1
|
Guimarães JG, de Campos GY, Machado MP, Oliveira Brito PKM, dos Reis TF, Goldman GH, Bonini Palma PV, de Campos Fraga-Silva TF, Cavallin DCU, Venturini J, da Silva TA. A novel mannan-specific chimeric antigen receptor M-CAR redirects T cells to interact with Candida spp. hyphae and Rhizopus oryzae spores. Bioengineered 2025; 16:2458786. [PMID: 39891522 PMCID: PMC11792852 DOI: 10.1080/21655979.2025.2458786] [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: 04/05/2024] [Revised: 07/24/2024] [Accepted: 08/28/2024] [Indexed: 02/03/2025] Open
Abstract
Invasive fungal infections (IFIs) are responsible for elevated rates of morbidity and mortality, causing around of 1.5 million deaths annually worldwide. One of the main causative agents of IFIs is Candida albicans, and non-albicans Candida species have emerged as a spreading global public health concernment. Furthermore, COVID-19 has contributed to a boost in the incidence of IFIs, such as mucormycosis, in which Rhizopus oryzae is the most prevalent causative agent. The effector host immune response against IFIs depends on the activity of T cells, which are susceptible to the regulatory effects triggered by fungal virulence factors. The fungal cell wall plays a crucial role as a virulence factor, and its remodeling compromises the development of a specific T-cell response. The redirection of Jurkat T cells to target Candida spp. by recognizing targets expressed on the fungal cell wall can be facilitated using chimeric antigen receptor (CAR) technology. This study generated an M-CAR that contains an scFv with specificity to α-1,6 mannose backbone of fungal mannan, and the expression of M-CAR on the surface of modified Jurkat cells triggered a strong activation against Candida albicans (hyphae form), Candida tropicalis (hyphae form), Candida parapsilosis (pseudohyphal form), and Candida glabrata (yeast form). Moreover, M-CAR Jurkat cells recognized Rhizopus oryzae spores, which induced high expression of cell activation markers. Thus, a novel Mannan-specific CAR enabled strong signal transduction in modified Jurkat cells in the presence of Candida spp. or R. oryzae.
Collapse
Affiliation(s)
- Júlia Garcia Guimarães
- Department of Cellular and Molecular Biology, Ribeirao Preto Medical School, University of São Paulo, São Paulo, Brazil
- Department of Clinical Analysis, School of Pharmaceutical Sciences in Araraquara, Sao Paulo State University, São Paulo, Brazil
| | - Gabriela Yamazaki de Campos
- Department of Cellular and Molecular Biology, Ribeirao Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Michele Procópio Machado
- Department of Cellular and Molecular Biology, Ribeirao Preto Medical School, University of São Paulo, São Paulo, Brazil
| | | | | | | | - Patricia Vianna Bonini Palma
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | | | | | - James Venturini
- Faculty of Medicine, Federal University of Mato Grosso do Sul, Campo Grande, Brazil
| | - Thiago Aparecido da Silva
- Department of Cellular and Molecular Biology, Ribeirao Preto Medical School, University of São Paulo, São Paulo, Brazil
- Department of Clinical Analysis, School of Pharmaceutical Sciences in Araraquara, Sao Paulo State University, São Paulo, Brazil
| |
Collapse
|
2
|
Qin H, Zhou Z, Shi R, Mai Y, Xu Y, Peng F, Cheng G, Zhang P, Chen W, Chen Y, Chen Y, Xu R, Lu Q. Insights into next-generation immunotherapy designs and tools: molecular mechanisms and therapeutic prospects. J Hematol Oncol 2025; 18:62. [PMID: 40483473 PMCID: PMC12145627 DOI: 10.1186/s13045-025-01701-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/11/2025] [Indexed: 06/11/2025] Open
Abstract
Immunotherapy has revolutionized the oncology treatment paradigm, and CAR-T cell therapy in particular represents a significant milestone in treating hematological malignancies. Nevertheless, tumor resistance due to target heterogeneity or mutation remains a Gordian knot for immunotherapy. This review elucidates molecular mechanisms and therapeutic potential of next-generation immunotherapeutic tools spanning genetically engineered immune cells, multi-specific antibodies, and cell engagers, emphasizing multi-targeting strategies to enhance personalized immunotherapy efficacy. Development of logic gate modulation-based circuits, adapter-mediated CARs, multi-specific antibodies, and cell engagers could minimize adverse effects while recognizing tumor signals. Ultimately, we highlight gene delivery, gene editing, and other technologies facilitating tailored immunotherapy, and discuss the promising prospects of artificial intelligence in gene-edited immune cells.
Collapse
Affiliation(s)
- Hongzhuo Qin
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zhaokai Zhou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Run Shi
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yumiao Mai
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yudi Xu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Fu Peng
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, School of Pharmacy, Sichuan University, Chengdu, 610041, West China, China
| | - Guangyang Cheng
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wenjie Chen
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Yun Chen
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Yajun Chen
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, 410011, People's Republic of China
- Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, People's Republic of China
| | - Ran Xu
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, People's Republic of China.
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China.
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, 410011, People's Republic of China.
| |
Collapse
|
3
|
Park B, Kim J, Baylink DJ, Hino C, Kwon C, Tran V, Xiao J, Cao H, Lee S, Tan L, Chang A, Saca L, Matus M, Lobo Moreno P, Schill-Depew A, Abdel-Azim H, Mirshahidi H, Xu Y. Nutrient-gene therapy as a strategy to enhance CAR T cell function and overcome barriers in the tumor microenvironment. J Transl Med 2025; 23:633. [PMID: 40481543 PMCID: PMC12144745 DOI: 10.1186/s12967-025-06606-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 05/12/2025] [Indexed: 06/11/2025] Open
Abstract
Cancer immunotherapy is transforming the treatment landscape of both hematological and solid cancers. Although T-cell-based adoptive cell transfer (ACT) therapies have demonstrated initial success, several recurrent obstacles limit their long-term anti-tumor efficacy, including: (1) lack of antigen specificity; (2) poor long-term survival of transplanted T cells in vivo; and (3) a hostile tumor microenvironment (TME). While numerous approaches have been explored to enhance the antigen specificity of Chimeric Antigen Receptor (CAR) T-cell therapies, the field still lacks an effective strategy to optimize the long-term retention and in vivo expansion of engrafted T cells within the TME-a critical factor for the durable efficacy of T-cell-based immunotherapies for both blood and solid cancers. Here, we hypothesize that the success of CAR T-cell therapy can be enhanced by targeting donor T cells' ability to compete with cancer cells for key nutrients, thereby overcoming T-cell exhaustion and sustaining durable anti-tumor function in the TME. To explore this hypothesis, we first provide a comprehensively review of the current understanding of the metabolic interactions (e.g., glucose metabolism) between T cells and tumor cells. To address the challenges, we propose an innovative strategy: utilizing nutrient gene therapy (genetic overexpression of glucose transporter 1, GLUT1) to fortify the metabolic competency of adoptive CAR T-cells, deprive tumors of critical metabolites and ATP, and disrupt the TME. Altogether, our proposed approach combining precision medicine (adoptive CAR T-cell therapy) with tumor metabolism-targeting strategies offers a promising and cost-effective solution to enhance the efficacy and durability of ACT therapies, ultimately improving outcomes for cancer patients.
Collapse
Affiliation(s)
- Brandon Park
- Division of Discovery, Innovation and Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA
| | - Joshua Kim
- Division of Discovery, Innovation and Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA
| | - David J Baylink
- Division of Discovery, Innovation and Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA
| | - Christopher Hino
- Division of Discovery, Innovation and Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA
| | - Cedric Kwon
- Division of Discovery, Innovation and Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA
| | - Victoria Tran
- Division of Discovery, Innovation and Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA
| | - Jeffrey Xiao
- Division of Discovery, Innovation and Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA
| | - Huynh Cao
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA
- Loma Linda University Cancer Center, Loma Linda, CA, 92354, USA
| | - Scott Lee
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA
| | - Laren Tan
- Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA
| | - Andrew Chang
- Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA
| | - Luis Saca
- Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA
| | - Michael Matus
- Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA
| | - Pamela Lobo Moreno
- Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA
| | - Amy Schill-Depew
- Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA
| | - Hisham Abdel-Azim
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA
- Loma Linda University Cancer Center, Loma Linda, CA, 92354, USA
| | - Hamid Mirshahidi
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA
- Loma Linda University Cancer Center, Loma Linda, CA, 92354, USA
| | - Yi Xu
- Division of Discovery, Innovation and Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA.
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA92354, USA.
- Loma Linda University Cancer Center, Loma Linda, CA, 92354, USA.
| |
Collapse
|
4
|
Peng JJ, Ding JY, Xu Y, Shih HP, Lin YN, Wu TY, Lo YF, Lo CC, Yeh CF, Kuo CY, Tu KH, Wang SY, Lei WT, Wu TS, Lin HS, Lee CH, Huang WC, Chen YC, Liu YM, Shi ZY, Chang YT, Syue LS, Chen PL, Teh SH, Chou CH, Ho MW, Chi CY, Ho PC, Ku CL. Chimeric autoantibody receptor T cells clonally eliminate B cells producing autoantibodies against IFN-γ. Sci Immunol 2025; 10:eadm8186. [PMID: 40344086 DOI: 10.1126/sciimmunol.adm8186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/04/2024] [Accepted: 03/27/2025] [Indexed: 05/11/2025]
Abstract
Neutralizing anti-interferon-γ (IFN-γ) autoantibodies (nAIGAs) impair IFN-γ-mediated immunity, predisposing patients with nAIGAs to infection by nontuberculous mycobacteria, Talaromyces marneffei, and other intracellular pathogens. Current clinical management relies on continuous antimicrobial therapy, with no treatment offering sustained benefits. Here, we developed human chimeric autoantibody receptor (CAAR) T cells targeting autoreactive B cells expressing nAIGA B cell receptors (BCRs) using an IFN-γ receptor-irresponsive IFN-γ variant as bait. By exploiting a mouse model of nAIGA BCR-expressing B cell leukemia, we found that IFN-γ CAAR T cells lack off-target toxicity, including IFN-γ receptor cross-reactive toxicity and Fc-redirected toxicity. IFN-γ CAAR T cells substantially reduced circulating AIGAs secreted from target cells in vivo. Further, IFN-γ CAAR T cells effectively eliminated autoreactive B cells in ex vivo cultures of peripheral blood mononuclear cells from patients with nAIGAs. Together, these results demonstrate that IFN-γ CAAR T cells may be a promising strategy to ameliorate nAIGA-associated infections by eliminating autoreactive B cells.
Collapse
Affiliation(s)
- Jhan-Jie Peng
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
- Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
| | - Jing-Ya Ding
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Yingxi Xu
- Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
| | - Han-Po Shih
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - You-Ning Lin
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Tsai-Yi Wu
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Fang Lo
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Chi Lo
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Chu-Fu Yeh
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
- Division of Infectious Diseases, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chen-Yen Kuo
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
- Division of Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kun-Hua Tu
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
- Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shang-Yu Wang
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
- Division of General Surgery, Department of Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Wei-Te Lei
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
- Department of Pediatrics, Division of Pediatric Allergy, Immunology, and Rheumatology, Hsinchu Municipal MacKay Children's Hospital, Hsinchu, Taiwan
| | - Ting-Shu Wu
- Division of Infectious Diseases, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Huang-Shen Lin
- Division of Infectious Diseases, Department of Internal Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Chen-Hsiang Lee
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Wen-Chi Huang
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yi-Chun Chen
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yuag-Meng Liu
- Division of Infectious Diseases, Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Zhi-Yuan Shi
- Division of Infectious Diseases, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ya-Ting Chang
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Ling-Shan Syue
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Po-Lin Chen
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Soon-Hian Teh
- Division of Infectious Diseases, Department of Internal Medicine, Hualien Tzu Chi Hospital, Hualien, Taiwan
| | - Chia-Huei Chou
- Division of Infectious Diseases, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Mao-Wang Ho
- Division of Infectious Diseases, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Yu Chi
- Division of Infectious Diseases, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Ping-Chih Ho
- Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
| | - Cheng-Lung Ku
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
- Division of Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Elixiron Immunotherapeutics, Taipei, Taiwan
| |
Collapse
|
5
|
Lee SS, Martinez Peña EG, Willis AA, Wang CC, Haddad NR, Garza LA. Cell Therapy and the Skin: Great Potential but in Need of Optimization. J Invest Dermatol 2025; 145:1033-1038. [PMID: 39530953 PMCID: PMC12018158 DOI: 10.1016/j.jid.2024.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/21/2024] [Accepted: 09/09/2024] [Indexed: 11/16/2024]
Abstract
Cell therapy is rapidly growing owing to its therapeutic potential for diseases with currently poor outcomes. Cell therapy encompasses both nonengineered and engineered cells and possesses unique abilities such as sense-and-respond functions and long-term engraftment for persistent curative potential. Cell therapy capabilities have expanded to address a wide spectrum of diseases, and our review is focused on dermatological applications. The use of fibroblasts and keratinocytes as cell therapy has shown promise in skin disorders such as epidermolysis bullosa. Future efforts include testing the ability of fibroblasts to reprogram nonvolar to volar skin to reduce stump dermatoses in patients with limb loss using prosthetics.
Collapse
Affiliation(s)
- Sam S Lee
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | | | - Aiden A Willis
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chen Chia Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nina Rossa Haddad
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Luis A Garza
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
6
|
Cao L, Liu Y, Lin G. Strategies for Altering Delivery Technologies to Optimize CAR Therapy. Int J Mol Sci 2025; 26:3206. [PMID: 40244018 PMCID: PMC11989270 DOI: 10.3390/ijms26073206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has been proven to be an effective strategy for the treatment of hematological malignancies. At present, how to prepare CAR-T cells efficiently, quickly, and safely is one of the urgent problems to be solved. The durability and activity of engineered T cells in solid tumors need to be further improved, and the strategy of T cells penetrating the tumor microenvironment also needs to be improved. In addition, although the problems mainly caused by T-cell biology are being solved, the manufacturing mode and process still need to be improved to ensure that CAR-T cell therapy can be widely used. This paper summarizes some strategies that can improve the efficacy of CAR-T cells.
Collapse
Affiliation(s)
- Lili Cao
- Student Counseling Center, Shandong University, Jinan 250012, China;
| | - Yingying Liu
- School of Pharmaceutical Science, Shandong University, Jinan 250012, China;
| | - Guimei Lin
- School of Pharmaceutical Science, Shandong University, Jinan 250012, China;
| |
Collapse
|
7
|
Dumbuya JS, Zeng C, Deng L, Li Y, Chen X, Ahmad B, Lu J. The impact of rare diseases on the quality of life in paediatric patients: current status. Front Public Health 2025; 13:1531583. [PMID: 40196857 PMCID: PMC11973084 DOI: 10.3389/fpubh.2025.1531583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/07/2025] [Indexed: 04/09/2025] Open
Abstract
Rare diseases, also known as orphan diseases, are a group of disorders that affect a small percentage of the population. Despite individually affecting a small number of people, collectively, they impact millions worldwide. This is particularly significant in paediatric patients, highlighting the global scale of the issue. This review delves into the exact prevalence of rare diseases among children and adolescents and their diverse impact on the quality of life of patients and their families. The review sheds light on the complex interplay of genetic and environmental factors contributing to these conditions and the diagnostic challenges and delays often encountered in identifying and categorising these diseases. It is noted that although there have been significant strides in the field of genomic medicine and the development of orphan drugs, effective treatments remain limited. This necessitates a comprehensive, multidisciplinary approach to management involving various specialities working closely together to provide holistic care. Furthermore, the review addresses the psychosocial and economic burdens faced by families with paediatric patients suffering from rare diseases, highlighting the urgent need for enhanced support mechanisms. Recent technological and therapeutic advancements, including genomic sequencing and personalized medicine, offer promising avenues for improving patient outcomes. Additionally, the review underscores the role of policy and advocacy in advancing research, ensuring healthcare access, and supporting affected families. It emphasises the importance of increased awareness, education, and collaboration among healthcare providers, researchers, policymakers, and patient advocacy groups. It stresses the pivotal role each group plays in improving the diagnosis, treatment, and overall quality of life for paediatric patients with rare diseases.
Collapse
Affiliation(s)
- John Sieh Dumbuya
- Department of Paediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Cizheng Zeng
- Department of Paediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lin Deng
- Department of Paediatrics, The 958 Hospital of the People’s Liberation Army, Chongqing, China
| | - Yuanglong Li
- Hainan Women and Children’s Medical Center, Haikou, China
| | - Xiuling Chen
- Department of Paediatrics, Haikou Affiliated Hospital of Central South University, Xiangya School of Medicine, Haikou, China
| | - Bashir Ahmad
- Department of Paediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jun Lu
- Department of Paediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
8
|
Ji X, Sun Y, Xie Y, Gao J, Zhang J. Advance in chimeric antigen receptor T therapy in autoimmune diseases. Front Immunol 2025; 16:1533254. [PMID: 40103816 PMCID: PMC11913860 DOI: 10.3389/fimmu.2025.1533254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
Autoimmune diseases are a group of diseases in which the body's immune system misrecognizes its own antigens resulting in an abnormal immune response, which can lead to pathological damage to or abnormal functioning of its own tissues. Current treatments are mainly hormones and broad-spectrum immunosuppressants, but these can lead to a decline in the patient's immunity. Chimeric antigen receptor T (CAR-T) Cell therapy has emerged, and now the structure of CAR has changed from first generation to fourth generation of CAR. The significant achievement of CAR-T therapy to B-cell leukemia has also inspired the treatment of autoimmune diseases, and by investigating the mechanisms of different autoimmune diseases, different designs of CAR-T can be used to specifically treat autoimmune diseases. In this review, we will discuss the therapeutic strategies of CAR-T cells in different autoimmune diseases and the limitations of the treatment.
Collapse
Affiliation(s)
- Xiaolan Ji
- Department of Ophthalmology, The Second Affiliated Hospital of Suzhou University, Suzhou, China
| | - Yunfan Sun
- The First Clinical College of Nanjing Medical University, Nanjing, China
| | - Yuyang Xie
- The First Clinical Medicine School, Suzhou Medical College, Soochow University, Suzhou, China
| | - Jianling Gao
- Department of Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Ji Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Suzhou University, Suzhou, China
| |
Collapse
|
9
|
Caël B, Bôle-Richard E, Garnache Ottou F, Aubin F. Chimeric antigen receptor-modified T-cell therapy: Recent updates and challenges in autoimmune diseases. J Allergy Clin Immunol 2025; 155:688-700. [PMID: 39675682 DOI: 10.1016/j.jaci.2024.12.1066] [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: 09/02/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy (CAR-T) has revolutionized the treatment of hematologic malignancies, demonstrating significant clinical efficacy and leading to US Food and Drug Administration approval of several CAR T-cell-based products. This success has prompted exploration of CAR-T in other disease areas, including autoimmune diseases (AIDs). CAR-T targeting B cells has been shown to provide clinical and biological improvements in patients with refractory AIDs. The aim of this review is to discuss promising strategies involving CAR-T in AIDs, such as those targeting B cells and T cells, and to explore new approaches targeting fibroblasts or plasmacytoid dendritic cells. Despite these advances, the application of CAR-T in AIDs faces several unique challenges. The quality and functionality of T cells in patients with AIDs may be compromised as a result of previous treatments and the underlying inflammatory state, affecting the generation and efficacy of CAR-T. In addition, achieving adequate tissue biodistribution and persistence of CAR T cells in affected tissues remains a major challenge. Finally, the high costs associated with T-cell production pose economic problems, particularly in the context of chronic diseases, which are far more numerous than the hematologic diseases for which CAR-Ts have been granted marketing authorization to date. If the indications for CAR-T increase significantly, production costs will have to drop drastically in order to obtain reliable economic models.
Collapse
Affiliation(s)
- Blandine Caël
- Université Marie et Louis Pasteur, INSERM, EFS BFC, UMR1098, Besançon, France; Centre Hospitalier Universitaire (CHU) Besançon, Laboratoire Biologie Médicale, Autoimmunité/Allergologie, Besançon, France.
| | - Elodie Bôle-Richard
- Université Marie et Louis Pasteur, INSERM, EFS BFC, UMR1098, Besançon, France; Franche-Comte' Innov, Bionoveo, Besançon, France
| | | | - François Aubin
- Université Marie et Louis Pasteur, INSERM, EFS BFC, UMR1098, Besançon, France; Service de Dermatologie, CHU Besançon, Besançon, France
| |
Collapse
|
10
|
Perico L, Casiraghi F, Benigni A, Remuzzi G. Is there a place for engineered immune cell therapies in autoimmune diseases? Trends Mol Med 2025:S1471-4914(25)00011-5. [PMID: 39984382 DOI: 10.1016/j.molmed.2025.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 02/23/2025]
Abstract
The ability to engineer immune cells yielded a transformative era in oncology. Early clinical trials demonstrated the efficacy of chimeric antigen receptor (CAR) T cells in resetting the immune system, motivating the expansion of this treatment beyond cancer, including autoimmune conditions. In this review, we discuss the current state of CAR T cell research in autoimmune diseases, examining the main challenges that limit widespread adoption of this therapy, such as complex isolation protocols, stringent immunosuppression, risk of secondary malignancies, and variable efficacy. We also review the studies addressing these limitations by development of off-the-shelf allogeneic CAR T cells, tunable safety systems, and antigen-specific therapies, which hold the potential to improve safety and accessibility of this treatment in clinical practice.
Collapse
Affiliation(s)
- Luca Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy.
| | | | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| |
Collapse
|
11
|
Wang H, Li F, Feng Y, Ma W, Li Y, Zhao X, Wu J, Shi C, Zong L, Li J, Cong J, Wang X. Cbl-b inhibition improves manufacturing efficiency and antitumoral efficacy of anti-CD19 CAR-T cells. Int Immunopharmacol 2025; 147:113971. [PMID: 39752754 DOI: 10.1016/j.intimp.2024.113971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/06/2024] [Accepted: 12/26/2024] [Indexed: 01/29/2025]
Abstract
Chimeric antigen receptor T (CAR-T) cells represent a promising approach for cancer immunotherapy, yet their efficacy is hindered by immunosuppressive signals in the tumor microenvironment. Casitas B-cell lymphoma protein b (Cbl-b) is a key negative regulator of T cell function. This study investigated whether inhibiting Cbl-b enhances the antitumor activity of human CAR-T cells. The Cbl-b inhibitor NX-1607 was shown to significantly improve CAR-T cell production and function. When applied during the manufacturing phase, NX-1607 increased the yield of anti-CD19 CAR-T cells. Treatment during the expansion phase enhanced cytokine secretion and cytotoxic activity. Notably, continuous NX-1607 treatment throughout manufacturing and expansion maximized CAR-T cell yield, cytokine production, and cytotoxicity. In vivo, NX-1607-treated CAR-T cells exhibited superior efficacy against hematological malignancies. These findings highlight Cbl-b as a therapeutic target for enhancing CAR-T cell manufacturing efficiency and antitumor efficacy, underscoring its potential for clinical applications.
Collapse
Affiliation(s)
- Haoqi Wang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, China
| | - Fei Li
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, China
| | - Yuanyuan Feng
- Department of Hematology, Anhui Provincial Cancer Hospital, Hefei, China
| | - Wenqiang Ma
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, China
| | - Yuanhao Li
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, China
| | - Xueqin Zhao
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, China
| | - Jingyi Wu
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, China
| | - Chenxi Shi
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, China
| | - Lu Zong
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jing Li
- School of Life Sciences, Anhui Medical University, Hefei, Anhui 230032, China.
| | - Jingjing Cong
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, China.
| | - Xuefu Wang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, China; Institute of Clinical Immunology, Anhui Medical University, Hefei, Anhui 230032, China.
| |
Collapse
|
12
|
Yin H, Wei X. The design of retroviral vectors used in the CAR-T products, risk management, and future perspective. MedComm (Beijing) 2025; 6:e70067. [PMID: 39866836 PMCID: PMC11758153 DOI: 10.1002/mco2.70067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/27/2024] [Accepted: 12/18/2024] [Indexed: 01/28/2025] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy is a revolutionary approach in cancer treatment. More than 10 CAR-T products have already approved on market worldly wide, and they use either gamma retroviral vectors or lentiviral vectors to deliver the CAR gene. Both vectors have the ability to effectively and persistently integrate the CAR gene into T cells. Despite the advancements in CAR-T therapy, the potential risks associated with the vectors, particularly the risks of the secondary malignancies, still remain as a concern. This article compares the characteristics of gamma retroviral and lentiviral vectors, discusses the development of vector packaging systems, and examines the design of self-inactivating (SIN) vectors. It also addresses the risks of secondary malignancies that might possibly be associated with the retroviral vectors, and the strategies to decrease the risks and increase the safer clinical use of the vectors. This article also discusses the current regulatory landscape and management approaches aiming to mitigate these risks through stringent safety measures and ongoing monitoring. Future perspectives focus on improving the safety profiles of the vectors and broadening their scope of use. The article provides a thorough overview of the most recent research discoveries and regulatory updates in the field of CAR-T therapy, highlighting the significance of a balanced strategy that strikes a balance between innovation and patient safety in the development and implementation of CAR-T therapy.
Collapse
Affiliation(s)
- Huifang Yin
- Office of Pharmaceutical ScienceYangtze River Delta Center for Drug Evaluation and InspectionNational Medical Products AdministrationShanghaiChina
| | - Xuejing Wei
- Office of Pharmaceutical ScienceYangtze River Delta Center for Drug Evaluation and InspectionNational Medical Products AdministrationShanghaiChina
| |
Collapse
|
13
|
Rafiq Z, Kang M, Barsoumian HB, Manzar GS, Hu Y, Leuschner C, Huang A, Masrorpour F, Lu W, Puebla-Osorio N, Welsh JW. Enhancing immunotherapy efficacy with synergistic low-dose radiation in metastatic melanoma: current insights and prospects. J Exp Clin Cancer Res 2025; 44:31. [PMID: 39881333 PMCID: PMC11781074 DOI: 10.1186/s13046-025-03281-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025] Open
Abstract
Recent advances in oncology research have highlighted the promising synergy between low-dose radiation therapy (LDRT) and immunotherapies, with growing evidence highlighting the unique benefits of the combination. LDRT has emerged as a potent tool for stimulating the immune system, triggering systemic antitumor effects by remodeling the tumor microenvironment. Notably, LDRT demonstrates remarkable efficacy even in challenging metastatic sites such as the liver (uveal) and brain (cutaneous), particularly in advanced melanoma stages. The increasing interest in utilizing LDRT for secondary metastatic sites of uveal, mucosal, or cutaneous melanomas underscores its potential efficacy in combination with various immunotherapies. This comprehensive review traverses the journey from laboratory research to clinical applications, elucidating LDRT's immunomodulatory role on the tumor immune microenvironment (TIME) and systemic immune responses. We meticulously examine the preclinical evidence and ongoing clinical trials, throwing light on the promising prospects of LDRT as a complementary therapy in melanoma treatment. Furthermore, we explore the challenges associated with LDRT's integration into combination therapies, addressing crucial factors such as optimal dosage, fractionation, treatment frequency, and synergy with other pharmacological agents. Considering its low toxicity profile, LDRT presents a compelling case for application across multiple lesions, augmenting the antitumor immune response in poly-metastatic disease scenarios. The convergence of LDRT with other disciplines holds immense potential for developing novel radiotherapy-combined modalities, paving the way for more effective and personalized treatment strategies in melanoma and beyond. Moreover, the dose-related toxicities of immunotherapies may be reduced by synergistic amplification of antitumor efficacy with LDRT.
Collapse
Affiliation(s)
- Zahid Rafiq
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 500 W. University Ave, El Paso, TX, 79968, USA
| | - Mingyo Kang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hampartsoum B Barsoumian
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Gohar S Manzar
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yun Hu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Carola Leuschner
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ailing Huang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Fatemeh Masrorpour
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Weiqin Lu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 500 W. University Ave, El Paso, TX, 79968, USA
| | - Nahum Puebla-Osorio
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - James W Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
14
|
Hernández-López A, Olaya-Vargas A, Bustamante-Ogando JC, Meneses-Acosta A. Expanding the Horizons of CAR-T Cell Therapy: A Review of Therapeutic Targets Across Diverse Diseases. Pharmaceuticals (Basel) 2025; 18:156. [PMID: 40005970 PMCID: PMC11858291 DOI: 10.3390/ph18020156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
CAR-T cell therapy has shown promising results in treating malignant hematologic diseases. The principle of this therapy is based on the use of genetically modified T lymphocytes to express a Chimeric Antigen Receptor (CAR) on their membrane that specifically recognizes an antigen predominantly expressed on target cells. The molecular design of the CAR, along with advancements in molecular techniques and the development of "omics", has opened the possibility of discovering new therapeutic targets and thereby expanding the range of diseases treated with CAR-T cells beyond the use of anti-CD19 and anti-BCMA for hematologic cancer. This review summarizes the novel therapeutic targets that are currently used in clinical trials with CAR-T cell therapy on autoimmune diseases and other challenging conditions, such as cardiac fibrosis, and different infections. Additionally, challenges and novel opportunities are discussed for expanding clinical access to this innovative therapy.
Collapse
Affiliation(s)
- Alejandrina Hernández-López
- Laboratorio 7 of Biotecnología Farmacéutica, Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca 62210, Mexico;
- Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT), Mexico City 03940, Mexico
| | - Alberto Olaya-Vargas
- Programa de Trasplante de Células Madre Hematopoyéticas y Terapia Celular, Instituto Nacional de Pediatría, Mexico City 04530, Mexico;
| | - Juan Carlos Bustamante-Ogando
- Laboratorio de Investigación en Inmunodeficiencias y Departamento de Inmunología Clínica, Instituto Nacional de Pediatría, Mexico City 04530, Mexico;
| | - Angélica Meneses-Acosta
- Laboratorio 7 of Biotecnología Farmacéutica, Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca 62210, Mexico;
| |
Collapse
|
15
|
Dingfelder J, Taubmann J, von Heydebrand F, Aigner M, Bergmann C, Knitza J, Park S, Cheng JK, Van Blarcom T, Schett G, Mackensen A, Lutzny-Geier G. Exploring CAR T-Cell Dynamics: Balancing Potent Cytotoxicity and Controlled Inflammation in CAR T-Cells Derived from Systemic Sclerosis and Myositis Patients. Int J Mol Sci 2025; 26:467. [PMID: 39859183 PMCID: PMC11765450 DOI: 10.3390/ijms26020467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
Systemic lupus erythematosus (SLE), systemic sclerosis (SSc), and idiopathic inflammatory myositis (IIM) are autoimmune diseases managed with long-term immunosuppressive therapies. Hu19-CD828Z, a fully human anti-CD19 chimeric antigen receptor (CAR) with a CD28 costimulatory domain, is engineered to potently deplete B-cells. In this study, we manufactured Hu19-CD828Z CAR T-cells from peripheral blood of SLE, IIM, and SSc patients and healthy donors (HDs). CAR-mediated, CD19-specific activity of these cells was evaluated in vitro by assessing cytotoxicity, cytokine release, and proliferation assays in response to autologous CD19+ B-cells, the CD19+ NALM-6 B-cell line, or a CD19- U937 non-B-cell line as targets. The results demonstrated an increased proliferation of Hu19-CD828Z CAR T-cells and dose-dependent cytotoxicity against primary autologous and NALM-6 B-cells compared to non-transduced controls or co-cultures with non-B-cells. Notably, autoimmune-patient-derived CAR T-cells produced lower levels of inflammatory cytokines than healthy-donor-derived CAR T-cells in response to CD19+ B-cell targets. These data support the potential of Hu19-CD828Z and its therapeutic cell product KYV-101 as a therapeutic strategy to achieve deep B-cell depletion in SLE, IIM, and SSc patients, and highlights its promise for broader application in B-cell-driven autoimmune disorders.
Collapse
Affiliation(s)
- Janin Dingfelder
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (J.D.); (F.v.H.); (M.A.); (A.M.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (J.T.); (C.B.); (G.S.)
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Jule Taubmann
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (J.T.); (C.B.); (G.S.)
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Franziska von Heydebrand
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (J.D.); (F.v.H.); (M.A.); (A.M.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (J.T.); (C.B.); (G.S.)
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Michael Aigner
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (J.D.); (F.v.H.); (M.A.); (A.M.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (J.T.); (C.B.); (G.S.)
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Christina Bergmann
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (J.T.); (C.B.); (G.S.)
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Johannes Knitza
- Institute for Digital Medicine, University Hospital Giessen-Marburg, Philipps University Marburg, 35043 Marburg, Germany;
| | - Soo Park
- Kyverna Therapeutics, Emeryville, CA 94608, USA; (S.P.); (J.K.C.); (T.V.B.)
| | - Joseph K. Cheng
- Kyverna Therapeutics, Emeryville, CA 94608, USA; (S.P.); (J.K.C.); (T.V.B.)
| | - Thomas Van Blarcom
- Kyverna Therapeutics, Emeryville, CA 94608, USA; (S.P.); (J.K.C.); (T.V.B.)
| | - Georg Schett
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (J.T.); (C.B.); (G.S.)
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (J.D.); (F.v.H.); (M.A.); (A.M.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (J.T.); (C.B.); (G.S.)
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Gloria Lutzny-Geier
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (J.D.); (F.v.H.); (M.A.); (A.M.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (J.T.); (C.B.); (G.S.)
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| |
Collapse
|
16
|
Ransohoff RM. Selected Aspects of the Neuroimmunology of Cell Therapies for Neurologic Disease: Perspective. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200352. [PMID: 39671535 PMCID: PMC11649171 DOI: 10.1212/nxi.0000000000200352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 10/24/2024] [Indexed: 12/15/2024]
Abstract
Neurologic disease remains a cause of incalculable suffering, a formidable public health burden, and a wilderness of complex biology and medicine. At the same time, advances in basic science, technology, and the clinical development toolkit bring meaningful benefit for patients along with realistic hope for those whose conditions remain inadequately treated. This perspective focuses on cell-based therapies for neurologic disease, with particular emphasis on neuroimmunologic disorders and on the immunologic considerations of cell therapy for nonimmune conditions. I will consider the use of chimeric antigen receptor (CAR)-T effector cells and regulatory T-cell therapies for autoimmune conditions. I will briefly discuss the immune aspects of pluripotent stem cell (PSC)-derived neuronal therapies. With apologies for the omission, we do not discuss mesenchymal stem cells, glial progenitor cells, or CAR-NK cells, primarily for space limitations.
Collapse
|
17
|
Aundhia C, Shah N, Talele C, Zanwar A, Kumari M, Patil S. Enhancing Gene Therapy through Ultradeformable Vesicles for Efficient siRNA Delivery. Pharm Nanotechnol 2025; 13:55-69. [PMID: 38284710 DOI: 10.2174/0122117385271654231215064542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/30/2023] [Accepted: 11/08/2023] [Indexed: 01/30/2024]
Abstract
Gene therapy is a revolutionary approach aimed at treating various diseases by manipulating the expression of specific genes. The composition and formulation of ultra-deformable vesicles play a crucial role in determining their properties and performance as siRNA delivery vectors. In the development of ultra-deformable vesicles for siRNA delivery, careful lipid selection and optimization are crucial for achieving desirable vesicle characteristics and efficient siRNA encapsulation and delivery. The stratum corneum acts as a protective barrier, limiting the penetration of molecules, including siRNA, into the deeper layers of the skin. Ultradeformable vesicles offer a promising solution to overcome this barrier and facilitate efficient siRNA delivery to target cells in the skin. The stratum corneum, the outermost layer of the skin, acts as a significant barrier to the penetration of siRNA.These engineering approaches enable the production of uniform and well-defined vesicles with enhanced deformability and improved siRNA encapsulation efficiency. Looking ahead, advancements in ultra-deformable vesicle design and optimization, along with continued exploration of combination strategies and regulatory frameworks, will further drive the field of ultra-deformable vesicle-based siRNA delivery.
Collapse
Affiliation(s)
- Chintan Aundhia
- Department of Pharmacy, Sumandeep Vidyapeeth Deemed to be University, Piparia, Waghodia, Vadodara, 391760, Gujarat, India
| | - Nirmal Shah
- Department of Pharmacy, Sumandeep Vidyapeeth Deemed to be University, Piparia, Waghodia, Vadodara, 391760, Gujarat, India
| | - Chitrali Talele
- Department of Pharmacy, Sumandeep Vidyapeeth Deemed to be University, Piparia, Waghodia, Vadodara, 391760, Gujarat, India
| | - Aarti Zanwar
- Department of Pharmacy, Sumandeep Vidyapeeth Deemed to be University, Piparia, Waghodia, Vadodara, 391760, Gujarat, India
| | - Mamta Kumari
- Department of Pharmacy, Sumandeep Vidyapeeth Deemed to be University, Piparia, Waghodia, Vadodara, 391760, Gujarat, India
| | - Sapana Patil
- Department of Pharmacy, Sumandeep Vidyapeeth Deemed to be University, Piparia, Waghodia, Vadodara, 391760, Gujarat, India
| |
Collapse
|
18
|
Lescoat A, Ghosh M, Kadauke S, Khanna D. Innovative cell therapies for systemic sclerosis: available evidence and new perspectives. Expert Rev Clin Immunol 2025; 21:29-43. [PMID: 39279565 DOI: 10.1080/1744666x.2024.2402494] [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: 04/30/2024] [Accepted: 09/03/2024] [Indexed: 09/18/2024]
Abstract
INTRODUCTION Systemic sclerosis (SSc) is the rheumatic disease with the highest individual mortality rate with a detrimental impact on quality of life. Cell-based therapies may offer new perspectives for this disease as recent phase I trials support the safety of IV infusion of allogeneic mesenchymal stromal cells in SSc and case reports highlight the potential use of Chimeric Antigen Receptor (CAR)-T cells targeting CD19 in active SSc patients who have not responded to conventional immunosuppressive therapies. AREAS COVERED This narrative review highlights the most recent evidence supporting the use of cellular therapies in SSc as well as their potential mechanisms of action and discusses future perspectives for cell-based therapies in SSc. Medline/PubMed was used to identify the articles of interest, using the keywords 'Cellular therapies,' 'Mesenchymal stromal cells,' 'Chimeric Antigen Receptor' AND 'systemic sclerosis.' Milestones articles reported by the authors were also used. EXPERT OPINION Cellular therapies may represent an opportunity for long-term remission/cure in patients with different autoimmune diseases, including SSc who have not responded to conventional therapies. Multiple ongoing phase I/II trials will provide greater insights into the efficacy and toxicity of cellular therapies.
Collapse
Affiliation(s)
- Alain Lescoat
- Inserm, EHESP, Irset -Institut de Recherche en Santé, Environnement et Travail-UMRS, University of Rennes CHU Rennes, Rennes, France
- Department of Internal Medicine and Clinical Immunology, CHU Rennes, Rennes, France
| | - Monalisa Ghosh
- Blood and Marrow Transplant Program, University of Michigan, Ann Arbor, MI, USA
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Stephan Kadauke
- Division of Transfusion Medicine, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dinesh Khanna
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Scleroderma Program, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
19
|
Wu Z, Wang Y, Jin X, Wang L. Universal CAR cell therapy: Challenges and expanding applications. Transl Oncol 2025; 51:102147. [PMID: 39413693 PMCID: PMC11525228 DOI: 10.1016/j.tranon.2024.102147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 09/20/2024] [Accepted: 09/28/2024] [Indexed: 10/18/2024] Open
Abstract
Chimeric Antigen Receptor (CAR) T cell therapy has gained success in adoptive cell therapy for hematological malignancies. Although most CAR cell therapies in clinical trials or markets remain autologous, their acceptance has been limited due to issues like lengthy manufacturing, poor cell quality, and demanding cost. Consequently, "Off-the-shelf", universal CAR (UCAR) cell therapy has emerged. Current concerns with UCAR therapies revolve around side effects such as graft versus host disease (GVHD) and host versus graft response (HVGR). Preclinical research on UCAR cell therapies aims to enhance efficacy and minimize these side effects. Common approaches involve gene editing techniques to knock out T cell receptor (TCR), human leukocyte antigen (HLA), and CD52 expression to mitigate GVHD and HVGR risks. However, these methods carry drawbacks including potential genotoxicity of the edited cells. Most recently, novel editing techniques, such as epigenetic editing and RNA writer systems, have been developed to reduce the risk of GVHD and HVGR, allowing for multiplex editing at different sites. Additionally, incorporating more cell types into UCAR cell therapies, like T-cell subtypes (DNT, γδT, virus-specific T cells) and NK cells, can efficiently target tumors without triggering side effects. In addition, the limited efficacy of T cells and NK cells against solid tumors is being addressed through CAR-Macrophages. In summary, CAR cell therapy has evolved to accommodate multiple cell types while expanding applications to various diseases, including hematologic malignancies and solid tumors, which holds tremendous growth potential and is promised to improve the lives of more patients in the future.
Collapse
Affiliation(s)
- Ziyu Wu
- Department of Hematology I, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Yifan Wang
- Department of Translational Medicine, Research Ward, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, China.
| | - Xin Jin
- Department of Translational Medicine, Research Ward, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, China.
| | - Luqiao Wang
- Department of Hematology I, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
20
|
Michaels YS, Major MC, Bonham-Carter B, Zhang J, Heydari T, Edgar JM, Siu MM, Greenstreet L, Vilarrasa-Blasi R, Kim S, Castle EL, Forrow A, Ibanez-Rios MI, Zimmerman C, Chung Y, Stach T, Werschler N, Knapp DJHF, Vento-Tormo R, Schiebinger G, Zandstra PW. Tracking the gene expression programs and clonal relationships that underlie mast, myeloid, and T lineage specification from stem cells. Cell Syst 2024; 15:1245-1263.e10. [PMID: 39615483 DOI: 10.1016/j.cels.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 07/03/2024] [Accepted: 11/01/2024] [Indexed: 12/21/2024]
Abstract
T cells develop from hematopoietic progenitors in the thymus and protect against pathogens and cancer. However, the emergence of human T cell-competent blood progenitors and their subsequent specification to the T lineage have been challenging to capture in real time. Here, we leveraged a pluripotent stem cell differentiation system to understand the transcriptional dynamics and cell fate restriction events that underlie this critical developmental process. Time-resolved single-cell RNA sequencing revealed that downregulation of the multipotent hematopoietic program, upregulation of >90 lineage-associated transcription factors, and cell-cycle exit all occur within a highly coordinated developmental window. Gene-regulatory network inference uncovered a role for YBX1 in T lineage specification. We mapped the differentiation cell fate hierarchy using transcribed lineage barcoding and discovered that mast and myeloid potential bifurcate from each other early in hematopoiesis, upstream of T lineage restriction. Our systems-level analyses provide a quantitative, time-resolved model of human T cell fate specification. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Yale S Michaels
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada; Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Matthew C Major
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada; Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Becca Bonham-Carter
- Department of Mathematics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| | - Jingqi Zhang
- Department of Mathematics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| | - Tiam Heydari
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| | - John M Edgar
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| | - Mona M Siu
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| | - Laura Greenstreet
- Department of Mathematics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| | | | - Seungjoon Kim
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Elizabeth L Castle
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| | - Aden Forrow
- Department of Mathematics and Statistics, University of Maine, Orono, ME 04469-5752, USA
| | - M Iliana Ibanez-Rios
- Institut de recherche en immunologie et en cancérologie and Département de pathologie et biologie cellulaire, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Carla Zimmerman
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| | - Yvonne Chung
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| | - Tara Stach
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| | - Nico Werschler
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada; Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - David J H F Knapp
- Institut de recherche en immunologie et en cancérologie and Département de pathologie et biologie cellulaire, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Geoffrey Schiebinger
- Department of Mathematics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada.
| | - Peter W Zandstra
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| |
Collapse
|
21
|
Zhou S, Lin W, Jin X, Niu R, Yuan Z, Chai T, Zhang Q, Guo M, Kim SS, Liu M, Deng Y, Park JB, Choi SI, Shi B, Yin J. CD97 maintains tumorigenicity of glioblastoma stem cells via mTORC2 signaling and is targeted by CAR Th9 cells. Cell Rep Med 2024; 5:101844. [PMID: 39637858 PMCID: PMC11722114 DOI: 10.1016/j.xcrm.2024.101844] [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] [Revised: 08/19/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024]
Abstract
Glioblastoma (GBM) stem cells (GSCs) contribute to poor prognosis in patients with GBM. Identifying molecular markers is crucial for developing targeted therapies. Here, we identify cluster of differentiation 97 (CD97) as an optimal GSC surface antigen for potential targeting by chimeric antigen receptor (CAR) T cell therapy through in vitro antibody screening. CD97 is consistently expressed in all validated patient-derived GSCs and positively correlated with known intracellular GSC markers. Silencing CD97 reduces GSC tumorigenicity-related activities, including self-renewal, proliferation, and tumor progression. Transcriptome analysis reveals that CD97 activates mTORC2, leading to AKT S473 phosphorylation and enhanced expression of the downstream genes ARHGAP1, BZW1, and BZW2. Inhibiting mTORC2 with JR-AB2-011 suppresses GSC tumorigenicity and downstream gene expression. We develop CD97-CAR T helper (Th) 9 cells, which exhibit potent cytotoxic effects in vitro and extend survival in mice. These findings suggest that CD97 is a promising GSC-enriched antigen and that targeting it with CAR Th9 cells offers a potential therapeutic strategy for GBM.
Collapse
MESH Headings
- Glioblastoma/pathology
- Glioblastoma/metabolism
- Glioblastoma/immunology
- Glioblastoma/genetics
- Humans
- Animals
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/immunology
- Signal Transduction
- Mice
- Mechanistic Target of Rapamycin Complex 2/metabolism
- Mechanistic Target of Rapamycin Complex 2/genetics
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/immunology
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Carcinogenesis/pathology
- Carcinogenesis/genetics
- Cell Proliferation
- Cell Line, Tumor
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
- Brain Neoplasms/pathology
- Brain Neoplasms/immunology
- Brain Neoplasms/metabolism
- Brain Neoplasms/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Mice, Inbred NOD
- Immunotherapy, Adoptive/methods
- Gene Expression Regulation, Neoplastic
- GTPase-Activating Proteins/metabolism
- GTPase-Activating Proteins/genetics
- Proto-Oncogene Proteins c-akt/metabolism
Collapse
Affiliation(s)
- Shuchang Zhou
- The Zhongzhou Laboratory for Integrative Biology, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Weiwei Lin
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China; Research Institute, National Cancer Center, Goyang, Gyeonggi 10408, Republic of Korea
| | - Xiong Jin
- The Zhongzhou Laboratory for Integrative Biology, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; School of Pharmacy, Henan University, Kaifeng, Henan 475004, China
| | - Rui Niu
- The Zhongzhou Laboratory for Integrative Biology, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Zheng Yuan
- The Zhongzhou Laboratory for Integrative Biology, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Tianran Chai
- The Zhongzhou Laboratory for Integrative Biology, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Republic of Korea
| | - Qi Zhang
- The Zhongzhou Laboratory for Integrative Biology, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Meixia Guo
- The Zhongzhou Laboratory for Integrative Biology, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Sung Soo Kim
- Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Republic of Korea
| | - Meichen Liu
- The Zhongzhou Laboratory for Integrative Biology, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Republic of Korea
| | - Yilin Deng
- The Zhongzhou Laboratory for Integrative Biology, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Republic of Korea
| | - Jong Bae Park
- Research Institute, National Cancer Center, Goyang, Gyeonggi 10408, Republic of Korea; Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Republic of Korea
| | - Sun Il Choi
- The Zhongzhou Laboratory for Integrative Biology, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; School of Pharmacy, Henan University, Kaifeng, Henan 475004, China.
| | - Bingyang Shi
- The Zhongzhou Laboratory for Integrative Biology, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China.
| | - Jinlong Yin
- The Zhongzhou Laboratory for Integrative Biology, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China; Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
22
|
Honaker Y, Gruber D, Jacobs C, Yu-Hong Cheng R, Patel S, Galvan CZ, Khan IF, Zhou K, Sommer K, Astrakhan A, Cook PJ, James RG, Rawlings DJ. Targeting human plasma cells using regulated BCMA CAR T cells eliminates circulating antibodies in humanized mice. Mol Ther 2024:S1525-0016(24)00817-7. [PMID: 39673129 DOI: 10.1016/j.ymthe.2024.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/22/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024] Open
Abstract
Pathogenic long-lived plasma cells (LLPCs) secrete autoreactive antibodies, exacerbating autoimmune diseases and complicating solid organ transplantation. Targeted elimination of the autoreactive B cell pool represents a promising therapeutic strategy, yet current treatment modalities fall short in depleting mature PCs. Here, we demonstrate that chimeric antigen receptor (CAR) T cells, targeting B cell maturation antigen (BCMA) utilizing a split-receptor design, offer a controlled and effective therapeutic strategy against LLPCs. Dimerizing agent-regulated immune-receptor complex (DARIC) T cells demonstrated robust rapamycin-dependent targeting of tumor and PCs. Notably, in humanized mouse models, DARIC T cells regulated peripheral human immunoglobulin levels through specific elimination of human LLPCs from the bone marrow. Furthermore, DARIC constructs were efficiently integrated into the T cell receptor α constant (TRAC) locus while maintaining potent antigen-specific cytotoxicity. These findings underscore the potential of split-receptor CAR T cells in autoimmune and transplant medicine, highlighting their versatility in applications beyond oncology.
Collapse
Affiliation(s)
- Yuchi Honaker
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - David Gruber
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Chester Jacobs
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Rene Yu-Hong Cheng
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Shivani Patel
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Christopher Zavala Galvan
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Iram F Khan
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Kevin Zhou
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Karen Sommer
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | | | - Peter J Cook
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Richard G James
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98105, USA
| | - David J Rawlings
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98105, USA; Department of Immunology, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
23
|
Bartoli-Leonard F, Pennel T, Caputo M. Immunotherapy in the Context of Aortic Valve Diseases. Cardiovasc Drugs Ther 2024; 38:1173-1185. [PMID: 39017904 PMCID: PMC11680629 DOI: 10.1007/s10557-024-07608-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/08/2024] [Indexed: 07/18/2024]
Abstract
PURPOSE Aortic valve disease (AVD) affects millions of people around the world, with no pharmacological intervention available. Widely considered a multi-faceted disease comprising both regurgitative pathogenesis, in which retrograde blood flows back through to the left ventricle, and aortic valve stenosis, which is characterized by the thickening, fibrosis, and subsequent mineralization of the aortic valve leaflets, limiting the anterograde flow through the valve, surgical intervention is still the main treatment, which incurs considerable risk to the patient. RESULTS Though originally thought of as a passive degeneration of the valve or a congenital malformation that has occurred before birth, the paradigm of AVD is shifting, and research into the inflammatory drivers of valve disease as a potential mechanism to modulate the pathobiology of this life-limiting pathology is taking center stage. Following limited success in mainstay therapeutics such as statins and mineralisation inhibitors, immunomodulatory strategies are being developed. Immune cell therapy has begun to be adopted in the cancer field, in which T cells (chimeric antigen receptor (CAR) T cells) are isolated from the patient, programmed to attack the cancer, and then re-administered to the patient. Within cardiac research, a novel T cell-based therapeutic approach has been developed to target lipid nanoparticles responsible for increasing cardiac fibrosis in a failing heart. With clonally expanded T-cell populations recently identified within the diseased valve, their unique epitope presentation may serve to identify novel targets for the treatment of valve disease. CONCLUSION Taken together, targeted T-cell therapy may hold promise as a therapeutic platform to target a multitude of diseases with an autoimmune aspect, and this review aims to frame this in the context of cardiovascular disease, delineating what is currently known in the field, both clinically and translationally.
Collapse
Affiliation(s)
- Francesca Bartoli-Leonard
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, UK.
- Bristol Heart Institute, University Hospital Bristol and Weston NHS Foundation Trust, Bristol, UK.
- Chris Barnard Division of Cardiothoracic Surgery, University of Cape Town, Cape Town, South Africa.
| | - Tim Pennel
- Chris Barnard Division of Cardiothoracic Surgery, University of Cape Town, Cape Town, South Africa
| | - Massimo Caputo
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, UK
- Bristol Heart Institute, University Hospital Bristol and Weston NHS Foundation Trust, Bristol, UK
| |
Collapse
|
24
|
Chang Y, Chang M, Bao X, Dong C. Advancements in adoptive CAR immune cell immunotherapy synergistically combined with multimodal approaches for tumor treatment. Bioact Mater 2024; 42:379-403. [PMID: 39308543 PMCID: PMC11415837 DOI: 10.1016/j.bioactmat.2024.08.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/26/2024] [Accepted: 08/31/2024] [Indexed: 09/25/2024] Open
Abstract
Adoptive immunotherapy, notably involving chimeric antigen receptor (CAR)-T cells, has obtained Food and Drug Administration (FDA) approval as a treatment for various hematological malignancies, demonstrating promising preclinical efficacy against cancers. However, the intricate and resource-intensive autologous cell processing, encompassing collection, expansion, engineering, isolation, and administration, hamper the efficacy of this therapeutic modality. Furthermore, conventional CAR T therapy is presently confined to addressing solid tumors due to impediments posed by physical barriers, the potential for cytokine release syndrome, and cellular exhaustion induced by the immunosuppressive and heterogeneous tumor microenvironment. Consequently, a strategic integration of adoptive immunotherapy with synergistic multimodal treatments, such as chemotherapy, radiotherapy, and vaccine therapy etc., emerges as a pivotal approach to surmount these inherent challenges. This collaborative strategy holds the key to addressing the limitations delineated above, thereby facilitating the realization of more precise personalized therapies characterized by heightened therapeutic efficacy. Such synergistic strategy not only serves to mitigate the constraints associated with adoptive immunotherapy but also fosters enhanced clinical applicability, thereby advancing the frontiers of therapeutic precision and effectiveness.
Collapse
Affiliation(s)
- Yun Chang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China
| | - Mingyang Chang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
| | - Xiaoping Bao
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Purdue University Institute for Cancer Research, West Lafayette, IN, 47907, USA
| | - Cheng Dong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China
| |
Collapse
|
25
|
Bharadwaj S, Lowsky R, Mikkilineni L, Smith M, Weng WK. A 17-Year Experience of a Large Dedicated Fellowship in Blood and Marrow Transplantation and Cellular Therapy: A Blueprint for Modern Day Training Program. JOURNAL OF CANCER EDUCATION : THE OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER EDUCATION 2024:10.1007/s13187-024-02545-3. [PMID: 39604772 DOI: 10.1007/s13187-024-02545-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
The field of allogeneic blood and marrow transplantation-cellular therapy (BMT-CT) has evolved through incremental advances. Engineered donor grafts, gene editing and chimeric antigen receptor T-cells are all standard clinical practice. Consequently, the scientific knowledge and complexity of clinical skills needed for next generation of BMT-CT physicians have increased. We report a 17-year experience of arguably the largest 12-month BMT-CT clinical fellowship program in the USA. Seventy-three (73) trainees were accepted and 2 cohorts that reflected different time periods (2007-1016 and 2017-2024, inclusive) and different core training curriculum were compared. The cohorts were equivalent in terms of demographics; notably, most (70%) had graduated from international medical schools and trained in the US on a non-immigrant J1 visa. In 2015, we introduced a structured mentoring program to address the desire of trainees for experience with scholarly activities. There was a high rate of successful academic careers with a trend toward a higher likelihood of academic retention following structured mentoring (70% vs 89%). In this report, we included our detailed core curriculum and highlight potential future changes as a blueprint for modern day programs to ensure that graduate "transplant docs" can continue to contribute at the highest academic level.
Collapse
Affiliation(s)
- Sushma Bharadwaj
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA94304, USA
| | - Robert Lowsky
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA94304, USA
| | - Lekha Mikkilineni
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA94304, USA
| | - Melody Smith
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA94304, USA
| | - Wen-Kai Weng
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA94304, USA.
| |
Collapse
|
26
|
Liu J, Zhao Y, Zhao H. Chimeric antigen receptor T-cell therapy in autoimmune diseases. Front Immunol 2024; 15:1492552. [PMID: 39628482 PMCID: PMC11611814 DOI: 10.3389/fimmu.2024.1492552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 10/28/2024] [Indexed: 12/06/2024] Open
Abstract
The administration of T cells that have been modified to carry chimeric antigen receptors (CARs) aimed at B cells has been an effective strategy in treating B cell malignancies. This breakthrough has spurred the creation of CAR T cells intended to specifically reduce or alter the faulty immune responses associated with autoimmune disorders. Early positive outcomes from clinical trials involving CAR T cells that target the B cell protein CD19 in patients suffering from autoimmune diseases driven by B cells have been reported. Additional strategies are being developed to broaden the use of CAR T cell therapy and enhance its safety in autoimmune conditions. These include employing chimeric autoantireceptors (CAAR) to specifically eliminate B cells that are reactive to autoantigens, and using regulatory T cells (Tregs) engineered to carry antigen-specific CARs for precise immune modulation. This discussion emphasizes key factors such as choosing the right target cell groups, designing CAR constructs, defining tolerable side effects, and achieving a lasting immune modification, all of which are critical for safely integrating CAR T cell therapy in treating autoimmune diseases.
Collapse
MESH Headings
- Humans
- Autoimmune Diseases/therapy
- Autoimmune Diseases/immunology
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Animals
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- T-Lymphocytes, Regulatory/immunology
- B-Lymphocytes/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Autoantigens/immunology
- Antigens, CD19/immunology
Collapse
Affiliation(s)
- Jie Liu
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yan Zhao
- Department of Respiratory, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Hai Zhao
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
27
|
Robinson WH, Fiorentino D, Chung L, Moreland LW, Deodhar M, Harler MB, Saulsbery C, Kunder R. Cutting-edge approaches to B-cell depletion in autoimmune diseases. Front Immunol 2024; 15:1454747. [PMID: 39445025 PMCID: PMC11497632 DOI: 10.3389/fimmu.2024.1454747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024] Open
Abstract
B-cell depletion therapy (BCDT) has been employed to treat autoimmune disease for ~20 years. Immunoglobulin G1 (IgG1) monoclonal antibodies targeting CD20 and utilizing effector function (eg, antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, antibody-dependent cellular phagocytosis) to eliminate B cells have historically been the predominant therapeutic approaches. More recently, diverse BCDT approaches targeting a variety of B-cell surface antigens have been developed for use in hematologic malignancies, including effector-function-enhanced monoclonal antibodies, chimeric antigen receptor T-cell (CAR-T) treatment, and bispecific T-cell engagers (TCEs). The latter category of antibodies employs CD3 engagement to augment the killing of target cells. Given the improvement in B-cell depletion observed with CAR-T and TCEs compared with conventional monospecific antibodies for treatment of hematologic malignancies and the recent case reports demonstrating therapeutic benefit of CAR-T in autoimmune disease, there is potential for these mechanisms to be effective for B-cell-mediated autoimmune disease. In this review, we discuss the various BCDTs that are being developed in autoimmune diseases, describing the molecule designs, depletion mechanisms, and potential advantages and disadvantages of each approach as they pertain to safety, efficacy, and patient experience. Additionally, recent advances and strategies with TCEs are presented to help broaden understanding of the potential for bispecific antibodies to safely and effectively engage T cells for deep B-cell depletion in autoimmune diseases.
Collapse
Affiliation(s)
- William H. Robinson
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA, United States
| | - David Fiorentino
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA, United States
| | - Lorinda Chung
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA, United States
- Palo Alto VA Health Care System, Palo Alto, CA, United States
| | - Larry W. Moreland
- Division of Rheumatology, School of Medicine, University of Colorado Anschutz, Aurora, CO, United States
| | | | | | | | | |
Collapse
|
28
|
Cheever A, Kang CC, O’Neill KL, Weber KS. Application of novel CAR technologies to improve treatment of autoimmune disease. Front Immunol 2024; 15:1465191. [PMID: 39445021 PMCID: PMC11496059 DOI: 10.3389/fimmu.2024.1465191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has become an important treatment for hematological cancers, and its success has spurred research into CAR T cell therapies for other diseases, including solid tumor cancers and autoimmune diseases. Notably, the development of CAR-based treatments for autoimmune diseases has shown great progress recently. Clinical trials for anti-CD19 and anti-BCMA CAR T cells in treating severe B cell-mediated autoimmune diseases, like systemic lupus erythematosus (SLE), have shown lasting remission thus far. CAR T cells targeting autoreactive T cells are beginning clinical trials for treating T cell mediated autoimmune diseases. Chimeric autoantigen receptor (CAAR) T cells specifically target and eliminate only autoreactive B cells, and they have shown promise in treating mucosal pemphigus vulgaris and MuSK myasthenia gravis. Regulatory CAR T cells have also been developed, which show potential in altering autoimmune affected areas by creating a protective barrier as well as helping decrease inflammation. These new treatments are only the beginning of potential CAR T cell applications in treating autoimmune disease. Novel CAR technologies have been developed that increase the safety, potency, specificity, and efficacy of CAR T cell therapy. Applying these novel modifications to autoimmune CARs has the potential to enhance the efficacy and applicability of CAR therapies to autoimmune disease. This review will detail several recently developed CAR technologies and discuss how their application to autoimmune disease will improve this emerging field. These include logic-gated CARs, soluble protein-secreting CARs, and modular CARs that enable CAR T cell therapies to be more specific, reach a wider span of target cells, be safer for patients, and give a more potent cytotoxic response. Applying these novel CAR technologies to the treatment of autoimmune diseases has the potential to revolutionize this growing application of CAR T cell therapies.
Collapse
|
29
|
Cao H, Xiao J, Baylink DJ, Nguyen V, Shim N, Lee J, Mallari DJR, Wasnik S, Mirshahidi S, Chen CS, Abdel-Azim H, Reeves ME, Xu Y. Development of a Competitive Nutrient-Based T-Cell Immunotherapy Designed to Block the Adaptive Warburg Effect in Acute Myeloid Leukemia. Biomedicines 2024; 12:2250. [PMID: 39457563 PMCID: PMC11504511 DOI: 10.3390/biomedicines12102250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Background: T-cell-based adoptive cell therapies have emerged at the forefront of cancer immunotherapies; however, failed long-term survival and inevitable exhaustion of transplanted T lymphocytes in vivo limits clinical efficacy. Leukemia blasts possess enhanced glycolysis (Warburg effect), exploiting their microenvironment to deprive nutrients (e.g., glucose) from T cells, leading to T-cell dysfunction and leukemia progression. Methods: Thus, we explored whether genetic reprogramming of T-cell metabolism could improve their survival and empower T cells with a competitive glucose-uptake advantage against blasts and inhibit their uncontrolled proliferation. Results: Here, we discovered that high-glucose concentration reduced the T-cell expression of glucose transporter GLUT1 (SLC2A1) and TFAM (mitochondrion transcription factor A), an essential transcriptional regulator of mitochondrial biogenesis, leading to their impaired expansion ex vivo. To overcome the glucose-induced genetic deficiency in metabolism, we engineered T cells with lentiviral overexpression of SLC2A1 and/or TFAM transgene. Multi-omics analyses revealed that metabolic reprogramming promoted T-cell proliferation by increasing IL-2 release and reducing exhaustion. Moreover, the engineered T cells competitively deprived glucose from allogenic blasts and lessened leukemia burden in vitro. Conclusions: Our findings propose a novel T-cell immunotherapy that utilizes a dual strategy of starving blasts and cytotoxicity for preventing uncontrolled leukemia proliferation.
Collapse
Affiliation(s)
- Huynh Cao
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Jeffrey Xiao
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - David J. Baylink
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Vinh Nguyen
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Nathan Shim
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Jae Lee
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Dave J. R. Mallari
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Samiksha Wasnik
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Saied Mirshahidi
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
- Biospecimen Laboratory, Department of Medicine and Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Chien-Shing Chen
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Hisham Abdel-Azim
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Division of Transplant and Cell Therapy, Loma Linda University Cancer Center, Loma Linda, CA 92354, USA
- Division of Hematology and Oncology, Department of Pediatrics, Loma Linda University, Loma Linda, CA 92354, USA
| | - Mark E. Reeves
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Yi Xu
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| |
Collapse
|
30
|
Abdulla A, Abudureheman T, Chen K, Ghalandari B, Yan H, Zhou H, Su H, Zhang Y, Duan CW, Ding X. Efficient Capture and Traceless Release of Functional CD8 + T Cells with a Microfluidic Chip for Enhanced In Vitro and In Vivo CD4-CAR Transduction. Anal Chem 2024. [PMID: 39269432 DOI: 10.1021/acs.analchem.4c03135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
The chimeric antigen receptor (CAR) T cells targeting CD4 expressed cells in acute lymphoblastic leukemia (T-ALL) and acute myeloid leukemia (AML) could reduce the risk of off target effects in normal tissues. However, the efficacy of adoptive cell therapy is predominantly attributed to CD8+ T cells, necessitating their purification before lentivirus transfection to enhance the production of CD4-CAR-T cells. In this study, we developed a microfluidic chip functionalized with an optimized CD8 aptamer, A3t-MU, to facilitate the enrichment and purification of CD8+ T cells. The presented chip showed efficient capture and seamless release of CD8+ T cells from cultured T cells and peripheral blood mononuclear cells (PBMCs). The purity of the released CD8+ T cells reached 98.1%, representing a 13% improvement over the conventional magnetic bead separation method. CD4-CAR was efficiently transduced into the purified CD8+ T cells to construct CAR-T cells. We evaluated the antitumor capability of the CD4-CAR transduced CD8+ T cells (anti-CD4 CD8-CAR T cells) both in vitro and in vivo. The anti-CD4 CD8-CAR T cells exhibited significant cancer-cell-killing capacity across multiple tumor cell lines, including CEM, Jurkat, and MV4-11. Meanwhile, anti-CD4 CD8-CAR T cells significantly inhibited tumor growth in vivo. In conclusion, the presented microfluidic chip offers a cost-effective and high-purity approach for CD8+ T cell separation, enhancing CD4-CAR transduction and achieving efficient antitumor capability both in vitro and in vivo.
Collapse
Affiliation(s)
- Aynur Abdulla
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200092, China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Tuersunayi Abudureheman
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Kaiming Chen
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Behafarid Ghalandari
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Haoni Yan
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Hang Zhou
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Hengxing Su
- Department of Neurosurgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200092, China
| | - Yunqian Zhang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Cai-Wen Duan
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Xianting Ding
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200092, China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
31
|
Yang L, Wu M, Yang H, Sun X, Xing L, Liu D, Xing L, Yu J. Case report: Bridging radiation therapy before chimeric antigen receptor T-cell therapy induces sustained remission in patients with relapsed/refractory double-expressor diffuse large B-cell lymphoma with localized compressive symptoms. Front Immunol 2024; 15:1441404. [PMID: 39290703 PMCID: PMC11405209 DOI: 10.3389/fimmu.2024.1441404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Background High-risk double-expressor diffuse large B-cell lymphoma has an inferior prognosis following standard first-line therapy. After failure of second-line therapy, treatment options are limited if accompanied by localized compressive symptoms. Chimeric Antigen Receptor T cell (CAR-T) therapy preceded by bridging radiotherapy may be an effective emerging therapy. Case presentation We report a 66-year-old female patient diagnosed with stage IV double-expressor diffuse large B-cell lymphoma. The patient achieved progressive disease after two cycles of rituximab, cyclophosphamide, liposomal doxorubicin, vincristine, and prednisone and continued to develop cervical lymph node recurrence after second-line therapy. The patient was infused with CAR-T cells after receiving focal bridging radiotherapy and remained in complete response more than 9 months after treatment. In addition, the patients did not experience serious adverse reactions related to radiotherapy as well as CAR-T cell therapy. Conclusions In this article, we describe a patient with double-expressor diffuse large B-cell lymphoma with localized compression symptoms after second-line treatment failure who benefited from CAR-T combined with focal bridging radiotherapy.
Collapse
Affiliation(s)
- Liying Yang
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mengdi Wu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Graduate, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Hao Yang
- Department of Graduate, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiaorong Sun
- Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Lijie Xing
- Department of Hematology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Dan Liu
- Department of Hematology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ligang Xing
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jinming Yu
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
32
|
Li J, Chen P, Ma W. The next frontier in immunotherapy: potential and challenges of CAR-macrophages. Exp Hematol Oncol 2024; 13:76. [PMID: 39103972 PMCID: PMC11302330 DOI: 10.1186/s40164-024-00549-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/30/2024] [Indexed: 08/07/2024] Open
Abstract
Chimeric antigen receptor macrophage (CAR-MΦ) represents a significant advancement in immunotherapy, especially for treating solid tumors where traditional CAR-T therapies face limitations. CAR-MΦ offers a promising approach to target and eradicate tumor cells by utilizing macrophages' phagocytic and antigen-presenting abilities. However, challenges such as the complex tumor microenvironment (TME), variability in antigen expression, and immune suppression limit their efficacy. This review addresses these issues, exploring mechanisms of CAR-MΦ action, optimal construct designs, and interactions within the TME. It also delves into the ex vivo manufacturing challenges of CAR-MΦ, discussing autologous and allogeneic sources and the importance of stringent quality control. The potential synergies of integrating CAR-MΦ with existing cancer therapies like checkpoint inhibitors and conventional chemotherapeutics are examined to highlight possible enhanced treatment outcomes. Furthermore, regulatory pathways for CAR-MΦ therapies are scrutinized alongside established protocols for CAR-T cells, identifying unique considerations essential for clinical trials and market approval. Proposed safety monitoring frameworks aim to manage potential adverse events, such as cytokine release syndrome, crucial for patient safety. Consolidating current research and clinical insights, this review seeks to refine CAR-MΦ therapeutic applications, overcome barriers, and suggest future research directions to transition CAR-MΦ therapies from experimental platforms to standard cancer care options.
Collapse
Affiliation(s)
- Jing Li
- The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Ping Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Union Hospital, Fujian Medical University Fuzhou, Fujian, 350001, China
| | - Wenxue Ma
- Sanford Stem Cell Institute, Moores Cancer Center, University of California San Diego, CA, 92093, La Jolla, USA.
| |
Collapse
|
33
|
Soroudi S, Jaafari MR, Arabi L. Lipid nanoparticle (LNP) mediated mRNA delivery in cardiovascular diseases: Advances in genome editing and CAR T cell therapy. J Control Release 2024; 372:113-140. [PMID: 38876358 DOI: 10.1016/j.jconrel.2024.06.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/16/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of global mortality among non-communicable diseases. Current cardiac regeneration treatments have limitations and may lead to adverse reactions. Hence, innovative technologies are needed to address these shortcomings. Messenger RNA (mRNA) emerges as a promising therapeutic agent due to its versatility in encoding therapeutic proteins and targeting "undruggable" conditions. It offers low toxicity, high transfection efficiency, and controlled protein production without genome insertion or mutagenesis risk. However, mRNA faces challenges such as immunogenicity, instability, and difficulty in cellular entry and endosomal escape, hindering its clinical application. To overcome these hurdles, lipid nanoparticles (LNPs), notably used in COVID-19 vaccines, have a great potential to deliver mRNA therapeutics for CVDs. This review highlights recent progress in mRNA-LNP therapies for CVDs, including Myocardial Infarction (MI), Heart Failure (HF), and hypercholesterolemia. In addition, LNP-mediated mRNA delivery for CAR T-cell therapy and CRISPR/Cas genome editing in CVDs and the related clinical trials are explored. To enhance the efficiency, safety, and clinical translation of mRNA-LNPs, advanced technologies like artificial intelligence (AGILE platform) in RNA structure design, and optimization of LNP formulation could be integrated. We conclude that the strategies to facilitate the extra-hepatic delivery and targeted organ tropism of mRNA-LNPs (SORT, ASSET, SMRT, and barcoded LNPs) hold great prospects to accelerate the development and translation of mRNA-LNPs in CVD treatment.
Collapse
Affiliation(s)
- Setareh Soroudi
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
34
|
Abedini A, Levinsohn J, Klötzer KA, Dumoulin B, Ma Z, Frederick J, Dhillon P, Balzer MS, Shrestha R, Liu H, Vitale S, Bergeson AM, Devalaraja-Narashimha K, Grandi P, Bhattacharyya T, Hu E, Pullen SS, Boustany-Kari CM, Guarnieri P, Karihaloo A, Traum D, Yan H, Coleman K, Palmer M, Sarov-Blat L, Morton L, Hunter CA, Kaestner KH, Li M, Susztak K. Single-cell multi-omic and spatial profiling of human kidneys implicates the fibrotic microenvironment in kidney disease progression. Nat Genet 2024; 56:1712-1724. [PMID: 39048792 PMCID: PMC11592391 DOI: 10.1038/s41588-024-01802-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 05/15/2024] [Indexed: 07/27/2024]
Abstract
Kidneys are intricate three-dimensional structures in the body, yet the spatial and molecular principles of kidney health and disease remain inadequately understood. We generated high-quality datasets for 81 samples, including single-cell, single-nuclear, spot-level (Visium) and single-cell resolution (CosMx) spatial-RNA expression and single-nuclear open chromatin, capturing cells from healthy, diabetic and hypertensive diseased human kidneys. Combining these data, we identify cell types and map them to their locations within the tissue. Unbiased deconvolution of the spatial data identifies the following four distinct microenvironments: glomerular, immune, tubule and fibrotic. We describe the complex organization of microenvironments in health and disease and find that the fibrotic microenvironment is able to molecularly classify human kidneys and offers an improved prognosis compared to traditional histopathology. We provide a comprehensive spatially resolved molecular roadmap of the human kidney and the fibrotic process, demonstrating the clinical utility of spatial transcriptomics.
Collapse
Affiliation(s)
- Amin Abedini
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Jonathan Levinsohn
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Konstantin A Klötzer
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Bernhard Dumoulin
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Ziyuan Ma
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Julia Frederick
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Poonam Dhillon
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael S Balzer
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Nephrology, Charité - Universitätsmedizin, Berlin, Germany
| | - Rojesh Shrestha
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Hongbo Liu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Steven Vitale
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Andi M Bergeson
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Paola Grandi
- Genomic Sciences, GSK-Cellzome, Heidelberg, Germany
| | | | - Erding Hu
- Research and Development, GSK, Crescent Drive, Philadelphia, PA, USA
| | - Steven S Pullen
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | - Carine M Boustany-Kari
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | - Paolo Guarnieri
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | | | - Daniel Traum
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Hanying Yan
- Department of Epidemiology, Biostatistics and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kyle Coleman
- Department of Epidemiology, Biostatistics and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Matthew Palmer
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Lea Sarov-Blat
- Research and Development, GSK, Crescent Drive, Philadelphia, PA, USA
| | - Lori Morton
- Cardiovascular and Renal Research, Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Christopher A Hunter
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Klaus H Kaestner
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Mingyao Li
- Department of Epidemiology, Biostatistics and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
35
|
Olson EN. Myocardial Regeneration: Feasible or Fantasy? Circulation 2024; 150:347-349. [PMID: 39074179 PMCID: PMC11335024 DOI: 10.1161/circulationaha.124.070136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Affiliation(s)
- Eric N Olson
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
36
|
Ramírez-Valle F, Maranville JC, Roy S, Plenge RM. Sequential immunotherapy: towards cures for autoimmunity. Nat Rev Drug Discov 2024; 23:501-524. [PMID: 38839912 DOI: 10.1038/s41573-024-00959-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2024] [Indexed: 06/07/2024]
Abstract
Despite major progress in the treatment of autoimmune diseases in the past two decades, most therapies do not cure disease and can be associated with increased risk of infection through broad suppression of the immune system. However, advances in understanding the causes of autoimmune disease and clinical data from novel therapeutic modalities such as chimeric antigen receptor T cell therapies provide evidence that it may be possible to re-establish immune homeostasis and, potentially, prolong remission or even cure autoimmune diseases. Here, we propose a 'sequential immunotherapy' framework for immune system modulation to help achieve this ambitious goal. This framework encompasses three steps: controlling inflammation; resetting the immune system through elimination of pathogenic immune memory cells; and promoting and maintaining immune homeostasis via immune regulatory agents and tissue repair. We discuss existing drugs and those in development for each of the three steps. We also highlight the importance of causal human biology in identifying and prioritizing novel immunotherapeutic strategies as well as informing their application in specific patient subsets, enabling precision medicine approaches that have the potential to transform clinical care.
Collapse
|
37
|
Elsallab M, Bourgeois F, Maus MV. National Survey of FACT-Accredited Cell Processing Facilities: Assessing Preparedness for Local Manufacturing of Immune Effector Cells. Transplant Cell Ther 2024; 30:626.e1-626.e11. [PMID: 38494077 DOI: 10.1016/j.jtct.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/05/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
The utilization of the human immune system as a therapeutic modality has materialized in the form of novel biologics known as immune effector cells (IECs). However, currently approved IECs rely on autologous cells for manufacturing that are funneled through costly centralized supply chains leading to long wait times and potentially increased mortality. Alternative models for manufacturing at or near the point-of-care in a distributed and local approach are being proposed to overcome such a bottleneck. Cell processing facilities for minimally manipulated products, as well as academic good manufacturing practice facilities, are being considered for such manufacturing tasks. However, the infrastructure and the practices of these facilities remains unstudied. Here, we surveyed the cell processing facilities accredited by the Foundation for Accreditation of Cellular Therapy (FACT) in the United States to better understand their preparedness for local manufacturing of IECs. A structured survey consisting of 40 items was distributed to the directors of 157 facilities. The survey evaluated 6 domains, including facility characteristics, quality practices, personnel, use of automation, experience with IECs, and the perception of the point-of-care model. Thirty-eight facilities completed the survey (24.2%). Most facilities were involved in handling IEC products (35/38, 92.1%), and the majority had infrastructure to support basic operations and quality control such as viability (36/36, 100%), identity (33/36, 91.7%), and sterility (33/36, 91.7%). The quality practices varied among the facilities depending on the types of products processed. A slight majority implemented automation in their workflows (22/38, 57.9%). Facilities expressed a general interest in adopting point-of-care models (23/38, 61%), with financial and human resources identified as the most significant constraints. In conclusion, FACT-accredited cell processing facilities may provide the infrastructure required for local manufacturing. However, there is a need for standardization and minimum quality requirements to effectively implement such models.
Collapse
Affiliation(s)
- Magdi Elsallab
- Harvard-MIT Center for Regulatory Science, Harvard Medical School, Boston, Massachusetts; Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Florence Bourgeois
- Harvard-MIT Center for Regulatory Science, Harvard Medical School, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Computational Health Informatics Program, Boston Children's Hospital, Boston, Massachusetts; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Cancer Center, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
38
|
Yang Z, Liu Y, Zhao H. CAR T treatment beyond cancer: Hope for immunomodulatory therapy of non-cancerous diseases. Life Sci 2024; 344:122556. [PMID: 38471620 DOI: 10.1016/j.lfs.2024.122556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/28/2024] [Accepted: 03/07/2024] [Indexed: 03/14/2024]
Abstract
Engineering a patient's own T cells to accurately identify and eliminate cancer cells has effectively cured individuals afflicted with previously incurable hematologic cancers. These findings have stimulated research into employing chimeric antigen receptor (CAR) T therapy across various areas within the field of oncology. However, evidence from both clinical and preclinical investigations emphasize the broader potential of CAR T therapy, extending beyond oncology to address autoimmune disorders, persistent infections, cardiac fibrosis, age-related ailments and other conditions. Concurrently, the advent of novel technologies and platforms presents additional avenues for utilizing CAR T therapy in non-cancerous contexts. This review provides an overview of the rationale behind CAR T therapy, delineates ongoing challenges in its application to cancer treatment, summarizes recent findings in non-cancerous diseases, and engages in discourse regarding emerging technologies that bear relevance. The review delves into prospective applications of this therapeutic approach across a diverse range of scenarios. Lastly, the review underscores concerns related to precision and safety, while also outlining the envisioned trajectory for extending CAR T therapy beyond cancer treatment.
Collapse
Affiliation(s)
- Zhibo Yang
- Department of Neurosurgery, 3201 Hospital of Xi'an Jiaotong University Health Science Center, Hanzhong, Shaanxi 723000, China
| | - Yingfeng Liu
- Department of Neurosurgery, Tianshui First People's Hospital, Tianshui, Gansu 741000, China
| | - Hai Zhao
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266005, China.
| |
Collapse
|
39
|
Enriquez-Rodriguez L, Attia N, Gallego I, Mashal M, Maldonado I, Puras G, Pedraz JL. Expanding the horizon of transient CAR T therapeutics using virus-free technology. Biotechnol Adv 2024; 72:108350. [PMID: 38537878 DOI: 10.1016/j.biotechadv.2024.108350] [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: 12/07/2023] [Revised: 03/14/2024] [Accepted: 03/24/2024] [Indexed: 04/01/2024]
Abstract
The extraordinary success that chimeric antigen receptor (CAR) T cell therapies have shown over the years on fighting hematological malignancies is evidenced by the six FDA-approved products present on the market. CAR T treatments have forever changed the way we understand cellular immunotherapies, as current research in the topic is expanding even outside the field of cancer with very promising results. Until now, virus-based strategies have been used for CAR T cell manufacturing. However, this methodology presents relevant limitations that need to be addressed prior to wide spreading this technology to other pathologies and in order to optimize current cancer treatments. Several approaches are being explored to overcome these challenges such as virus-free alternatives that additionally offer the possibility of developing transient CAR expression or in vivo T cell modification. In this review, we aim to spotlight a pivotal juncture in the history of medicine where a significant change in perspective is occurring. We review the current progress made on viral-based CAR T therapies as well as their limitations and we discuss the future outlook of virus-free CAR T strategies to overcome current challenges and achieve affordable immunotherapies for a wide variety of pathologies, including cancer.
Collapse
Affiliation(s)
- Lucia Enriquez-Rodriguez
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Noha Attia
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Histology and Cell Biology Department, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Idoia Gallego
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Mohamed Mashal
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Iván Maldonado
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Gustavo Puras
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - José Luis Pedraz
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
| |
Collapse
|
40
|
Shumnalieva R, Velikova T, Monov S. Expanding the role of CAR T-cell therapy: From B-cell hematological malignancies to autoimmune rheumatic diseases. Int J Rheum Dis 2024; 27:e15182. [PMID: 38742463 DOI: 10.1111/1756-185x.15182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 04/04/2024] [Accepted: 04/28/2024] [Indexed: 05/16/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a form of immunotherapy where the lymphocytes, mostly T-cells, are redirected to specifically recognize and eliminate a target antigen by coupling them with CARs. The binding of CAR and target cell surface antigens leads to vigorous T cell activation and robust anti-tumor immune responses. Areas of implication of CAR T-cell therapies include mainly hematological malignancies (i.e., advanced B-cell cancers); however, recent studies have proven the unprecedented success of the new immunotherapy also in autoimmune rheumatic diseases. We aim to review the recent advances in CAR T-cell therapies in rheumatology but also to address the limitations of their use in the real clinical practice based on the data on their efficacy and safety.
Collapse
Affiliation(s)
- Russka Shumnalieva
- Department of Rheumatology, Clinic of Rheumatology, Medical University-Sofia, Faculty of Medicine, Sofia, Bulgaria
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University "St. Kliment Ohridski"- Sofia, Sofia, Bulgaria
| | - Simeon Monov
- Department of Rheumatology, Clinic of Rheumatology, Medical University-Sofia, Faculty of Medicine, Sofia, Bulgaria
| |
Collapse
|
41
|
Hassan SH, Alshahrani MY, Saleh RO, Mohammed BA, Kumar A, Almalki SG, Alkhafaji AT, Ghildiyal P, Al-Tameemi AR, Elawady A. A new vision of the efficacy of both CAR-NK and CAR-T cells in treating cancers and autoimmune diseases. Med Oncol 2024; 41:127. [PMID: 38656354 DOI: 10.1007/s12032-024-02362-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/19/2024] [Indexed: 04/26/2024]
Abstract
Chimeric Antigen Receptor (CAR) based therapies are becoming increasingly important in treating patients. CAR-T cells have been shown to be highly effective in the treatment of hematological malignancies. However, harmful therapeutic barriers have been identified, such as the potential for graft-versus-host disease (GVHD), neurotoxicity, and cytokine release syndrome (CRS). As a result, CAR NK-cell therapy is expected to be a new therapeutic option. NK cells act as cytotoxic lymphocytes, supporting the innate immune response against autoimmune diseases and cancer cells by precisely detecting and eliminating malignant cells. Genetic modification of these cells provides a dual approach to the treatment of AD and cancer. It can be used through both CAR-independent and CAR-dependent mechanisms. The use of CAR-based cell therapies has been successful in treating cancer patients, leading to further investigation of this innovative treatment for alternative diseases, including AD. The complementary roles of CAR T and CAR NK cells have stimulated exploration in this area. Our study examines the latest research on the therapeutic effectiveness of these cells in treating both cancer and ADs.
Collapse
Affiliation(s)
- Salim Hussein Hassan
- Community Health Department, Technical Institute of Karbala, AL-Furat Al-Awsat Technical University, Najaf, Iraq.
| | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | | | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named After the First President of Russia Boris Yeltsin, Ekaterinburg, 620002, Russia
| | - Sami G Almalki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, 11952, Majmaah, Saudi Arabia
| | | | - Pallavi Ghildiyal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - Ahmed Elawady
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
42
|
Rojas-Quintero J, Díaz MP, Palmar J, Galan-Freyle NJ, Morillo V, Escalona D, González-Torres HJ, Torres W, Navarro-Quiroz E, Rivera-Porras D, Bermúdez V. Car T Cells in Solid Tumors: Overcoming Obstacles. Int J Mol Sci 2024; 25:4170. [PMID: 38673757 PMCID: PMC11050550 DOI: 10.3390/ijms25084170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/28/2024] Open
Abstract
Chimeric antigen receptor T cell (CAR T cell) therapy has emerged as a prominent adoptive cell therapy and a therapeutic approach of great interest in the fight against cancer. This approach has shown notorious efficacy in refractory hematological neoplasm, which has bolstered its exploration in the field of solid cancers. However, successfully managing solid tumors presents considerable intrinsic challenges, which include the necessity of guiding the modified cells toward the tumoral region, assuring their penetration and survival in adverse microenvironments, and addressing the complexity of identifying the specific antigens for each type of cancer. This review focuses on outlining the challenges faced by CAR T cell therapy when used in the treatment of solid tumors, as well as presenting optimizations and emergent approaches directed at improving its efficacy in this particular context. From precise localization to the modulation of the tumoral microenvironment and the adaptation of antigen recognition strategies, diverse pathways will be examined to overcome the current limitations and buttress the therapeutic potential of CAR T cells in the fight against solid tumors.
Collapse
Affiliation(s)
- Joselyn Rojas-Quintero
- Medicine, Pulmonary, Critical Care, and Sleep Medicine Department, Baylor College of Medicine, Houston, TX 77030, USA;
| | - María P. Díaz
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | - Jim Palmar
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | - Nataly J. Galan-Freyle
- Centro de Investigaciones en Ciencias de la Vida, Universidad Simón Bolívar, Barranquilla 080002, Colombia; (N.J.G.-F.); (E.N.-Q.)
| | - Valery Morillo
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | - Daniel Escalona
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | | | - Wheeler Torres
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | - Elkin Navarro-Quiroz
- Centro de Investigaciones en Ciencias de la Vida, Universidad Simón Bolívar, Barranquilla 080002, Colombia; (N.J.G.-F.); (E.N.-Q.)
- Facultad de Ciencias Básicas y Biomédicas, Barranquilla 080002, Colombia
| | - Diego Rivera-Porras
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Cúcuta 540001, Colombia;
| | - Valmore Bermúdez
- Centro de Investigaciones en Ciencias de la Vida, Universidad Simón Bolívar, Barranquilla 080002, Colombia; (N.J.G.-F.); (E.N.-Q.)
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 080002, Colombia;
| |
Collapse
|
43
|
Chen Y, Liu C, Fang Y, Chen W, Qiu J, Zhu M, Wei W, Tu J. Developing CAR-immune cell therapy against SARS-CoV-2: Current status, challenges and prospects. Biochem Pharmacol 2024; 222:116066. [PMID: 38373592 DOI: 10.1016/j.bcp.2024.116066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
Chimeric antigen receptor (CAR)-immune cell therapy has revolutionized the anti-tumor field, achieving efficient and precise tumor clearance by directly guiding immune cell activity to target tumors. In addition, the use of CAR-immune cells to influence the composition and function of the immune system and ultimately achieve virus clearance and immune system homeostasis has attracted the interest of researchers. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) triggered a global pandemic of coronavirus disease 2019 (COVID-19). To date, the rapidly mutating SARS-CoV-2 continues to challenge existing therapies and has raised public concerns regarding reinfection. In patients with COVID-19, the interaction of SARS-CoV-2 with the immune system influences the course of the disease, and the coexistence of over-activated immune system components, such as macrophages, and severely compromised immune system components, such as natural killer cells, reveals a dysregulated immune system. Dysregulated immune-induced inflammation may impair viral clearance and T-cell responses, causing cytokine storms and ultimately leading to patient death. Here, we summarize the research progress on the use of CAR-immune cells against SARS-CoV-2 infection. Furthermore, we discuss the feasibility, challenges and prospect of CAR-immune cells as a new immune candidate therapy against SARS-CoV-2.
Collapse
Affiliation(s)
- Yizhao Chen
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Chong Liu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Yilong Fang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Weile Chen
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Jiaqi Qiu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Mengjuan Zhu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Wei Wei
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China.
| | - Jiajie Tu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
44
|
Abraham D, Lescoat A, Stratton R. Emerging diagnostic and therapeutic challenges for skin fibrosis in systemic sclerosis. Mol Aspects Med 2024; 96:101252. [PMID: 38325132 DOI: 10.1016/j.mam.2024.101252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/29/2024] [Indexed: 02/09/2024]
Abstract
Systemic sclerosis (also called scleroderma, SSc) is a chronic autoimmune disorder characterized by excessive collagen deposition leading to skin fibrosis and various internal organ manifestations. The emergent diagnostics and therapeutic strategies for scleroderma focus on early detection and targeted interventions to improve patient outcomes and quality of life. Diagnostics for SSc have evolved significantly in recent years, driven by advancements in serological markers and imaging techniques. Autoantibody profiling, especially antinuclear antibodies (ANA) and specific scleroderma-associated autoantibodies, aids in identifying subsets of scleroderma and predicting disease progression. Furthermore, novel imaging modalities, such as high-frequency ultrasonography and optical coherence tomography, enable early detection of skin fibrosis and internal organ involvement, enhancing the diagnostic precision and allowing for tailored management. Therapeutic strategies for SSc are multifaceted, targeting immune dysregulation, vascular abnormalities, and fibrotic processes. Emerging biologic agents have shown promise in clinical trials, including monoclonal antibodies directed against key cytokines involved in fibrosis, such as transforming growth factor-β (TGF-β) and interleukin-6 (IL-6). Additionally, small-molecule inhibitors that disrupt fibrotic pathways, like tyrosine kinase inhibitors, have exhibited potential in limiting collagen deposition and preventing disease progression. Stem cell therapy, cell ablation and gene editing techniques hold great potential in regenerating damaged tissue and halting fibrotic processes. Early intervention remains crucial in managing SSc, as irreversible tissue damage often occurs in advanced stages. Novel diagnostic methods, such as biomarkers and gene expression profiling, are being explored to identify individuals at high risk for developing progressive severe disease and intervene proactively. Furthermore, patient-tailored therapeutic approaches, employing a combination of immunosuppressive agents and targeted anti-fibrotic therapies, are being investigated to improve treatment efficacy while minimizing adverse effects. The emergent diagnostics and therapeutic strategies in scleroderma are transforming the management of this challenging disease. Nevertheless, ongoing research and clinical trials are needed to optimize the efficacy and safety of these novel approaches in the complex and diverse spectrum of SSc manifestations.
Collapse
Affiliation(s)
- David Abraham
- UCL Centre for Rheumatology, Royal Free Hospital, UCL Division of Medicine, Department of Inflammation, London, UK
| | - Alain Lescoat
- Department of Internal Medicine and Clinical Immunology, Rennes University Hospital, Rennes, France
| | - Richard Stratton
- UCL Centre for Rheumatology, Royal Free Hospital, UCL Division of Medicine, Department of Inflammation, London, UK.
| |
Collapse
|
45
|
Hou F, Guo Z, Ho MT, Hui Y, Zhao CX. Particle-Based Artificial Antigen-Presenting Cell Systems for T Cell Activation in Adoptive T Cell Therapy. ACS NANO 2024; 18:8571-8599. [PMID: 38483840 DOI: 10.1021/acsnano.3c10180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
T cell-based adoptive cell therapy (ACT) has emerged as a promising treatment for various diseases, particularly cancers. Unlike other immunotherapy modalities, ACT involves directly transferring engineered T cells into patients to eradicate diseased cells; hence, it necessitates methods for effectively activating and expanding T cells in vitro. Artificial antigen-presenting cells (aAPCs) have been widely developed based on biomaterials, particularly micro- and nanoparticles, and functionalized with T cell stimulatory antibodies to closely mimic the natural T cell-APC interactions. Due to their vast clinical utility, aAPCs have been employed as an off-the-shelf technology for T cell activation in FDA-approved ACTs, and the development of aAPCs is constantly advancing with the emergence of aAPCs with more sophisticated designs and additional functionalities. Here, we review the recent advancements in particle-based aAPCs for T cell activation in ACTs. Following a brief introduction, we first describe the manufacturing processes of ACT products. Next, the design and synthetic strategies for micro- and nanoparticle-based aAPCs are discussed separately to emphasize their features, advantages, and limitations. Then, the impact of design parameters of aAPCs, such as size, shape, ligand density/mobility, and stiffness, on their functionality and biomedical performance is explored to provide deeper insights into the design concepts and principles for more efficient and safer aAPCs. The review concludes by discussing current challenges and proposing future perspectives for the development of more advanced aAPCs.
Collapse
Affiliation(s)
- Fei Hou
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Zichao Guo
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Minh Trang Ho
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Yue Hui
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Chun-Xia Zhao
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
46
|
Appalaneni R, Achanta N, Mohan C. Chimeric antigen receptor T-cell therapy in rheumatology: B-cell depletion 2.0. Curr Opin Rheumatol 2024; 36:126-133. [PMID: 38099466 DOI: 10.1097/bor.0000000000000994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
PURPOSE OF REVIEW Chimeric antigen receptor T-cell therapy (CAR-T) has revolutionized cancer treatment by harnessing the immune system's power to target malignancies. CD19, a B-cell surface antigen, a key target for CAR-T cell therapy in hematological malignancies, displayed remarkable clinical responses. Recently, there has been a growing interest in exploring the application of CD19 CAR-T cell therapy beyond oncology. The rationale for investigating CD19 CAR-T cells in Rheumatology stems from their ability to selectively target B cells, which play a central pathogenic role through autoantibody-dependent and independent mechanisms. RECENT FINDINGS Preclinical and five completed clinical studies have shown remarkable efficacy and safety in diseases such as systemic lupus erythematosus, antisynthetase syndrome, and systemic sclerosis. It is thus not surprising that 17 active clinical trials exploring CAR-T cells in Rheumatology are in progress. SUMMARY Although CAR-T therapy holds great promise in Rheumatology, many challenges loom. Whether this new way to deplete B-cells is superior to conventional antibody-based B-cell depletion in rheumatic diseases will be closely watched in the coming years.
Collapse
Affiliation(s)
- Rohith Appalaneni
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| | | | | |
Collapse
|
47
|
Liu Q, Chen G, Liu X, Tao L, Fan Y, Xia T. Tolerogenic Nano-/Microparticle Vaccines for Immunotherapy. ACS NANO 2024. [PMID: 38323542 DOI: 10.1021/acsnano.3c11647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Autoimmune diseases, allergies, transplant rejections, generation of antidrug antibodies, and chronic inflammatory diseases have impacted a large group of people across the globe. Conventional treatments and therapies often use systemic or broad immunosuppression with serious efficacy and safety issues. Tolerogenic vaccines represent a concept that has been extended from their traditional immune-modulating function to induction of antigen-specific tolerance through the generation of regulatory T cells. Without impairing immune homeostasis, tolerogenic vaccines dampen inflammation and induce tolerogenic regulation. However, achieving the desired potency of tolerogenic vaccines as preventive and therapeutic modalities calls for precise manipulation of the immune microenvironment and control over the tolerogenic responses against the autoantigens, allergens, and/or alloantigens. Engineered nano-/microparticles possess desirable design features that can bolster targeted immune regulation and enhance the induction of antigen-specific tolerance. Thus, particle-based tolerogenic vaccines hold great promise in clinical translation for future treatment of aforementioned immune disorders. In this review, we highlight the main strategies to employ particles as exciting tolerogenic vaccines, with a focus on the particles' role in facilitating the induction of antigen-specific tolerance. We describe the particle design features that facilitate their usage and discuss the challenges and opportunities for designing next-generation particle-based tolerogenic vaccines with robust efficacy to promote antigen-specific tolerance for immunotherapy.
Collapse
Affiliation(s)
- Qi Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Guoqiang Chen
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Xingchi Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Lu Tao
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Yubo Fan
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Tian Xia
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Division of NanoMedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
48
|
Wu J, Wu W, Zhou B, Li B. Chimeric antigen receptor therapy meets mRNA technology. Trends Biotechnol 2024; 42:228-240. [PMID: 37741706 DOI: 10.1016/j.tibtech.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/25/2023]
Abstract
Genetically engineered immune cells expressing chimeric antigen receptors (CARs) have emerged as a new game changer in cancer immunotherapy. The utility of CAR T cell therapy against hematological malignancies has been validated in clinical practice. Other CAR immune cells are currently under investigation to improve the potency of CAR therapy in solid tumors. As a new class of therapeutic modalities, mRNA-based therapeutics hold enormous potential beyond COVID-19 mRNA vaccines. Arming immune cells with mRNA-encoded CARs represents a new frontier in cancer and beyond, enabling in vivo generation of CAR cells without causing transgene integration. In this review, we summarize recent advances in mRNA-based CAR immunotherapies and highlight their opportunities and challenges for the development of a new generation of living drugs.
Collapse
Affiliation(s)
- Jiacai Wu
- Department of Infectious Disease, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology and The Second Clinical Medical College of Jinan University, Shenzhen 518020, China; School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Weigang Wu
- Department of Infectious Disease, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology and The Second Clinical Medical College of Jinan University, Shenzhen 518020, China
| | - Boping Zhou
- Department of Infectious Disease, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology and The Second Clinical Medical College of Jinan University, Shenzhen 518020, China; School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Bin Li
- Department of Infectious Disease, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology and The Second Clinical Medical College of Jinan University, Shenzhen 518020, China; School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
49
|
Gokemeijer J, Balasubramanian N, Ogasawara K, Grudzinska-Goebel J, Upreti VV, Mody H, Kasar S, Vepachedu VR, Xu W, Gupta S, Tarcsa E, Dodge R, Herr K, Yang TY, Tourdot S, Jawa V. An IQ Consortium Perspective on Best Practices for Bioanalytical and Immunogenicity Assessment Aspects of CAR-T and TCR-T Cellular Therapies Development. Clin Pharmacol Ther 2024; 115:188-200. [PMID: 37983584 DOI: 10.1002/cpt.3111] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/05/2023] [Indexed: 11/22/2023]
Abstract
CAR-T therapies have shown remarkable efficacy against hematological malignancies in the clinic over the last decade and new studies indicate that progress is being made to use these novel therapies to target solid tumors as well as treat autoimmune disease. Innovation in the field, including TCR-T, allogeneic or "off the shelf" CAR-T, and autoantigen/armored CAR-Ts are likely to increase the efficacy and applications of these therapies. The unique aspects of these cell-based therapeutics; patient-derived cells, intracellular expression, in vivo expansion, and phenotypic changes provide unique bioanalytical challenges to develop pharmacokinetic and immunogenicity assessments. The International Consortium for Innovation and Quality in Pharmaceutical Development (IQ) Translational and ADME Sciences Leadership Group (TALG) has brought together a group of industry experts to discuss and consider these challenges. In this white paper, we present the IQ consortium perspective on the best practices and considerations for bioanalytical and immunogenicity aspects toward the optimal development of CAR-T and TCR-T cell therapies.
Collapse
Affiliation(s)
- Jochem Gokemeijer
- Discovery Biotherapeutics, Bristol Myers Squibb, Cambridge, Massachusetts, USA
| | - Nanda Balasubramanian
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | - Ken Ogasawara
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | | | - Vijay V Upreti
- Clinical Pharmacology, Modeling & Simulation, Amgen, Inc., South San Francisco, California, USA
| | - Hardik Mody
- Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Siddha Kasar
- Oncology Precision & Translational Medicine, Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Venkata R Vepachedu
- Bioanalytical Discovery & Development Sciences, Johnson & Johnson Innovative Medicine, Spring House, Pennsylvania, USA
| | - Weifeng Xu
- Preclinical Development, Bioanalytical, Merck & Co., Inc., Rahway, New Jersey, USA
| | - Swati Gupta
- Development Biological Sciences, Immunology, AbbVie, Irvine, California, USA
| | - Edit Tarcsa
- Abbvie Bioresearch Center, Worcester, Massachusetts, USA
| | - Robert Dodge
- Novartis Institutes for BioMedical Research, One Health Plaza, East Hanover, New Jersey, USA
| | - Kate Herr
- Bioanalytical Discovery & Development Sciences, Johnson & Johnson Innovative Medicine, Spring House, Pennsylvania, USA
| | - Tong-Yuan Yang
- Bioanalytical Discovery & Development Sciences, Johnson & Johnson Innovative Medicine, Spring House, Pennsylvania, USA
| | - Sophie Tourdot
- BioMedicine Design, Pfizer Inc., Andover, Massachusetts, USA
| | - Vibha Jawa
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| |
Collapse
|
50
|
Kalim M, Jing R, Li X, Jiang Z, Zheng N, Wang Z, Wei G, Lu Y. Essentials of CAR-T Therapy and Associated Microbial Challenges in Long Run Immunotherapy. JOURNAL OF CELLULAR IMMUNOLOGY 2024; 6:22-50. [PMID: 38883270 PMCID: PMC11172397 DOI: 10.33696/immunology.6.189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has shown potential in improving outcomes for individuals with hematological malignancies. However, achieving long-term full remission for blood cancer remains challenging due to severe life-threatening toxicities such as limited anti-tumor efficacy, antigen escape, trafficking restrictions, and limited tumor invasion. Furthermore, the interactions between CAR-T cells and their host tumor microenvironments have a significant impact on CAR-T function. To overcome these considerable hurdles, fresh methodologies and approaches are needed to produce more powerful CAR-T cells with greater anti-tumor activity and less toxicity. Despite advances in CAR-T research, microbial resistance remains a significant obstacle. In this review, we discuss and describe the basics of CAR-T structures, generations, challenges, and potential risks of infections in CAR-T cell therapy.
Collapse
Affiliation(s)
- Muhammad Kalim
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| | - Rui Jing
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| | - Xin Li
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| | - Zhiwu Jiang
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| | - Ningbo Zheng
- Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Ziyu Wang
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| | - Guo Wei
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| | - Yong Lu
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| |
Collapse
|