1
|
Nardone V, Ruggiero D, Chini MG, Bruno I, Lauro G, Terracciano S, Nebbioso A, Bifulco G, Cappabianca S, Reginelli A. From Bench to Bedside: Translational Approaches to Cardiotoxicity in Breast Cancer, Lung Cancer, and Lymphoma Therapies. Cancers (Basel) 2025; 17:1059. [PMID: 40227572 PMCID: PMC11987928 DOI: 10.3390/cancers17071059] [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: 12/12/2024] [Revised: 03/10/2025] [Accepted: 03/19/2025] [Indexed: 04/15/2025] Open
Abstract
Cardiotoxicity represents a critical challenge in cancer therapy, particularly in the treatment of thoracic tumors, such as lung cancer and lymphomas, as well as breast cancer. These malignancies stand out for their high prevalence and the widespread use of cardiotoxic treatments, such as chemotherapy, radiotherapy, and immunotherapy. This work underscores the importance of preclinical models in uncovering the mechanisms of cardiotoxicity and developing targeted prevention and mitigation strategies. In vitro models provide valuable insights into cellular processes, enabling the observation of changes in cell viability and function following exposure to various drugs or ionizing radiation. Complementarily, in vivo animal models offer a broader perspective, allowing for evaluating of both short- and long-term effects and a better understanding of chronic toxicity and cardiac diseases. By integrating these approaches, researchers can identify potential mechanisms of cardiotoxicity and devise effective prevention strategies. This analysis highlights the central role of preclinical models in advancing knowledge of cardiotoxic effects associated with common therapeutic regimens for thoracic and breast cancers.
Collapse
Affiliation(s)
- Valerio Nardone
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (V.N.); (D.R.); (A.N.); (S.C.); (A.R.)
| | - Dafne Ruggiero
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (V.N.); (D.R.); (A.N.); (S.C.); (A.R.)
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (I.B.); (G.L.); (S.T.); (G.B.)
| | - Maria Giovanna Chini
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, Pesche, 86090 Isernia, Italy
| | - Ines Bruno
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (I.B.); (G.L.); (S.T.); (G.B.)
| | - Gianluigi Lauro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (I.B.); (G.L.); (S.T.); (G.B.)
| | - Stefania Terracciano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (I.B.); (G.L.); (S.T.); (G.B.)
| | - Angela Nebbioso
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (V.N.); (D.R.); (A.N.); (S.C.); (A.R.)
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (I.B.); (G.L.); (S.T.); (G.B.)
| | - Salvatore Cappabianca
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (V.N.); (D.R.); (A.N.); (S.C.); (A.R.)
| | - Alfonso Reginelli
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (V.N.); (D.R.); (A.N.); (S.C.); (A.R.)
| |
Collapse
|
2
|
Sun W, Wang S, Han J, Zhuo L, Cao J, Zhou F. Symptoms of Hematologic Tumors Patients after CAR-T Therapy: A Systematic Review and Meta-Analysis. J Pain Symptom Manage 2025; 69:304-317. [PMID: 39547263 DOI: 10.1016/j.jpainsymman.2024.11.002] [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: 09/19/2024] [Revised: 10/28/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
CONTEXT Patients with hematologic neoplasms after Chimeric antigen receptor T-cell (CAR-T) therapy have multiple syndromes, with corresponding symptoms. OBJECTIVES The review aimed to integrate the severity and incidences of symptoms in these patients, and to investigate the difference of the symptoms among different geographic locations, types of hematological tumors, evaluating instruments, and evaluating time, to provide a theoretical basis for symptom management. METHODS A literature search of PubMed, Web of Science, Embase, the Cochrane Library, China National Knowledge Internet, SinoMed, VIP, and WANFANG DATA was performed for studies reporting symptom scores or symptom incidences of these patients published before November 9, 2023. Heterogeneity between studies was assessed by Higgins' I2. A random effects model was used for studies with I2 > 50%. Methodological quality of included studies was assessed using the Joanna Briggs Institute Critical Appraisal Checklist. RESULTS Eight studies were included. Among the reported symptoms, sleep disturbance, fatigue and depression were of higher severity, with the standardized scores exceeding 50. Sadness, problem with concentration, problem with memory, cough and nausea were the top five symptoms in incidence, which exceeded 50%. The symptom scores and incidences assessed by the patient-reported outcomes were higher. Within 90 days of CAR-T infusion, these patients reported a significantly higher severity and incidence of multiple symptoms. CONCLUSION Patients with hematologic neoplasms treated by CAR-T suffer from multiple symptoms, including depression, fatigue, and so on. Instruments used to evaluate symptoms and the evaluating time may influence the outcome of symptom assessment.
Collapse
Affiliation(s)
- Wan Sun
- School of Nursing (W.S., S.W., J.H., F.Z.), Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Shuo Wang
- School of Nursing (W.S., S.W., J.H., F.Z.), Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Jiachen Han
- School of Nursing (W.S., S.W., J.H., F.Z.), Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Lang Zhuo
- School of Public Health (L.Z.), Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Jiang Cao
- Department of Hematology (J.C.), The Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Fang Zhou
- School of Nursing (W.S., S.W., J.H., F.Z.), Xuzhou Medical University, Xuzhou City, Jiangsu Province, China.
| |
Collapse
|
3
|
Zhu S, Zhou Z, Gu R, Zhao Z, Zhang Y, Miao Y, Lei Q, Liu T, Wang G, Dai C, Huo Y, You J, Lv L, Li C, Yin M, Wang C, Deng H. TLR7/8 signaling activation enhances the potency of human pluripotent stem cell-derived eosinophils in cancer immunotherapy for solid tumors. Exp Hematol Oncol 2025; 14:26. [PMID: 40025520 PMCID: PMC11871822 DOI: 10.1186/s40164-025-00613-y] [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: 07/19/2024] [Accepted: 02/11/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Efficient tumor T-cell infiltration is crucial for the effectiveness of T-cell-based therapies against solid tumors. Eosinophils play crucial roles in recruiting T cells in solid tumors. Our group has previously generated induced eosinophils (iEOs) from human pluripotent stem cells and exhibited synergistic efficacy with CAR-T cells in solid tumor inhibition. However, administrated eosinophils might influx into inflammatory lungs, posing a potential safety risk. Mitigating the safety concern and enhancing efficacy is a promising development direction for further application of eosinophils. METHODS We developed a new approach to generate eosinophils with enhanced potency from human chemically reprogrammed induced pluripotent stem cells (hCiPSCs) with the Toll-like receptor (TLR) 7/8 signaling agonist R848. RESULTS R848-activated iEOs (R-iEOs) showed significantly decreased influx to the inflamed lungs, indicating a lower risk of causing airway disorders. Furthermore, these R-iEOs had enhanced anti-tumor functions, preferably accumulated at tumor sites, and further increased T-cell infiltration. The combination of R-iEOs and CAR-T cells suppressed tumor growth in mice. Moreover, the chemo-trafficking signaling increased in R-iEOs, which may contribute to the decreased lung influx of R-iEOs and the increased tumor recruitment of T cells. CONCLUSION Our study provides a novel approach to alleviate the potential safety concerns associated with eosinophils while increasing T-cell infiltration in solid tumors. This finding offers a prospective strategy for incorporating eosinophils to improve CAR-T-cell immunotherapy for solid tumors in the future.
Collapse
Affiliation(s)
- Sheng Zhu
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Zhengyang Zhou
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Ruixin Gu
- Center for Bioinformatics, Center for Statistical Science, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Zixin Zhao
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Yingfeng Zhang
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Yudi Miao
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Qi Lei
- State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University Health Science Center, Peking University, Beijing, 100191, China
| | - Tianxing Liu
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Guokai Wang
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Chenyi Dai
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Yi Huo
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Jinghao You
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Lejun Lv
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Cheng Li
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- Center for Bioinformatics, Center for Statistical Science, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ming Yin
- Beijing Vitalstar Biotechnology, Beijing, 100012, China
| | - Chengyan Wang
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
| | - Hongkui Deng
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
- State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University Health Science Center, Peking University, Beijing, 100191, China.
| |
Collapse
|
4
|
Lazaro Sanchez AD, Benitez Fuentes JD, Gil GL, García MTG, Moreno EF, Zamora PC, Yago JB, Mohamed KM, Arroyo Rodríguez AB. Clinical and Proteomic Insights into a Cytokine Release Syndrome Triggered by Tebentafusp in a Metastatic Uveal Melanoma Patient: Case Report. J Clin Med 2025; 14:1333. [PMID: 40004863 PMCID: PMC11856639 DOI: 10.3390/jcm14041333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/25/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Uveal melanoma is the most common primary intraocular cancer in adults; however, it remains rare. Despite its rarity, metastatic uveal melanoma poses significant treatment challenges. Tebentafusp, a T-cell receptor-bispecific molecule targeting glycoprotein 100 and CD3, has shown substantial survival benefits for HLA-A*02:01 positive patients. A notable complication associated with tebentafusp and similar immunotherapies is cytokine release syndrome (CRS), occurring in nearly 90% of tebentafusp-treated patients. Although typically mild, severe CRS (grade 3) affects around 1% of patients. The unpredictable nature of CRS complicates patient management during treatment. Methods: Monitoring cytokine levels, as key indicators of inflammation, may therefore be crucial for understanding and managing CRS. Advanced proteomic technologies enable the simultaneous measurement of multiple cytokines, providing a comprehensive view of inflammatory responses. Results: In this case, a patient with metastatic uveal melanoma developed CRS after tebentafusp treatment. A proteomic analysis tracked the cytokine changes from baseline to post-treatment, revealing significant elevations in inflammatory markers. Conclusions: These findings suggest potential strategies for more personalized CRS management in similar therapies.
Collapse
Affiliation(s)
- Antonio David Lazaro Sanchez
- Department of Medical Oncology, Morales Meseguer General University Hospital, 30008 Murcia, Spain
- Laboratory Medicine and Pathology Department, Group of Molecular Pathology and Pharmacogenetics, Biomedical Research Institute from Murcia (IMIB), Santa Lucía General University Hospital, 30202 Cartagena, Spain; (G.L.G.); (P.C.Z.); (A.B.A.R.)
| | | | - Ginés Luengo Gil
- Laboratory Medicine and Pathology Department, Group of Molecular Pathology and Pharmacogenetics, Biomedical Research Institute from Murcia (IMIB), Santa Lucía General University Hospital, 30202 Cartagena, Spain; (G.L.G.); (P.C.Z.); (A.B.A.R.)
- Health Sciences Faculty, Universidad Católica de Murcia (UCAM), 30107 Guadalupe, Spain; (M.T.G.G.); (E.F.M.); (J.B.Y.)
| | - María Teresa García García
- Health Sciences Faculty, Universidad Católica de Murcia (UCAM), 30107 Guadalupe, Spain; (M.T.G.G.); (E.F.M.); (J.B.Y.)
- Department of Medical Oncology, Santa Lucia General University Hospital, 30202 Cartagena, Spain
| | - Eduardo Feliciangeli Moreno
- Health Sciences Faculty, Universidad Católica de Murcia (UCAM), 30107 Guadalupe, Spain; (M.T.G.G.); (E.F.M.); (J.B.Y.)
- Department of Medical Oncology, Santa Lucia General University Hospital, 30202 Cartagena, Spain
| | - Pablo Conesa Zamora
- Laboratory Medicine and Pathology Department, Group of Molecular Pathology and Pharmacogenetics, Biomedical Research Institute from Murcia (IMIB), Santa Lucía General University Hospital, 30202 Cartagena, Spain; (G.L.G.); (P.C.Z.); (A.B.A.R.)
- Health Sciences Faculty, Universidad Católica de Murcia (UCAM), 30107 Guadalupe, Spain; (M.T.G.G.); (E.F.M.); (J.B.Y.)
| | - José Balsalobre Yago
- Health Sciences Faculty, Universidad Católica de Murcia (UCAM), 30107 Guadalupe, Spain; (M.T.G.G.); (E.F.M.); (J.B.Y.)
- Department of Medical Oncology, Santa Lucia General University Hospital, 30202 Cartagena, Spain
| | - Kauzar Mohamed Mohamed
- Department of Immunology, IML and IdISSC, Hospital Clinico San Carlos, 28040 Madrid, Spain;
| | - Ana Belén Arroyo Rodríguez
- Laboratory Medicine and Pathology Department, Group of Molecular Pathology and Pharmacogenetics, Biomedical Research Institute from Murcia (IMIB), Santa Lucía General University Hospital, 30202 Cartagena, Spain; (G.L.G.); (P.C.Z.); (A.B.A.R.)
- Health Sciences Faculty, Universidad Católica de Murcia (UCAM), 30107 Guadalupe, Spain; (M.T.G.G.); (E.F.M.); (J.B.Y.)
| |
Collapse
|
5
|
Ploch W, Sadowski K, Olejarz W, Basak GW. Advancement and Challenges in Monitoring of CAR-T Cell Therapy: A Comprehensive Review of Parameters and Markers in Hematological Malignancies. Cancers (Basel) 2024; 16:3339. [PMID: 39409959 PMCID: PMC11475293 DOI: 10.3390/cancers16193339] [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: 08/31/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has revolutionized the treatment for relapsed/refractory B-cell lymphomas. Despite its success, this therapy is accompanied by a significant frequency of adverse events, including cytokine release syndrome (CRS), immune-effector-cell-associated neurotoxicity syndrome (ICANS), or cytopenias, reaching even up to 80% of patients following CAR-T cell therapy. CRS results from the uncontrolled overproduction of proinflammatory cytokines, which leads to symptoms such as fever, headache, hypoxia, or neurological complications. CAR-T cell detection is possible by the use of flow cytometry (FC) or quantitative polymerase chain reaction (qPCR) assays, the two primary techniques used for CAR-T evaluation in peripheral blood, bone marrow (BM), and cerebrospinal fluid (CSF). State-of-the-art imaging technologies play a crucial role in monitoring the distribution and persistence of CAR-T cells in clinical trials. Still, they can also be extended with the use of FC and digital PCR (dPCR). Monitoring the changes in cell populations during disease progression and treatment gives an important insight into how the response to CAR-T cell therapy develops on a cellular level. It can help improve the therapeutic design and optimize CAR-T cell therapy to make it more precise and personalized, which is crucial to overcoming the problem of tumor relapse.
Collapse
Affiliation(s)
- Weronika Ploch
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (W.P.); (K.S.)
- Department of Hematology, Transplantation and Internal Medicine, 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; (W.P.); (K.S.)
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (W.P.); (K.S.)
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Grzegorz W. Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| |
Collapse
|
6
|
Oluwole OO, Ray MD, Rosettie KL, Ball G, Jacob J, Bilir SP, Patel AR, Jacobson CA. Cost-Effectiveness of Axicabtagene Ciloleucel for Adult Patients With Relapsed or Refractory Follicular Lymphoma in the United States. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2024; 27:1030-1038. [PMID: 38641058 DOI: 10.1016/j.jval.2024.04.003] [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: 10/16/2023] [Revised: 03/25/2024] [Accepted: 04/08/2024] [Indexed: 04/21/2024]
Abstract
OBJECTIVES The results of a recent single-arm trial (ZUMA-5) of axicabtagene ciloleucel (axi-cel) for relapsed/refractory (r/r) follicular lymphoma (FL) demonstrated high rates of durable response and tolerable toxicity among treated patients. To quantify the value of axi-cel compared with standard of care (SOC) to manage r/r FL patients who have had at least 2 prior lines of systemic therapy (3L+), a cost-effectiveness model was developed from a US third-party payer perspective. METHODS A 3-state partitioned-survival cost-effectiveness model was developed with a lifetime horizon. Patient-level analyses of the 36-month ZUMA-5 (axi-cel) and SCHOLAR-5 (SOC) studies were used to extrapolate progression-free and overall survivals. After 5 years of survival, an estimated 40% of the modeled population was assumed to experience long-term remission based on literature. Results include the incremental cost-effectiveness ratio (ICER) measured as incremental cost per quality-adjusted life year (QALY) gained. One-way sensitivity analysis, probabilistic sensitivity analysis, and scenario analyses were performed. All outcomes were discounted 3% per year. RESULTS Axi-cel led to an increase of 4.28 life-years, 3.64 QALYs, and a total cost increase of $321 192 relative to SOC, resulting in an ICER of $88 300 per QALY. Across all parameters varied in the one-way sensitivity analysis, the ICER varied between $133 030 and $67 277. In the probabilistic sensitivity analysis, axi-cel had a 99% probability of being cost-effective across 5000 iterations using a $150 000 willingness-to-pay threshold. CONCLUSIONS Given the robustness of the model results and sensitivity analyses, axi-cel is expected to be a cost-effective treatment in 3L+ r/r FL.
Collapse
Affiliation(s)
- Olalekan O Oluwole
- Vanderbilt University Medical Center, School of Medicine, Nashville, TN, USA.
| | | | | | - Graeme Ball
- Kite, A Gilead Company, Santa Monica, CA, USA
| | | | | | | | | |
Collapse
|
7
|
Géraud A, Hueso T, Laparra A, Bige N, Ouali K, Cauquil C, Stoclin A, Danlos FX, Hollebecque A, Ribrag V, Gazzah A, Goldschmidt V, Baldini C, Suzzoni S, Bahleda R, Besse B, Barlesi F, Lambotte O, Massard C, Marabelle A, Castilla-Llorente C, Champiat S, Michot JM. Reactions and adverse events induced by T-cell engagers as anti-cancer immunotherapies, a comprehensive review. Eur J Cancer 2024; 205:114075. [PMID: 38733717 DOI: 10.1016/j.ejca.2024.114075] [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: 03/01/2024] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024]
Abstract
T-cell engagers (TCE) are cancer immunotherapies that have recently demonstrated meaningful benefit for patients with hematological malignancies and solid tumors. The anticipated widespread use of T cell engagers poses implementation challenges and highlights the need for guidance to anticipate, mitigate, and manage adverse events. By mobilizing T-cells directly at the contact of tumor cells, TCE mount an obligatory and immediate anti-tumor immune response that could result in diverse reactions and adverse events. Cytokine release syndrome (CRS) is the most common reaction and is largely confined to the first drug administrations during step-up dosage. Cytokine release syndrome should be distinguished from infusion related reaction by clinical symptoms, timing to occurrence, pathophysiological aspects, and clinical management. Other common reactions and adverse events with TCE are immune effector Cell-Associated Neurotoxicity Syndrome (ICANS), infections, tumor flare reaction and cytopenias. The toxicity profiles of TCE and CAR-T cells have commonalities and distinctions that we sum-up in this review. As compared with CAR-T cells, TCE are responsible for less frequently severe CRS or ICANS. This review recapitulates terminology, pathophysiology, severity grading system and management of reactions and adverse events related to TCE.
Collapse
Affiliation(s)
- Arthur Géraud
- Gustave Roussy, Département d'Innovation Thérapeutique et d'Essais Précoces, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Thomas Hueso
- Gustave Roussy, Département d'Innovation Thérapeutique et d'Essais Précoces, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Ariane Laparra
- Gustave Roussy, Departement Interdisciplinaire d'Organisation des Parcours Patients, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Naike Bige
- Gustave Roussy, Service de réanimation et de soins intensifs, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Kaissa Ouali
- Gustave Roussy, Département d'Innovation Thérapeutique et d'Essais Précoces, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Cécile Cauquil
- Hôpital Universitaire du Kremlin Bicêtre, Service de Neurologie, 94270 Le Kremlin-Bicêtre, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Annabelle Stoclin
- Gustave Roussy, Service de réanimation et de soins intensifs, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - François-Xavier Danlos
- Gustave Roussy, Département d'Innovation Thérapeutique et d'Essais Précoces, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Antoine Hollebecque
- Gustave Roussy, Département d'Innovation Thérapeutique et d'Essais Précoces, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Vincent Ribrag
- Gustave Roussy, Department Hématologie, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Anas Gazzah
- Gustave Roussy, Département d'Innovation Thérapeutique et d'Essais Précoces, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Vincent Goldschmidt
- Gustave Roussy, Département d'Innovation Thérapeutique et d'Essais Précoces, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Capucine Baldini
- Gustave Roussy, Département d'Innovation Thérapeutique et d'Essais Précoces, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Steve Suzzoni
- Gustave Roussy, Department of Pharmacy, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Rastislav Bahleda
- Gustave Roussy, Département d'Innovation Thérapeutique et d'Essais Précoces, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Benjamin Besse
- Gustave Roussy, Department de Médecine Oncologique, 94805 Villejuif, France; Université Paris-Saclay, Gustave Roussy, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Fabrice Barlesi
- Gustave Roussy, Department de Médecine Oncologique, 94805 Villejuif, France; Université Paris-Saclay, Gustave Roussy, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Olivier Lambotte
- Université Paris-Saclay, Gustave Roussy, 94805 Villejuif, France; Hôpital Universitaire du Kremlin Bicêtre, Service de Médecine Interne, 94270 Le Kremlin-Bicêtre, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Christophe Massard
- Gustave Roussy, Département d'Innovation Thérapeutique et d'Essais Précoces, 94805 Villejuif, France; Université Paris-Saclay, Gustave Roussy, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Aurélien Marabelle
- Gustave Roussy, Département d'Innovation Thérapeutique et d'Essais Précoces, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Cristina Castilla-Llorente
- Gustave Roussy, Department Hématologie, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Stéphane Champiat
- Gustave Roussy, Département d'Innovation Thérapeutique et d'Essais Précoces, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France
| | - Jean-Marie Michot
- Gustave Roussy, Département d'Innovation Thérapeutique et d'Essais Précoces, 94805 Villejuif, France; Gustave Roussy, Department d'Hématologie Clinique, 94805 Villejuif, France.
| |
Collapse
|
8
|
Shumnalieva R, Velikova T, Monov S. Expanding the role of CAR T-cell therapy: From B-cell hematological malignancies to autoimmune rheumatic diseases. Int J Rheum Dis 2024; 27:e15182. [PMID: 38742463 DOI: 10.1111/1756-185x.15182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 04/04/2024] [Accepted: 04/28/2024] [Indexed: 05/16/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a form of immunotherapy where the lymphocytes, mostly T-cells, are redirected to specifically recognize and eliminate a target antigen by coupling them with CARs. The binding of CAR and target cell surface antigens leads to vigorous T cell activation and robust anti-tumor immune responses. Areas of implication of CAR T-cell therapies include mainly hematological malignancies (i.e., advanced B-cell cancers); however, recent studies have proven the unprecedented success of the new immunotherapy also in autoimmune rheumatic diseases. We aim to review the recent advances in CAR T-cell therapies in rheumatology but also to address the limitations of their use in the real clinical practice based on the data on their efficacy and safety.
Collapse
Affiliation(s)
- Russka Shumnalieva
- Department of Rheumatology, Clinic of Rheumatology, Medical University-Sofia, Faculty of Medicine, Sofia, Bulgaria
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University "St. Kliment Ohridski"- Sofia, Sofia, Bulgaria
| | - Simeon Monov
- Department of Rheumatology, Clinic of Rheumatology, Medical University-Sofia, Faculty of Medicine, Sofia, Bulgaria
| |
Collapse
|
9
|
Appelbaum J, Price AE, Oda K, Zhang J, Leung WH, Tampella G, Xia D, So PP, Hilton SK, Evandy C, Sarkar S, Martin U, Krostag AR, Leonardi M, Zak DE, Logan R, Lewis P, Franke-Welch S, Ngwenyama N, Fitzgerald M, Tulberg N, Rawlings-Rhea S, Gardner RA, Jones K, Sanabria A, Crago W, Timmer J, Hollands A, Eckelman B, Bilic S, Woodworth J, Lamble A, Gregory PD, Jarjour J, Pogson M, Gustafson JA, Astrakhan A, Jensen MC. Drug-regulated CD33-targeted CAR T cells control AML using clinically optimized rapamycin dosing. J Clin Invest 2024; 134:e162593. [PMID: 38502193 PMCID: PMC11060733 DOI: 10.1172/jci162593] [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: 06/13/2022] [Accepted: 03/08/2024] [Indexed: 03/21/2024] Open
Abstract
Chimeric antigen receptor (CAR) designs that incorporate pharmacologic control are desirable; however, designs suitable for clinical translation are needed. We designed a fully human, rapamycin-regulated drug product for targeting CD33+ tumors called dimerizaing agent-regulated immunoreceptor complex (DARIC33). T cell products demonstrated target-specific and rapamycin-dependent cytokine release, transcriptional responses, cytotoxicity, and in vivo antileukemic activity in the presence of as little as 1 nM rapamycin. Rapamycin withdrawal paused DARIC33-stimulated T cell effector functions, which were restored following reexposure to rapamycin, demonstrating reversible effector function control. While rapamycin-regulated DARIC33 T cells were highly sensitive to target antigen, CD34+ stem cell colony-forming capacity was not impacted. We benchmarked DARIC33 potency relative to CD19 CAR T cells to estimate a T cell dose for clinical testing. In addition, we integrated in vitro and preclinical in vivo drug concentration thresholds for off-on state transitions, as well as murine and human rapamycin pharmacokinetics, to estimate a clinically applicable rapamycin dosing schedule. A phase I DARIC33 trial has been initiated (PLAT-08, NCT05105152), with initial evidence of rapamycin-regulated T cell activation and antitumor impact. Our findings provide evidence that the DARIC platform exhibits sensitive regulation and potency needed for clinical application to other important immunotherapy targets.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Immunotherapy, Adoptive
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Receptors, Chimeric Antigen/immunology
- Sialic Acid Binding Ig-like Lectin 3/immunology
- Sialic Acid Binding Ig-like Lectin 3/metabolism
- Sirolimus/pharmacology
- Sirolimus/administration & dosage
- T-Lymphocytes/immunology
- T-Lymphocytes/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jacob Appelbaum
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
- Division of Hematology/Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Seattle Children’s Hospital, Seattle, Washington, USA
| | | | - Kaori Oda
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Joy Zhang
- 2seventy bio, Cambridge, Massachusetts, USA
| | | | - Giacomo Tampella
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Dong Xia
- 2seventy bio, Cambridge, Massachusetts, USA
| | | | | | - Claudya Evandy
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Semanti Sarkar
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | | | | | - Marissa Leonardi
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | | | - Rachael Logan
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | | | | | | | - Michael Fitzgerald
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Niklas Tulberg
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Stephanie Rawlings-Rhea
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Rebecca A. Gardner
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Kyle Jones
- Inhibrx, Torrey Pines Science Park, La Jolla, California, USA
| | | | - William Crago
- Inhibrx, Torrey Pines Science Park, La Jolla, California, USA
| | - John Timmer
- Inhibrx, Torrey Pines Science Park, La Jolla, California, USA
| | - Andrew Hollands
- Inhibrx, Torrey Pines Science Park, La Jolla, California, USA
| | | | | | | | - Adam Lamble
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
- Seattle Children’s Hospital, Seattle, Washington, USA
| | | | | | | | - Joshua A. Gustafson
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | | | - Michael C. Jensen
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| |
Collapse
|
10
|
Liu Y, Peng C, Ahad F, Ali Zaidi SA, Muluh TA, Fu Q. Advanced Strategies of CAR-T Cell Therapy in Solid Tumors and Hematological Malignancies. Recent Pat Anticancer Drug Discov 2024; 19:557-572. [PMID: 38213150 DOI: 10.2174/0115748928277331231218115402] [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: 09/18/2023] [Revised: 10/30/2023] [Accepted: 11/10/2023] [Indexed: 01/13/2024]
Abstract
Chimeric antigen receptor T-cells, known as CAR-T cells, represent a promising breakthrough in the realm of adoptive cell therapy. These T-cells are genetically engineered to carry chimeric antigen receptors that specifically target tumors. They have achieved notable success in the treatment of blood-related cancers, breathing new life into this field of medical research. However, numerous obstacles limit chimeric antigen receptors T-cell therapy's efficacy, such as it cannot survive in the body long. It is prone to fatigue and exhaustion, leading to difficult tumor elimination and repeated recurrence, affecting solid tumors and hematological malignancies. The challenges posed by solid tumors, especially in the context of the complex solid-tumor microenvironment, require specific strategies. This review outlines recent advancements in improving chimeric antigen receptors T-cell therapy by focusing on the chimeric antigen receptors protein, modifying T-cells, and optimizing the interaction between T-cells and other components within the tumor microenvironment. This article aims to provide an extensive summary of the latest discoveries regarding CAR-T cell therapy, encompassing its application across various types of human cancers. Moreover, it will delve into the obstacles that have emerged in recent times, offering insights into the challenges faced by this innovative approach. Finally, it highlights novel therapeutic options in treating hematological and solid malignancies with chimeric antigen receptors T-cell therapies.
Collapse
Affiliation(s)
- Yangjie Liu
- Department of Pharmacy, Luzhou People's Hospital, Luzhou 646000, Sichuan, PRC China
| | - Cao Peng
- Department of Pharmacy, Luzhou People's Hospital, Luzhou 646000, Sichuan PRC China
| | - Faiza Ahad
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Syed Aqib Ali Zaidi
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Tobias Achu Muluh
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Qiuxia Fu
- Department of Pharmacy, Luzhou People's Hospital, Luzhou 646000, Sichuan PRC China
| |
Collapse
|
11
|
Hajibabaie F, Abedpoor N, Haghjooy Javanmard S, Hasan A, Sharifi M, Rahimmanesh I, Shariati L, Makvandi P. The molecular perspective on the melanoma and genome engineering of T-cells in targeting therapy. ENVIRONMENTAL RESEARCH 2023; 237:116980. [PMID: 37648188 DOI: 10.1016/j.envres.2023.116980] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/19/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023]
Abstract
Melanoma, an aggressive malignant tumor originating from melanocytes in humans, is on the rise globally, with limited non-surgical treatment options available. Recent advances in understanding the molecular and cellular mechanisms underlying immune escape, tumorigenesis, drug resistance, and cancer metastasis have paved the way for innovative therapeutic strategies. Combination therapy targeting multiple pathways simultaneously has been shown to be promising in treating melanoma, eliciting favorable responses in most melanoma patients. CAR T-cells, engineered to overcome the limitations of human leukocyte antigen (HLA)-dependent tumor cell detection associated with T-cell receptors, offer an alternative approach. By genetically modifying apheresis-collected allogeneic or autologous T-cells to express chimeric antigen receptors, CAR T-cells can appreciate antigens on cell surfaces independently of major histocompatibility complex (MHC), providing a significant cancer cell detection advantage. However, identifying the most effective target antigen is the initial step, as it helps mitigate the risk of toxicity due to "on-target, off-tumor" and establishes a targeted therapeutic strategy. Furthermore, evaluating signaling pathways and critical molecules involved in melanoma pathogenesis remains insufficient. This study emphasizes the novel approaches of CAR T-cell immunoediting and presents new insights into the molecular signaling pathways associated with melanoma.
Collapse
Affiliation(s)
- Fatemeh Hajibabaie
- Department of Biology, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran; Department of Medical Biotechnology, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran.
| | - Navid Abedpoor
- Department of Sports Physiology, Faculty of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran; Department of Medical Biotechnology, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran.
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha, 2713, Qatar; Biomedical Research Center, Qatar University, Doha, 2713, Qatar.
| | - Mehran Sharifi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Ilnaz Rahimmanesh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Laleh Shariati
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran; Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, China; School of Engineering, Institute for Bioengineering, The University of Edinburgh, Edinburgh, EH9 3JL, UK.
| |
Collapse
|
12
|
Khanam R, Faiman B, Batool S, Najmuddin MM, Usman R, Kuriakose K, Ahmed A, Rehman MEU, Roksana Z, Syed Z, Anwer F, Raza S. Management of Adverse Reactions for BCMA-Directed Therapy in Relapsed Multiple Myeloma: A Focused Review. J Clin Med 2023; 12:5539. [PMID: 37685606 PMCID: PMC10487885 DOI: 10.3390/jcm12175539] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/26/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Anti-B-cell maturation antigen therapies consisting of bispecific antibodies, antibody-drug conjugates, and chimeric antigen receptor T cells have shown promising results in relapsed refractory multiple myeloma (RRMM). However, the severe side effects include cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, cytopenia(s), infections, hemophagocytic lymphohistiocytosis, and organ toxicity, which could sometimes be life-threatening. This review focuses on these most common complications post-BCMA therapy. We discussed the risk factors, pathogenesis, clinical features associated with these complications, and how to prevent and treat them. We included four original studies for this focused review. All four agents (idecabtagene vicleucel, ciltacabtagene autoleucel, teclistamab, belantamab mafodotin) have received FDA approval for adult RRMM patients. We went through the FDA access data packages of the approved agents to outline stepwise management of the complications for better patient outcomes.
Collapse
Affiliation(s)
- Razwana Khanam
- Department of Hospital Medicine, Baystate Medical Center, Springfield, MA 01199, USA
| | - Beth Faiman
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH 44106, USA; (B.F.); (F.A.); (S.R.)
| | - Saba Batool
- Department of Hospital Medicine, Carle Health Methodist Hospital, Peoria, IL 61636, USA;
| | | | - Rana Usman
- University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Kiran Kuriakose
- Department of Hospital Medicine, UPMC Mercy Hospital, Pittsburgh, PA 15219, USA;
| | - Arooj Ahmed
- Department of Translational Hematology and Oncology, Cleveland Clinic Taussig Cancer Center, Cleveland, OH 44195, USA;
| | | | - Zinath Roksana
- Sheikh Hasina National Institute of Burn and Plastic Surgery, Dhaka 1217, Bangladesh;
| | - Zain Syed
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Faiz Anwer
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH 44106, USA; (B.F.); (F.A.); (S.R.)
| | - Shahzad Raza
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH 44106, USA; (B.F.); (F.A.); (S.R.)
| |
Collapse
|
13
|
Dubbs SB, Falat C, Rosenblatt L. Immune-based Therapies-What the Emergency Physician Needs to Know. Immunol Allergy Clin North Am 2023; 43:569-582. [PMID: 37394260 DOI: 10.1016/j.iac.2022.10.004] [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: 07/04/2023]
Abstract
Immunotherapy is a treatment modality that has a broad and rapidly growing range of applications to treat both chronic and acute diseases, including rheumatoid arthritis, Crohn disease, cancer, and COVID-19. Emergency physicians must be aware of the breadth of applications and be able to consider the effects of immunotherapies when patients on these treatments present to the hospital. This article provides a review of the mechanisms of action, indications for use, and potential complications of immunotherapy treatments that are relevant in the emergency care setting.
Collapse
Affiliation(s)
- Sarah B Dubbs
- Department of Emergency Medicine, University of Maryland School of Medicine, 110 South Paca Street, 6th Floor, Suite 200, Baltimore, MD 21201, USA.
| | - Cheyenne Falat
- Department of Emergency Medicine, University of Maryland School of Medicine, 110 South Paca Street, 6th Floor, Suite 200, Baltimore, MD 21201, USA
| | - Lauren Rosenblatt
- Department of Emergency Medicine, University of Maryland School of Medicine, 110 South Paca Street, 6th Floor, Suite 200, Baltimore, MD 21201, USA
| |
Collapse
|
14
|
Rafaniello C, Liguori V, Zinzi A, Gaio M, Falco A, Di Costanzo L, Gargano F, Trimarco V, Cataldi M, Capuano A. A Pharmacovigilance Study on the Safety of Axicabtagene Ciloleucel Based on Spontaneous Reports from the EudraVigilance Database. Biomedicines 2023; 11:2162. [PMID: 37626659 PMCID: PMC10452324 DOI: 10.3390/biomedicines11082162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/27/2023] [Accepted: 07/29/2023] [Indexed: 08/27/2023] Open
Abstract
During pre-approval clinical trials, the safety of axi-cel, a second-generation CAR-T-cell therapy directed against CD19, which dramatically improved the prognosis of intractable B-cell lymphomas, has been investigated only in about 400 patients. Therefore, additional information on this issue is urgently needed. In the present paper, we evaluated the 2905 ICSRs with axi-cel as the suspected drug that had been uploaded in the EudraVigilance database from 1 January 2018 to 31 December 2022. About 80% of the reported adverse events were serious, and about 20% of them did not fully resolve or caused death. The adverse events most-frequently reported were Nervous system disorders (25.6%) and, among them, immune-effector-cell-associated neurotoxicity syndrome, followed by Immune system disorders (23.1%), General disorders and administration site conditions (12.0%), Blood and lymphatic system disorders (7.2%), and Infections and infestations (5.8%). Disproportionality analysis showed that the frequency of reported adverse events related to the nervous system was higher with axi-cel than with the other approved CAR-T-cells, except brexu-cel. In conclusion, real-world pharmacovigilance data showed that nervous system and immune system disorders are the adverse events most reported in axi-cel-related ICSRs and suggest that axi-cel could be more neurotoxic than other CAR-T-cells.
Collapse
Affiliation(s)
- Concetta Rafaniello
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; (C.R.); (V.L.); (A.Z.); (M.G.); (A.F.)
- Section of Pharmacology “L. Donatelli”, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Valerio Liguori
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; (C.R.); (V.L.); (A.Z.); (M.G.); (A.F.)
- Section of Pharmacology “L. Donatelli”, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Alessia Zinzi
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; (C.R.); (V.L.); (A.Z.); (M.G.); (A.F.)
- Section of Pharmacology “L. Donatelli”, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Mario Gaio
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; (C.R.); (V.L.); (A.Z.); (M.G.); (A.F.)
- Section of Pharmacology “L. Donatelli”, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Angela Falco
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; (C.R.); (V.L.); (A.Z.); (M.G.); (A.F.)
- Section of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, Federico II University of Naples, Via Sergio Pansini 5, 80131 Naples, Italy;
| | - Luigi Di Costanzo
- Section of Pharmacology “L. Donatelli”, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Francesca Gargano
- Department of Anesthesia and Resuscitation, Biomedical Campus University of Rome, 00128 Rome, Italy;
| | - Valentina Trimarco
- Section of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, Federico II University of Naples, Via Sergio Pansini 5, 80131 Naples, Italy;
| | - Mauro Cataldi
- Section of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, Federico II University of Naples, Via Sergio Pansini 5, 80131 Naples, Italy;
| | - Annalisa Capuano
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy; (C.R.); (V.L.); (A.Z.); (M.G.); (A.F.)
- Section of Pharmacology “L. Donatelli”, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| |
Collapse
|
15
|
Worel N, Holbro A, Vrielink H, Ootjers C, Le Poole K, Beer-Wekking I, Rintala T, Lozano M, Bonig H. A guide to the collection of T-cells by apheresis for ATMP manufacturing-recommendations of the GoCART coalition apheresis working group. Bone Marrow Transplant 2023; 58:742-748. [PMID: 37024570 DOI: 10.1038/s41409-023-01957-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/03/2023] [Accepted: 03/13/2023] [Indexed: 04/08/2023]
Abstract
Autologous chimeric antigen receptor-modified T-cells (CAR-T) provide meaningful benefit for otherwise refractory malignancies. As clinical indications for CAR-T cells are expanding, hospitals hitherto not active in the field of immune effector cell therapy will need to build capacity and expertise. The GoCART Coalition seeks to disseminate knowledge and skills to facilitate the introduction of CAR-T cells and to standardize management and documentation of CAR-T cell recipients, in order to optimize outcomes and to be able to benchmark clinical results against other centers. Apheresis generates the starting material for CAR-T cell manufacturing. This guide provides some initial suggestions for patient's apheresis readiness and performance to collect starting material and should thus facilitate the implementation of a CAR-T-starting material apheresis facility. It cannot replace, of course, the extensive training needed to perform qualitative apheresis collections in compliance with national and international regulations and assess their cellular composition and biological safety.
Collapse
Affiliation(s)
- Nina Worel
- Department for Transfusion Medicine and Cell Therapy, Medical University of Vienna, Vienna, Austria.
| | - Andreas Holbro
- Regional Blood Transfusion Service, Swiss Red Cross, Basel, Switzerland
- Division of Hematology, University Hospital Basel and University Basel, Basel, Switzerland
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
| | - Hans Vrielink
- Department for Transfusion Medicine, Sanquin Blood Supply, Amsterdam, The Netherlands
| | - Claudia Ootjers
- Department of Hematology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Kaatje Le Poole
- Department for Transfusion Medicine, Sanquin Blood Supply, Amsterdam, The Netherlands
| | - Ingrid Beer-Wekking
- Department of Hematology, Leiden University Medical Centre, Leiden, The Netherlands
| | | | - Miquel Lozano
- Apheresis and Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, University Clinic Hospital, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology, Goethe University, Frankfurt a.M., Germany
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA
| |
Collapse
|
16
|
Amberger M, Grueso E, Ivics Z. CRISISS: A Novel, Transcriptionally and Post-Translationally Inducible CRISPR/Cas9-Based Cellular Suicide Switch. Int J Mol Sci 2023; 24:9799. [PMID: 37372948 DOI: 10.3390/ijms24129799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/23/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
With the ever-increasing developing rate of gene and cellular therapy applications and growing accessibility due to products receiving regulatory approval, the need for effective and reliable safety mechanisms to prevent or eliminate potentially fatal side effects is of the utmost importance. In this study, we present the CRISPR-induced suicide switch (CRISISS) as a tool to eliminate genetically modified cells in an inducible and highly efficient manner by targeting Cas9 to highly repetitive Alu retrotransposons in the human genome, causing irreparable genomic fragmentation by the Cas9 nuclease and resulting cell death. The suicide switch components, including expression cassettes for a transcriptionally and post-translationally inducible Cas9 and an Alu-specific single-guide RNA, were integrated into the genome of target cells via Sleeping-Beauty-mediated transposition. The resulting transgenic cells did not show signs of any impact on overall fitness when uninduced, as unintended background expression, background DNA damage response and background cell killing were not observed. When induced, however, a strong expression of Cas9, a strong DNA damage response and a rapid halt of cell proliferation coupled with near complete cell death within four days post-induction were seen. With this proof-of-concept study, we present a novel and promising approach for a robust suicide switch with potential utility for gene and cell therapy in the future.
Collapse
Affiliation(s)
- Maximilian Amberger
- Research Center, Division of Hematology, Gene and Cell Therapy, Paul-Ehrlich-Institute, 63225 Langen, Germany
| | - Esther Grueso
- Research Center, Division of Hematology, Gene and Cell Therapy, Paul-Ehrlich-Institute, 63225 Langen, Germany
| | - Zoltán Ivics
- Research Center, Division of Hematology, Gene and Cell Therapy, Paul-Ehrlich-Institute, 63225 Langen, Germany
| |
Collapse
|
17
|
Gu C, Wu Q, Zhang J, Kang L, Yu L, Qiu H, Wu D, Chen S, Yang X. Successful treatment of severe cytokine release syndrome after CAR-T therapy by ruxolitinib without compromising CAR-T efficacy. Leuk Lymphoma 2023; 64:495-498. [PMID: 36395278 DOI: 10.1080/10428194.2022.2148209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Chengyuan Gu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Qian Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jingren Zhang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Liqing Kang
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China.,Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd, Shanghai, China
| | - Lei Yu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China.,Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd, Shanghai, China
| | - Huiying Qiu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Suning Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xiaofei Yang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
18
|
Gribkova IV. CAR NK-сells for the treatment of hematological malignancies: A review. JOURNAL OF MODERN ONCOLOGY 2022. [DOI: 10.26442/18151434.2022.3.201699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hematological malignant neoplasms include more than a hundred different subtypes and account for about 4.8% of all neoplastic diseases in Russia. Despite significant advances in diagnosis and treatment, many of them remain incurable. In recent years, cell-based therapy appears to be a promising approach to the treatment of these incurable hematologic malignancies, showing striking results in various clinical trials. The most studied and advanced cell therapy is the therapy with T-lymphocytes modified with chimeric antigen receptors (CAR). However, although the US Food and Drug Administration has approved CAR T cells for the treatment of B-cell lymphoma and acute lymphoblastic leukemia, significant problems remain in terms of production, cost, and serious side effects. An alternative to the use of T cells can be the use of innate immune cells, in particular natural killer cells (NK), which have a high antitumor potential. Recent studies have shown the antitumor efficacy of a therapy that uses genetically modified natural killer cells CAR NK cells. The purpose of this review was to describe and systematize the experience of using CAR NK cells for the treatment of hematological neoplasms. The review presents the advantages and disadvantages of this method, as well as the problems that still have to be solved for its widespread introduction into clinical practice.
Collapse
|
19
|
The use of ICU resources in CAR-T cell recipients: a hospital-wide study. Ann Intensive Care 2022; 12:75. [PMID: 35976532 PMCID: PMC9385897 DOI: 10.1186/s13613-022-01036-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/17/2022] [Indexed: 11/10/2022] Open
Abstract
Background CAR-T cell (chimeric antigen receptor T) therapy has emerged as an effective treatment of refractory hematological malignancies. Intensive care management is intrinsic to CAR-T cell therapy. We aim to describe and to assess outcomes in critically ill CAR-T cell recipients. Study design and methods Hospital-wide retrospective study. Consecutive CAR-T cell recipients requiring ICU admission from July 2017 and December 2020 were included. Results 71 patients (median age 60 years [37–68]) were admitted to the ICU 6 days [4–7] after CAR-T cell infusion. Underlying malignancies included diffuse large B cell lymphoma (n = 53, 75%), acute lymphoblastic leukemia (17 patients, 24%) and multiple myeloma (n = 1, 1.45%). Performance status (PS) was 1 [1–2]. Shock was the main reason for ICU admission (n = 40, 48%). Isolated cytokine release syndrome (CRS) was the most common complication (n = 33, 46%), while 21 patients (30%) had microbiologically documented bacterial infection (chiefly catheter-related infection). Immune effector cell-associated neurotoxicity syndrome was reported in 26 (37%) patients. At ICU admission, vasopressors were required in 18 patients (25%) and invasive mechanical ventilation in two. Overall, 49 (69%) and 40 patients (56%) received tocilizumab or steroids, respectively. Determinant of mortality were the reason for ICU admission (disease progression vs. sepsis or CRS (HR 4.02 [95%CI 1.10–14.65]), Performance status (HR 1.97/point [95%CI 1.14–3.41]) and SOFA score (HR 1.16/point [95%CI 1.01–1.33]). Conclusions Meaningful survival could be achieved in up to half the CAR-T cell recipients. The severity of organ dysfunction is a major determinant of death, especially in patients with altered performance status or disease progression. Supplementary Information The online version contains supplementary material available at 10.1186/s13613-022-01036-2.
Collapse
|
20
|
Ferreros P, Trapero I. Interleukin Inhibitors in Cytokine Release Syndrome and Neurotoxicity Secondary to CAR-T Therapy. Diseases 2022; 10:41. [PMID: 35892735 PMCID: PMC9326641 DOI: 10.3390/diseases10030041] [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: 06/06/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Chimeric antigen receptor T-cell (CAR-T) therapy is an innovative therapeutic option for addressing certain recurrent or refractory hematological malignancies. However, CAR-T cells also cause the release of pro-inflammatory cytokines that lead to life-threatening cytokine release syndrome and neurotoxicity. OBJECTIVE To study the efficacy of interleukin inhibitors in addressing cytokine release syndrome (CRS) and neurotoxicity secondary to CAR-T therapy. METHODOLOGY The authors conducted a bibliographic review in which 10 articles were analyzed. These included cut-off studies, case reports, and clinical trials involving 11 cancer centers and up to 475 patients over 18 years of age. RESULTS Tocilizumab is the only interleukin inhibitor approved to address CRS secondary to CAR-T therapy due to its efficacy and safety. Other inhibitors, such as siltuximab and anakinra, could be useful in combination with tocilizumab for preventing severe cytokine release and neurotoxicity. In addition, the new specific inhibitors could be effective in mitigating CRS without affecting the cytotoxic efficacy of CAR-T therapy. CONCLUSION More lines of research should be opened to elucidate the true implications of these drugs in treating the side effects of CAR-T therapy.
Collapse
Affiliation(s)
- Puri Ferreros
- Nursing Department, Faculty of Nursing and Podiatry, University of Valencia, 46010 Valencia, Spain;
| | | |
Collapse
|
21
|
Crofton KM, Bassan A, Behl M, Chushak YG, Fritsche E, Gearhart JM, Marty MS, Mumtaz M, Pavan M, Ruiz P, Sachana M, Selvam R, Shafer TJ, Stavitskaya L, Szabo DT, Szabo ST, Tice RR, Wilson D, Woolley D, Myatt GJ. Current status and future directions for a neurotoxicity hazard assessment framework that integrates in silico approaches. COMPUTATIONAL TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 22:100223. [PMID: 35844258 PMCID: PMC9281386 DOI: 10.1016/j.comtox.2022.100223] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
Neurotoxicology is the study of adverse effects on the structure or function of the developing or mature adult nervous system following exposure to chemical, biological, or physical agents. The development of more informative alternative methods to assess developmental (DNT) and adult (NT) neurotoxicity induced by xenobiotics is critically needed. The use of such alternative methods including in silico approaches that predict DNT or NT from chemical structure (e.g., statistical-based and expert rule-based systems) is ideally based on a comprehensive understanding of the relevant biological mechanisms. This paper discusses known mechanisms alongside the current state of the art in DNT/NT testing. In silico approaches available today that support the assessment of neurotoxicity based on knowledge of chemical structure are reviewed, and a conceptual framework for the integration of in silico methods with experimental information is presented. Establishing this framework is essential for the development of protocols, namely standardized approaches, to ensure that assessments of NT and DNT based on chemical structures are generated in a transparent, consistent, and defendable manner.
Collapse
Affiliation(s)
| | - Arianna Bassan
- Innovatune srl, Via Giulio Zanon 130/D, 35129 Padova,
Italy
| | - Mamta Behl
- Division of the National Toxicology Program, National
Institutes of Environmental Health Sciences, Durham, NC 27709, USA
| | - Yaroslav G. Chushak
- Henry M Jackson Foundation for the Advancement of Military
Medicine, Wright-Patterson AFB, OH 45433, USA
| | - Ellen Fritsche
- IUF – Leibniz Research Institute for Environmental
Medicine & Medical Faculty Heinrich-Heine-University, Düsseldorf,
Germany
| | - Jeffery M. Gearhart
- Henry M Jackson Foundation for the Advancement of Military
Medicine, Wright-Patterson AFB, OH 45433, USA
| | | | - Moiz Mumtaz
- Agency for Toxic Substances and Disease Registry, US
Department of Health and Human Services, Atlanta, GA, USA
| | - Manuela Pavan
- Innovatune srl, Via Giulio Zanon 130/D, 35129 Padova,
Italy
| | - Patricia Ruiz
- Agency for Toxic Substances and Disease Registry, US
Department of Health and Human Services, Atlanta, GA, USA
| | - Magdalini Sachana
- Environment Health and Safety Division, Environment
Directorate, Organisation for Economic Co-Operation and Development (OECD), 75775
Paris Cedex 16, France
| | - Rajamani Selvam
- Office of Clinical Pharmacology, Office of Translational
Sciences, Center for Drug Evaluation and Research (CDER), U.S. Food and Drug
Administration (FDA), Silver Spring, MD 20993, USA
| | - Timothy J. Shafer
- Biomolecular and Computational Toxicology Division, Center
for Computational Toxicology and Exposure, US EPA, Research Triangle Park, NC,
USA
| | - Lidiya Stavitskaya
- Office of Clinical Pharmacology, Office of Translational
Sciences, Center for Drug Evaluation and Research (CDER), U.S. Food and Drug
Administration (FDA), Silver Spring, MD 20993, USA
| | | | | | | | - Dan Wilson
- The Dow Chemical Company, Midland, MI 48667, USA
| | | | - Glenn J. Myatt
- Instem, Columbus, OH 43215, USA
- Corresponding author.
(G.J. Myatt)
| |
Collapse
|
22
|
Paul B, Rodriguez C, Usmani SZ. BCMA-Targeted Biologic Therapies: The Next Standard of Care in Multiple Myeloma Therapy. Drugs 2022; 82:613-631. [PMID: 35412114 PMCID: PMC9554894 DOI: 10.1007/s40265-022-01697-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2022] [Indexed: 11/03/2022]
Abstract
With recent advances in myeloma therapy, patients can achieve long-term remissions, but eventually relapses will occur. Triple-class refractory myeloma (disease that is refractory to an immunomodulatory agent, a proteasome inhibitor, and an anti-CD38 monoclonal antibody) and penta-refractory myeloma (disease that is refractory to two proteasome inhibitors, two immunomodulatory agents, and an anti-CD38 antibody) are associated with a particularly poor prognosis, and novel treatments are desperately needed for these patients. Targeting B cell maturation antigen (BCMA), which is ubiquitously expressed on plasma cells, has emerged as a well-tolerated and highly efficacious strategy in patients with relapsed and refractory myeloma. Several mechanisms of targeting BCMA are currently under investigation, including antibody-drug conjugates, bispecific antibodies, and chimeric antigen receptor T cells and natural killer (NK) cells, all with unique side effect profiles. Early phase clinical trials showed unprecedented response rates in highly refractory myeloma patients, leading to the recent approvals of some of these agents. Still, many questions remain with regard to this target, including how best to target it, how to treat patients who have progressed on a BCMA-targeting therapy, and whether response rates will deepen if these agents are used in earlier lines of therapy. In this review, we examine the rationale for targeting BCMA and summarize the data for several agents across multiple classes of BCMA-targeting therapeutics, paying special attention to the diverse mechanisms and unique challenges of each therapeutic class.
Collapse
Affiliation(s)
- Barry Paul
- Division of Plasma Cell Disorders, Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute/Atrium Health, Charlotte, NC, USA
| | | | - Saad Z Usmani
- Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
23
|
Reikvam H, Hatfield KJ, Wendelbo Ø, Lindås R, Lassalle P, Bruserud Ø. Endocan in Acute Leukemia: Current Knowledge and Future Perspectives. Biomolecules 2022; 12:biom12040492. [PMID: 35454082 PMCID: PMC9027427 DOI: 10.3390/biom12040492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 11/16/2022] Open
Abstract
Endocan is a soluble dermatan sulfate proteoglycan expressed by endothelial cells and detected in serum/plasma. Its expression is increased in tumors/tumor vessels in several human malignancies, and high expression (high serum/plasma levels or tumor levels) has an adverse prognostic impact in several malignancies. The p14 endocan degradation product can also be detected in serum/plasma, but previous clinical studies as well as previously unpublished results presented in this review suggest that endocan and p14 endocan fragment levels reflect different biological characteristics, and the endocan levels seem to reflect the disease heterogeneity in acute leukemia better than the p14 fragment levels. Furthermore, decreased systemic endocan levels in previously immunocompetent sepsis patients are associated with later severe respiratory complications, but it is not known whether this is true also for immunocompromised acute leukemia patients. Finally, endocan is associated with increased early nonrelapse mortality in (acute leukemia) patients receiving allogeneic stem cell transplantation, and this adverse prognostic impact seems to be independent of the adverse impact of excessive fluid overload. Systemic endocan levels may also become important to predict cytokine release syndrome after immunotherapy/haploidentical transplantation, and in the long-term follow-up of acute leukemia survivors with regard to cardiovascular risk. Therapeutic targeting of endocan is now possible, and the possible role of endocan in acute leukemia should be further investigated to clarify whether the therapeutic strategy should also be considered.
Collapse
Affiliation(s)
- Håkon Reikvam
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway;
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (Ø.W.); (R.L.)
| | - Kimberley Joanne Hatfield
- Department of Transfusion Medicine and Immunology, Haukeland University Hospital, 5021 Bergen, Norway;
| | - Øystein Wendelbo
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (Ø.W.); (R.L.)
| | - Roald Lindås
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (Ø.W.); (R.L.)
| | - Philippe Lassalle
- Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, U1019-UMR9017, University of Lille, 59000 Lille, France;
- Center for Infection and Immunity, le Centre Nationale de la Recherche Scientifique, Univeristy of Lille, 59000 Lille, France
- Centre d’Infection et d’Immunité de Lille, Equipe Immunité Pulmonaire, University of Lille, 59000 Lille, France
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway;
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (Ø.W.); (R.L.)
- Correspondence:
| |
Collapse
|
24
|
Obaisi O, Fontillas RC, Patel K, Ngo-Huang A. Rehabilitation Needs for Patients Undergoing CAR T-Cell Therapy. Curr Oncol Rep 2022; 24:741-749. [PMID: 35267151 PMCID: PMC8907385 DOI: 10.1007/s11912-022-01240-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2022] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Chimeric antigen receptor (CAR) T-cell therapy is a relatively new, innovative treatment strategy to manage refractory hematological cancers, including some types of leukemia, lymphoma, and multiple myeloma. This article outlines the CAR T-cell therapy process, toxicity, and complications, along with an overview of the currently known short- and long-term physical and functional sequelae that will be helpful for general or oncology rehabilitation specialists caring for these patients. RECENT FINDINGS There is a dearth of literature on the topic of rehabilitation of patients receiving CAR T-cell therapy. Rehabilitation practices can be extrapolated from the limited functional information on patients who have completed treatment for lymphoma and multiple myeloma. Patients present with cognitive impairment, muscle weakness, reduced exercise capacity, neuropathy, and cancer-related fatigue. Physical activity and rehabilitation programs may be beneficial to address fatigue, psychological symptoms, and quality of life. There is limited rehabilitation research in patients receiving CAR T-cell therapy. These patients may present with general deconditioning and neurological complications which translate to neuromuscular and cognitive impairment that benefit from multidisciplinary rehabilitation intervention prior to, during, and after treatment. Studies measuring the impairments at baseline and evaluation of the impact of rehabilitation practices are much needed to support this.
Collapse
Affiliation(s)
- Obada Obaisi
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Unit 1414, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Rhodora C Fontillas
- Department of Rehabilitation Services, The University of Texas MD Anderson Cancer Center, Unit 0322, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Krina Patel
- Department of Lymphoma-Myeloma, The University of Texas MD Anderson Cancer Center, Unit 0429, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - An Ngo-Huang
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Unit 1414, 1515 Holcombe Boulevard, Houston, TX, 77030, USA.
| |
Collapse
|
25
|
Mirzaee Godarzee M, Mahmud Hussen B, Razmara E, Hakak‐Zargar B, Mohajerani F, Dabiri H, Fatih Rasul M, Ghazimoradi MH, Babashah S, Sadeghizadeh M. Strategies to overcome the side effects of chimeric antigen receptor T cell therapy. Ann N Y Acad Sci 2022; 1510:18-35. [DOI: 10.1111/nyas.14724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/05/2021] [Accepted: 10/22/2021] [Indexed: 11/26/2022]
Affiliation(s)
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy Hawler Medical University Erbil Iraq
| | - Ehsan Razmara
- Australian Regenerative Medicine Institute Monash University, Clayton, Victoria, Australia, 3800
| | | | - Fatemeh Mohajerani
- Department of Molecular Genetics, Faculty of Biological Sciences Tarbiat Modares University Tehran Iran
| | - Hamed Dabiri
- Department of Molecular Genetics, Faculty of Biological Sciences Tarbiat Modares University Tehran Iran
| | - Mohammed Fatih Rasul
- Department of Medical Analysis, Faculty of Sciences Tishk International University Erbil Iraq
| | | | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences Tarbiat Modares University Tehran Iran
| | - Majid Sadeghizadeh
- Department of Molecular Genetics, Faculty of Biological Sciences Tarbiat Modares University Tehran Iran
| |
Collapse
|
26
|
Hwang EI, Sayour EJ, Flores CT, Grant G, Wechsler-Reya R, Hoang-Minh LB, Kieran MW, Salcido J, Prins RM, Figg JW, Platten M, Candelario KM, Hale PG, Blatt JE, Governale LS, Okada H, Mitchell DA, Pollack IF. The current landscape of immunotherapy for pediatric brain tumors. NATURE CANCER 2022; 3:11-24. [PMID: 35121998 DOI: 10.1038/s43018-021-00319-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/24/2021] [Indexed: 02/06/2023]
Abstract
Pediatric central nervous system tumors are the most common solid malignancies in childhood, and aggressive therapy often leads to long-term sequelae in survivors, making these tumors challenging to treat. Immunotherapy has revolutionized prospects for many cancer types in adults, but the intrinsic complexity of treating pediatric patients and the scarcity of clinical studies of children to inform effective approaches have hampered the development of effective immunotherapies in pediatric settings. Here, we review recent advances and ongoing challenges in pediatric brain cancer immunotherapy, as well as considerations for efficient clinical translation of efficacious immunotherapies into pediatric settings.
Collapse
Affiliation(s)
- Eugene I Hwang
- Division of Oncology, Brain Tumor Institute, Children's National Hospital, Washington, DC, USA.
| | - Elias J Sayour
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Catherine T Flores
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Gerald Grant
- Division of Pediatric Neurosurgery, Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA, USA
| | - Robert Wechsler-Reya
- Tumor Initiation & Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Lan B Hoang-Minh
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | | | | | - Robert M Prins
- Departments of Neurosurgery and Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - John W Figg
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University and CCU Brain Tumor Immunology, DKFZ, Heidelberg, Germany
| | - Kate M Candelario
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Paul G Hale
- Children's Brain Trust, Coral Springs, FL, USA
| | - Jason E Blatt
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Lance S Governale
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Hideho Okada
- Department of Neurosurgery, University of California, San Francisco, CA, USA
| | - Duane A Mitchell
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Ian F Pollack
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
27
|
Attiq A, Yao LJ, Afzal S, Khan MA. The triumvirate of NF-κB, inflammation and cytokine storm in COVID-19. Int Immunopharmacol 2021; 101:108255. [PMID: 34688149 PMCID: PMC8516728 DOI: 10.1016/j.intimp.2021.108255] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/30/2021] [Accepted: 10/09/2021] [Indexed: 01/08/2023]
Abstract
The coronavirus disease (COVID-19) has once again reminded us of the significance of host immune response and consequential havocs of the immune dysregulation. The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) inflicts severe complications to the infected host, including cough, dyspnoea, fever, septic shock, acute respiratory distress syndrome (ARDs), and multiple organ failure. These manifestations are the consequence of the dysregulated immune system, which gives rise to excessive and unattended production of pro-inflammatory mediators. Elevated circulatory cytokine and chemokine levels are accompanied by spontaneous haemorrhage, thrombocytopenia and systemic inflammation, which are the cardinal features of life-threatening cytokine storm syndrome in advanced COVID-19 diseases. Coronavirus hijacked NF-kappa B (NF-κB) is responsible for upregulating the expressions of inflammatory cytokine, chemokine, alarmins and inducible enzymes, which paves the pathway for cytokine storm. Given the scenario, the systemic approach of simultaneous inhibition of NF-κB offers an attractive therapeutic intervention. Targeted therapies with proteasome inhibitor (VL-01, bortezomib, carfilzomib and ixazomib), bruton tyrosine kinase inhibitor (acalabrutinib), nucleotide analogue (remdesivir), TNF-α monoclonal antibodies (infliximab and adalimumab), N-acetylcysteine and corticosteroids (dexamethasone), focusing the NF-κB inhibition have demonstrated effectiveness in terms of the significant decrease in morbidity and mortality in severe COVID-19 patients. Hence, this review highlights the activation, signal transduction and cross-talk of NF-κB with regard to cytokine storm in COVID-19. Moreover, the development of therapeutic strategies based on NF-κB inhibition are also discussed herein.
Collapse
Affiliation(s)
- Ali Attiq
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, MAHSA University, Bandar Saujana Putra, 42610 Jenjarom, Selangor, Malaysia.
| | - Lui Jin Yao
- Kuala Balah Health Clinic (Klinik Kesihatan Kuala Balah), Kuala Balah, 17600 Jeli, Kelantan, Malaysia
| | - Sheryar Afzal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, MAHSA University, Bandar Saujana Putra, 42610 Jenjarom, Selangor, Malaysia
| | - Mansoor Ali Khan
- COVID-19 Vaccination Centres, University College London Hospitals, National Health Service, N10QH London, England
| |
Collapse
|
28
|
Schaefer A, Huang Y, Kittai A, Maakaron JE, Saygin C, Brammer J, Penza S, Saad A, Jaglowski SM, William BM. Cytopenias After CD19 Chimeric Antigen Receptor T-Cells (CAR-T) Therapy for Diffuse Large B-Cell Lymphomas or Transformed Follicular Lymphoma: A Single Institution Experience. Cancer Manag Res 2021; 13:8901-8906. [PMID: 34876852 PMCID: PMC8643129 DOI: 10.2147/cmar.s321202] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/18/2021] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Patients with relapsed/refractory diffuse large B-cell lymphoma (DLBCL) have poor outcomes. Treatment with CD19 chimeric antigen receptor (CAR-T) cells, tisagenlecleucel and axicabtagene ciloleucel, has been associated with improved outcomes. Cytopenias were observed in clinical trials with both products; however, little is known regarding the patterns and outcomes of these cytopenias. SUBJECTS AND METHODS We reviewed DLBCL patients (n=32) receiving either product between January and September 2018 at our institution. RESULTS Median duration of leukopenia, neutropenia, lymphopenia, anemia, and thrombocytopenia was 49, 9, 117.5, 125, and 95.5 days after CAR-T infusion, respectively. Filgrastim was used in 63% of patients, and 50% of patients received red cell or platelet transfusions. With the exception of neutropenia, increase in the duration of cytopenia of any lineage was associated with improvement in progression-free survival, and in overall survival in case of anemia. There was no association between the duration of cytopenias with either cytokine release syndrome or neurotoxicity. DISCUSSION Our data suggest a correlation between cytopenias and survival outcomes after CD19 CAR-T therapy. If validated, cytopenia may be proven useful as a biomarker of response and survival after CAR-T therapy.
Collapse
Affiliation(s)
- Andrew Schaefer
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Ying Huang
- Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Adam Kittai
- Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Joseph E Maakaron
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Caner Saygin
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Jonathan Brammer
- Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Sam Penza
- Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Ayman Saad
- Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Basem M William
- OhioHealth Blood and Marrow Transplant Program, Columbus, OH, USA
| |
Collapse
|
29
|
Dubbs SB, Falat C, Rosenblatt L. Immune-based Therapies-What the Emergency Physician Needs to Know. Emerg Med Clin North Am 2021; 40:135-148. [PMID: 34782084 DOI: 10.1016/j.emc.2021.08.011] [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: 11/03/2022]
Abstract
Immunotherapy is a treatment modality that has a broad and rapidly growing range of applications to treat both chronic and acute diseases, including rheumatoid arthritis, Crohn disease, cancer, and COVID-19. Emergency physicians must be aware of the breadth of applications and be able to consider the effects of immunotherapies when patients on these treatments present to the hospital. This article provides a review of the mechanisms of action, indications for use, and potential complications of immunotherapy treatments that are relevant in the emergency care setting.
Collapse
Affiliation(s)
- Sarah B Dubbs
- Department of Emergency Medicine, University of Maryland School of Medicine, 110 South Paca Street, 6th Floor, Suite 200, Baltimore, MD 21201, USA.
| | - Cheyenne Falat
- Department of Emergency Medicine, University of Maryland School of Medicine, 110 South Paca Street, 6th Floor, Suite 200, Baltimore, MD 21201, USA
| | - Lauren Rosenblatt
- Department of Emergency Medicine, University of Maryland School of Medicine, 110 South Paca Street, 6th Floor, Suite 200, Baltimore, MD 21201, USA
| |
Collapse
|
30
|
Castaneda-Puglianini O, Chavez JC. Assessing and Management of Neurotoxicity After CAR-T Therapy in Diffuse Large B-Cell Lymphoma. J Blood Med 2021; 12:775-783. [PMID: 34466048 PMCID: PMC8403007 DOI: 10.2147/jbm.s281247] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/31/2021] [Indexed: 11/23/2022] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy represents the most important advances in cancer immunotherapy, especially in hematological malignancies such as B-cell lymphomas. CAR-T cell therapy has significant activity in poor risk B-cell lymphomas. CAR-T cell therapy is associated with potentially life-threatening toxicities such as cytokine release syndrome (CRS) and neurotoxicity (NT). While CRS pathophysiology and management are well established, the understanding and treatment of NT continues to develop. All current CAR-T products approved for DLBCL have been associated with NT with some differences in their severity. As cell therapies continue to advance and its access broadening, it will be imperative for clinicians to be aware of the signs and symptoms of NT, its stratification and basic management.
Collapse
Affiliation(s)
- Omar Castaneda-Puglianini
- Virginia Commonwealth University, Massey Cancer Center, Cellular Immunotherapies and Transplant Program, Richmond, VA, USA
| | - Julio C Chavez
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
31
|
Cosenza M, Sacchi S, Pozzi S. Cytokine Release Syndrome Associated with T-Cell-Based Therapies for Hematological Malignancies: Pathophysiology, Clinical Presentation, and Treatment. Int J Mol Sci 2021; 22:ijms22147652. [PMID: 34299273 PMCID: PMC8305850 DOI: 10.3390/ijms22147652] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/15/2021] [Accepted: 07/15/2021] [Indexed: 01/08/2023] Open
Abstract
Cytokines are a broad group of small regulatory proteins with many biological functions involved in regulating the hematopoietic and immune systems. However, in pathological conditions, hyperactivation of the cytokine network constitutes the fundamental event in cytokine release syndrome (CRS). During the last few decades, the development of therapeutic monoclonal antibodies and T-cell therapies has rapidly evolved, and CRS can be a serious adverse event related to these treatments. CRS is a set of toxic adverse events that can be observed during infection or following the administration of antibodies for therapeutic purposes and, more recently, during T-cell-engaging therapies. CRS is triggered by on-target effects induced by binding of chimeric antigen receptor (CAR) T cells or bispecific antibody to its antigen and by subsequent activation of bystander immune and non-immune cells. CRS is associated with high circulating concentrations of several pro-inflammatory cytokines, including interleukins, interferons, tumor necrosis factors, colony-stimulating factors, and transforming growth factors. Recently, considerable developments have been achieved with regard to preventing and controlling CRS, but it remains an unmet clinical need. This review comprehensively summarizes the pathophysiology, clinical presentation, and treatment of CRS caused by T-cell-engaging therapies utilized in the treatment of hematological malignancies.
Collapse
|
32
|
Meng Y, Deng B, Rong L, Li C, Song W, Ling Z, Xu J, Duan J, Wang Z, Chang AH, Feng X, Xiong X, Chen X, Pan J. Short-Interval Sequential CAR-T Cell Infusion May Enhance Prior CAR-T Cell Expansion to Augment Anti-Lymphoma Response in B-NHL. Front Oncol 2021; 11:640166. [PMID: 34277400 PMCID: PMC8279746 DOI: 10.3389/fonc.2021.640166] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/25/2021] [Indexed: 11/13/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy emerges as a new treatment for refractory or relapsed (r/r) B-cell non-Hodgkin lymphoma (B-NHL); however, the overall response rate (ORR) of which in the B-NHL patients is much lower compared to the patients with r/r B acute lymphoblastic leukemia (B-ALL). We previously confirmed that sequential infusions of CD20 and CD22 CAR-T cells significantly improved the prognosis of the B-NHL patients, while some advanced patients still progressed to death during these CAR-T cell treatments. In this study, we showed that timely sequential administration of the second CAR-T cells could enhance expansion of prior CAR-T cells with stronger tumor-killing capacity in vitro and in vivo. We further conducted compassionate treatments on two advanced B-NHL patients with short-interval sequential infusions of CD19/22/20 CAR-T cells. Disease progression was observed in both patients after primary CAR-T cell infusion but robust re-expansion of prior CAR-T cells and anti-tumor effects was induced by infusion of a secondary CAR-T cells. These results indicate sequential infusions of CAR-T cells with a short interval may improve therapeutic efficacy in the B-NHL patients by promoting expansion of prior CAR-T cells.
Collapse
Affiliation(s)
- Yuan Meng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Biping Deng
- Cytology Laboratory, Beijing Boren Hospital, Beijing, China
| | - Luan Rong
- Cytology Laboratory, Beijing Boren Hospital, Beijing, China
| | - Chuo Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Weiliang Song
- Department of Hematology, Beijing Boren Hospital, Beijing, China
| | - Zhuojun Ling
- Department of Hematology, Beijing Boren Hospital, Beijing, China
| | - Jinlong Xu
- Department of Hematology, Beijing Boren Hospital, Beijing, China
| | - Jiajia Duan
- Department of Hematology, Beijing Boren Hospital, Beijing, China
| | - Zelin Wang
- Department of Hematology, Beijing Boren Hospital, Beijing, China
| | - Alex H Chang
- Clinical Translational Research Center, Tongji University School of Medicine, Shanghai, China
| | - Xiaoming Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Xiujuan Xiong
- Department of Pathology, Basic Medical College of Nanchang University, Nanchang, China
| | - Xiaoli Chen
- Ganzhou Key Laboratory of Molecular Medicine, the Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, China
| | - Jing Pan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,State Key Laboratory of Experimental Hematology, Boren Clinical Translational Center, Department of Hematology, Beijing Boren Hospital, Beijing, China
| |
Collapse
|
33
|
Bichon A, Bourenne J, Gainnier M, Carvelli J. Capillary leak syndrome: State of the art in 2021. Rev Med Interne 2021; 42:789-796. [PMID: 34099313 DOI: 10.1016/j.revmed.2021.05.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/11/2021] [Accepted: 05/18/2021] [Indexed: 02/06/2023]
Abstract
Capillary leak syndrome (CLS) is an increasingly acknowledged multifaceted and potentially lethal disease. Initial nonspecific symptoms are followed by the intriguing CLS hallmark: the double paradox associating diffuse severe edema and hypovolemia, along with hemoconcentration and hypoalbuminemia. Spontaneous resolutive phase is often associated with poor outcome due to iatrogenic fluid overload during leak phase. CLS is mainly triggered by drugs (anti-tumoral therapies), malignancy, infections (mostly viruses) and inflammatory diseases. Its idiopathic form is named after its eponymous finder: Clarkson's disease. CLS pathophysiology involves a severe, transient and multifactorial endothelial disruption which mechanisms are still unclear. Empirical and based-on-experience treatment implies symptomatic care during the acute phase (with the eventual addition of drugs amplifying cAMP levels in the severest cases), and the prophylactic use of monthly polyvalent immunoglobulins to prevent relapses. As CLS literature is scattered, we aimed to collect and summarize the current knowledge on CLS to facilitate its diagnosis, understanding and management.
Collapse
Affiliation(s)
- A Bichon
- Service de réanimation des urgences, hôpital de la Timone, AP-HM, 264, rue Saint Pierre, 13005 Marseille, France.
| | - J Bourenne
- Service de réanimation des urgences, hôpital de la Timone, AP-HM, 264, rue Saint Pierre, 13005 Marseille, France
| | - M Gainnier
- Service de réanimation des urgences, hôpital de la Timone, AP-HM, 264, rue Saint Pierre, 13005 Marseille, France
| | - J Carvelli
- Service de réanimation des urgences, hôpital de la Timone, AP-HM, 264, rue Saint Pierre, 13005 Marseille, France
| |
Collapse
|
34
|
Akhoundi M, Mohammadi M, Sahraei SS, Sheykhhasan M, Fayazi N. CAR T cell therapy as a promising approach in cancer immunotherapy: challenges and opportunities. Cell Oncol (Dordr) 2021; 44:495-523. [PMID: 33759063 DOI: 10.1007/s13402-021-00593-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-modified T cell therapy has shown great potential in the immunotherapy of patients with hematologic malignancies. In spite of this striking achievement, there are still major challenges to overcome in CAR T cell therapy of solid tumors, including treatment-related toxicity and specificity. Also, other obstacles may be encountered in tackling solid tumors, such as their immunosuppressive microenvironment, the heterogeneous expression of cell surface markers, and the cumbersome arrival of T cells at the tumor site. Although several strategies have been developed to overcome these challenges, aditional research aimed at enhancing its efficacy with minimum side effects, the design of precise yet simplified work flows and the possibility to scale-up production with reduced costs and related risks is still warranted. CONCLUSIONS Here, we review main strategies to establish a balance between the toxicity and activity of CAR T cells in order to enhance their specificity and surpass immunosuppression. In recent years, many clinical studies have been conducted that eventually led to approved products. To date, the FDA has approved two anti-CD19 CAR T cell products for non-Hodgkin lymphoma therapy, i.e., axicbtagene ciloleucel and tisagenlecleucel. With all the advances that have been made in the field of CAR T cell therapy for hematologic malignancies therapy, ongoing studies are focused on optimizing its efficacy and specificity, as well as reducing the side effects. Also, the efforts are poised to broaden CAR T cell therapeutics for other cancers, especially solid tumors.
Collapse
Affiliation(s)
- Maryam Akhoundi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahsa Mohammadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Seyedeh Saeideh Sahraei
- Department of Reproductive Biology, Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran
| | - Mohsen Sheykhhasan
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran.
| | - Nashmin Fayazi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
35
|
Li C, Qi Y, Zhang Y, Chen Y, Feng J, Zhang X. Artificial Engineering of Immune Cells for Improved Immunotherapy. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Chuxin Li
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry Wuhan University Wuhan 430072 P.R. China
| | - Yongdan Qi
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry Wuhan University Wuhan 430072 P.R. China
| | - Yu Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry Wuhan University Wuhan 430072 P.R. China
| | - Yingge Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry Wuhan University Wuhan 430072 P.R. China
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry Wuhan University Wuhan 430072 P.R. China
| | - Xianzheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry Wuhan University Wuhan 430072 P.R. China
| |
Collapse
|
36
|
A Novel off-the-Shelf Trastuzumab-Armed NK Cell Therapy (ACE1702) Using Antibody-Cell-Conjugation Technology. Cancers (Basel) 2021; 13:cancers13112724. [PMID: 34072864 PMCID: PMC8199224 DOI: 10.3390/cancers13112724] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary Chimeric antigen receptor T cell therapy has shown its potency against hematologic malignancies in autologous settings but also limited success against solid tumors with severe adverse events, including fatal cases of cytokine releasing syndrome. The aim of this research is to develop a novel off-the-shelf natural killer cell therapy against HER2-expressing cancers using Antibody-Cell Conjugation (ACC) technology and the endogenous CD16-expressing oNK cell line. ACE1702, trastuzumab-armed oNK cells with γ irradiation and cryopreservation, present superior in vitro and in vivo potency against HER2-expressing cancer cells and shows no tumorigenic potential, indicating the clinical application fighting HER2-expressing solid tumors. These findings suggest that ACC technology can be applied to allogeneic immune cells to provide off-the-shelf therapies for cancer patients. Abstract Natural killer (NK) cells harbor efficient cytotoxicity against tumor cells without causing life-threatening cytokine release syndrome (CRS) or graft-versus-host disease (GvHD). When compared to chimeric antigen receptor (CAR) technology, Antibody-Cell Conjugation (ACC) technology has been developed to provide an efficient platform to arm immune cells with cancer-targeting antibodies to recognize and attack cancer cells. Recently, we established an endogenous CD16-expressing oNK cell line (oNK) with a favorable expression pattern of NK activation/inhibitory receptors. In this study, we applied ACC platform to conjugate oNK with trastuzumab and an anti-human epidermal growth factor receptor 2 (HER2) antibody. Trastuzumab-conjugated oNK, ACE-oNK-HER2, executed in vitro and in vivo cytotoxicity against HER2-expressing cancer cells and secretion of IFNγ. The irradiated and cryopreserved ACE-oNK-HER2, designated as ACE1702, retained superior HER2-specific in vitro and in vivo potency with no tumorigenic potential. In conclusion, this study provides the evidence to support the potential clinical application of ACE1702 as a novel off-the-shelf NK cell therapy against HER2-expressing solid tumors.
Collapse
|
37
|
Abstract
A paradigm shift has recently occurred in the field of cancer therapeutics. Traditional anticancer agents, such as chemotherapy, radiotherapy and small-molecule drugs targeting specific signalling pathways, have been joined by cellular immunotherapies based on T cell engineering. The rapid adoption of novel, patient-specific cellular therapies builds on scientific developments in tumour immunology, genetic engineering and cell manufacturing, best illustrated by the curative potential of chimeric antigen receptor (CAR) T cell therapy targeting CD19-expressing malignancies. However, the clinical benefit observed in many patients may come at a cost. In up to one-third of patients, significant toxicities occur that are directly associated with the induction of powerful immune effector responses. The most frequently observed immune-mediated toxicities are cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. This Review discusses our current understanding of their pathophysiology and clinical features, as well as the development of novel therapeutics for their prevention and/or management. This Review discusses our current understanding of the pathophysiological mechanisms of cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome associated with chimeric antigen receptor (CAR) T cell therapies, and how this might be used for the prevention or management of these toxicities.
Collapse
|
38
|
Luo Y, Song G, Liang S, Li F, Liu K. Research advances in chimeric antigen receptor-modified T-cell therapy (Review). Exp Ther Med 2021; 21:484. [PMID: 33790993 PMCID: PMC8005741 DOI: 10.3892/etm.2021.9915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
Chimeric antigen receptor (CAR)-modified T-cells are T-cells that have been genetically engineered to express CAR molecules to target specific surface antigens on tumor cells. CAR T-cell therapy, a novel cancer immunotherapy, has been attracting increasing attention, since it exhibited notable efficacy in the treatment of hematological tumors in clinical trials. However, for this type of therapy, challenges must be overcome in the treatment of solid tumors. Furthermore, certain side effects associated with CAR T-cell therapy, including cytokine release syndrome, immune effector cell-related neurotoxicity syndrome, tumor lysis syndrome and on-target off-tumor toxicity, must be taken into consideration. The present study provides a systematic review of the principle, clinical application, current challenges, possible solutions and future perspectives for CAR T-cell therapy.
Collapse
Affiliation(s)
- Yuxi Luo
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China.,The First Clinic of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Guiqin Song
- Department of Biology, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Shichu Liang
- The First Clinic of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Feifei Li
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
39
|
Johnson PC, Jacobson C, Yi A, Saucier A, Dhawale TM, Nelson A, Lavoie MW, Reynolds MJ, Topping CE, Frigault MJ, El-Jawahri A. Healthcare Utilization and End-of-Life Outcomes in Patients Receiving CAR T-Cell Therapy. J Natl Compr Canc Netw 2021; 19:928-934. [PMID: 33706257 PMCID: PMC11221604 DOI: 10.6004/jnccn.2020.7678] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/26/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND CAR T-cell therapy has revolutionized the treatment of patients with hematologic malignancies, but it can result in prolonged hospitalizations and serious toxicities. However, data on the impact of CAR T-cell therapy on healthcare utilization and end-of-life (EoL) outcomes are lacking. METHODS We conducted a retrospective analysis of 236 patients who received CAR T-cell therapy at 2 tertiary care centers from February 2016 through December 2019. We abstracted healthcare utilization and EoL outcomes from the electronic health record, including hospitalizations, receipt of ICU care, hospitalization and receipt of systemic therapy in the last 30 days of life, palliative care, and hospice referrals. RESULTS Most patients (81.4%; n=192) received axicabtagene ciloleucel. Overall, 28.1% of patients experienced a hospital readmission and 15.5% required admission to the ICU within 3 months of CAR T-cell therapy. Among the deceased cohort, 58.3% (49/84) were hospitalized and 32.5% (26/80) received systemic therapy in the last 30 days of life. Rates of palliative care and hospice referrals were 47.6% and 30.9%, respectively. In multivariable logistic regression, receipt of bridging therapy (odds ratio [OR], 3.15; P=.041), index CAR-T hospitalization length of stay >14 days (OR, 4.76; P=.009), hospital admission within 3 months of CAR T-cell infusion (OR, 4.29; P=.013), and indolent lymphoma transformed to diffuse large B-cell lymphoma (OR, 9.83; P=.012) were associated with likelihood of hospitalization in the last 30 days of life. CONCLUSIONS A substantial minority of patients receiving CAR T-cell therapy experienced hospital readmission or ICU utilization in the first 3 months after CAR T-cell therapy, and most deceased recipients of CAR T-cell therapy received intensive EoL care. These findings underscore the need for interventions to optimize healthcare delivery and EoL care for this population.
Collapse
Affiliation(s)
- P. Connor Johnson
- Department of Medicine, Division of Hematology & Oncology, Massachusetts General Hospital Cancer Center & Harvard Medical School
| | - Caron Jacobson
- Department of Medical Oncology, Center for Lymphoma, Dana-Farber Cancer Institute & Harvard Medical School
| | - Alisha Yi
- Department of Medicine, Division of Hematology & Oncology, Massachusetts General Hospital Cancer Center & Harvard Medical School
| | - Anna Saucier
- Department of Medical Oncology, Center for Lymphoma, Dana-Farber Cancer Institute & Harvard Medical School
| | - Tejaswini M. Dhawale
- Department of Medicine, Division of Hematology & Oncology, Massachusetts General Hospital Cancer Center & Harvard Medical School
| | - Ashley Nelson
- Department of Psychiatry, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts
| | - Mitchell W. Lavoie
- Department of Psychiatry, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts
| | - Mathew J. Reynolds
- Department of Psychiatry, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts
| | - Carlisle E.W. Topping
- Department of Psychiatry, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts
| | - Matthew J. Frigault
- Department of Medicine, Division of Hematology & Oncology, Massachusetts General Hospital Cancer Center & Harvard Medical School
| | - Areej El-Jawahri
- Department of Medicine, Division of Hematology & Oncology, Massachusetts General Hospital Cancer Center & Harvard Medical School
| |
Collapse
|
40
|
Plaze M, Attali D, Prot M, Petit AC, Blatzer M, Vinckier F, Levillayer L, Chiaravalli J, Perin-Dureau F, Cachia A, Friedlander G, Chrétien F, Simon-Loriere E, Gaillard R. Inhibition of the replication of SARS-CoV-2 in human cells by the FDA-approved drug chlorpromazine. Int J Antimicrob Agents 2021; 57:106274. [PMID: 33387629 PMCID: PMC7772996 DOI: 10.1016/j.ijantimicag.2020.106274] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 12/14/2020] [Accepted: 12/20/2020] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Urgent action is needed to fight the ongoing coronavirus disease 2019 (COVID-19) pandemic by reducing the number of infected cases, contagiousness and severity. Chlorpromazine (CPZ), an antipsychotic from the phenothiazine group, is known to inhibit clathrin-mediated endocytosis and has antiviral activity against severe acute respiratory syndrome coronavirus-1 (SARS-CoV-1) and Middle East respiratory syndrome coronavirus. The aim of this in-vitro study was to test CPZ against SARS-CoV-2 in monkey and human cells. MATERIALS AND METHODS Monkey VeroE6 cells and human alveolar basal epithelial A549-ACE2 cells were infected with SARS-CoV-2 in the presence of various concentrations of CPZ. Supernatants were harvested at day 2 and analysed by quantitative reverse transcription polymerase chain reaction (RT-qPCR) for the presence of SARS-CoV-2 RNA. Cell viability was assessed in non-infected cells. RESULTS CPZ was found to have antiviral activity against SARS-CoV-2 in monkey VeroE6 cells, with a half maximal inhibitory concentration (IC50) of 8.2 µM, half maximal cytotoxic concentration (CC50) of 13.5 µM, and selectivity index (SI) of 1.65. In human A549-ACE2 cells, CPZ was also found to have anti-SARS-CoV-2 activity, with IC50 of 11.3 µM, CC50 of 23.1 µM and SI of 2.04. DISCUSSION Although the measured SI values are low, the IC50 values measured in vitro may translate to CPZ dosages used in routine clinical practice because of the high biodistribution of CPZ in lungs and saliva. Also, the distribution of CPZ in brain could be of interest for treating or preventing neurological and psychiatric forms of COVID-19. CONCLUSIONS These preclinical findings support clinical investigation of the repurposing of CPZ, a drug with mild side effects, in the treatment of patients with COVID-19.
Collapse
Affiliation(s)
- Marion Plaze
- GHU Paris Psychiatrie & Neurosciences, Hôpital Sainte-Anne, Service Hospitalo-Universitaire, Pôle Hospitalo-Universitaire Paris 15, Paris, France; Université de Paris, Paris, France
| | - David Attali
- GHU Paris Psychiatrie & Neurosciences, Hôpital Sainte-Anne, Service Hospitalo-Universitaire, Pôle Hospitalo-Universitaire Paris 15, Paris, France; Université de Paris, Paris, France; Physics for Medicine Paris, INSERM, ESPCI Paris, CNRS, PSL Research University, Université Paris Diderot, Sorbonne Paris Cite, Paris, France
| | - Matthieu Prot
- Institut Pasteur, G5 Evolutionary Genomics of RNA Viruses, Paris, France
| | - Anne-Cécile Petit
- GHU Paris Psychiatrie & Neurosciences, Hôpital Sainte-Anne, Service Hospitalo-Universitaire, Pôle Hospitalo-Universitaire Paris 15, Paris, France; Institut Pasteur, Experimental Neuropathology Unit, Paris, France
| | - Michael Blatzer
- Institut Pasteur, Experimental Neuropathology Unit, Paris, France
| | - Fabien Vinckier
- GHU Paris Psychiatrie & Neurosciences, Hôpital Sainte-Anne, Service Hospitalo-Universitaire, Pôle Hospitalo-Universitaire Paris 15, Paris, France; Université de Paris, Paris, France
| | - Laurine Levillayer
- Institut Pasteur, Functional Genetics of Infectious Diseases Unit, Paris, France
| | - Jeanne Chiaravalli
- Institut Pasteur, Chemogenomic and Biological Screening Core Facility, C2RT, Paris, France
| | - Florent Perin-Dureau
- Fondation Rothschild, Department of Anaesthesiology, ASMR-II Consulting, Regstem, Paris, France
| | - Arnaud Cachia
- Université de Paris, Laboratoire de Psychologie du développement et de l'Education de l'Enfant, CNRS, Paris, France; Université de Paris, Institut de Psychiatrie et Neurosciences de Paris, INSERM, Paris, France
| | | | - Fabrice Chrétien
- Université de Paris, Paris, France; Institut Pasteur, Experimental Neuropathology Unit, Paris, France; GHU Paris Psychiatrie & Neurosciences, Hôpital Sainte-Anne, Service de Neuropathologie, Paris, France
| | | | - Raphaël Gaillard
- GHU Paris Psychiatrie & Neurosciences, Hôpital Sainte-Anne, Service Hospitalo-Universitaire, Pôle Hospitalo-Universitaire Paris 15, Paris, France; Université de Paris, Paris, France; Institut Pasteur, Experimental Neuropathology Unit, Paris, France.
| |
Collapse
|
41
|
Ying Z, He T, Wang X, Zheng W, Lin N, Tu M, Xie Y, Ping L, Zhang C, Liu W, Deng L, Wu M, Feng F, Leng X, Du T, Qi F, Hu X, Ding Y, Lu XA, Song Y, Zhu J. Distribution of chimeric antigen receptor-modified T cells against CD19 in B-cell malignancies. BMC Cancer 2021; 21:198. [PMID: 33632155 PMCID: PMC7908740 DOI: 10.1186/s12885-021-07934-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 02/18/2021] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The unprecedented efficacy of chimeric antigen receptor T (CAR-T) cell immunotherapy of CD19+ B-cell malignancies has opened a new and useful way for the treatment of malignant tumors. Nonetheless, there are still formidable challenges in the field of CAR-T cell therapy, such as the biodistribution of CAR-T cells in vivo. METHODS NALM-6, a human B-cell acute lymphoblastic leukemia (B-ALL) cell line, was used as target cells. CAR-T cells were injected into a mice model with or without target cells. Then we measured the distribution of CAR-T cells in mice. In addition, an exploratory clinical trial was conducted in 13 r/r B-cell non-Hodgkin lymphoma (B-NHL) patients, who received CAR-T cell infusion. The dynamic changes in patient blood parameters over time after infusion were detected by qPCR and flow cytometry. RESULTS CAR-T cells still proliferated over time after being infused into the mice without target cells within 2 weeks. However, CAR-T cells did not increase significantly in the presence of target cells within 2 weeks after infusion, but expanded at week 6. In the clinical trial, we found that CAR-T cells peaked at 7-21 days after infusion and lasted for 420 days in peripheral blood of patients. Simultaneously, mild side effects were observed, which could be effectively controlled within 2 months in these patients. CONCLUSIONS CAR-T cells can expand themselves with or without target cells in mice, and persist for a long time in NHL patients without serious side effects. TRIAL REGISTRATION The registration date of the clinical trial is May 17, 2018 and the trial registration numbers is NCT03528421 .
Collapse
Affiliation(s)
- Zhitao Ying
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Ting He
- Beijing Immunochina Pharmaceuticals Co., Ltd., Beijing, China
| | - Xiaopei Wang
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Wen Zheng
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Ningjing Lin
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Meifeng Tu
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yan Xie
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Lingyan Ping
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Chen Zhang
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Weiping Liu
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Lijuan Deng
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Meng Wu
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Feier Feng
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xin Leng
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Tingting Du
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Feifei Qi
- Beijing Immunochina Pharmaceuticals Co., Ltd., Beijing, China
| | - Xuelian Hu
- Beijing Immunochina Pharmaceuticals Co., Ltd., Beijing, China
| | - Yanping Ding
- Beijing Immunochina Pharmaceuticals Co., Ltd., Beijing, China
| | - Xin-An Lu
- Beijing Immunochina Pharmaceuticals Co., Ltd., Beijing, China.
| | - Yuqin Song
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China.
| | - Jun Zhu
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
42
|
Cheng ZF, Li HK, Yang HP, Lee CY, Tang SW, Lin YL, Hsiao SC. A novel endogenous CD16-Expressing Natural Killer Cell for cancer immunotherapy. Biochem Biophys Rep 2021; 26:100935. [PMID: 33644421 PMCID: PMC7889824 DOI: 10.1016/j.bbrep.2021.100935] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/25/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Natural killer (NK) cells, as a potential source for off-the-shelf cell therapy, attack tumor cells with low risk of severe cytokine release syndrome (CRS) or graft-versus-host disease (GvHD). Fcγ receptor IIIA, also known as CD16, further confers NK cells with antibody-dependent cell-mediated cytotoxicity (ADCC), one mechanism of action of antibody-based immunotherapy. Here, we establish a novel human NK cell line, oNK-1, endogenously expressing CD16 along with high levels of NK activation markers and low levels of NK inhibitory markers. The long-term expansion and CD16 expression of oNK-1 cells were demonstrated. Furthermore, oNK-1 cells elicit superior cytotoxicity against cancer cells than primary NK cells. In conclusion, this study suggests that endogenous CD16-expressing oNK-1 has the potential to develop an effective NK-based therapy.
Collapse
Affiliation(s)
| | - Hao-Kang Li
- Acepodia Biotech Inc. San Mateo, California, USA
| | | | - Chia-Yun Lee
- Acepodia Biotech Inc. San Mateo, California, USA
| | - Sai-Wen Tang
- Acepodia Biotech Inc. San Mateo, California, USA
| | | | | |
Collapse
|
43
|
Abstract
Patients with chronic lymphocytic leukemia can be divided into three categories: those who are minimally affected by the problem, often never requiring therapy; those that initially follow an indolent course but subsequently progress and require therapy; and those that from the point of diagnosis exhibit an aggressive disease necessitating treatment. Likewise, such patients pass through three phases: development of the disease, diagnosis, and need for therapy. Finally, the leukemic clones of all patients appear to require continuous input from the exterior, most often through membrane receptors, to allow them to survive and grow. This review is presented according to the temporal course that the disease follows, focusing on those external influences from the tissue microenvironment (TME) that support the time lines as well as those internal influences that are inherited or develop as genetic and epigenetic changes occurring over the time line. Regarding the former, special emphasis is placed on the input provided via the B-cell receptor for antigen and the C-X-C-motif chemokine receptor-4 and the therapeutic agents that block these inputs. Regarding the latter, prominence is laid upon inherited susceptibility genes and the genetic and epigenetic abnormalities that lead to the developmental and progression of the disease.
Collapse
MESH Headings
- Disease Progression
- Humans
- Immunotherapy
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Mutation
- PAX5 Transcription Factor/metabolism
- Receptors, Antigen, B-Cell
- Signal Transduction
- Tumor Microenvironment
Collapse
Affiliation(s)
- Nicholas Chiorazzi
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York 11030, USA
| | - Shih-Shih Chen
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York 11030, USA
| | - Kanti R Rai
- The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York 11549, USA
| |
Collapse
|
44
|
Zhou X, Rasche L, Kortüm KM, Danhof S, Hudecek M, Einsele H. Toxicities of Chimeric Antigen Receptor T Cell Therapy in Multiple Myeloma: An Overview of Experience From Clinical Trials, Pathophysiology, and Management Strategies. Front Immunol 2021; 11:620312. [PMID: 33424871 PMCID: PMC7793717 DOI: 10.3389/fimmu.2020.620312] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 11/24/2020] [Indexed: 12/15/2022] Open
Abstract
In the last few years, monoclonal antibodies (mAbs) such as elotuzumab and daratutumab have brought the treatment of multiple myeloma (MM) into the new era of immunotherapy. More recently, chimeric antigen receptor (CAR) modified T cell, a novel cellular immunotherapy, has been developed for treatment of relapsed/refractory (RR) MM, and early phase clinical trials have shown promising efficacy of CAR T cell therapy. Many patients with end stage RRMM regard CAR T cell therapy as their “last chance” and a “hope of cure”. However, severe adverse events (AEs) and even toxic death related to CAR T cell therapy have been observed. The management of AEs related to CAR T cell therapy represents a new challenge, as the pathophysiology is not fully understood and there is still no well-established standard of management. With regard to CAR T cell associated toxicities in MM, in this review, we will provide an overview of experience from clinical trials, pathophysiology, and management strategies.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Leo Rasche
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - K Martin Kortüm
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Sophia Danhof
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Michael Hudecek
- 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
|
45
|
Maus MV, Alexander S, Bishop MR, Brudno JN, Callahan C, Davila ML, Diamonte C, Dietrich J, Fitzgerald JC, Frigault MJ, Fry TJ, Holter-Chakrabarty JL, Komanduri KV, Lee DW, Locke FL, Maude SL, McCarthy PL, Mead E, Neelapu SS, Neilan TG, Santomasso BD, Shpall EJ, Teachey DT, Turtle CJ, Whitehead T, Grupp SA. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune effector cell-related adverse events. J Immunother Cancer 2020; 8:jitc-2020-001511. [PMID: 33335028 PMCID: PMC7745688 DOI: 10.1136/jitc-2020-001511] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2020] [Indexed: 12/20/2022] Open
Abstract
Immune effector cell (IEC) therapies offer durable and sustained remissions in significant numbers of patients with hematological cancers. While these unique immunotherapies have improved outcomes for pediatric and adult patients in a number of disease states, as 'living drugs,' their toxicity profiles, including cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), differ markedly from conventional cancer therapeutics. At the time of article preparation, the US Food and Drug Administration (FDA) has approved tisagenlecleucel, axicabtagene ciloleucel, and brexucabtagene autoleucel, all of which are IEC therapies based on genetically modified T cells engineered to express chimeric antigen receptors (CARs), and additional products are expected to reach marketing authorization soon and to enter clinical development in due course. As IEC therapies, especially CAR T cell therapies, enter more widespread clinical use, there is a need for clear, cohesive recommendations on toxicity management, motivating the Society for Immunotherapy of Cancer (SITC) to convene an expert panel to develop a clinical practice guideline. The panel discussed the recognition and management of common toxicities in the context of IEC treatment, including baseline laboratory parameters for monitoring, timing to onset, and pharmacological interventions, ultimately forming evidence- and consensus-based recommendations to assist medical professionals in decision-making and to improve outcomes for patients.
Collapse
Affiliation(s)
- Marcela V Maus
- Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - Sara Alexander
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael R Bishop
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | | | - Colleen Callahan
- Cancer Immunotherapy Program, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Marco L Davila
- Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA
| | - Claudia Diamonte
- Cellular Therapeutics Center, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jorg Dietrich
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Julie C Fitzgerald
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Matthew J Frigault
- Bone Marrow Transplant and Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Terry J Fry
- Pediatric Hematology/Oncology/BMT, Children's Hospital Colorado and University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
| | - Jennifer L Holter-Chakrabarty
- Department of Hematology/Oncology/Bone Marrow Transplant and Cellular Therapy, The University of Oklahoma Stephenson Cancer Center, Oklahoma City, Oklahoma, USA
| | - Krishna V Komanduri
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, USA
| | - Daniel W Lee
- Department of Pediatrics, University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA
| | - Shannon L Maude
- Cancer Immunotherapy Program, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Philip L McCarthy
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Elena Mead
- Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sattva S Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tomas G Neilan
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Bianca D Santomasso
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David T Teachey
- Cancer Center, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cameron J Turtle
- Clinical Research Division, Fred Hutchinson Cancer Research Center Division of Medical Oncology, University of Washington, Seattle, Washington, USA
| | - Tom Whitehead
- Emily Whitehead Foundation, Phillipsburg, Pennsylvania, USA
| | - Stephan A Grupp
- Cancer Immunotherapy Program, Division of Oncology, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
46
|
Acharya UH, Walter RB. Chimeric Antigen Receptor (CAR)-Modified Immune Effector Cell Therapy for Acute Myeloid Leukemia (AML). Cancers (Basel) 2020; 12:E3617. [PMID: 33287224 PMCID: PMC7761730 DOI: 10.3390/cancers12123617] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
Despite the availability of an increasing number of targeted therapeutics and wider use of allogeneic hematopoietic stem cell transplantation, many patients with acute myeloid leukemia (AML) ultimately succumb to this disease. Given their remarkable efficacy in B-acute lymphoblastic leukemia and other CD19-expressing B cell malignancies, there is hope adoptive cellular transfer, particularly chimeric antigen receptor (CAR)-modified immune effector cell (IEC) therapies, may afford a novel, potent immune-based approach for the treatment of AML that complements or replaces existing ones and improves cure rates. However, it is unclear how best to translate the success of these therapies from B cell malignancies, where use of highly potent immunotherapies is facilitated by identified target antigens with near ubiquitous expression on malignant cells and non-fatal consequences from "on-target, off-tumor cell" toxicities. Herein, we review the current status of CAR-modified IEC therapies for AML, with considerations regarding suitable, relatively leukemia-restricted target antigens, expected toxicities, and interactions of the engineered cells with a profoundly immunosuppressive tumor microenvironment that restricts their therapeutic efficacy. With these challenges in mind, we will discuss possible strategies to improve the cells' potency as well as their therapeutic window for optimal clinical use in AML.
Collapse
Affiliation(s)
- Utkarsh H. Acharya
- Divisions of Hematologic Malignancies & Immune Effector Cell Therapy, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Roland B. Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA;
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA 98195, USA
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA 98195, USA
- Department of Epidemiology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
47
|
Wu W, Huo Y, Ding X, Zhou Y, Gu S, Gao Y. Identification of the risks in CAR T-cell therapy clinical trials in China: a Delphi study. Ther Adv Med Oncol 2020; 12:1758835920966574. [PMID: 33149770 PMCID: PMC7580145 DOI: 10.1177/1758835920966574] [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: 01/31/2020] [Accepted: 07/13/2020] [Indexed: 11/16/2022] Open
Abstract
Aims: Within the past few years, there has been tremendous growth in clinical trials of chimeric antigen receptor (CAR) T-cell therapies. Unlike those of many small-molecule pharmaceuticals, CAR T-cell therapy clinical trials are fraught with risks due to the use of live cell products. The aim of this study is to reach a consensus with experts on the most relevant set of risks that practically occur in CAR T-cell therapy clinical trials. Methods: A Delphi method of consensus development was used to identify the risks in CAR T-cell therapy clinical trials, comprising three survey rounds. The expert panel consisted of principal investigators, clinical research physicians, members of institutional ethics committees, and Good Clinical Practice managers. Results: Of the 24 experts invited to participate in this Delphi study, 20 participants completed Round 1, Round 2, and Round 3. Finally, consensus (defined as >80% agreement) was achieved for 54 risks relating to CAR T-cell clinical trials. Effective interventions related to these risks are needed to ensure the proper protection of subject health and safety. Conclusion: The Delphi method was successful in gaining a consensus on risks relevant to CAR T-cell clinical trials in a geographically diverse expert association. It is hoped that this work can benefit future risk-based quality management in clinical trials and can potentially promote the better development of CAR T-cell therapy products.
Collapse
Affiliation(s)
- Weijia Wu
- Department of Clinical Pharmacy and Pharmaceutical Management, School of Pharmacy, Fudan University, Shanghai, China
| | - Yan Huo
- National Institution of Food and Drug Control, National Medical Products Administration, Beijing, China
| | - Xueying Ding
- Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee
| | - Yuhong Zhou
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shengying Gu
- Clinical Research Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Gao
- Department of Clinical Pharmacy and Pharmaceutical Management, School of Pharmacy, Fudan University, Pudong District, Shanghai, 200433, China
| |
Collapse
|
48
|
Photoswitchable CAR-T Cell Function In Vitro and In Vivo via a Cleavable Mediator. Cell Chem Biol 2020; 28:60-69.e7. [PMID: 33113407 DOI: 10.1016/j.chembiol.2020.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/07/2020] [Accepted: 10/07/2020] [Indexed: 01/20/2023]
Abstract
Chimeric antigen receptor (CAR)-T-based therapeutics are a breakthrough in cancer treatment; however, they are hampered by constitutive activation, which leads to worrisome side effects. Engineering CAR-T cells to be as tightly controllable as possible remains a topic of ongoing investigation. Here, we report a photoswitchable approach that uses a mediator for the at-will regulation of CAR-T cells. This mediator carries dual folate and fluorescein isothiocyanate moieties tethered by an ortho-nitrobenzyl ester photocleavable linker. CAR-T cells were shown to be highly cytotoxic to targeted cells only in the presence of the mediator and acted in a dose-dependent manner. The toxicity of CAR-T cells can be rapidly terminated by cleavage of the mediator, and the effects of CAR-T cells can be activated again by resupplementation with the mediator without compromising tumor therapy. The approach described here provides a direction for enhancing the controllability of CAR-T cells and can likely be applied in other immunotherapies.
Collapse
|
49
|
Schubert ML, Schmitt M, Wang L, Ramos CA, Jordan K, Müller-Tidow C, Dreger P. Side-effect management of chimeric antigen receptor (CAR) T-cell therapy. Ann Oncol 2020; 32:34-48. [PMID: 33098993 DOI: 10.1016/j.annonc.2020.10.478] [Citation(s) in RCA: 294] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/15/2020] [Accepted: 10/15/2020] [Indexed: 12/18/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells directed against the B-cell marker CD19 are currently changing the landscape for treatment of patients with refractory and/or relapsed B-cell malignancies. Due to the nature of CAR T cells as living drugs, they display a unique toxicity profile. As CAR T-cell therapy is extending towards other diseases and being more broadly employed in hematology and oncology, optimal management strategies of side-effects associated with CAR T-cell therapy are of high relevance. Cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and cytopenias constitute challenges in the treatment of patients with CAR T cells. This review summarizes the current understanding of CAR T-cell toxicity and its management.
Collapse
Affiliation(s)
- M-L Schubert
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany.
| | - M Schmitt
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany; National Centre for Tumor Diseases (NCT), Heidelberg, Germany
| | - L Wang
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - C A Ramos
- Center for Cell Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, Texas, USA
| | - K Jordan
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - C Müller-Tidow
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany; National Centre for Tumor Diseases (NCT), Heidelberg, Germany
| | - P Dreger
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany; National Centre for Tumor Diseases (NCT), Heidelberg, Germany
| |
Collapse
|
50
|
Boyiadzis MM, Aksentijevich I, Arber DA, Barrett J, Brentjens RJ, Brufsky J, Cortes J, De Lima M, Forman SJ, Fuchs EJ, Fukas LJ, Gore SD, Litzow MR, Miller JS, Pagel JM, Waller EK, Tallman MS. The Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immunotherapy for the treatment of acute leukemia. J Immunother Cancer 2020; 8:jitc-2020-000810. [PMID: 33077513 PMCID: PMC7574947 DOI: 10.1136/jitc-2020-000810] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2020] [Indexed: 12/29/2022] Open
Abstract
Acute leukemia is a constellation of rapidly progressing diseases that affect a wide range of patients regardless of age or gender. Traditional treatment options for patients with acute leukemia include chemotherapy and hematopoietic cell transplantation. The advent of cancer immunotherapy has had a significant impact on acute leukemia treatment. Novel immunotherapeutic agents including antibody-drug conjugates, bispecific T cell engagers, and chimeric antigen receptor T cell therapies have efficacy and have recently been approved by the US Food and Drug Administration (FDA) for the treatment of patients with acute leukemia. The Society for Immunotherapy of Cancer (SITC) convened a panel of experts to develop a clinical practice guideline composed of consensus recommendations on immunotherapy for the treatment of acute lymphoblastic leukemia and acute myeloid leukemia.
Collapse
Affiliation(s)
- Michael M Boyiadzis
- Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Daniel A Arber
- Department of Pathology, University of Chicago, Chicago, Illinois, USA
| | - John Barrett
- Stem Cell Allotransplantation Section, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Renier J Brentjens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jill Brufsky
- Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jorge Cortes
- Department of Medicine, Division of Hematology/Oncology, Georgia Cancer Center, Augusta, Georgia, USA
| | - Marcos De Lima
- Division of Hematology, University Hospitals of Cleveland and Case Western Reserve University, Cleveland, Ohio, USA
| | - Stephen J Forman
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Ephraim J Fuchs
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Linda J Fukas
- Clinical Research Services, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Steven D Gore
- Investigational Drug Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Mark R Litzow
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jeffrey S Miller
- Division of Hematology, Oncology and Transplantation, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - John M Pagel
- Center for Blood Disorders and Stem Cell Transplantation, Swedish Cancer Institute, Seattle, Washington, USA
| | - Edmund K Waller
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Martin S Tallman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|