1
|
Velasco Santiago M, Aehnlich P, Hulen T, Jensen K, Holmen Olofsson G, Met Ö, thor Straten P. Overcoming antigen loss in CAR T therapy with Vγ9Vδ2 CAR T-cells. IMMUNO-ONCOLOGY TECHNOLOGY 2025; 26:101053. [PMID: 40271017 PMCID: PMC12013397 DOI: 10.1016/j.iotech.2025.101053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Background Vγ9Vδ2 T-cells demonstrate potent antitumor activity in vitro but, despite successful safety studies, the clinical benefit of Vγ9Vδ2 in adoptive cell therapy has been limited. One approach to enhance the therapeutic potential of Vγ9Vδ2 T-cells while maintaining their safety profile is genetic engineering to express a chimeric antigen receptor (CAR). Vγ9Vδ2 CAR T-cells retain the ability to target tumor cells even after target antigen loss, a major cause of CAR treatment relapse. Methods Vγ9Vδ2 T-cells were expanded from peripheral blood mononuclear cells in the presence of high levels of interleukin 2 (IL-2) or IL-2 in combination with IL-15. Cells were then virally transduced with a CD19-directed CAR and underwent antigen-specific stimulation to enrich CAR-expressing cells. Results Vγ9Vδ2 CAR T-cells showed similar cytotoxic activity to conventional αβ-CAR T-cells against CD19-positive tumor cells. They demonstrated superior responses against CD19-negative tumor cells, however, particularly when IL-15 was included during expansion. This enhanced function was further confirmed in co-culture assays with mixed CD19-positive and CD19-negative tumor populations, simulating antigen loss. Conclusions Vγ9Vδ2 CAR T-cell therapy presents a promising strategy for B-cell malignancies, offering sustained antitumor activity even after antigen loss. This approach may help overcome a major limitation of conventional CAR T-cell therapy, potentially improving clinical outcomes.
Collapse
Affiliation(s)
- M. Velasco Santiago
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, University Hospital Herlev, Herlev, Denmark
| | - P. Aehnlich
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, University Hospital Herlev, Herlev, Denmark
| | - T.M. Hulen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, University Hospital Herlev, Herlev, Denmark
| | - K.M. Jensen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, University Hospital Herlev, Herlev, Denmark
| | - G. Holmen Olofsson
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, University Hospital Herlev, Herlev, Denmark
| | - Ö. Met
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, University Hospital Herlev, Herlev, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - P. thor Straten
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, University Hospital Herlev, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Arroyo-Ródenas J, Falgas A, Díez-Alonso L, Martinez-Moreno A, Roca-Ho H, Gil-Etayo FJ, Pérez-Pons A, Aguilar-Sopeña Ó, Velasco-Sidro M, Gómez-Rosel M, Jiménez-Matías B, Muñoz-Sánchez G, Pacheco Y, Bravo-Martín C, Ramírez-Fernández Á, Jiménez-Reinoso A, González-Navarro EA, Juan M, Orfao A, Blanco B, Roda-Navarro P, Bueno C, Menéndez P, Álvarez-Vallina L. CD22 CAR-T cells secreting CD19 T-cell engagers for improved control of B-cell acute lymphoblastic leukemia progression. J Immunother Cancer 2025; 13:e009048. [PMID: 40306957 DOI: 10.1136/jitc-2024-009048] [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] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND CD19-directed cancer immunotherapies, based on engineered T cells bearing chimeric antigen receptors (CARs, CAR-T cells) or the systemic administration of bispecific T cell-engaging (TCE) antibodies, have shown impressive clinical responses in relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL). However, more than half of patients relapse after CAR-T or TCE therapy, with antigen escape or lineage switching accounting for one-third of disease recurrences. To minimize tumor escape, dual-targeting CAR-T cell therapies simultaneously targeting CD19 and CD22 have been developed and validated both preclinically and clinically. METHODS We have generated the first dual-targeting strategy for B-cell malignancies based on CD22 CAR-T cells secreting an anti-CD19 TCE antibody (CAR-STAb-T) and conducted a comprehensive preclinical characterization comparing its therapeutic potential in B-ALL with that of previously validated dual-targeting CD19/CD22 tandem CAR cells (TanCAR-T cells) and co-administration of two single-targeting CD19 and CD22 CAR-T cells (pooled CAR-T cells). RESULTS We demonstrate that CAR-STAb-T cells efficiently redirect bystander T cells, resulting in higher cytotoxicity of B-ALL cells than dual-targeting CAR-T cells at limiting effector:target ratios. Furthermore, when antigen loss was replicated in a heterogeneous B-ALL cell model, CAR-STAb T cells induced more potent and effective cytotoxic responses than dual-targeting CAR-T cells in both short- and long-term co-culture assays, reducing the risk of CD19-positive leukemia escape. In vivo, CAR-STAb-T cells also controlled leukemia progression more efficiently than dual-targeting CAR-T cells in patient-derived xenograft mouse models under T cell-limiting conditions. CONCLUSIONS CD22 CAR-T cells secreting CD19 T-cell engagers show an enhanced control of B-ALL progression compared with CD19/CD22 dual CAR-based therapies, supporting their potential for clinical testing.
Collapse
Affiliation(s)
- Javier Arroyo-Ródenas
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Aida Falgas
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Díez-Alonso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alba Martinez-Moreno
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III, Madrid, Spain
| | - Heleia Roca-Ho
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco J Gil-Etayo
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Alba Pérez-Pons
- Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca, Salamanca, Spain
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC; CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
- Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - Óscar Aguilar-Sopeña
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense, Madrid, Spain
- Lymphocyte Immunobiology Group, Instituto de Investigación Sanitaria 12 de Octubre (i+12), Madrid, Spain
| | - Miriam Velasco-Sidro
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Marina Gómez-Rosel
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Beatriz Jiménez-Matías
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | | | - Yedra Pacheco
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Clara Bravo-Martín
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense, Madrid, Spain
- Lymphocyte Immunobiology Group, Instituto de Investigación Sanitaria 12 de Octubre (i+12), Madrid, Spain
| | - Ángel Ramírez-Fernández
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Anaïs Jiménez-Reinoso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Manel Juan
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Inmunología, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Alberto Orfao
- Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca, Salamanca, Spain
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC; CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
- Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - Belén Blanco
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Pedro Roda-Navarro
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense, Madrid, Spain
- Lymphocyte Immunobiology Group, Instituto de Investigación Sanitaria 12 de Octubre (i+12), Madrid, Spain
| | - Clara Bueno
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC; CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
| | - Pablo Menéndez
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC; CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Biomedicine, School of Medicine, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Hospital Sant Joan de Déu-Pediatric Cancer Center Barcelona (SJD-PCCB), Barcelona, Spain
| | - Luis Álvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- CNIO-HMRIB Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Hospital del Mar Research Institute Barcelona (HMRIB), Madrid/Barcelona, Spain
- Banc de Sang i Teixits (BST), Barcelona, Spain
| |
Collapse
|
3
|
Domizi P, Sarno J, Jager A, Merchant M, Pacheco KZB, Yamada-Hunter SA, Rotiroti MC, Liu Y, Baskar R, Reynolds WD, Sworder BJ, Sahaf B, Bendall SC, Mullighan CG, Alizadeh AA, Leahy AB, Myers RM, Yates B, Wang HW, Shah NN, Majzner RG, Mackall CL, Grupp SA, Barrett DM, Sotillo E, Davis KL. IKAROS levels are associated with antigen escape in CD19- and CD22-targeted therapies for B-cell malignancies. Nat Commun 2025; 16:3800. [PMID: 40268897 PMCID: PMC12019336 DOI: 10.1038/s41467-025-58868-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 04/02/2025] [Indexed: 04/25/2025] Open
Abstract
Antigen escape relapse is a major challenge in targeted immunotherapies, including CD19- and CD22-directed chimeric antigen receptor (CAR) T-cell for B-cell acute lymphoblastic leukemia (B-ALL). To identify tumor-intrinsic factors driving antigen loss, we perform single-cell analyses on 61 B-ALL patient samples treated with CAR T cells. Here we show that low levels of IKAROS in pro-B-like B-ALL cells before CAR T treatment correlate with antigen escape. IKAROSlow B-ALL cells undergo epigenetic and transcriptional changes that diminish B-cell identity, making them resemble progenitor cells. This shift leads to reduced CD19 and CD22 surface expression. We demonstrate that CD19 and CD22 expression is IKAROS dose-dependent and reversible. Furthermore, IKAROSlow cells exhibit higher resistance to CD19- and CD22-targeted therapies. These findings establish a role for IKAROS as a regulator of antigens targeted by widely used immunotherapies and in the risk of antigen escape relapse, identifying it as a potential prognostic target.
Collapse
Affiliation(s)
- Pablo Domizi
- Department of Pediatrics, Hematology, Oncology, Stem Cell Transplant and Regenerative Medicine, Stanford University, Stanford, CA, USA.
| | - Jolanda Sarno
- Department of Pediatrics, Hematology, Oncology, Stem Cell Transplant and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20126, Milan, Italy
| | - Astraea Jager
- Department of Pediatrics, Hematology, Oncology, Stem Cell Transplant and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Milton Merchant
- Department of Pediatrics, Hematology, Oncology, Stem Cell Transplant and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Kaithlen Zen B Pacheco
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Sean A Yamada-Hunter
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Yuxuan Liu
- Department of Pediatrics, Hematology, Oncology, Stem Cell Transplant and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Reema Baskar
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Warren D Reynolds
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Brian J Sworder
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Bita Sahaf
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Sean C Bendall
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ash A Alizadeh
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Allison B Leahy
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Regina M Myers
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bonnie Yates
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hao-Wei Wang
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Robbie G Majzner
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Crystal L Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Stephan A Grupp
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Elena Sotillo
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Kara L Davis
- Department of Pediatrics, Hematology, Oncology, Stem Cell Transplant and Regenerative Medicine, Stanford University, Stanford, CA, USA.
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
4
|
Hu X, Wang Z, Zhu Y, Li Z, Yan H, Zhao X, Wang Q. Advancements in molecular imaging for the diagnosis and treatment of pancreatic ductal adenocarcinoma. NANOSCALE ADVANCES 2025:d4na01080a. [PMID: 40270837 PMCID: PMC12012634 DOI: 10.1039/d4na01080a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/03/2025] [Indexed: 04/25/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor characterized by poor overall patient survival and prognosis, largely due to challenges in early diagnosis, limited surgical options, and a high propensity for therapy resistance. The integration of various imaging modalities through molecular imaging techniques, particularly multimodal molecular imaging, offers the potential to provide more precise and comprehensive information about the lesion. With advances in nanomedicine, new imaging and drug delivery approaches that allow the development of multifunctional theranostic agents offer opportunities for improving pancreatic cancer treatment using precision oncology. Herein, we review the diagnostic and therapeutic applications of molecular imaging for PDAC and discuss the adoption of multimodal imaging approaches that combine the strengths of different imaging techniques to enhance diagnostic accuracy and therapeutic efficacy. We emphasize the significant role of nanomedicine technology in advancing multimodal molecular imaging and theranostics, and their potential impact on PDAC management. This comprehensive review aims to serve as a valuable reference for researchers and clinicians, offering insights into the current state of molecular imaging in PDAC and outlining future directions for improving early diagnosis, combination therapies, and prognostic evaluations.
Collapse
Affiliation(s)
- Xun Hu
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021 China
| | - Zihua Wang
- School of Basic Medical Sciences, Fujian Medical University Fuzhou 350122 Fujian Province China
| | - Yuting Zhu
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021 China
| | - Zhangfu Li
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital Shenzhen Guangdong 518036 China
| | - Hao Yan
- Tsinghua Shenzhen International Graduate School/Tsinghua University Shenzhen 518055 China
| | - Xinming Zhao
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021 China
| | - Qian Wang
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021 China
| |
Collapse
|
5
|
Lei W, Liu H, Deng W, Chen W, Liang Y, Gao W, Yuan X, Guo S, Li P, Wang J, Tong X, Sun YE, Liang A, Qian W. Safety and feasibility of 4-1BB co-stimulated CD19-specific CAR-NK cell therapy in refractory/relapsed large B cell lymphoma: a phase 1 trial. NATURE CANCER 2025:10.1038/s43018-025-00940-3. [PMID: 40251398 DOI: 10.1038/s43018-025-00940-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/05/2025] [Indexed: 04/20/2025]
Abstract
Chimeric antigen receptor (CAR)-modified NK (CAR-NK) cells are candidates for next-generation cancer immunotherapies. Here we generated CD19-specific CAR-NK cells with 4-1BB and CD3ζ signaling endo-domains (CD19-BBz CAR-NK) by transduction of cord blood-derived NK cells using baboon envelope pseudotyped lentiviral vectors and demonstrated their antitumor activity in preclinical B cell lymphoma models in female mice. We next conducted a phase 1 dose-escalation trial involving repetitive administration of CAR-NK cells in 8 patients with relapsed/refractory large B cell lymphoma (NCT05472558). Primary end points were safety, maximum tolerated dose, and overall response rate. Secondary end points included duration of response, overall survival, and progression-free survival. No dose-limiting toxicities occurred, and the maximum tolerated dose was not reached. No cases of cytokine release syndrome, neurotoxicity, or graft-versus-host disease were observed. Results showed an overall response rate of 62.5% at day 30, with 4 patients (50%) achieving complete response. The median progression-free survival was 9.5 months, and the median overall survival was not reached. A post hoc exploratory single-cell RNA sequencing analysis revealed molecular features of CAR-NK cells associated with therapeutic efficacy and efficacy-related immune cell interaction networks. This study met the pre-specified end points. In conclusion, CD19-BBz CAR-NK cells were feasible and therapeutically safe, capable of inducing durable response in patients with B cell lymphoma.
Collapse
Affiliation(s)
- Wen Lei
- Department of Hematology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education; Biotherapy Research Center, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hui Liu
- Department of Hematology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Wenhai Deng
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wei Chen
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Yun Liang
- Department of Hematology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Wenxia Gao
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Xianggui Yuan
- Department of Hematology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Shanshan Guo
- Department of Hematology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ping Li
- Department of Hematology, Tongji Hospital of Tongji University, Shanghai, China
| | - Jinyong Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xiangmin Tong
- Department of Hematology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
| | - Yi Eve Sun
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University, School of Medicine, Shanghai, China.
| | - Aibin Liang
- Department of Hematology, Tongji Hospital of Tongji University, Shanghai, China.
| | - Wenbin Qian
- Department of Hematology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education; Biotherapy Research Center, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
6
|
Cimons JM, DeGolier KR, Burciaga SD, Yarnell MC, Novak AJ, Rivera-Reyes AM, Kohler ME, Fry TJ. T-bet overexpression enhances CAR T cell effector functions and antigen sensitivity. J Immunother Cancer 2025; 13:e010962. [PMID: 40246581 PMCID: PMC12007057 DOI: 10.1136/jitc-2024-010962] [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: 11/01/2024] [Accepted: 04/07/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND T cells modified to express a chimeric antigen receptor (CAR) are successful against B-lineage malignancies but fail to induce durable remissions in up to half of patients and have shown limited efficacy against other types of cancer. Strategies to improve CAR T cell potency and responses to low antigen densities without inducing CAR T cell dysfunction or limiting persistence are necessary to expand durability of remissions. METHODS We overexpressed T-bet in human and mouse CAR T cells to mimic exposure to signal 3 cytokines during T cell priming to promote T helper cell 1 (Th1) polarization of CD4+CAR T cells with the goal of enhancing antitumor activity. Using human CAR T cells and xenograft models we interrogated the impact of T-bet overexpression on CAR T cell antitumor activity in vitro and in vivo. We also used a syngeneic murine CAR T cell model to study the impact of T-bet overexpression on long-term persistence and secondary responses to tumor rechallenge. RESULTS T-bet overexpression reduced expression of the Th2 cytokine interleukin 4 and promoted polyfunctional production of Th1-associated cytokines in response to CAR stimulation. T-bet overexpression enhanced some effector functions in vitro but did not improve CAR T cell-mediated control of leukemia expressing high levels of antigen in vivo. T-bet overexpression also improved effector function of murine CD19 CAR T cells with no impairment to the persistence or ability of persistent CAR T cells to re-expand and clear a secondary leukemia challenge. Finally, T-bet overexpression promoted enhanced in vitro function against leukemia expressing low levels of CD19, which translated to improved control of CD19lo leukemia in vivo by human C19 CAR T cells containing a 4-1BB costimulatory domain. CONCLUSIONS Together, our data demonstrate that T-bet overexpression induces a reduction in Th2 cytokine production, an increase in polyfunctional Th1 cytokine production and enhances 4-1BB CAR T cell activity against cancers expressing low levels of target antigen without promoting a loss in functional CAR T cell persistence.
Collapse
Affiliation(s)
- Jennifer M Cimons
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Kole R DeGolier
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Samuel D Burciaga
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Michael C Yarnell
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Amanda J Novak
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Amalia M Rivera-Reyes
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - M Eric Kohler
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
- Pediatrics, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Terry J Fry
- University of Colorado Denver Children's Hospital Colorado Research Institute, Aurora, Colorado, USA
| |
Collapse
|
7
|
Passweg JR, Baldomero H, Atlija M, Kleovoulou I, Witaszek A, Alexander T, Angelucci E, Averbuch D, Bazarbachi A, Ciceri F, Greco R, Hazenberg MD, Kalwak K, McLornan DP, Neven B, Perić Z, Risitano AM, Ruggeri A, Sánchez-Ortega I, Snowden JA, Sureda A. The 2023 EBMT report on hematopoietic cell transplantation and cellular therapies. Increased use of allogeneic HCT for myeloid malignancies and of CAR-T at the expense of autologous HCT. Bone Marrow Transplant 2025; 60:519-528. [PMID: 39939433 PMCID: PMC11971038 DOI: 10.1038/s41409-025-02524-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/13/2025] [Accepted: 01/30/2025] [Indexed: 02/14/2025]
Abstract
In 2023, 47,731 HCT (20,485 (42.9%) allogeneic and 27,246 (57.1%) autologous) in 43,902 patients were reported by 696 European centers. 6042 patients received advanced cellular therapies, 4888 of which were CAR-T. Compared to the previous year there was an increase in CAR-T (+52.5%), in allogeneic HCT (+7.8%) but none in autologous HCT (+0.4%). Main indications for allogeneic HCT were myeloid (11,748; 60.7%), lymphoid malignancies (4,850; 25.0%), and non-malignant disorders (2558; 13.2%). Use of allogeneic HCT increased for AML (+12.1%) and for NHL (+11.0%), particularly in T-NHL (+25.6%). Main indications for autologous HCT were lymphomas (7890; 32.2%), PCD (14,271; 58.2%), and solid tumors (1608; 6.6%) with recovering numbers for autoimmune diseases. In patients with allogeneic HCT, the use of sibling donors increased by +1.0%, haploidentical donors by +11.7%, and unrelated donors by +11.1%. Cord blood HCT decreased again by -5.4%. Pediatric HCT activity increased slightly (5455; +0.1%) with differences between allogeneic (4111; -0.5%) and autologous HCT (1344: +1.7%). Use of CAR-T increased to a cumulative total of 13,927 patients including patients treated for autoimmune diseases. Overall, numbers show a complete recovery from the pandemic dip with increased cellular therapy at the expense of autologous HCT. Allogeneic HCT activity focuses on myeloid malignancies.
Collapse
Affiliation(s)
- Jakob R Passweg
- EBMT Activity Survey Office Basel, Hematology Division, University Hospital, Basel, Switzerland
| | - Helen Baldomero
- EBMT Activity Survey Office Basel, Hematology Division, University Hospital, Basel, Switzerland.
| | - Marina Atlija
- EBMT Activity Survey Office Leiden, Leiden, The Netherlands
| | | | | | - Tobias Alexander
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Emanuele Angelucci
- Hematology and Cellular Therapy Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Dina Averbuch
- Faculty of Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ali Bazarbachi
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Fabio Ciceri
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Raffaella Greco
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Mette D Hazenberg
- Department of Hematology, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands
- Department of Hematopoiesis, Sanquin Research, Amsterdam, The Netherlands
| | - Krzysztof Kalwak
- Clinical Department of Pediatric BMT, Hematology and Oncology, Wroclaw Medical University, Wroclaw, Poland
| | - Donal P McLornan
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Bénédicte Neven
- Pediatric Immune-Hematology Unit, Necker Children Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | | | - Antonio M Risitano
- Hematology and Hematopoietic Transplant Unit, Azienda Ospedaliera di Rilievo Nazionale San Giuseppe Moscati" (A.O.R.N. Giuseppe Moscati), Avellino, Italy
| | - Annalisa Ruggeri
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | | | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Anna Sureda
- Clinical Hematology Department, Institut Català d'Oncologia-Hospitalet, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), University of Barcelona, Barcelona, Spain
| |
Collapse
|
8
|
Shi X, He X, Xu C. Charge-based immunoreceptor signalling in health and disease. Nat Rev Immunol 2025; 25:298-311. [PMID: 39528837 DOI: 10.1038/s41577-024-01105-6] [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: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Immunoreceptors have crucial roles in sensing environmental signals and initiating immune responses to protect the host. Dysregulation of immunoreceptor signalling can therefore lead to a range of diseases, making immunoreceptor-based therapies a promising frontier in biomedicine. A common feature of various immunoreceptors is the basic-residue-rich sequence (BRS), which is a largely unexplored aspect of immunoreceptor signalling. The BRS is typically located in the cytoplasmic juxtamembrane region of immunoreceptors, where it forms dynamic interactions with neighbouring charged molecules to regulate signalling. Loss or gain of the basic residues in an immunoreceptor BRS has been linked to severe human diseases, such as immunodeficiency and autoimmunity. In this Perspective, we describe the role of BRSs in various immunoreceptors, elucidating their signalling mechanisms and biological functions. Furthermore, we highlight pathogenic mutations in immunoreceptor BRSs and discuss the potential of leveraging BRS signalling in engineered T cell-based therapies.
Collapse
Affiliation(s)
- Xiaoshan Shi
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Xing He
- Key Laboratory of Multi-Cell Systems, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Chenqi Xu
- Key Laboratory of Multi-Cell Systems, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
9
|
Santurio DS, Barros LRC, Glauche I, Fassoni AC. Mathematical modeling unveils the timeline of CAR-T cell therapy and macrophage-mediated cytokine release syndrome. PLoS Comput Biol 2025; 21:e1012908. [PMID: 40203243 PMCID: PMC11981663 DOI: 10.1371/journal.pcbi.1012908] [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: 09/02/2024] [Accepted: 02/24/2025] [Indexed: 04/11/2025] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy holds significant potential for cancer treatment, although disease relapse and cytokine release syndrome (CRS) remain as frequent clinical challenges. To better understand the mechanisms underlying the temporal dynamics of CAR-T cell therapy response and CRS, we developed a novel multi-layer mathematical model incorporating antigen-mediated CAR-T cell expansion, antigen-negative resistance, and macrophage-associated cytokine release. Three key mechanisms of macrophage activation are considered: release of damage-associated molecular patterns, antigen-binding mediated activation, and CD40-CD40L contact. The model accurately describes 25 patient time courses with different responses and IL-6 cytokine kinetics. We successfully link the dynamic shape of the response to interpretable model parameters and investigate the influence of CAR-T cell dose and initial tumor burden on the occurrence of cytokine release and treatment outcome. By disentangling the timeline of macrophage activation, the model identified distinct contributions of each activation mechanism, suggesting the CD40-CD40L axis as a major driver of cytokine release and a clinically feasible target to control the activation process and modulate cytokine peak height. Our multi-layer model provides a comprehensive framework for understanding the complex interactions between CAR-T cells, tumor cells, and macrophages during therapy.
Collapse
Affiliation(s)
| | | | - Ingmar Glauche
- Institute for Medical Informatics and Biometry, Technische Universität Dresden, Dresden, Germany
| | - Artur c Fassoni
- Institute for Medical Informatics and Biometry, Technische Universität Dresden, Dresden, Germany
- Instituto de Matemática e Computação, Universidade Federal de Itajubá, Itajubá, Brazil
| |
Collapse
|
10
|
Zhang Q, Dai J, Liu T, Rao W, Li D, Gu Z, Huang L, Wang J, Hou X. Targeting cardiac fibrosis with Chimeric Antigen Receptor-Engineered Cells. Mol Cell Biochem 2025; 480:2103-2116. [PMID: 39460827 DOI: 10.1007/s11010-024-05134-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024]
Abstract
Cardiac fibrosis poses a significant challenge in cardiovascular diseases due to its intricate pathogenesis, and there is currently no standardized and effective treatment approach. The fibrotic process entails the involvement of various cell types and molecular mechanisms, such as fibroblast activation and proliferation, increased collagen synthesis, and extracellular matrix rearrangement. Traditional therapies often fall short in efficacy or carry substantial side effects. However, recent studies have shown that Chimeric Antigen Receptor T (CAR-T) cells can selectively target and eliminate activated cardiac fibroblasts (CFs) in mice, leading to reduced cardiac fibrosis and improved myocardial tissue compliance. This breakthrough presents a new and promising avenue for treating cardiac fibrosis. Currently, CAR-T cell-based therapy for cardiac fibrosis is undergoing animal experimentation, indicating ample scope for enhancement. Future investigations could explore the application of CAR cell therapy in cardiac fibrosis treatment, including the potential of CAR-natural killer (CAR-NK) cells and CAR macrophages (CAR-M), offering novel insights and strategies for combating cardiac fibrosis.
Collapse
Affiliation(s)
- Qinghang Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University of Medicine, Shanghai, 200030, China
| | - Jinjie Dai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University of Medicine, Shanghai, 200030, China
| | - Tianbao Liu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University of Medicine, Shanghai, 200030, China
| | - Wutian Rao
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University of Medicine, Shanghai, 200030, China
| | - Dan Li
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Zhengying Gu
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Lin Huang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Jiayi Wang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Xumin Hou
- Hospital's Office, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| |
Collapse
|
11
|
Corona M, Ip A, Brown S, Luna A, Khatib H, Flynn JR, Devlin SM, Landego I, Cassanello G, Rejeski K, Zuckerman T, Dahi PB, Scordo M, Lin RJ, Kabat M, Luttwak E, Pavkovic E, Palomba ML, Park J, Salles G, Schoder H, Leithner D, Leslie LA, Perales MA, Beyar-Katz O, Shah GL, Shouval R. Treatment failure patterns in early versus late introduction of CAR T-cell therapy in large B-cell lymphoma. Bone Marrow Transplant 2025; 60:491-498. [PMID: 39893244 DOI: 10.1038/s41409-025-02519-z] [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: 10/02/2024] [Revised: 12/16/2024] [Accepted: 01/23/2025] [Indexed: 02/04/2025]
Abstract
CD19-directed chimeric antigen receptor T-cell (CAR-T) therapy has recently been approved as second-line treatment for relapsed/refractory large B-cell lymphoma (LBCL). This study compares patterns of disease relapse and progression across patients receiving CAR-T as second-line (early administration) versus third or subsequent lines (late administration). We analyzed 354 patients treated with Axicabtagene ciloleucel (71%) and Lisocabtagene maraleucel (29%); 80 (23%) received early administration, and 274 (77%) late administration. One-year overall survival was higher in the early group (82% [95% CI 72-93] vs. 71% [95% CI 66-77], p = 0.048). However, the survival benefit was not sustained in multivariable Cox regression modeling and propensity score matching. One-year cumulative incidences of relapse were similar (37% [95% CI 24-50] vs. 43% [95% CI 37-49], p = 0.2), as were 1-year progression-free survival probabilities (62% [95% CI 50-76] vs. 50% [95% CI 44-57], p = 0.14). The early group exhibited a favorable toxicity profile, with lower rate of grade ≥2 cytokine release syndrome (26% vs. 39%, p = 0.031) and reduced cumulative incidence of severe neutropenia (41% [95% CI 30-52] vs. 55% [95% CI 49-60], p = 0.027). Our results indicate favorable outcomes with CAR-T irrespective of treatment line. The equivalence in disease control suggests that CAR-T resistance mechanisms persist in LBCL failing first-line therapy.
Collapse
Affiliation(s)
- Magdalena Corona
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew Ip
- Lymphoma Service, Hackensack Meridian Health, New Jersey, NJ, USA
| | - Samantha Brown
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alejandro Luna
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Hematology Service, Hospital Ramon y Cajal, Madrid, Spain
| | - Hazim Khatib
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Jessica R Flynn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sean M Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ivan Landego
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Giulio Cassanello
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Kai Rejeski
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tsila Zuckerman
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Parastoo B Dahi
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Michael Scordo
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Richard J Lin
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Maciej Kabat
- Lymphoma Service, Hackensack Meridian Health, New Jersey, NJ, USA
| | - Efrat Luttwak
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Emma Pavkovic
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - M Lia Palomba
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jae Park
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gilles Salles
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Heiko Schoder
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Doris Leithner
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Lori A Leslie
- Lymphoma Service, Hackensack Meridian Health, New Jersey, NJ, USA
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Ofrat Beyar-Katz
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Gunjan L Shah
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Roni Shouval
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
12
|
Wang Z, Wang M, Wang M, Zhou R, Deng X, Ouyang X, Chu M, Wei X, Yang L, Liu J, Xu Y. From molecular design to clinical translation: dual-targeted CAR-T strategies in cancer immunotherapy. Int J Biol Sci 2025; 21:2676-2691. [PMID: 40303292 PMCID: PMC12035882 DOI: 10.7150/ijbs.108036] [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: 12/02/2024] [Accepted: 03/10/2025] [Indexed: 05/02/2025] Open
Abstract
The pathogenesis of tumors involves various abnormalities at both the cellular and genetic levels. Chimeric antigen receptor (CAR)-T cell immunotherapy has emerged as a transformative treatment strategy that effectively addresses these challenges. While CAR-T therapy has shown remarkable success in treating hematological malignancies, limitations have been identified, particularly in single antigen-targeting CAR-T therapies. These limitations include antigenic mutation or loss, reduced efficacy against leukemia, and poor results in solid tumors due to factors like low CAR-T cell persistence, limited tumor infiltration, rapid cell exhaustion, the suppressive tumor microenvironment, and heterogeneous tumor antigen expression. In recent years, multi-antigen targeted CAR-T therapies have garnered significant attention for their potential to prevent tumor relapse and progression. This review outlines the fundamental design of dual CAR structures and summarizes the major advancements in both preclinical studies and clinical trials of dual-targeted CAR-T cell therapy, categorized by cancer type. Additionally, it discusses the challenges associated with dual-targeted CAR-T therapy and the strategies to enhance its efficacy and applicability in treating both hematologic and solid tumors. In conclusion, the progress in dual-targeted CAR-T cell therapy presents a promising therapeutic avenue for multiple malignancies, offering insights into future modifications of immunotherapy to advance the field.
Collapse
Affiliation(s)
- Zhenrong Wang
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Mengyi Wang
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Mengting Wang
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Ruijie Zhou
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Xiaotong Deng
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Xin Ouyang
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Minghui Chu
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Xinyu Wei
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Lei Yang
- People's Hospital of Jingyang County, Xianyang, Shaanxi, 713700, China
| | - Jinbiao Liu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, Hubei, 430068, China
- Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
| | - Yao Xu
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| |
Collapse
|
13
|
Bragasin EI, Cheng J, Ford L, Poei D, Ali S, Hsu R. Advances in adoptive cell therapies in small cell lung cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002302. [PMID: 40160238 PMCID: PMC11949692 DOI: 10.37349/etat.2025.1002302] [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: 11/18/2024] [Accepted: 02/10/2025] [Indexed: 04/02/2025] Open
Abstract
Small cell lung cancer (SCLC) is an aggressive tumor characterized by early metastasis and resistance to treatment, making it a prime target for therapeutic investigation. The current standard of care for frontline treatment involves a combination of chemotherapeutic agents and immune checkpoint inhibitors (ICIs), though durability of response remains limited. The genetic heterogeneity of SCLC also complicates the development of new therapeutic options. Adoptive cell therapies show promise by targeting specific mutations in order to increase efficacy and minimize toxicity. There has been significant investigation in three therapeutic classes for application towards SCLC: antibody drug conjugates (ADCs), bispecific T-cell engagers (BiTEs), and chimeric antigen receptor (CAR)-T cell therapies. This review summarizes the recent advances and challenges in the development of adoptive cell therapies. Genetic targets such as delta-like ligand 3 (DLL3), trophoblast cell surface antigen 2 (Trop2), B7-H3 (CD276), gangliosides disialoganglioside GD2 (GD2) and ganglioside GM2 (GM2) have been found to be expressed in SCLC, which makes them prime targets for therapy development. While investigated therapies such as rovalpituzumab tesirine (Rova-T) have failed, several insights from these trials have led to the development of compelling new agents such as sacituzumab govitecan (SG), ifinatamab deruxtecan (I-DXd), tarlatamab, and DLL3-targeted CAR-T cells. Advancing development of molecular testing and improving targeted approaches remain integral to pushing forward the progress of adoptive cell therapies in SCLC.
Collapse
Affiliation(s)
- Eljie Isaak Bragasin
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Justin Cheng
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Lauren Ford
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Darin Poei
- Department of Internal Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Sana Ali
- Department of Medicine, Division of Medical Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Robert Hsu
- Department of Medicine, Division of Medical Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| |
Collapse
|
14
|
Pham-Danis C, Novak AJ, Danis E, McClellan SM, Leach L, Yarnell MC, Ebmeier CC, Tasian SK, Kohler ME. Restoration of LAT activity improves CAR T cell sensitivity and persistence in response to antigen-low acute lymphoblastic leukemia. Cancer Cell 2025; 43:482-502.e9. [PMID: 40068599 PMCID: PMC12002840 DOI: 10.1016/j.ccell.2025.02.008] [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] [Received: 02/21/2024] [Revised: 11/11/2024] [Accepted: 02/07/2025] [Indexed: 04/18/2025]
Abstract
Chimeric antigen receptor (CAR) T cells induce responses in patients with relapsed/refractory leukemia; however, long-term efficacy is frequently limited by relapse. The inability to target antigen-low cells is an intrinsic vulnerability of second-generation CAR T cells and underlies most relapses following CD22BBz CAR T cell therapy. Here, we interrogate CD22BBz CAR signaling in response to low antigen and find inefficient phosphorylation of the linker for activation of T cells (LAT) limiting downstream signaling. To overcome this, we designed the adjunctive LAT-activating CAR T cell (ALA-CART) platform, pairing a second-generation CAR with a LAT-CAR incorporating the intracellular domain of LAT. ALA-CART cells demonstrate reduced differentiation during manufacturing and increased LAT phosphorylation, MAPK signaling, and AP-1 activity. ALA-CART cells show improved cytotoxicity, proliferation, persistence, and efficacy against antigen-low leukemias that were refractory to clinically active CD22BBz CAR T cells. Restoration of LAT signaling through the ALA-CART platform represents a promising strategy for overcoming multiple mechanisms of CAR T cell failure.
Collapse
Affiliation(s)
- Catherine Pham-Danis
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Amanda J Novak
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Etienne Danis
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Biostatistics & Bioinformatics Shared Resource, University of Colorado Cancer Center, Aurora, CO 80045, USA
| | - Samantha M McClellan
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lillie Leach
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michael C Yarnell
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Christopher C Ebmeier
- Proteomics and Mass Spectrometry Core, Department of Biochemistry, University of Colorado, Boulder, CO 80309, USA
| | - Sarah K Tasian
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics & Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - M Eric Kohler
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Division of Blood and Marrow Transplantation & Cellular Therapy, Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO 80045, USA.
| |
Collapse
|
15
|
Zhu J, Zhou J, Liang X, An F, Ding Y, Jiao X, Xiao M, Wu F, Li Y, Xiao H, Pan Y, Wang H, Zhai Z. Elevated CD10 - neutrophils correlate with non-response and poor prognosis of CD19 CAR T-cell therapy for B-cell acute lymphoblastic leukemia. BMC Med 2025; 23:138. [PMID: 40038688 DOI: 10.1186/s12916-025-03968-5] [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] [Received: 09/04/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND The primary challenges in CD19-specific chimeric antigen receptor T-cell (CD19 CAR T) therapy for patients with refractory/relapsed B-cell acute lymphoblastic leukemia (r/r B-ALL) are non-response and relapse; it is urgent to reveal these mechanisms. Neutrophils play a critical role in the immunosuppressive tumor microenvironment (TME), which can hinder CAR T efficacy. Our previous research identified a subset of immunosuppressive neutrophils with a special phenotype (CD14-CD10-CD45-HLA-DR-SSC++, termed CD10- neuts), which suppress T cell function. Therefore, we speculate that CD10- neuts may also influence CAR T efficacy, and this study aims to clinically validate this hypothesis. METHODS We enrolled 44 patients with r/r B-ALL undergoing CD19 CAR T therapy and 47 healthy controls (HCs). Peripheral blood samples were obtained prior to CAR T infusion to detect CD10- neuts levels by flow cytometry. Key parameters included the percentage of CD10- neuts in neutrophils (CD10- neuts/neutrophils), in all nucleated cells (CD10- neuts/nucleated cells), and the absolute count of CD10- neuts. We analyzed the correlations between these indicators and therapeutic response, relapse-free survival (RFS), overall survival (OS), and CAR T cell persistence time. RESULTS CD10- neuts levels were significantly elevated in patients with r/r B-ALL compared to HCs. Additionally, non-responding patients exhibited higher CD10- neuts levels than those in remission. Specifically, CD10- neuts/neutrophils, CD10- neuts/nucleated cells, and absolute CD10- neuts count were 64.44% vs. 25.43% (p = 0.004), 28.61% vs. 9.81% (p = 0.018), and 766.1/μL vs. 152.9/μL (p = 0.04), respectively. Among these indices, only CD10- neuts/neutrophils emerged as an independent risk factor for CAR T response (OR = 19.8, p = 0.013), relapse (HR = 4.704, p = 0.004), and survival (HR = 6.417, p = 0.001). Patients with CD10- neuts/neutrophils ≥ 21.57% demonstrated significantly shorter RFS and OS compared to those with lower levels (p = 0.001; p = 0.0002). Furthermore, CD10- neuts/neutrophils were negatively correlated with the persistence time of CAR T cells. CONCLUSIONS As one of the key factors in the TME, abnormally elevated CD10- neuts correlate with CAR T therapy resistance. Targeting these neutrophils could enhance the effectiveness of CAR T treatment.
Collapse
Affiliation(s)
- Jinli Zhu
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
| | - Ji Zhou
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
- School of Nursing, Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
| | - Xue Liang
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
- Hematology Diagnosis and Treatment Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
| | - Furun An
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
- Hematology Diagnosis and Treatment Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
| | - Yangyang Ding
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
| | - Xunyi Jiao
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
| | - Meng Xiao
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
- Hematology Diagnosis and Treatment Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
| | - Fan Wu
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
- Hematology Diagnosis and Treatment Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
| | - Yingwei Li
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
- Hematology Diagnosis and Treatment Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
| | - Hao Xiao
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
- Hematology Diagnosis and Treatment Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
| | - Ying Pan
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
- Hematology Diagnosis and Treatment Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China
| | - Huiping Wang
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China.
- Hematology Diagnosis and Treatment Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China.
| | - Zhimin Zhai
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China.
- Hematology Diagnosis and Treatment Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China.
- Center of Hematology Research, Anhui Medical University, Hefei, Anhui, 230000, People's Republic of China.
- Institute of Hematology and Clinical Immunology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People's Republic of China.
| |
Collapse
|
16
|
Du Y, Yang Y, Zheng B, Zhang Q, Zhou S, Zhao L. Finding a needle in a haystack: functional screening for novel targets in cancer immunology and immunotherapies. Oncogene 2025; 44:409-426. [PMID: 39863748 PMCID: PMC11810799 DOI: 10.1038/s41388-025-03273-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/06/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Genome-wide functional genetic screening has been widely used in the biomedicine field, which makes it possible to find a needle in a haystack at the genetic level. In cancer research, gene mutations are closely related to tumor development, metastasis, and recurrence, and the use of state-of-the-art powerful screening technologies, such as clustered regularly interspaced short palindromic repeat (CRISPR), to search for the most critical genes or coding products provides us with a new possibility to further refine the cancer mapping and provide new possibilities for the treatment of cancer patients. The use of CRISPR screening for the most critical genes or coding products has further refined the cancer atlas and provided new possibilities for the treatment of cancer patients. Immunotherapy, as a highly promising cancer treatment method, has been widely validated in the clinic, but it could only meet the needs of a small proportion of cancer patients. Finding new immunotherapy targets is the key to the future of tumor immunotherapy. Here, we revisit the application of functional screening in cancer immunology from different perspectives, from the selection of diverse in vitro and in vivo screening models to the screening of potential immune checkpoints and potentiating genes for CAR-T cells. The data will offer fresh therapeutic clues for cancer patients.
Collapse
Affiliation(s)
- Yi Du
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second Hospital, State Key Laboratory of Biotherapy, and Department of Neurosurgery, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, P. R. China
| | - Yang Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second Hospital, State Key Laboratory of Biotherapy, and Department of Neurosurgery, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, P. R. China
| | - Bohao Zheng
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second Hospital, State Key Laboratory of Biotherapy, and Department of Neurosurgery, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, P. R. China
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Qian Zhang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second Hospital, State Key Laboratory of Biotherapy, and Department of Neurosurgery, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, P. R. China.
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second Hospital, State Key Laboratory of Biotherapy, and Department of Neurosurgery, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, P. R. China.
| | - Linjie Zhao
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second Hospital, State Key Laboratory of Biotherapy, and Department of Neurosurgery, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, P. R. China.
| |
Collapse
|
17
|
Miklos DB, Riedell PA, Bokun A, Chavez JC, Schuster SJ. Leveraging the Immunomodulatory Potential of Ibrutinib for Improved Outcomes of T Cell-Mediated Therapies of B Cell Malignancies: A Narrative Review. Target Oncol 2025; 20:217-234. [PMID: 40035913 PMCID: PMC11933223 DOI: 10.1007/s11523-025-01133-9] [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: 02/10/2025] [Indexed: 03/06/2025]
Abstract
Standard treatment options for B cell malignancies include immunochemotherapies and/or targeted therapies, which often provide temporary disease remission. However, many patients do not achieve complete remission with these treatments, develop resistance, and eventually experience disease relapse. New immunomodulatory treatments, such as T cell-based therapies, show promise in treating various types of blood cancers, including B cell malignancies. However, their effectiveness is often limited by the immunosuppressive tumor microenvironment and altered function of patient-derived T cells. Ibrutinib, a Bruton tyrosine kinase inhibitor, has been shown to restore immune balance and function in patients with chronic lymphocytic leukemia. Ibrutinib is being studied as adjuvant or combinatorial therapy with chimeric antigen receptor (CAR) T cells or T cell-engaging bispecific antibodies for the treatment of B cell malignancies. Current evidence suggests that ibrutinib could be beneficial when used before, during, or after CAR T cell administration, potentially providing higher complete response rates and reduced toxicity. In conclusion, existing evidence strongly supports the combined use of ibrutinib and T cell therapies. However, additional clinical trials are needed to further validate the effectiveness of this treatment strategy in patients with various B cell malignancies.
Collapse
Affiliation(s)
- David B Miklos
- Stanford University School of Medicine, Stanford, CA, USA
| | - Peter A Riedell
- David and Etta Jonas Center for Cellular Therapy, The University of Chicago, Chicago, IL, USA
| | - Alex Bokun
- Janssen Biotech, Inc., a Johnson & Johnson company, Horsham, PA, USA.
| | - Julio C Chavez
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Stephen J Schuster
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
18
|
Zelikson V, Gurumurthi A, Sawalha Y, Annunzio K, Saha A, Dong N, Qualls D, Amoozgar B, Kahl B, Baird J, Challa P, Huntington SF, Santos J, Bair S, Narkhede M, Li S, Frosch Z, Ho C, Smith SD, Winter A, Landsburg D, Furqan F, Hamadani M, Baird K, Romancik J, Alharthy H, Law J, Bojanini L, Advani R, Hu B, Johnson PC, Grover NS, Merril M, Crombie JL, Shafagati N, Sterling C, Nastoupil LJ, Epperla N, Ayers EC. Loncastuximab in high-risk and heavily pretreated relapsed/refractory diffuse large B-cell lymphoma: a realworld analysis from 21 US centers. Haematologica 2025; 110:706-714. [PMID: 39540227 PMCID: PMC11873705 DOI: 10.3324/haematol.2024.285977] [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: 06/06/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Outcomes in patients with relapsed/refractory (R/R) diffuse large B-cell lymphoma (DLBCL) are poor. Loncastuximab- teserine (Lonca) is an antibody-drug conjugate which was approved by the Food and Drug Administration for the treatment of patients with R/R DLBCL who have received at least two prior lines of therapy, based on the results of the LOTIS-2 trial. However, there are limited data regarding its efficacy in the real-world setting. This retrospective study included 21 US centers and evaluated outcomes of patients with R/R DLBCL treated with Lonca. Our analysis comprises 187 patients with notably higher-risk baseline features compared to those of the LOTIS-2 population, including a higher proportion of patients with bulky disease (17% vs. 0%), high-grade B-cell histology (22% vs. 8%), and increased number of prior lines of therapy (median 4 vs. 3). The complete response rate was 14% and overall response rate was 32%. The median event-free survival and overall survival were 2.1 and 4.6 months, respectively. Those with bulky disease and high-grade B-cell histology had significantly worse outcomes, and those with non-germinal center cell of origin and a complete response to the most recent line of therapy demonstrated superior outcomes. In summary, in this largest retrospective cohort study of Lonca in the real-world setting, the response rates, event-free survival and overall survival were lower than those reported in LOTIS-2, which is likely reflective of its use in higher risk and more heavily pre-treated patients in the real world compared to the patients enrolled on a clinical study.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Male
- Female
- Aged
- Middle Aged
- United States/epidemiology
- Retrospective Studies
- Adult
- Aged, 80 and over
- Immunoconjugates/therapeutic use
- Immunoconjugates/adverse effects
- Immunoconjugates/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Drug Resistance, Neoplasm
- Treatment Outcome
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Benzodiazepines
Collapse
Affiliation(s)
| | - Ashwath Gurumurthi
- MD Anderson Cancer Center, Houston, USA; Memorial Sloan Kettering Cancer Center, New York
| | - Yazeed Sawalha
- The Ohio State University Comprehensive Cancer Center, Columbus
| | | | | | | | | | - Behzad Amoozgar
- Siteman Cancer Center, Washington University of St Louis, St. Louis
| | - Brad Kahl
- Siteman Cancer Center, Washington University of St Louis, St. Louis
| | | | | | | | - Jennifer Santos
- University of Colorado Cancer Center, Anschutz Medical Campus, Denver
| | - Steven Bair
- University of Colorado Cancer Center, Anschutz Medical Campus, Denver
| | | | | | | | - Carrie Ho
- Fred Hutchinson Cancer Center, Seattle
| | | | | | - Daniel Landsburg
- Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | | | | | | | | | - Hanan Alharthy
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore
| | - Jennie Law
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore
| | | | | | - Boyu Hu
- Hunstman Cancer Institute, University of Utah, Salt Lake City
| | | | - Natalie S Grover
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill
| | | | | | - Nazila Shafagati
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore
| | - Cole Sterling
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore
| | | | | | - Emily C Ayers
- University of Virginia Health System, Charlottesville, GA.
| |
Collapse
|
19
|
Ruan L, Wang L. Adoptive cell therapy against tumor immune evasion: mechanisms, innovations, and future directions. Front Oncol 2025; 15:1530541. [PMID: 40094019 PMCID: PMC11906336 DOI: 10.3389/fonc.2025.1530541] [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: 11/19/2024] [Accepted: 02/06/2025] [Indexed: 03/19/2025] Open
Abstract
Tumors employ a range of strategies to evade detection and eradication by the host's immune system. These include downregulating antigen expression, altering antigen presentation processes, and inhibiting immune checkpoint pathways. etc. Adoptive Cell Therapy (ACT) represents a strategy that boosts anti-tumor immunity. This is achieved by amplifying or genetically engineering immune cells, which are either sourced from the patient or a donor, in a laboratory setting. Subsequently, these cells are reintroduced into the patient to bolster their immune response against cancer. ACT has successfully restored anti-tumor immune responses by amplifying the activity of T cells from patients or donors. This review focuses on the mechanisms underlying tumor escape, including alterations in tumor cell antigens, the immunosuppressive tumor microenvironment (TME), and modulation of immune checkpoint pathways. It further explores how ACT can avddress these factors to enhance therapeutic efficacy. Additionally, the review discusses the application of gene-editing technologies (such as CRISPR) in ACT, highlighting their potential to strengthen the anti-tumor capabilities of T cells. Looking forward, the personalized design of ACT, combined with immune checkpoint inhibitors and targeted therapies, is expected to significantly improve treatment outcomes, positioning this approach as a key strategy in the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Liqin Ruan
- Department of Hepatobiliary Surgery, JiuJiang City Key Laboratory of Cell Therapy, JiuJiang No.1 People's Hospital, Jiujiang, Jiangxi, China
| | - Lu Wang
- Department of Oncology, JiuJiang City Key Laboratory of Cell Therapy, JiuJiang No.1 People's Hospital, Jiujiang, Jiangxi, China
| |
Collapse
|
20
|
Yu T, Jiao JH, Wu MF. CAR-T cells in the treatment of multiple myeloma: an encouraging cell therapy. Front Immunol 2025; 16:1499590. [PMID: 40078993 PMCID: PMC11897482 DOI: 10.3389/fimmu.2025.1499590] [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: 09/21/2024] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Multiple myeloma (MM) is a malignant disease of plasma cells that accounts for approximately 10% of all hematological malignancies and is characterized by a clonal proliferation of malignant plasma cells in the bone marrow. Numerous therapeutic strategies, including proteasome inhibitors, immunomodulators, monoclonal antibodies against CD38 and autologous stem cell transplantation, have prolonged the median survival of MM patients. Nevertheless, almost all MM patients suffer disease relapses due to drug resistance and eventually die from MM or MM-related complications. Chimeric antigen receptor (CAR) T-cell therapy is a novel immunotherapy strategy for MM and has shown encouraging results in several clinical trials. However, the use of CAR T-cell therapy for the treatment of MM is still associated with several difficulties, including antigen escape, poor persistence, an immunosuppressive microenvironment, cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, CAR T-cell-associated encephalopathy syndrome, cytopenia, and infections. In this review, we describe in detail the target antigens of CAR T cells in MM. We also comprehensively discuss recent innovations in the development of CAR T cells to improve clinical efficacy and strategies to overcome the limitations of CAR T-cell therapy in MM.
Collapse
Affiliation(s)
| | - Jian-Hang Jiao
- Department of Orthopaedic Medical Center, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin, China
| | - Min-Fei Wu
- Department of Orthopaedic Medical Center, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
21
|
Wang Y, Lv L, Song Y, Wei X, Zhou H, Liu Q, Xu K, Yan D, Zhang C, Liu S, Jin J, Mei H, Niu T, Liang A, Gu R, Ren J, Feng Y, Jin W, Zhou Y, Deng Y, Wang J. Inaticabtagene autoleucel in adult relapsed or refractory B-cell acute lymphoblastic leukemia. Blood Adv 2025; 9:836-843. [PMID: 39626300 PMCID: PMC11872425 DOI: 10.1182/bloodadvances.2024014182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/06/2024] [Indexed: 03/05/2025] Open
Abstract
ABSTRACT Before November 2023, CD19 chimeric antigen receptor (CAR) T-cell therapies had not been approved in China for patients with relapsed or refractory B-cell acute lymphoblastic leukemia (R/R B-ALL), leaving a significant unmet need. In response, inaticabtagene autoleucel (Inati-cel), a novel CD19 CAR T-cell therapy with a distinct single-chain variable fragment (HI19α), was developed and showed promising efficacy in preliminary clinical research. We conducted a phase 2, single-arm, multicenter study of Inati-cel in adult CD19+ R/R B-ALL in China. The primary end point was the overall remission rate (ORR) at the end of month 3. Forty-eight patients who underwent Inati-cel infusion were evaluated for both efficacy and safety. Among them, 34 patients achieved and maintained remission beyond 3 months, with a 3-month ORR of 70.8% (95% confidence interval [CI], 55.9-83.1). The best ORR was 85.4%, with all responders reaching minimal residual disease negativity. With a median follow-up of 23.7 months, the median duration of remission was 20.7 months (95% CI, 6.4 to not reached), and the median overall survival was not reached (95% CI, 13.0 months to not reached). Additionally, grade ≥3 cytokine release syndrome and neurologic events occurred in 12.5% and 6.2% of patients, respectively. The 2-year follow-up data suggest that Inati-cel demonstrates encouraging and durable responses with manageable safety profiles in R/R B-ALL. Based on the data from this pivotal trial, Inati-cel was approved as the first CAR T-cell therapy for adult R/R B-ALL in China and underscores its potential therapeutic benefits for this patient population. This trial was registered at www.ClinicalTrials.gov as #NCT04684147.
Collapse
Affiliation(s)
- Ying Wang
- Leukemia Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center of Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Lulu Lv
- Juventas Cell Therapy Ltd, Tianjin, China
| | - Yongping Song
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xudong Wei
- Department of Hematology, Henan Cancer Hospital, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongsheng Zhou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dongmei Yan
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Cheng Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
| | - Shuangyou Liu
- Department of Hematology, Beijing Gobroad Boren Hospital, Beijing, China
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Heng Mei
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Aibin Liang
- Department of Hematology, Tongji Hospital of Tongji University, Shanghai, China
| | - Runxia Gu
- Leukemia Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center of Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jienan Ren
- Juventas Cell Therapy Ltd, Tianjin, China
| | - Yi Feng
- Juventas Cell Therapy Ltd, Tianjin, China
| | - Wei Jin
- Juventas Cell Therapy Ltd, Tianjin, China
| | - Yan Zhou
- Juventas Cell Therapy Ltd, Tianjin, China
| | | | - Jianxiang Wang
- Leukemia Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center of Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
22
|
Le X, Zhang Y, Ma J. Comprehensive analysis of adverse events associated with T-cell engagers using the FAERS database. Expert Opin Drug Saf 2025:1-10. [PMID: 39982395 DOI: 10.1080/14740338.2025.2470875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND T-cell engagers (TCEs) are transformative immunotherapies with significant potential in treating hematologic malignancies and solid tumors. However, their real-world safety profiles remain inadequately characterized. RESEARCH DESIGN AND METHODS Using the FDA Adverse Event Reporting System (FAERS) database (October 2019 - September 2024, 8,747,158 reports), we analyzed adverse events (AEs) associated with nine TCEs. Disproportionality analysis identified overreported AEs, with 11,963 unique reports analyzed after deduplication. RESULTS Blinatumomab was the most reported TCE (n = 4,950), and Tarlatamab the least (n = 185). Predominant AEs included immune system disorders, particularly cytokine release syndrome (IC025 range: 6.08-7.47). Drug-specific signals included reproductive system and breast disorders (IC025: 2.74) and vascular disorders (IC025: 2.25) with Tebentafusp, renal and urinary disorders with Epcoritamab (IC025: 1.84), and eye disorders with Elranatamab (IC025: 1.81). Novel AEs were also uncovered, including second malignant neoplasms, vasogenic cerebral edema with Mosunetuzumab (IC025: 5.77, ROR025: 56.29), and hydronephrosis with Epcoritamab (IC025: 7.50, ROR025: 180.70). Early-onset events (0.5-9.5 days) were linked to four TCEs, while delayed-onset events (>20 days) were linked to five others. CONCLUSIONS This study highlights diverse AE profiles of TCEs, providing insights for clinicians to optimize their safe use in practice.
Collapse
Affiliation(s)
- Xiangyang Le
- Department of Pharmacy, Yiyang Central Hospital, Yiyang, Hunan, China
| | - Yefu Zhang
- Quality Control Department, Yiyang Central Hospital, Yiyang, Hunan, China
| | - Junlong Ma
- Center of Clinical Pharmacology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
23
|
Nasiri F, Safarzadeh Kozani P, Salem F, Mahboubi Kancha M, Dashti Shokoohi S, Safarzadeh Kozani P. Mechanisms of antigen-dependent resistance to chimeric antigen receptor (CAR)-T cell therapies. Cancer Cell Int 2025; 25:64. [PMID: 39994651 PMCID: PMC11849274 DOI: 10.1186/s12935-025-03697-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/14/2025] [Indexed: 02/26/2025] Open
Abstract
Cancer immunotherapy has reshaped the landscape of cancer treatment over the past decades. Genetic manipulation of T cells to express synthetic receptors, known as chimeric antigen receptors (CAR), has led to the creation of tremendous commercial and therapeutic success for the treatment of certain hematologic malignancies. However, since the engagement of CAR-T cells with their respective antigens is solely what triggers their cytotoxic reactions against target cells, the slightest changes to the availability and/or structure of the target antigen often result in the incapacitation of CAR-T cells to enforce tumoricidal responses. This results in the resistance of tumor cells to a particular CAR-T cell therapy that requires meticulous heeding to sustain remissions in cancer patients. In this review, we highlight the antigen-dependent resistance mechanisms by which tumor cells dodge being recognized and targeted by CAR-T cells. Moreover, since substituting the target antigen is the most potent strategy for overcoming antigen-dependent disease relapse, we tend to highlight the current status of some target antigens that might be considered suitable alternatives to the currently available antigens in various cancers. We also propose target antigens whose targeting might reduce the off-tumor adverse events of CAR-T cells in certain malignancies.
Collapse
Affiliation(s)
- Fatemeh Nasiri
- Department of Internal Medicine, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran
| | - Pouya Safarzadeh Kozani
- Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran.
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Faeze Salem
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maral Mahboubi Kancha
- Faculty of Engineering and Science, School of Science, University of Greenwich, Chatham Maritime, Chatham, Kent, ME4 4TB, UK
| | | | - Pooria Safarzadeh Kozani
- Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran.
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
24
|
Ma M, Liu C, Jiang L, Liu D, Zhang P, Tao M, Zhang M, Gong J, Peng Z, Zhang X, Li J, Zheng C, Deng M, Shen L, Qi C. Exploring the therapeutic efficacy difference in claudin18.2-targeted cell therapy revealed by single-cell sequencing. iScience 2025; 28:111768. [PMID: 39925434 PMCID: PMC11804729 DOI: 10.1016/j.isci.2025.111768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 09/02/2024] [Accepted: 01/06/2025] [Indexed: 02/11/2025] Open
Abstract
Chimeric antigen receptor T cell (CAR T) therapy has been successfully used to treat hematological malignancies. Nonetheless, its application to solid tumors remains challenging. Our previous analysis of the ongoing clinical trial (NCT03874897) demonstrated promising results in patients with advanced CLDN18.2-positive gastric cancer who received CT041 CAR T treatment. Here, we collected peripheral blood and ascites from five patients from the clinical trial 3 and 7 days (d) after CT041 infusion. Patients with a high proportion of naïve-like T cells were more likely to benefit from CT041 treatment. We found that high expression of CLDN18 in ascites epithelial cells correlated with a favorable prognosis, whereas ascites epithelial cells with high MYC expression and strong interactions between tumor cells and T cells were adverse prognostic factors for CT041 treatment. These findings may provide theoretical evidence for the screening of populations that can benefit from CAR T therapy and improve the efficacy of CAR T therapy.
Collapse
Affiliation(s)
- Mingyang Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
| | - Chang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
| | - Lei Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
| | - Dan Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
| | - Panpan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
| | - Min Tao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
| | - Miao Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
| | - Jifang Gong
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhi Peng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiaotian Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jian Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Chunhong Zheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
- International Cancer Institute, Peking University, No. 38 Xueyuan Road, Beijing 100191, China
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Mi Deng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
- International Cancer Institute, Peking University, No. 38 Xueyuan Road, Beijing 100191, China
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lin Shen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Changsong Qi
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
25
|
Wang Z, Han M, Wang Y, Wang N, Yang Y, Shao B, Miao Q, Shi Z, Yan F, Feng S. UiO-66 MOFs-Based "Epi-Nano-Sonosensitizer" for Ultrasound-Driven Cascade Immunotherapy against B-Cell Lymphoma. ACS NANO 2025; 19:6282-6298. [PMID: 39920081 DOI: 10.1021/acsnano.4c15761] [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: 02/09/2025]
Abstract
B-cell lymphoma (BCL) is a hematological malignancy with high heterogeneity and represents an aggressive proliferation of mature B-cells. Despite the initial success of traditional treatments for BCL in clinical trials, a majority of patients eventually develop resistance to therapy and have poor clinical outcomes. Epigenetic dysregulation is a major contributor to the pathogenesis of BCL, and therapies targeting epigenetic pathways is a promising alternative strategy for treating BCL. Herein, we developed a metal-organic framework (MOF)-based nano-sonosensitizer for ultrasound-driven cascade immunotherapy against BCL. The nano-sonosensitizer was synthesized by encapsulating copper complex of the m6A-mRNA demethylase inhibitor into UiO-66-NH2, which possesses a Z-scheme heterostructure and allows efficient electron-hole pair separation for generating reactive oxygen species (ROS) under ultrasound activation. These CuR@UiO66 sonosensitizers were functionalized with mPEG-PO3 and anti-CD19 antibody, and the resulting CRUPPA19 particles could specifically accumulate in the BCL tissue and also target lymphoma cells that infiltrated into the bone marrow. Once internalized, CRUPPA19 could induce intracellular ROS production and apoptosis under ultrasound irradiation. Subsequently, ultrasonic stimulation triggered autophagy-mediated release of Cu and Rhein from CRUPPA19, thereby increasing protein lipoylation and global mRNA methylation, which led to cuproptosis and the transcriptional repression PDL1, respectively. These cascades synergistically induced immunogenic cell death in the tumors and promoted activation of CD8+ T cells, eventually leading to an antilymphoma immune response. CRUPPA19-mediated sono-immunotherapy not only eliminated the primary and metastatic lymphomas but also cleared lymphoma cells from the bone marrow. This study provided an insight into a MOF-based nanoepigenetic therapy platform with ultrasound-triggered cascade amplification for enhanced antihematological tumor immunity.
Collapse
Affiliation(s)
- Zhihua Wang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Mingda Han
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Yiqiao Wang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Ning Wang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Yilin Yang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Bingru Shao
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Qiannan Miao
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Zhan Shi
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Fei Yan
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Shouhua Feng
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| |
Collapse
|
26
|
Caratelli S, De Paolis F, Silvestris DA, Baldari S, Salvatori I, Tullo A, Lanzilli G, Gurtner A, Ferri A, Valle C, Padovani S, Cesarini V, Sconocchia T, Cifaldi L, Arriga R, Spagnoli GC, Ferrone S, Venditti A, Rossi P, Pesole G, Toietta G, Sconocchia G. The CD64/CD28/CD3ζ chimeric receptor reprograms T-cell metabolism and promotes T-cell persistence and immune functions while triggering antibody-independent and antibody-dependent cytotoxicity. Exp Hematol Oncol 2025; 14:17. [PMID: 39962623 PMCID: PMC11834217 DOI: 10.1186/s40164-025-00601-2] [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: 10/30/2024] [Accepted: 01/19/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Recent studies have shown that CD32/CD8a/CD28/CD3ζ chimeric receptor cells directly kill breast cancer cells, suggesting the existence of cell surface myeloid FcγR alternative ligands (ALs). Here, we investigated the metabolism, ALs, cytotoxicity, and immunoregulatory functions of CD64/CD28/CD3ζ in colorectal cancer (CRC) and squamous cell carcinoma of the head and neck. METHODS The CD64/CD28/CD3ζ -SFG retroviral vector was used to produce viruses for T-cell transduction. T-cell expansion and differentiation were monitored via flow cytometry. Gene expression was assessed by RNA-seq. Bioenergetics were documented on a Seahorse extracellular flux analyzer. CD64/CD28/CD3ζ polarization was identified via confocal microscopy. Cytotoxicity was determined by MTT assay and bioluminescent imaging, and flow cytometry. Tridimensional antitumor activity of CD64/CD28/CD3ζ T cells was achieved by utilizing HCT116-GFP 3D spheroids via the IncuCyte S3 Live-Cell Analysis system. The intraperitoneal distribution and antitumor activity of NIR-CD64/CD28/CD3ζ and NIR-nontransduced T cells were investigated in CB17-SCID mice bearing subcutaneous FaDu Luc + cells by bioluminescent and fluorescent imaging. IFNγ was assessed by ELISA. RESULTS Compared to CD16/CD8a/CD28/CD3ζ T cells, CD32/CD8a/CD28/CD3ζ T cells, and non-transduced T cells, CD64/CD28/CD3ζ T cells exhibited the highest levels of cell expansion and persistence capacity. A total of 235 genes linked to cell division and 52 genes related to glycolysis were overexpressed. The glycolytic phenotype was confirmed by functional in vitro studies accompanied by preferential T-cell effector memory differentiation. Interestingly, oxamic acid was found to inhibit CD64-CR T cell proliferation, indicating the involvement of lactate. Upon CD64/CD28/CD3ζ T-cell conjugation with CRC cells, CD64/CD28/CD3ζ cells polarize at immunological synapses, leading to CRC cell death. CD64/CD28/CD3ζ T cells kill SCCHN cells, and in combination with the anti-B7-H3 mAb (376.96) or anti-EGFR mAb, these cells trigger antibody-dependent cellular cytotoxicity (ADCC) in vitro under 2D and 3D conditions. The 376.96 mAb combined with CD64/CD28/CD3ζ T cells had anti-SCCHN activity in vivo. In addition, they induce the upregulation of PD-L1 and HLA-DR expression in cancer cells via IFNγ. PD-L1 positive SCCHN cells in combination with anti-PD-L1 mAb and CD64-CR T cells were killed by ADCC, which enhanced direct cytotoxicity. These findings indicate that the glycolytic phenotype is involved in CD64-CR T cell proliferation/expansion. These cells mediate long-lasting HLA-independent cytotoxicity and ADCC in CRC and SCCHN cells. CONCLUSIONS CD64/CD28/CD3ζ T cells could significantly impact the rational design of personalized studies to treat CRC and SCCHN and the identification of novel FcγR ALs in cancer and healthy cells.
Collapse
Affiliation(s)
- Sara Caratelli
- Department of Biomedicine, Institute of Translational Pharmacology, Italian National Research Council (CNR), Via Fosso del Cavaliere 100, Rome, 00133, Italy
| | - Francesca De Paolis
- Department of Biomedicine, Institute of Translational Pharmacology, Italian National Research Council (CNR), Via Fosso del Cavaliere 100, Rome, 00133, Italy
| | | | - Silvia Baldari
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Apollonia Tullo
- Department of Biomedicine, Institute of Biomembranes Bioenergetics and Molecular Biotechnologies, National Research Council (CNR), Bari, Italy
| | - Giulia Lanzilli
- Department of Biomedicine, Institute of Translational Pharmacology, Italian National Research Council (CNR), Via Fosso del Cavaliere 100, Rome, 00133, Italy
| | - Aymone Gurtner
- Department of Biomedicine, Institute of Translational Pharmacology, Italian National Research Council (CNR), Via Fosso del Cavaliere 100, Rome, 00133, Italy
| | - Alberto Ferri
- Department of Biomedicine, Institute of Translational Pharmacology, Italian National Research Council (CNR), Via Fosso del Cavaliere 100, Rome, 00133, Italy
- IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Cristiana Valle
- Department of Biomedicine, Institute of Translational Pharmacology, Italian National Research Council (CNR), Via Fosso del Cavaliere 100, Rome, 00133, Italy
- IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Simona Padovani
- Department of Biomedicine, Institute of Translational Pharmacology, Italian National Research Council (CNR), Via Fosso del Cavaliere 100, Rome, 00133, Italy
| | - Valeriana Cesarini
- Department of Biomedicine, Institute of Translational Pharmacology, Italian National Research Council (CNR), Via Fosso del Cavaliere 100, Rome, 00133, Italy
- Saint Camillus, International Medical University (UNICAMILLUS), Rome, Italy
| | - Tommaso Sconocchia
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Loredana Cifaldi
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Roberto Arriga
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Giulio Cesare Spagnoli
- Department of Biomedicine, Institute of Translational Pharmacology, Italian National Research Council (CNR), Via Fosso del Cavaliere 100, Rome, 00133, Italy
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, NA, USA
| | - Adriano Venditti
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Piero Rossi
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Graziano Pesole
- Dipartimento di Bioscienze, Biotecnologie e Ambiente, University of Bari, Bari, Italy
- Department of Biomedicine, Institute of Biomembranes Bioenergetics and Molecular Biotechnologies, National Research Council (CNR), Bari, Italy
| | - Gabriele Toietta
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giuseppe Sconocchia
- Department of Biomedicine, Institute of Translational Pharmacology, Italian National Research Council (CNR), Via Fosso del Cavaliere 100, Rome, 00133, Italy.
- Saint Camillus, International Medical University (UNICAMILLUS), Rome, Italy.
| |
Collapse
|
27
|
Tufail M, Jiang CH, Li N. Tumor dormancy and relapse: understanding the molecular mechanisms of cancer recurrence. Mil Med Res 2025; 12:7. [PMID: 39934876 PMCID: PMC11812268 DOI: 10.1186/s40779-025-00595-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 01/26/2025] [Indexed: 02/13/2025] Open
Abstract
Cancer recurrence, driven by the phenomenon of tumor dormancy, presents a formidable challenge in oncology. Dormant cancer cells have the ability to evade detection and treatment, leading to relapse. This review emphasizes the urgent need to comprehend tumor dormancy and its implications for cancer recurrence. Despite notable advancements, significant gaps remain in our understanding of the mechanisms underlying dormancy and the lack of reliable biomarkers for predicting relapse. This review provides a comprehensive analysis of the cellular, angiogenic, and immunological aspects of dormancy. It highlights the current therapeutic strategies targeting dormant cells, particularly combination therapies and immunotherapies, which hold promise in preventing relapse. By elucidating these mechanisms and proposing innovative research methodologies, this review aims to deepen our understanding of tumor dormancy, ultimately facilitating the development of more effective strategies for preventing cancer recurrence and improving patient outcomes.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Can-Hua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, 410008, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Institute of Oral Precancerous Lesions, Central South University, Changsha, 410008, China.
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
28
|
Mitsuno K, Suematsu M, Naito Y, Mayumi A, Yoshida H, Osone S, Imamura T, Nakazawa Y, Yagyu S, Iehara T. Selective JAK2 pathway inhibition enhances anti-leukemic functionality in CD19 CAR-T cells. Cancer Immunol Immunother 2025; 74:79. [PMID: 39891728 PMCID: PMC11787079 DOI: 10.1007/s00262-024-03927-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 12/18/2024] [Indexed: 02/03/2025]
Abstract
The integration of molecular targeted therapeutics with chimeric antigen receptor T (CAR-T) cell therapy represents a novel strategy to amplify the anti-tumor efficacy of immunotherapy. While CD19-targeted CAR-T cells and Janus kinase (JAK) inhibitors have independently shown efficacy against certain B-cell leukemias, such as Philadelphia chromosome-like acute lymphoblastic leukemia, the concurrent use of JAK1/2 inhibitors, such as ruxolitinib, has been implicated in reducing CAR-T cell potency by inhibiting the JAK1-dependent T cell activation pathway. This study explores the combinatorial use of a selective type II JAK2 inhibitor, CHZ868, with CD19 CAR-T cells, revealing a synergistic enhancement of anti-leukemic activity across B-cell tumor models irrespective of JAK2 mutational status. CHZ868-mediated JAK2 inhibition did not induce the exhaustion of CAR-T cells, maintaining efficacy over repeated tumor challenges and significantly extending survival in mouse models engrafted with JAK2 inhibitor-resistant leukemia cells (median survival, CD19 CAR-T + CHZ868 vs. CD19 CAR-T + DMSO: 32 days vs. 26 days, p = 0.0303). Transcriptomic analyses suggest that CHZ868 impedes CAR-T cell differentiation while preserving their proliferative capacity, a crucial factor in maintaining CAR-T cell functionality. Therefore, the selective inhibition of the JAK2 pathway may potentiate CAR-T cell therapy and offer a viable treatment strategy for patients with resistant B-cell leukemias.
Collapse
Affiliation(s)
- Kohei Mitsuno
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masaya Suematsu
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Yuki Naito
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Azusa Mayumi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hideki Yoshida
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shinya Osone
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshihiko Imamura
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yozo Nakazawa
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan
| | - Shigeki Yagyu
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.
- Innovative Research and Liaison Organization, Shinshu University, Matsumoto, Japan.
| | - Tomoko Iehara
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
29
|
Ni Y, Zhang Q, Tang X, Li X, Ye S, Lu Y, Liang A, Li P. Upregulation of CD19 by low-dose chidamide promotes CAR T cells functionality in B-cell non-Hodgkin lymphoma. Discov Oncol 2025; 16:84. [PMID: 39853608 PMCID: PMC11759751 DOI: 10.1007/s12672-025-01810-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 01/13/2025] [Indexed: 01/26/2025] Open
Abstract
B-cell non-Hodgkin lymphoma (B-NHL) is a highly heterogeneous group of lymphopoietic malignancies that account for 85% to 90% of all non-Hodgkin lymphomas. In recent years, CD19 Chimeric antigen receptor T (CAR T) cell immunotherapy has significantly improved the cure rate of B-NHL patients, but there are still some patients who cannot achieve remission after treatment, or relapse after remission. Therefore, it is of great importance to overcome the drug resistance of CD19 CAR T cells after B-NHL treatment and reduce the recurrence rate of CD19 CAR T cells after B-NHL treatment. We found that low concentrations of chidamide did not enhance the ability of CD19 CAR T cells to kill B-NHL cells during and after preparation. B-NHL cells pretreated with chidamide were more likely to be killed by CD19 CAR T cells. CD19 CAR T cells secreted more cytokines (IL-2, TNF-α, and IFN-γ) after co-culture with B-NHL cells pretreated with chidamide. At the same time, the expression of CD19 on B-NHL cell surface was increased by chidamide. In vivo experiments showed that infusion of CD19 CAR T cells after chidamide bridging intervention can enhance the therapeutic effect of B-NHL and prolong the overall survival of mice. This study provides a new direction and theoretical foundation for CD19 CAR T cell therapy in B-NHL.
Collapse
Affiliation(s)
- Ying Ni
- Department of Hematology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qun Zhang
- Department of Digestive Surgical Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaochen Tang
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiuchun Li
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shiguang Ye
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yan Lu
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Aibin Liang
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Ping Li
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
30
|
Zhao ML, Lei YM, Tang JY, Li W, Cao XY, Liang WB, Yuan R, Yang C, Zhuo Y. DNA lesion-gated dumbbell nanodevices enable on-demand activation of the cGAS-STING pathway for enhancing cancer immunotherapy. Chem Sci 2025; 16:1783-1790. [PMID: 39720145 PMCID: PMC11664422 DOI: 10.1039/d4sc06493c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/13/2024] [Indexed: 12/26/2024] Open
Abstract
Utilizing the cGAS-STING pathway to combat immune evasion is one of the most promising strategies for enhancing cancer immunotherapy. However, current techniques for activating the cGAS-STING pathway often face a dilemma, mainly due to the balance between efficacy and safety. Here, we develop a uracil base lesion-gated dumbbell DNA nanodevice (UBLE) that allows on-demand activation and termination of the cGAS-STING pathway in tumor cells, thereby enhancing cancer immunotherapy. The UBLE integrates two deoxyuridines (dU) in the stem for DNA lesion recognition, two locked complementary primer sequences (primers A and B) for DNA self-assembly, and a Förster resonance energy transfer pair (Cy3 and Cy5) attached to the loop for activation assessment. Upon the orthogonal recognition of tumor-specific repair indicators (UDG and APE1), the UBLE undergoes a conformational change to create massive nicked double-stranded DNA (dsDNA) units. These units self-assemble to generate long fluorescent dsDNA structures, permitting selective evaluation and on-demand activation of the cGAS-STING pathway. Furthermore, we demonstrate that the UBLE can effectively activate the cGAS-STING pathway in tumor cells, enhancing NK cell-targeted cancer immunotherapy. This work develops a DNA lesion-gated strategy for on-demand activation and termination of the cGAS-STING pathway, affording an innovative avenue for enhancing cancer immunotherapy.
Collapse
Affiliation(s)
- Mei-Ling Zhao
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| | - Yan-Mei Lei
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 2002127 P. R. China
| | - Jing-Yi Tang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| | - Wen Li
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| | - Xin-Yu Cao
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| | - Wen-Bin Liang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| | - Ruo Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| | - Chaoyong Yang
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 2002127 P. R. China
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 P. R. China
| | - Ying Zhuo
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| |
Collapse
|
31
|
Chen L, Huang R, Huang C, Nong G, Mo Y, Ye L, Lin K, Chen A. Cell therapy for scleroderma: progress in mesenchymal stem cells and CAR-T treatment. Front Med (Lausanne) 2025; 11:1530887. [PMID: 39882532 PMCID: PMC11774712 DOI: 10.3389/fmed.2024.1530887] [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: 11/19/2024] [Accepted: 12/26/2024] [Indexed: 01/31/2025] Open
Abstract
Cell therapy is an emerging strategy for precision treatment of scleroderma. This review systematically summarizes the research progress of mesenchymal stem cell (MSC) and chimeric antigen receptor T cell (CAR-T) therapies in scleroderma and discusses the challenges and future directions for development. MSCs possess multiple functions, including immunomodulation, anti-fibrosis, and promotion of vascular regeneration, all of which can improve multiple pathological processes associated with scleroderma. Studies have demonstrated that MSCs can alleviate skin fibrosis by inhibiting CCL2 production and reducing the recruitment of pathological macrophages; their paracrine effects can exert extensive regulatory functions. CAR-T cell therapy ca specifically target and eliminate autoreactive immune cells, exhibiting enhanced specificity and personalized potential. Different cell therapies may have complementary and synergistic effects in treating scleroderma, such as MSCs exerting their effects through paracrine mechanisms while CAR-T cells specifically eliminate pathological cells. Furthermore, cell-free therapies derived from MSCs, such as extracellular vesicles or exosomes, may help circumvent the limitations of MSC therapy. Although cell therapy has opened new avenues for the precision treatment of scleroderma, it still faces numerous challenges. In the future, it is essential to strengthen integration of basic and clinical research, establish standardized protocols for cell preparation and quality control, develop personalized treatment plans, and rationally combine cell therapy with existing treatment methods to maximize its advantages and improve patient prognosis and quality of life.
Collapse
Affiliation(s)
- Liting Chen
- Department of Rheumatology and Immunology, Qinzhou First People's Hospital, Qinzhou, Guangxi, China
| | - Rongshan Huang
- Department of Rheumatology and Immunology, Qinzhou First People's Hospital, Qinzhou, Guangxi, China
| | - Chaoshuo Huang
- Department of Rheumatology and Immunology, Qinzhou First People's Hospital, Qinzhou, Guangxi, China
| | - Guiming Nong
- Department of Rheumatology and Immunology, Qinzhou First People's Hospital, Qinzhou, Guangxi, China
| | - Yuanyuan Mo
- Department of Rheumatology and Immunology, Qinzhou First People's Hospital, Qinzhou, Guangxi, China
| | - Lvyin Ye
- Department of Rheumatology and Immunology, Qinzhou First People's Hospital, Qinzhou, Guangxi, China
| | - Kunhong Lin
- Department of Rheumatology and Immunology, Qinzhou First People's Hospital, Qinzhou, Guangxi, China
| | - Anping Chen
- Department of Rheumatology and Immunology, Qinzhou First People's Hospital, Qinzhou, Guangxi, China
- Minda Hospital of Hubei Minzu University, Enshi, China
| |
Collapse
|
32
|
Zou Q, Liao K, Li G, Huang X, Zheng Y, Yang G, Luo M, Xue EY, Lan C, Wang S, Shen Y, Luo D, Ng DKP, Liu Q. Photo-metallo-immunotherapy: Fabricating Chromium-Based Nanocomposites to Enhance CAR-T Cell Infiltration and Cytotoxicity against Solid Tumors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2407425. [PMID: 38899741 PMCID: PMC11733712 DOI: 10.1002/adma.202407425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Indexed: 06/21/2024]
Abstract
The infiltration and cytotoxicity of chimeric antigen receptor (CAR)-T cells are crucial for effective elimination of solid tumors. While metallo-immunotherapy is a promising strategy that can activate the antitumor immunity, its role in promoting CAR-T cell therapy remains elusive. The first single-element nanomaterial based on chromium nanoparticles (Cr NPs) for cancer photo-metallo-immunotherapy has been reported previously. Herein, an extended study using biodegradable polydopamine as a versatile carrier for these nanoparticles, enabling synergistic CAR-T cell therapy, is reported. The results show that these nanocomposites with or without further encapsulation of the anticancer drug alpelisib can promote the CAR-T cell migration and antitumor effect. Upon irradiation with near-infrared light, they caused mild hyperthermia that can "warm" the "cold" tumor microenvironment (TME). The administration of B7-H3 CAR-T cells to NOD severe combined immunodeficiency gamma mice bearing a human hepatoma or PIK3CA-mutated breast tumor can significantly inhibit the tumor growth after the induction of tumor hyperthermia by the nanocomposites and promote the secretion of serum cytokines, including IL-2, IFN-γ, and TNF-α. The trivalent Cr3+ ions, which are the major degradation product of these nanocomposites, can increase the CXCL13 and CCL3 chemokine expressions to generate tertiary lymphoid structures (TLSs) in the tumor tissues, facilitating the CAR-T cell infiltration.
Collapse
Affiliation(s)
- Qingshuang Zou
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
- Department of ChemistryThe Chinese University of Hong KongShatin, N.T.Hong Kong999077China
| | - Ke Liao
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
- Institute of Pharmacy and PharmacologySchool of Pharmaceutical ScienceHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001China
| | - Guangchao Li
- Department of HematologyThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhou510317China
| | - Xin Huang
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
| | - Yongwei Zheng
- Research and Development DepartmentGuangzhou Bio‐Gene Technology Co. Ltd.Guangzhou510530China
| | - Gun Yang
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
| | - Min Luo
- Research and Development DepartmentGuangzhou Bio‐Gene Technology Co. Ltd.Guangzhou510530China
| | - Evelyn Y. Xue
- Department of ChemistryThe Chinese University of Hong KongShatin, N.T.Hong Kong999077China
| | - Chuanqing Lan
- Department of ChemistryThe Chinese University of Hong KongShatin, N.T.Hong Kong999077China
| | - Shuai Wang
- Department of ChemistryThe Chinese University of Hong KongShatin, N.T.Hong Kong999077China
| | - Yao Shen
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
| | - Dixian Luo
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
| | - Dennis K. P. Ng
- Department of ChemistryThe Chinese University of Hong KongShatin, N.T.Hong Kong999077China
| | - Quan Liu
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
| |
Collapse
|
33
|
Mestermann K, Garitano-Trojaola A, Hudecek M. Accelerating CAR-T Cell Therapies with Small-Molecule Inhibitors. BioDrugs 2025; 39:33-51. [PMID: 39589646 PMCID: PMC11750903 DOI: 10.1007/s40259-024-00688-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 11/27/2024]
Abstract
Chimeric antigen receptor T-cell therapies have markedly improved the survival rates of patients with B-cell malignancies. However, their efficacy in other hematological cancers, such as acute myeloid leukemia, and in solid tumors has been limited. Key obstacles include the downregulation or loss of antigen expression on cancer cells, restricted accessibility to target cells, and the poor persistence of these "living drugs" because of the highly immunosuppressive tumor microenvironment. Additionally, manufacturing these immunotherapies presents significant challenges, and patients frequently experience side effects such as cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. This review emphasizes the potential of small-molecule inhibitors, many of which are already approved for clinical use, to facilitate chimeric antigen receptor T-cell manufacturing, enhance their anti-tumor efficacy, and mitigate their side effects. Although substantial work remains, the robust pre-clinical data and the growing clinical interest suggest significant promise for using cancer signaling pathway inhibitors to enhance and refine chimeric antigen receptor T-cell therapy for both hematological and solid tumors. Exploring these combination strategies could lead to more effective therapies, offering new hope for patients with resistant forms of cancer.
Collapse
Affiliation(s)
- Katrin Mestermann
- Medizinische Klinik und Poliklinik II, Lehrstuhl für zelluläre Immuntherapie, Universitätsklinikum Würzburg, Haus E4-/Raum 4.06, Versbacher Straße 5, 97078, Würzburg, Germany.
- Fraunhofer-Institut für Zelltherapie und Immunologie, Außenstelle Zelluläre Immuntherapie, Würzburg, Germany.
| | - Andoni Garitano-Trojaola
- Medizinische Klinik und Poliklinik II, Lehrstuhl für zelluläre Immuntherapie, Universitätsklinikum Würzburg, Haus E4-/Raum 4.06, Versbacher Straße 5, 97078, Würzburg, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II, Lehrstuhl für zelluläre Immuntherapie, Universitätsklinikum Würzburg, Haus E4-/Raum 4.06, Versbacher Straße 5, 97078, Würzburg, Germany
- Fraunhofer-Institut für Zelltherapie und Immunologie, Außenstelle Zelluläre Immuntherapie, Würzburg, Germany
| |
Collapse
|
34
|
Ahmed G, Hamadani M, Al-Juhaishi T. The potential of antibody-drug conjugates for effective therapy in diffuse large B-cell lymphoma. Expert Opin Biol Ther 2025; 25:161-173. [PMID: 39798075 DOI: 10.1080/14712598.2025.2453524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/13/2025]
Abstract
INTRODUCTION Antibody-drug conjugates (ADCs) are a rapidly evolving class of anti-cancer drugs with a significant impact on management of hematological malignancies including diffuse large B-cell lymphoma (DLBCL). ADCs combine a cytotoxic drug (a.k.a. payload) attached through a linker to a monoclonal antibody specific to a particular cancer antigen. Payloads include microtubule disruptors or DNA damaging chemicals. After attaching to the antigen, the ADCs are internalized, and the payload is dissociated from ADC by lysozymes and delivered to the intended site for exerting cytotoxic effects. This unique molecular design permits a better balance of efficacy and safety. Loncastuximab tesirine and polatuzumab vedotin are two ADCs approved in the U.S.A. for treatment of DLBCL. AREAS COVERED This review covers the efficacy and safety data of these two drugs. We will review new ADC-based combination regimens and novel constructs in development. EXPERT OPINION ADCs have made a significant impact in improving outcomes of DLBCL patients. Both polatuzumab vedotin and loncastuximab tesirine are established as useful therapeutics options, with polatuzumab vedotin currently approved in first line and relapsed/refractory setting, while loncastuximab tesirine is approved in relapsed setting. ADCs are effective with tolerable safety profile and currently many more ADCs are undergoing clinical trials.
Collapse
MESH Headings
- Humans
- Immunoconjugates/adverse effects
- Immunoconjugates/administration & dosage
- Immunoconjugates/therapeutic use
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/immunology
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/therapeutic use
- Animals
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/therapeutic use
- Benzodiazepines
Collapse
Affiliation(s)
- Gulrayz Ahmed
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mehdi Hamadani
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | | |
Collapse
|
35
|
Firuzpour F, Saleki K, Aram C, Rezaei N. Nanocarriers in glioblastoma treatment: a neuroimmunological perspective. Rev Neurosci 2024:revneuro-2024-0097. [PMID: 39733347 DOI: 10.1515/revneuro-2024-0097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 12/08/2024] [Indexed: 12/31/2024]
Abstract
Glioblastoma multiforme (GBM) is the most fatal brain tumor with a poor prognosis with current treatments, mainly because of intrinsic resistance processes. GBM is also referred to as grade 4 astrocytoma, that makes up about 15.4 % of brain cancers globally as well as 60-75 % of astrocytoma. The most prevalent therapeutic choices for GBM comprise surgery in combination with radiotherapy and chemotherapy, providing patients with an average survival of 6-14 months. Nanocarriers provide various benefits such as enhanced drug solubility, biocompatibility, targeted activity, as well as minimized side effects. In addition, GBM treatment comes with several challenges such as the presence of the blood-brain barrier (BBB), blood-brain tumor barrier (BBTB), overexpressed efflux pumps, infiltration, invasion, drug resistance, as well as immune escape due to tumor microenvironment (TME) and cancer stem cells (CSC). Recent research has focused on nanocarriers due to their ability to self-assemble, improve bioavailability, provide controlled release, and penetrate the BBB. These nano-based components could potentially enhance drug accumulation in brain tumor tissues and reduce systemic toxicity, making them a compelling solution for GBM therapy. This review captures the complexities associated with multi-functional nano drug delivery systems (NDDS) in crossing the blood-brain barrier (BBB) and targeting cancer cells. In addition, it presents a succinct overview of various types of targeted multi-functional nano drug delivery system (NDDS) which has exhibited promising value for improving drug delivery to the brain.
Collapse
Affiliation(s)
- Faezeh Firuzpour
- USERN Office, Babol University of Medical Sciences, 47176-41367, Babol, Iran
- Student Research Committee, Babol University of Medical Sciences, 47176-41367, Babol, Iran
| | - Kiarash Saleki
- USERN Office, Babol University of Medical Sciences, 47176-41367, Babol, Iran
- Student Research Committee, Babol University of Medical Sciences, 47176-41367, Babol, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
| | - Cena Aram
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, 15719-14911, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
- Department of Immunology, School of Medicine, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
| |
Collapse
|
36
|
Kaviyarasan V, Das A, Deka D, Saha B, Banerjee A, Sharma NR, Duttaroy AK, Pathak S. Advancements in immunotherapy for colorectal cancer treatment: a comprehensive review of strategies, challenges, and future prospective. Int J Colorectal Dis 2024; 40:1. [PMID: 39731596 DOI: 10.1007/s00384-024-04790-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/15/2024] [Indexed: 12/30/2024]
Abstract
PURPOSE Colorectal cancer (CRC) remains one of the leading causes of cancer-related mortality worldwide. Metastatic colorectal cancer (mCRC) continues to present significant challenges, particularly in patients with proficient mismatch repair/microsatellite stable (pMMR/MSS) tumors. This narrative review aims to provide recent developments in immunotherapy for CRC treatment, focusing on its efficacy and challenges. METHODS This review discussed the various immunotherapeutic strategies for CRC treatment, including immune checkpoint inhibitors (ICIs) targeting PD-1 and PD-L1, combination therapies involving ICIs with other modalities, chimeric antigen receptor T-cell (CAR-T) cell therapy, and cancer vaccines. The role of the tumor microenvironment and immune evasion mechanisms was also explored to understand their impact on the effectiveness of these therapies. RESULTS This review provides a comprehensive update of recent advancements in immunotherapy for CRC, highlighting the potential of various immunotherapeutic approaches, including immune checkpoint inhibitors, combination therapies, CAR-T therapy, and vaccination strategies. The results of checkpoint inhibitors, particularly in patients with MSI-H/dMMR tumors, which have significant improvements in survival rates have been observed. Furthermore, this review also addresses the challenges faced in treating pMMR/MSS CRC, which remains resistant to immunotherapy. CONCLUSION Immunotherapy plays a significant role in the treatment of CRC, particularly in patients with MSI-H/dMMR tumors. However, many challenges remain, especially in treating pMMR/MSS CRC. This review discussed the need for further research into combination therapies, biomarker development, CAR-T cell therapy, and a deeper understanding of immune evasion mechanisms for CRC treatment.
Collapse
Affiliation(s)
- Vaishak Kaviyarasan
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Alakesh Das
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Dikshita Deka
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Biki Saha
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India.
| | - Neeta Raj Sharma
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India.
| |
Collapse
|
37
|
Yao CD, Davis KL. Correlative studies reveal factors contributing to successful CAR-T cell therapies in cancer. Cancer Metastasis Rev 2024; 44:15. [PMID: 39625613 DOI: 10.1007/s10555-024-10232-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024]
Abstract
Cellular and targeted immunotherapies have revolutionized cancer treatments in the last several decades. Successful cellular therapies require both effective and durable cytotoxic activity from the immune cells as well as an accessible and susceptible response from targeted cancer cells. Correlative studies from clinical trials as well as real-world data from FDA-approved therapies have revealed invaluable insights about immune cell factors and cancer cell factors that impact rates of response and relapse to cellular therapies. This review focuses on the flagship cellular therapy of engineered chimeric antigen receptor T-cells (CAR-T cells). Within the CAR-T cell compartment, we discuss discoveries about T-cell phenotype, transcriptome, epigenetics, cytokine signaling, and metabolism that inform the cell manufacturing process to produce the most effective and durable CAR-T cells. Within the cancer cell compartment, we discuss mechanisms of resistance and relapse caused by mutations, alternative splicing, post-transcriptional modifications, and cellular reprogramming. Continued correlative and mechanistic studies are required to help us further optimize cellular therapies in a variety of malignancies.
Collapse
Affiliation(s)
- Catherine D Yao
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplant and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Kara L Davis
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplant and Regenerative Medicine, Stanford University, Stanford, CA, USA.
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
38
|
Lei T, Wang Y, Zhang Y, Yang Y, Cao J, Huang J, Chen J, Chen H, Zhang J, Wang L, Xu X, Gale RP, Wang L. Leveraging CRISPR gene editing technology to optimize the efficacy, safety and accessibility of CAR T-cell therapy. Leukemia 2024; 38:2517-2543. [PMID: 39455854 PMCID: PMC11588664 DOI: 10.1038/s41375-024-02444-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024]
Abstract
Chimeric Antigen Receptor (CAR)-T-cell therapy has revolutionized cancer immune therapy. However, challenges remain including increasing efficacy, reducing adverse events and increasing accessibility. Use of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) technology can effectively perform various functions such as precise integration, multi-gene editing, and genome-wide functional regulation. Additionally, CRISPR screening using large-scale guide RNA (gRNA) genetic perturbation provides an unbiased approach to understanding mechanisms underlying anti-cancer efficacy of CAR T-cells. Several emerging CRISPR tools with high specificity, controllability and efficiency are useful to modify CAR T-cells and identify new targets. In this review we summarize potential uses of the CRISPR system to improve results of CAR T-cells therapy including optimizing efficacy and safety and, developing universal CAR T-cells. We discuss challenges facing CRISPR gene editing and propose solutions highlighting future research directions in CAR T-cell therapy.
Collapse
Affiliation(s)
- Tao Lei
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Yazhuo Wang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuchen Zhang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Yufei Yang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Jiaying Cao
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Jiansong Huang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Jiali Chen
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Huajing Chen
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Jiayi Zhang
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Luzheng Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Robert Peter Gale
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College of Science, Technology and Medicine, London, UK.
| | - Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
39
|
Zhang L, Norberg SM, Karimipour F, Davies JS, Kuznetsov A, Lassoued W, Burnett D, Homan P, Cam M, Sinkoe A, Xue P, Gulley JL, Hinrichs CS. Adoptive transfer of membrane-restricted IL-12-TCR T cells promotes antigen spreading and elimination of antigen-negative tumor variants. J Immunother Cancer 2024; 12:e009868. [PMID: 39557544 PMCID: PMC11574437 DOI: 10.1136/jitc-2024-009868] [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] [Accepted: 10/26/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Adoptive T-cell therapy has demonstrated clinical activity in B-cell malignancies, offering hope for its application to a broad spectrum of cancers. However, a significant portion of patients with solid tumors experience primary or secondary resistance to this treatment modality. Target antigen loss resulting either from non-uniform antigen expression or defects in antigen processing and presentation machinery is one well-characterized resistance mechanism. Constitutively expressed membrane-anchored interleukin-12 (caIL-12) has demonstrated enhanced antitumor activity and low systemic exposure in multiple preclinical adoptive T-cell treatment models with homogeneous tumor antigen expression. In this study, we assess the therapeutic impact of caIL-12 on target antigen-negative variants in syngeneic mouse models. METHODS Target antigen-positive tumors were generated by transducing B16F10 melanoma cells (B16) or Lewis Lung Carcinoma cells (LLC) with a construct expressing the OVA antigen, SIINFEKL, tagged to ubiquitin (B16-U-OVA, LLC-U-OVA), while B16 or LLC tumors served as antigen-negative variants. C57BL/6J mice were subcutaneously injected with heterogeneous tumors composed of 80% B16-U-OVA and 20% B16. Bilateral tumors were established by injecting the left flank with B16-U-OVA or LLC-U-OVA tumors and the right flank injected with B16 or LLC tumors. The tumor-bearing mice then underwent 5.5 Gy total body irradiation, followed by adoptive transfer of OT-I TCR-T cells engineered with or without caIL-12. RESULTS TCR-T cells (OT-I) delivered caIL-12 to the B16-U-OVA tumor sites and induced robust tumor control and survival benefits in mice bearing a heterogeneous tumor with OVA-negative variants. caIL-12 exerted its effect on OVA-negative B16 variants primarily by priming and activating endogenous antitumor CD8 T cells via antigen spreading. In addition, antigen spreading induced by OT-I-caIL-12 resulted in controlling OVA-negative tumors implanted at distant sites. This therapeutic effect required antigen-specific TCR-T cells and caIL-12 to colocalize at the tumor site, along with endogenous CD8 T cells capable of recognizing shared tumor antigens. CONCLUSION Expression of caIL-12 by tumor-targeting T cells demonstrated therapeutic effect against target-antigen-negative tumor variants, primarily through the induction of antigen spreading. These findings highlight the potential of caIL-12 to address challenges of antigen escape and tumor heterogeneity that may limit the efficacy of T-cell therapy against solid tumors.
Collapse
MESH Headings
- Animals
- Mice
- Interleukin-12/metabolism
- Antigens, Neoplasm/immunology
- Immunotherapy, Adoptive/methods
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Mice, Inbred C57BL
- Cell Line, Tumor
- Humans
- Adoptive Transfer/methods
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/therapy
Collapse
Affiliation(s)
- Ling Zhang
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Scott M Norberg
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Farrah Karimipour
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - John S Davies
- Department of Safety Assessment, Genentech Inc, South San Francisco, California, USA
| | - Alex Kuznetsov
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Wiem Lassoued
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniel Burnett
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Philip Homan
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Margaret Cam
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrew Sinkoe
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ping Xue
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - James L Gulley
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Christian S Hinrichs
- Duncan and Nancy MacMillan Cancer Immunology and Metabolism Center of Excellence, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| |
Collapse
|
40
|
Gao Q, Shen K, Xiao M. TET2 mutation in acute myeloid leukemia: biology, clinical significance, and therapeutic insights. Clin Epigenetics 2024; 16:155. [PMID: 39521964 PMCID: PMC11550532 DOI: 10.1186/s13148-024-01771-2] [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: 10/05/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
TET2 is a critical gene that regulates DNA methylation, encoding a dioxygenase protein that plays a vital role in the regulation of genomic methylation and other epigenetic modifications, as well as in hematopoiesis. Mutations in TET2 are present in 7%-28% of adult acute myeloid leukemia (AML) patients. Despite this, the precise mechanisms by which TET2 mutations contribute to malignant transformation and how these insights can be leveraged to enhance treatment strategies for AML patients with TET2 mutations remain unclear. In this review, we provide an overview of the functions of TET2, the effects of its mutations, its role in clonal hematopoiesis, and the possible mechanisms of leukemogenesis. Additionally, we explore the mutational landscape across different AML subtypes and present recent promising preclinical research findings.
Collapse
Affiliation(s)
- Qiang Gao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China
| | - Kefeng Shen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China.
| | - Min Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China.
| |
Collapse
|
41
|
Pal S, Firdous SM. Unraveling the role of heavy metals xenobiotics in cancer: a critical review. Discov Oncol 2024; 15:615. [PMID: 39495398 PMCID: PMC11535144 DOI: 10.1007/s12672-024-01417-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 10/01/2024] [Indexed: 11/05/2024] Open
Abstract
Cancer is a multifaceted disease characterized by the gradual accumulation of genetic and epigenetic alterations within cells, leading to uncontrolled cell growth and invasive behavior. The intricate interplay between environmental factors, such as exposure to carcinogens, and the molecular cascades governing cell growth, differentiation, and survival contributes to cancer's development and progression. This review offers a comprehensive overview of key molecular targets and their roles in cancer development. Peroxisome proliferator-activated receptors are implicated in various cancers due to their role in regulating lipid metabolism, inflammation, and cell proliferation. Nuclear factor erythroid 2-related factor 2 protects cells from oxidative damage but can also promote tumor cell survival. Cytochrome P450 1B1 metabolizes exogenous and endogenous substances, and its increased expression is observed in several cancers. The constitutive androstane receptor regulates gene expression, and its dysregulation can lead to liver cancer. Transforming growth factor-beta 2 is involved in the development and progression of various cancers by dysregulating cell proliferation, differentiation, and migration. Chelation treatment has been investigated for removing heavy metals, while genetically altered immune cells show promise in treating specific cancers. Metal-organic frameworks and fibronectin targeting represent new directions in cancer treatment. While some heavy metals, such as arsenic, chromium, nickel, and cadmium, are known to have carcinogenic properties, others, like zinc, Copper, gold, bismuth, and silver, have many uses that highlight their potential as effective cancer control tactics. There are a variety of heavy metal-based technologies that show potential for improving cancer treatment methods, including targeted drug delivery, improved radiation, and diagnostic tools.
Collapse
Affiliation(s)
- Sourav Pal
- Department of Pharmacology, Seacom Pharmacy College, Jaladhulagori, Sankrail, Howrah, West Bengal, 711302, India
| | - Sayed Mohammed Firdous
- Department of Pharmacology, Calcutta Institute of Pharmaceutical Technology & AHS, Uluberia, Howrah, West Bengal, 711316, India.
| |
Collapse
|
42
|
Katoh M, Katoh M. Claudin 1, 4, 6 and 18 isoform 2 as targets for the treatment of cancer (Review). Int J Mol Med 2024; 54:100. [PMID: 39301632 DOI: 10.3892/ijmm.2024.5424] [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: 07/24/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024] Open
Abstract
The 24 claudin (CLDN) genes in the human genome encode 26 representative CLDN family proteins. CLDNs are tetraspan‑transmembrane proteins at tight junctions. Because several CLDN isoforms, such as CLDN6 and CLDN18.2, are specifically upregulated in human cancer, CLDN‑targeting monoclonal antibodies (mAbs), antibody‑drug conjugates (ADCs), bispecific antibodies (bsAbs) and chimeric antigen receptor (CAR) T cells have been developed. In the present review, CLDN1‑, 4‑, 6‑ and 18.2‑targeting investigational drugs in clinical trials are discussed. CLDN18.2‑directed therapy for patients with gastric and other types of cancer is the most advanced area in this field. The mouse/human chimeric anti‑CLDN18.2 mAb zolbetuximab has a single‑agent objective response rate (ORR) of 9%, and increases progression‑free and overall survival in combination with chemotherapy. The human/humanized anti‑CLDN18.2 mAb osemitamab, and ADCs AZD0901, IBI343 and LM‑302, with single‑agent ORRs of 28‑60%, have been tested in phase III clinical trials. In addition, bsAbs, CAR T cells and their derivatives targeting CLDN4, 6 or 18.2 are in phase I and/or II clinical trials. AZD0901, IBI343, zolbetuximab and the anti‑CLDN1 mAb ALE.C04 have been granted fast track designation or priority review designation by the US Food and Drug Administration.
Collapse
Affiliation(s)
- Masuko Katoh
- Department of Global Network, M & M Precision Medicine, Tokyo 113‑0033, Japan
| | - Masaru Katoh
- Department of Global Network, M & M Precision Medicine, Tokyo 113‑0033, Japan
| |
Collapse
|
43
|
Sureda A, Lugtenburg PJ, Kersten MJ, Subklewe M, Spanjaart A, Shah NN, Kerbauy LN, Roddie C, Pennings ERA, Mahuad C, Poon M, Hendricks CL, Kamdar M, Jacobson C. Cellular therapy in lymphoma. Hematol Oncol 2024; 42:e3200. [PMID: 37382086 DOI: 10.1002/hon.3200] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/07/2023] [Accepted: 06/17/2023] [Indexed: 06/30/2023]
Abstract
CD19-directed chimeric antigen receptor (CAR) T-cell therapy has had a dramatic impact on the natural history and survival of patients with high-risk B-cell non-Hodgkin lymphoma. Accompanying this success has been the development of new fields of medicine and investigation into toxicity risks and mitigation therapies, mechanisms of resistance and the development of novel and next generation products and strategies in order to address relapse, and issues related to global access and health care economics. This article is a survey of each of these areas as it pertains to the rapidly evolving field of CAR T-cell therapy, written by an International community of lymphoma experts, who also happen to be women.
Collapse
Affiliation(s)
- Anna Sureda
- Clinical Hematology Department, Institut Catala d'Oncologia - Hospitalet, Institut d'Investigatcions Biomediques de Bellvitge (IDIBELL), Universitat de Barcelona, Barcelona, Spain
| | | | - Marie José Kersten
- Department of Hematology, Amsterdam Universitair Medische Centra, Amsterdam, The Netherlands
| | - Marion Subklewe
- Department of Medicine III, University Hospital, LMU, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Anne Spanjaart
- Department of Hematology, Amsterdam Universitair Medische Centra, Amsterdam, The Netherlands
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Lucila N Kerbauy
- Departments of Stem Cell Transplantation and Hemotherapy/Cellular Therapy, Hospital Israelita Albert Einstein, Sao Paolo, Brazil
| | - Clarie Roddie
- Research Department of Haematology, Cancer Institute, University College London, London, UK
| | - Elise R A Pennings
- Department of Hematology, Amsterdam Universitair Medische Centra, Amsterdam, The Netherlands
| | - Carolina Mahuad
- Hematology Service, Department of Internal Medicine, Deutsches Hospital, Buenos Aires, Argentina
| | - Michelle Poon
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Candice L Hendricks
- Institute for Cellular and Molecular Medicine, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | | | - Caron Jacobson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
44
|
Banerjee S, Booth CM, Bruera E, Büchler MW, Drilon A, Fry TJ, Ghobrial IM, Gianni L, Jain RK, Kroemer G, Llovet JM, Long GV, Pantel K, Pritchard-Jones K, Scher HI, Tabernero J, Weichselbaum RR, Weller M, Wu YL. Two decades of advances in clinical oncology - lessons learned and future directions. Nat Rev Clin Oncol 2024; 21:771-780. [PMID: 39354161 DOI: 10.1038/s41571-024-00945-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2024] [Indexed: 10/03/2024]
Affiliation(s)
- Susana Banerjee
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK.
- The Institute of Cancer Research, London, UK.
| | | | - Eduardo Bruera
- Department of Palliative, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer, Unit 1414, Houston, TX, USA.
| | - Markus W Büchler
- Botton-Champalimaud Pancreatic Cancer, Champalimaud Foundation, Lisbon, Portugal.
| | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA.
| | - Terry J Fry
- Department of Paediatrics and Immunology, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Aurora, CO, USA.
| | - Irene M Ghobrial
- Center for Prevention of Progression of Blood Cancers, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medical Oncology, Harvard Medical School, Boston, MA, USA.
| | | | - Rakesh K Jain
- Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Josep M Llovet
- Mount Sinai Liver Cancer Program, Divisions of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain.
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia.
| | - Klaus Pantel
- Institute of Tumour Biology, University Cancer Center Hamburg, University Medical Center Hamburg Eppendorf, Hamburg, Germany.
| | - Kathy Pritchard-Jones
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK.
| | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Josep Tabernero
- Medical Oncology Department, Vall d'Hebron University Hospital (HUVH), Barcelona, Spain.
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, USA.
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland.
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
45
|
Cai W, Tanaka K, Mi X, Rajasekhar VK, Khan JF, Yoo S, de Stanchina E, Rahman J, Mathew S, Abrahimi P, Souness S, Purdon TJ, McDowell JR, Meyerberg J, Fujino T, Healey JH, Abdel-Wahab O, Scheinberg DA, Brentjens RJ, Daniyan AF. Augmenting CAR T-cell Functions with LIGHT. Cancer Immunol Res 2024; 12:1361-1379. [PMID: 38959337 PMCID: PMC11444887 DOI: 10.1158/2326-6066.cir-24-0246] [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: 03/13/2024] [Revised: 04/23/2024] [Accepted: 07/02/2024] [Indexed: 07/05/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has resulted in remarkable clinical success in the treatment of B-cell malignancies. However, its clinical efficacy in solid tumors is limited, primarily by target antigen heterogeneity. To overcome antigen heterogeneity, we developed CAR T cells that overexpress LIGHT, a ligand of both lymphotoxin-β receptor on cancer cells and herpes virus entry mediator on immune cells. LIGHT-expressing CAR T cells displayed both antigen-directed cytotoxicity mediated by the CAR and antigen-independent killing mediated through the interaction of LIGHT with lymphotoxin-β receptor on cancer cells. Moreover, CAR T cells expressing LIGHT had immunostimulatory properties that improved the cells' proliferation and cytolytic profile. These data indicate that LIGHT-expressing CAR T cells may provide a way to eliminate antigen-negative tumor cells to prevent antigen-negative disease relapse.
Collapse
Affiliation(s)
- Winson Cai
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA 10021
| | - Kento Tanaka
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - Xiaoli Mi
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | | | - Jonathan F. Khan
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA 10021
| | - Sarah Yoo
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | | | - Jahan Rahman
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - Serena Mathew
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - Parwiz Abrahimi
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA 10021
| | - Sydney Souness
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | | | | | - Jeremy Meyerberg
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - Takeshi Fujino
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - John H. Healey
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - Omar Abdel-Wahab
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA 10021
| | - David A. Scheinberg
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA 10021
| | | | | |
Collapse
|
46
|
Toner K, McCann CD, Bollard CM. Applications of cell therapy in the treatment of virus-associated cancers. Nat Rev Clin Oncol 2024; 21:709-724. [PMID: 39160243 DOI: 10.1038/s41571-024-00930-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2024] [Indexed: 08/21/2024]
Abstract
A diverse range of viruses have well-established roles as the primary driver of oncogenesis in various haematological malignancies and solid tumours. Indeed, estimates suggest that approximately 1.5 million patients annually are diagnosed with virus-related cancers. The predominant human oncoviruses include Epstein-Barr virus (EBV), Kaposi sarcoma-associated herpesvirus (KSHV), hepatitis B and C viruses (HBV and HCV), human papillomavirus (HPV), human T-lymphotropic virus type 1 (HTLV1), and Merkel cell polyomavirus (MCPyV). In addition, although not inherently oncogenic, human immunodeficiency virus (HIV) is associated with immunosuppression that contributes to the development of AIDS-defining cancers (specifically, Kaposi sarcoma, aggressive B cell non-Hodgkin lymphoma and cervical cancer). Given that an adaptive T cell-mediated immune response is crucial for the control of viral infections, increasing research is being focused on evaluating virus-specific T cell therapies for the treatment of virus-associated cancers. In this Review, we briefly outline the roles of viruses in the pathogenesis of these malignancies before describing progress to date in the field of virus-specific T cell therapy and evaluating the potential utility of these therapies to treat or possibly even prevent virus-related malignancies.
Collapse
Affiliation(s)
- Keri Toner
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
- Department of Paediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Chase D McCann
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
- Department of Paediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA.
- Department of Paediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
47
|
Srinivasan S, Armitage J, Nilsson J, Waithman J. Transcriptional rewiring in CD8 + T cells: implications for CAR-T cell therapy against solid tumours. Front Immunol 2024; 15:1412731. [PMID: 39399500 PMCID: PMC11466849 DOI: 10.3389/fimmu.2024.1412731] [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: 04/05/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
T cells engineered to express chimeric-antigen receptors (CAR-T cells) can effectively control relapsed and refractory haematological malignancies in the clinic. However, the successes of CAR-T cell therapy have not been recapitulated in solid tumours due to a range of barriers such as immunosuppression, poor infiltration, and tumour heterogeneity. Numerous strategies are being developed to overcome these barriers, which include improving culture conditions and manufacturing protocols, implementing novel CAR designs, and novel approaches to engineering the T cell phenotype. In this review, we describe the various emerging strategies to improve CAR T cell therapy for solid tumours. We specifically focus on new strategies to modulate cell function and fate that have precipitated from the growing knowledge of transcriptional circuits driving T cell differentiation, with the ultimate goal of driving more productive anti-tumour T cell immunity. Evidence shows that enrichment of particular phenotypic subsets of T cells in the initial cell product correlates to improved therapeutic responses and clinical outcomes. Furthermore, T cell exhaustion and poor persistence are major factors limiting therapeutic efficacy. The latest preclinical work shows that targeting specific master regulators and transcription factors can overcome these key barriers, resulting in superior T cell therapeutic products. This can be achieved by targeting key transcriptional circuits promoting memory-like phenotypes or sustaining key effector functions within the hostile tumour microenvironment. Additional discussion points include emerging considerations for the field such as (i) targeting permutations of transcription factors, (ii) transient expression systems, (iii) tissue specificity, and (iv) expanding this strategy beyond CAR-T cell therapy and cancer.
Collapse
Affiliation(s)
- Shamini Srinivasan
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Jesse Armitage
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
- Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Jonas Nilsson
- Melanoma Discovery Lab, Harry Perkins Institute of Medical Research, Centre of Medical Research, The University of Western Australia, Perth, WA, Australia
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Jason Waithman
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
- Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| |
Collapse
|
48
|
Yoshimoto S, Kudo A, Rotolo A, Foos K, Olenick L, Takagi S, Mason NJ. Validation of a PD-1/CD28 chimeric switch receptor to augment CAR-T function in dogs with spontaneous B cell lymphoma. iScience 2024; 27:110863. [PMID: 39314237 PMCID: PMC11418608 DOI: 10.1016/j.isci.2024.110863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/07/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has achieved unprecedented clinical outcomes in patients with relapsed/refractory B cell leukemias; however, response rates in patients with large B cell lymphoma (LBCL) are less impressive. Expression of PD-1 on activated T cells and PD-L1 on malignant, stromal, and immune cells within the tumor microenvironment (TME) contribute to CAR-T exhaustion, hypofunction, and treatment failures. Here, a comparative approach is taken to develop a chimeric switch receptor (CSR) with potential to augment CAR-T persistence, function, and clinical efficacy in immune competent, pet dogs with spontaneous B cell lymphoma (BCL). We show that similar to human CAR-T cells, expression of a PD-1/CD28 CSR in canine CAR-T cells results in enhanced function against PD-L1+ targets and preserves central memory phenotype. We also demonstrate that these effects depend upon active CSR signaling. This work paves the way for in vivo studies in canine BCL patients to inform human trial design.
Collapse
Affiliation(s)
- Sho Yoshimoto
- Laboratory of Small Animal Surgery, Department of Veterinary Medicine, School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa, Japan
| | - Ayano Kudo
- Laboratory of Small Animal Surgery, Department of Veterinary Medicine, School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa, Japan
| | - Antonia Rotolo
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kay Foos
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lauren Olenick
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Satoshi Takagi
- Laboratory of Small Animal Surgery, Department of Veterinary Medicine, School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa, Japan
| | - Nicola J. Mason
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
49
|
Głowacki P, Tręda C, Rieske P. Regulation of CAR transgene expression to design semiautonomous CAR-T. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200833. [PMID: 39184876 PMCID: PMC11344471 DOI: 10.1016/j.omton.2024.200833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Effective transgene expression is critical for genetically engineered cell therapy. Therefore, one of CAR-T cell therapy's critical areas of interest, both in registered products and next-generation approaches is the expression of transgenes. It turns out that various constitutive promoters used in clinical products may influence CAR-T cell antitumor effectiveness and impact the manufacturing process. Furthermore, next-generation CAR-T starts to install remotely controlled inducible promoters or even autonomous expression systems, opening new ways of priming, boosting, and increasing the safety of CAR-T. In this article, a wide range of constitutive and inducible promoters has been grouped and structured, making it possible to compare their pros and cons as well as clinical usage. Finally, logic gates based on Synthetic Notch have been elaborated, demonstrating the coupling of desired external signals with genetically engineered cellular responses.
Collapse
Affiliation(s)
- Paweł Głowacki
- Department of Tumor Biology, Chair of Medical Biology, Medical University of Lodz, Zeligowskiego 7/9 Street, 90-752 Lodz, Poland
| | - Cezary Tręda
- Department of Tumor Biology, Chair of Medical Biology, Medical University of Lodz, Zeligowskiego 7/9 Street, 90-752 Lodz, Poland
- Department of Research and Development Personather Ltd, Inwestycyjna 7, 95-050 Konstantynow Lodzki, Poland
| | - Piotr Rieske
- Department of Tumor Biology, Chair of Medical Biology, Medical University of Lodz, Zeligowskiego 7/9 Street, 90-752 Lodz, Poland
- Department of Research and Development Personather Ltd, Inwestycyjna 7, 95-050 Konstantynow Lodzki, Poland
| |
Collapse
|
50
|
Stewart CM, Siegler EL, Sakemura RL, Cox MJ, Huynh T, Kimball B, Mai L, Can I, Manriquez Roman C, Yun K, Sirpilla O, Girsch JH, Ogbodo E, Mohammed Ismail W, Gaspar-Maia A, Budka J, Kim J, Scholler N, Mattie M, Filosto S, Kenderian SS. IL-4 drives exhaustion of CD8 + CART cells. Nat Commun 2024; 15:7921. [PMID: 39266501 PMCID: PMC11393358 DOI: 10.1038/s41467-024-51978-3] [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: 07/14/2023] [Accepted: 08/22/2024] [Indexed: 09/14/2024] Open
Abstract
Durable response to chimeric antigen receptor T (CART) cell therapy remains limited in part due to CART cell exhaustion. Here, we investigate the regulation of CART cell exhaustion with three independent approaches including: a genome-wide CRISPR knockout screen using an in vitro model for exhaustion, RNA and ATAC sequencing on baseline and exhausted CART cells, and RNA and ATAC sequencing on pre-infusion CART cell products from responders and non-responders in the ZUMA-1 clinical trial. Each of these approaches identify interleukin (IL)-4 as a regulator of CART cell dysfunction. Further, IL-4-treated CD8+ CART cells develop signs of exhaustion independently of the presence of CD4+ CART cells. Conversely, IL-4 pathway editing or the combination of CART cells with an IL-4 monoclonal antibody improves antitumor efficacy and reduces signs of CART cell exhaustion in mantle cell lymphoma xenograft mouse models. Therefore, we identify both a role for IL-4 in inducing CART exhaustion and translatable approaches to improve CART cell therapy.
Collapse
Affiliation(s)
- Carli M Stewart
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Elizabeth L Siegler
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - R Leo Sakemura
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | - Truc Huynh
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Brooke Kimball
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Long Mai
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Ismail Can
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | - Kun Yun
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Olivia Sirpilla
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - James H Girsch
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ekene Ogbodo
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Justin Budka
- Department of Oncology, Gilead Sciences Inc., Foster City, CA, USA
| | - Jenny Kim
- Department of Oncology, Gilead Sciences Inc., Foster City, CA, USA
| | | | - Mike Mattie
- Department of Oncology, Gilead Sciences Inc., Foster City, CA, USA
| | - Simone Filosto
- Department of Oncology, Gilead Sciences Inc., Foster City, CA, USA
| | - Saad S Kenderian
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA.
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|