete remissions do not relapse and that outcomes are similar between epcor and glofit. CD20xCD3 bsAb have a safety profile that reflect their mechanism of action, with cytokine release syndrome (CRS) the key management issue. Neurotoxicity is far less common than observed with CD19-directed CAR-T. BsAbs are attractive, rapidly available, treatment options for patients with RR DLBCL, without the practical and financial challenges seen with autologous CAR-T therapies. Recent data also demonstrate the feasibility and potential efficacy of bsAb in combination with chemoimmunotherapy with large randomized trials evaluating bsAb-chemotherapy combinations underway. There are open questions about the future role of bsAB for LBCL, the optimal duration of therapy, optimal CRS risk mitigation strategies, and potential resistance mechanisms. In this review we seek to describe the current evidence for bsAb in LBCL, and offer opinion regarding these open questions.
Collapse
Affiliation(s)
- Rory Bennett
- Department of Clinical Haematology, Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Victoria, Australia
| | - Michael Dickinson
- Department of Clinical Haematology, Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia.
| |
Collapse
|
25
|
Lancman G, Song K, White D, Crosbie T, Sharif I, Emond M, Saleem Raza M, Elias M, Kaedbey R, Chu MP. Recommendations for the effective use of T-cell-redirecting therapies: a Canadian consensus statement. Front Oncol 2024; 14:1446995. [PMID: 39659785 PMCID: PMC11628543 DOI: 10.3389/fonc.2024.1446995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 10/21/2024] [Indexed: 12/12/2024] Open
Abstract
Background T-cell-redirecting therapies, such as bispecific antibodies and chimeric antigen receptor T-cells, exploit the cytotoxic capabilities of the immune system to destroy cells expressing specific surface antigens, including malignant cells. These therapies have demonstrated unprecedented rates, depth, and duration of responses in relapsed and refractory multiple myeloma. However, there are significant challenges in implementing these therapies into practice, which require multidisciplinary and multicenter coordination and significant healthcare resources to effectively manage these patients. So far, there are no Canadian guidelines for the effective implementation and use of T-cell-redirecting therapies. Methods This consensus statement was developed based on three advisory meetings held in March, July, and November 2023. During these meetings, a panel of Canadian subject matter experts and representation from Myeloma Canada gathered to discuss the optimal procedures for the use of T-cell-redirecting therapies in the treatment of multiple myeloma. Members of the panel performed a thorough review of randomized clinical trials, real-world data, and other current literature, and provided their up-to-date clinical experience with T-cell-redirecting therapies in Canadian practice settings. Subsequently, asynchronous working groups were appointed to develop unified criteria for patient selection, appraise referral pathways, and devise strategies for management of short-term and long-term adverse events arising from the use of T-cell-redirecting therapies in multiple myeloma. Results Here, we present recommendations for optimizing patient selection, referral pathways, and adverse event management in the Canadian practice setting. These recommendations are relevant for hematologists/oncologists, oncology nurses, pharmacists, nurse practitioners, physician assistants, and other providers who treat patients with multiple myeloma, as well as individuals with multiple myeloma and their care partners. These recommendations will be of interest to clinicians who treat patients with MM at community clinics and hospitals and who may be interested in referring patients for T-cell-redirecting therapy.
Collapse
Affiliation(s)
- Guido Lancman
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Kevin Song
- The Leukemia/Bone Marrow Transplant Program of BC, BC Cancer Agency, Vancouver General Hospital, Vancouver, BC, Canada
| | - Darrell White
- Queen Elizabeth II Health Sciences Centre and Dalhousie University, Halifax, NS, Canada
| | - Tina Crosbie
- Pharmacy Department, The Ottawa Hospital, Ottawa, ON, Canada
| | | | - Marianne Emond
- Pharmacy Department, Centre Hospitalier Universitaire de Québec-Université Laval, Quebec City, QC, Canada
| | | | | | - Rayan Kaedbey
- Segal Cancer Centre, Jewish General Hospital, McGill University, Montréal, QC, Canada
| | | |
Collapse
|
26
|
Yang Y, Peng H, Wang J, Li F. New insights into CAR T-cell hematological toxicities: manifestations, mechanisms, and effective management strategies. Exp Hematol Oncol 2024; 13:110. [PMID: 39521987 PMCID: PMC11549815 DOI: 10.1186/s40164-024-00573-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [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
|
27
|
Rivetti di Val Cervo P, Alessi E, Lastella M, La Greca A, Trotta F. Advanced Therapy Medicinal Products: Availability, Access and Expenditure in Italy. BioDrugs 2024; 38:831-844. [PMID: 39404995 PMCID: PMC11530470 DOI: 10.1007/s40259-024-00683-0] [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: 09/16/2024] [Indexed: 11/03/2024]
Abstract
BACKGROUND Advanced therapy medicinal products (ATMPs) are an innovative output of biomedical research, characterized by a high level of uncertainty on long-term efficacy and safety, elevated price tags and often complex administration. All these elements compounded make their European authorization, national price negotiation for reimbursement and subsequent dispensation and administration to the patient less straightforward and often less successful than for less innovative drugs. To assess if these hurdles have affected patient access and how are ATMPs used in Italy, we have analysed availability, access and expenditure of ATMPs in the period spanning from 2016 to 2023. METHODS We have analysed real world data on the duration of ATMP regulatory evaluations for authorisation and reimbursement, time to first patient access and expenditure for ATMPs through the Italian National Health System (INHS) expenditure data flow, as well as information on patient mobility and availability of health facilities specialized in administering ATMPs. FINDINGS Of the 18 ATMPs currently authorized in Europe, 9 are reimbursed by the INHS, but only 6 were actually used, generating a cumulative expenditure of roughly 300 Mln€ from 2016 to 2023, largely owing to CAR-T therapies. Time to patient access reaches an average of 340.6 days from the day publication in the official Gazette of the reimbursement decision to first patient treatment in one of the 107 health facilities authorized for ATMP administration, after an even longer evaluation time by regulatory agencies. CONCLUSION Since the first reimbursement decision for an ATMP in Italy, back in 2016, these innovative drugs became progressively more and more available, both in terms of numbers and in terms of coverage across the country. Almost all Italian regions have at least one centre for ATMP administration and has performed a treatment in 2023. Notwithstanding their high per-treatment prices, ATMPs currently have a rather contained expenditure, however it is bound to keep growing in the next few years.
Collapse
Affiliation(s)
- Pia Rivetti di Val Cervo
- HTA and Pharmaceutical Economics Department, Italian Medicines Agency, Via del Tritone 181, 00187, Rome, Italy.
| | - Eva Alessi
- HTA and Pharmaceutical Economics Department, Italian Medicines Agency, Via del Tritone 181, 00187, Rome, Italy
| | - Marilena Lastella
- HTA and Pharmaceutical Economics Department, Italian Medicines Agency, Via del Tritone 181, 00187, Rome, Italy
| | - Antonio La Greca
- HTA and Pharmaceutical Economics Department, Italian Medicines Agency, Via del Tritone 181, 00187, Rome, Italy
| | - Francesco Trotta
- HTA and Pharmaceutical Economics Department, Italian Medicines Agency, Via del Tritone 181, 00187, Rome, Italy
| |
Collapse
|
28
|
Dandoy CE, Adams J, Artz A, Bredeson C, Dahi PB, Dodd T, Jaglowski S, Lehmann L, LeMaistre CF, Mian A, Neal A, Page K, Rizzo JD, Rotz S, Sorror M, Steinberg A, Viswabandya A, Howard DS. In Pursuit of Optimal Outcomes: A Framework for Quality Standards in Immune Effector Cell Therapy. Transplant Cell Ther 2024; 30:942-954. [PMID: 39067790 DOI: 10.1016/j.jtct.2024.07.011] [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: 05/18/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/30/2024]
Abstract
Immune effector cell (IEC) therapy represents a transformative advancement in oncology, leveraging the immune system to combat various malignancies. This article outlines a comprehensive framework for establishing and maintaining quality standards in IEC therapy amidst rapid scientific and clinical advancements. We emphasize the integration of structured process measures, robust quality assurance, and meticulous outcome evaluation to ensure treatment efficacy and safety. Key components include multidisciplinary expertise, stringent accreditation protocols, and advanced data management systems, which facilitate standardized reporting and continual innovation. The collaborative effort among stakeholders-ranging from patients and healthcare providers to regulatory bodies-is crucial in delivering high-quality IEC therapies. This framework aims to enhance patient outcomes and cement the role of IEC therapy as a cornerstone of modern oncology, promoting continuous improvement and adherence to high standards across the therapeutic spectrum.
Collapse
Affiliation(s)
- Christopher E Dandoy
- Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, Ohio.
| | - Joan Adams
- Stephenson Cancer Center, OU Health Science Center The University of Oklahoma, Oklahoma City, Oklahoma
| | - Andrew Artz
- Division of Leukemia, Department of Hematology and HCT, City of Hope, Duarte, California
| | - Christopher Bredeson
- Ottawa Hospital Research Institute, Division of Hematology, University of Ottawa, Ottawa, Canada
| | - Parastoo B Dahi
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Therese Dodd
- Sarah Cannon Transplant and Cellular Therapy Network, Nashville, Tennessee
| | - Samantha Jaglowski
- Department of Pediatrics and Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Leslie Lehmann
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Division of Hematology/Oncology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Amir Mian
- Department of Pediatric Hematology and Oncology, Department of Pediatrics at Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Alison Neal
- Department of Bone Marrow Transplant and Cellular Therapy, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Kristen Page
- Department of Pediatrics and Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - J Douglas Rizzo
- Department of Pediatrics and Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Seth Rotz
- Division of Pediatric Hematology, Oncology, and Blood and Marrow Transplantation, Cleveland Clinic, Cleveland, Ohio
| | - Mohamed Sorror
- Fred Hutchinson Cancer Center and University of Washington, Seattle, Washington
| | - Amir Steinberg
- Adult Stem Cell Transplantation, Westchester Medical Center, New York Medical College, Valhalla, New York
| | - Auro Viswabandya
- Department of Haematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Dianna S Howard
- Department of Internal Medicine, Section of Hematology and Oncology, Stem Cell Transplant and Cellular Therapy Program, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston Salem, North Carolina
| |
Collapse
|
29
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [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
|
30
|
Abdalhadi HM, Chatham WW, Alduraibi FK. CAR-T-Cell Therapy for Systemic Lupus Erythematosus: A Comprehensive Overview. Int J Mol Sci 2024; 25:10511. [PMID: 39408836 PMCID: PMC11476835 DOI: 10.3390/ijms251910511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disorder characterized by the production of autoreactive B and T cells and cytokines, leading to chronic inflammation affecting multiple organs. SLE is associated with significant complications that substantially increase morbidity and mortality. Given its complex pathogenesis, conventional treatments for SLE often have significant side effects and limited efficacy, necessitating the exploration of novel therapeutic strategies. One promising approach is the use of chimeric antigen receptor (CAR)-T-cell therapy, which has shown remarkable success in treating refractory hematological malignancies. This review provides a comprehensive analysis of the current use of CAR-T-cell therapy in SLE.
Collapse
Affiliation(s)
- Haneen M. Abdalhadi
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Walter W. Chatham
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Nevada, Las Vegas, NV 89102, USA;
| | - Fatima K. Alduraibi
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Department of Medicine, Division of Clinical Immunology and Rheumatology, Harvard Teaching Hospital, Boston, MA 02215, USA
- Department of Medicine, Division of Clinical Immunology and Rheumatology, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| |
Collapse
|
31
|
Beadnell TC, Jasti S, Wang R, Davis BH, Litwin V. Using Spectral Flow Cytometry for CAR T-Cell Clinical Trials: Game Changing Technologies Enabling Novel Therapies. Int J Mol Sci 2024; 25:10263. [PMID: 39408593 PMCID: PMC11476793 DOI: 10.3390/ijms251910263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/20/2024] Open
Abstract
Monitoring chimeric antigen redirected (CAR) T-cells post-infusion in clinical trials is a specialized application of flow cytometry. Unlike the CAR T-cell monitoring for individual patients conducted in clinical laboratories, the data generated during a clinical trial will be used not only to monitor the therapeutic response of a single patient, but determine the success of the therapy itself, or even of an entire class of therapeutic compounds. The data, typically acquired at multiple testing laboratories, will be compiled into a single database. The data may also be used for mathematical modeling of cellular kinetics or to identify predictive biomarkers. With the expanded context of use, a robust, standardized assay is mandatory in order to generate a valuable and reliable data set. Hence, the requirements for assay validation, traceable calibration, technology transfer, cross-instrument standardization and regulatory compliance are high.
Collapse
Affiliation(s)
| | - Susmita Jasti
- Eurofins Viracor Biopharma, Lenexa, KS 66219, USA; (T.C.B.); (S.J.)
| | - Ruqi Wang
- Eurofins Pharma Bioanalytical Services, St. Charles, MO 63304, USA;
| | | | - Virginia Litwin
- Eurofins Clinical Trial Solutions, Montreal, QC J2L 3N5, Canada
| |
Collapse
|
32
|
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
|
33
|
Guo Z, Ding Y, Wang M, Zhai Q, Liu J, Du Q. Comparing the Differences in Adverse Events among Chimeric Antigen Receptor T-Cell Therapies: A Real-World Pharmacovigilance Study. Pharmaceuticals (Basel) 2024; 17:1025. [PMID: 39204130 PMCID: PMC11359317 DOI: 10.3390/ph17081025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
In this study, we compared the similarities and differences in adverse events (AEs) among CAR T-cell products through signal mining via the FDA Adverse Event Reporting System (FAERS) and identified unknown AEs to provide a reference for safe clinical medication. Data from the FAERS database spanning from the fourth quarter of 2017 to the first quarter of 2024 were extracted. Signals were identified using the reporting odds ratio (ROR) method and the Medicines and Healthcare Products Regulatory Agency (MHRA) method. A total of 11,386 AE reports related to six CAR T-cell products were selected. The top three categories of AEs reported were nervous system disorders, immune system disorders, and general disorders and administration site conditions. However, there were variations in the AE spectra among the different CAR T-cell products. The BCMA-targeting drugs idecabtagene vicleucel (Ide-cel) and ciltacabtagene autoleucel (Cilta-cel) were found to be associated with parkinsonism, which were not observed in CD19-targeting drugs. Tisagenlecleucel (Tisa-cel) and axicabtagene ciloleucel (Axi-cel) exhibited cerebrovascular accident-related AEs, graft versus host disease, and abnormal coagulation indices. Cilta-cel was associated with cerebral hemorrhage, intracranial hemorrhage, cranial nerve disorder, and facial nerve disorder. Cardiopulmonary toxicity, including hypoxia, tachypnoea, cardiorenal syndrome, and hypotension, exhibited strong signal intensities and considerable overlap with CRS. The number of positive signals for cardiopulmonary toxicity associated with drugs targeting CD-19 is greater. Clinicians should assess patients prior to medication and closely monitor their vital signs, mental status, and laboratory parameters during treatment.
Collapse
Affiliation(s)
- Zihan Guo
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, China; (Z.G.); (Y.D.); (M.W.); (Q.Z.)
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yunlan Ding
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, China; (Z.G.); (Y.D.); (M.W.); (Q.Z.)
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Mengmeng Wang
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, China; (Z.G.); (Y.D.); (M.W.); (Q.Z.)
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Qing Zhai
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, China; (Z.G.); (Y.D.); (M.W.); (Q.Z.)
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jiyong Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, China; (Z.G.); (Y.D.); (M.W.); (Q.Z.)
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Qiong Du
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, China; (Z.G.); (Y.D.); (M.W.); (Q.Z.)
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
34
|
Gong IY, Tran D, Saibil S, Laister RC, Kuruvilla J. Biomarkers of outcome in patients undergoing CD19 CAR-T therapy for large B cell lymphoma. Hemasphere 2024; 8:e130. [PMID: 39175824 PMCID: PMC11339649 DOI: 10.1002/hem3.130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/12/2024] [Accepted: 06/26/2024] [Indexed: 08/24/2024] Open
Abstract
CD19-directed autologous chimeric antigen receptor T cell (CAR-T) therapy has transformed the management of relapsed/refractory (R/R) large B cell lymphoma (LBCL). Initially approved in the third line and beyond setting, CAR-T is now standard of care (SOC) for second-line treatment in patients with refractory disease or early relapse (progression within 12 months) following primary chemoimmunotherapy. Despite becoming SOC, most patients do not achieve complete response, and long-term cure is only observed in approximately 40% of patients. Accordingly, there is an urgent need to better understand the mechanisms of treatment failure and to identify patients that are unlikely to benefit from SOC CAR-T. The field needs robust biomarkers to predict treatment outcome, as better understanding of prognostic factors and mechanisms of resistance can inform on the design of novel treatment approaches for patients predicted to respond poorly to SOC CAR-T. This review aims to provide a comprehensive overview of clinical, molecular, imaging, and cellular features that have been shown to influence outcomes of CAR-T therapy in patients with R/R LBCL.
Collapse
Affiliation(s)
- Inna Y. Gong
- Princess Margaret Cancer CenterTorontoOntarioCanada
- Division of Medical Oncology and HematologyUniversity Health NetworkTorontoOntarioCanada
| | - Daisy Tran
- Princess Margaret Cancer CenterTorontoOntarioCanada
- Division of Medical Oncology and HematologyUniversity Health NetworkTorontoOntarioCanada
| | - Samuel Saibil
- Princess Margaret Cancer CenterTorontoOntarioCanada
- Division of Medical Oncology and HematologyUniversity Health NetworkTorontoOntarioCanada
- Department of ImmunologyUniversity of TorontoTorontoOntarioCanada
| | - Rob C. Laister
- Princess Margaret Cancer CenterTorontoOntarioCanada
- Division of Medical Oncology and HematologyUniversity Health NetworkTorontoOntarioCanada
| | - John Kuruvilla
- Princess Margaret Cancer CenterTorontoOntarioCanada
- Division of Medical Oncology and HematologyUniversity Health NetworkTorontoOntarioCanada
| |
Collapse
|
35
|
Wang M, Jia L, Dai X, Zhang X. Advanced strategies in improving the immunotherapeutic effect of CAR-T cell therapy. Mol Oncol 2024; 18:1821-1848. [PMID: 38456710 PMCID: PMC11306536 DOI: 10.1002/1878-0261.13621] [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: 08/12/2023] [Revised: 12/23/2023] [Accepted: 02/20/2024] [Indexed: 03/09/2024] Open
Abstract
Chimeric antigen receptor (CAR-T) cell therapy is a newly developed immunotherapy strategy and has achieved satisfactory outcomes in the treatment of hematological malignancies. However, some adverse effects related to CAR-T cell therapy have to be resolved before it is widely used in clinics as a cancer treatment. Furthermore, the application of CAR-T cell therapy in the treatment of solid tumors has been hampered by numerous limitations. Therefore, it is essential to explore novel strategies to improve the therapeutic effect of CAR-T cell therapy. In this review, we summarized the recently developed strategies aimed at optimizing the generation of CAR-T cells and improving the anti-tumor efficiency of CAR-T cell therapy. Furthermore, the discovery of new targets for CAR-T cell therapy and the combined treatment strategies of CAR-T cell therapy with chemotherapy, radiotherapy, cancer vaccines and nanomaterials are highlighted.
Collapse
Affiliation(s)
- Minmin Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationFirst Hospital of Jilin UniversityChangchunChina
- National‐Local Joint Engineering Laboratory of Animal Models for Human DiseaseFirst Hospital of Jilin UniversityChangchunChina
| | - Linzi Jia
- Department of General MedicineShanxi Province Cancer HospitalTaiyuanChina
| | - Xiangpeng Dai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationFirst Hospital of Jilin UniversityChangchunChina
- National‐Local Joint Engineering Laboratory of Animal Models for Human DiseaseFirst Hospital of Jilin UniversityChangchunChina
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationFirst Hospital of Jilin UniversityChangchunChina
- National‐Local Joint Engineering Laboratory of Animal Models for Human DiseaseFirst Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
36
|
Olejarz W, Sadowski K, Szulczyk D, Basak G. Advancements in Personalized CAR-T Therapy: Comprehensive Overview of Biomarkers and Therapeutic Targets in Hematological Malignancies. Int J Mol Sci 2024; 25:7743. [PMID: 39062986 PMCID: PMC11276786 DOI: 10.3390/ijms25147743] [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: 06/30/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy is a novel anticancer therapy using autologous or allogeneic T-cells. To date, six CAR-T therapies for specific B-cell acute lymphoblastic leukemia (B-ALL), non-Hodgkin lymphomas (NHL), and multiple myeloma (MM) have been approved by the Food and Drug Administration (FDA). Significant barriers to the effectiveness of CAR-T therapy include cytokine release syndrome (CRS), neurotoxicity in the case of Allogeneic Stem Cell Transplantation (Allo-SCT) graft-versus-host-disease (GVHD), antigen escape, modest antitumor activity, restricted trafficking, limited persistence, the immunosuppressive microenvironment, and senescence and exhaustion of CAR-Ts. Furthermore, cancer drug resistance remains a major problem in clinical practice. CAR-T therapy, in combination with checkpoint blockades and bispecific T-cell engagers (BiTEs) or other drugs, appears to be an appealing anticancer strategy. Many of these agents have shown impressive results, combining efficacy with tolerability. Biomarkers like extracellular vesicles (EVs), cell-free DNA (cfDNA), circulating tumor (ctDNA) and miRNAs may play an important role in toxicity, relapse assessment, and efficacy prediction, and can be implicated in clinical applications of CAR-T therapy and in establishing safe and efficacious personalized medicine. However, further research is required to fully comprehend the particular side effects of immunomodulation, to ascertain the best order and combination of this medication with conventional chemotherapy and targeted therapies, and to find reliable predictive biomarkers.
Collapse
Affiliation(s)
- Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Karol Sadowski
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Daniel Szulczyk
- Chair and Department of Biochemistry, The Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Grzegorz Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| |
Collapse
|
37
|
Fowler NH, Chavez JC, Riedell PA. Moving T-Cell Therapies into the Standard of Care for Patients with Relapsed or Refractory Follicular Lymphoma: A Review. Target Oncol 2024; 19:495-510. [PMID: 38896212 PMCID: PMC11271334 DOI: 10.1007/s11523-024-01070-z] [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] [Accepted: 05/06/2024] [Indexed: 06/21/2024]
Abstract
Patients with follicular lymphoma, an indolent form of non-Hodgkin lymphoma, typically experience multiple relapses over their disease course. Periods of remission become progressively shorter with worse clinical outcomes after each subsequent line of therapy. Currently, no clear standard of care/preferred treatment approach exists for patients with relapsed or refractory follicular lymphoma. As novel agents continue to emerge for treatment in the third-line setting, guidance is needed for selecting the most appropriate therapy for each patient. Several classes of targeted therapeutic agents, including monoclonal antibodies, phosphoinositide 3-kinase inhibitors, enhancer of zeste homolog 2 inhibitors, chimeric antigen receptor (CAR) T-cell therapies, and bispecific antibodies, have been approved by regulatory authorities based on clinical benefit in patients with relapsed or refractory follicular lymphoma. Additionally, antibody-drug conjugates and other immunocellular therapies are being evaluated in this setting. Effective integration of CAR-T cell therapy into the treatment paradigm after two or more prior therapies requires appropriate patient selection based on transformation status following a rebiopsy; a risk evaluation based on age, fitness, and remission length; and eligibility for CAR-T cell therapy. Consideration of important logistical factors (e.g., proximity to the treatment center and caregiver support during key periods of CAR-T cell therapy) is also critical. Overall, an individualized treatment plan that considers patient-related factors (e.g., age, disease status, tumor burden, comorbidities) and prior treatment types is recommended for patients with relapsed or refractory follicular lymphoma. Future analyses of real-world data and a better understanding of mechanisms of relapse are needed to further refine patient selection and identify optimal sequencing of therapies in this setting.
Collapse
Affiliation(s)
| | - Julio C Chavez
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Peter A Riedell
- David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL, USA
| |
Collapse
|
38
|
Cui C, Feng C, Rosenthal N, Wade SW, Curry L, Fu C, Shah GL. Hospital healthcare resource utilization and costs for chimeric antigen T-cell therapy and autologous hematopoietic cell transplant in patients with large B-cell lymphoma in the United States. Leuk Lymphoma 2024; 65:922-931. [PMID: 38567630 DOI: 10.1080/10428194.2024.2331084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
The efficacy of chimeric antigen receptor (CAR) T-cell therapy for large B-cell lymphoma (LBCL) is well-established. This study, using the Premier PINC AI Healthcare Database, assessed hospital costs and healthcare resource utilization (HRU) between CAR T-cell therapy and autologous hematopoietic cell transplant (AHCT) for 733 LBCL patients from 01/01/2017-04/30/2021 (166 CAR T and 567 AHCT from 37 US hospital systems. CAR T-cell therapy had higher index costs but lower non-pharmacy costs, shorter hospital stays, lower ICU utilization than AHCT. The CAR T-cell cohort also presented fewer preparatory costs and HRU. At a 180-day follow-up, AHCT had lower hospitalization rates and costs. Overall, despite higher index costs, CAR T-cell therapy has lower non-pharmacy costs and HRU during the index procedure and requires less preparation time with lower preparation HRUs and costs than AHCT. This has important implications for resource management and informed decision-making for stakeholders.
Collapse
Affiliation(s)
- Chendi Cui
- PINC AI Applied Sciences, Premier Inc, Charlotte, NC, USA
| | | | - Ning Rosenthal
- PINC AI Applied Sciences, Premier Inc, Charlotte, NC, USA
| | - Sally W Wade
- Wade Outcomes Research and Consulting, Salt Lake City, UT, USA
| | - Laura Curry
- PINC AI Applied Sciences, Premier Inc, Charlotte, NC, USA
| | | | - Gunjan L Shah
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
39
|
Tane M, Kosako H, Sonoki T, Hosoi H. TAFRO Syndrome and COVID-19. Biomedicines 2024; 12:1287. [PMID: 38927495 PMCID: PMC11200813 DOI: 10.3390/biomedicines12061287] [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: 04/30/2024] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
TAFRO syndrome is a systemic inflammatory disease characterized by thrombocytopenia and anasarca. It results from hyperinflammation and produces severe cytokine storms. Severe acute respiratory syndrome coronavirus 2, which led to the coronavirus disease 2019 (COVID-19) pandemic, also causes cytokine storms. COVID-19 was reported to be associated with various immune-related manifestations, including multisystem inflammatory syndrome, hemophagocytic syndrome, vasculitis, and immune thrombocytopenia. Although the pathogenesis and complications of COVID-19 have not been fully elucidated, the pathogeneses of excessive immunoreaction after COVID-19 and TAFRO syndrome both involve cytokine storms. Since the COVID-19 pandemic, there have been a few case reports about the onset of TAFRO syndrome after COVID-19 or COVID-19 vaccination. Castleman disease also presents with excessive cytokine production. We reviewed the literature about the association between TAFRO syndrome or Castleman disease and COVID-19 or vaccination against it. While the similarities and differences between the pathogeneses of TAFRO syndrome and COVID-19 have not been investigated previously, the cytokines and genetic factors associated with TAFRO syndrome and COVID-19 were reviewed by examining case reports. Investigation of TAFRO-like manifestations after COVID-19 or vaccination against COVID-19 may contribute to understanding the pathogenesis of TAFRO syndrome.
Collapse
Affiliation(s)
- Misato Tane
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama 641-8509, Japan; (M.T.)
- Department of Hematology, Kinan Hospital, Wakayama 646-8588, Japan
| | - Hideki Kosako
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama 641-8509, Japan; (M.T.)
- Department of Hematology, Kinan Hospital, Wakayama 646-8588, Japan
| | - Takashi Sonoki
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama 641-8509, Japan; (M.T.)
- Department of Transfusion Medicine, Wakayama Medical University Hospital, Wakayama 641-8510, Japan
| | - Hiroki Hosoi
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama 641-8509, Japan; (M.T.)
- Department of Transfusion Medicine, Wakayama Medical University Hospital, Wakayama 641-8510, Japan
| |
Collapse
|
40
|
Passweg JR, Baldomero H, Ciceri F, de la Cámara R, Glass B, Greco R, Hazenberg MD, Kalwak K, McLornan DP, Neven B, Perić Z, Risitano AM, Ruggeri A, Snowden JA, Sureda A. Hematopoietic cell transplantation and cellular therapies in Europe 2022. CAR-T activity continues to grow; transplant activity has slowed: a report from the EBMT. Bone Marrow Transplant 2024; 59:803-812. [PMID: 38438647 PMCID: PMC11161408 DOI: 10.1038/s41409-024-02248-9] [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: 02/01/2024] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 03/06/2024]
Abstract
In 2022, 46,143 HCT (19,011 (41.2%) allogeneic and 27,132 (58.8%) autologous) in 41,854 patients were reported by 689 European centers. 4329 patients received advanced cellular therapies, 3205 of which were CAR-T. An additional 2854 patients received DLI. Changes compared to the previous year were an increase in CAR-T treatments (+27%) and decrease in allogeneic (-4.0%) and autologous HCT (-1.7%). Main indications for allogeneic HCT were myeloid malignancies (10,433; 58.4%), lymphoid malignancies (4,674; 26.2%) and non-malignant disorders (2572; 14.4%). Main indications for autologous HCT were lymphomas (7897; 32.9%), PCD (13,694; 57.1%) and solid tumors (1593; 6.6%). In allogeneic HCT, use of sibling donors decreased by -7.7%, haploidentical donors by -6.3% and unrelated donors by -0.9%. Overall cord blood HCT decreased by -16.0%. Use of allogeneic, and to a lesser degree autologous HCT, decreased for lymphoid malignancies likely reflecting availability of new treatment modalities, including small molecules, bispecific antibodies, and CAR-T cells. Pediatric HCT activity remains stable (+0.3%) with differences between allogeneic and autologous HCT. Use of CAR-T continues to increase and reached a cumulative total of 9039 patients treated with wide differences across European countries. After many years of continuous growth, increase in application of HCT seems to have slowed down.
Collapse
Affiliation(s)
- Jakob R Passweg
- EBMT Activity Survey Office, Hematology Division, University Hospital, Basel, Switzerland
| | - Helen Baldomero
- EBMT Activity Survey Office, Hematology Division, University Hospital, Basel, Switzerland.
| | - Fabio Ciceri
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | | | - Bertram Glass
- Klinik für Hämatologie und Stammzelltransplantation, HELIOS Klinikum Berlin-Buch, Berlin, Germany
| | - Raffaella Greco
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Mette D Hazenberg
- Department of Hematology, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands
| | - Krzysztof Kalwak
- Clinical Department of Pediatric BMT, Hematology and Oncology, Wroclaw Medical University, Wroclaw, Poland
| | - Donal P McLornan
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Bénédicte Neven
- Pediatric immune-hematology unit, Necker Children Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Zinaida Perić
- School of Medicine, University of Zagreb, University Hospital Center Zagreb, Zagreb, Croatia
| | - Antonio M Risitano
- Hematology and Hematopoietic Transplant Unit, Azienda Ospedaliera di Rilievo Nazionale "San Giuseppe Moscati" (A.O.R.N. Giuseppe Moscati), Avellino, Italy
| | - Annalisa Ruggeri
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Anna Sureda
- Clinical Hematology Department, Institut Català d'Oncologia-Hospitalet, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), University of Barcelona, Barcelona, Spain
| |
Collapse
|
41
|
An J, Zhao J, Zou P, Zhang Y, Wei J, Tian W, Wei J. Infections associated with CAR-T cell therapy in patients with relapsed refractory multiple myeloma: Risks and prevention strategies. Cancer Med 2024; 13:e7372. [PMID: 38923216 PMCID: PMC11196838 DOI: 10.1002/cam4.7372] [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: 11/29/2023] [Revised: 05/13/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Chimeric antigen receptor T (CAR-T) cell therapy has emerged as a potent treatment for relapsed or refractory multiple myeloma, demonstrating significant clinical efficacy. Despite these advances, treatment-related toxicities, particularly infections, pose a significant challenge to patient safety. METHODS This review synthesizes current knowledge on the mechanisms underlying post-CAR-T therapy infections, focusing on the interplay between immune dysfunction, host factors, and treatment-induced toxicity. It provides a comprehensive analysis of the temporal and individual variability in infection characteristics and the confounding clinical presentation of cytokine release syndrome. RESULTS The review identifies that patients receiving CAR-T cells are at increased risk of concurrent infections due to the heterogeneity in infection characteristics across different time periods, individuals, and patient groups. It highlights the diagnostic and therapeutic complexities introduced by the overlapping symptoms of infection and cytokine release syndrome. CONCLUSION To enhance the infection control post-CAR-T therapy, this review proposes preventive strategies tailored to the early and long-term management of patients. It underscores the need for a nuanced understanding of infection mechanisms and the importance of personalized prevention plans to improve clinical outcomes in multiple myeloma treatment.
Collapse
Affiliation(s)
- Jing An
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical SciencesThird Hospital of Shanxi Medical University, Tongji Shanxi HospitalTaiyuanShanxiChina
- School of Public HealthShanxi Medical UniversityTaiyuanShanxiChina
| | - Jie Zhao
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical SciencesThird Hospital of Shanxi Medical University, Tongji Shanxi HospitalTaiyuanShanxiChina
- Sino‐German Joint Oncological Research LaboratoryShanxi Bethune Hospital, Shanxi Academy of Medical SciencesTaiyuanShanxiChina
| | - Ping Zou
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
- Immunotherapy Research Center for Hematologic Diseases of Hubei ProvinceWuhanHubeiChina
| | - Junni Wei
- School of Public HealthShanxi Medical UniversityTaiyuanShanxiChina
| | - Weiwei Tian
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical SciencesThird Hospital of Shanxi Medical University, Tongji Shanxi HospitalTaiyuanShanxiChina
- School of Public HealthShanxi Medical UniversityTaiyuanShanxiChina
- Sino‐German Joint Oncological Research LaboratoryShanxi Bethune Hospital, Shanxi Academy of Medical SciencesTaiyuanShanxiChina
| | - Jia Wei
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical SciencesThird Hospital of Shanxi Medical University, Tongji Shanxi HospitalTaiyuanShanxiChina
- Sino‐German Joint Oncological Research LaboratoryShanxi Bethune Hospital, Shanxi Academy of Medical SciencesTaiyuanShanxiChina
- Department of Hematology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
- Immunotherapy Research Center for Hematologic Diseases of Hubei ProvinceWuhanHubeiChina
| |
Collapse
|
42
|
Bishop MR, Kay GE. CAR T-cell therapy: A collaboration between authorized treatment centers and community oncologists. Semin Oncol 2024; 51:87-94. [PMID: 38531760 DOI: 10.1053/j.seminoncol.2024.02.001] [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: 10/05/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 03/28/2024]
Abstract
With the approval of the first CAR T-cell products for hematological malignancies in 2017, these autologous cell therapies have changed the treatment paradigm for patients with relapsed or refractory (r/r) non-Hodgkin lymphoma (NHL), who have a poor prognosis and few effective treatment options. Despite the demonstrated clinical benefit in patients with r/r diffuse large B-cell lymphoma, mantle cell lymphoma, and follicular lymphoma, many patients who are eligible for CAR T-cell therapies do not receive them or are treated with CAR T cells as a later line of therapy at advanced stages of disease. Several barriers exist for referring patients to an authorized treatment center (ATC) for CAR T-cell therapy. Although most patients with NHL are treated by community-based oncologists, educational gaps may exist for some community oncologists about the availability of CAR T-cell therapies in certain indications, the overall treatment process, and how they can access these therapies for their patients. In addition to navigation of the referral process from the community setting to the ATC, other barriers include timely identification of candidates eligible for CAR T-cell therapy and logistical and reimbursement concerns. Here, we examine the patient CAR T-cell experience, which begins and ends in the community setting, and identify and discuss opportunities for improved collaboration between community oncologists and ATC physicians to help address barriers to treatment and enhance patient outcomes. Treatment decisions for a patient's second or third line of therapy for NHL are critically important, owing to declining probabilities for favorable outcomes with each successive line of therapy. For patients who are eligible, CAR T-cell therapies should be considered as early as possible in their treatment course. A better understanding of the CAR T-cell process, the patient's experience, and the collaboration necessary for timely patient identification, better access, and successful outcomes will enable more patients to benefit from CAR T-cell therapies.
Collapse
Affiliation(s)
- Michael R Bishop
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL.
| | - Gary E Kay
- Northwest Oncology and Hematology, Rolling Meadows, IL
| |
Collapse
|
43
|
Kamel T, Boulain T. Pneumocystis pneumonia in French intensive care units in 2013-2019: mortality and immunocompromised conditions. Ann Intensive Care 2024; 14:80. [PMID: 38776012 PMCID: PMC11111439 DOI: 10.1186/s13613-024-01309-y] [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: 02/05/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
PURPOSE The recent epidemiology of Pneumocystis pneumonia (PCP) requiring intensive care unit (ICU) admission and the associated spectrum of immunocompromising conditions are poorly described. METHODS We analyzed all adult PCP cases admitted to French ICUs via the French medical database system (PMSI), over the period from 2013 to 2019. RESULTS French ICUs admitted a total of 4055 adult patients with PCP. Among all hospitalized PCP cases, the proportion requiring ICU admission increased from 17.8 in 2014 to 21.3% in 2019 (P < 0.001). The incidence of severe PCP rose from 0.85 in 2013 to 1.32/100,000 adult inhabitants in 2019 (P < 0.0001), primarily due to the proportion of HIV-negative patients that increased from 60.6% to 74.4% (P < 0.0001). Meanwhile, the annual number of severe PCP cases among patients with HIV infection remained stable over the years. In-hospital mortality of severe PCP cases was 28.5% in patients with HIV infection and 49.7% in patients without. Multivariable logistic analysis showed that patients with HIV infection had a lower adjusted risk of death than patients without HIV infection (Odds Ratio [OR]: 0.30, 95% confidence interval [95CI]: 0.17-0.55). Comorbidities or conditions strongly associated with hospital mortality included the patient's age, Simplified Acute Physiologic Score II, congestive heart failure, coagulopathy, solid organ cancer, and cirrhosis. A vast array of autoimmune inflammatory diseases affected 19.9% of HIV-negative patients. CONCLUSIONS The number of PCP cases requiring ICU admission in France has risen sharply. While the yearly count of severe PCP cases in HIV-infected patients has remained steady, this rise predominantly affects cancer patients, with a recent surge observed in patients with autoimmune inflammatory diseases, affecting one in five individuals.
Collapse
Affiliation(s)
- Toufik Kamel
- Service de Médecine Intensive Réanimation, Centre Hospitalier Universitaire d'Orléans, 14 Avenue de L'Hôpital CS 86709, 45067, Orleans Cedex 2, France
| | - Thierry Boulain
- Service de Médecine Intensive Réanimation, Centre Hospitalier Universitaire d'Orléans, 14 Avenue de L'Hôpital CS 86709, 45067, Orleans Cedex 2, France.
| |
Collapse
|
44
|
Lyu X, Gupta L, Tholouli E, Chinoy H. Chimeric antigen receptor T cell therapy: a new emerging landscape in autoimmune rheumatic diseases. Rheumatology (Oxford) 2024; 63:1206-1216. [PMID: 37982747 PMCID: PMC11065442 DOI: 10.1093/rheumatology/kead616] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/21/2023] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy, an innovative immune cell therapy, has revolutionized the treatment landscape of haematological malignancies. The past 2 years has witnessed the successful application of CD19-targeting CAR constructs in refractory cases of autoimmune rheumatic diseases, including systemic lupus erythematosus, systemic sclerosis and anti-synthetase syndrome. In comparison with existing B cell depletion therapies, targeting CD19 has demonstrated a more rapid and profound therapeutic effect, enabling drug-free remission with manageable adverse events. These promising results necessitate validation through long-term, large-sample randomized controlled studies. Corroborating the role of CAR-T therapy in refractory rheumatological disorders and affirming safety, efficacy and durability of responses are the aims of future clinical studies. Optimizing the engineering strategies and better patient selection are also critical to further refining the successful clinical implementation of CAR-T therapy.
Collapse
MESH Headings
- Humans
- Rheumatic Diseases/therapy
- Rheumatic Diseases/immunology
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/therapeutic use
- Autoimmune Diseases/therapy
- Autoimmune Diseases/immunology
- Immunotherapy, Adoptive/methods
- Antigens, CD19/immunology
- Antigens, CD19/therapeutic use
- Lupus Erythematosus, Systemic/therapy
- Lupus Erythematosus, Systemic/immunology
- Receptors, Antigen, T-Cell/therapeutic use
- Receptors, Antigen, T-Cell/immunology
- Scleroderma, Systemic/therapy
- Scleroderma, Systemic/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
Collapse
Affiliation(s)
- Xia Lyu
- Department of Rheumatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Epidemiology and Public Health Group, School of Health Sciences, The University of Manchester, Manchester, UK
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Latika Gupta
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Rheumatology, Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, UK
| | - Eleni Tholouli
- Department of Haematology, Manchester Royal Infirmary, Manchester, UK
| | - Hector Chinoy
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Rheumatology, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, Salford, UK
| |
Collapse
|
45
|
Ahmed N, Oluwole O, Mahmoudjafari Z, Suleman N, McGuirk JP. Managing Infection Complications in the Setting of Chimeric Antigen Receptor T cell (CAR-T) Therapy. Clin Hematol Int 2024; 6:31-45. [PMID: 38817309 PMCID: PMC11086990 DOI: 10.46989/001c.115932] [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: 12/06/2023] [Accepted: 01/23/2024] [Indexed: 06/01/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR T-cell) therapy has changed the paradigm of management of non-Hodgkin's lymphoma (NHL) and Multiple Myeloma. Infection complications have emerged as a concern that can arise in the setting of therapy and lead to morbidity and mortality. In this review, we classified infection complications into three categories, pre-infusion phase from the time pre- lymphodepletion (LD) up to day zero, early phase from day of infusion to day 30 post-infusion, and late phase after day 30 onwards. Infections arising in the pre-infusion phase are closely related to previous chemotherapy and bridging therapy. Infections arising in the early phase are more likely related to LD chemo and the expected brief period of grade 3-4 neutropenia. Infections arising in the late phase are particularly worrisome because they are associated with adverse risk features including prolonged neutropenia, dysregulation of humoral and adaptive immunity with lymphopenia, hypogammaglobinemia, and B cell aplasia. Bacterial, respiratory and other viral infections, protozoal and fungal infections can occur during this time . We recommend enhanced supportive care including prompt recognition and treatment of neutropenia with growth factor support, surveillance testing for specific viruses in the appropriate instance, management of hypogammaglobulinemia with repletion as appropriate and extended antimicrobial prophylaxis in those at higher risk (e.g. high dose steroid use and prolonged cytopenia). Finally, we recommend re-immunizing patients post CAR-T based on CDC and transplant guidelines.
Collapse
Affiliation(s)
- Nausheen Ahmed
- Hematologic Malignancies and Cellular TherapeuticsUniversity of Kansas Cancer Center
| | - Olalekan Oluwole
- Medicine, Hematology and OncologyVanderbilt University Medical Center
| | - Zahra Mahmoudjafari
- Hematologic Malignancies and Cellular TherapeuticsUniversity of Kansas Cancer Center
| | - Nahid Suleman
- Hematologic Malignancies and Cellular TherapeuticsUniversity of Kansas Cancer Center
| | - Joseph P McGuirk
- Hematologic Malignancies and Cellular TherapeuticsUniversity of Kansas Cancer Center
| |
Collapse
|
46
|
Han Z, Ma X, Ma G. Improving cell reinfusion to enhance the efficacy of chimeric antigen receptor T-cell therapy and alleviate complications. Heliyon 2024; 10:e28098. [PMID: 38560185 PMCID: PMC10981037 DOI: 10.1016/j.heliyon.2024.e28098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 02/24/2024] [Accepted: 03/12/2024] [Indexed: 04/04/2024] Open
Abstract
Adoptive cell therapy (ACT) is a rapidly expanding area within the realm of transfusion medicine, focusing on the delivery of lymphocytes to trigger responses against tumors, viruses, or inflammation. This area has quickly evolved from its initial promise in immuno-oncology during preclinical trials to commercial approval of chimeric antigen receptor (CAR) T-cell therapies for leukemia and lymphoma (Jun and et al., 2018) [1]. CAR T-cell therapy has demonstrated success in treating hematological malignancies, particularly relapsed/refractory B-cell acute lymphoblastic leukemia and non-Hodgkin's lymphoma (Qi and et al., 2022) [2]. However, its success in treating solid tumors faces challenges due to the short-lived presence of CAR-T cells in the body and diminished T cell functionality (Majzner and Mackall, 2019) [3]. CAR T-cell therapy functions by activating immune effector cells, yet significant side effects and short response durations remain considerable obstacles to its advancement. A prior study demonstrated that the therapeutic regimen can induce systemic inflammatory reactions, such as cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), tumor lysis syndrome (TLS), off-target effects, and other severe complications. This study aims to explore current research frontiers in this area.
Collapse
Affiliation(s)
- Zhihao Han
- Department of Nursing, Zhejiang Chinese Medical University, Hangzhou City, Zhejiang Province, China
| | - Xiaoqin Ma
- Department of Nursing, Zhejiang Chinese Medical University, Hangzhou City, Zhejiang Province, China
| | - Guiyue Ma
- Department of Nursing, Zhejiang Chinese Medical University, Hangzhou City, Zhejiang Province, China
| |
Collapse
|
47
|
Tu S, Li P, Mei H, Liu Y, Hu Y, Liu P, Zou D, Niu T, Xu K, Wang L, Yang J, Zhao M, Huang X, Wang J, Hu Y, Zhao W, Wu D, Ma J, Qian W, Han W, Li Y, Liang A. Chinese expert consensus on integrated case management by a multidisciplinary team in CAR-T cell therapy for lymphoma. Chin Med J (Engl) 2024:00029330-990000000-01025. [PMID: 38570196 DOI: 10.1097/cm9.0000000000003085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Indexed: 04/05/2024] Open
Affiliation(s)
- Sanfang Tu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou,Guangdong 510280, China
| | - Ping Li
- Department of Hematology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Heng Mei
- Institute of Hematology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yang Liu
- Department of Bio-therapeutic, Chinese PLA General Hospital, Beijing 100853, China
| | - Yongxian Hu
- Center for Bone Marrow Transplantation, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Peng Liu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032,China
| | - Dehui Zou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Division of Pediatric Blood Disease Center, Institute of Hematology & Blood Disease Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, China
| | - Li Wang
- Department of Hematology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jianmin Yang
- Department of Hematology, Changhai Hospital, The Naval Medical University, Shanghai 210000, China
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, The First Affiliated Central Hospital of Nankai University, The First Central Clinical College of Tianjin Medical University, Tianjin 300192, China
| | - Xiaojun Huang
- Department of Hematology, Peking University People's Hospital, Peking University Institute of Hematology Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation Research Unit of Key Technique for Diagnosis and Treatments of Haematologic Malignancies, Chinese Academt of Medical Sciences, Peking University, Beijing 100044, China
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Division of Pediatric Blood Disease Center, Institute of Hematology & Blood Disease Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Yu Hu
- Institute of Hematology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Weili Zhao
- Department of Hematology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Depei Wu
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Jun Ma
- Harbin Institute of Hematology and Cancer, Harbin, Heilongjiang 150001, China
| | - Wenbin Qian
- Department of Hematology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Weidong Han
- Department of Bio-therapeutic, Chinese PLA General Hospital, Beijing 100853, China
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou,Guangdong 510280, China
| | - Aibin Liang
- Department of Hematology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| |
Collapse
|
48
|
Qian H, Yang X, Zhang T, Zou P, Zhang Y, Tian W, Mao Z, Wei J. Improving the safety of CAR-T-cell therapy: The risk and prevention of viral infection for patients with relapsed or refractory B-cell lymphoma undergoing CAR-T-cell therapy. Am J Hematol 2024; 99:662-678. [PMID: 38197307 DOI: 10.1002/ajh.27198] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/05/2023] [Accepted: 12/15/2023] [Indexed: 01/11/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy, an innovative immunotherapeutic against relapsed/refractory B-cell lymphoma, faces challenges due to frequent viral infections. Despite this, a comprehensive review addressing risk assessment, surveillance, and treatment management is notably absent. This review elucidates immune response compromises during viral infections in CAR-T recipients, collates susceptibility risk factors, and deliberates on preventive strategies. In the post-pandemic era, marked by the Omicron variant, new and severe threats to CAR-T therapy emerge, necessitating exploration of preventive and treatment measures for COVID-19. Overall, the review provides recommendations for viral infection prophylaxis and management, enhancing CAR-T product safety and recipient survival.
Collapse
Affiliation(s)
- Hu Qian
- 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
| | - Xingcheng Yang
- 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
| | - Tingting Zhang
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- 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
| | - Ping Zou
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yicheng Zhang
- 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
| | - Weiwei Tian
- 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
| | - Zekai Mao
- 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
| | - Jia Wei
- 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
- 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
| |
Collapse
|
49
|
Gambella M, Carlomagno S, Mangerini R, Colombo N, Parodi A, Ghiggi C, Giannoni L, Coviello E, Setti C, Luchetti S, Serio A, Laudisi A, Passannante M, Bo A, Tedone E, Sivori S, Angelucci E, Raiola AM. Early CAR - CD4 + T-lymphocytes recovery following CAR-T cell infusion: A worse outcome in diffuse large B cell lymphoma. EJHAEM 2024; 5:360-368. [PMID: 38633118 PMCID: PMC11020131 DOI: 10.1002/jha2.871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/05/2024] [Indexed: 04/19/2024]
Abstract
CAR- CD4+ T cell lymphopenia is an emerging issue following CAR-T cell therapy. We analyzed the determinants of CD4+ T cell recovery and a possible association with survival in 31 consecutive patients treated with commercial CAR-T for diffuse large B-cell (DLBCL) or mantle cell lymphoma. Circulating immune subpopulations were characterized through multiparametric-flow cytometry. Six-month cumulative incidence of CAR- CD4+ T cell recovery (≥200 cells/μL) was 0.43 (95% confidence interval [CI]: 0.28-0.65). Among possible determinants of CD4+ T cell recovery, we recognized infusion of a 4-1BB product (tisagenlecleucel, TSA) in comparison with a CD28 (axicabtagene/brexucabtagene, AXI/BRX) (hazard ratio [HR] [95% CI]: 5.79 [1.16-24.12] p = 0.016). Higher CD4+ T cell counts resulted with TSA at month-1, -2 and -3. Moderate-to-severe infections were registered with prolonged CD4+ T cell lymphopenia. Early, month-1 CD4+ T cell recovery was associated with a worse outcome in the DLBCL cohort, upheld in a multivariate regression model for overall survival (HR: 4.46 [95% CI: 1.12-17.71], p = 0.03). We conclude that a faster CAR- CD4+ T cell recovery is associated with TSA as compared to AXI/BRX. Month-1 CAR- CD4+ T cell subset recovery could represent a "red flag" for CAR-T cell therapy failure in DLBCL patients.
Collapse
Affiliation(s)
- Massimiliano Gambella
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
- Department of Experimental Medicine (DIMES)University of GenoaGenovaItaly
| | | | - Rosa Mangerini
- Anatomia Patologica OspedalieraIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Nicoletta Colombo
- Anatomia Patologica OspedalieraIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Alessia Parodi
- Anatomia Patologica OspedalieraIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Chiara Ghiggi
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Livia Giannoni
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Elisa Coviello
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Chiara Setti
- Department of Experimental Medicine (DIMES)University of GenoaGenovaItaly
| | - Silvia Luchetti
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Alberto Serio
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Antonella Laudisi
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Monica Passannante
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Alessandra Bo
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Elisabetta Tedone
- Anatomia Patologica OspedalieraIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Simona Sivori
- Department of Experimental Medicine (DIMES)University of GenoaGenovaItaly
- IRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Emanuele Angelucci
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Anna Maria Raiola
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| |
Collapse
|
50
|
Kegyes D, Ghiaur G, Bancos A, Tomuleasa C, Gale RP. Immune therapies of B-cell acute lymphoblastic leukaemia in children and adults. Crit Rev Oncol Hematol 2024; 196:104317. [PMID: 38437908 DOI: 10.1016/j.critrevonc.2024.104317] [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: 10/14/2023] [Revised: 01/26/2024] [Accepted: 02/27/2024] [Indexed: 03/06/2024] Open
Abstract
B-cell acute lymphoblastic leukaemia (B-cell ALL) is a common haematologic cancer in children and adults. About 10 percent of children and 50 percent of adults fail to achieve a histological complete remission or subsequently relapse despite current anti-leukaemia drug therapies and/or haematopoietic cell transplants. Several new immune therapies including monoclonal antibodies and chimeric antigen receptor (CAR)-T-cells are proved safe and effective in this setting. We review data on US Food and Drug Administration (FDA)-approved immune therapies for B-cell ALL in children and adults including blinatumomab, inotuzumab ozogamicin, tisagenlecleucel, and brexucabtagene autoleucel. We also summarize pharmaco-dynamics, pharmaco-kinetics, and pharmaco-economics of these interventions.
Collapse
Affiliation(s)
- David Kegyes
- Department of Hematology-Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj Napoca, Romania; Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania; Academy of Romanian Scientists, Bucharest, Romania
| | - Gabriel Ghiaur
- Department of Hematology-Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj Napoca, Romania; Department of Leukemia, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University, Baltimore, MD, USA
| | - Anamaria Bancos
- Department of Hematology-Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj Napoca, Romania; Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania
| | - Ciprian Tomuleasa
- Department of Hematology-Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj Napoca, Romania; Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania; Academy of Romanian Scientists, Bucharest, Romania.
| | - Robert Peter Gale
- Centre for Haematology, Imperial College of Science, Technology and Medicine, London, UK; Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Hematology, Peking University People's Hospital, Beijing, China
| |
Collapse
|