1
|
Nie J, Zhou L, Tian W, Liu X, Yang L, Yang X, Zhang Y, Wei S, Wang DW, Wei J. Deep insight into cytokine storm: from pathogenesis to treatment. Signal Transduct Target Ther 2025; 10:112. [PMID: 40234407 PMCID: PMC12000524 DOI: 10.1038/s41392-025-02178-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/22/2024] [Accepted: 02/12/2025] [Indexed: 04/17/2025] Open
Abstract
Cytokine storm (CS) is a severe systemic inflammatory syndrome characterized by the excessive activation of immune cells and a significant increase in circulating levels of cytokines. This pathological process is implicated in the development of life-threatening conditions such as fulminant myocarditis (FM), acute respiratory distress syndrome (ARDS), primary or secondary hemophagocytic lymphohistiocytosis (HLH), cytokine release syndrome (CRS) associated with chimeric antigen receptor-modified T (CAR-T) therapy, and grade III to IV acute graft-versus-host disease following allogeneic hematopoietic stem cell transplantation. The significant involvement of the JAK-STAT pathway, Toll-like receptors, neutrophil extracellular traps, NLRP3 inflammasome, and other signaling pathways has been recognized in the pathogenesis of CS. Therapies targeting these pathways have been developed or are currently being investigated. While novel drugs have demonstrated promising therapeutic efficacy in mitigating CS, the overall mortality rate of CS resulting from underlying diseases remains high. In the clinical setting, the management of CS typically necessitates a multidisciplinary team strategy encompassing the removal of abnormal inflammatory or immune system activation, the preservation of vital organ function, the treatment of the underlying disease, and the provision of life supportive therapy. This review provides a comprehensive overview of the key signaling pathways and associated cytokines implicated in CS, elucidates the impact of dysregulated immune cell activation, and delineates the resultant organ injury associated with CS. In addition, we offer insights and current literature on the management of CS in cases of FM, ARDS, systemic inflammatory response syndrome, treatment-induced CRS, HLH, and other related conditions.
Collapse
Grants
- 82070217, 81873427 National Natural Science Foundation of China (National Science Foundation of China)
- 82100401 National Natural Science Foundation of China (National Science Foundation of China)
- 81772477, 81201848, 82473220 National Natural Science Foundation of China (National Science Foundation of China)
- 82330010,81630010,81790624 National Natural Science Foundation of China (National Science Foundation of China)
- National High Technology Research and Development Program of China, Grant number: 2021YFA1101500.
- The Hubei Provincial Natural Science Foundation (No.2024AFB050)
- Project of Shanxi Bethune Hospital, Grant Numbber: 2023xg02); Fundamental Research Program of Shanxi Province, Grant Numbber: 202303021211224
- The Key Scientific Research Project of COVID-19 Infection Emergency Treatment of Shanxi Bethune Hospital (2023xg01), 2023 COVID-19 Research Project of Shanxi Provincial Health Commission (No.2023XG001, No. 2023XG005), Four “Batches” Innovation Project of Invigorating Medical through Science and Technology of Shanxi Province (2023XM003), Cancer special Fund research project of Shanxi Bethune Hospital (No. 2020-ZL04), and External Expert Workshop Fund Program of Shanxi Provincial Health Commission(Proteomics Shanxi studio for Huanghe professor)
- Fundamental Research Program of Shanxi Province(No.202303021221192); 2023 COVID-19 Emergency Project of Shanxi Health Commission (Nos.2023XG001,2023XG005)
Collapse
Affiliation(s)
- Jiali Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Ling Zhou
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China
| | - Weiwei Tian
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xiansheng Liu
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Liping Yang
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xingcheng Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China.
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China.
| | - Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
2
|
Ruemmele T, Macedo R, Stein MN, Chan HT, Mapara MY, Jacquemont CF, Reshef R. Emapalumab for severe cytokine release syndrome in solid tumor CAR-T: a case report. Front Oncol 2025; 15:1543622. [PMID: 40236652 PMCID: PMC11997384 DOI: 10.3389/fonc.2025.1543622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/06/2025] [Indexed: 04/17/2025] Open
Abstract
Chimeric Antigen Receptor T (CAR-T) cell therapy significantly and rapidly changed the treatment paradigm for lymphoma, myeloma and leukemia, and the recent approvals of the first cellular immunotherapies in melanoma and synovial sarcoma demonstrate the potential success of this approach in solid tumors. Though the therapeutic potential of CAR-T is impressive, severe cytokine release syndrome (CRS) remains an ongoing challenge. Here we report a patient who received an investigational CAR-T product for metastatic castration-resistant prostate cancer who developed multi-drug refractory, life-threatening CRS, which was successfully treated with the interferon (IFN)-γ antagonist emapalumab. Within 12 hours after the first dose of emapalumab, there was a dramatic improvement in hemodynamic status and the patient was weaned off all four vasopressors. The hemodynamic improvement was associated with a decrease in IFN-γ and CXCL10 levels but no other cytokines. Not only was emapalumab the only drug effective at treating this case of refractory CRS, but it did not appear to reduce the activity of the CAR-T product, as the CAR-T vector copy numbers remained persistent and the patient's PSA levels remained low. This case demonstrates the clinical use of emapalumab to treat refractory cytokine release syndrome in a solid tumor CAR-T while potentially preserving therapeutic efficacy of CAR-T therapy. Further studies with larger patient populations are needed to evaluate the use of emapalumab as a treatment for CRS.
Collapse
Affiliation(s)
- Tyler Ruemmele
- Division of Hematology/Oncology, New-York Presbyterian Hospital/Columbia University Irving Medical Center, New York, NY, United States
| | - Rodney Macedo
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, United States
| | - Mark N. Stein
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, United States
| | - Hei Ton Chan
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, United States
| | - Markus Y. Mapara
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, United States
| | | | - Ran Reshef
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
3
|
Zhang S, Chen W, Zhou J, Liang Q, Zhang Y, Su M, Zhang Z, Qu J. The Benefits and Safety of Monoclonal Antibodies: Implications for Cancer Immunotherapy. J Inflamm Res 2025; 18:4335-4357. [PMID: 40162076 PMCID: PMC11952073 DOI: 10.2147/jir.s499403] [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: 10/05/2024] [Accepted: 03/06/2025] [Indexed: 04/02/2025] Open
Abstract
Monoclonal antibodies (mAbs) have transformed cancer treatment by providing highly targeted and effective therapies that specifically attack cancer cells, thus reducing the likelihood of adverse events (AEs) in patients. mAbs exert their action through various mechanisms, such as receptor blockade, antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), and inhibition of immune checkpoints (eg, PD-1, PD-L1, and CTLA-4). These therapies have led to significant improvements in the treatment of several cancers, including HER2-positive breast cancer, non-small cell lung cancer (NSCLC), and melanoma. The efficacy of mAb therapy in cancer treatment is influenced by various intrinsic and extrinsic factors, such as environmental exposures, psychosocial factors, infection status, ways of life, and tumor microenvironment (TME), all of which can impact immune responses and treatment outcomes. Notably, the therapeutic benefits of mAbs are often accompanied by immune-related AEs (irAEs), which can vary from mild to severe and affect multiple organ systems. The dual nature of mAbs-stimulating antitumor immune responses while also inducing immune-related side effects-presents a notable challenge in clinical practice. This review highlights the importance of proactive strategies for managing irAEs, such as early detection, corticosteroid use, targeted immunosuppressive treatments, and the urgent need for reliable predictive biomarkers to improve treatment outcomes. Advancements in the prevention, prediction, and management of irAEs are essential to enhance the safety and effectiveness of mAb-based therapies, ultimately aiming to improve cancer patient outcomes.
Collapse
Affiliation(s)
- Shuguang Zhang
- Department of Pharmacy, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Wenying Chen
- Department of Pharmacy, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People’s Republic of China
| | - Jihong Zhou
- Department of Respiratory and Critical Care Medicine, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Qi Liang
- Department of Pharmacy, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Yu Zhang
- Department of Intensive Care Unit, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Ming Su
- Department of Respiratory and Critical Care Medicine, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Zilong Zhang
- Department of Pharmacy, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Jian Qu
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Institute of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, People’s Republic of China
| |
Collapse
|
4
|
Zhou Q, An Y, Zhang X, Xiao X, Bai X, Liu P, Pu Y, Meng J, Zhu H, Lyu C, Zhang H, Zhang Y, Xie T, Meng H, Lyu H. Efficacy and safety of tocilizumab in managing cytokine release syndrome after CD19 CAR-T therapy for relapsed or refractory B-cell acute lymphoblastic leukemia. Front Immunol 2025; 16:1530623. [PMID: 40160812 PMCID: PMC11949925 DOI: 10.3389/fimmu.2025.1530623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Purpose CD19 chimeric antigen receptor T (CAR-T) cell therapy has shown promise in treating relapsed or refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL), but cytokine release syndrome (CRS) remains a significant side effect. Methods This retrospective cohort study investigated the use of tocilizumab for managing CAR-T-related CRS in 45 R/R B-ALL patients. Results Of these, 17 patients received tocilizumab, resulting in a significant reduction in the duration of grade 3 CRS compared to those who did not receive the drug. Additionally, 10 patients showed decreased cytokine levels.Importantly, tocilizumab did not impair CAR-T cell expansion or efficacy, nor did it increase the incidence of adverse events. Conclusion These findings suggest that tocilizumab may be an effective and safe strategy for mitigating CAR-T-related CRS in R/R B-ALL patients, potentially improving patient outcomes and survival.
Collapse
Affiliation(s)
- Qianyi Zhou
- First Center Clinical College, Tianjin Medical University, Tianjin, China
| | - Yuxin An
- First Center Clinical College, Tianjin Medical University, Tianjin, China
| | - Xiaomei Zhang
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Xia Xiao
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Xue Bai
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Pengjiang Liu
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yedi Pu
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Juanxia Meng
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Haibo Zhu
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Cuicui Lyu
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Huan Zhang
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yu Zhang
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Tianle Xie
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Haotian Meng
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Hairong Lyu
- Nankai University School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
5
|
Wang JM, Jiang HW, Zhang YQ, Hu Y, Mei H. Hemophagocytic lymphohistiocytosis post chimeric antigen receptor T cell therapies. Expert Rev Clin Immunol 2025; 21:277-289. [PMID: 39727348 DOI: 10.1080/1744666x.2024.2444673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
INTRODUCTION Besides cytokine release syndromes (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), immune effector cell-associated HLH-like syndrome (IEC-HS) is increasingly recognized across CAR-T recipients. This emergent and fatal syndrome is difficult to separate from other disorders during the early phase, and urgently requires more integrated diagnostic and therapeutic frameworks. AREAS COVERED Existing literature has pointed out the potential role of unbridled proliferation of cytotoxic T lymphocytes, lymphopenia of natural killing cells, and hypercytokinemia in triggering the IEC-HS. The onset time of IEC-HS usually overlaps with CRS or be delayed from CRS. Clinical features include hyperferritinemia, hepatic and renal dysfunctions, cytopenias, coagulopathy, and hemophagocytosis. Multiple diagnostic criteria are based predominantly on ferritin elevation and prerequisite CRS. Corticosteroids are the cornerstone for IEC-HS treatment, while cytokine-targeted agents and pathway inhibitors offer great promise in alleviating IEC-HS syndromes. EXPERT OPINIONS Several controversial predisposing factors of IEC-HS such as disease burden should be further investigated. Future research is anticipated to identify the real-time biomarkers, as well as develop a more sophisticated grading and management network.
Collapse
Affiliation(s)
- Jing-Ming Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Hui-Wen Jiang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Yin-Qiang Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| |
Collapse
|
6
|
Evangelidis P, Tragiannidis K, Vyzantiadis A, Evangelidis N, Kalmoukos P, Vyzantiadis TA, Tragiannidis A, Kourti M, Gavriilaki E. Invasive Fungal Disease After Chimeric Antigen Receptor-T Immunotherapy in Adult and Pediatric Patients. Pathogens 2025; 14:170. [PMID: 40005545 PMCID: PMC11858289 DOI: 10.3390/pathogens14020170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 01/25/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Invasive fungal diseases (IFDs) have been documented among the causes of post-chimeric antigen receptor-T (CAR-T) cell immunotherapy complications, with the incidence of IFDs in CAR-T cell therapy recipients being measured between 0% and 10%, globally. IFDs are notorious for their potentially life-threatening nature and challenging diagnosis and treatment. In this review, we searched the recent literature aiming to examine the risk factors and epidemiology of IFDs post-CAR-T infusion. Moreover, the role of antifungal prophylaxis is investigated. CAR-T cell therapy recipients are especially vulnerable to IFDs due to several risk factors that contribute to the patient's immunosuppression. Those include the underlying hematological malignancies, the lymphodepleting chemotherapy administered before the treatment, existing leukopenia and hypogammaglobinemia, and the use of high-dose corticosteroids and interleukin-6 blockers as countermeasures for immune effector cell-associated neurotoxicity syndrome and cytokine release syndrome, respectively. IFDs mostly occur within the first 60 days following the infusion of the T cells, but cases even a year after the infusion have been described. Aspergillus spp., Candida spp., and Pneumocystis jirovecii are the main cause of these infections following CAR-T cell therapy. More real-world data regarding the epidemiology of IFDs and the role of antifungal prophylaxis in this population are essential.
Collapse
Affiliation(s)
- Paschalis Evangelidis
- 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (P.E.); (N.E.); (P.K.)
| | - Konstantinos Tragiannidis
- Children & Adolescent Hematology-Oncology Unit, Second Department of Pediatrics, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.T.); (A.T.); (M.K.)
| | - Athanasios Vyzantiadis
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.V.); (T.-A.V.)
| | - Nikolaos Evangelidis
- 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (P.E.); (N.E.); (P.K.)
| | - Panagiotis Kalmoukos
- 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (P.E.); (N.E.); (P.K.)
| | - Timoleon-Achilleas Vyzantiadis
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.V.); (T.-A.V.)
| | - Athanasios Tragiannidis
- Children & Adolescent Hematology-Oncology Unit, Second Department of Pediatrics, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.T.); (A.T.); (M.K.)
| | - Maria Kourti
- Children & Adolescent Hematology-Oncology Unit, Second Department of Pediatrics, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.T.); (A.T.); (M.K.)
| | - Eleni Gavriilaki
- 2nd Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (P.E.); (N.E.); (P.K.)
- Hematology Department and Bone Marrow Transplant (BMT) Unit, G. Papanicolaou Hospital, 57010 Thessaloniki, Greece
| |
Collapse
|
7
|
Cai W, Lu Y, He H, Li J, Liu S, Geng H, Yang Q, Zeng L, Wu D, Li C. Efficacy of emapalumab in the management of anti‑CD19 chimeric antigen receptor T‑cell therapy‑associated cytokine release syndrome: A report of two cases. Oncol Lett 2025; 29:71. [PMID: 39628826 PMCID: PMC11612719 DOI: 10.3892/ol.2024.14817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/01/2024] [Indexed: 12/06/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is an effective treatment for diffuse large B-cell lymphoma (DLBCL). However, it may activate the systemic immune system of the patient, resulting in cytokine release syndrome (CRS). Emapalumab is a human monoclonal antibody targeting interferon-γ, inhibiting its interaction with cell surface receptors and the subsequent activation of inflammatory pathways. The present report describes the cases of 2 patients with relapsed DLBCL treated with CAR T-cell therapy, in which the severe CRS associated with CAR T-cell therapy was attenuated without compromising antitumor efficacy after receiving emapalumab. Further prospective clinical trials are warranted to determine the role of emapalumab in CAR T-cell therapy.
Collapse
Affiliation(s)
- Wenzhi Cai
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Institute of Blood and Marrow Transplantation, Suzhou University Medical College, Suzhou, Jiangsu 215006, P.R. China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yutong Lu
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Institute of Blood and Marrow Transplantation, Suzhou University Medical College, Suzhou, Jiangsu 215006, P.R. China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Haiju He
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Institute of Blood and Marrow Transplantation, Suzhou University Medical College, Suzhou, Jiangsu 215006, P.R. China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jiaqi Li
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Institute of Blood and Marrow Transplantation, Suzhou University Medical College, Suzhou, Jiangsu 215006, P.R. China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Shuangzhu Liu
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Institute of Blood and Marrow Transplantation, Suzhou University Medical College, Suzhou, Jiangsu 215006, P.R. China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Hongzhi Geng
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Qin Yang
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Liangyu Zeng
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Depei Wu
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Institute of Blood and Marrow Transplantation, Suzhou University Medical College, Suzhou, Jiangsu 215006, P.R. China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Caixia Li
- National Clinical Research Center for Hematological Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
8
|
Athanassiou P, Athanassiou L, Kostoglou-Athanassiou I, Shoenfeld Y. Targeted Cellular Treatment of Systemic Lupus Erythematosus. Cells 2025; 14:210. [PMID: 39937001 PMCID: PMC11816398 DOI: 10.3390/cells14030210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/18/2025] [Accepted: 01/30/2025] [Indexed: 02/13/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease affecting all organ systems. The disease preferentially affects females of childbearing age. It runs a variable course. It may run a mild course that may never lead to severe disease and manifestations from critical organ systems. However, it may also run an undulating course with periods of mild and severe disease. It may run as a mild disease, quickly deteriorating to severe disease and affecting multiple organ systems. Various immune pathways related both to the innate and adaptive immune response are involved in the pathogenesis of SLE. Various drugs have been developed targeting cellular and molecular targets in these pathways. Interferons are involved in the pathogenesis of SLE, and various drugs have been developed to target this pathway. T and B lymphocytes are involved in the pathophysiology of SLE. Various treatment modalities targeting cellular targets are available for the treatment of SLE. These include biologic agents targeting B lymphocytes. However, some patients have disease refractory to these treatment modalities. For these patients, cell-based therapies may be used. Hematopoietic stem cell transplantation involving autologous cells is an option in the treatment of refractory SLE. Mesenchymal stem cells are also applied in the treatment of SLE. Chimeric antigen receptor (CAR)-T cell therapy is a novel treatment also used in SLE management. This novel treatment method holds major promise for the management of autoimmune diseases and, in particular, SLE. Major hurdles to be overcome are the logistics involved, as well as the need for specialized facilities. This review focuses on novel treatment modalities in SLE targeting cellular and molecular targets in the immune system.
Collapse
Affiliation(s)
| | - Lambros Athanassiou
- Department of Rheumatology, Asclepeion Hospital, Voula, 16673 Athens, Greece;
| | | | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Reichman University, Herzliya 4610101, Israel;
| |
Collapse
|
9
|
Huntington SF, Lin D, Lafeuille M, Thompson‐Leduc P, Shah A, Kim N, Hester L, Tardif‐Samson A, Moore B, Fowler J, Marshall A, Zhang X, Gifkins D, Wu B. Identification of cytokine release syndrome and indicators of severity in retrospective databases among patients receiving immunotherapy. Pharmacol Res Perspect 2024; 12:e70024. [PMID: 39487576 PMCID: PMC11530407 DOI: 10.1002/prp2.70024] [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: 06/07/2024] [Revised: 09/05/2024] [Accepted: 09/24/2024] [Indexed: 11/04/2024] Open
Abstract
Cytokine release syndrome (CRS) can occur following cancer immunotherapies, but is most often mild and of limited duration. International Classification of Diseases (ICD)-10 codes allowing identification of CRS were introduced in 2020 but may be underutilized. We evaluated the performance of a published claims-based algorithm to detect CRS (any grade) and high-grade CRS (HG, grades 2-5), as well as identified indicators of HG CRS in retrospective data. Adults with low-grade and HG CRS during an encounter coinciding with administrations of blinatumomab or chimeric antigen receptor-T therapy were identified in three types of retrospective databases (hospital chargemaster data, electronic health records, and administrative claims). The algorithm's sensitivity in detecting any CRS and HG CRS was reported. A least absolute shrinkage and selection operator (LASSO) regression model was developed to identify indicators of HG CRS. Performance of the model was evaluated using area under the curve (AUC). The sensitivity of the algorithm to detect any grade CRS ranged between 77%-100% and between 8%-80% for HG CRS, depending on the type of database. The LASSO model identified hypotension, positive pressure (including mechanical ventilation), tocilizumab, and vasopressors as indicators of HG CRS. AUC varied between 60% and 75%. The algorithm accurately detected any grade CRS for over three-quarters of instances, but was not as reliable for HG CRS. Results varied based on database attributes. Hypotension, vasopressors, positive pressure, and tocilizumab were associated with HG CRS and may be methodologically helpful signals of CRS severity in retrospective data.
Collapse
Affiliation(s)
| | - Dee Lin
- Janssen Scientific Affairs, LLC, a Johnson and Johnson CompanyHorshamPennsylvaniaUSA
| | | | | | - Aditi Shah
- Analysis Group, Inc.MontrealQuebecCanada
| | - Nina Kim
- Janssen Scientific Affairs, LLC, a Johnson and Johnson CompanyHorshamPennsylvaniaUSA
| | - Laura Hester
- Janssen Scientific Affairs, LLC, a Johnson and Johnson CompanyHorshamPennsylvaniaUSA
| | | | | | - Jessica Fowler
- Janssen Scientific Affairs, LLC, a Johnson and Johnson CompanyHorshamPennsylvaniaUSA
| | - Alexander Marshall
- Janssen Global Services, LLC, a Johnson and Johnson CompanyRaritanPennsylvaniaUSA
| | - Xinke Zhang
- Janssen Scientific Affairs, LLC, a Johnson and Johnson CompanyHorshamPennsylvaniaUSA
| | - Dina Gifkins
- Janssen Research and Development, LLC, a Johnson and Johnson CompanyRaritanPennsylvaniaUSA
| | - Bingcao Wu
- Janssen Scientific Affairs, LLC, a Johnson and Johnson CompanyHorshamPennsylvaniaUSA
| |
Collapse
|
10
|
Huang L, Zhu L, Zhao Z, Jiang S. Hyperactive browning and hypermetabolism: potentially dangerous element in critical illness. Front Endocrinol (Lausanne) 2024; 15:1484524. [PMID: 39640882 PMCID: PMC11617193 DOI: 10.3389/fendo.2024.1484524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Brown/beige adipose tissue has attracted much attention in previous studies because it can improve metabolism and combat obesity through non-shivering thermogenesis. However, recent studies have also indicated that especially in critical illness, overactivated brown adipose tissue or extensive browning of white adipose tissue may bring damage to individuals mainly by exacerbating hypermetabolism. In this review, the phenomenon of fat browning in critical illness will be discussed, along with the potential harm, possible regulatory mechanism and corresponding clinical treatment options of the induction of fat browning. The current research on fat browning in critical illness will offer more comprehensive understanding of its biological characteristics, and inspire researchers to develop new complementary treatments for the hypermetabolic state that occurs in critically ill patients.
Collapse
Affiliation(s)
- Lu Huang
- Department of Basic Medical Sciences, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Lili Zhu
- Department of Plastic and Reconstructive Surgery, Taizhou Enze Hospital, Taizhou, China
| | - Zhenxiong Zhao
- Department of Basic Medical Sciences, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Shenglu Jiang
- Department of Basic Medical Sciences, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| |
Collapse
|
11
|
Almskog LM, Sjöström A, Sundén-Cullberg J, Taxiarchis A, Ågren A, Freyland S, Börjesson M, Wikman A, Wahlgren CM, Wanecek M, van der Linden J, Antovic J, Lampa J, Magnusson M. Tocilizumab reduces hypercoagulation in COVID-19 - Perspectives from the coagulation and immunomodulation Covid assessment (Coag-ImmCovA) clinical trial. Thromb Res 2024; 243:109135. [PMID: 39226747 DOI: 10.1016/j.thromres.2024.109135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND Despite medical interventions, COVID-19 continues to persist at pandemic proportions. A hypercoagulation state was rapidly observed in the severely ill, and the incidence of thromboembolic events remains elevated. Interleukin inhibitors have demonstrated positive effects on the hyperactivation of the immune system in COVID-19, with the interleukin-6 inhibitor tocilizumab showing promising results in reducing mortality. Nevertheless, the impact of interleukin inhibitors on the coagulation system remains incompletely understood. METHODS In this clinical trial conducted in Stockholm, Sweden, interleukin inhibitors, namely anakinra (ANA) or tocilizumab (TOCI), were randomly administered in addition to standard care (SC) to hospitalized patients with COVID-19. A control group received only SC. The primary outcome sought to measure effects on global hemostasis, as indicated by changes in functional coagulation tests, specifically Rotational Thromboelastometry (ROTEM) or Overall Hemostatic Potential (OHP), visualized through scanning electron microscopy images. Secondary outcomes included effects on conventional coagulation laboratory tests. RESULTS The study enrolled 74 patients who were randomized to receive either ANA or TOCI in addition to SC, or SC alone. In the TOCI group, ROTEM variables exhibited less hypercoagulation after 29 days compared with ANA or SC treatment groups, characterized by prolonged clot formation time and decreased clot firmness. OHP decreased, but there were no significant differences among the three treatment groups. Plasma fibrinogen levels, initially elevated, decreased significantly in TOCI recipients over time. CONCLUSION Tocilizumab treatment demonstrated a significant reduction of hypercoagulation in hospitalized COVID-19 patients, by improvements in both global coagulation tests and conventional laboratory tests, in comparison with anakinra or SC alone. This finding underscores the significance of tocilizumab as a viable treatment option in severe COVID-19 cases, with the potential to decrease thrombosis incidence.
Collapse
Affiliation(s)
- Lou M Almskog
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden.
| | - Anna Sjöström
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Sundén-Cullberg
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Apostolos Taxiarchis
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Ågren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Sciences, Danderyd Hospital, Division of Cardiovascular Medicine, Karolinska Institutet, Stockholm, Sweden; Coagulation Unit, Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Sara Freyland
- Division of Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Madeleine Börjesson
- Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Agneta Wikman
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Center for Hematology and Regenerative Medicine (HERM), Karolinska Institutet, Stockholm, Sweden
| | - Carl Magnus Wahlgren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Vascular Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Michael Wanecek
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Intensive Care Unit, Capio St Göran's Hospital, Stockholm, Sweden
| | - Jan van der Linden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jovan Antovic
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry, Karolinska University Hospital, Stockholm, Sweden
| | - Jon Lampa
- Rheumatology Division, Department of Medicine Solna, Center for Molecular Medicine (CMM), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Magnusson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Coagulation Unit, Department of Hematology, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
12
|
Injinari N, Asadollahi S, Sefid F, Arshadi M, Hosseini SS, Ghoshouni H, Soltani F, Namiranian N, Sheikhha MH, Aghaeimeybodi F. Impact of FCGR2A rs1801274 and IL-6R rs2228145 polymorphisms on tocilizumab response in the Iranian population with severe COVID-19. BMC Infect Dis 2024; 24:1168. [PMID: 39415081 PMCID: PMC11481263 DOI: 10.1186/s12879-024-10073-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 10/09/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Although several genetic biomarkers have been reported in the tocilizumab (TCZ) response in rheumatoid arthritis, no studies have addressed the pharmacogenomics effect of TCZ in COVID-19. METHODS In this prospective longitudinal study, 95 individuals with severe COVID-19 were selected between 2020-2022. The recovery process was measured at 24 h, 48 h, and 10 days before and after taking TCZ. All participants were genotyped using RFLP-PCR. Different genotypes of FCGR2A rs1801274 and IL-6R rs2228145 were compared in terms of the recovery process. RESULTS 43.2% of patients were male and 56.8% were female with an average age of 58.20(± 16.214) years. The GA genotype for FCGR2A rs1801274 increased the risk of death (OR = 2.83, P = 0.038) and ventilation (OR = 2.71, P = 0.047) in TCZ-treated individuals. However, there was no risk of death and ventilation with IL-6R rs2228145 (P > 0.05). Additionally, docking analysis showed that not only IL6R but also FCGR2A can be a ligand for TCZ. CONCLUSION This study provides valuable insights into the impact of genetic variations on the response rate of TCZ in COVID-19 patients. The GA genotype for FCGR2A rs1801274 was associated with poor treatment outcomes.
Collapse
Affiliation(s)
- Nastaran Injinari
- Diabetes Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Samira Asadollahi
- Research Center for Food Hygiene and Safety, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Sefid
- Department of Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Maedeh Arshadi
- Department of Epidemiology and Biostatistics, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Saeedeh Sadat Hosseini
- Research Center for Food Hygiene and Safety, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hamed Ghoshouni
- Diabetes Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Soltani
- Diabetes Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Nasim Namiranian
- Diabetes Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Hasan Sheikhha
- Abortion Research Centre, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Science, Yazd, Iran
| | - Fatemeh Aghaeimeybodi
- Department of Internal Medicine, Shahid Sadoughi University of Medical Sciences, Daneshjoo Blvd, Yazd, Iran.
| |
Collapse
|
13
|
Herrera M, Pretelli G, Desai J, Garralda E, Siu LL, Steiner TM, Au L. Bispecific antibodies: advancing precision oncology. Trends Cancer 2024; 10:893-919. [PMID: 39214782 DOI: 10.1016/j.trecan.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/29/2024] [Accepted: 07/12/2024] [Indexed: 09/04/2024]
Abstract
Bispecific antibodies (bsAbs) are engineered molecules designed to target two different epitopes or antigens. The mechanism of action is determined by the bsAb molecular targets and structure (or format), which can be manipulated to create variable and novel functionalities, including linking immune cells with tumor cells, or dual signaling pathway blockade. Several bsAbs have already changed the treatment landscape of hematological malignancies and select solid cancers. However, the mechanisms of resistance to these agents are understudied and the management of toxicities remains challenging. Herein, we review the principles in bsAb engineering, current understanding of mechanisms of action and resistance, data for clinical application, and provide a perspective on ongoing challenges and future developments in this field.
Collapse
Affiliation(s)
- Mercedes Herrera
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Giulia Pretelli
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jayesh Desai
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Elena Garralda
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Lillian L Siu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Thiago M Steiner
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia; Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Lewis Au
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia; Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
| |
Collapse
|
14
|
Saeki K, Nakagama H, Tanaka Y, Goto Y, Kaneshiro K, Kono H, Yanai K, Yamamoto H, Yoneda R, Shimakawa T, Ueki T. Rectum necrosis in a patient with severe COVID19 infection after CAR-T therapy: a case report. Surg Case Rep 2024; 10:227. [PMID: 39325308 PMCID: PMC11427651 DOI: 10.1186/s40792-024-02026-1] [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: 04/02/2024] [Accepted: 09/14/2024] [Indexed: 09/27/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID19) can cause gastrointestinal complications as well as respiratory tract disease. Coagulation abnormalities and thrombosis frequently occur in COVID19, especially in cases with severe clinical outcome. The relationship between gastrointestinal perforation and coagulopathy due to COVID19 remains unclear. CASE PRESENTATION A 49-year-old female received Chimeric antigen receptor T (CAR-T) therapy for an early recurrence of diffuse large B-cell lymphoma (DLBCL) that was refractory to chemotherapy. She was diagnosed with cytokine release syndrome (CRS) because of a fever and oxygen desaturation, and administered tocilizumab. Forty days after completing CAR-T therapy, she was infected with COVID19 and transferred to our hospital. Her general condition worsened and she developed COVID19 pneumonia, and then steroid pulse therapy was started. While her respiratory condition improved, she experienced pain in the anal region and computed tomography (CT) revealed a rectal perforation. An emergency surgery was undertaken, and the lower rectum wall was found to be completely necrotic. Removal of the necrotic part of the rectum tissue, and drainage and lavage of necrotic tissue in the pelvic cavity were performed. The remaining rectum was resected with partial sigmoidectomy, but we could not make the anal stump closed. In addition, an end colostomy in the sigmoid colon was performed. Histopathological findings showed thromboses in the rectal mesentery veins. After the first surgery, the pelvic abscess cavity persisted and her high-grade fever continued. Reoperation was laparoscopically performed, and she underwent a resection of anal canal with residual necrotic rectal and mesorectal tissue, and a drainage of the pelvic abscess. After the reoperation, her general condition improved and CT showed that the abscess cavity had significantly improved. CONCLUSIONS Gastrointestinal perforation, especially rectal necrosis due to coagulopathy caused by severe COVID19 infection, is a rare but life-threatening complication. Physicians should have a high degree of clinical suspicion for timely diagnosis and management, and surgical intervention is necessary in cases of rectal necrosis.
Collapse
Affiliation(s)
- Kiyoshi Saeki
- Department of Surgery, Hamanomachi General Hospital, 3-3-1, Nagahama, Fukuoka, 810-8539, Japan.
| | - Hidenobu Nakagama
- Department of Surgery, Hamanomachi General Hospital, 3-3-1, Nagahama, Fukuoka, 810-8539, Japan
| | - Yuichi Tanaka
- Department of Surgery, Hamanomachi General Hospital, 3-3-1, Nagahama, Fukuoka, 810-8539, Japan
| | - Yoshitaka Goto
- Department of Surgery, Hamanomachi General Hospital, 3-3-1, Nagahama, Fukuoka, 810-8539, Japan
| | - Kazuhisa Kaneshiro
- Department of Surgery, Hamanomachi General Hospital, 3-3-1, Nagahama, Fukuoka, 810-8539, Japan
| | - Hiroshi Kono
- Department of Surgery, Hamanomachi General Hospital, 3-3-1, Nagahama, Fukuoka, 810-8539, Japan
| | - Kosuke Yanai
- Department of Surgery, Hamanomachi General Hospital, 3-3-1, Nagahama, Fukuoka, 810-8539, Japan
| | - Hirofumi Yamamoto
- Department of Surgery, Hamanomachi General Hospital, 3-3-1, Nagahama, Fukuoka, 810-8539, Japan
| | - Reiko Yoneda
- Department of Pathology, Hamanomachi General Hospital, Fukuoka, Japan
| | - Takashi Shimakawa
- Department of Hematology, Hamanomachi General Hospital, Fukuoka, Japan
| | - Takashi Ueki
- Department of Surgery, Hamanomachi General Hospital, 3-3-1, Nagahama, Fukuoka, 810-8539, Japan
| |
Collapse
|
15
|
Schroeder T, Martens T, Fransecky L, Valerius T, Schub N, Pott C, Baldus C, Stölzel F. Management of chimeric antigen receptor T (CAR-T) cell-associated toxicities. Intensive Care Med 2024; 50:1459-1469. [PMID: 39172238 PMCID: PMC11377606 DOI: 10.1007/s00134-024-07576-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/21/2024] [Indexed: 08/23/2024]
Abstract
The use of chimeric antigen receptor T (CAR-T) cells is a significant therapeutic improvement increasing the prognosis for patients with a variety of hematological malignancies. However, this therapy has also sometimes life-threatening, complications. Therefore, knowledge of the treatment and management of these complications, especially in treatment centers and intensive care units, respectively, is of outstanding importance. This review provides recommendations for the diagnosis, management, and treatment of CAR-T cell-associated complications such as cytokine release syndrome, immune effector cell associated neurotoxicity syndrome, hematotoxicity, hypogammaglobulinemia, and CAR-T cell-induced pseudo-progression amongst others for physicians treating patients with CAR-T cell-associated complications and intensivists.
Collapse
Affiliation(s)
- Torsten Schroeder
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Tjark Martens
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Lars Fransecky
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Thomas Valerius
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Natalie Schub
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Christiane Pott
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Claudia Baldus
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany
| | - Friedrich Stölzel
- Department of Hematology and Oncology, Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein Kiel, Kiel University, Arnold-Heller-Str. 3, 24106, Kiel, Germany.
| |
Collapse
|
16
|
Maali A, Noei A, Feghhi-Najafabadi S, Sharifzadeh Z. A Systematic Review on the Dual Role of Interleukin-1 in CAR T-Cell Therapy: Enhancer and Mitigator. IRANIAN BIOMEDICAL JOURNAL 2024; 28:221-34. [PMID: 39891450 PMCID: PMC11829154 DOI: 10.61186/ibj.4444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 10/29/2024] [Indexed: 02/10/2025]
Abstract
Chimeric antigen receptor T-cell therapy is a groundbreaking approach for treating certain hematologic malignancies and solid tumors. However, its application is limited by severe toxicities, particularly CRS and ICANS, dramatically limit its broader application. IL-1 plays a crucial role in both enhancing CAR T-cell efficacy and driving these toxic effects. This review systematically examines the dual functions of IL-1, highlighting its role in promoting CAR T-cell activation and persistence while contributing to CRS and ICANS pathogenesis. Strategies to mitigate IL-1-driven toxicities, including IL-1 receptor antagonists, monoclonal antibodies, IL-1 trapping, and interference with IL-1 production, show promise in reducing adverse effects without compromising therapeutic efficacy. Understanding the complex role of IL-1 in CAR T-cell therapy may lead to optimized treatment strategies, improving safety and expanding clinical applicability. Further research is essential to refine IL-1-targeted interventions and enhance the therapeutic potential of CAR T-cell therapy. Chimeric antigen receptor (CAR) T-cell therapy is a groundbreaking approach for treating certain hematologic malignancies and solid tumors. However, its application is limited by severe toxicities, particularly cytokine release syndrome (CRS) and cell-associated neurotoxicity syndrome (ICANS), dramatically limit its broader application. IL-1 plays a crucial role in both enhancing CAR T-cell efficacy and driving these toxic effects. This review systematically examines the dual functions of IL-1, highlighting its role in promoting CAR T-cell activation and persistence while contributing to CRS and ICANS pathogenesis. Strategies to mitigate IL-1-driven toxicities, including IL-1 receptor antagonists, monoclonal antibodies, IL-1 trapping, and interference with IL-1 production, show promise in reducing adverse effects without compromising therapeutic efficacy. Understanding the complex role of IL-1 in CAR T-cell therapy may lead to optimized treatment strategies, improving safety and expanding clinical applicability. Further research is essential to refine IL-1-targeted interventions and enhance the therapeutic potential of CAR T-cell therapy.
Collapse
Affiliation(s)
- Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Ahmad Noei
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Saba Feghhi-Najafabadi
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
17
|
Cheng Z, Cui X, Li S, Liang Y, Yang W, Ouyang J, Wei M, Yan Z, Yu W. Harnessing cytokines to optimize chimeric antigen receptor-T cell therapy for gastric cancer: Current advances and innovative strategies. Biomed Pharmacother 2024; 178:117229. [PMID: 39096620 DOI: 10.1016/j.biopha.2024.117229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/20/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024] Open
Abstract
Enormous patients with gastric cancer (GC) are insensitive to chemotherapy and targeted therapy without the chance of radical surgery, so immunotherapy may supply a novel choice for them. Chimeric antigen receptor (CAR)-T cell therapy has the advantages of higher specificity, stronger lethality, and longer-lasting efficacy, and it has the potential for GC in the future. However, its application still faces numerous obstacles in terms of accuracy, efficacy, and safety. Cytokines can mediate the migration, proliferation, and survival of immune cells, regulate the duration and strength of immune responses, and are involved in the occurrence of severe side effects in CAR-T cell therapy. The expression levels of specific cytokines are associated with the genesis, invasion, metastasis, and prognosis of GC. Applications of cytokines and their receptors in CAR-T cell therapy have emerged, and various cytokines and their receptors have contributed to improving CAR-T cell anti-tumor capabilities. Large amounts of central cytokines in this therapy include chemokines, interleukins (ILs), transforming growth factor-β (TGF-β), and colony-stimulating factors (CSFs). Meanwhile, researchers have explored the combination therapy in treating GC, and several approaches applied to other malignancies can also be considered as references. Therefore, our review comprehensively outlines the biological functions and clinical significance of cytokines and summarizes current advances and innovative strategies for harnessing cytokines to optimize CAR-T cell therapy for GC.
Collapse
Affiliation(s)
- Zewei Cheng
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaohan Cui
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Song Li
- Department of Medical Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yize Liang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Wenshuo Yang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jun Ouyang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Meng Wei
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhibo Yan
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Wenbin Yu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
18
|
Wang X, Zhang B, Zhang Q, Zhou H, Sun Q, Zhou Y, Li T, Zhou D, Shen Z, Zhang J, Li P, Liang A, Zhou K, Han L, Hu Y, Yang Y, Cao J, Li Z, Xu K, Sang W. Impact of tocilizumab on anti-CD19 chimeric antigen receptor T-cell therapy in B-cell acute lymphoblastic leukemia. Cancer 2024; 130:2660-2669. [PMID: 38578977 DOI: 10.1002/cncr.35316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 04/07/2024]
Abstract
BACKGROUND Tocilizumab is commonly used for the management of chimeric antigen receptor (CAR) T-cell therapy-associated cytokine release syndrome (CRS). However, it remains unknown whether tocilizumab or its dosage affects the efficacy and safety of CAR T-cell therapy. The objective of this multicenter retrospective study was to explore the impact of tocilizumab on CAR T-cell therapy. METHODS In total, 93 patients with B-cell acute lymphoblastic leukemia (B-ALL) receiving humanized anti-CD19 CAR T cells were recruited from May 2016 to November 2022. Forty-five patients received tocilizumab (tocilizumab group), whereas 48 patients did not (nontocilizumab group). Thirteen patients received >1 dose of tocilizumab. The primary end point was the effect of tocilizumab on the efficacy and safety of CAR T cells. Additionally, proliferation, killing, and cytokine assays of CAR T cells were performed in vitro in the presence of tocilizumab. RESULTS The median age of the patients was 33 years, with 47 males and 46 females. Patients in the tocilizumab group showed similar complete response (CR) rate, overall survival (OS), and event-free survival (EFS) compared with the nontocilizumab group. Compared with patients who received ≤1 dose of tocilizumab, receiving >1 dose of tocilizumab did not affect their CR rate, OS, or EFS. In the tocilizumab group, all patients experienced CRS and 26.7% experienced immune effector cell-associated neurotoxicity syndrome (ICANS). In the nontocilizumab group, 64.6% of patients experienced CRS and 8.3% experienced ICANS. Up to 75% of ICANS and 87.5% of grade ≥3 ICANS occurred in the tocilizumab group. In vitro, tocilizumab did not impair the proliferation and killing effects of CAR T cells. CONCLUSIONS Tocilizumab does not affect the efficacy of CAR T cells but may increase the likelihood of ICANS.
Collapse
Affiliation(s)
- Xiangmin Wang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Bingpei Zhang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Qing Zhang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Hongyuan Zhou
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Qian Sun
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Yi Zhou
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Tianci Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Dian Zhou
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Ziyuan Shen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Jiaoli Zhang
- Department of Rehabilitation, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ping Li
- Department of Hematology, Tongji Hospital of Tongji University, Shanghai, China
| | - Aibin Liang
- Department of Hematology, Tongji Hospital of Tongji University, Shanghai, China
| | - Keshu Zhou
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Lu Han
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongxian Hu
- Department of Hematology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yun Yang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiang Cao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Zhenyu Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Wei Sang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
19
|
Wang X, Zhang C, Su J, Ren S, Wang X, Zhang Y, Yuan Z, He X, Wu X, Li M, Du F, Chen Y, Deng S, Zhao Y, Wang X, Sun Y, Shen J, Ji H, Hou Y, Xiao Z. Rejuvenation Strategy for Inducing and Enhancing Autoimmune Response to Eliminate Senescent Cells. Aging Dis 2024:AD.2024.0579. [PMID: 39122450 DOI: 10.14336/ad.2024.0579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
The process of aging, which involves progressive changes in the body over time, is closely associated with the development of age-related diseases. Cellular senescence is a pivotal hallmark and mechanism of the aging process. The accumulation of senescent cells can significantly contribute to the onset of age-related diseases, thereby compromising overall health. Conversely, the elimination of senescent cells enhances the body's regenerative and reparative capacity, thereby retarding the aging process. Here, we present a brief overview of 12 Hallmarks of aging and subsequently emphasize the potential of immune checkpoint blockade, innate immune cell therapy (including T cells, iNKT cells, macrophages, and NK cells), as well as CAR-T cell therapy for inducing and augmenting immune responses aimed at eliminating senescent cells. In addition to CAR-T cells, we also explore the possibility of engineered immune cells such as CAR-NK and CAR-M cells to eliminate senescent cells. In summary, immunotherapy, as an emerging strategy for the treatment of aging, offers new prospects for age-related research.
Collapse
Affiliation(s)
- Xingyue Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Chengyu Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jiahong Su
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Siqi Ren
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yinping Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Zijun Yuan
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xinyu He
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Shuai Deng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Xiaodong Wang
- Department of Hepatobiliary Disease, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Huijiao Ji
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yunqing Hou
- LongmaTan District People's Hospital of Luzhou City, Luzhou 646600, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
- Department of Pharmacology, School of Pharmacy, Sichuan College of Traditional Chinese Medicine, Mianyang 621000, China
| |
Collapse
|
20
|
Ercilla-Rodríguez P, Sánchez-Díez M, Alegría-Aravena N, Quiroz-Troncoso J, Gavira-O'Neill CE, González-Martos R, Ramírez-Castillejo C. CAR-T lymphocyte-based cell therapies; mechanistic substantiation, applications and biosafety enhancement with suicide genes: new opportunities to melt side effects. Front Immunol 2024; 15:1333150. [PMID: 39091493 PMCID: PMC11291200 DOI: 10.3389/fimmu.2024.1333150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 06/14/2024] [Indexed: 08/04/2024] Open
Abstract
Immunotherapy has made significant strides in cancer treatment with strategies like checkpoint blockade antibodies and adoptive T cell transfer. Chimeric antigen receptor T cells (CAR-T) have emerged as a promising approach to combine these strategies and overcome their limitations. This review explores CAR-T cells as a living drug for cancer treatment. CAR-T cells are genetically engineered immune cells designed to target and eliminate tumor cells by recognizing specific antigens. The study involves a comprehensive literature review on CAR-T cell technology, covering structure optimization, generations, manufacturing processes, and gene therapy strategies. It examines CAR-T therapy in haematologic cancers and solid tumors, highlighting challenges and proposing a suicide gene-based mechanism to enhance safety. The results show significant advancements in CAR-T technology, particularly in structure optimization and generation. The manufacturing process has improved for broader clinical application. However, a series of inherent challenges and side effects still need to be addressed. In conclusion, CAR-T cells hold great promise for cancer treatment, but ongoing research is crucial to improve efficacy and safety for oncology patients. The proposed suicide gene-based mechanism offers a potential solution to mitigate side effects including cytokine release syndrome (the most common toxic side effect of CAR-T therapy) and the associated neurotoxicity.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Genes, Transgenic, Suicide
- Neoplasms/therapy
- Neoplasms/immunology
- Neoplasms/genetics
- T-Lymphocytes/immunology
- Animals
- Genetic Therapy/adverse effects
- Genetic Therapy/methods
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
| | - Marta Sánchez-Díez
- ETSIAAB, Universidad Politécnica de Madrid, Madrid, Spain
- Laboratorio Cancer Stem Cell, HST group, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
| | - Nicolás Alegría-Aravena
- Grupo de Biología y Producción de Cérvidos, Instituto de Desarrollo Regional, Universidad de Castilla-La Mancha, Albacete, Spain
- Asociación Española Contra el Cáncer (AECC)-Fundación Científica AECC, Albacete, Spain
| | - Josefa Quiroz-Troncoso
- ETSIAAB, Universidad Politécnica de Madrid, Madrid, Spain
- Laboratorio Cancer Stem Cell, HST group, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
| | - Clara E. Gavira-O'Neill
- Laboratorio Cancer Stem Cell, HST group, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
- Sección de Oncología, Instituto de Investigación Sanitaria San Carlos, Madrid, Spain
| | - Raquel González-Martos
- ETSIAAB, Universidad Politécnica de Madrid, Madrid, Spain
- Laboratorio Cancer Stem Cell, HST group, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
| | - Carmen Ramírez-Castillejo
- ETSIAAB, Universidad Politécnica de Madrid, Madrid, Spain
- Laboratorio Cancer Stem Cell, HST group, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
- Sección de Oncología, Instituto de Investigación Sanitaria San Carlos, Madrid, Spain
| |
Collapse
|
21
|
Kurnik M, Peter F, Matej P. Tocilizumab and CytoSorb for delayed severe cytokine release syndrome after ipilimumab plus nivolumab immunotherapy. Immunotherapy 2024; 16:791-801. [PMID: 39016056 PMCID: PMC11457641 DOI: 10.1080/1750743x.2024.2370180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Cytokine release syndrome (CRS) is immune dysregulation phenomenon that is associated with immune checkpoint inhibitors. It is still difficult to distinguish CRS from other dangerous, acute and life-threatening medical disorders.We present a case of delayed grade 4 CRS following treatment of lung adenocarcinoma with ipilimumab plus nivolumab that warranted intensive care level treatment with abundant fluid resuscitation, two-tire vasopressor support, high-flow nasal oxygenation, corticosteroids in high dosages, as well as sustained low-efficiency daily diafiltration with CytoSorb hemadsorption and tocilizumab. Initial treatment of presumed septic shock of unknown origin did not yield results.After initiation of corticosteroids and particularly CytoSorb hemadsorption and tocilizumab, prompt clinical and laboratory improvement was observed.
Collapse
Affiliation(s)
- Marko Kurnik
- General Hospital Celje, Department of Internal Intensive Medicine, Celje, Slovenia
| | - Fazarinc Peter
- General Hospital Celje, Department of Hematology & Oncology, Celje, Slovenia
| | - Podbregar Matej
- General Hospital Celje, Department of Internal Intensive Medicine, Celje, Slovenia
- University of Ljubljana, Medical Faculty, Ljubljana, Slovenia
| |
Collapse
|
22
|
Liu B, Zhou H, Tan L, Siu KTH, Guan XY. Exploring treatment options in cancer: Tumor treatment strategies. Signal Transduct Target Ther 2024; 9:175. [PMID: 39013849 PMCID: PMC11252281 DOI: 10.1038/s41392-024-01856-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 07/18/2024] Open
Abstract
Traditional therapeutic approaches such as chemotherapy and radiation therapy have burdened cancer patients with onerous physical and psychological challenges. Encouragingly, the landscape of tumor treatment has undergone a comprehensive and remarkable transformation. Emerging as fervently pursued modalities are small molecule targeted agents, antibody-drug conjugates (ADCs), cell-based therapies, and gene therapy. These cutting-edge treatment modalities not only afford personalized and precise tumor targeting, but also provide patients with enhanced therapeutic comfort and the potential to impede disease progression. Nonetheless, it is acknowledged that these therapeutic strategies still harbour untapped potential for further advancement. Gaining a comprehensive understanding of the merits and limitations of these treatment modalities holds the promise of offering novel perspectives for clinical practice and foundational research endeavours. In this review, we discussed the different treatment modalities, including small molecule targeted drugs, peptide drugs, antibody drugs, cell therapy, and gene therapy. It will provide a detailed explanation of each method, addressing their status of development, clinical challenges, and potential solutions. The aim is to assist clinicians and researchers in gaining a deeper understanding of these diverse treatment options, enabling them to carry out effective treatment and advance their research more efficiently.
Collapse
Affiliation(s)
- Beilei Liu
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| | - Hongyu Zhou
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Licheng Tan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Kin To Hugo Siu
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China.
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, China.
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China.
| |
Collapse
|
23
|
Hughes AD, Teachey DT, Diorio C. Riding the storm: managing cytokine-related toxicities in CAR-T cell therapy. Semin Immunopathol 2024; 46:5. [PMID: 39012374 PMCID: PMC11252192 DOI: 10.1007/s00281-024-01013-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/18/2024] [Indexed: 07/17/2024]
Abstract
The advent of chimeric antigen receptor T cells (CAR-T) has been a paradigm shift in cancer immunotherapeutics, with remarkable outcomes reported for a growing catalog of malignancies. While CAR-T are highly effective in multiple diseases, salvaging patients who were considered incurable, they have unique toxicities which can be life-threatening. Understanding the biology and risk factors for these toxicities has led to targeted treatment approaches which can mitigate them successfully. The three toxicities of particular interest are cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and immune effector cell-associated hemophagocytic lymphohistiocytosis (HLH)-like syndrome (IEC-HS). Each of these is characterized by cytokine storm and hyperinflammation; however, they differ mechanistically with regard to the cytokines and immune cells that drive the pathophysiology. We summarize the current state of the field of CAR-T-associated toxicities, focusing on underlying biology and how this informs toxicity management and prevention. We also highlight several emerging agents showing promise in preclinical models and the clinic. Many of these established and emerging agents do not appear to impact the anti-tumor function of CAR-T, opening the door to additional and wider CAR-T applications.
Collapse
Affiliation(s)
- Andrew D Hughes
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David T Teachey
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Caroline Diorio
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Immune Dysregulation Frontier Program, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Chang JS, Kim JH. Cytokine Release Syndrome in a Patient With Metastatic Triple-Negative Breast Cancer Treated With Hypofractionated Radiation Therapy, Who Had Previously Undergone Immunotherapy: A Case Report. Adv Radiat Oncol 2024; 9:101513. [PMID: 38883994 PMCID: PMC11179535 DOI: 10.1016/j.adro.2024.101513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/03/2024] [Indexed: 06/18/2024] Open
Affiliation(s)
- Jee Suk Chang
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, Korea
| | - Jee Hung Kim
- Division of Medical Oncology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
25
|
Wu Y, Chen D, Gao Y, Xu Q, Zhou Y, Ni Z, Na M. Immunosuppressive regulatory cells in cancer immunotherapy: restrain or modulate? Hum Cell 2024; 37:931-943. [PMID: 38814516 DOI: 10.1007/s13577-024-01083-w] [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: 04/14/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
Immunosuppressive regulatory cells (IRCs) play important roles in negatively regulating immune response, and are mainly divided into myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs). Large numbers of preclinical and clinical studies have shown that inhibition or reduction of IRCs could effectively elevate antitumor immune responses. However, several studies also reported that excessive inhibition of IRCs function is one of the main reasons causing the side effects of cancer immunotherapy. Therefore, the reasonable regulation of IRCs is crucial for improving the safety and efficiency of cancer immunotherapy. In this review, we summarised the recent research advances in the cancer immunotherapy by regulating the proportion of IRCs, and discussed the roles of IRCs in regulating tumour immune evasion and drug resistance to immunotherapies. Furthermore, we also discussed how to balance the potential opportunities and challenges of using IRCs to improve the safety of cancer immunotherapies.
Collapse
Affiliation(s)
- Yan Wu
- Department of Burn and Plastic Surgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, 210031, Jiangsu, People's Republic of China
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Dongfeng Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Yang Gao
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, China
| | - Qinggang Xu
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Yang Zhou
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Zhong Ni
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Manli Na
- Department of Burn and Plastic Surgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, 210031, Jiangsu, People's Republic of China.
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China.
- International Genome Center, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China.
| |
Collapse
|
26
|
Karthikeyan B, Sunder SS, Puzanov I, Olejniczak SH, Pokharel S, Sharma UC. Cardiotoxic profiles of CAR-T therapy and bispecific T-cell engagers in hematological cancers. COMMUNICATIONS MEDICINE 2024; 4:116. [PMID: 38871977 PMCID: PMC11176393 DOI: 10.1038/s43856-024-00540-9] [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: 10/26/2023] [Accepted: 05/29/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T-cell therapy and bispecific T-cell engagers, which redirect T-cells to tumor antigens, have immensely benefitted patients with relapsed/refractory B-cell cancers. How these therapies differ in cardiotoxicity is underexplored. We used the World Health Organization pharmacovigilance database, VigiBase, to compare cardiotoxicity profiles between CD19-targeted CAR-T therapy and blinatumomab (a CD19/CD3-targeted bispecific T-cell engager). METHODS Safety reports in VigiBase were filtered for diffuse large B-cell lymphoma (DLBCL, n = 17,479) and acute lymphocytic leukemia (ALL, n = 28,803) for all adverse reactions. Data were further filtered for patients taking CAR-T therapy or blinatumomab. Reporting odds ratios (ROR) and fatality rates were compared between CAR-T cell products (e.g. tisagenlecleucel and axicabtagene ciloleucel), and between CAR-T therapy and blinatumomab. RESULTS Tisagenlecleucel is associated with cardiac failure (IC025 = 0.366) with fatality rates of 85.7% and 80.0% in DLBCL and pediatric ALL patients respectively. For DLBCL patients, axicabtagene ciloleucel has greater reporting for hypotension than tisagenlecleucel (ROR: 2.54; 95% CI: 1.28-5.03; p = 0.012), but tisagenlecleucel has higher fatality rates for hypotension than axicabtagene ciloleucel [50.0% (tisagenlecleucel) vs 5.6% (axicabtagene ciloleucel); p < 0.001]. Blinatumomab and tisagenlecleucel have similar fatality rates for hypotension in pediatric ALL patients [34.7% (tisagenlecleucel) vs 20.0% (blinatumomab); p = 0.66]. CONCLUSIONS Tisagenlecleucel is associated with severe and fatal adverse cardiac events, with higher fatality rates for hypotension compared to axicabtagene ciloleucel in DLBCL patients, but similar hypotension fatality rates compared to blinatumomab in pediatric ALL patients. Effective management necessitates experienced physicians, including cardio-oncologists, skilled in interdisciplinary approaches to manage these toxicities.
Collapse
Affiliation(s)
- Badri Karthikeyan
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, 14203, USA
- Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Sunitha Shyam Sunder
- Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Igor Puzanov
- Department of Medicine, Division of Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Scott H Olejniczak
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Saraswati Pokharel
- Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Umesh C Sharma
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, 14203, USA.
| |
Collapse
|
27
|
Shen J, Li J, Lei Y, Chen Z, Wu L, Lin C. Frontiers and hotspots evolution in cytokine storm: A bibliometric analysis from 2004 to 2022. Heliyon 2024; 10:e30955. [PMID: 38774317 PMCID: PMC11107250 DOI: 10.1016/j.heliyon.2024.e30955] [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: 07/17/2023] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 05/24/2024] Open
Abstract
Background As a fatal disease, cytokine storm has garnered research attention in recent years. Nonetheless, as the body of related studies expands, a thorough and impartial evaluation of the current status of research on cytokine storms remains absent. Consequently, this study aimed to thoroughly explore the research landscape and evolution of cytokine storm utilizing bibliometric and knowledge graph approaches. Methods Research articles and reviews centered on cytokine storms were retrieved from the Web of Science Core Collection database. For bibliometric analysis, tools such as Excel 365, CiteSpace, VOSviewer, and the Bibliometrix R package were utilized. Results This bibliometric analysis encompassed 6647 articles published between 2004 and 2022. The quantity of pertinent articles and citation frequency exhibited a yearly upward trend, with a sharp increase starting in 2020. Network analysis of collaborations reveals that the United States holds a dominant position in this area, boasting the largest publication count and leading institutions. Frontiers in Immunology ranks as the leading journal for the largest publication count in this area. Stephan A. Grupp, a prominent researcher in this area, has authored the largest publication count and has the second-highest citation frequency. Research trends and keyword evaluations show that the connection between cytokine storm and COVID-19, as well as cytokine storm treatment, are hot topics in research. Furthermore, research on cytokine storm and COVID-19 sits at the forefront in this area. Conclusion This study employed bibliometric analysis to create a visual representation of cytokine storm research, revealing current trends and burgeoning topics in this area for the first time. It will provide valuable insights, helping scholars pinpoint critical research areas and potential collaborators.
Collapse
Affiliation(s)
- Junyi Shen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiaming Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yuqi Lei
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhengrui Chen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Lingling Wu
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Chunyan Lin
- Department of Teaching and Research Section of Internal Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
28
|
Yoshikawa T, Ito Y, Wu Z, Kasuya H, Nakashima T, Okamoto S, Amaishi Y, Zhang H, Li Y, Matsukawa T, Inoue S, Kagoya Y. Development of a chimeric cytokine receptor that captures IL-6 and enhances the antitumor response of CAR-T cells. Cell Rep Med 2024; 5:101526. [PMID: 38670095 PMCID: PMC11148643 DOI: 10.1016/j.xcrm.2024.101526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 10/06/2023] [Accepted: 04/03/2024] [Indexed: 04/28/2024]
Abstract
The efficacy of chimeric antigen receptor (CAR)-engineered T cell therapy is suboptimal in most cancers, necessitating further improvement in their therapeutic actions. However, enhancing antitumor T cell response inevitably confers an increased risk of cytokine release syndrome associated with monocyte-derived interleukin-6 (IL-6). Thus, an approach to simultaneously enhance therapeutic efficacy and safety is warranted. Here, we develop a chimeric cytokine receptor composed of the extracellular domains of GP130 and IL6RA linked to the transmembrane and cytoplasmic domain of IL-7R mutant that constitutively activates the JAK-STAT pathway (G6/7R or G6/7R-M452L). CAR-T cells with G6/7R efficiently absorb and degrade monocyte-derived IL-6 in vitro. The G6/7R-expressing CAR-T cells show superior expansion and persistence in vivo, resulting in durable antitumor response in both liquid and solid tumor mouse models. Our strategy can be widely applicable to CAR-T cell therapy to enhance its efficacy and safety, irrespective of the target antigen.
Collapse
Affiliation(s)
- Toshiaki Yoshikawa
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan; Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan
| | - Yusuke Ito
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan; Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan
| | - Zhiwen Wu
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan
| | - Hitomi Kasuya
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan
| | - Takahiro Nakashima
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan; Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan; Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, Nagoya 467-8601, Japan
| | | | | | - Haosong Zhang
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan; Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan; Division of Cellular Oncology, Department of Cancer Diagnostics and Therapeutics, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yang Li
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan; Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan; Division of Cellular Oncology, Department of Cancer Diagnostics and Therapeutics, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Tetsuya Matsukawa
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan; Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan; Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Satoshi Inoue
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan; Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan
| | - Yuki Kagoya
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan; Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan; Division of Cellular Oncology, Department of Cancer Diagnostics and Therapeutics, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
29
|
Huang Y, Qin Y, He Y, Qiu D, Zheng Y, Wei J, Zhang L, Yang DH, Li Y. Advances in molecular targeted drugs in combination with CAR-T cell therapy for hematologic malignancies. Drug Resist Updat 2024; 74:101082. [PMID: 38569225 DOI: 10.1016/j.drup.2024.101082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/03/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
Molecular targeted drugs and chimeric antigen receptor (CAR) T cell therapy represent specific biological treatments that have significantly improved the efficacy of treating hematologic malignancies. However, they face challenges such as drug resistance and recurrence after treatment. Combining molecular targeted drugs and CAR-T cells could regulate immunity, improve tumor microenvironment (TME), promote cell apoptosis, and enhance sensitivity to tumor cell killing. This approach might provide a dual coordinated attack on cancer cells, effectively eliminating minimal residual disease and overcoming therapy resistance. Moreover, molecular targeted drugs can directly or indirectly enhance the anti-tumor effect of CAR-T cells by inducing tumor target antigen expression, reversing CAR-T cell exhaustion, and reducing CAR-T cell associated toxic side effects. Therefore, combining molecular targeted drugs with CAR-T cells is a promising and novel tactic for treating hematologic malignancies. In this review article, we focus on analyzing the mechanism of therapy resistance and its reversal of CAR-T cell therapy resistance, as well as the synergistic mechanism, safety, and future challenges in CAR-T cell therapy in combination with molecular targeted drugs. We aim to explore the benefits of this combination therapy for patients with hematologic malignancies and provide a rationale for subsequent clinical studies.
Collapse
Affiliation(s)
- Yuxian Huang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China.
| | - Yinjie Qin
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China
| | - Yingzhi He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China
| | - Dezhi Qiu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China
| | - Yeqin Zheng
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China
| | - Jiayue Wei
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China
| | - Lenghe Zhang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China
| | - Dong-Hua Yang
- New York College of Traditional Chinese Medicine, Mineola, NY, USA.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China.
| |
Collapse
|
30
|
Ntwali F, Gilliaux Q, Honoré PM. Nivolumab-Induced Cytokine Release Syndrome: A Case Report and Literature Review. AMERICAN JOURNAL OF CASE REPORTS 2024; 25:e941835. [PMID: 38625840 PMCID: PMC11034389 DOI: 10.12659/ajcr.941835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 03/03/2024] [Accepted: 01/17/2024] [Indexed: 04/18/2024]
Abstract
BACKGROUND CRS (cytokine release syndrome) is a massive activation of the inflammatory system characterized by a supra-physiological rate of inflammatory cytokines. The interleukin 6 cytokine plays a central role in CRS. The main clinical sign of CRS is fever, but CRS can lead to multiple organ failure in severe cases. CRS is usually described in sepsis, more recently in SARS COV-2 infection, and in chimeric antigen receptor T-cell therapy. However, it can also be associated with immune checkpoint inhibitors (ICIs), which is infrequently described. ICI have growing indications and can lead to CRS by causing an uncontrolled activation of the immune system. There are currently no treatment guidelines for ICI-induced CRS. CASE REPORT We report a rare case of grade 3 CRS induced by nivolumab associated with 5-fluorouracil and oxaliplatin for gastric cancer. The patient was 65-year-old man with an adenocarcinoma of the cardia. CRS developed during the tenth course of treatment and was characterized by fever, hypotension requiring vasopressors, hypoxemia, acute kidney injury, and thrombopenia. The patient was transferred quickly to the Intensive Care Unit. He was treated for suspected sepsis, but it was ruled out after multiple laboratory examinations. There was rapid resolution after infusion of hydrocortisone. CONCLUSIONS The use of ICIs is expanding. Nivolumab-induced CRS is rarely described but can be severe and lead to multiple organ dysfunction; therefore, intensive care practitioners should be informed about this adverse effect. More studies are needed to better understand this condition and establish treatment guidelines.
Collapse
Affiliation(s)
- Francis Ntwali
- Intensive Care Unit, UCL Namur University Hospital, Yvoir, Belgium
| | - Quentin Gilliaux
- Department of Oncology, UCL Namur University Hospital, Yvoir, Belgium
| | | |
Collapse
|
31
|
Zhang J, Ding X, Ding X. Exploring the efficacy and safety of anti-BCMA chimeric antigen receptor T-cell therapy for multiple myeloma: Systematic review and meta-analysis. Cytojournal 2024; 21:13. [PMID: 38628287 PMCID: PMC11021094 DOI: 10.25259/cytojournal_64_2023] [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: 08/23/2023] [Accepted: 10/31/2023] [Indexed: 04/19/2024] Open
Abstract
Objective Multiple myeloma (MM) is a bone marrow cancer that profoundly affects plasma cells involved in the immune response. Myeloma cells alter the average production of cells in the bone marrow. Anti-B-cell maturation antigen (BCMA) chimeric antigen receptor (CAR) T-cell therapy allows genetic modifications of an individual's T-cells to increase the expression of CARs used to identify and attach BCMA proteins to the malignant cells. Our main objective is to perform a systematic review and meta-analysis to explore the efficacy and safety of anti-BCMA CAR T-cell therapy for MM. Material and Methods We searched five databases, PubMed, CNKI, EMBASE, Cochrane, Web of Science, and CNKI, for studies published on anti-BCMA,CAR-T-cell treatment for MM. Inclusion criteria involved prospective single-arm studies either single or multi-center, in various MM phases and studies that reported anti-BCMA,CAR-T-cell treatment for MM. We excluded non-English publications and conference papers. All statistical analyses were performed in R software and Review Manager 5.4.1. Results Thirteen articles were included in the analysis. We found that the overall response survival complete response increase was statistically significant. Similarly, the reduction in cytokine release syndrome grades 3 and 4 and neurotoxicity after follow-up was statistically significant. However, the reduction in minimal residual disease negativity (MRDN) was not statistically significant. Conclusion Using anti-BCMA CAR T-cell therapy in MM was highly efficacious and safe in lowering the adverse outcomes and improving the survival outcomes, complete response, and overall response.
Collapse
Affiliation(s)
- Jia Zhang
- Department of Hematology and Oncology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, China
| | - Xinhua Ding
- Department of Oncology, Tiantai People’s Hospital of Zhejiang Province, Taizhou, China
| | - Xiaoxiao Ding
- Department of Hematology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| |
Collapse
|
32
|
Sun Z, Zhao H, Ma L, Shi Y, Ji M, Sun X, Ma D, Zhou W, Huang T, Zhang D. The quest for nanoparticle-powered vaccines in cancer immunotherapy. J Nanobiotechnology 2024; 22:61. [PMID: 38355548 PMCID: PMC10865557 DOI: 10.1186/s12951-024-02311-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
Despite recent advancements in cancer treatment, this disease still poses a serious threat to public health. Vaccines play an important role in preventing illness by preparing the body's adaptive and innate immune responses to combat diseases. As our understanding of malignancies and their connection to the immune system improves, there has been a growing interest in priming the immune system to fight malignancies more effectively and comprehensively. One promising approach involves utilizing nanoparticle systems for antigen delivery, which has been shown to potentiate immune responses as vaccines and/or adjuvants. In this review, we comprehensively summarized the immunological mechanisms of cancer vaccines while focusing specifically on the recent applications of various types of nanoparticles in the field of cancer immunotherapy. By exploring these recent breakthroughs, we hope to identify significant challenges and obstacles in making nanoparticle-based vaccines and adjuvants feasible for clinical application. This review serves to assess recent breakthroughs in nanoparticle-based cancer vaccinations and shed light on their prospects and potential barriers. By doing so, we aim to inspire future immunotherapies for cancer that harness the potential of nanotechnology to deliver more effective and targeted treatments.
Collapse
Affiliation(s)
- Zhe Sun
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Hui Zhao
- Department of Endodontics, East Branch of Jinan Stomatological Hospital, Jinan, 250000, Shandong, China
| | - Li Ma
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yanli Shi
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Mei Ji
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Xiaodong Sun
- Department of Endodontics, Gaoxin Branch of Jinan Stomatological Hospital, Jinan, 250000, Shandong, China
| | - Dan Ma
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Wei Zhou
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Tao Huang
- Department of Biomedical Engineering, Graeme Clark Institute, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Dongsheng Zhang
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
33
|
Chen Z, Hu Y, Mei H. Harnessing Biomaterials for Safeguarding Chimeric Antigen Receptor T Cell Therapy: An Artful Expedition in Mitigating Adverse Effects. Pharmaceuticals (Basel) 2024; 17:139. [PMID: 38276012 PMCID: PMC10819334 DOI: 10.3390/ph17010139] [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: 12/20/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy has emerged as a groundbreaking approach in cancer treatment, showcasing remarkable efficacy. However, the formidable challenge lies in taming the formidable side effects associated with this innovative therapy, among which cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS) and on-target off-tumor toxicities (OTOT) are typical representatives. Championing the next frontier in cellular immunotherapy, this comprehensive review embarks on an artistic exploration of leveraging biomaterials to meticulously navigate the intricate landscape of CAR-T cell therapy. Unraveling the tapestry of potential toxicities, our discourse unveils a symphony of innovative strategies designed to elevate the safety profile of this revolutionary therapeutic approach. Through the lens of advanced medical science, we illuminate the promise of biomaterial interventions in sculpting a safer and more efficacious path for CAR-T cell therapy, transcending the boundaries of conventional treatment paradigms.
Collapse
Affiliation(s)
- Zhaozhao Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China;
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China;
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China;
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| |
Collapse
|
34
|
Willbanks A, Seals M, Karmali R, Roy I. Harnessing the Systemic Biology of Functional Decline and Cachexia to Inform more Holistic Therapies for Incurable Cancers. Cancers (Basel) 2024; 16:360. [PMID: 38254849 PMCID: PMC10814065 DOI: 10.3390/cancers16020360] [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: 12/13/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Options for treatment of incurable cancer remain scarce and are largely focused on limited therapeutic mechanisms. A new approach specific to advanced cancers is needed to identify new and effective treatments. Morbidity in advanced cancer is driven by functional decline and a number of systemic conditions, including cachexia and fatigue. This review will focus on these clinical concepts, describe our current understanding of their underlying biology, and then propose how future therapeutic strategies, including pharmaceuticals, exercise, and rehabilitation, could target these mechanisms as an alternative route to addressing incurable cancer.
Collapse
Affiliation(s)
| | - Mina Seals
- Shirley Ryan AbilityLab, Chicago, IL 60611, USA
| | - Reem Karmali
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Ishan Roy
- Shirley Ryan AbilityLab, Chicago, IL 60611, USA
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
35
|
Saedmocheshi S, Amiri E, Mehdipour A, Stefani GP. The Effect of Vitamin D Consumption on Pro-Inflammatory Cytokines in Athletes: A Systematic Review of Randomized Controlled Trials. Sports (Basel) 2024; 12:32. [PMID: 38251306 PMCID: PMC10821273 DOI: 10.3390/sports12010032] [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: 12/11/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/23/2024] Open
Abstract
Vitamin D is essential for the optimal health of the skeletal system. However, this vitamin also plays a role in other functions of the human body, such as muscle, immune, and inflammatory functions. Some studies have reported that adequate levels of vitamin D improve immune system function by reducing the levels of certain pro-inflammatory cytokines, which can protect against the risk of post-exercise illness. This systematic review aims to investigate the effects of vitamin D supplementation on pro-inflammatory cytokines in athletes. This study was conducted using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. A literature search was conducted in SPORTDiscus, PubMed, ScienceDirect, and Google Scholar up to 1 October 2023. The quality of the articles was evaluated using the Risk of Bias 2 Tool. After searching the databases, a total of 7417 studies were identified, 6 of which met the eligibility criteria, and their outcomes were presented. The six studies included 176 participants. All six studies are randomized control trials, including a total of 176 subjects, primarily men (81%). Regarding the types of athletes, most participants were endurance athletes. Our investigation in this systematic review demonstrated that out of the six studies, only two of them reported significant changes in IL-6 and TNF-α levels after taking high-dose vitamin D. Other studies did not present any significant changes after vitamin D supplementation in athletes with respect to IL-6 and TNF-α levels. Further studies are needed to determine the effectiveness of vitamin D supplementation for athletes as a disease-prone community.
Collapse
Affiliation(s)
- Saber Saedmocheshi
- Department of Physical Education and Sport Sciences, Faculty of Humanities and Social Sciences, University of Kurdistan, Sanandaj 6617715175, Iran; (S.S.)
| | - Ehsan Amiri
- Department of Physical Education and Sport Sciences, Faculty of Humanities and Social Sciences, University of Kurdistan, Sanandaj 6617715175, Iran; (S.S.)
| | - Aref Mehdipour
- Department of Physical Education and Sport Sciences, Faculty of Humanities and Social Sciences, University of Kurdistan, Sanandaj 6617715175, Iran; (S.S.)
| | - Giuseppe Potrick Stefani
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619-900, Brazil
| |
Collapse
|
36
|
Raghani NR, Chorawala MR, Mahadik M, Patel RB, Prajapati BG, Parekh PS. Revolutionizing cancer treatment: comprehensive insights into immunotherapeutic strategies. Med Oncol 2024; 41:51. [PMID: 38195781 DOI: 10.1007/s12032-023-02280-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/02/2023] [Indexed: 01/11/2024]
Abstract
Cancer, characterized by the uncontrolled proliferation of aberrant cells, underscores the imperative for innovative therapeutic approaches. Immunotherapy has emerged as a pivotal constituent in cancer treatment, offering improved prognostic outcomes for a substantial patient cohort. Noteworthy for its precision, immunotherapy encompasses strategies such as adoptive cell therapy and checkpoint inhibitors, orchestrating the immune system to recognize and selectively target malignant cells. Exploiting the specificity of the immune response renders immunotherapy efficacious, as it selectively targets the body's immune milieu. Diverse mechanisms underlie cancer immunotherapies, leading to distinct toxicity profiles compared to conventional treatments. A remarkable clinical stride in the anticancer resources is immunotherapy. Remarkably, certain recalcitrant cancers like skin malignancies exhibit resistance to radiation or chemotherapy, yet respond favorably to immunotherapeutic interventions. Notably, combination therapies involving chemotherapy and immunotherapy have exhibited synergistic effects, enhancing overall therapeutic efficacy. Understanding the pivotal role of immunotherapy elucidates its complementary value, bolstering the therapeutic landscape. In this review, we elucidate the taxonomy of cancer immunotherapy, encompassing adoptive cell therapy and checkpoint inhibitors, while scrutinizing their distinct adverse event profiles. Furthermore, we expound on the unprecedented potential of immunogenic vaccines to bolster the anticancer immune response. This comprehensive analysis underscores the significance of immunotherapy in modern oncology, unveiling novel prospects for tailored therapeutic regimens.
Collapse
Affiliation(s)
- Neha R Raghani
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Mayuresh Mahadik
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Mehsana, Gujarat, India
| | - Rakesh B Patel
- Department of Internal Medicine, Division of Hematology and Oncology, UI Carver College of Medicine: The University of Iowa Roy J and Lucille A Carver College of Medicine, 375 Newton Rd, Iowa City, IA, 52242, USA
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Mehsana, Gujarat, India.
| | - Priyajeet S Parekh
- A V Pharma LLC, 1545 University Blvd N Ste A, Jacksonville, FL, 32211, USA
| |
Collapse
|
37
|
Dou B, Ren S, Qiu L, Zhang X, Zhang N, Cai J, Chen D, Zhang Q, Yao H, Fan F. Prophylactic use of interleukin 6 monoclonal antibody can reduce CRS response of CAR-T cell therapy. Front Med (Lausanne) 2024; 10:1265835. [PMID: 38264058 PMCID: PMC10804994 DOI: 10.3389/fmed.2023.1265835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/30/2023] [Indexed: 01/25/2024] Open
Abstract
Background Chimeric antigen receptor T (CAR-T) cell immunotherapy is becoming one of the most promising treatments for hematological malignancies, however, complications such as cytokine release syndrome (CRS) seriously threaten the lives of patients. Interleukin 6(IL-6) monoclonal antibody is the common and useful treatment of CRS, however, it is not clear whether prophylactic use IL-6 monoclonal antibody before CAR-T therapy can reduce the incidence of CRS. Purpose This study aims to systematically evaluate whether the prophylactic use of IL-6 monoclonal antibody can reduce the incidence of CRS. Data sources and methods We searched the PubMed, Embase, web of Science, and Cochrane Library databases for studies that reported the prophylactic use of IL-6 monoclonal antibody in the treatment of CRS-related complications of CAR-T cell immunotherapy before December 2022. The literature is screened according to the established inclusion and exclusion criteria, relevant data are extracted, and the quality of the literature is evaluated using the scale Cochrane bias risk assessment tool, and the Review Manager 5.3 is used to draw for related charts. Since the two experimental data only provide the median, the maximum and minimum values of the data, the mean and standard (Standard Deviation, SD) are calculated by this document Delai, and finally use Review Manager for data processing, and STATA software for supplementation. Results A total of 2 trials with a total of 37 participants were included in this study. Meta-analysis showed that compared with no use of IL-6 monoclonal antibody to prevent CRS, IL-6 monoclonal antibody was given to patients at 8 mg/kg one hour before CAR-T cell infusion, which reduced the incidence of CRS [RR: 0.41 95% confidence interval (0.20, 0.86) I[2] = 0.0% P = 0.338 z = -2.369 (p = 0.018)]. In subgroup analysis, compared with those who did not use IL-6 monoclonal antibody to prevent CRS, IL-6 monoclonal antibody was given to patients at 8 mg/kg one hour before CAR-T cell infusion, which reduced lactate dehydrogenase (LDH)[MD: -617.21, 95% confidence interval (-1104.41, -130.01) I[2] = 0% P = 0.88 Z = 2.48 (P = 0.01)], prophylactic use of IL-6 monoclonal antibody has a significant effect on reducing peak C-reactive protein (CRP) after CAR-T therapy [MD: -11.58, 95% confidence interval (-15.28, -7.88) I[2] = 0.0% P = 0.73 z = 6.14 (p < 0.00001)]. Conclusion The prophylactic use of IL-6 monoclonal antibody can significantly reduce the incidence of CRS complications after CAR-T therapy, can also reduce LDH vaule and peak CRP vaule after CAR-T therapy. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42023487662, identifier CRD42023487662.
Collapse
Affiliation(s)
- Baitao Dou
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
- General Hospital of the Chinese People’s Liberation Army Western Theater, Chengdu, Sichuan, China
| | - Shihui Ren
- General Hospital of the Chinese People’s Liberation Army Western Theater, Chengdu, Sichuan, China
| | - Ling Qiu
- General Hospital of the Chinese People’s Liberation Army Western Theater, Chengdu, Sichuan, China
| | - Xupai Zhang
- General Hospital of the Chinese People’s Liberation Army Western Theater, Chengdu, Sichuan, China
| | - Nan Zhang
- General Hospital of the Chinese People’s Liberation Army Western Theater, Chengdu, Sichuan, China
| | - Jiao Cai
- General Hospital of the Chinese People’s Liberation Army Western Theater, Chengdu, Sichuan, China
| | - Dan Chen
- General Hospital of the Chinese People’s Liberation Army Western Theater, Chengdu, Sichuan, China
| | - Qian Zhang
- General Hospital of the Chinese People’s Liberation Army Western Theater, Chengdu, Sichuan, China
| | - Hao Yao
- General Hospital of the Chinese People’s Liberation Army Western Theater, Chengdu, Sichuan, China
| | - Fangyi Fan
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
- General Hospital of the Chinese People’s Liberation Army Western Theater, Chengdu, Sichuan, China
| |
Collapse
|
38
|
Schulert GS, Zhang K. Genetics of Acquired Cytokine Storm Syndromes : Secondary HLH Genetics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1448:103-119. [PMID: 39117810 DOI: 10.1007/978-3-031-59815-9_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Secondary hemophagocytic lymphohistiocytosis (sHLH) has historically been defined as a cytokine storm syndrome (CSS) occurring in the setting of triggers leading to strong and dysregulated immunological activation, without known genetic predilection. However, recent studies have suggested that existing underlying genetic factors may synergize with particular diseases and/or environmental triggers (including infection, autoimmune/autoinflammatory disorder, certain biologic therapies, or malignant transformation), leading to sHLH. With the recent advances in genetic testing technology, more patients are examined for genetic variations in primary HLH (pHLH)-associated genes, including through whole exome and whole genome sequencing. This expanding genetic and genomic evidence has revealed HLH as a more complex phenomenon, resulting from specific immune challenges in patients with a susceptible genetic background. Rather than a simple, binary definition of pHLH and sHLH, HLH represents a spectrum of diseases, from a severe complication of common infections (EBV, influenza) to early onset familial diseases that can only be cured by transplantation.
Collapse
Affiliation(s)
- Grant S Schulert
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Kejian Zhang
- Sema4 and Department of Genetic and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
39
|
Edwards KM. The Therapy of SARS-CoV-2 Infection in Children. J Clin Med 2023; 13:120. [PMID: 38202127 PMCID: PMC10779459 DOI: 10.3390/jcm13010120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/11/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
The impact of SARS-CoV-2 infections in children has fortunately been lower than what has been seen in adults. However, even previously healthy children have developed severe disease, sometimes with subsequent mortality, and those who are infants or adolescents, are from racial and ethnic minority groups, or have certain chronic conditions are at higher risk of these outcomes. During the pandemic, extensive studies of therapeutic agents, including antivirals and immunomodulators, were conducted in adults. Few trials included children, and most were in older children and adolescents. Thus, the potential benefits of therapies in children must be extrapolated from adult evidence. Despite these limitations, advisory committees of the National Institute of Health (NIH), the Infectious Disease Society of America (IDSA), and the Pediatric Infectious Diseases Society (PIDS) were constituted, and expert consensus guidelines were developed. This review provides a synthesis of those comprehensive recommendations for therapy in children. These address treatment during the early infectious period with antiviral agents, including remdesivir and nirmatrelvir/ritonavir, as well as treatment in the later period of immune dysregulation with corticosteroids and immunomodulators. In addition, the therapeutic approach for multisystem inflammatory syndrome in children (MIS-C), also referred to as Pediatric Inflammatory Multisystem Syndrome temporally associated with SARS-CoV-2 (PIMS-TS), is also provided.
Collapse
Affiliation(s)
- Kathryn M Edwards
- Division of Infectious Diseases, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| |
Collapse
|
40
|
Li S, Zhang H, Shang G. Current status and future challenges of CAR-T cell therapy for osteosarcoma. Front Immunol 2023; 14:1290762. [PMID: 38187386 PMCID: PMC10766856 DOI: 10.3389/fimmu.2023.1290762] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/07/2023] [Indexed: 01/09/2024] Open
Abstract
Osteosarcoma, the most common bone malignancy in children and adolescents, poses considerable challenges in terms of prognosis, especially for patients with metastatic or recurrent disease. While surgical intervention and adjuvant chemotherapy have improved survival rates, limitations such as impractical tumor removal or chemotherapy resistance hinder the treatment outcomes. Chimeric antigen receptor (CAR)-T cell therapy, an innovative immunotherapy approach that involves targeting tumor antigens and releasing immune factors, has shown significant advancements in the treatment of hematological malignancies. However, its application in solid tumors, including osteosarcoma, is constrained by factors such as low antigen specificity, limited persistence, and the complex tumor microenvironment. Research on osteosarcoma is ongoing, and some targets have shown promising results in pre-clinical studies. This review summarizes the current status of research on CAR-T cell therapy for osteosarcoma by compiling recent literature. It also proposes future research directions to enhance the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Shizhe Li
- Department of Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Orthopaedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - He Zhang
- Department of Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Guanning Shang
- Department of Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
41
|
Davoutis E, Panou C, Stachika N, Dalla C, Kokras N. Drug-drug interactions between COVID-19 drug therapies and antidepressants. Expert Opin Drug Metab Toxicol 2023; 19:937-950. [PMID: 37934891 DOI: 10.1080/17425255.2023.2280750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
INTRODUCTION Antidepressants are widely used for the pharmacological treatment of anxiety and mood disorders. Since the eruption of the SARS-COV-2 pandemic and the later development of targeted treatments against COVID-19, inevitably many patients receive antidepressants as well as targeted treatments against COVID-19 against COVID-19. Co-administration of antidepressants with COVID-19 therapeutics has the potential of drug-drug interactions, of varying severity and clinical significance. AREAS COVERED This is a curated narrative review of the current state of the art regarding drug-drug interactions between COVID-19 therapeutics and medications licensed for the pharmacotherapy of depression. A systematic search of electronic databases, using as keywords the international nonproprietaty names of currently approved COVID-19 therapeutics and antidepressants was performed, and additionally online interaction checker tools were consulted. Derived data were synthesized for each COVID-19 therapeutic and presented with up-to-date guidance. EXPERT OPINION Several COVID-19 therapeutics have potential for drug-drug interactions with antidepressants. Remdesivir and Nirmatrelvir-Ritonavir have the higher risk, whereas several monoclonal antibodies appear safer. The most serious drug-drug interactions (serotonin syndrome and QTc prolongation) require close monitoring; however, DDI toward reducing the efficacy of antidepressants may be difficult to recognize. As COVID-19 treatment protocols take precedence, psychiatrists should exert flexibility in antidepressant use and proactively monitor treatment progress.
Collapse
Affiliation(s)
- Efstathia Davoutis
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Chrysa Panou
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolina Stachika
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
42
|
Martinez-King C, Chung SH, McCartney SA. Adult-Onset Still's Disease in Pregnancy: Lessons Learned and an Approach to Subsequent Pregnancies. Reprod Sci 2023; 30:3515-3519. [PMID: 37464203 DOI: 10.1007/s43032-023-01296-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 07/09/2023] [Indexed: 07/20/2023]
Abstract
Adult-onset Still's disease (AOSD) is a rare autoinflammatory disorder with potential for life-threatening complications in pregnancy. Recently, biologic therapeutics have been increasingly used for treatment of AOSD, but there is little available data on the treatment of AOSD in pregnancy. Here we report a 23-year-old primigravid patient with a history of AOSD who presented at 20 weeks of gestation with fever, arthralgias, rash, fatigue, and highly elevated ferritin, concerning for AOSD flare. She was treated with tocilizumab, an interleukin-6 receptor antagonist, with rapid clinical and laboratory improvement; however, she underwent iatrogenic preterm delivery at 34 weeks of gestation for fetal distress, which was attributed to placental injury. In a subsequent pregnancy, she was treated with tocilizumab throughout and had an uncomplicated term delivery with normal labs and no AOSD flare. This case highlights that the use of tocilizumab may be effective to reduce the risk of AOSD flare during pregnancy.
Collapse
Affiliation(s)
- Carolina Martinez-King
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, University of Washington, 1959 NE Pacific St, Box 356460, Seattle, WA, 98195, USA
| | - Sarah H Chung
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Stephen A McCartney
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, University of Washington, 1959 NE Pacific St, Box 356460, Seattle, WA, 98195, USA.
| |
Collapse
|
43
|
Li T, Wang D, Wei H, Xu X. Cytokine storm and translating IL-6 biology into effective treatments for COVID-19. Front Med 2023; 17:1080-1095. [PMID: 38157195 DOI: 10.1007/s11684-023-1044-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/23/2023] [Indexed: 01/03/2024]
Abstract
As of May 3, 2023, the Coronavirus disease 2019 (COVID-19) pandemic has resulted in more than 760 million confirmed cases and over 6.9 million deaths. Several patients have developed pneumonia, which can deteriorate into acute respiratory distress syndrome. The primary etiology may be attributed to cytokine storm, which is triggered by the excessive release of proinflammatory cytokines and subsequently leads to immune dysregulation. Considering that high levels of interleukin-6 (IL-6) have been detected in several highly pathogenic coronavirus-infected diseases, such as severe acute respiratory syndrome in 2002, the Middle East respiratory syndrome in 2012, and COVID-19, the IL-6 pathway has emerged as a key in the pathogenesis of this hyperinflammatory state. Thus, we review the history of cytokine storm and the process of targeting IL-6 signaling to elucidate the pivotal role played by tocilizumab in combating COVID-19.
Collapse
Affiliation(s)
- Tiantian Li
- Department of Geriatric Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Dongsheng Wang
- Respiratory and Critical Care Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Haiming Wei
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Science and Medical Center, University of Science and Technology of China, Hefei, 230001, China
- Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230001, China
| | - Xiaoling Xu
- Respiratory and Critical Care Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| |
Collapse
|
44
|
Cao Y, Efetov SK, He M, Fu Y, Beeraka NM, Zhang J, Zhang X, Bannimath N, Chen K. Updated Clinical Perspectives and Challenges of Chimeric Antigen Receptor-T Cell Therapy in Colorectal Cancer and Invasive Breast Cancer. Arch Immunol Ther Exp (Warsz) 2023; 71:19. [DOI: https:/doi.org/10.1007/s00005-023-00684-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 06/28/2023] [Indexed: 09/20/2024]
|
45
|
Ekpa QL, Akahara PC, Anderson AM, Adekoya OO, Ajayi OO, Alabi PO, Okobi OE, Jaiyeola O, Ekanem MS. A Review of Acute Lymphocytic Leukemia (ALL) in the Pediatric Population: Evaluating Current Trends and Changes in Guidelines in the Past Decade. Cureus 2023; 15:e49930. [PMID: 38179374 PMCID: PMC10766210 DOI: 10.7759/cureus.49930] [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] [Accepted: 12/04/2023] [Indexed: 01/06/2024] Open
Abstract
Acute lymphocytic leukemia (ALL) is a commonly diagnosed cancer in children. Despite technological advancements to improve treatment and survival rates, there has been a steady increase in the incidence of ALL and treatment failures. This paper discusses the pathogenic interaction between genetic and environmental factors leading to childhood ALL. It evaluates the current treatment guidelines and notable obstacles leading to resistance, relapse, and treatment toxicities. The review evaluates a 10-year trend in the management guidelines of pediatric ALL through a systematic literature review of records from 2012 to 2023. Findings show that improvement in the five-year survival rates, notwithstanding rates of relapse and incurable diseases, is still high. Furthermore, several risk factors, including an interplay between genetic and environmental factors, are largely contributory to the outcome of ALL treatments and its overall incidence. Moreover, huge financial costs have remained a significant challenge in outcomes. There remains a need to provide individualized treatment plans, shared decision-making, and goals of care as parts of the management guidelines for the best possible outcomes. We expect that future advancements will increase overall survival rates and disease-free years.
Collapse
Affiliation(s)
- Queen L Ekpa
- General Practice, Conestoga College, Kitchener, CAN
| | | | - Alexis M Anderson
- Pediatric Medicine, St. George's University, School of Medicine, St. George's, GRD
| | | | - Olamide O Ajayi
- Pediatrics, Medway Maritime Hospital, Kent, GBR
- Internal Medicine, Obafemi Awolowo College of Health Sciences, Olabisi Onabanjo University, Sagamu, NGA
| | - Peace O Alabi
- Pediatrics, University of Abuja Teaching Hospital, Abuja, NGA
| | - Okelue E Okobi
- Family Medicine, Larkin Community Hospital Palm Springs Campus, Hialeah, USA
- Family Medicine, Medficient Health Systems, Laurel, USA
- Family Medicine, Lakeside Medical Center, Belle Glade, USA
| | | | - Medara S Ekanem
- General Medicine, Babcock University Teaching Hospital, Ilishan-Remo, NGA
| |
Collapse
|
46
|
Cuvelier GDE, Paulson K, Bow EJ. Updates in hematopoietic cell transplant and cellular therapies that enhance the risk for opportunistic infections. Transpl Infect Dis 2023; 25 Suppl 1:e14101. [PMID: 37461887 DOI: 10.1111/tid.14101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/21/2023] [Accepted: 06/28/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Infectious disease physicians may be asked to evaluate and manage a variety of infections in immunocompromised hosts undergoing hematopoietic cell transplant (HCT) and cellular therapies. Over the last decade, several advances in cellular therapy have occurred, with implications for the types of infectious complications that may be seen. AIMS The purpose of this review is to update the infectious disease physician on newer advances in HCT and cellular therapy, including haploidentical transplant, expanding indications for transplant in older individuals and children, and chimeric antigen receptor T-cells. We will review how these advances might influence infectious disease complications following HCT. We will also provide a perspective that infectious disease physicians can use to evaluate the degree of immune suppression in an individual patient to help determine the type of infections that may be encountered.
Collapse
Affiliation(s)
- Geoffrey D E Cuvelier
- Department of Paediatrics and Child Health, Section of Paediatric Haematology/Oncology-BMT, Max Rady College of Medicine, the University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Paediatric Haematology/Oncology-BMT, CancerCare Manitoba, Winnipeg, Manitoba, Canada
- Manitoba Blood and Marrow Transplant Programme, CancerCare Manitoba, Winnipeg, Manitoba, Canada
| | - Kristjan Paulson
- Manitoba Blood and Marrow Transplant Programme, CancerCare Manitoba, Winnipeg, Manitoba, Canada
- Section of Haematology/Oncology, Department of Internal Medicine, Max Rady College of Medicine, the University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Oncology and Haematology, CancerCare Manitoba, Winnipeg, Manitoba, Winnipeg, Manitoba, Canada
| | - Eric J Bow
- Manitoba Blood and Marrow Transplant Programme, CancerCare Manitoba, Winnipeg, Manitoba, Canada
- Section of Haematology/Oncology, Department of Internal Medicine, Max Rady College of Medicine, the University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Oncology and Haematology, CancerCare Manitoba, Winnipeg, Manitoba, Winnipeg, Manitoba, Canada
- Section of Infectious Diseases, Department of Internal Medicine, Max Rady College of Medicine, The University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
47
|
Yang C, Nguyen J, Yen Y. Complete spectrum of adverse events associated with chimeric antigen receptor (CAR)-T cell therapies. J Biomed Sci 2023; 30:89. [PMID: 37864230 PMCID: PMC10590030 DOI: 10.1186/s12929-023-00982-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 10/12/2023] [Indexed: 10/22/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapies have been approved by FDA to treat relapsed or refractory hematological malignancies. However, the adverse effects of CAR-T cell therapies are complex and can be challenging to diagnose and treat. In this review, we summarize the major adverse events, including cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and CAR T-cell associated HLH (carHLH), and discuss their pathophysiology, symptoms, grading, and diagnosis systems, as well as management. In a future outlook, we also provide an overview of measures and modifications to CAR-T cells that are currently being explored to limit toxicity.
Collapse
Affiliation(s)
- Chieh Yang
- Department of Internal Medicine, School of Medicine, University of California Riverside, Riverside, CA USA
| | - John Nguyen
- Covina Discovery Center, Theragent Inc., Covina, CA USA
| | - Yun Yen
- College of Medical Technology, Taipei Medical University, Taipei City, Taiwan
| |
Collapse
|
48
|
Amicucci M, Simioli V, De Cecco V, Orlando L, Ciaralli I, Buccino A, Guidi B, Locatelli G, Palmieri C, Piazzalunga M, Proietti R, Pucci A, Botti S. Nursing Management in Pediatric Patients Undergoing Chimeric Antigen Receptor T (CAR-T) Cell Therapy: A Systematic Literature Review. Semin Oncol Nurs 2023; 39:151478. [PMID: 37544775 DOI: 10.1016/j.soncn.2023.151478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 06/08/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023]
Abstract
OBJECTIVES This systematic review aims to describe an overview of the overall care, patient and parent education, staff training, and management of complications from a nursing perspective of pediatric patients undergoing chimeric antigen receptor T (CAR-T) cell infusion in order to provide an updated summary of the approach to the management of these patients. CAR-T cellular therapy represents an innovation within pediatric hematology and oncology used to treat relapse and refractory leukemias, solid tumors, and lymphomas when standard therapy has not worked. However, this type of therapy could lead to the onset of some clinical complications that must be managed appropriately and promptly. Although their use is constantly increasing, the knowledge and resources in the literature are still limited. DATA SOURCES The review was conducted from January 2022 to July 2022 in PubMed, CINAHL, Scopus, and Cochrane and produced 502 articles. Based on the selection criteria and after removing duplicate articles, 26 articles were included in the study. CONCLUSION From these analyzed articles, it was possible to have an overview regarding the management, patient and parent education, staff training, and management of complications from a nursing perspective of pediatric patients undergoing CAR-T cell infusion. IMPLICATIONS FOR NURSING PRACTICE The management of hematology-oncology patients undergoing CAR-T cell therapy from a nursing perspective is not simple. We hope this review can be used as a tool to guide nursing staff. In this regard, we have developed a summary table with the actions to be taken in the case of assisting a pediatric patient being treated with CAR-T.
Collapse
Affiliation(s)
- Matteo Amicucci
- Department of Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS; and Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy.
| | - Valentina Simioli
- Department of Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Valentina De Cecco
- Department of Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Laura Orlando
- BMT Unit Coordinator and Quality Manager, Department of Nursing, Oncology Institute of Southern Switzerland (IOSI), EOC, Bellinzona, Switzerland
| | - Italo Ciaralli
- Department of Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Barbara Guidi
- Department of Pediatric Hematology Oncology - Cell and Gene Therapy, Meyer Children's Hospital, IRCCS, Florence, Italy
| | - Giovanna Locatelli
- Pediatric Hemato-Oncology, Fondazione IRCSS San Gerardo dei Tintori-Monza, Monza, Italy
| | - Claudia Palmieri
- Department of Pediatric Hematology Oncology - Cell and Gene Therapy, Meyer Children's Hospital, IRCCS, Florence, Italy
| | - Martina Piazzalunga
- Pediatric Hemato-Oncology, Fondazione IRCSS San Gerardo dei Tintori-Monza, Monza, Italy
| | - Romina Proietti
- Department of Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Agnese Pucci
- Pediatric Hematology and Oncology Stem Cell Transplantation and Cellular Therapy Division of the Regina Margherita Children's Hospital, A.O.U. Città della Salute e della Scienza, Turin, Italy
| | - Stefano Botti
- Hematology Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
49
|
Milošević I, Barać A, Jovanović J, Vujović A, Stevanović G, Todorović N, Milošević B. A single-center experience of early administration of tocilizumab and corticosteroids in patients with COVID-19 pneumonia. Trans R Soc Trop Med Hyg 2023; 117:668-672. [PMID: 37103332 DOI: 10.1093/trstmh/trad026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 02/16/2023] [Accepted: 04/12/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND We investigated the therapeutic response of tocilizumab (TCZ) therapy in patients with coronavirus disease 2019 (COVID-19) pneumonia. METHODS This observational retrospective study included 205 patients with confirmed COVID-19 pneumonia with SpO2˂93% and a markedly increased level of at least two biomarkers of inflammation. The TCZ was given in combination with corticosteroids. Clinical and laboratory results were analyzed and compared before TCZ therapy and 7 d after. RESULTS The mean value of C-reactive protein (CRP) was significantly lower (p=0.001) on the seventh day after administration of TCZ compared with before (10.7 and 173.6 mg/L, respectively). Only in 9/205 (4.3%) patients, the CRP level did not decrease during the week-long period, and this was related to disease progression. The mean level of interleukin-6 before TCZ administration was 88±113 pg/mL, while after it was 32.7±21.7 pg/mL (p=0.01). After 7 d of TCZ therapy, almost 50% of patients who needed high-flow oxygen or ventilation support started to receive low-flow oxygen, while 73/205 (35.6%) patients who received low-flow oxygen before TCZ administration did not receive further oxygen support anymore (p=0.001). Although they received TCZ treatment, 38/205 (18.5%) severely sick patients died. CONCLUSIONS Tocilizumab improves clinical outcomes in hospitalized COVID-19 patients. These advantages were evident independent of the patient's comorbidities and were in addition to the advantages of systemic corticosteroids. In COVID-19 patients at risk of cytokine storms, TCZ appears to be an effective therapy choice.
Collapse
Affiliation(s)
- Ivana Milošević
- Clinic for Infectious and Tropical Diseases, University Clinical Center of Serbia, Bul. Oslobodjenja 16, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotica 1, 11000 Belgrade , Serbia
| | - Aleksandra Barać
- Clinic for Infectious and Tropical Diseases, University Clinical Center of Serbia, Bul. Oslobodjenja 16, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotica 1, 11000 Belgrade , Serbia
| | - Jaroslava Jovanović
- Clinic for Infectious and Tropical Diseases, University Clinical Center of Serbia, Bul. Oslobodjenja 16, 11000 Belgrade, Serbia
| | - Ankica Vujović
- Clinic for Infectious and Tropical Diseases, University Clinical Center of Serbia, Bul. Oslobodjenja 16, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotica 1, 11000 Belgrade , Serbia
| | - Goran Stevanović
- Clinic for Infectious and Tropical Diseases, University Clinical Center of Serbia, Bul. Oslobodjenja 16, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotica 1, 11000 Belgrade , Serbia
| | - Nevena Todorović
- Clinic for Infectious and Tropical Diseases, University Clinical Center of Serbia, Bul. Oslobodjenja 16, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotica 1, 11000 Belgrade , Serbia
| | - Branko Milošević
- Clinic for Infectious and Tropical Diseases, University Clinical Center of Serbia, Bul. Oslobodjenja 16, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotica 1, 11000 Belgrade , Serbia
| |
Collapse
|
50
|
Dharra R, Kumar Sharma A, Datta S. Emerging aspects of cytokine storm in COVID-19: The role of proinflammatory cytokines and therapeutic prospects. Cytokine 2023; 169:156287. [PMID: 37402337 PMCID: PMC10291296 DOI: 10.1016/j.cyto.2023.156287] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 06/24/2023] [Indexed: 07/06/2023]
Abstract
COVID-19 has claimed millions of lives during the last 3 years since initial cases were reported in Wuhan, China, in 2019. Patients with COVID-19 suffer from severe pneumonia, high fever, acute respiratory distress syndrome (ARDS), and multiple-organ dysfunction, which may also result in fatality in extreme cases. Cytokine storm (CS) is hyperactivation of the immune system, wherein the dysregulated production of proinflammatory cytokines could result in excessive immune cell infiltrations in the pulmonary tissues, resulting in tissue damage. The immune cell infiltration could also occur in other tissues and organs and result in multiple organs' dysfunction. The key cytokines implicated in the onset of disease severity include TNF-α, IFN-γ, IL-6, IL-1β, GM-CSF, and G-CSF. Controlling the CS is critical in treating COVID-19 disease. Therefore, different strategies are employed to mitigate the effects of CS. These include using monoclonal antibodies directed against soluble cytokines or the cytokine receptors, combination therapies, mesenchymal stem cell therapy, therapeutic plasma exchange, and some non-conventional treatment methods to improve patient immunity. The current review describes the role/s of critical cytokines in COVID-19-mediated CS and the respective treatment modalities.
Collapse
Affiliation(s)
- Renu Dharra
- CSIR-Institute of Microbial Technology, Sector 39 A, Chandigarh 160036, India
| | - Anil Kumar Sharma
- Department of Bio-Science and Technology, M. M. Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India
| | - Sonal Datta
- Department of Bio-Science and Technology, M. M. Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India.
| |
Collapse
|