1
|
Gandhi M, Sharma B, Nair S, Vaidya ADB. Current Insights into CAR T-Cell-Based Therapies for Myelodysplastic Syndrome. Pharm Res 2024; 41:1757-1773. [PMID: 39187686 DOI: 10.1007/s11095-024-03761-8] [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: 03/30/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024]
Abstract
Myelodysplastic syndromes (MDS) are due to defective hematopoiesis in bone marrow characterized by cytopenia and dysplasia of blood cells, with a varying degree of risk of acute myeloid leukemia (AML). Currently, the only potentially curative strategy is hematopoietic stem cell transplantation (HSCT). Many patients are ineligible for HSCT, due to late diagnosis, presence of co-morbidities, old age and complications likely due to graft-versus-host disease (GvHD). As a consequence, patients with MDS are often treated conservatively with blood transfusions, chemotherapy, immunotherapy etc. based on the grade and manifestations of MDS. The development of chimeric antigen receptor (CAR)-T cell therapy has revolutionized immunotherapy for hematological malignancies, as evidenced by a large body of literature. However, resistance and toxicity associated with it are also a challenge. Hence, there is an urgent need to develop new strategies for immunological and hematopoetic management of MDS. Herein, we discuss current limitations of CAR T-cell therapy and summarize novel approaches to mitigate this. Further, we discuss the in vivo activation of tumor-specific T cells, immune check inhibitors (ICI) and other approaches to normalize the bone marrow milieu for the management of MDS.
Collapse
Affiliation(s)
- Manav Gandhi
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Bhirisha Sharma
- University of Mumbai, Santa Cruz (East), Mumbai, 400055, India
| | - Sujit Nair
- Viridis Biopharma Pvt. Ltd, Mumbai, 400022, India.
- Phytoveda Pvt. Ltd, Mumbai, 400022, India.
| | - Ashok D B Vaidya
- Kasturba Health Society-Medical Research Centre, Vile Parle (West), Mumbai, 400056, India
| |
Collapse
|
2
|
Ozma MA, Moaddab SR, Hosseini H, Khodadadi E, Ghotaslou R, Asgharzadeh M, Abbasi A, Kamounah FS, Aghebati Maleki L, Ganbarov K, Samadi Kafil H. A critical review of novel antibiotic resistance prevention approaches with a focus on postbiotics. Crit Rev Food Sci Nutr 2024; 64:9637-9655. [PMID: 37203933 DOI: 10.1080/10408398.2023.2214818] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Antibiotic resistance is a significant public health issue, causing illnesses that were once easily treatable with antibiotics to develop into dangerous infections, leading to substantial disability and even death. To help fight this growing threat, scientists are developing new methods and techniques that play a crucial role in treating infections and preventing the inappropriate use of antibiotics. These effective therapeutic methods include phage therapies, quorum-sensing inhibitors, immunotherapeutics, predatory bacteria, antimicrobial adjuvants, haemofiltration, nanoantibiotics, microbiota transplantation, plant-derived antimicrobials, RNA therapy, vaccine development, and probiotics. As a result of the activity of probiotics in the intestine, compounds derived from the structure and metabolism of these bacteria are obtained, called postbiotics, which include multiple agents with various therapeutic applications, especially antimicrobial effects, by using different mechanisms. These compounds have been chosen in particular because they don't promote the spread of antibiotic resistance and don't include substances that can increase antibiotic resistance. This manuscript provides an overview of the novel approaches to preventing antibiotic resistance with emphasis on the various postbiotic metabolites derived from the gut beneficial microbes, their activities, recent related progressions in the food and medical fields, as well as concisely giving an insight into the new concept of postbiotics as "hyperpostbiotic".
Collapse
Affiliation(s)
- Mahdi Asghari Ozma
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyyed Reza Moaddab
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hedayat Hosseini
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ehsaneh Khodadadi
- Material Science and Engineering, Department of Chemistry and Biochemistry, University of Arkansas-Fayetteville, Fayetteville, AR, USA
| | - Reza Ghotaslou
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Asgharzadeh
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Abbasi
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fadhil S Kamounah
- Department of Chemistry, University of Copenhagen, Copenhagen, Denmark
| | | | - Khudaverdi Ganbarov
- Research Laboratory of Microbiology and Virology, Baku State University, Baku, Republic of Azerbaijan
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Tardif M, Usmani N, Krajinovic M, Bittencourt H. Cytokine release syndrome after CAR T-cell therapy for B-cell acute lymphoblastic leukemia in children and young adolescents: storms make trees take deeper roots. Expert Opin Pharmacother 2024; 25:1497-1506. [PMID: 39087712 DOI: 10.1080/14656566.2024.2387673] [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: 05/26/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/02/2024]
Abstract
INTRODUCTION Chimeric antigen receptor (CAR) T-cells have revolutionized cancer treatment, showing significant success, including treatment of pediatric B-cell acute lymphoblastic leukemia (B-ALL). Despite their efficacy, cytokine release syndrome (CRS) emerges as a common early adverse effect that can be life threatening in severe cases, resulting from the immune system's targeted activation against tumors. AREAS COVERED This review concentrates on CRS in children and young adults undergoing CAR T-cell therapy for B-ALL. It explores CRS pathophysiology, clinical presentation, and incidence, emphasizing the importance of a consensus definition and grading to homogenize the treatment according to the severity of symptoms. We will discuss the standard of care treatment of CRS but also novel approaches. We will highlight the importance of managing CRS without compromising the efficacy of immune cell activation against tumors. EXPERT OPINION As CAR T-cell therapy in pediatric B-ALL become increasingly available and used, optimal management of CRS becomes increasingly important. Early recognition and timely management has improved. Further information will aid us to identify optimal timing of tocilizumab and corticosteroids. Continued bench research coupled with clinical studies and biomarker discovery will allow for valuable insights into CRS pathophysiology and patient and/or cell-targeted treatments.
Collapse
Affiliation(s)
- Magalie Tardif
- Hematology/Oncology Division, CHU Sainte-Justine, Montreal, Quebec, Canada
| | - Nida Usmani
- Hematology/Oncology Division, CHU Sainte-Justine, Montreal, Quebec, Canada
| | - Maja Krajinovic
- Hematology/Oncology Division, CHU Sainte-Justine, Montreal, Quebec, Canada
- Research Center - CHU Sainte-Justine, Montreal, Quebec, Canada
- School of Medicine, Université de Montreal, Montreal, Quebec, Canada
| | - Henrique Bittencourt
- Hematology/Oncology Division, CHU Sainte-Justine, Montreal, Quebec, Canada
- Research Center - CHU Sainte-Justine, Montreal, Quebec, Canada
- School of Medicine, Université de Montreal, Montreal, Quebec, Canada
| |
Collapse
|
4
|
Quílez Trasobares N, González-Fernández M, Barea-Mendoza JA, Arias-Verdú MD, Barrueco-Francioni JE, Seller-Pérez G, Molina-Collado Z, Lesmes González-de Aledo A, Herrera-Gutiérrez M, Sánchez-Izquierdo Riera JÁ. The Role of Immunomodulatory Therapy with Oxiris in COVID-19 with Renal Failure and Immune Dysfunction. Blood Purif 2024; 53:804-812. [PMID: 38991521 DOI: 10.1159/000539833] [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: 09/15/2023] [Accepted: 06/10/2024] [Indexed: 07/13/2024]
Abstract
INTRODUCTION The main objective of this study was to evaluate the impact of hemoadsorption on the elimination of inflammatory mediators. METHODS A prospective, bicenter, observational cohort study was conducted between March 2020 and February 2022 to explore the immunomodulatory response, demographic and clinical characteristics of individuals with COVID-19 admitted to the ICU with severe acute respiratory failure and in need of CRRT with Oxiris® with or without AKI. RESULTS Sixty-four patients were analyzed. Statistically significant differences were observed between exposed and unexposed groups, in relation to the reduction in D-dimer levels -15,614 (24,848.9) versus -4,136.5 (9,913.47) (p 0.031, d: 1.59, 95% CI: -21,830, -1,126). An increase in PCT was observed 0.47 (2.08) versus -0.75 (2.3) (p 0.044 95% CI: 0.03, 2.44). No differences were found in a decrease in CRP -4.21 (7.29) versus -1.6 (9.02) (p 0.22) nor in the rest of inflammatory parameters fibrinogen, IL-6, ferritin, lymphocytes, and neutrophils. Subgroup analysis in patients exposed to therapy also showed a significant decrease in D-dimer of 55% from baseline: 6,000 (1,984.5-27,750) pre-therapy versus 2,700 (2,119.5-6,145) (95% CI: -23,000, -2,489) post-therapy with a strong effect size (p 0.001, d: 0.65). CONCLUSION The hemoadsorptive therapy in COVID-19 was associated with a significant decrease in D-dimer parameters without showing decreases in the rest of the clinical, inflammatory parameters and severity scales analyzed.
Collapse
|
5
|
Wang Y, Dong H, Dong T, Zhao L, Fan W, Zhang Y, Yao W. Treatment of cytokine release syndrome-induced vascular endothelial injury using mesenchymal stem cells. Mol Cell Biochem 2024; 479:1149-1164. [PMID: 37392343 DOI: 10.1007/s11010-023-04785-1] [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: 04/13/2023] [Accepted: 06/04/2023] [Indexed: 07/03/2023]
Abstract
Cytokine release syndrome (CRS) is an acute systemic inflammatory reaction in which hyperactivated immune cells suddenly release a large amount of cytokines, leading to exaggerated inflammatory responses, multiple organ dysfunction, and even death. Although palliative treatment strategies have significantly reduced the overall mortality, novel targeted treatment regimens with superior therapy efficacy are urgently needed. Vascular endothelial cells (ECs) are important target cells of systemic inflammation, and their destruction is considered to be the initiating event underlying many serious complications of CRS. Mesenchymal stem/stromal cells (MSCs) are multipotent cells with self-renewing differentiation capacity and immunomodulatory properties. MSC transplantation can effectively suppress the activation of immune cells, reduce the bulk release of cytokines, and repair damaged tissues and organs. Here, we review the molecular mechanisms underlying CRS-induced vascular endothelial injury and discuss potential treatments using MSCs. Preclinical studies demonstrate that MSC therapy can effectively repair endothelium damage and thus reduce the incidence and severity of ensuing CRS-induced complications. This review highlights the therapeutic role of MSCs in fighting against CRS-induced EC damage, and summarizes the possible therapeutic formulations of MSCs for improved efficacy in future clinical trials.
Collapse
Affiliation(s)
- Yuyan Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Yangtze University, Jingzhou, China
- Health Science Center, Yangtze University, Jingzhou, China
| | - Haibo Dong
- Wuhan Optics Valley Vcanbiopharma Co., Ltd, Wuhan, China
- Key Industrial Base for Stem Cell Engineering Products, Tianjin, China
| | - Tengyun Dong
- Wuhan Optics Valley Vcanbiopharma Co., Ltd, Wuhan, China
- Key Industrial Base for Stem Cell Engineering Products, Tianjin, China
| | - Lulu Zhao
- Wuhan Optics Valley Vcanbiopharma Co., Ltd, Wuhan, China
- Key Industrial Base for Stem Cell Engineering Products, Tianjin, China
| | - Wen Fan
- Department of Laboratory Medicine, The First Affiliated Hospital of Yangtze University, Jingzhou, China.
| | - Yu Zhang
- Wuhan Optics Valley Vcanbiopharma Co., Ltd, Wuhan, China.
- Key Industrial Base for Stem Cell Engineering Products, Tianjin, China.
- Haihe Laboratory of Cell Ecosystem, Tianjin, China.
| | - Weiqi Yao
- Wuhan Optics Valley Vcanbiopharma Co., Ltd, Wuhan, China.
- Key Industrial Base for Stem Cell Engineering Products, Tianjin, China.
- Department of Biology and Medicine, Hubei University of Technology, Wuhan, China.
| |
Collapse
|
6
|
Walton ZE, Frigault MJ, Maus MV. Current and emerging pharmacotherapies for cytokine release syndrome, neurotoxicity, and hemophagocytic lymphohistiocytosis-like syndrome due to CAR T cell therapy. Expert Opin Pharmacother 2024; 25:263-279. [PMID: 38588525 DOI: 10.1080/14656566.2024.2340738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/01/2024] [Indexed: 04/10/2024]
Abstract
INTRODUCTION Chimeric antigen receptor (CAR) T cells have revolutionized the treatment of multiple hematologic malignancies. Engineered cellular therapies now offer similar hope to transform the management of solid tumors and autoimmune diseases. However, toxicities can be serious and often require hospitalization. AREAS COVERED We review the two chief toxicities of CAR T therapy, cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), and the rarer immune effector cell-associated hemophagocytic lymphohistiocytosis-like syndrome. We discuss treatment paradigms and promising future pharmacologic strategies. Literature and therapies reviewed were identified by PubMed search, cited references therein, and review of registered trials. EXPERT OPINION Management of CRS and ICANS has improved, aided by consensus definitions and guidelines that facilitate recognition and timely intervention. Further data will define optimal timing of tocilizumab and corticosteroids, current foundations of management. Pathophysiologic understanding has inspired off-label use of IL-1 receptor antagonism, IFNγ and IL-6 neutralizing antibodies, and janus kinase inhibitors, with data emerging from ongoing clinical trials. Further strategies to reduce toxicities include novel pharmacologic targets and safety features engineered into CAR T cells themselves. As these potentially curative therapies are used earlier in oncologic therapy and even in non-oncologic indications, effective accessible strategies to manage toxicities are critical.
Collapse
Affiliation(s)
- Zandra E Walton
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Division of Rheumatology, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Matthew J Frigault
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
McNerney KO, Hsieh EM, Shalabi H, Epperly R, Wolters PL, Hill JA, Gardner R, Talleur AC, Shah NN, Rossoff J. INSPIRED Symposium Part 3: Prevention and Management of Pediatric Chimeric Antigen Receptor T Cell-Associated Emergent Toxicities. Transplant Cell Ther 2024; 30:38-55. [PMID: 37821079 PMCID: PMC10842156 DOI: 10.1016/j.jtct.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023]
Abstract
Chimeric antigen receptor (CAR) T cell (CAR-T) therapy has emerged as a revolutionary cancer treatment modality, particularly in children and young adults with B cell malignancies. Through clinical trials and real-world experience, much has been learned about the unique toxicity profile of CAR-T therapy. The past decade brought advances in identifying risk factors for severe inflammatory toxicities, investigating preventive measures to mitigate these toxicities, and exploring novel strategies to manage refractory and newly described toxicities, infectious risks, and delayed effects, such as cytopenias. Although much progress has been made, areas needing further improvements remain. Limited guidance exists regarding initial administration of tocilizumab with or without steroids and the management of inflammatory toxicities refractory to these treatments. There has not been widespread adoption of preventive strategies to mitigate inflammation in patients at high risk of severe toxicities, particularly children. Additionally, the majority of research related to CAR-T toxicity prevention and management has focused on adult populations, with only a few pediatric-specific studies published to date. Given that children and young adults undergoing CAR-T therapy represent a unique population with different underlying disease processes, physiology, and tolerance of toxicities than adults, it is important that studies be conducted to evaluate acute, delayed, and long-term toxicities following CAR-T therapy in this younger age group. In this pediatric-focused review, we summarize key findings on CAR-T therapy-related toxicities over the past decade, highlight emergent CAR-T toxicities, and identify areas of greatest need for ongoing research.
Collapse
Affiliation(s)
- Kevin O McNerney
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.
| | - Emily M Hsieh
- Pediatric Hematology/Oncology, Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, California
| | - Haneen Shalabi
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Rebecca Epperly
- Department of Bone Marrow Transplant, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Pamela L Wolters
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Joshua A Hill
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Rebecca Gardner
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Aimee C Talleur
- Department of Bone Marrow Transplant, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jenna Rossoff
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| |
Collapse
|
8
|
K S, Vasanthrao R, Chattopadhyay I. Impact of environment on transmission of antibiotic-resistant superbugs in humans and strategies to lower dissemination of antibiotic resistance. Folia Microbiol (Praha) 2023; 68:657-675. [PMID: 37589876 DOI: 10.1007/s12223-023-01083-7] [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: 12/26/2022] [Accepted: 08/02/2023] [Indexed: 08/18/2023]
Abstract
Antibiotics are the most efficient type of therapy developed in the twentieth century. From the early 1960s to the present, the rate of discovery of new and therapeutically useful classes of antibiotics has significantly decreased. As a result of antibiotic use, novel strains emerge that limit the efficiency of therapies in patients, resulting in serious consequences such as morbidity or mortality, as well as clinical difficulties. Antibiotic resistance has created major concern and has a greater impact on global health. Horizontal and vertical gene transfers are two mechanisms involved in the spread of antibiotic resistance genes (ARGs) through environmental sources such as wastewater treatment plants, agriculture, soil, manure, and hospital-associated area discharges. Mobile genetic elements have an important part in microbe selection pressure and in spreading their genes into new microbial communities; additionally, it establishes a loop between the environment, animals, and humans. This review contains antibiotics and their resistance mechanisms, diffusion of ARGs, prevention of ARG transmission, tactics involved in microbiome identification, and therapies that aid to minimize infection, which are explored further below. The emergence of ARGs and antibiotic-resistant bacteria (ARB) is an unavoidable threat to global health. The discovery of novel antimicrobial agents derived from natural products shifts the focus from chemical modification of existing antibiotic chemical composition. In the future, metagenomic research could aid in the identification of antimicrobial resistance genes in the environment. Novel therapeutics may reduce infection and the transmission of ARGs.
Collapse
Affiliation(s)
- Suganya K
- Department of Biotechnology, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur, 610101, India
| | - Ramavath Vasanthrao
- Department of Biotechnology, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur, 610101, India
| | - Indranil Chattopadhyay
- Department of Biotechnology, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur, 610101, India.
| |
Collapse
|
9
|
Daei Sorkhabi A, Mohamed Khosroshahi L, Sarkesh A, Mardi A, Aghebati-Maleki A, Aghebati-Maleki L, Baradaran B. The current landscape of CAR T-cell therapy for solid tumors: Mechanisms, research progress, challenges, and counterstrategies. Front Immunol 2023; 14:1113882. [PMID: 37020537 PMCID: PMC10067596 DOI: 10.3389/fimmu.2023.1113882] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/28/2023] [Indexed: 04/07/2023] Open
Abstract
The successful outcomes of chimeric antigen receptor (CAR) T-cell therapy in treating hematologic cancers have increased the previously unprecedented excitement to use this innovative approach in treating various forms of human cancers. Although researchers have put a lot of work into maximizing the effectiveness of these cells in the context of solid tumors, few studies have discussed challenges and potential strategies to overcome them. Restricted trafficking and infiltration into the tumor site, hypoxic and immunosuppressive tumor microenvironment (TME), antigen escape and heterogeneity, CAR T-cell exhaustion, and severe life-threatening toxicities are a few of the major obstacles facing CAR T-cells. CAR designs will need to go beyond the traditional architectures in order to get over these limitations and broaden their applicability to a larger range of malignancies. To enhance the safety, effectiveness, and applicability of this treatment modality, researchers are addressing the present challenges with a wide variety of engineering strategies as well as integrating several therapeutic tactics. In this study, we reviewed the antigens that CAR T-cells have been clinically trained to recognize, as well as counterstrategies to overcome the limitations of CAR T-cell therapy, such as recent advances in CAR T-cell engineering and the use of several therapies in combination to optimize their clinical efficacy in solid tumors.
Collapse
Affiliation(s)
- Amin Daei Sorkhabi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Aila Sarkesh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Mardi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Aghebati-Maleki
- Stem Cell Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- *Correspondence: Leili Aghebati-Maleki, ; Behzad Baradaran,
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- *Correspondence: Leili Aghebati-Maleki, ; Behzad Baradaran,
| |
Collapse
|
10
|
Nasiri F, Kazemi M, Mirarefin SMJ, Mahboubi Kancha M, Ahmadi Najafabadi M, Salem F, Dashti Shokoohi S, Evazi Bakhshi S, Safarzadeh Kozani P, Safarzadeh Kozani P. CAR-T cell therapy in triple-negative breast cancer: Hunting the invisible devil. Front Immunol 2022; 13. [DOI: https:/doi.org/10.3389/fimmu.2022.1018786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is known as the most intricate and hard-to-treat subtype of breast cancer. TNBC cells do not express the well-known estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 (HER2) expressed by other breast cancer subtypes. This phenomenon leaves no room for novel treatment approaches including endocrine and HER2-specific antibody therapies. To date, surgery, radiotherapy, and systemic chemotherapy remain the principal therapy options for TNBC treatment. However, in numerous cases, these approaches either result in minimal clinical benefit or are nonfunctional, resulting in disease recurrence and poor prognosis. Nowadays, chimeric antigen receptor T cell (CAR-T) therapy is becoming more established as an option for the treatment of various types of hematologic malignancies. CAR-Ts are genetically engineered T lymphocytes that employ the body’s immune system mechanisms to selectively recognize cancer cells expressing tumor-associated antigens (TAAs) of interest and efficiently eliminate them. However, despite the clinical triumph of CAR-T therapy in hematologic neoplasms, CAR-T therapy of solid tumors, including TNBC, has been much more challenging. In this review, we will discuss the success of CAR-T therapy in hematological neoplasms and its caveats in solid tumors, and then we summarize the potential CAR-T targetable TAAs in TNBC studied in different investigational stages.
Collapse
|
11
|
Nasiri F, Kazemi M, Mirarefin SMJ, Mahboubi Kancha M, Ahmadi Najafabadi M, Salem F, Dashti Shokoohi S, Evazi Bakhshi S, Safarzadeh Kozani P, Safarzadeh Kozani P. CAR-T cell therapy in triple-negative breast cancer: Hunting the invisible devil. Front Immunol 2022; 13:1018786. [PMID: 36483567 PMCID: PMC9722775 DOI: 10.3389/fimmu.2022.1018786] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 10/24/2022] [Indexed: 11/23/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is known as the most intricate and hard-to-treat subtype of breast cancer. TNBC cells do not express the well-known estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 (HER2) expressed by other breast cancer subtypes. This phenomenon leaves no room for novel treatment approaches including endocrine and HER2-specific antibody therapies. To date, surgery, radiotherapy, and systemic chemotherapy remain the principal therapy options for TNBC treatment. However, in numerous cases, these approaches either result in minimal clinical benefit or are nonfunctional, resulting in disease recurrence and poor prognosis. Nowadays, chimeric antigen receptor T cell (CAR-T) therapy is becoming more established as an option for the treatment of various types of hematologic malignancies. CAR-Ts are genetically engineered T lymphocytes that employ the body's immune system mechanisms to selectively recognize cancer cells expressing tumor-associated antigens (TAAs) of interest and efficiently eliminate them. However, despite the clinical triumph of CAR-T therapy in hematologic neoplasms, CAR-T therapy of solid tumors, including TNBC, has been much more challenging. In this review, we will discuss the success of CAR-T therapy in hematological neoplasms and its caveats in solid tumors, and then we summarize the potential CAR-T targetable TAAs in TNBC studied in different investigational stages.
Collapse
Affiliation(s)
- Fatemeh Nasiri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Mehrasa Kazemi
- Department of Laboratory Medicine, Thalassemia Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Maral Mahboubi Kancha
- Department of Medical Nanotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Milad Ahmadi Najafabadi
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Faeze Salem
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Setareh Dashti Shokoohi
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sahar Evazi Bakhshi
- Department of Anatomical Sciences, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pouya Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pooria Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
12
|
Li Y, Ming Y, Fu R, Li C, Wu Y, Jiang T, Li Z, Ni R, Li L, Su H, Liu Y. The pathogenesis, diagnosis, prevention, and treatment of CAR-T cell therapy-related adverse reactions. Front Pharmacol 2022; 13:950923. [PMID: 36313336 PMCID: PMC9616161 DOI: 10.3389/fphar.2022.950923] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy is effective in the treatment of refractory/relapsed (r/r) hematological malignancies (r/r B-cell lymphoblastic leukemia, B-cell lymphoma, and multiple myeloma). In addition, it is being explored as a treatment option for solid tumors. As of 31 March 2022, seven CAR-T therapies for hematological malignancies have been approved worldwide. Although CAR-T therapy is an effective treatment for many malignancies, it also causes adverse effects. The incidence of cytokine release syndrome (CRS), the most common adverse reaction after infusion of CAR-T cells, is as high as 93%.CRS, is the leading risk factor of immune effector cell-associated neurotoxicity syndrome (ICANS), as well as cardiovascular, hematological, hepatorenal, skin, pulmonary, and gastrointestinal toxicity. Severe adverse reactions complicated by CRS severely impede the widespread application of CAR-T therapy. The CAR-T product was initially approved in 2017; however, only limited studies have investigated the adverse reactions owing to CAR-T therapy compared to that of clinically approved drugs. Thus, we aimed to elucidate the mechanisms, risk factors, diagnostic criteria, and treatment of toxicities concurrent with CRS, thereby providing a valuable reference for the safe, effective, and widespread application of CAR-T therapy.
Collapse
|
13
|
Zhang F, Jia XL, Zuo YX, Lu AD, Zhang PF, Xue L, Zhang LP. Continuous blood purification successfully treated severe cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome after chimeric antigen receptor T-cell therapy: A case report. Pediatr Blood Cancer 2022; 69:e29563. [PMID: 35044056 DOI: 10.1002/pbc.29563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/24/2022]
Affiliation(s)
- Feng Zhang
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Xin-Lei Jia
- Department of Pediatrics Intensive Care Unit, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Ying-Xi Zuo
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Ai-Dong Lu
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Peng-Fei Zhang
- Department of Pediatrics Intensive Care Unit, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Lian Xue
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Le-Ping Zhang
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| |
Collapse
|
14
|
Chen SY, Chen WH, Wan XC, Du X, Luo CR, Li XQ, Zhang XH. [Three cases of hemodiafiltration for the treatment of CAR-T related grade 3 - 4 cytokine release syndrome after ineffective treatment with IL-6 receptor inhibitors]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2022; 43:494-498. [PMID: 35968593 PMCID: PMC9800215 DOI: 10.3760/cma.j.issn.0253-2727.2022.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Indexed: 01/01/2023]
Abstract
Objective: To investigate the efficacy and safety of hemodiafiltration (HDF) in treating CAR-T related grade 3-4 cytokine release syndrome after ineffective treatment with IL-6 receptor inhibitors. Methods: Between July 2015 and July 2021, retrospective analysis of hemodiafiltration for the treatment of 3 patients, including 2 cases of acute B-lymphoblastic leukemia and 1 case of diffuse large B-cell lymphoma, with grade 3-4 CRS after CAR-T cell therapy and ineffective treatment with IL-6 receptor inhibitor was carried out. Results: The patient's clinical symptoms, including body temperature, blood pressure, and blood oxygen, were relieved within 12 hours of all treatments, and the cytokines (IL-6, IL-10, TNF-α, INF-γ) and C-reactive protein (CRP) levels decreased significantly. No adverse side effects were observed during the follow-up period of 3 months. Conclusion: HDF can be a safe and feasible method to treat CAR-T related grade 3- 4 CRS after ineffective treatment with IL-6 receptor inhibitors.
Collapse
Affiliation(s)
- S Y Chen
- Department of Hematology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - W H Chen
- Department of Hematology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - X C Wan
- Protein and Cell Drug Research Center, Institute of Biomedicine and Technology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - X Du
- Department of Hematology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - C R Luo
- Department of Hematology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - X Q Li
- Department of Hematology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - X H Zhang
- Department of Hematology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| |
Collapse
|
15
|
Thompson JA, Schneider BJ, Brahmer J, Achufusi A, Armand P, Berkenstock MK, Bhatia S, Budde LE, Chokshi S, Davies M, Elshoury A, Gesthalter Y, Hegde A, Jain M, Kaffenberger BH, Lechner MG, Li T, Marr A, McGettigan S, McPherson J, Medina T, Mohindra NA, Olszanski AJ, Oluwole O, Patel SP, Patil P, Reddy S, Ryder M, Santomasso B, Shofer S, Sosman JA, Wang Y, Zaha VG, Lyons M, Dwyer M, Hang L. Management of Immunotherapy-Related Toxicities, Version 1.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2022; 20:387-405. [PMID: 35390769 DOI: 10.6004/jnccn.2022.0020] [Citation(s) in RCA: 164] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The aim of the NCCN Guidelines for Management of Immunotherapy-Related Toxicities is to provide guidance on the management of immune-related adverse events resulting from cancer immunotherapy. The NCCN Management of Immunotherapy-Related Toxicities Panel is an interdisciplinary group of representatives from NCCN Member Institutions, consisting of medical and hematologic oncologists with expertise across a wide range of disease sites, and experts from the areas of dermatology, gastroenterology, endocrinology, neurooncology, nephrology, cardio-oncology, ophthalmology, pulmonary medicine, and oncology nursing. The content featured in this issue is an excerpt of the recommendations for managing toxicities related to CAR T-cell therapies and a review of existing evidence. For the full version of the NCCN Guidelines, including recommendations for managing toxicities related to immune checkpoint inhibitors, visit NCCN.org.
Collapse
Affiliation(s)
- John A Thompson
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | - Julie Brahmer
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | | | | | | | - Saurin Chokshi
- St. Jude Children's Research Hospital/The University of Tennessee Health Science Center
| | | | | | | | | | | | - Benjamin H Kaffenberger
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | | | | | | | | | | | | | - Nisha A Mohindra
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | | - Pradnya Patil
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | | | | | - Jeffrey A Sosman
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | - Vlad G Zaha
- UT Southwestern Simmons Comprehensive Cancer Center; and
| | | | | | - Lisa Hang
- National Comprehensive Cancer Network
| |
Collapse
|
16
|
Xiao X, Huang S, Chen S, Wang Y, Sun Q, Xu X, Li Y. Mechanisms of cytokine release syndrome and neurotoxicity of CAR T-cell therapy and associated prevention and management strategies. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:367. [PMID: 34794490 PMCID: PMC8600921 DOI: 10.1186/s13046-021-02148-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/20/2021] [Indexed: 02/08/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has yielded impressive outcomes and transformed treatment algorithms for hematological malignancies. To date, five CAR T-cell products have been approved by the US Food and Drug Administration (FDA). Nevertheless, some significant toxicities pose great challenges to the development of CAR T-cell therapy, most notably cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Understanding the mechanisms underlying these toxicities and establishing prevention and treatment strategies are important. In this review, we summarize the mechanisms underlying CRS and ICANS and provide potential treatment and prevention strategies.
Collapse
Affiliation(s)
- Xinyi Xiao
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Shengkang Huang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Sifei Chen
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Yazhuo Wang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China.,Medical College of Rehabilitation, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Qihang Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510623, People's Republic of China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China. .,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, 510005, People's Republic of China.
| |
Collapse
|
17
|
Safarzadeh Kozani P, Safarzadeh Kozani P, Rahbarizadeh F. Optimizing the Clinical Impact of CAR-T Cell Therapy in B-Cell Acute Lymphoblastic Leukemia: Looking Back While Moving Forward. Front Immunol 2021; 12:765097. [PMID: 34777381 PMCID: PMC8581403 DOI: 10.3389/fimmu.2021.765097] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/07/2021] [Indexed: 12/13/2022] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has been successful in creating extraordinary clinical outcomes in the treatment of hematologic malignancies including relapsed or refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL). With several FDA approvals, CAR-T therapy is recognized as an alternative treatment option for particular patients with certain conditions of B-ALL, diffuse large B-cell lymphoma, mantle cell lymphoma, follicular lymphoma, or multiple myeloma. However, CAR-T therapy for B-ALL can be surrounded by challenges such as various adverse events including the life-threatening cytokine release syndrome (CRS) and neurotoxicity, B-cell aplasia-associated hypogammaglobulinemia and agammaglobulinemia, and the alloreactivity of allogeneic CAR-Ts. Furthermore, recent advances such as improvements in media design, the reduction of ex vivo culturing duration, and other phenotype-determining factors can still create room for a more effective CAR-T therapy in R/R B-ALL. Herein, we review preclinical and clinical strategies with a focus on novel studies aiming to address the mentioned hurdles and stepping further towards a milestone in CAR-T therapy of B-ALL.
Collapse
Affiliation(s)
- Pouya Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran.,Student Research Committee, Medical Biotechnology Research Center, School of Nursing, Midwifery, and Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pooria Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.,Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
18
|
Shin YH, Tian X, Park JJ, Kim GYG, Aboujaoude E, Sturgill MG. Management of chimeric antigen receptor T-cell induced cytokine release syndrome: Current and emerging approaches. J Oncol Pharm Pract 2021; 28:159-174. [PMID: 34586003 DOI: 10.1177/10781552211039238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The most common adverse event associated with chimeric antigen receptor T-cell therapy is cytokine release syndrome, which is characterized by fever, hypoxia, and hypotension in varying degrees of severity. In severe cases, cytokine release syndrome can result in life-threatening symptoms such as multi-organ failure. The widely accepted first-line therapy for cytokine release syndrome management is tocilizumab with or without corticosteroids, but there is very limited guidance on the proper management of patients unresponsive to this regimen. There are emerging strategies that target cytokine release syndrome through novel mechanisms, showing promise in treating or preventing severe cytokine release syndrome. Although further clinical investigation is necessary to assess the applicability of the emerging approaches, these exploratory therapies may shape the future landscape of chimeric antigen receptor T-cell induced cytokine release syndrome management. This review article provides a comprehensive overview of the current and emerging therapies for the management of chimeric antigen receptor T-cell induced cytokine release syndrome, especially cases that are refractory to tocilizumab and steroids.
Collapse
Affiliation(s)
- Yunjung H Shin
- 15484Rutgers University Ernest Mario School of Pharmacy, USA
| | - Xiaofan Tian
- 15484Rutgers University Ernest Mario School of Pharmacy, USA
| | - Jiyeon J Park
- 15484Rutgers University Ernest Mario School of Pharmacy, USA.,145249Rutgers Cancer Institute of New Jersey, USA
| | - Gee Y Geeny Kim
- 15484Rutgers University Ernest Mario School of Pharmacy, USA.,3673Hackensack University Medical Center, USA
| | - Emily Aboujaoude
- 15484Rutgers University Ernest Mario School of Pharmacy, USA.,25044Robert Wood Johnson University Hospital, USA
| | - Marc G Sturgill
- 15484Rutgers University Ernest Mario School of Pharmacy, USA
| |
Collapse
|
19
|
Constantinescu C, Pasca S, Tat T, Teodorescu P, Vlad C, Iluta S, Dima D, Tomescu D, Scarlatescu E, Tanase A, Sigurjonsson OE, Colita A, Einsele H, Tomuleasa C. Continuous renal replacement therapy in cytokine release syndrome following immunotherapy or cellular therapies? J Immunother Cancer 2021; 8:jitc-2020-000742. [PMID: 32474415 PMCID: PMC7264828 DOI: 10.1136/jitc-2020-000742] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2020] [Indexed: 02/07/2023] Open
Abstract
Recently, an increasing number of novel drugs were approved in oncology and hematology. Nevertheless, pharmacology progress comes with a variety of side effects, of which cytokine release syndrome (CRS) is a potential complication of some immunotherapies that can lead to multiorgan failure if not diagnosed and treated accordingly. CRS generally occurs with therapies that lead to highly activated T cells, like chimeric antigen receptor T cells or in the case of bispecific T-cell engaging antibodies. This, in turn, leads to a proinflammatory state with subsequent organ damage. To better manage CRS there is a need for specific therapies or to repurpose strategies that are already known to be useful in similar situations. Current management strategies for CRS are represented by anticytokine directed therapies and corticosteroids. Based on its pathophysiology and the resemblance of CRS to sepsis and septic shock, as well as based on the principles of initiation of continuous renal replacement therapy (CRRT) in sepsis, we propose the rationale of using CRRT therapy as an adjunct treatment in CRS where all the other approaches have failed in controlling the clinically significant manifestations.
Collapse
Affiliation(s)
- Catalin Constantinescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Department of Anesthesia - Intensive Care, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Sergiu Pasca
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Tiberiu Tat
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Patric Teodorescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Catalin Vlad
- Department of Surgery, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Sabina Iluta
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Dana Tomescu
- Department of Anesthesia - Intensive Care, Carol Davila University of Medicine and Pharmacy, Bucuresti, Romania.,Department of Anesthesia - Intensive Care, Fundeni Clinical Institute, Bucuresti, Romania
| | - Ecaterina Scarlatescu
- Department of Stem Cell Transplantation, Clinical Institute Fundeni, Bucuresti, Romania
| | - Alina Tanase
- Department of Stem Cell Transplantation, Clinical Institute Fundeni, Bucuresti, Romania
| | - Olafur Eysteinn Sigurjonsson
- University of Reykjavik, Reykjavik, Iceland.,Bloodbank, Landspitali National University Hospital of Iceland, Reykjavik, Iceland
| | - Anca Colita
- Department of Stem Cell Transplantation, Clinical Institute Fundeni, Bucuresti, Romania
| | - Hermann Einsele
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Wurzburg, Bayern, Germany
| | - Ciprian Tomuleasa
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| |
Collapse
|
20
|
Miao L, Zhang Z, Ren Z, Li Y. Reactions Related to CAR-T Cell Therapy. Front Immunol 2021; 12:663201. [PMID: 33995389 PMCID: PMC8113953 DOI: 10.3389/fimmu.2021.663201] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
The application of chimeric antigen receptor (CAR) T-cell therapy as a tumor immunotherapy has received great interest in recent years. This therapeutic approach has been used to treat hematological malignancies solid tumors. However, it is associated with adverse reactions such as, cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), off-target effects, anaphylaxis, infections associated with CAR-T-cell infusion (CTI), tumor lysis syndrome (TLS), B-cell dysplasia, hemophagocytic lymphohistiocytosis (HLH)/macrophage activation syndrome (MAS) and coagulation disorders. These adverse reactions can be life-threatening, and thus they should be identified early and treated effectively. In this paper, we review the adverse reactions associated with CAR-T cells, the mechanisms driving such adverse reactions, and strategies to subvert them. This review will provide important reference data to guide clinical application of CAR-T cell therapy.
Collapse
Affiliation(s)
- Lele Miao
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Zhengchao Zhang
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Zhijian Ren
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Yumin Li
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
21
|
Kumar M, Sarma DK, Shubham S, Kumawat M, Verma V, Nina PB, JP D, Kumar S, Singh B, Tiwari RR. Futuristic Non-antibiotic Therapies to Combat Antibiotic Resistance: A Review. Front Microbiol 2021; 12:609459. [PMID: 33574807 PMCID: PMC7870489 DOI: 10.3389/fmicb.2021.609459] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/04/2021] [Indexed: 12/26/2022] Open
Abstract
The looming problem of resistance to antibiotics in microorganisms is a global health concern. The drug-resistant microorganisms originating from anthropogenic sources and commercial livestock farming have posed serious environmental and health challenges. Antibiotic-resistant genes constituting the environmental "resistome" get transferred to human and veterinary pathogens. Hence, deciphering the origin, mechanism and extreme of transfer of these genetic factors into pathogens is extremely important to develop not only the therapeutic interventions to curtail the infections, but also the strategies to avert the menace of microbial drug-resistance. Clinicians, researchers and policymakers should jointly come up to develop the strategies to prevent superfluous exposure of pathogens to antibiotics in non-clinical settings. This article highlights the present scenario of increasing antimicrobial-resistance in pathogenic bacteria and the clinical importance of unconventional or non-antibiotic therapies to thwart the infectious pathogenic microorganisms.
Collapse
Affiliation(s)
- Manoj Kumar
- ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | | | - Swasti Shubham
- ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Manoj Kumawat
- ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Vinod Verma
- Stem Cell Research Centre, Department of Hematology, SGPGIMS, Lucknow, India
| | | | - Devraj JP
- ICMR- National Institute of Nutrition, Hyderabad, India
| | - Santosh Kumar
- ICMR- National Institute of Nutrition, Hyderabad, India
| | - Birbal Singh
- ICAR-Indian Veterinary Research Institute, Regional Station, Palampur, India
| | | |
Collapse
|
22
|
Gust J, Ponce R, Liles WC, Garden GA, Turtle CJ. Cytokines in CAR T Cell-Associated Neurotoxicity. Front Immunol 2020; 11:577027. [PMID: 33391257 PMCID: PMC7772425 DOI: 10.3389/fimmu.2020.577027] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cells provide new therapeutic options for patients with relapsed/refractory hematologic malignancies. However, neurotoxicity is a frequent, and potentially fatal, complication. The spectrum of manifestations ranges from delirium and language dysfunction to seizures, coma, and fatal cerebral edema. This novel syndrome has been designated immune effector cell-associated neurotoxicity syndrome (ICANS). In this review, we draw an arc from our current understanding of how systemic and potentially local cytokine release act on the CNS, toward possible preventive and therapeutic approaches. We systematically review reported correlations of secreted inflammatory mediators in the serum/plasma and cerebrospinal fluid with the risk of ICANS in patients receiving CAR T cell therapy. Possible pathophysiologic impacts on the CNS are covered in detail for the most promising candidate cytokines, including IL-1, IL-6, IL-15, and GM-CSF. To provide insight into possible final common pathways of CNS inflammation, we place ICANS into the context of other systemic inflammatory conditions that are associated with neurologic dysfunction, including sepsis-associated encephalopathy, cerebral malaria, thrombotic microangiopathy, CNS infections, and hepatic encephalopathy. We then review in detail what is known about systemic cytokine interaction with components of the neurovascular unit, including endothelial cells, pericytes, and astrocytes, and how microglia and neurons respond to systemic inflammatory challenges. Current therapeutic approaches, including corticosteroids and blockade of IL-1 and IL-6 signaling, are reviewed in the context of what is known about the role of cytokines in ICANS. Throughout, we point out gaps in knowledge and possible new approaches for the investigation of the mechanism, prevention, and treatment of ICANS.
Collapse
Affiliation(s)
- Juliane Gust
- Department of Neurology, University of Washington, Seattle, WA, United States
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA, United States
| | | | - W. Conrad Liles
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Gwenn A. Garden
- Department of Neurology, University of North Carolina, Chapel Hill, NC, United States
| | - Cameron J. Turtle
- Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| |
Collapse
|
23
|
Li W, Wu L, Huang C, Liu R, Li Z, Liu L, Shan B. Challenges and strategies of clinical application of CAR-T therapy in the treatment of tumors-a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1093. [PMID: 33145312 PMCID: PMC7575947 DOI: 10.21037/atm-20-4502] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has achieved good therapeutic efficacy in the treatment of hematological malignancies. In August 2017, Novartis Kymriah (CAR-T cells targeting CD19) was approved by the FDA, indicating the real entry of CAR-T cell therapy into clinical applications and making CAR-T cell therapy the most attractive technology in the field of tumor treatment. In October 2017, the FDA approved the world’s second CAR-T cell therapy—Yescarta. The launch of these products has attracted wide attention to CAR-T cell therapy. CAR-T cell therapy has achieved significant effect in the treatment of tumors, however, CAR-T therapy also faces clinical problems, such as cytokine release syndrome (CRS), poor therapeutic efficacy in solid tumors, and high rates of tumor recurrence. At present, the side effects of CAR-T therapy have attracted a large amount of attention, which has resulted in investigations into strategy establishment. With a deepening understanding of CAR-T therapy and the continuous optimization of therapeutic regimens, its toxicity and side effects have been partially controlled. This study set out to analyze the problems in the clinical application of CAR-T therapy encountered in recent years and to introduce corresponding strategies, with the aim of providing a basis of reference for clinicians and scientists in the management of CAR-T therapy in clinical practice and in the CAR-T therapy research.
Collapse
Affiliation(s)
- Weijing Li
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lili Wu
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chen Huang
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ruixia Liu
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zheng Li
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lihong Liu
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Baoen Shan
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
24
|
Chimeric antigen receptor T cell therapy and nephrotoxicity: From diagnosis to treatment strategies. Int Immunopharmacol 2020; 89:107072. [PMID: 33059198 DOI: 10.1016/j.intimp.2020.107072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/13/2020] [Accepted: 10/01/2020] [Indexed: 01/28/2023]
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy is a breakthrough in cancer treatment. With the widespread use of this therapy, increasing evidence is available that CAR-T cell therapy is associated with acute kidney injury (AKI). Nephrologists need to understand the potential nephrotoxicity arising from CAR-T cell therapy. Determining the cause of AKI is a key factor of clinical management. This review focuses on the clinical use of CAR-T cell therapy and the cause and outcomes of nephrotoxicity with its use. We also provide clinical suggestions for clinicians towards both better diagnosis and management of AKI in those receiving CAR-T cell therapy.
Collapse
|
25
|
Liu Y, Fang Y, Chen X, Wang Z, Liang X, Zhang T, Liu M, Zhou N, Lv J, Tang K, Xie J, Gao Y, Cheng F, Zhou Y, Zhang Z, Hu Y, Zhang X, Gao Q, Zhang Y, Huang B. Gasdermin E-mediated target cell pyroptosis by CAR T cells triggers cytokine release syndrome. Sci Immunol 2020; 5:5/43/eaax7969. [PMID: 31953257 DOI: 10.1126/sciimmunol.aax7969] [Citation(s) in RCA: 311] [Impact Index Per Article: 77.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/28/2019] [Accepted: 12/19/2019] [Indexed: 12/16/2022]
Abstract
Cytokine release syndrome (CRS) counteracts the effectiveness of chimeric antigen receptor (CAR) T cell therapy in cancer patients, but the mechanism underlying CRS remains unclear. Here, we show that tumor cell pyroptosis triggers CRS during CAR T cell therapy. We find that CAR T cells rapidly activate caspase 3 in target cells through release of granzyme B. The latter cleaves gasdermin E (GSDME), a pore-forming protein highly expressed in B leukemic and other target cells, which results in extensive pyroptosis. Consequently, pyroptosis-released factors activate caspase 1 for GSDMD cleavage in macrophages, which results in the release of cytokines and subsequent CRS. Knocking out GSDME, depleting macrophages, or inhibiting caspase 1 eliminates CRS occurrence in mouse models. In patients, GSDME and lactate dehydrogenase levels are correlated with the severity of CRS. Notably, we find that the quantity of perforin/granzyme B used by CAR T cells rather than existing CD8+ T cells is critical for CAR T cells to induce target cell pyroptosis.
Collapse
Affiliation(s)
- Yuying Liu
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China.,Clinical Immunology Center, CAMS, Beijing 100005, China
| | - Yiliang Fang
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China
| | - Xinfeng Chen
- Biotherapy Center and Cancer Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Zhenfeng Wang
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China
| | - Xiaoyu Liang
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China
| | - Tianzhen Zhang
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China
| | - Mengyu Liu
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China
| | - Nannan Zhou
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China
| | - Jiadi Lv
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China
| | - Ke Tang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jing Xie
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China
| | - Yunfeng Gao
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China
| | - Feiran Cheng
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China
| | - Yabo Zhou
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China
| | - Zhen Zhang
- Biotherapy Center and Cancer Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China
| | - Quanli Gao
- Department of Immunology, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Bo Huang
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing 100005, China. .,Clinical Immunology Center, CAMS, Beijing 100005, China.,Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
26
|
Zhang Z, Chen X, Tian Y, Li F, Zhao X, Liu J, Yao C, Zhang Y. Point mutation in CD19 facilitates immune escape of B cell lymphoma from CAR-T cell therapy. J Immunother Cancer 2020; 8:jitc-2020-001150. [PMID: 33023981 PMCID: PMC7539592 DOI: 10.1136/jitc-2020-001150] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tumor relapse due to mutation in CD19 can hinder the efficacy of chimeric antigen receptor (CAR)-T cell therapy. Herein, we focused on lymphoma patients whose B cells exhibited a point mutation in CD19 of B cells after CAR-T cell infusion. METHODS The CAR-T and CD19+ B cells from peripheral blood or bone marrow were assessed using flow cytometry. Genome sequencing was conducted to identify the molecular characteristics of CAR-T and CD19+ B cells from pre-relapse and postrelapse samples. CD19 in CARs comprising single chain fragments variable (scFV) antibody with FMC63 or 21D4 was constructed. The cytotoxic efficacy of CAR-T cells was also evaluated via in vitro and in vivo experiments. RESULTS A patient with high-grade B cell lymphoma exhibited complete response, but the lymphoma relapsed in her left breast at 6 months after CD19 CAR (FMC63)-T cell infusion. A mutation was found in exon 3 of CD19 (p.163. R-L) in malignant B cells of the patient. In two lymphoma patients who exhibited resistance to CAR-T cell therapy, a mutation was detected in exon 3 of CD19 (p.174. L-V). Functional analysis revealed that FMC63 CAR-T cells exhibited antitumor ability against wild-type CD19+ cells but were unable to eradicate these two types of mutated CD19+ cells. Interestingly, 21D4 CAR-T cells were potentially capable of eradicating these mutated CD19+ cells and exhibiting high antitumor capacity against CD19+ cells with loss of exon 1, 2, or 3. CONCLUSIONS These findings suggest that point mutation can facilitate immune escape from CAR-T cell therapy and that alternative CAR-T cells can effectively eradicate the mutated B cells, providing an individualized therapeutic approach for lymphoma patients showing relapse.
Collapse
Affiliation(s)
- Zhen Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xinfeng Chen
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yonggui Tian
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feng Li
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xuan Zhao
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jinyan Liu
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chang Yao
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, China
| |
Collapse
|
27
|
Cai C, Tang D, Han Y, Shen E, Abdihamid O, Guo C, Shen H, Zeng S. A comprehensive analysis of the fatal toxic effects associated with CD19 CAR-T cell therapy. Aging (Albany NY) 2020; 12:18741-18753. [PMID: 32973124 PMCID: PMC7585129 DOI: 10.18632/aging.104058] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/25/2020] [Indexed: 01/24/2023]
Abstract
To determine the incidence, spectrum, timing, and clinical features of CD19 Chimeric antigen receptor (CAR-T) cell therapy-associated fatal toxic effects. We initiated a comprehensive analysis. First, we retrospectively queried the real-world data from a World Health Organization (WHO) pharmacovigilance database (Vigilyze). Furthermore, we performed a meta-analysis of published trials of CD19 CAR-T cell therapy. From screening the WHO database, we identified 1200 patients: 499 received Kymriah therapy, and 701 received Yescarta therapy. We compared the adverse reactions of the two drugs. We evaluated all published clinical trials, comprising 19 trials and 890 patients. Our meta-analysis showed that the incidence of fatal toxic effects associated with death was 5.4%. Infections and infestations appeared to present the highest risk of death. The toxic effect specific median time to death was 30, 30, and 68 days for total, cytokine release syndrome (CRS), and nervous system disorders (NSD), respectively. We observed a high mortality rate for some toxic effects and an early onset of death with varied causes, indicating the need for clinicians to pay more attention to the monitoring and treatment of these fatal toxic effects when using CD19 CAR-T cell therapy, especially for infections and infestations.
Collapse
Affiliation(s)
- Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China,Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Diya Tang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China,Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China,Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Edward Shen
- Department of Life Science, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Omar Abdihamid
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Cao Guo
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China,Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China,Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China,Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
| |
Collapse
|
28
|
Safari S, Salimi A, Zali A, Jahangirifard A, Bastanhagh E, Aminnejad R, Dabbagh A, Lotfi AH, Saeidi M. Extracorporeal Hemoperfusion as a Potential Therapeutic Option for Severe COVID-19 patients; a Narrative Review. ARCHIVES OF ACADEMIC EMERGENCY MEDICINE 2020; 8:e67. [PMID: 33134963 PMCID: PMC7587998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The 2019 novel coronavirus (officially known as severe acute respiratory syndrome coronavirus 2, SARS-CoV2) was first found in Wuhan, China. On February 11, 2020, the World Health Organization (WHO) has declared the outbreak of the disease caused by SARS-CoV2, named coronavirus disease 2019 (COVID-19), as an emergency of international concern. Based on the current epidemiological surveys, some COVID-19 patients with severe infection gradually develop impairment of the respiratory system, acute kidney injury (AKI), multiple organ failure, and ultimately, death. Currently, there is no established pharmacotherapy available for COVID-19. As seen in influenza, immune damage mediated by excessive production of inflammatory mediators contributes to high incidence of complications and poor prognosis. Thus, removal or blocking the overproduction of these mediators potentially aids in reducing the deleterious cytokine storm and improving critically ill patients' outcomes. Based on previous experience of blood purification to treat cytokine storm syndrome (CSS) in severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS), here we aimed to review the current literature on extracorporeal hemoperfusion as a potential therapeutic option for CSS-associated conditions, with a focus on severe COVID-19.
Collapse
Affiliation(s)
- Saeid Safari
- Functional Neurosurgery Research Center, Shohada Tajrish Neurosurgical Comprehensive Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Salimi
- Department of Anesthesiology and Critical Care, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Zali
- Functional Neurosurgery Research Center, Shohada Tajrish Neurosurgical Comprehensive Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Jahangirifard
- Chronic Respiratory Disease Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ehsan Bastanhagh
- Department of Anesthesiology and Critical Care, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Aminnejad
- Department of Anesthesiology and Critical Care, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Anesthesiology and Critical Care, Qom University of Medical Sciences, Qom, Iran
| | - Ali Dabbagh
- Anesthesiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Saeidi
- Department of Anesthesiology and Critical Care, Qom University of Medical Sciences, Qom, Iran
| |
Collapse
|
29
|
The model of cytokine release syndrome in CAR T-cell treatment for B-cell non-Hodgkin lymphoma. Signal Transduct Target Ther 2020; 5:134. [PMID: 32728035 PMCID: PMC7388484 DOI: 10.1038/s41392-020-00256-x] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/12/2020] [Accepted: 07/15/2020] [Indexed: 02/05/2023] Open
Abstract
Chimeric antigen receptor T (CAR T) cell therapy has demonstrated efficacy in the treatment of haematologic malignancies. However, the accompanying adverse events, the most common of which is cytokine release syndrome (CRS), substantially limit its wide application. Due to its unique physiological characteristics, CRS in CAR T-cell treatment for B-cell non-Hodgkin lymphoma (B-NHL) may exhibit some special features. Although existing guidelines had greatly promoted the recognition and management of CRS, many recommendations are not fully applicable to B-NHL. Therefore, it is imperative to identify responses that are specific to CRS observed following CAR T treatment for B-NHL. Based on underlying biological processes and known pathophysiological mechanisms, we tentatively propose a new model to illustrate the occurrence and evolution of CAR T-cell-therapy-related CRS in B-NHL. In this model, tumour burden and bone marrow suppression are considered determinants of CRS. Novel phenomena after CAR T-cell infusion (such as local inflammatory response) are further identified. The proposed model will help us better understand the basic biology of CRS and recognize and manage it more rationally.
Collapse
|
30
|
Huang C, Zhang HC, Ho JY, Liu RX, Wang L, Kuang N, Zheng MR, Liu LH, Li JQ. Dual specific CD19/CD22-targeted chimeric antigen receptor T-cell therapy for refractory diffuse large B-cell lymphoma: A case report. Oncol Lett 2020; 20:21. [PMID: 32774494 PMCID: PMC7405542 DOI: 10.3892/ol.2020.11882] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 04/09/2020] [Indexed: 12/21/2022] Open
Abstract
Clinical trials of chimeric antigen receptors (CARs) targeting CD19 have produced impressive results in hematological malignancies, including diffuse large B-cell lymphoma (DLBCL). However, a notable number of patients with DLBCL fail to achieve remission after CD19 CAR T-cell therapy and may therefore require a dual targeted CAR T-cell therapy. A 31-year-old man with refractory DLBCL was assessed in the present case report. The patient was treated with sequential infusion of single CD19 CAR T cells followed by dual CD19/CD22-targeted CAR T cells. The outcome was that the patient achieved partial remission after the first single CD19 CAR T-cell infusion and complete remission after the dual CD19/CD22-targeted CAR T-cell infusion. Grade 1 cytokine release syndrome (CRS) was observed after the single CD19 CAR T-cell infusion, while grade 3 CRS and hemophagocytic syndrome were observed after the dual targeted CAR T-cell infusion, but these adverse effects alleviated after the treatments. To the best of our knowledge, the present case report is the first to describe the successful application of dual CD19/CD22-targeted CAR T-cell therapy for the treatment of refractory DLBCL. The report suggests that dual CD19/CD22-targeted CAR T-cell therapy may represent a promising option for the treatment of refractory DLBCL; however, caution should be taken due to potential CRS development.
Collapse
Affiliation(s)
- Chen Huang
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Hui-Chao Zhang
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Jin-Yuan Ho
- Hebei Senlang Biotechnology Co., Ltd., Shijiazhuang, Hebei 050011, P.R. China
| | - Rui-Xia Liu
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Lin Wang
- Hebei Senlang Biotechnology Co., Ltd., Shijiazhuang, Hebei 050011, P.R. China
| | - Na Kuang
- Hebei Senlang Biotechnology Co., Ltd., Shijiazhuang, Hebei 050011, P.R. China
| | - Mei-Rong Zheng
- Hebei Senlang Biotechnology Co., Ltd., Shijiazhuang, Hebei 050011, P.R. China
| | - Li-Hong Liu
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Jian-Qiang Li
- Hebei Senlang Biotechnology Co., Ltd., Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
31
|
Wang L, Tan Su Yin E, Zhao H, Ni F, Hu Y, Huang H. CAR-T cells: the Chinese experience. Expert Opin Biol Ther 2020; 20:1293-1308. [PMID: 32605454 DOI: 10.1080/14712598.2020.1790521] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Chimeric antigen receptor T (CAR-T) cells are harnessed to identify and lyse malignant cells specifically, efficiently, and independently of the major histocompatibility complex (MHC). As a result, prognoses of relapsed or refractory (R/R) B cell hematological malignancies as well as limited types of solid tumors, have been ameliorated to a great extent. In China, a rising number of clinical trials that contribute to the development of novel CAR-T therapeutic strategies have been conducted on an extensive scale. AREAS COVERED We summarize registered clinical trials related to CAR-T therapy conducted in China by evaluating various parameters such as distribution, study phase, CAR structure, target antigen, and disease. The efficacy, toxicity, and, more importantly, the new strategies for optimization of CAR-T therapy of Chinese studies and clinical trials are elaborated in detail. EXPERT OPINION In terms of the number of CAR-T clinical trials, China is second to the USA, registering approximately 33% of trials worldwide. China's extensive explorations and breakthroughs in the search of novel target antigens, optimization of CAR structure, cocktail CAR-T therapy, combination therapy, and extension of CAR-T cell applications, imply that we are currently on the verge of a revolution in CAR-T therapy.
Collapse
Affiliation(s)
- Linqin Wang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, China.,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy , Hangzhou, China.,Institute of Hematology, Zhejiang University , Hangzhou, China
| | - Elaine Tan Su Yin
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, China.,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy , Hangzhou, China.,Institute of Hematology, Zhejiang University , Hangzhou, China
| | - Houli Zhao
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, China.,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy , Hangzhou, China.,Institute of Hematology, Zhejiang University , Hangzhou, China
| | - Fang Ni
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, China.,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy , Hangzhou, China.,Institute of Hematology, Zhejiang University , Hangzhou, China
| | - Yongxian Hu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, China.,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy , Hangzhou, China.,Institute of Hematology, Zhejiang University , Hangzhou, China
| | - He Huang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, China.,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy , Hangzhou, China.,Institute of Hematology, Zhejiang University , Hangzhou, China
| |
Collapse
|
32
|
Chen X, Li X, Liu Y, Zhang Z, Zhang X, Huang J, Li H, Li F, Zhang L, Li L, Wu X, Ma W, Sun Z, Yu H, Zhou Z, Feng X, Cui K, Li Z, Zhang H, Zeng Y, Wan X, Chen YH, Zhang M, Zhang Y. A Phase I clinical trial of chimeric antigen receptor-modified T cells in patients with relapsed and refractory lymphoma. Immunotherapy 2020; 12:681-696. [PMID: 32580597 DOI: 10.2217/imt-2020-0022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: CD19 chimeric antigen receptor (CAR) T cells have been approved by the US FDA for treatment of relapsed and refractory (R/R) B-cell malignancies. Patients & methods: This study investigated the safety and efficacy of autologous 4-1BB costimulatory domain-engineered CD19 CAR-T cells in R/R B-cell lymphoma. Results: After CD19 CAR-T-cell infusion, severe cytokine release syndrome occurred in 28.6% (4/14) of the patients. The overall response rate was 77% with complete remission observed in 6/14 patients at 3 months. A higher tumor burden and grade 3-4 of myelosuppression after chemotherapy were associated with severe cytokine-release syndrome. Notably, combining CD19 CAR-T cells and PD-1 blockade, but not CD19 CAR-T cells alone, reduced intracranial tumor burden in a patient with central invasion of lymphoma. Conclusion: CD19 CAR-T cells could effectively induce tumor remission and PD-1 blockade might improve the efficacy in Chinese patients with R/R B-cell lymphoma.
Collapse
Affiliation(s)
- Xinfeng Chen
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.,Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xin Li
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yanfen Liu
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.,Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Zhen Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.,Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xudong Zhang
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jianmin Huang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Hong Li
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Feng Li
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Lei Zhang
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Ling Li
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xiaolong Wu
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Wang Ma
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Zhenchang Sun
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Hui Yu
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Zhiyuan Zhou
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xiaoyan Feng
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Kang Cui
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Zhaoming Li
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | | | - Ying Zeng
- Binde Biotech Inc., Shenzhen 518055, Guangdong, China
| | - Xiaochun Wan
- Center for Antibody Drug Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
| | - Youhai H Chen
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mingzhi Zhang
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yi Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.,Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.,School of Life Sciences, Zhengzhou University, Zhengzhou 450052, Henan, China.,Engineering Key Laboratory for Cell Therapy of Henan Province, Zhengzhou 450052, Henan, China
| |
Collapse
|
33
|
Chou CK, Turtle CJ. Assessment and management of cytokine release syndrome and neurotoxicity following CD19 CAR-T cell therapy. Expert Opin Biol Ther 2020; 20:653-664. [PMID: 32067497 PMCID: PMC7393694 DOI: 10.1080/14712598.2020.1729735] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 02/11/2020] [Indexed: 12/14/2022]
Abstract
Introduction: The success of CD19 chimeric antigen receptor (CAR)-T cell therapy for treatment of CD19 positive malignancies has led to the FDA approval of two CD19 CAR-T cell products, tisagenlecleucel and axicabtagene ciloleucel, and ongoing clinical trials of new products. Cytokine release syndrome (CRS) and neurotoxicity are common toxicities associated with CD19 CAR-T cell therapies.Areas covered: This review will discuss CRS and neurotoxicity associated with CD19 CAR-T cell therapies, including clinical presentation, risk factors, pathophysiology, and therapeutic or prophylactic interventions.Expert opinion: In conjunction with improved understanding of the pathophysiology of CRS and neurotoxicity, we expect that the recent development of consensus guidelines for the evaluation of these toxicities will enhance management of patients undergoing CD19 CAR-T cell therapies.
Collapse
Affiliation(s)
- Cassie K. Chou
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Pediatrics, University of Washington, Seattle, Washington
| | - Cameron J. Turtle
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
34
|
Cardiovascular Events Among Adults Treated With Chimeric Antigen Receptor T-Cells (CAR-T). J Am Coll Cardiol 2020; 74:3099-3108. [PMID: 31856966 DOI: 10.1016/j.jacc.2019.10.038] [Citation(s) in RCA: 218] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 09/09/2019] [Accepted: 10/01/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Chimeric antigen receptors redirect T cells (CAR-T) to target cancer cells. There are limited data characterizing cardiac toxicity and cardiovascular (CV) events among adults treated with CAR-T. OBJECTIVES The purpose of this study was to evaluate the possible cardiac toxicities of CAR-T. METHODS The registry included 137 patients who received CAR-T. Covariates included the occurrence and grade of cytokine release syndrome (CRS) and the administration of tocilizumab for CRS. Cardiac toxicity was defined as a decrease in the left ventricular ejection fraction or an increase in serum troponin. Cardiovascular events were a composite of arrhythmias, decompensated heart failure, and CV death. RESULTS The median age was 62 years (interquartile range [IQR]: 54 to 70 years), 67% were male, 88% had lymphoma, and 8% had myeloma. Approximately 50% were treated with commercial CAR-T (Yescarta or Kymriah), and the remainder received noncommercial products. CRS, occurring a median of 5 days (IQR: 2 to 7 days) after CAR-T, occurred in 59%, and 39% were grade ≥2. Tocilizumab was administered to 56 patients (41%) with CRS, at a median of 27 h (IQR: 16 to 48 h) after onset. An elevated troponin occurred in 29 of 53 tested patients (54%), and a decreased left ventricular ejection fraction in 8 of 29 (28%); each occurred only in patients with grade ≥2 CRS. There were 17 CV events (12%, 6 CV deaths, 6 decompensated heart failure, and 5 arrhythmias; median time to event of 21 days), all occurred with grade ≥2 CRS (31% patients with grade ≥2 CRS), and 95% of events occurred after an elevated troponin. The duration between CRS onset and tocilizumab administration was associated with CV events, where the risk increased 1.7-fold with each 12-h delay to tocilizumab. CONCLUSIONS Among adults, cardiac injury and CV events are common post-CAR-T. There was a graded relationship among CRS, elevated troponin, and CV events, and a shorter time from CRS onset to tocilizumab was associated with a lower rate of CV events.
Collapse
|
35
|
Boyarinov G, Zubeyev P, Mokrov K, Voyennov O. Hemofiltration in Patients with Severe Acute Pancreatitis (Review). Sovrem Tekhnologii Med 2020; 12:105-121. [PMID: 34513045 PMCID: PMC8353697 DOI: 10.17691/stm2020.12.1.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Indexed: 11/19/2022] Open
Abstract
Questions regarding the application of extracorporeal detoxification to patients with severe acute pancreatitis have been considered. Hemodialysis, the historically first method of extracorporeal detoxification for such patients, has been also described in the review. Appropriateness of using renal replacement therapy methods and among them continued renal replacement therapy has been shown. Hemofiltration and hemodiafiltration technologies are described in detail including different modes of their application and the possibility of using various types of filters. Available data on hemofiltration for patients with severe acute pancreatitis have been analyzed. Great attention is paid to the unsolved aspects of hemofiltration in severe acute pancreatitis such as determining renal and extrarenal indices; time of starting hemofiltration; selection of volume replacement modes and a buffer system; procedure duration; anticoagulation measures, defining criteria to assess the adequacy of hemofiltration, state severity, and organ dysfunction degree. Further multicenter investigations are necessary to be able to assess the efficacy of the hemofiltration procedures on the basis of the thoroughly worked out and pathogenically grounded protocol using adequate control methods taking into consideration endogenic intoxication phases and intensity of the multiple organ failure syndrome.
Collapse
Affiliation(s)
- G.A. Boyarinov
- Professor, Head of the Department of Anesthesiology and Resuscitation, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - P.S. Zubeyev
- Professor, Head of the Department of Emergency Medical Care, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - K.V. Mokrov
- Head of the Resuscitation and Anesthesiology Unit, City Hospital No.33, 54 Lenin Avenue, Nizhny Novgorod, 603076, Russia
| | - O.V. Voyennov
- Professor, Department of Anesthesiology and Resuscitation, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| |
Collapse
|
36
|
Mihăilă RG. Chimeric Antigen Receptor-Engineered T-Cells - A New Way and Era for Lymphoma Treatment. Recent Pat Anticancer Drug Discov 2019; 14:312-323. [PMID: 31642414 DOI: 10.2174/1574892814666191022164641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 10/16/2019] [Accepted: 10/19/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Patients with refractory or relapsed diffuse large B-cell lymphoma have a poor prognosis with the current standard of care. OBJECTIVE Chimeric Antigen Receptor T-cells (CAR T-cells) are functionally reprogrammed lymphocytes, which are able to recognize and kill tumor cells. The aim of this study is to make progress in this area. METHODS A mini-review was achieved using the articles published in Web of Science and PubMed in the last year and the new patents were made in this field. RESULTS The responses to CAR T-cell products axicabtagene ciloleucel and tisagenlecleucel are promising; the objective response rate can reach up to 83%, and the complete response rate ranges between 40 and 58%. About half of the patients may have serious side effects, such as cytokine release syndrome and neurotoxicity. Current and future developments include the improvement of CAR T-cell expansion and polyfunctionality, the combined use of CAR T-cells with a fusion protein between interferon and an anti-CD20 monoclonal antibody, with checkpoint inhibitors or small molecule sensitizers that have apoptotic-regulatory effects. Furthermore, the use of IL-12-expressing CAR T-cells, an improved technology for the production of CAR T-cells based on targeted nucleases, the widespread use of allogeneic CAR T-cells or universal CAR T-cells obtained from genetically engineered healthy donor T-cells are future developments actively considered. CONCLUSION CAR T-cell therapy significantly improved the outcome of patients with relapsed or refractory diffuse large B-cell lymphoma. The advances in CAR T-cells production technology will improve the results and enable the expansion of this new immunotherapy.
Collapse
Affiliation(s)
- Romeo G Mihăilă
- "Lucian Blaga" University of Sibiu, Faculty of Medicine, Emergency County Clinical Hospital Sibiu, Sibiu 550169, Romania
| |
Collapse
|
37
|
Constantinescu C, Bodolea C, Pasca S, Teodorescu P, Dima D, Rus I, Tat T, Achimas-Cadariu P, Tanase A, Tomuleasa C, Einsele H. Clinical Approach to the Patient in Critical State Following Immunotherapy and/or Stem Cell Transplantation: Guideline for the On-Call Physician. J Clin Med 2019; 8:E884. [PMID: 31226876 PMCID: PMC6616972 DOI: 10.3390/jcm8060884] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 12/11/2022] Open
Abstract
: The initial management of the hematology patient in a critical state is crucial and poses a great challenge both for the hematologist and the intensive care unit (ICU) physician. After years of clinical practice, there is still a delay in the proper recognition and treatment of critical situations, which leads to late admission to the ICU. There is a much-needed systematic ABC (Airway, Breathing, Circulation) approach for the patients being treated on the wards as well as in the high dependency units because the underlying hematological disorder, as well as disease-related complications, have an increasing frequency. Focusing on score-based decision-making on the wards (Modified Early Warning Score (MEWS), together with Quick Sofa score), active sepsis screening with inflammation markers (C-reactive protein, procalcitonin, and presepsin), and assessment of microcirculation, organ perfusion, and oxygen supply by using paraclinical parameters from the ICU setting (lactate, central venous oxygen saturation (ScVO2), and venous-to-arterial carbon dioxide difference), hematologists can manage the immediate critical patient and improve the overall outcome.
Collapse
Affiliation(s)
- Catalin Constantinescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania.
- Intensive Care Unit, Ion Chiricuta Clinical Cancer Center, 400005 Cluj Napoca, Romania.
| | - Constantin Bodolea
- Department of Anesthesia, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania.
| | - Sergiu Pasca
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania.
| | - Patric Teodorescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania.
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, 400005 Cluj Napoca, Romania.
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, 400005 Cluj Napoca, Romania.
| | - Ioana Rus
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, 400005 Cluj Napoca, Romania.
| | - Tiberiu Tat
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, 400005 Cluj Napoca, Romania.
| | - Patriciu Achimas-Cadariu
- Department of Surgery, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania.
| | - Alina Tanase
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, 022328 Bucharest, Romania.
| | - Ciprian Tomuleasa
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400124 Cluj Napoca, Romania.
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, 400005 Cluj Napoca, Romania.
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Victor Babes Street, 400124 Cluj Napoca, Romania.
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Wuerzburg, 97070 Wuerzburg, Germany.
| |
Collapse
|
38
|
Garcia Borrega J, Gödel P, Rüger MA, Onur ÖA, Shimabukuro-Vornhagen A, Kochanek M, Böll B. In the Eye of the Storm: Immune-mediated Toxicities Associated With CAR-T Cell Therapy. Hemasphere 2019; 3:e191. [PMID: 31723828 PMCID: PMC6746039 DOI: 10.1097/hs9.0000000000000191] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 02/15/2019] [Accepted: 02/15/2019] [Indexed: 12/16/2022] Open
Abstract
The success of chimeric antigen receptor (CAR)-T cell therapy with impressive response rates in hematologic malignancies but also promising data in solid tumors came along with the cognition of unexpected, potentially life-threatening immune-mediated toxicities, namely the cytokine release syndrome (CRS) and neurotoxicity recently referred to as "immune effector cell-associated neurotoxicity syndrome" (ICANS). These toxicities require urgent diagnostic and therapeutic interventions and targeted modulation of key cytokine pathways represents the mainstay of CRS treatment. However, as the underlying mechanisms of ICANS are not well understood, treatment options remain limited and further investigation is warranted. Importantly, after the recent market approval of 2 CAR-T cell constructs, the application of CAR-T cells will expand to nonacademic centers with limited experience in the management of CAR-T cell-associated toxicities. Here, we review the current evidence of CRS and ICANS pathophysiology, diagnostics, and treatment.
Collapse
Affiliation(s)
- Jorge Garcia Borrega
- Department I of Internal Medicine, Hematology-Oncology, Center of Integrated Oncology Cologne-Bonn, University Hospital of Cologne, Cologne, Germany
- Intensive Care Program, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Philipp Gödel
- Department I of Internal Medicine, Hematology-Oncology, Center of Integrated Oncology Cologne-Bonn, University Hospital of Cologne, Cologne, Germany
- Intensive Care Program, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
- Cologne Translational Immunology, University Hospital of Cologne, Cologne, Germany
| | - Maria Adele Rüger
- Department of Neurology, University Hospital of Cologne, Cologne, Germany
| | - Özgür A. Onur
- Department of Neurology, University Hospital of Cologne, Cologne, Germany
| | - Alexander Shimabukuro-Vornhagen
- Department I of Internal Medicine, Hematology-Oncology, Center of Integrated Oncology Cologne-Bonn, University Hospital of Cologne, Cologne, Germany
- Intensive Care Program, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
- Intensive Care in HematoOncologic Patients (iCHOP) Collaborative Group
| | - Matthias Kochanek
- Department I of Internal Medicine, Hematology-Oncology, Center of Integrated Oncology Cologne-Bonn, University Hospital of Cologne, Cologne, Germany
- Intensive Care Program, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
- Intensive Care in HematoOncologic Patients (iCHOP) Collaborative Group
| | - Boris Böll
- Department I of Internal Medicine, Hematology-Oncology, Center of Integrated Oncology Cologne-Bonn, University Hospital of Cologne, Cologne, Germany
- Intensive Care Program, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
- Intensive Care in HematoOncologic Patients (iCHOP) Collaborative Group
| |
Collapse
|
39
|
Yakoub-Agha I, Moreau AS, Ahmad I, Borel C, Hadhoum N, Masouridi-Levrat S, Naudin J, Nicolas-Virelizier E, Ouachée-Chardin M, Platon L, Quessar A, Roth-Guepin G, Beauvais D, Baruchel A, Cornillon J. [Management of cytokine release syndrome in adult and pediatric patients undergoing CAR-T cell therapy for hematological malignancies: Recommendation of the French Society of Bone Marrow and cellular Therapy (SFGM-TC)]. Bull Cancer 2019; 106:S102-S109. [PMID: 30661749 DOI: 10.1016/j.bulcan.2018.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 12/02/2018] [Indexed: 12/12/2022]
Abstract
The cytokine release syndrome (CRS) is the most common complication after adoptive immunotherapies such as chimeric antigen receptor T cells (CAR-T). The incidence varies from 30 to 100% depending on the CAR-T construct, cell doses and the underlying disease. Severe cases may involve 10 to 30% of patients. The triggering event is the activation of the CAR-T, after meeting with their target. The T cell activation leads to the release of effector cytokines, such as IFNγ, TNFα and IL2, that are responsible for the activating of monocyte/macrophage system, resulting in the production of pro-inflammatory cytokines, (including IL6, IFN-γ, IL10, MCP1) and associated with a significant elevation of CRP and ferritin. The CRS usually appears between 1 and 14days after the infusion of the cells and can last from 1 to 10days. Rare fatal cases have been reported in the literature. The first symptom is often a fever, sometimes very high, which must alert and reinforce the surveillance. In moderate forms, one can find fatigue, headache, rash, arthralgia and myalgia. T cell-related encephalopathy (CRES) syndrome may occur concomitantly. In case of aggravation, a vasoplegic shock associating capillary leakage and respiratory distress can occur. Close clinical monitoring is essential right from the injection to quickly detect the first symptoms. The treatment of severe forms, in addition to symptomatic management involves monoclonal antibodies targeting the IL6 or IL6 receptor, and sometimes steroids. Close cooperation with intensive care units is essential since 20 to 50% of patients require intensive care unit transfer.
Collapse
Affiliation(s)
| | - Anne-Sophie Moreau
- CHU de Lille, hôpital Salengro, centre de réanimation, 1, rue Émile-Laine, 59000 Lille, France
| | - Imran Ahmad
- Université de Montréal, hôpital Maisonneuve-Rosemont, service d'hématologie et d'oncologie médicale, 5415, boulevard de l'Assomption, Montréal, Québec H1T 2M4, Canada
| | - Cécile Borel
- Institut universitaire du cancer de Toulouse, service d'hématologie, 1, avenue Irène-Joliot-Curie, 31100 Toulouse, France
| | - Nawal Hadhoum
- CHU de Lille, hôpital Roger-Salengro, neurologie D/pathologies neuro-inflammatoires, 59037 Lille cedex, France
| | - Stavroula Masouridi-Levrat
- Hôpitaux universitaires de Genève, département d'oncologie, service d'hématologie, 4, rue Gabrielle-Perret-Gentil, 1205 Genève, Suisse
| | - Jérôme Naudin
- AP-HP, hôpital Robert-Debré, service de réanimation et surveillance continue pédiatrique, 48, boulevard Serrurier, 75019 Paris, France
| | | | - Marie Ouachée-Chardin
- IHOPe, service d'immuno-hématologie pédiatrie, 1, place Joseph-Renault, 69008 Lyon, France
| | - Lara Platon
- CHU Lapeyronie, service de réanimation médicale et médecine intensive, 371, avenue du Doyen-Gaston-Giraud, 34090 Montpellier, France
| | - Asmaa Quessar
- CHU Ibn Rochd, hôpital 20-Août, service d'hématologie et d'oncologie pédiatrique, rue Lahcen Al Arjoun, Casablanca 20000, Maroc
| | | | - Davis Beauvais
- CHU de Lille, maladies du sang, 2, avenue Oscar-Lambret, 59037 Lille, France
| | - André Baruchel
- AP-HP, hôpital universitaire Robert-Debré, hématologie-immunologie pédiatrique, 48, boulevard Sérurier, 75935 Paris cedex 19, France
| | - Jérôme Cornillon
- Institut de cancérologie Lucien-Neuwirth, département d'hématologie clinique, 108 Bis, avenue Albert-Raimond, 42271 Saint-Priest-en-Jarez, France.
| |
Collapse
|