1
|
Wang Q, He J, Lei T, Li X, Yue S, Liu C, Hu Q. New insights into cancer immune checkpoints landscape from single-cell RNA sequencing. Biochim Biophys Acta Rev Cancer 2025; 1880:189298. [PMID: 40088992 DOI: 10.1016/j.bbcan.2025.189298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/07/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
Immune checkpoint blockade (ICB) therapy represents a pivotal advancement in tumor immunotherapy by restoring the cytotoxic lymphocytes' anti-tumor activity through the modulation of immune checkpoint functions. Nevertheless, many patients experience suboptimal therapeutic outcomes, likely due to the immunosuppressive tumor microenvironment, drug resistance, and other factors. Single-cell RNA sequencing has assisted to precisely investigate the immune infiltration patterns before and after ICB treatment, enabling a high-resolution depiction of previously unrecognized functional interaction among immune checkpoints. This review addresses the heterogeneity between tumor microenvironments that respond to or resist ICB therapy, highlighting critical factors underlying the variation in immunotherapy efficacy and elucidating treatment failure. Furthermore, a comprehensive examination is provided of how specific ICBs modulate immune and tumor cells to achieve anti-tumor effects and generate treatment resistance, alongside a summary of emerging immune checkpoints identified as promising targets for cancer immunotherapy through single-cell RNA sequencing applications.
Collapse
Affiliation(s)
- Qian Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiahui He
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tianyu Lei
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaohui Li
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China
| | - Shengqin Yue
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Chao Liu
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China.
| | - Qinyong Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Renmin Hospital of Wuhan Economic and Technological Development Zone (Hannan), Wuhan 430090, China.
| |
Collapse
|
2
|
Shen Z, Meng X, Rautela J, Chopin M, Huntington ND. Adjusting the scope of natural killer cells in cancer therapy. Cell Mol Immunol 2025:10.1038/s41423-025-01297-4. [PMID: 40410571 DOI: 10.1038/s41423-025-01297-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 05/06/2025] [Indexed: 05/25/2025] Open
Abstract
Natural killer (NK) cells have evolved to detect abnormalities in tissues arising from infection with pathogens, genomic damage, or transformation and respond rapidly to the production of potent proinflammatory and cytolytic mediators. While this acute proinflammatory response is highly efficient at orchestrating sterilizing immunity to pathogens in a matter of days, cellular transformation often avoids the innate detection mechanisms of NK cells. When cellular transformation results in malignancy, tumor cells and/or the tumor microenvironment can evolve additional mechanisms to circumvent NK cell responses, and cancer is now a dominant disease burden worldwide. Here, we review recent advances in our understanding of the combined relationship between malignancies and natural killer (NK) cells, learn from recent clinical efforts in therapeutically targeting natural killer (NK) cells in cancer and outline some emerging therapeutic concepts that aim to improve the innate immune response against cancer.
Collapse
Affiliation(s)
- Zihen Shen
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Xiangpeng Meng
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Jai Rautela
- oNKo-Innate Pty Ltd., Moonee Ponds, VIC, Australia
| | - Michael Chopin
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Nicholas D Huntington
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia.
- oNKo-Innate Pty Ltd., Moonee Ponds, VIC, Australia.
| |
Collapse
|
3
|
Yu H, Jin S, Zeng M, Yang Z, Wang X. TIGIT antibody with PVR competitive ability enhances cancer immunotherapy and capable of eliciting anti-tumour immune memory. Br J Cancer 2025:10.1038/s41416-025-03046-w. [PMID: 40394151 DOI: 10.1038/s41416-025-03046-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 04/01/2025] [Accepted: 04/25/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND T-cell immunoreceptor with immunoglobulin (Ig) and ITIM domains (TIGIT) is a checkpoint receptor thought to be involved in mediating T-cell exhaustion and dysfunction of natural killer (NK) cells in tumours and is emerging as novel promising targets in immunotherapy, however, the ligand binding and the efficacy of its antibody still need to be further explored. METHODS Four different TIGIT antibodies in characteristics of antigen binding, in vitro effects on activated T cells, Fc region functions and tumour inhibition in animal models were compared. The antibody as monotherapy and combined with anti-PD-L1 antibody, effects on PBMC in ex vivo coculture with autologous human CRC organoids as well as PK profile were evaluated. RESULTS Studies demonstrated that TIGIT antibody with PVR-competitive ability as monotherapy resulted in inhibition of tumour growth, sustained anti-tumour immune memory in tumour re-challenge mice, enhanced anti-tumour therapy in combination with anti-PD-L1. Ex vivo coculture assay suggested that TIGIT antibody treatment activated immune cells and promoted infiltration and tumour killing ability of autologous PBMC in human CRC organoids. CONCLUSIONS Our study broadens the knowledge of TIGIT antibody in cancer immunotherapy and may benefit future development of next-generation checkpoint inhibitors with improved clinical outcomes.
Collapse
Affiliation(s)
- Huijuan Yu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China
| | - Shaowen Jin
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Min Zeng
- Guangdong Annpobio Co., Ltd, Guangzhou, China
| | | | - Xiaofei Wang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China.
- Guangdong Annpobio Co., Ltd, Guangzhou, China.
| |
Collapse
|
4
|
Liu Y, Du Z, Li L, Huang J, Liu S, Lu B, Duan Y, Cheng Y, Li T, Zhang J, Mo J, Yang Y, Wang W, Zou H, Liang T, Jiang M, Yang M, Chen Y, Ouyang C, Chen C. scRNA-seq reveals an immune microenvironment and JUN-mediated NK cell exhaustion in relapsed T-ALL. Cell Rep Med 2025; 6:102098. [PMID: 40306275 DOI: 10.1016/j.xcrm.2025.102098] [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: 08/30/2024] [Revised: 01/21/2025] [Accepted: 04/08/2025] [Indexed: 05/02/2025]
Abstract
T cell acute lymphoblastic leukemia (T-ALL) is a heterogeneous disease characterized by a high relapse rate. By single-cell transcriptome analysis, we characterize the bone marrow immune microenvironment in patients with T-ALL, identifying 13 major cell clusters. These patients exhibited abnormally expanded hematopoietic stem cells (HSCs) and granulocyte-monocyte progenitors (GMPs), immunosuppressive traits in CD4+ T, CD8+ T, and natural killer (NK) cells. Subdividing CD4+ T cells reveal two subsets transitioning between T helper (Th)1/Th2, Annexin-A1 (ANXA1)-GATA3-CD4+ T, and ANXA1+GATA3+CD4+ T. Additionally, NK cells demonstrate exhaustion in the tumor microenvironment of patients with relapsed T-ALL, with JUN identified as a critical factor. Additionally, JUN is also highly expressed in T-ALL and is crucial for maintaining its proliferation. The JUN inhibitor exhibited successful lethality toward leukemia cells and ameliorated NK cell exhaustion in relapsed T-ALL cell line, as well as in cell-derived tumor xenograft (CDX), patient-derived tumor xenograft (PDX), and NOTCH1-mutant mouse models. In summary, our findings enhance the understanding of T-ALL relapse mechanisms and support the development of innovative immunotherapies for patients with relapsed T-ALL.
Collapse
Affiliation(s)
- Yong Liu
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Zefan Du
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Lindi Li
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Junbin Huang
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Su Liu
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Bo Lu
- Department of Haematology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Yifei Duan
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Yucai Cheng
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Tianwen Li
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Jing Zhang
- Department of Thyroid and Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Jiani Mo
- Department of Hematology, Affiliated Hospital of Guangdong Medical University (GDMU), Zhanjiang 524001, Guangdong, China
| | - Yalin Yang
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Wengqing Wang
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Hailin Zou
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Tianqi Liang
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Meng Jiang
- School of Medicine, Sun Yat-sen University, Shenzhen 518107, Guangdong, China
| | - Mo Yang
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China; Department of Thyroid and Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China.
| | - Yun Chen
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China.
| | - Cheng Ouyang
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China.
| | - Chun Chen
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China.
| |
Collapse
|
5
|
Razmara AM, Lammers M, Judge SJ, Murphy WJ, Gaskill CE, Culp WT, Gingrich AA, Morris ZS, Rebhun RB, Brown CT, Vail DM, Kent MS, Canter RJ. Single cell atlas of canine natural killer cells identifies distinct circulating and tissue resident gene profiles. Front Immunol 2025; 16:1571085. [PMID: 40443661 PMCID: PMC12119461 DOI: 10.3389/fimmu.2025.1571085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/22/2025] [Indexed: 06/02/2025] Open
Abstract
Introduction Natural killer (NK) cells in mice and humans are key effectors of the innate immune system with complex immunoregulatory functions, and diverse subsets have been identified with distinct characteristics and roles. Companion dogs with spontaneous cancer have been validated as models of human disease, including cancer immunology and immunotherapy, and greater understanding of NK cell heterogeneity in dogs can inform NK biology across species and optimize NK immunotherapy for both dogs and people. Methods Here, we assessed canine NK cell populations by single-cell RNA sequencing (scRNAseq) across blood, lung, liver, spleen, and placenta with comparison to human NK cells from blood and the same tissues to better characterize the differential gene expression of canine and human NK cells regarding ontogeny, heterogeneity, patterns of activation, inhibition, and tissue residence. Results Overall, we observed tissue-specific NK cell signatures consistent with immature NK cells in the placenta, mature and activated NK cells in the lung, and NK cells with a mixed activated and inhibited signature in the liver with significant cross-species homology. Discussion Together, our results point to heterogeneous canine NK populations highly comparable to human NK cells, and we provide a comprehensive atlas of canine NK cells across organs which will inform future cross-species NK studies and further substantiate the spontaneous canine model to optimize NK immunotherapy across species.
Collapse
Affiliation(s)
- Aryana M. Razmara
- Division of Surgical Oncology, Department of Surgery, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Marshall Lammers
- Division of Surgical Oncology, Department of Surgery, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Sean J. Judge
- Division of Surgical Oncology, Department of Surgery, University of California Davis School of Medicine, Sacramento, CA, United States
| | - William J. Murphy
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Cameron E. Gaskill
- Division of Surgical Oncology, Department of Surgery, University of California Davis School of Medicine, Sacramento, CA, United States
| | - William T.N. Culp
- Department of Surgical and Radiological Sciences, University of California Davis School of Veterinary Medicine, Davis, CA, United States
| | - Alicia A. Gingrich
- Division of Surgical Oncology, Department of Surgery, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Robert B. Rebhun
- Department of Surgical and Radiological Sciences, University of California Davis School of Veterinary Medicine, Davis, CA, United States
| | - C. Titus Brown
- Department Population Health and Reproduction, University of California Davis School of Veterinary Medicine, Davis, CA, United States
| | - David M. Vail
- Department of Medical Sciences, University of Wisconsin School of Veterinary Medicine, Madison, WI, United States
| | - Michael S. Kent
- Department of Surgical and Radiological Sciences, University of California Davis School of Veterinary Medicine, Davis, CA, United States
| | - Robert J. Canter
- Division of Surgical Oncology, Department of Surgery, University of California Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
6
|
Politis A, Stavrinou L, Kalyvas A, Boviatsis E, Piperi C. Glioblastoma: molecular features, emerging molecular targets and novel therapeutic strategies. Crit Rev Oncol Hematol 2025; 212:104764. [PMID: 40368035 DOI: 10.1016/j.critrevonc.2025.104764] [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: 03/04/2025] [Revised: 05/01/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025] Open
Abstract
Glioblastomas (GBMs) constitute the most common malignant tumors of the Central Nervous System (CNS) with a complex molecular, genetic and histological profile and extensive heterogenicity. GBMs are notoriously difficult to treat, with morbidity and mortality rate that remain high and practically unchanged, despite the aggressive and multimodal treatment strategies. Keeping up with current research and emerging scientific data is of primary importance for the detection of new molecular targets, enabling the design of novel therapeutic strategies. Herein, we discuss current data on the cellular and molecular features that contribute to GBM pathophysiological mechanisms in an effort to reveal emerging molecular targets with therapeutic potential as well as effective immunotherapeutic approaches, including chimeric antigen receptor (CAR) T-cell therapy and adaptive immune modulation with immune checkpoint inhibitors. Enhanced drug delivery strategies such as ultrasound-assisted technologies to overcome drug resistance are also discussed, aiming to provide an overall translational perspective that bridges molecular insights with practical therapeutic implications.
Collapse
Affiliation(s)
- Anastasios Politis
- Second Department of Neurosurgery, "Attikon" University Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece; Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Lampis Stavrinou
- Second Department of Neurosurgery, "Attikon" University Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Aristotelis Kalyvas
- Second Department of Neurosurgery, "Attikon" University Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece; Division of Neurosurgery, Department of Surgery, Temetry Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Efstathios Boviatsis
- Second Department of Neurosurgery, "Attikon" University Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| |
Collapse
|
7
|
Mao X, Yang R, Yan Y, Zeng Y, Bao M, Huang R, Dai Y, Zhang Q, Ye Y, Cheng J, Mo Z, Zhang H. Single-cell analysis reveals potential therapeutic markers of peripheral blood mononuclear cells from bladder cancer patients. Braz J Med Biol Res 2025; 58:e14002. [PMID: 40367012 PMCID: PMC12068768 DOI: 10.1590/1414-431x2025e14002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 03/20/2025] [Indexed: 05/16/2025] Open
Abstract
Bladder cancer is the most prevalent malignancy of the urinary tract, with significant advancements in treatment achieved over recent decades. Nonetheless, the immunological mechanisms underlying bladder cancer progression remain elusive, and only a limited number of patients derive benefit from current immune checkpoint inhibitors. Here, we conducted a single-cell RNA sequencing analysis of 44,022 cells from peripheral blood mononuclear cell samples of bladder cancer patients and a healthy donor. Our findings indicated that the proportions of T cells and neutrophils are higher in bladder cancer patients than in the healthy donor. LAG3, HAVCR2, and CTLA4 had elevated expression levels in CD8-T2-GZMK cell clusters from patients. CD8-T7-STMN1 cells highly expressed ITGAE, CD38, and STMN1. Furthermore, NK3-CMC1, more prevalent in patients, showed a high expression of TIGIT. Additionally, Bcell2-TCL1A and Bcell3-MS4A1 were characterized by the high expression of inhibitory receptor marker genes. Gene set variation analysis suggested that Mono4-THBS1 may play a role in promoting tumor hypoxia and angiogenesis. Neu-FCGR3B exhibited high levels of IL4R and CD274 expression. Our study indicated that LAG-3 and TIM-3 may serve as novel potential immune checkpoint inhibitors in bladder cancer treatment. The phenotypes of NK3-CMC1, Bcell2-TCL1A, and Bcell3-MS4A1 might be altered by tumor progression. Mono4-THBS1 could potentially be a source of tumor-enriched monocyte-like cells. Neu-FCGR3B may play a detrimental role in the anti-tumor response and could emerge as a predictive marker for bladder cancer. Overall, these high-resolution transcriptomic data offer invaluable insights for identifying new therapeutic targets and biomarkers in bladder cancer immunotherapy.
Collapse
Affiliation(s)
- Xingning Mao
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Rirong Yang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Yunkun Yan
- Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Yanyu Zeng
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Mengying Bao
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Rong Huang
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Yan Dai
- Emergency Department, the Second Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Qingyun Zhang
- Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Yu Ye
- Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Emergency Department, the Second Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Jiwen Cheng
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Zengnan Mo
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Haiying Zhang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
8
|
Parra-Tello B, García-Gómez M, Rekus-Polanska E, Malgue F, Charlín S, Hernández-Oliveras A, Reyes-Alvarez J, Rosemblatt M, Lundqvist A, Lladser A, Bono MR, Sauma D. CD73 promotes the maturation of murine NK cells and their survival in the tumor microenvironment. J Leukoc Biol 2025; 117:qiaf011. [PMID: 39901844 DOI: 10.1093/jleuko/qiaf011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/19/2024] [Accepted: 01/31/2025] [Indexed: 02/05/2025] Open
Abstract
Natural Killer (NK) cells are crucial in recognizing and eliminating tumor cells, making them pivotal in antitumor responses. Adenosine, the product of ATP hydrolysis mediated by CD39 and CD73 ectonucleotidases, has been reported to reduce the proliferation and maturation of NK cells. In this study, we investigate the expression of CD73 in NK cells and its impact on maturation, phenotype, survival, and function. Our findings reveal that while splenic NK cells express minimal levels of CD73, its expression is induced upon activation and in the tumor microenvironment upon adoptive transfer to tumor-bearing mice. Notably, within the tumor microenvironment, CD73 expression in NK cells correlates with elevated levels of PD-L1 and CD226. Accordingly, analysis of human melanoma datasets uncovers a subset of immature tumor-infiltrating NK cells expressing CD73. To further understand the role of CD73 on NK cells, we used a CD73 knockout (KO) murine model and observed that CD73-deficient NK cells display a more immature phenotype and heightened proliferative activity than wild-type (WT) NK cells. Additionally, CD73-deficient NK cells exhibit elevated levels of P2X7R and reduced CD39 expression, suggesting an increased susceptibility to ATP-induced death. Following adoptive transfer to tumor-bearing mice, CD73KO NK cells are present at a lower frequency but demonstrate similar control over tumor growth compared with WT NK cells. In conclusion, our study demonstrates the upregulation of CD73 in NK cells infiltrating tumors and underscores its role as a checkpoint regulating the functional maturation of NK cells.
Collapse
Affiliation(s)
- Brian Parra-Tello
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, 7800003 Santiago, Chile
| | - Moira García-Gómez
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, 7800003 Santiago, Chile
| | - Eva Rekus-Polanska
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, 7800003 Santiago, Chile
| | - Felipe Malgue
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, 7800003 Santiago, Chile
| | - Sebastián Charlín
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, 7800003 Santiago, Chile
| | - Andrés Hernández-Oliveras
- Centro Ciencia & Vida, Avda. del Valle Norte N° 725, Huechuraba, 8580702 Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastian, Lota 2465, Providencia, 7510602 Santiago, Chile
| | - Javiera Reyes-Alvarez
- Centro Ciencia & Vida, Avda. del Valle Norte N° 725, Huechuraba, 8580702 Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastian, Lota 2465, Providencia, 7510602 Santiago, Chile
| | - Mario Rosemblatt
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, 7800003 Santiago, Chile
- Centro Ciencia & Vida, Avda. del Valle Norte N° 725, Huechuraba, 8580702 Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastian, Lota 2465, Providencia, 7510602 Santiago, Chile
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Alvaro Lladser
- Centro Ciencia & Vida, Avda. del Valle Norte N° 725, Huechuraba, 8580702 Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastian, Lota 2465, Providencia, 7510602 Santiago, Chile
| | - María Rosa Bono
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, 7800003 Santiago, Chile
- Centro Ciencia & Vida, Avda. del Valle Norte N° 725, Huechuraba, 8580702 Santiago, Chile
| | - Daniela Sauma
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, 7800003 Santiago, Chile
- Centro Ciencia & Vida, Avda. del Valle Norte N° 725, Huechuraba, 8580702 Santiago, Chile
| |
Collapse
|
9
|
Zhang ZF, Zhang Y, Chen YW, Cao GS, Zheng XD, Sun R, Peng H, Tian ZG, Sun HY. CD200R blockade enhances anti-tumor immunity by unleashing NK and CD8 + T cells in tumor. Acta Pharmacol Sin 2025:10.1038/s41401-025-01556-0. [PMID: 40329005 DOI: 10.1038/s41401-025-01556-0] [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: 11/09/2024] [Accepted: 03/30/2025] [Indexed: 05/08/2025]
Abstract
Immune checkpoint inhibitors have revolutionized cancer therapy, but a large proportion of patients do not respond well to current checkpoint immunotherapies. CD200R (also known as OX2R) is a transmembrane glycoprotein of the immunoglobulin superfamily that is mainly expressed on myeloid and lymphoid-derived immunocompetent cells such as myeloid cells, natural killer (NK), and CD8+ T cells. In this study, we investigated the therapeutic potential and cellular mechanisms of targeting CD200R in tumor immunotherapy. We established 4 subcutaneous tumor mouse models using MC38 (colon cancer), MCA205 (fibrosarcoma), LLC (lung cancer), and EO771 (mammary cancer) cell lines. We found that CD200R was highly expressed on tumor-infiltrating NK and CD8+ T cells with exhausted phenotypes in the four subcutaneous tumor mouse models. Either genetic ablation or antibody blockade of CD200R retarded tumor growth and prolonged the survival of tumor-bearing mice by preventing or reversing exhaustion of both NK cells and CD8+ T cells. The combined therapy of CD200R antibody with anti-PD-1/anti-PD-L1 synergistically inhibited tumor growth. By depletion of NK or/and CD8+ T cells, we demonstrated that both cell types contributed to the anti-tumor efficacy of CD200R blockade in tumor-bearing mice. Further, the blockade of human CD200R significantly enhanced human NK cell function and inhibited human tumor growth in PBMC-reconstituted xenograft mice. Our results demonstrate that CD200R is a potential immune checkpoint molecule that can suppress the tumoricidal activities of NK and CD8+ T cells, and could thus be exploited as a therapeutic target in the future.
Collapse
Affiliation(s)
- Zheng-Feng Zhang
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Yu Zhang
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Ya-Wen Chen
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Guo-Shuai Cao
- Hefei TG ImmunoPharma Corporation Limited, Hefei, 230027, China
| | - Xiao-Dong Zheng
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Rui Sun
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Hui Peng
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Zhi-Gang Tian
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Hao-Yu Sun
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
10
|
Wei H, Suo C, Gu X, Shen S, Lin K, Zhu C, Yan K, Bian Z, Chen L, Zhang T, Yan R, Yang Z, Yu Y, Li Z, Liu R, He J, He Q, Zhong X, Jia W, Wong CM, Dong Z, Cao J, Sun L, Zhang H, Gao P. AKR1D1 suppresses liver cancer progression by promoting bile acid metabolism-mediated NK cell cytotoxicity. Cell Metab 2025; 37:1103-1118.e7. [PMID: 40010348 DOI: 10.1016/j.cmet.2025.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 07/31/2024] [Accepted: 01/14/2025] [Indexed: 02/28/2025]
Abstract
Bile acid metabolism and antitumor immunity are both disrupted during liver cancer progression. However, the complex regulatory relationship between them remains largely unclear. Here, we find that loss of aldo-keto reductase 1D1 (AKR1D1) promotes the accumulation of isolithocholic acid (iso-LCA) through gut microbiome dysregulation, thereby impairing the cytotoxic function of natural killer (NK) cells and leading to the accelerated development of hepatocellular carcinoma (HCC). Mechanistically, AKR1D1 deficiency leads to an increased proportion of Bacteroidetes ovatus (B. ovatus), which breaks down chenodeoxycholic acid (CDCA) into iso-LCA. Moreover, accumulated iso-LCA impairs the antitumor activity of hepatic NK cells in a phosphorylated-CREB1 (p-CREB1)-dependent manner. The potassium-sparing diuretic spironolactone treatment significantly enhances the inhibitory effect of anti-PD1 antibody on HCC progression by targeting iso-LCA-mediated tumor immune escape. Taken together, our results uncover a previously unappreciated link between AKR1D1 and HCC and suggest that targeting iso-LCA produced by B. ovatus might be a promising strategy to activate NK cell cytotoxicity to treat HCC.
Collapse
Affiliation(s)
- Haoran Wei
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China; National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Caixia Suo
- Department of Colorectal Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Xuemei Gu
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Shengqi Shen
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Kashuai Lin
- Department of Colorectal Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Chuxu Zhu
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Kai Yan
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhenhua Bian
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Liang Chen
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Tong Zhang
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ronghui Yan
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhiyi Yang
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Yingxuan Yu
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhikun Li
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Rui Liu
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Junming He
- School of Medicine and Institute for Immunology, Beijing Key Laboratory for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Qiwei He
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiuying Zhong
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Weidong Jia
- Anhui Key Laboratory of Hepatopancreatobiliary Surgery, Department of General Surgery, Anhui Provincial Hospital, the First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Chun-Ming Wong
- State Key Laboratory of Liver Research, Department of Pathology, LKS Faculty of Medicine, the University of Hong Kong, Hong Kong, China
| | - Zhongjun Dong
- School of Medicine and Institute for Immunology, Beijing Key Laboratory for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Jie Cao
- Department of Colorectal Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Linchong Sun
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Huafeng Zhang
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Hepatopancreatobiliary Surgery, Department of General Surgery, Anhui Provincial Hospital, the First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Ping Gao
- Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan 610212, China.
| |
Collapse
|
11
|
Xie J, Liu XF, Zhou T, Liu L, Hou RQ, Yu XX, Fan ZY, Shang QN, Chang YJ, Zhao XS, Wang Y, Xu LP, Zhang XH, Huang XJ, Zhao XY. Overexpressing natural killer group 2 member A drives natural killer cell exhaustion in relapsed acute myeloid leukemia. Signal Transduct Target Ther 2025; 10:143. [PMID: 40320412 PMCID: PMC12050333 DOI: 10.1038/s41392-025-02228-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 05/08/2025] Open
Abstract
Acute myeloid leukemia (AML) relapse is associated with poor prognosis. While natural killer (NK) cell therapy can induce leukemia remission, infused NK cells are prone to exhaustion. Elucidating the molecular mechanisms driving NK cell exhaustion in AML patients could provide critical insights for developing novel strategies to optimize NK cell-based immunotherapies. In this study, we systematically investigated NK cell exhaustion in relapsed AML patients following allogeneic hematopoietic stem cell transplantation (allo-HSCT) through phenotypic assessments, functional assays, and RNA sequencing analyses. Compared to NK cells from complete remission patients and healthy controls, NK cells from relapsed AML patients exhibited an exhausted phenotype, marked by reduced maturity, elevated expression of the inhibitory receptor NKG2A, impaired cytotoxicity, and suppression of the PI3K-AKT pathway. Notably, NKG2A expression levels on NK cells correlated with disease progression. Blockade or genetic knockout of NKG2A effectively reversed NK cell exhaustion both in vitro and in an AML mouse model. Furthermore, activation of the PI3K-AKT pathway significantly enhanced cytotoxicity in exhausted NK cells. We found that excessive activation of the NKG2A/HLA-E axis was associated with PI3K-AKT pathway inhibition, and blocking the NKG2A/HLA-E interaction or knocking out NKG2A restored AKT phosphorylation in exhausted NK cells. In summary, AML cells drive NK cell exhaustion through overactivation of the NKG2A/HLA-E axis and suppression of the PI3K-AKT pathway. Targeting the NKG2A/HLA-E axis represents a promising therapeutic approach to restore PI3K-AKT signaling and reverse NK cell exhaustion.
Collapse
MESH Headings
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/pathology
- Humans
- Animals
- Mice
- NK Cell Lectin-Like Receptor Subfamily C/genetics
- NK Cell Lectin-Like Receptor Subfamily C/immunology
- Male
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/immunology
- Female
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/immunology
- Signal Transduction
- Hematopoietic Stem Cell Transplantation
- Middle Aged
Collapse
Grants
- This work was supported by the Noncommunicable Chronic Diseases-National Science and Technology Major Project (No. 2023ZD0501200), National Key Research and Development Program of China (grant 2022YFA1103300), National Natural Science Foundation of China (grants 82350105, 82070184 and 82270228), Science, Technology &Innovation Project of Xiongan New area (No. 2023XACX0004), Beijing Outstanding Young Scientists Project(JWZQ20240101001); Beijing Natural Science Foundation (Z240019), and Clinical Medicine Plus X-Young Scholars Project of Peking University (grant PKU2024LCXQ002).
- This work was supported by the Noncommunicable Chronic Diseases-National Science and Technology Major Project (No. 2023ZD0501200), National Key Research and Development Program of China (grant 2022YFA1103300), National Natural Science Foundation of China (grants 82350105, 82070184 and 82270228), Science, Technology & Innovation Project of Xiongan New area (No. 2023XACX0004), Beijing Outstanding Young Scientists Project(JWZQ20240101001); Beijing Natural Science Foundation (Z240019), and Clinical Medicine Plus X-Young Scholars Project of Peking University (grant PKU2024LCXQ002).
Collapse
Affiliation(s)
- Juan Xie
- Peking University People's Hospital, Institute of Hematology, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, National Clinical Research Centre for Hematologic Disease; No. 11 South Street of Xizhimen, Xicheng District, 100044, Beijing, China
| | - Xue-Fei Liu
- Peking University People's Hospital, Institute of Hematology, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, National Clinical Research Centre for Hematologic Disease; No. 11 South Street of Xizhimen, Xicheng District, 100044, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, 100871, Beijing, China
| | - Tong Zhou
- Peking University People's Hospital, Institute of Hematology, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, National Clinical Research Centre for Hematologic Disease; No. 11 South Street of Xizhimen, Xicheng District, 100044, Beijing, China
| | - Long Liu
- Peking University People's Hospital, Institute of Hematology, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, National Clinical Research Centre for Hematologic Disease; No. 11 South Street of Xizhimen, Xicheng District, 100044, Beijing, China
| | - Rui-Qin Hou
- Peking University People's Hospital department of blood transfusion, No. 11 South Street of Xizhimen, Xicheng District, 100044, Beijing, China
| | - Xing-Xing Yu
- Peking University People's Hospital, Institute of Hematology, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, National Clinical Research Centre for Hematologic Disease; No. 11 South Street of Xizhimen, Xicheng District, 100044, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, 100871, Beijing, China
| | - Ze-Ying Fan
- Peking University People's Hospital, Institute of Hematology, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, National Clinical Research Centre for Hematologic Disease; No. 11 South Street of Xizhimen, Xicheng District, 100044, Beijing, China
| | - Qian-Nan Shang
- Peking University People's Hospital, Institute of Hematology, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, National Clinical Research Centre for Hematologic Disease; No. 11 South Street of Xizhimen, Xicheng District, 100044, Beijing, China
| | - Ying-Jun Chang
- Peking University People's Hospital, Institute of Hematology, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, National Clinical Research Centre for Hematologic Disease; No. 11 South Street of Xizhimen, Xicheng District, 100044, Beijing, China
| | - Xiao-Su Zhao
- Peking University People's Hospital, Institute of Hematology, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, National Clinical Research Centre for Hematologic Disease; No. 11 South Street of Xizhimen, Xicheng District, 100044, Beijing, China
| | - Yu Wang
- Peking University People's Hospital, Institute of Hematology, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, National Clinical Research Centre for Hematologic Disease; No. 11 South Street of Xizhimen, Xicheng District, 100044, Beijing, China
| | - Lan-Ping Xu
- Peking University People's Hospital, Institute of Hematology, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, National Clinical Research Centre for Hematologic Disease; No. 11 South Street of Xizhimen, Xicheng District, 100044, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Institute of Hematology, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, National Clinical Research Centre for Hematologic Disease; No. 11 South Street of Xizhimen, Xicheng District, 100044, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Institute of Hematology, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, National Clinical Research Centre for Hematologic Disease; No. 11 South Street of Xizhimen, Xicheng District, 100044, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, 100871, Beijing, China.
| | - Xiang-Yu Zhao
- Peking University People's Hospital, Institute of Hematology, Beijing Key Laboratory of Cell and Gene Therapy for Hematologic Malignancies, National Clinical Research Centre for Hematologic Disease; No. 11 South Street of Xizhimen, Xicheng District, 100044, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, 100871, Beijing, China.
| |
Collapse
|
12
|
Tong J, Tan Y, Ouyang W, Chang H. Targeting immune checkpoints in hepatocellular carcinoma therapy: toward combination strategies with curative potential. Exp Hematol Oncol 2025; 14:65. [PMID: 40317077 PMCID: PMC12046748 DOI: 10.1186/s40164-025-00636-5] [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: 01/20/2025] [Accepted: 03/07/2025] [Indexed: 05/04/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a primary liver cancer characterized by poor immune cell infiltration and a strongly immunosuppressive microenvironment. Traditional treatments have often yielded unsatisfactory outcomes due to the insidious onset of the disease. Encouragingly, the introduction of immune checkpoint inhibitors (ICIs) has significantly transformed the approach to HCC treatment. Moreover, combining ICIs with other therapies or novel materials is considered the most promising opportunity in HCC, with some of these combinations already being evaluated in large-scale clinical trials. Unfortunately, most clinical trials fail to meet their endpoints, and the few successful ones also face challenges. This indicates that the potential of ICIs in HCC treatment remains underutilized, prompting a reevaluation of this promising therapy. Therefore, this article provides a review of the role of immune checkpoints in cancer treatment, the research progress of ICIs and their combination application in the treatment of HCC, aiming to open up avenues for the development of safer and more efficient immune checkpoint-related strategies for HCC treatment.
Collapse
Affiliation(s)
- Jing Tong
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
| | - Yongci Tan
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
| | - Wenwen Ouyang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
| | - Haocai Chang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China.
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
13
|
Mortezaee K. T-cell immunoglobulin and ITIM domain as a target in combo anti-PD-(L)1 cancer therapy. Int J Biol Macromol 2025; 310:143557. [PMID: 40294684 DOI: 10.1016/j.ijbiomac.2025.143557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/20/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
Immunoregulatory roles of T-cell immunoglobulin and ITIM domain (TIGIT) in solid tumors, and its interactions with other checkpoints is a focus of research in cancer immunotherapy. The increased activity of TIGIT/CD155 promotes dendritic cell (DC) tolerance and CD8+ T cell exclusion/energy/exhaustion. Increased TIGIT activity also hampers natural killer (NK) cell function and increases immunosuppressive activity of myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs), with the latter serving as a key cell type to pursue TIGIT regulatory effects in tumor immune ecosystem. Frequent co-expression of TIGIT with programmed death-1 (PD-1) on CD8+ T cells along with the increased TIGIT expression in Tregs after anti-PD-1 therapy, the stimulatory effect of TIGIT+ Tregs on T-cell immunoglobulin and mucin-domain containing-3 (TIM-3), and the inducible effect of anti-programmed death-ligand 1 (PD-L1) on CD155 are all rationalizing a possibility for application of anti-TIGIT as a desired combinatory with anti-PD-(L)1 drugs in cancer immunotherapy. TIGIT can also be a target for development of bispecific antibodies to simultaneously target activities within the TIGIT/CD155 and PD-1/PD-L1 axes or for dual targeting of two inhibitory receptors, such as TIGIT/anti-poliovirus receptor-related immunoglobulin domain-containing protein (PVRIG), with the latter also acting to hamper activation of other inhibitory receptors occurring secondary to the anti-TIGIT therapy.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran; Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
14
|
Chai J, Li L, Wu Q, Zhang S. CD96: immunoregulation and its role and prospect in immunotherapy of primary hepatocellular carcinoma. Eur J Gastroenterol Hepatol 2025; 37:534-539. [PMID: 39976070 DOI: 10.1097/meg.0000000000002916] [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] [Indexed: 02/21/2025]
Abstract
Recently, immune checkpoint inhibitors have been widely used in the treatment of advanced liver cancer. Immune checkpoints are a type of molecules that play an important role in the self-regulation of the immune system. In tumor immunity, their activation by immune checkpoints leads to the inhibition of effector lymphocyte activation or the mediation of cytotoxic T cell dysfunction, resulting in immune escape. These immune checkpoints include programmed death receptor 1 (PD-1) and its ligand PD-L1, as well as cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and others. Immune checkpoint inhibitors block the interaction between immune checkpoint receptors and ligands, thereby relieving the immune suppression caused by immune checkpoints, and reactivating immune cells to exert antitumor effects. With the continuous progress of immunotherapy research, drugs targeting PDL-1, PD-1, and CTLA-4 have played an important role in clinical treatment. However, some patients still cannot benefit from immunotherapy; therefore, multitarget immunotherapy is an important way to improve the response rate of immunotherapy. CD96 is one of the members of the immunoglobulin superfamily receptors, which mainly functions by regulating natural killer cells and CD8+ T cells, and is expected to become a new generation of immune checkpoints. This article reviews the molecular structure of CD96, its role in tumor immunity, and its application in hepatocellular carcinoma, hoping to provide reference for related research.
Collapse
Affiliation(s)
- Jiaqi Chai
- Department of Colorectal Surgery, 731 Hospital of China Aerospace Science and Industry Group
| | - Luyang Li
- Department of Hepatobiliary Surgery, Air Force Medical Center, Air Force Medical University Beijing
| | - Qimei Wu
- Graduate School of China Medical University, Shenyang, China
| | - Shuhan Zhang
- Department of Hepatobiliary Surgery, Air Force Medical Center, Air Force Medical University Beijing
| |
Collapse
|
15
|
Li L, Pu H, Zhang X, Guo X, Li G, Zhang M. Resistance to PD-1/PD-L1 immune checkpoint blockade in advanced non-small cell lung cancer. Crit Rev Oncol Hematol 2025; 209:104683. [PMID: 40024354 DOI: 10.1016/j.critrevonc.2025.104683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
Lung cancer is one of the most common malignant tumors, of which non-small cell lung cancer (NSCLC) accounts for about 85 %. Although immune checkpoint inhibitors (ICIs), particularly PD-1/PD-L1 inhibitors, have significantly improved the prognosis of patients with NSCLC. There are still many patients do not benefit from ICIs. Primary resistance remains a major challenge in advanced NSCLC. The cancer-immunity cycle describes the process from antigen release to T cell recognition and killing of the tumor, which provides a framework for understanding anti-tumor immunity. The classical cycle consists of seven steps, and alterations at each stage can result in resistance. This review examines the current status of PD-1/PD-L1 blockade in the treatment of advanced NSCLC and explores potential mechanisms of resistance. We summarize the latest clinical trials of PD-1/PD-L1 inhibitors combined with other therapies and explore potential targets for overcoming primary resistance to PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Lijun Li
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Haihong Pu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xiaoxin Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xiaotian Guo
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Guangrui Li
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Minghui Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
16
|
Weng D, Guo R, Gao Y, Xu S, Li Y, Zhou J, An R, Xu H. Immuno-PET Imaging of a 68Ga-Labeled Single-Domain Antibody for Detecting Tumor TIGIT Expression. Mol Pharm 2025. [PMID: 40298304 DOI: 10.1021/acs.molpharmaceut.4c00900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Preclinical studies have shown that the expression of T cell immunoglobulin and the immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) in the tumor microenvironment is associated with the efficacy of anti-TIGIT-based immunotherapy. This study aimed to develop a TIGIT single-domain antibody (sdAb)-based positron emission tomography (PET) radiotracer, [68Ga]Ga-NOTA-NABT3A1, and evaluate its characteristics. The NABT3A1 was modified with a NOTA derivative and radiolabeled with 68Ga. In vitro stability of [68Ga]Ga-NOTA-NABT3A1 was assessed in phosphate-buffered saline (PBS) and fetal bovine serum (FBS), along with its specificity for TIGIT stably transfected A375 Human melanoma cells (A375-TIGIT) was performed. In vivo imaging was conducted on A375-TIGIT tumor-bearing nude mice at different time points after the injection of [68Ga]Ga-NOTA-NABT3A1. The synthesized [68Ga]Ga-NOTA-NABT3A1 achieved a radiochemical yield of 70.56 ± 1.14% and purity levels of 95.80 ± 0.58% in PBS and 96.79 ± 1.69% in FBS at 2 h. Immuno-PET imaging revealed specific accumulation of [68Ga]Ga-NOTA-NABT3A1 in A375-TIGIT tumor-bearing nude mice, with a maximum uptake of 3.86 ± 0.29% injected dose/g at 0.5 h. Biodistribution and immunohistochemical analyses confirmed the in vivo imaging results. In conclusion, we successfully synthesized an NABT3A1-derived PET radiotracer with the potential to noninvasively assess TIGIT expression in tumors.
Collapse
Affiliation(s)
- Dinghu Weng
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 201318 Shanghai, China
- Department of Medical Imaging, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, 430071 Wuhan, China
- Wuhan Clinical Research and Development Center of Brain Resuscitation and Functional Imaging, 430071 Wuhan, China
| | - Rong Guo
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430000, China
| | - Yu Gao
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430000, China
| | - Shasha Xu
- Beijing Novaboody Biotechnological Ltd., Beijing 102600, China
| | - Yingying Li
- Beijing Novaboody Biotechnological Ltd., Beijing 102600, China
| | - Jun Zhou
- Interventional Diagnostic and Therapeutic Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Rui An
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430000, China
| | - Haibo Xu
- Department of Medical Imaging, Zhongnan Hospital of Wuhan University, 430071 Wuhan, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, 430071 Wuhan, China
- Wuhan Clinical Research and Development Center of Brain Resuscitation and Functional Imaging, 430071 Wuhan, China
| |
Collapse
|
17
|
Liu R, Jiang X, Dong R, Zhang Y, Gai C, Wei P. Revealing the mechanisms and therapeutic potential of immune checkpoint proteins across diverse protein families. Front Immunol 2025; 16:1499663. [PMID: 40356928 PMCID: PMC12066663 DOI: 10.3389/fimmu.2025.1499663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 03/28/2025] [Indexed: 05/15/2025] Open
Abstract
Host immune responses to antigens are tightly regulated through the activation and inhibition of synergistic signaling networks that maintain homeostasis. Stimulatory checkpoint molecules initiate attacks on infected or tumor cells, while inhibitory molecules halt the immune response to prevent overreaction and self-injury. Multiple immune checkpoint proteins are grouped into families based on common structural domains or origins, yet the variability within and between these families remains largely unexplored. In this review, we discuss the current understanding of the mechanisms underlying the co-suppressive functions of CTLA-4, PD-1, and other prominent immune checkpoint pathways. Additionally, we examine the IgSF, PVR, TIM, SIRP, and TNF families, including key members such as TIGIT, LAG-3, VISTA, TIM-3, SIRPα, and OX40. We also highlight the unique dual role of VISTA and SIRPα in modulating immune responses under specific conditions, and explore potential immunotherapeutic pathways tailored to the distinct characteristics of different immune checkpoint proteins. These insights into the unique advantages of checkpoint proteins provide new directions for drug discovery, emphasizing that emerging immune checkpoint molecules could serve as targets for novel therapies in cancer, autoimmune diseases, infectious diseases, and transplant rejection.
Collapse
Affiliation(s)
| | | | | | | | - Cong Gai
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Peng Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
18
|
He Y, Tian T, Li Y, Zeng Y, Wang X, Qian L, Tian T, Jiang M, Li L. From neglect to necessity: the role of innate immunity in cutaneous squamous cell carcinoma therapy. Front Immunol 2025; 16:1570032. [PMID: 40352926 PMCID: PMC12061915 DOI: 10.3389/fimmu.2025.1570032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 04/03/2025] [Indexed: 05/14/2025] Open
Abstract
As the second most common non-melanoma skin cancer, cutaneous squamous cell carcinoma (cSCC) has experienced a significant increase in incidence. Although clinical detection is relatively easy, a considerable number of patients are diagnosed at an advanced stage, featuring local tissue infiltration and distant metastasis. Cemiplimab, along with other immune checkpoint inhibitors, enhances T cell activation by blocking the PD-1 pathway, resulting in notable improvements in clinical outcomes. Nonetheless, approximately 50% of the patients with advanced cSCC remain unresponsive to this therapeutic approach. It emphasizes the importance of finding innovative therapeutic targets and strategies to boost the success of immunotherapy across a wider range of patients. Therefore, we focused on frequently neglected functions of innate immune cells. Emerging evidence indicates that innate immune cells exhibit considerable heterogeneity and plasticity, fundamentally contributing to tumor initiation and development. The identification and eradication of cancer cells, along with the modulation of adaptive immune responses, are essential roles of these cells. Consequently, targeting innate immune cells to activate anti-tumor immune responses presents significant potential for enhancing immunotherapeutic strategies in cSCC.
Collapse
Affiliation(s)
- Yong He
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Ting Tian
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Yuancheng Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Yong Zeng
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, The Second Xiangya Hospital of Central South University, Changsha, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Xiaoke Wang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Leqi Qian
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Tian Tian
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Mingjun Jiang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Liming Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| |
Collapse
|
19
|
Kabut J, Gorzelak-Magiera A, Gisterek-Grocholska I. New Therapeutic Targets TIGIT, LAG-3 and TIM-3 in the Treatment of Advanced, Non-Small-Cell Lung Cancer. Int J Mol Sci 2025; 26:4096. [PMID: 40362333 PMCID: PMC12072094 DOI: 10.3390/ijms26094096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/20/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
The introduction of immunotherapy and target therapy into clinical practice has become a chance for many patients with cancer to prolong their survival while maintaining optimal quality of life. Treatment of lung cancer is excellent evidence of the progress of medical therapies. An understanding of the mechanisms of tumor development has led to the evolution of new methods of treatment. Immunoreceptors of T cells with the immunoglobulin domain ITIM, TIM-3 (T-cell immunoglobulin- and mucin domain-3-containing molecule 3), and LAG-3 (lymphocyte activation gene-3) represent new interesting therapeutic targets. The combination of anti-PD-1 and anti-CTLA-4 blockade has proven the possibility of strengthening the anti-tumor response by acting via two separate mechanisms. Adding additional checkpoints to the PD-1 blockade offers hope for further improvements in the effects of the treatment of patients and expanding the group responding to immunotherapy. This paper presents new promising molecular targets along with studies demonstrating the treatment results using them.
Collapse
Affiliation(s)
- Jacek Kabut
- Department of Oncology and Radiotherapy, Medical University of Silesia, 40-514 Katowice, Poland;
| | | | | |
Collapse
|
20
|
Ma S, Yu J, Caligiuri MA. Natural killer cell-based immunotherapy for cancer. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf036. [PMID: 40246292 DOI: 10.1093/jimmun/vkaf036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/18/2025] [Indexed: 04/19/2025]
Abstract
Natural killer (NK) cells are emerging as a promising tool for cancer immunotherapy due to their innate ability to selectively recognize and eliminate cancer cells. Over the past 3 decades, strategies to harness NK cells have included cytokines, small molecules, antibodies, and the adoptive transfer of autologous or allogeneic NK cells, both unmodified and genetically engineered. Despite favorable safety profiles in clinical trials, challenges such as limited in vivo persistence, exhaustion, and the suppressive tumor microenvironment continue to hinder their efficacy and durability. This review categorizes NK cell-based therapies into 3 major approaches: (i) cellular therapies, including unmodified and chimeric antigen receptor-engineered NK cells; (ii) cytokine-based strategies such as interleukin-2 and interleukin-15 derivatives; and (iii) antibody-based therapies, including immune checkpoint inhibitors and NK cell engagers. We highlight these advancements, discuss current limitations, and propose strategies to optimize NK cell-based therapies for improved cancer treatment outcomes.
Collapse
Affiliation(s)
- Shoubao Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, United States
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, United States
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, United States
| | - Jianhua Yu
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, University of California, Irvine, CA, United States
- Institute for Precision Cancer Therapeutics and Immuno-Oncology, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, United States
- Clemons Family Center for Transformative Cancer Research, University of California, Irvine, Irvine, CA, United States
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, United States
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, United States
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, United States
| |
Collapse
|
21
|
Chen J, Li H, Jin Q, Li X, Zhang Y, Shen J, Huang G, Yin J, Zou C, Li X, He X, Xie X, Lin T. Troxerutin suppresses the stemness of osteosarcoma via the CD155/SRC/β-catenin signaling axis. Cell Mol Biol Lett 2025; 30:45. [PMID: 40217455 PMCID: PMC11992710 DOI: 10.1186/s11658-025-00724-8] [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: 10/08/2024] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Osteosarcoma is the most prevalent primary malignant bone tumor affecting pediatric and adolescent individuals. However, despite the passage of three decades, there has been no notable enhancement in the overall survival rate of patients with osteosarcoma. In recent years, CD155 has been reported to exhibit abnormal amplification in a range of tumors, yet the precise underlying mechanism remains elusive. The objective of this study is to investigate the role of CD155 in osteosarcoma, and to identify drugs that specifically target this molecule, thereby offering a novel direction for the treatment of osteosarcoma. METHODS The prognosis of patients with osteosarcoma with high and low expression of CD155 was verified by immunohistochemistry. CCK-8 and colony formation assays were used to detect cell proliferation and drug resistance. Transwell experiments were used to detect cell migration and invasion. The sphere formation experiment was used to evaluate the stemness of tumor cells. Additionally, in vivo animal models were utilized to assess the functional role of CD155 in a biological context. RNA-seq and co-immunoprecipitation methods were used to search for downstream target molecules and signaling pathways of CD155. Finally, virtual screening was used to find drugs targeting CD155. RESULTS In this study, we have established the significant amplification of CD155 in osteosarcoma. Utilizing a comprehensive array of experimental methods, including CCK-8 assay, colony formation assay, Transwell assay, and in vivo animal models, we unequivocally demonstrate that CD155 significantly potentiates the malignancy of osteosarcoma both in vitro and in vivo. Additionally, our findings reveal that CD155 promotes osteosarcoma stemness by modulating the Wnt/β-catenin signaling pathway. Advanced molecular techniques, such as RNA sequencing and co-immunoprecipitation, have been instrumental in elucidating the mechanism of CD155 in activating the Wnt/β-catenin pathway via the SRC/AKT/GSK3β signaling axis, thereby enhancing the stem-cell-like properties of osteosarcoma cells. To explore targeted therapeutic options, we conducted virtual screening and identified troxerutin as a promising CD155 inhibitor. CONCLUSIONS Our findings reveal that troxerutin effectively inhibits CD155, attenuates the SRC/AKT/GSK3β signaling cascade, diminishes the nuclear localization of β-catenin, and consequently mitigates osteosarcoma stemness. These discoveries position troxerutin as a promising candidate for targeted osteosarcoma therapy.
Collapse
Affiliation(s)
- Junkai Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hongbo Li
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qinglin Jin
- Department of Musculoskeletal Oncology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xiaoguang Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yiwen Zhang
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jingnan Shen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Gang Huang
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Junqiang Yin
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Changye Zou
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xinyu Li
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xin He
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Xianbiao Xie
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Tiao Lin
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
22
|
Balkhi S, Zuccolotto G, Di Spirito A, Rosato A, Mortara L. CAR-NK cell therapy: promise and challenges in solid tumors. Front Immunol 2025; 16:1574742. [PMID: 40260240 PMCID: PMC12009813 DOI: 10.3389/fimmu.2025.1574742] [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: 02/11/2025] [Accepted: 03/17/2025] [Indexed: 04/23/2025] Open
Abstract
Over the past few years, cellular immunotherapy has emerged as a promising treatment for certain hematologic cancers, with various CAR-T therapies now widely used in clinical settings. However, challenges related to the production of autologous cell products and the management of CAR-T cell toxicity highlight the need for new cell therapy options that are universal, safe, and effective. Natural killer (NK) cells, which are part of the innate immune system, offer unique advantages, including the potential for off-the-shelf therapy. A recent first-in-human trial of CD19-CAR-NK infusion in patients with relapsed/refractory lymphoid malignancies demonstrated safety and promising clinical activity. Building on these positive clinical outcomes, current research focuses on enhancing CAR-NK cell potency by increasing their in vivo persistence and addressing functional exhaustion. There is also growing interest in applying the successes seen in hematologic malignancies to solid tumors. This review discusses current trends and emerging concepts in the engineering of next-generation CAR- NK therapies. It will cover the process of constructing CAR-NK cells, potential targets for their manufacturing, and their role in various solid tumors. Additionally, it will examine the mechanisms of action and the research status of CAR-NK therapies in the treatment of solid tumors, along with their advantages, limitations, and future challenges. The insights provided may guide future investigations aimed at optimizing CAR-NK therapy for a broader range of malignancies.
Collapse
Affiliation(s)
- Sahar Balkhi
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Gaia Zuccolotto
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Anna Di Spirito
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| |
Collapse
|
23
|
Chen YZ, Meng ZS, Zhang YN, Xiang ZL. Natural Killer Cell-Associated Radiogenomics Model for Hepatocellular Carcinoma: Integrating CD2 and Enhanced CT-Derived Radiomics Signatures. Acad Radiol 2025; 32:1981-1992. [PMID: 39542805 DOI: 10.1016/j.acra.2024.10.043] [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: 09/14/2024] [Revised: 10/13/2024] [Accepted: 10/24/2024] [Indexed: 11/17/2024]
Abstract
RATIONALE AND OBJECTIVES Hepatocellular carcinoma (HCC) is a leading cause of cancer mortality. Natural Killer (NK) cells play a crucial role in immune defense against HCC, but their activity is often impaired by the tumor microenvironment (TME). This study aims to integrate radiomics and transcriptomics to develop a prognostic model linking NK cell characteristics to clinical outcomes in HCC. METHODS Transcriptomic data from five cohorts (734 HCC patients) from the Gene Expression Omnibus and The Cancer Genome Atlas databases were analyzed using the Microenvironment Cell Populations-counter algorithm. NK cell-related prognostic biomarkers were identified via weighted gene co-expression network analysis and LASSO-Cox regression. Radiomics models were established using CT imaging features from 239 patients in three datasets from The Cancer Imaging Archive and Shanghai East Hospital. HCC radiogenomic subtypes were proposed by integrating genetic biomarkers and radiomics models. RESULTS CD2 expression was identified as an independent NK cell-related prognostic biomarker, with a positive impact on prognosis and a strong correlation with NK cell-associated biological processes in HCC. A robust radiomics model was constructed, and the integration of CD2 expression with radioscore identified potential radiogenomic subtypes of HCC. CONCLUSION Radiomics has potential to link TME immune phenotypes with HCC prognosis. CD2 is a key biomarker connecting NK cells with radiomic features, offering a new classification of HCC into radiogenomic subtypes. This approach supports the use of radiogenomics in personalized HCC treatment.
Collapse
Affiliation(s)
- Yan-Zhu Chen
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China (Y.Z.C., Z.L.X.)
| | - Zhi-Shang Meng
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China (Z.S.M.)
| | - Yan-Nan Zhang
- Department of Radiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China (Y.N.Z.)
| | - Zuo-Lin Xiang
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China (Y.Z.C., Z.L.X.); Department of Radiation Oncology, Shanghai East Hospital Ji'an Hospital, Ji'an, China (Z.L.X.).
| |
Collapse
|
24
|
Mousavi S, Khazaee-Nasirabadi MH, Seyedmehdi MS, Bazi A, Mirzaee Khalilabadi R. Natural killer cells: a new promising source for developing chimeric antigen receptor anti-cancer cells in hematological malignancies. Leuk Lymphoma 2025; 66:594-616. [PMID: 39656564 DOI: 10.1080/10428194.2024.2438802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 11/18/2024] [Accepted: 12/01/2024] [Indexed: 12/17/2024]
Abstract
In recent times, the application of CAR-T cell treatment has significantly progressed, showing auspicious treatment outcomes in hematologic malignancies. However, along with these advances, certain limitations and challenges hurdle the widespread utilization of this technology. Recently, CAR-NK cells have gained attention in cancer treatment, as this approach has an important advantage over CART therapy (i.e. no need for HLA matching) for targeting foreign cells. This review aims to explore the benefits of CAR NK cell therapy, and generation strategies, as well as the challenges and limitations hindering the application of CAR NK cells in experimental studies and trials on hematologic malignancies.
Collapse
Affiliation(s)
- Shahrzad Mousavi
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Maryam Sadat Seyedmehdi
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Technology, Shahid Beheshti University, Tehran, Islamic Republic of Iran
| | - Ali Bazi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Faculty of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Roohollah Mirzaee Khalilabadi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
25
|
Chuang CH, Guo JC, Kato K, Hsu CH. Exploring novel immunotherapy in advanced esophageal squamous cell carcinoma: Is targeting TIGIT an answer? Esophagus 2025; 22:139-147. [PMID: 39847233 PMCID: PMC11929690 DOI: 10.1007/s10388-024-01105-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/26/2024] [Indexed: 01/24/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a prevalent and highly lethal malignancy in Asia. Recent advancements in immune checkpoint inhibitors (ICIs) have markedly transformed the systemic therapy landscape for ESCC. Anti-PD-1-based combination with chemotherapy or with ipilimumab, an anti-CTLA-4 antibody, have been established as the new standard first-line treatments for patients with advanced ESCC. Moreover, anti-PD-1 monotherapy has demonstrated improved efficacy and survival compared with second-line chemotherapy in previously treated patients with ESCC. Novel ICIs targeting other immune checkpoints also show potential for enhancing anticancer therapy in advanced ESCC.The TIGIT/PVR pathway represents a new immune checkpoint. Preclinical studies have indicated that the dual blockade of TIGIT and PD-1 can enhance antitumor immune responses. Clinical trials have reported that combining anti-TIGIT with anti-PD-1/PD-L1 antibodies elicited clinical responses in patients with advanced ESCC. In the first-line systemic therapy setting, combinations of dual ICIs targeting TIGIT and PD-1/PD-L1 plus platinum-based chemotherapy have demonstrated acceptable toxicity profiles and promising antitumor activity in several phase II trials and one phase III study. However, the role of adding an anti-TIGIT antibody to the current standard of anti-PD-1/PD-L1 plus platinum-based chemotherapy in first-line therapy for advanced ESCC remains to be fully determined, necessitating further clinical trials. Ongoing studies are also investigating the role of anti-TIGIT, with or without anti-PD-1/PD-L1, in locoregional ESCC. Additional research is essential to optimize the potential of anti-TIGIT therapy in ESCC and other malignancies by identifying predictive biomarkers, determining optimal antibody types, and gaining key mechanistic insights.
Collapse
Affiliation(s)
- Chien-Huai Chuang
- Department of Medical Oncology, National Taiwan University Cancer Center, 7 Chung-Shan South Road, Taipei, 10002, Taiwan
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jhe-Cyuan Guo
- Department of Medical Oncology, National Taiwan University Cancer Center, 7 Chung-Shan South Road, Taipei, 10002, Taiwan
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ken Kato
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan.
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Chih-Hung Hsu
- Department of Medical Oncology, National Taiwan University Cancer Center, 7 Chung-Shan South Road, Taipei, 10002, Taiwan.
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
26
|
Qian L, Wu L, Miao X, Xu J, Zhou Y. The role of TIGIT-CD226-PVR axis in mediating T cell exhaustion and apoptosis in NSCLC. Apoptosis 2025; 30:784-804. [PMID: 39725799 DOI: 10.1007/s10495-024-02052-2] [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: 11/24/2024] [Indexed: 12/28/2024]
Abstract
The treatment of non-small cell lung cancer (NSCLC) remains a critical challenge in oncology, primarily due to the dysfunction and exhaustion of T cells within the tumor microenvironment, which greatly limits the effectiveness of immunotherapy. This study investigates the regulatory role of the T cell immunoglobulin and ITIM domain (TIGIT)-CD226-PVR signaling axis in the exhaustion and apoptosis of cluster of differentiation (CD)27+/CD127+T cells in NSCLC. Utilizing single-cell sequencing technology, we conducted a comprehensive gene expression analysis of T cells in a mouse model of NSCLC. Bioinformatics analysis revealed that the TIGIT-CD226-PVR signaling axis is highly active in the CD27+/CD127+T cell subset and is closely associated with their functional decline and exhaustion. In vitro experiments further demonstrated that inhibiting the TIGIT-PVR pathway while activating the CD226-PVR pathway significantly restored T cell proliferation and effector function. Importantly, in vivo studies showed that targeting this axis can significantly alleviate T cell exhaustion, enhance their cytotoxicity against NSCLC cells, and promote apoptosis, thereby improving the efficacy of immunotherapy.
Collapse
MESH Headings
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Apoptosis/genetics
- Animals
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- T Lineage-Specific Activation Antigen 1
- Humans
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Antigens, Differentiation, T-Lymphocyte/genetics
- Mice
- Receptors, Virus/metabolism
- Receptors, Virus/genetics
- Signal Transduction
- Cell Line, Tumor
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Microenvironment/immunology
- Cell Proliferation
- T-Cell Exhaustion
Collapse
Affiliation(s)
- Liang Qian
- Department of Respiratory and Critical Care Medicine, WuJin Hospital Afliated With Jiangsu University, WuJin Clinical College of Xuzhou Medical University, No.2, Yongning North Road, Changzhou, 213017, Jiangsu, China
| | - Ling Wu
- Department of Respiratory and Critical Care Medicine, WuJin Hospital Afliated With Jiangsu University, WuJin Clinical College of Xuzhou Medical University, No.2, Yongning North Road, Changzhou, 213017, Jiangsu, China
| | - Xiaohui Miao
- Department of Respiratory and Critical Care Medicine, WuJin Hospital Afliated With Jiangsu University, WuJin Clinical College of Xuzhou Medical University, No.2, Yongning North Road, Changzhou, 213017, Jiangsu, China
| | - Jiao Xu
- Department of Respiratory and Critical Care Medicine, WuJin Hospital Afliated With Jiangsu University, WuJin Clinical College of Xuzhou Medical University, No.2, Yongning North Road, Changzhou, 213017, Jiangsu, China
| | - Yao Zhou
- Department of Respiratory and Critical Care Medicine, WuJin Hospital Afliated With Jiangsu University, WuJin Clinical College of Xuzhou Medical University, No.2, Yongning North Road, Changzhou, 213017, Jiangsu, China.
| |
Collapse
|
27
|
Nair R, Somasundaram V, Kuriakose A, Krishn SR, Raben D, Salazar R, Nair P. Deciphering T-cell exhaustion in the tumor microenvironment: paving the way for innovative solid tumor therapies. Front Immunol 2025; 16:1548234. [PMID: 40236693 PMCID: PMC11996672 DOI: 10.3389/fimmu.2025.1548234] [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: 12/19/2024] [Accepted: 03/14/2025] [Indexed: 04/17/2025] Open
Abstract
In solid tumors, the tumor microenvironment (TME) is a complex mix of tumor, immune, stromal cells, fibroblasts, and the extracellular matrix. Cytotoxic T lymphocytes (CTLs) constitute a fraction of immune cells that may infiltrate into the TME. The primary function of these T-cells is to detect and eliminate tumor cells. However, due to the immunosuppressive factors present in the TME primarily mediated by Myeloid-Derived Suppressor Cells (MDSCs), Tumor associated macrophages (TAMs), Cancer Associated Fibroblasts (CAFs) as well as the tumor cells themselves, T-cells fail to differentiate into effector cells or become dysfunctional and are unable to eliminate the tumor. In addition, chronic antigen stimulation within the TME also leads to a phenomenon, first identified in chronic lymphocytic choriomeningitis virus (LCMV) infection in mice, where the T-cells become exhausted and lose their effector functions. Exhausted T-cells (Tex) are characterized by the presence of remarkably conserved inhibitory receptors, transcription and signaling factors and the downregulation of key effector molecules. Tex cells have been identified in various malignancies, including melanoma, colorectal and hepatocellular cancers. Recent studies have indicated novel strategies to reverse T-cell exhaustion. These include checkpoint inhibitor blockade targeting programmed cell death protein 1 (PD-1), T-cell immunoglobulin and mucin-domain containing-3 (Tim-3), cytotoxic T-lymphocyte associated protein 4 (CTLA-4), or combinations of different immune checkpoint therapies (ICTs) or combination of ICTs with cytokine co-stimulation. In this review, we discuss aspects of T-cell dysfunction within the TME with a focus on T-cell exhaustion. We believe that gaining insight into the mechanisms of T-cell exhaustion within the TME of human solid tumors will pave the way for developing therapeutic strategies to target and potentially re-invigorate exhausted T-cells in cancer.
Collapse
Affiliation(s)
- Reshmi Nair
- Syngene International Limited, Bengaluru, India
| | | | | | | | - David Raben
- Bicara Therapeutics, Boston, MA, United States
| | | | - Pradip Nair
- Syngene International Limited, Bengaluru, India
| |
Collapse
|
28
|
Rothfuß C, Baumann T, Donakonda S, Brauchle B, Marcinek A, Urban C, Mergner J, Pedde AM, Hirschberger A, Krupka C, Neumann AS, Hänel G, Merten C, Öllinger R, Hecker JS, Bauer T, Schmid C, Götze KS, Altomonte J, Bücklein V, Jacobs R, Rad R, Dawid C, Simeoni L, Schraven B, Pichlmair A, Subklewe M, Knolle PA, Böttcher JP, Höchst B. Two-layered immune escape in AML is overcome by Fcγ receptor activation and inhibition of PGE2 signaling in NK cells. Blood 2025; 145:1395-1406. [PMID: 39840945 DOI: 10.1182/blood.2024025706] [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: 06/13/2024] [Revised: 11/13/2024] [Accepted: 11/29/2024] [Indexed: 01/23/2025] Open
Abstract
ABSTRACT Loss of anticancer natural killer (NK) cell function in patients with acute myeloid leukemia (AML) is associated with fatal disease progression and remains poorly understood. Here, we demonstrate that AML blasts isolated from patients rapidly inhibit NK cell function and escape NK cell-mediated killing. Transcriptome analysis of NK cells exposed to AML blasts revealed increased CREM expression and transcriptional activity, indicating enhanced cyclic adenosine monophosphate (cAMP) signaling, confirmed by uniform production of the cAMP-inducing prostanoid prostaglandin E2 (PGE2) by all AML-blast isolates from patients. Phosphoproteome analysis disclosed that PGE2 induced a blockade of lymphocyte-specific protein tyrosine kinase (LCK)-extracellular signal-regulated kinase signaling that is crucial for NK cell activation, indicating a 2-layered escape of AML blasts with low expression of NK cell-activating ligands and inhibition of NK cell signaling. To evaluate the therapeutic potential to target PGE2 inhibition, we combined Fcγ-receptor-mediated activation with the prevention of inhibitory PGE2 signaling. This rescued NK cell function and restored the killing of AML blasts. Thus, we identify the PGE2-LCK signaling axis as the key barrier for NK cell activation in 2-layered immune escape of AML blasts that can be targeted for immune therapy to reconstitute anticancer NK cell immunity in patients with AML.
Collapse
MESH Headings
- Humans
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/pathology
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Dinoprostone/immunology
- Dinoprostone/metabolism
- Signal Transduction/immunology
- Receptors, IgG/immunology
- Receptors, IgG/metabolism
- Tumor Escape/immunology
- Lymphocyte Activation/immunology
Collapse
Affiliation(s)
- Charlotte Rothfuß
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Tobias Baumann
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Sainitin Donakonda
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Bettina Brauchle
- Gene Center, Laboratory for Translational Cancer Immunology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Anetta Marcinek
- Gene Center, Laboratory for Translational Cancer Immunology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christian Urban
- Institute of Virology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Julia Mergner
- Bavarian Center for Biomolecular Mass Spectrometry at Munich Institute of Robotics and Machine Intelligence, Technical University of Munich, Munich, Germany
| | - Anna-Marie Pedde
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Anna Hirschberger
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Christina Krupka
- Gene Center, Laboratory for Translational Cancer Immunology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Anne-Sophie Neumann
- Gene Center, Laboratory for Translational Cancer Immunology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Gerulf Hänel
- Gene Center, Laboratory for Translational Cancer Immunology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Camilla Merten
- Institut of Molecular and Clinical Immunology, Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany
| | - Rupert Öllinger
- Institute of Molecular Oncology and Functional Genomics, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Judith S Hecker
- Department of Medicine III, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Tanja Bauer
- Institute of Virology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Christian Schmid
- Food Chemistry and Molecular Sensory Science, Technical University of Munich, Munich, Germany
| | - Katharina S Götze
- Department of Medicine III, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Jennifer Altomonte
- Department of Internal Medicine II, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Veit Bücklein
- Gene Center, Laboratory for Translational Cancer Immunology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Roland Jacobs
- Department of Rheumatology and Clinical Immunology, Hannover Medical School, Hannover, Germany
| | - Roland Rad
- Institute of Molecular Oncology and Functional Genomics, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Corina Dawid
- Food Chemistry and Molecular Sensory Science, Technical University of Munich, Munich, Germany
| | - Luca Simeoni
- Institut of Molecular and Clinical Immunology, Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany
| | - Burkhart Schraven
- Institut of Molecular and Clinical Immunology, Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany
| | - Andreas Pichlmair
- Institute of Virology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Marion Subklewe
- Gene Center, Laboratory for Translational Cancer Immunology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Percy A Knolle
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Molecular Immunology, School of Life Science, Technical University of Munich, Munich, Germany
| | - Jan P Böttcher
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Department of Experimental Immunology, Institute of Immunology, University of Tübingen, Tübingen, Germany
| | - Bastian Höchst
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| |
Collapse
|
29
|
Nie Z, Chang J, Yang Z, Zeng K, Liu Y, Tu Q, Wang C, Yan Q, Shi H, Guo Y. PRMT5 highly expressed on CD16 + CD56- natural killer cells is correlated with NK cells exhaustion in colorectal cancer mesenchyme. Cancer Immunol Immunother 2025; 74:139. [PMID: 40056169 PMCID: PMC11890482 DOI: 10.1007/s00262-025-03981-w] [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: 12/25/2024] [Accepted: 02/11/2025] [Indexed: 03/10/2025]
Abstract
OBJECTIVE To investigate the relationships between changes in the phenotype of natural killer cells (NK cells) in the microenvironment of colorectal cancer (CRC) and the expression of important immune checkpoints. To assess the expression level of CD16 bright CD56 negative (CD16 + CD56-) NK cell-associated immune checkpoints, including protein arginine methyltransferase 5 (PRMT5) and T-cell immunoreceptor with Ig and ITIM domains (TIGIT), single-immunoglobulin interleukin-1-related receptor (SIGIRR), in CRC mesenchyme. METHODS A total of 194 patients who were diagnosed with CRC were screened. The percentage of NK cells and the expression levels of their surface receptors, including PRMT5, CD56, CD69, TIGIT, CD16, IFN-γ, and SIGIRR, in the tumor microenvironment (TME) of CRC were assessed. Immunohistochemical staining, multiplex immunohistochemistry, and single-cell sequencing were performed. RESULTS Compared with normal mesenchyme, NK cells were less in CRC mesenchyme. The percentage of CD16 + CD56- NK cells in tumor mesenchyme was significantly higher, the number of CD16 + NK cells was more, and the number of CD56 + NK cells was less in CRC mesenchyme. High expression of TIGIT and PRMT5 expression affected the progression of CRC. The expression of PRMT5 and SIGIRR expression was significantly increased in CD16 + CD56- NK cells, and both genes were identified as important morbidity factors. PRMT5 and SIGIRR may contribute to the phenotype changes of NK cells in CRC. CONCLUSION The microenvironment of CRC is in an immunosuppressive state characterized mainly by high expression of TIGIT, CD16, PRMT5, and SIGIRR; low expression of CD56, IFN-γ, and CD69; significantly decreased percentage of CD56 + NK cells; and significantly increased percentage of CD16 + CD56- NK cells with weakened killing ability. PRMT5 and TIGIT may be closely related to the formation of CD16 + CD56- NK cells with weakened killing ability.
Collapse
Affiliation(s)
- Zunzhen Nie
- Xi'an Daxing Hospital Affiliated to Yan'an University, Xi'an, People's Republic of China
| | - Juanjuan Chang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, People's Republic of China
| | - Zhiqin Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, People's Republic of China
| | - Kaixuan Zeng
- Precision Medical Research Institute, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yuangang Liu
- Xi'an Daxing Hospital Affiliated to Yan'an University, Xi'an, People's Republic of China
| | - Qian Tu
- Xi'an Daxing Hospital Affiliated to Yan'an University, Xi'an, People's Republic of China
| | - Chao Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, People's Republic of China
| | - Qingguo Yan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, People's Republic of China
| | - Hai Shi
- Department of Surgery, Xi'an Daxing Hospital, Xi'an, People's Republic of China
| | - Ying Guo
- Xi'an Daxing Hospital Affiliated to Yan'an University, Xi'an, People's Republic of China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, People's Republic of China.
| |
Collapse
|
30
|
Wang W, Wu H, Zhang X, Hong Y, Tao S, Cao X, Wang S, Zha L, Zha Z. Whole-Component Antigen Nanovaccines Combined With aTIGIT for Enhanced Innate and Adaptive Anti-tumor Immunity. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2412800. [PMID: 39967373 DOI: 10.1002/smll.202412800] [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: 12/30/2024] [Revised: 01/27/2025] [Indexed: 02/20/2025]
Abstract
Using entire tumor cells or tissues that display both common and patient-specific antigens can potentially trigger a comprehensive and long-lasting anti-tumor immune response. However, the limited immunogenicity, low uptake efficiency, and susceptibility to degradation of whole-component antigens present significant challenges. In this study, we employed tumor lysates (TLs) as whole-component antigens, in conjunction with MgAl-layered double hydroxide (MA) as nanoadjuvants and Mn2+ as immunostimulants, to create personalized MMAT (Mn2+-MA-TLs) nanovaccines. After subcutaneous injection of MMAT nanovaccines, the high local concentrations of TLs and Mn2+ facilitated the recruitment and activation of antigen-presenting cells (APCs), thereby inducing a robust adaptive immune response. Remarkably, MMAT nanovaccines enabled lysosomal escape, enhanced antigen cross-presentation, and activated the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway in APCs. Furthermore, MMAT nanovaccines, when combined with the anti-TIGIT monoclonal antibody (aTIGIT), an immune checkpoint inhibitor, not only stimulated T-cell-based adaptive anti-tumor immune responses but also activated the NK-cell-based innate anti-tumor immunity, effectively suppressing tumor growth, recurrence, and metastasis. Thus, the ternary MMAT nanovaccines developed here introduced a pioneered paradigm for the rapid preparation of whole-component tumor antigens with nanoadjuvants and immunostimulants into nanovaccines, offering new prospects for clinical immunotherapies.
Collapse
Affiliation(s)
- Weitao Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Haitao Wu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Xuan Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Yang Hong
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Shi Tao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Xiangjing Cao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Shipeng Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Lisha Zha
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Zhengbao Zha
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| |
Collapse
|
31
|
Gao R, Wu P, Yin X, Zhuang L, Meng X. Deep analysis of the trials and major challenges in the first-line treatment for patients with extensive-stage small cell lung cancer. Int Immunopharmacol 2025; 148:114116. [PMID: 39847950 DOI: 10.1016/j.intimp.2025.114116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 01/25/2025]
Abstract
The median overall survival (OS) is approximately 10 months when chemotherapy alone is the first-line treatment for extensive-stage small cell lung cancer (ES-SCLC). The approval of the two PD-L1 inhibitors, atezolizumab and durvalumab, marked the beginning of the immunotherapy era for ES-SCLC. Serplulimab, as the first PD-1 inhibitor to achieve success in the first-line treatment of ES-SCLC, has not only demonstrated significant improvements in patient survival outcomes but also ushered in a new era for PD-1 inhibitors in the treatment of ES-SCLC. Recently, antiangiogenic agents with chemo-immunotherapy have achieved breakthroughs in first-line ES-SCLC treatment. Improving the clinical benefits of individualized treatment for patients with ES-SCLC remains challenging. Challenges include identifying biomarkers for targeted therapy, exploring new treatments, developing new medicines, and classifying SCLC molecular subtypes. This review provides an in-depth analysis of research on first-line ES-SCLC treatment. Additionally, it discusses advances in ES-SCLC treatment.
Collapse
Affiliation(s)
- Ran Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Peizhu Wu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Xiaoyan Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Lulu Zhuang
- Cheeloo College of Cancer Center, Shandong University, Jinan, Shandong, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China.
| |
Collapse
|
32
|
Kalfakakou D, Cameron DC, Kawaler EA, Tsuda M, Wang L, Jing X, Hajdu C, Tamayo DL, Shim Y, Ackermann A, Weissinger D, Zimny H, Hernandez R, Beier M, Dimartino D, Meyn P, Rice K, Selvaraj S, Loomis C, Heguy A, Lund AW, Sears RC, Welling TH, Dolgalev I, Tsirigos A, Simeone DM. Clonal Heterogeneity in Human Pancreatic Ductal Adenocarcinoma and Its Impact on Tumor Progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637729. [PMID: 39990481 PMCID: PMC11844494 DOI: 10.1101/2025.02.11.637729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer characterized by profound desmoplasia and cellular heterogeneity, which cannot be fully resolved using traditional bulk sequencing approaches. To understand the contribution of this heterogeneity to PDAC biology, we analyzed a large cohort of primary human PDAC samples (n = 62), profiling 443,451 single cells and 53,236 spatial transcriptomic spots using a combined single-cell RNA sequencing and spatial transcriptomics approach. Our analysis revealed significant intratumoral heterogeneity, with multiple genetically distinct neoplastic clones co-existing within individual tumors. These clones exhibited diverse transcriptional states and subtype profiles, challenging the traditional binary classification of PDAC into basal and classical subtypes; instead, our findings support a transcriptional continuum influenced by clonal evolution and spatial organization. Additionally, these clones each interacted uniquely with surrounding cell types in the tumor microenvironment. Phylogenetic analysis uncovered a rare but consistent classical-to-basal clonal transition associated with MYC amplification and immune response depletion, which were validated experimentally, suggesting a mechanism driving the emergence of a more aggressive basal clonal phenotype. Spatial analyses further revealed dispersed clones enriched for epithelial-to-mesenchymal transition (EMT) activity and immune suppression, correlating with metastatic potential and colonization of lymph node niches. These dispersed clones tended to transition toward a basal phenotype, contributing to disease progression. Our findings highlight the critical role of clonal diversity, transcriptional plasticity, and TME interactions in shaping human PDAC biology. This work provides new insights into the molecular and spatial heterogeneity of PDAC and offers potential avenues for therapeutic intervention targeting clonal evolution and the mechanisms driving metastasis.
Collapse
|
33
|
Zhang M, Yang Y, Liu J, Guo L, Guo Q, Liu W. Bone marrow immune cells and drug resistance in acute myeloid leukemia. Exp Biol Med (Maywood) 2025; 250:10235. [PMID: 40008144 PMCID: PMC11851207 DOI: 10.3389/ebm.2025.10235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
In recent years, the relationship between the immunosuppressive niche of the bone marrow and therapy resistance in acute myeloid leukemia (AML) has become a research focus. The abnormal number and function of immunosuppressive cells, including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), along with the dysfunction and exhaustion of immunological effector cells, including cytotoxic T lymphocytes (CTLs), dendritic cells (DCs) and natural killer cells (NKs), can induce immune escape of leukemia cells and are closely linked to therapy resistance in leukemia. This article reviews the research progress on the relationship between immune cells in the marrow microenvironment and chemoresistance in AML, aiming to provide new ideas for the immunotherapy of AML.
Collapse
Affiliation(s)
- Miao Zhang
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - You Yang
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jing Liu
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ling Guo
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qulian Guo
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wenjun Liu
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
34
|
Dehghan F, Metanat Y, Askarizadeh M, Ahmadi E, Moradi V. Novel gene manipulation approaches to unlock the existing bottlenecks of CAR-NK cell therapy. Front Cell Dev Biol 2025; 12:1511931. [PMID: 40007761 PMCID: PMC11850336 DOI: 10.3389/fcell.2024.1511931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/10/2024] [Indexed: 02/27/2025] Open
Abstract
Currently, CAR-T cell therapy is known as an efficacious treatment for patients with relapsed/refractory hematologic malignancies. Nonetheless, this method faces several bottlenecks, including low efficacy for solid tumors, lethal adverse effects, high cost of autologous products, and the risk of GvHD in allogeneic settings. As a potential alternative, CAR-NK cell therapy can overcome most of the limitations of CAR-T cell therapy and provide an off-the-shelf, safer, and more affordable product. Although published results from preclinical and clinical studies with CAR-NK cells are promising, several bottlenecks must be unlocked to maximize the effectiveness of CAR-NK cell therapy. These bottlenecks include low in vivo persistence, low trafficking into tumor sites, modest efficacy in solid tumors, and sensitivity to immunosuppressive tumor microenvironment. In recent years, advances in gene manipulation tools and strategies have laid the groundwork to overcome the current bottlenecks of CAR-NK cell therapy. This review will introduce the existing gene manipulation tools and discuss their advantages and disadvantages. We will also explore how these tools can enhance CAR-NK cell therapy's safety and efficacy.
Collapse
Affiliation(s)
- Fatemeh Dehghan
- Department of Anatomy and Molecular Biology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Yekta Metanat
- Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Sistan and Baluchestan Province, Iran
| | - Mandana Askarizadeh
- School of Biology and Ecology, University of Maine, Orono, ME, United States
| | - Ehsan Ahmadi
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Moradi
- Department of Hematology and Blood Transfusion Sciences, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Luo X, Li C, Qin G. Multiple machine learning-based integrations of multi-omics data to identify molecular subtypes and construct a prognostic model for HNSCC. Hereditas 2025; 162:17. [PMID: 39910672 PMCID: PMC11800565 DOI: 10.1186/s41065-025-00380-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/27/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Immunotherapy has introduced new breakthroughs in improving the survival of head and neck squamous cell carcinoma (HNSCC) patients, yet drug resistance remains a critical challenge. Developing personalized treatment strategies based on the molecular heterogeneity of HNSCC is essential to enhance therapeutic efficacy and prognosis. METHODS We integrated four HNSCC datasets (TCGA-HNSCC, GSE27020, GSE41613, and GSE65858) from TCGA and GEO databases. Using 10 multi-omics consensus clustering algorithms via the MOVICS package, we identified two molecular subtypes (CS1 and CS2) and validated their stability. A machine learning-driven prognostic signature was constructed by combining 101 algorithms, ultimately selecting 30 prognosis-related genes (PRGs) with the Elastic Net model. This signature was further linked to immune infiltration, functional pathways, and therapeutic sensitivity. RESULTS CS1 exhibited superior survival outcomes in both TCGA and META-HNSCC cohorts. The PRG-based signature stratified patients into low- and high-risk groups, with the low-risk group showing prolonged survival, enhanced immune cell infiltration (B cells, T cells, monocytes), and activated immune functions (cytolytic activity, T cell co-stimulation). High-risk patients were more sensitive to radiotherapy and chemotherapy (e.g., Cisplatin, 5-Fluorouracil), while low-risk patients responded better to immunotherapy and targeted therapies. CONCLUSION Our study delineates two molecular subtypes of HNSCC and establishes a robust prognostic model using multi-omics data and machine learning. These findings provide a framework for personalized treatment selection, offering clinical insights to optimize therapeutic strategies for HNSCC patients.
Collapse
Affiliation(s)
- Xiaoqin Luo
- Department of Otolaryngology, University of Electronic Science and Technology of China, Chengdu, 611731, China
- Department of Otolaryngology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Otolaryngology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
- Head and Neck Surgery Department, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Chao Li
- Department of Otolaryngology, University of Electronic Science and Technology of China, Chengdu, 611731, China.
- Department of Otolaryngology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
- Head and Neck Surgery Department, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| | - Gang Qin
- Department of Otolaryngology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
36
|
Guo P, Zhong L, Wang T, Luo W, Zhou A, Cao D. NK cell-based immunotherapy for hepatocellular carcinoma: Challenges and opportunities. Scand J Immunol 2025; 101:e13433. [PMID: 39934640 DOI: 10.1111/sji.13433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/22/2024] [Accepted: 01/01/2025] [Indexed: 02/13/2025]
Abstract
Hepatocellular carcinoma (HCC) remains one of the most challenging malignancies globally, characterized by significant heterogeneity, late-stage diagnosis, and resistance to treatment. In recent years, the advent of immune-checkpoint blockades (ICBs) and targeted immune cell therapies has marked a substantial advancement in HCC treatment. However, the clinical efficacy of these existing therapies is still limited, highlighting the urgent need for new breakthroughs. Natural killer (NK) cells, a subset of the innate lymphoid cell family, have shown unique advantages in the anti-tumour response. With increasing evidence suggesting the crucial role of dysfunctional NK cells in the pathogenesis and progression of HCC, considerable efforts have been directed toward exploring NK cells as a potential therapeutic target for HCC. In this review, we will provide an overview of the role of NK cells in normal liver immunity and in HCC, followed by a detailed discussion of various NK cell-based immunotherapies and their potential applications in HCC treatment.
Collapse
Affiliation(s)
- Pei Guo
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Liyuan Zhong
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Tao Wang
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weijia Luo
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Aiqiang Zhou
- Guangzhou Hospital of Integrated Chinese and Western Medicine, Guangzhou, Guangdong, P.R China
| | - Deliang Cao
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
37
|
Struckmeier AK, Gosau M, Smeets R. Immunotherapeutic strategies beyond the PD-1/PD-L1 pathway in head and neck squamous cell carcinoma - A scoping review on current developments in agents targeting TIM-3, TIGIT, LAG-3, and VISTA. Oral Oncol 2025; 161:107145. [PMID: 39705929 DOI: 10.1016/j.oraloncology.2024.107145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/24/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) poses a considerable challenge due to its high incidence and mortality rates. Immunotherapy targeting PD-(L)1 emerges as a promising approach for HNSCC, as it has the potential to trigger a broad and long-lasting anti-tumor response. Nevertheless, the effectiveness of immunotherapy encounters hurdles, and only a small proportion of patients benefit, with many eventually experiencing relapse. Consequently, there is a pursuit of strategies to enhance overall treatment outcomes. Understanding the mechanisms driving resistance to PD-(L)1 inhibition and devising strategies to overcome these challenges are vital for advancing more effective treatments. Furthermore, gaining insights into the mechanisms of action and safety profiles of novel combination therapies is critical for their successful adoption in clinical practice. As a result, current research is dedicated to investigating various immunotherapeutic agents beyond the PD-1/PD-L1 axis. This review offers a comprehensive overview of the existing immunotherapy strategies in HNSCC with a focus on TIM-3, TIGIT, LAG-3, and VISTA. The aim is to lay a strong foundation for the continual advancement of therapies for HNSCC.
Collapse
Affiliation(s)
- Ann-Kristin Struckmeier
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany.
| | - Martin Gosau
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Ralf Smeets
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany; Department of Oral and Maxillofacial Surgery, Division of Regenerative Orofacial Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
38
|
Das L, Das S. A comprehensive insights of cancer immunotherapy resistance. Med Oncol 2025; 42:57. [PMID: 39883235 DOI: 10.1007/s12032-025-02605-8] [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: 12/07/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025]
Abstract
Cancer is a major global health issue that is usually treated with multiple therapies, such as chemotherapy and targeted therapies like immunotherapy. Immunotherapy is a new and alternative approach to treating various types of cancer that are difficult to treat with other methods. Although immune checkpoint inhibitors have shown promise for long-term efficacy, they have limited effectiveness in common cancer types such as breast, prostate, and lung. Some patients do not respond to immunotherapy, while others develop resistance to the treatment over time, which is classified as primary or acquired resistance. Cancer immunotherapy, specifically immune checkpoint inhibitor-based resistance involves multiple factors such as genes, metabolism, inflammation, and angiogenesis. However, cutting-edge research has identified the mechanisms of immunotherapy resistance and possible solutions. Current research may improve biomarker identification and modify treatment strategies, which will lead to better clinical outcomes. This review provides a comprehensive discussion of the current mechanisms of immunotherapy resistance, related biomarker modulation, and strategies to overcome resistance.
Collapse
Affiliation(s)
- Laavanya Das
- Department of Food and Nutrition, Brainware University, 398, Ramkrishnapur Rd, Barasat, Kolkata, West Bengal, 700125, India
| | - Subhadip Das
- Department of In Vivo Pharmacology, TCG Lifesciences Pvt. Ltd, BN 7, Sector V, Salt Lake City, Kolkata, West Bengal, 700091, India.
| |
Collapse
|
39
|
Godfroid C, Romero J, Labiano S, Chuang CH, Kelemen A, Wyss T, Roh V, Verdeil G, Klein C, Codarri Deak L, Umaña P, Tolstonog GV, Trumpfheller C, Vozenin MC, Romero PJ. PD-1 cis-targeted IL-2v in combination with radiotherapy inhibits lung cancer growth and remodels the immune microenvironment. J Immunother Cancer 2025; 13:e009832. [PMID: 39848686 PMCID: PMC11759878 DOI: 10.1136/jitc-2024-009832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 12/27/2024] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND More efficient therapeutic options for non-small cell lung cancer (NSCLC) are needed as the survival at 5 years of metastatic disease is near zero. In this regard, we used a preclinical model of metastatic lung adenocarcinoma (SV2-OVA) to assess the safety and efficacy of novel radio-immunotherapy combining hypofractionated radiotherapy (HRT) with muPD1-IL2v immunocytokine and muFAP-CD40 bispecific antibody. METHODS We evaluated the changes in the lung immune microenvironment at multiple timepoints following combination therapies and investigated their underlying antitumor mechanisms. Additionally, we analyzed the tumor clonal heterogeneity upon the combination treatments to explore potential mechanisms associated with the lack of complete response. RESULTS The combination of HRT with muPD1-IL2v had a potent antitumor effect and increased survival in the SV2-OVA lung cancer model. Importantly, this combination therapy was devoid of measurable toxicity. It induced remodeling of the immune contexture through the increase of CD8+ T and natural killer (NK) cells. The addition of muFAP-CD40 to the combination treatment further increased infiltrating CD8+ T cells, expressing high levels of effector molecules, both in the periphery and core tumor regions. An accumulation of CD8+ PD-1+ TOX+ (exhausted) T cells, already at the 'early' timepoint, is consistent with the limited clinical benefits provided by the various combination treatments in this model. The study of the clonal dynamics of tumor cells during disease progression and therapy highlighted a clonal selection upon HRT+muPD1-IL2v therapy. CONCLUSIONS We demonstrated that HRT+muPD1-IL2v combination is a potent therapeutic strategy to delay tumor growth and increase survival in a metastatic lung cancer model, but additional studies are required to completely understand the resistance mechanisms associated with the lack of complete response in this model.
Collapse
Affiliation(s)
- Céline Godfroid
- Department of Oncology, University of Lausanne, Epalinges, Switzerland
- Department of Radiation Oncology, CHUV, Lausanne, Switzerland
| | | | - Sara Labiano
- Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Chia-Hsien Chuang
- Department of Otolaryngology-Head and Neck Surgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- AGORA, Lausanne, Switzerland
| | - Andrea Kelemen
- Department of Otolaryngology-Head and Neck Surgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- AGORA, Lausanne, Switzerland
| | - Tania Wyss
- Department of Oncology, University of Lausanne, Epalinges, Switzerland
| | | | - Grégory Verdeil
- Department of Oncology, University of Lausanne, Epalinges, Switzerland
| | | | | | | | - Genrich V Tolstonog
- Department of Otolaryngology-Head and Neck Surgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- AGORA, Lausanne, Switzerland
| | | | - Marie-Catherine Vozenin
- Sector of Radiobiology Applied to Radiotherapy, Radiation Oncology Service, Geneva University Hospital, Geneva, Switzerland
| | - Pedro J Romero
- Route de la Corniche 3B, Novigenix SA, 1066, Epalinges, Switzerland
| |
Collapse
|
40
|
Wlosik J, Orlanducci F, Richaud M, Demerle C, Amara AB, Rouviere MS, Livrati P, Gorvel L, Hospital MA, Dulphy N, Devillier R, Vey N, Olive D, Chretien AS. CD56 neg CD16 + cells represent a distinct mature NK cell subset with altered phenotype and are associated with adverse clinical outcome upon expansion in AML. Front Immunol 2025; 15:1487792. [PMID: 39867888 PMCID: PMC11760599 DOI: 10.3389/fimmu.2024.1487792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/02/2024] [Indexed: 01/28/2025] Open
Abstract
Introduction Acute myeloid leukemia (AML) is a rare haematological cancer with poor 5-years overall survival (OS) and high relapse rate. Leukemic cells are sensitive to Natural Killer (NK) cell mediated killing. However, NK cells are highly impaired in AML, which promote AML immune escape from NK cell immune surveillance. We made the first report of CD56neg CD16+ NK cells expansion in AML. This unconventional subset has been reported to expand in some chronic viral infections. Although it is unclear whether CD56neg NK cells expansion mechanism is common across diseases, it seems more relevant than ever to further investigate this subset, representing a potential therapeutic target. Methods We used PBMCs from AML patients and HV to perform mass cytometry, spectral flow cytometry, bulk RNA-seq and in vitro assays in order to better characterize CD56neg CD16+ NK cells that expand in AML. Results We confirmed that CD56neg CD16+ NK cells represent a unique NK cell subset coexpressing Eomes and T-bet. CD56neg CD16+ NK cells could recover CD56 expression in vitro where they displayed unaltered NK cell functions. We previously demonstrated that CD56neg CD16+ NK cells expansion at diagnosis was associated with adverse clinical outcome in AML. Here, we validated our findings in a validation cohort of N=38 AML patients. AML patients with CD56neg CD16+ NK cells expansion at diagnosis had decreased overall survival (HR[CI95]=5.5[1.2-24.5], p=0.0251) and relapse-free survival (HR[CI95]=13.1[1.9-87.5], p=0.0079) compared to AML patients without expansion after 36 months follow-up. RNA-seq unveiled that CD56neg CD16+ NK cells were mature circulating NK cells with functional capacities. Upon expansion, CD56neg CD16+ NK cells from AML patients showed altered proteomic phenotype, with increased frequency of terminally mature CD56neg CD16+ NK cells expressing TIGIT along with decreased frequency of Siglec-7+ CD56neg CD16+ NK cells. Discussion Taken together, our results suggest that we could harness CD56neg CD16+ NK cells cytotoxic potential in vitro to restore NK cell anti-tumor response in AML patients with CD56neg CD16+ NK cells expansion and improve patients' prognosis. To conclude, CD56neg CD16+ NK cells represent a relevant target for future NK-cell-based immunotherapies in AML.
Collapse
Affiliation(s)
- Julia Wlosik
- Team Immunity and Cancer, Cancer Research Center of Marseille (CRCM), Inserm U1068, CNRS UMR7258, Paoli-Calmettes Institute, University of Aix-Marseille UM105, Marseille, France
- Immunomonitoring Department, Paoli-Calmettes Institute, Marseille, France
| | - Florence Orlanducci
- Team Immunity and Cancer, Cancer Research Center of Marseille (CRCM), Inserm U1068, CNRS UMR7258, Paoli-Calmettes Institute, University of Aix-Marseille UM105, Marseille, France
- Immunomonitoring Department, Paoli-Calmettes Institute, Marseille, France
| | - Manon Richaud
- Cytometry Platform, Cancer Research Center of Marseille (CRCM), Inserm U1068, CNRS UMR7258, Paoli-Calmettes Institute, University of Aix-Marseille UM105, Marseille, France
| | - Clemence Demerle
- Centre for Clinical Investigation in Biotherapy, Paoli-Calmettes Institute, University of Aix-Marseille, Inserm CBT 1409, Marseille, France
| | - Amira Ben Amara
- Team Immunity and Cancer, Cancer Research Center of Marseille (CRCM), Inserm U1068, CNRS UMR7258, Paoli-Calmettes Institute, University of Aix-Marseille UM105, Marseille, France
- Immunomonitoring Department, Paoli-Calmettes Institute, Marseille, France
| | - Marie-Sarah Rouviere
- Team Immunity and Cancer, Cancer Research Center of Marseille (CRCM), Inserm U1068, CNRS UMR7258, Paoli-Calmettes Institute, University of Aix-Marseille UM105, Marseille, France
- Immunomonitoring Department, Paoli-Calmettes Institute, Marseille, France
| | - Philippe Livrati
- Team Immunity and Cancer, Cancer Research Center of Marseille (CRCM), Inserm U1068, CNRS UMR7258, Paoli-Calmettes Institute, University of Aix-Marseille UM105, Marseille, France
- Immunomonitoring Department, Paoli-Calmettes Institute, Marseille, France
| | - Laurent Gorvel
- Team Immunity and Cancer, Cancer Research Center of Marseille (CRCM), Inserm U1068, CNRS UMR7258, Paoli-Calmettes Institute, University of Aix-Marseille UM105, Marseille, France
- Immunomonitoring Department, Paoli-Calmettes Institute, Marseille, France
| | - Marie-Anne Hospital
- Hematology Department, CRCM, Inserm U1068, CNRS UMR7258, Paoli-Calmettes Institute, Marseille, France
| | - Nicolas Dulphy
- Paris Cité University, Saint-Louis Research Institute, Inserm UMRS1160, Paris, France
- Immunology and Histocompatibility Laboratory, Assistance Publique-Hôpitaux de Paris, Saint-Louis Hospital, Paris, France
| | - Raynier Devillier
- Hematology Department, CRCM, Inserm U1068, CNRS UMR7258, Paoli-Calmettes Institute, Marseille, France
| | - Norbert Vey
- Hematology Department, CRCM, Inserm U1068, CNRS UMR7258, Paoli-Calmettes Institute, Marseille, France
| | - Daniel Olive
- Team Immunity and Cancer, Cancer Research Center of Marseille (CRCM), Inserm U1068, CNRS UMR7258, Paoli-Calmettes Institute, University of Aix-Marseille UM105, Marseille, France
- Immunomonitoring Department, Paoli-Calmettes Institute, Marseille, France
| | - Anne-Sophie Chretien
- Team Immunity and Cancer, Cancer Research Center of Marseille (CRCM), Inserm U1068, CNRS UMR7258, Paoli-Calmettes Institute, University of Aix-Marseille UM105, Marseille, France
- Immunomonitoring Department, Paoli-Calmettes Institute, Marseille, France
| |
Collapse
|
41
|
Zhang Y, Li Y, Zhang Z, Zheng X, Peng H, Tian Z, Sun R, Sun H. CD49a Targeting Enhances NK Cell Function and Antitumor Immunity. Cancer Immunol Res 2025; 13:139-151. [PMID: 39570767 DOI: 10.1158/2326-6066.cir-24-0124] [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: 02/04/2024] [Revised: 06/04/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025]
Abstract
Approximately 70% of patients receiving immune checkpoint blockade therapies develop treatment resistance. Thus, there is a need for the identification of additional immunotherapeutic targets. CD49a is a membrane protein expressed on NK cells and T cells. In this study, we found that CD49a was highly expressed on the surface of tumor-infiltrating NK cells in various mouse tumor models and that CD49a+ tumor-infiltrating NK cells were more exhausted than CD49a- tumor-infiltrating NK cells. Furthermore, CD49a or NK-specific CD49a deficiency slowed tumor growth and prolonged survival in several mouse tumor models, primarily through the essential role played by NK cells in antitumor activities. Blockade of CD49a using an mAb suppressed tumor development in mice, and combination treatment with anti-PD-L1 further enhanced antitumor efficacy. Our research reveals CD49a on NK cells as an immunotherapeutic target and highlights the potential clinical applications of CD49a-targeted therapies.
Collapse
Affiliation(s)
- Yu Zhang
- Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, The Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
| | - Yangyang Li
- Hefei TG ImmunoPharma Corporation Limited, Hefei, China
| | - Zhengfeng Zhang
- Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, The Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
| | - Xiaodong Zheng
- Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, The Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
| | - Hui Peng
- Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, The Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
- Division of Life Sciences and Medicine, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China, Hefei, China
- Research Unit of NK Cell Study, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhigang Tian
- Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, The Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
- Research Unit of NK Cell Study, Chinese Academy of Medical Sciences, Beijing, China
| | - Rui Sun
- Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, The Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
- Division of Life Sciences and Medicine, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China, Hefei, China
| | - Haoyu Sun
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Dai Y, Dong C, Wang Z, Zhou Y, Wang Y, Hao Y, Chen P, Liang C, Li G. Infiltrating T lymphocytes and tumor microenvironment within cholangiocarcinoma: immune heterogeneity, intercellular communication, immune checkpoints. Front Immunol 2025; 15:1482291. [PMID: 39845973 PMCID: PMC11750830 DOI: 10.3389/fimmu.2024.1482291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Cholangiocarcinoma is the second most common primary liver cancer, and its global incidence has increased in recent years. Radical surgical resection and systemic chemotherapy have traditionally been the standard treatment options. However, the complexity of cholangiocarcinoma subtypes often presents a challenge for early diagnosis. Additionally, high recurrence rates following radical treatment and resistance to late-stage chemotherapy limit the benefits for patients. Immunotherapy has emerged as an effective strategy for treating various types of cancer, and has shown efficacy when combined with chemotherapy for cholangiocarcinoma. Current immunotherapies targeting cholangiocarcinoma have predominantly focused on T lymphocytes within the tumor microenvironment, and new immunotherapies have yielded unsatisfactory results in clinical trials. Therefore, it is essential to achieve a comprehensive understanding of the unique tumor microenvironment of cholangiocarcinoma and the pivotal role of T lymphocytes within it. In this review, we describe the heterogeneous immune landscape and intercellular communication in cholangiocarcinoma and summarize the specific distribution of T lymphocytes. Finally, we review potential immune checkpoints in cholangiocarcinoma.
Collapse
Affiliation(s)
- Yunyan Dai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Chenyang Dong
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Zhiming Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yunpeng Zhou
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yi Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yi Hao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Pinggui Chen
- Department of Nuclear Medicine, Nanyang First People’s Hospital, Nanyang, Henan, China
| | - Chaojie Liang
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
- Department of biliary and Pancreatic Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Gaopeng Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
43
|
Meggyes M, Nagy DU, Mezosi L, Polgar B, Szereday L. CD8+ and CD8- NK Cells and Immune Checkpoint Networks in Peripheral Blood During Healthy Pregnancy. Int J Mol Sci 2025; 26:428. [PMID: 39796279 PMCID: PMC11720283 DOI: 10.3390/ijms26010428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Pregnancy involves significant immunological changes to support fetal development while protecting the mother from infections. A growing body of evidence supports the importance of immune checkpoint pathways, especially at the maternal-fetal interface, although limited information is available about the peripheral expression of these molecules by CD8+ and CD8- NK cell subsets during the trimesters of pregnancy. Understanding the dynamics of these immune cells and their checkpoint pathways is crucial for elucidating their roles in pregnancy maintenance and potential complications. This study aims to investigate the peripheral expression and functional characteristics of CD8+ and CD8- NK cell subsets throughout pregnancy, providing insights into their contributions to maternal and fetal health. A total of 34 healthy women were enrolled from the first, 30 from the second and 40 from the third trimester of pregnancy. At the same time, 35 healthy age-matched non-pregnant women formed the control group. From peripheral blood, mononuclear cells were separated and stored at -80 °C. CD8+ and CD8- NK cell subsets were analyzed from freshly thawed samples, and surface and intracellular staining was performed using flow cytometric analyses. The proportions of CD56+ NK cells in peripheral blood were similar across groups. While CD8- NKdim cells increased significantly in all trimesters compared to non-pregnant controls, CD8+ NKdim cells showed no significant changes. CD8- NKbright cells had higher frequencies throughout pregnancy, whereas CD8+ NKbright cells significantly increased only in the first and second trimesters. The expression levels of immune checkpoint molecules, such as PD-1 and PD-L1, and cytotoxic-activity-related molecules were stable, with notable perforin and granzyme B increases in CD8- NKbright cells throughout pregnancy. Our study shows that peripheral NK cell populations, especially CD8- subsets, are predominant during pregnancy. This shift suggests a crucial role for CD8- NK cells in balancing maternal immune tolerance and surveillance. The stable expression of immune checkpoint molecules indicates that other regulatory mechanisms may be at work. These findings enhance our understanding of peripheral immune dynamics in pregnancy and suggest that targeting CD8- NKbright cell functions could help manage pregnancy-related immune complications. This research elucidates the stable distribution and functional characteristics of peripheral NK cells during pregnancy, with CD8- subsets being more prevalent. The increased activity of CD8- NKbright cells suggests their critical role in maintaining immune surveillance. Our findings provide a basis for future studies to uncover the mechanisms regulating NK cell function in pregnancy, potentially leading to new treatments for immune-related pregnancy complications.
Collapse
Affiliation(s)
- Matyas Meggyes
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 12 Szigeti Street, 7624 Pecs, Hungary
- Janos Szentagothai Research Centre, 20 Ifjusag Street, 7624 Pecs, Hungary
| | - David U. Nagy
- Institute of Geobotany/Plant Ecology, Martin-Luther-University, Große Steinstraße 79/80, D-06108 Halle, Germany
| | - Livia Mezosi
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 12 Szigeti Street, 7624 Pecs, Hungary
| | - Beata Polgar
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 12 Szigeti Street, 7624 Pecs, Hungary
| | - Laszlo Szereday
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 12 Szigeti Street, 7624 Pecs, Hungary
- Janos Szentagothai Research Centre, 20 Ifjusag Street, 7624 Pecs, Hungary
| |
Collapse
|
44
|
Zhou H, Bao G, Zhao J, Zhu X. Nuclear Molecular Imaging for Evaluating T Cell Exhaustion. Mol Pharm 2025; 22:103-112. [PMID: 39586059 DOI: 10.1021/acs.molpharmaceut.4c00970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
T cells are indispensable for the therapeutic efficacy of cancer immunotherapies, including immune checkpoint blockade. However, prolonged antigen exposure also drives T cells into exhaustion, which is characterized by upregulated inhibitory molecules, impaired effector functions, reduced cytotoxicity, altered metabolism, etc. Noninvasive monitoring of T cell exhaustion allows a timely identification of cancer patients that are most likely to benefit from immunotherapies. In this Review, we briefly explain the biological cascades underlying the modulation of inhibitory molecules, present a concise update on the nuclear molecular imaging tracers of T cell exhaustion, and then discuss the potential opportunities for future development.
Collapse
Affiliation(s)
- Huimin Zhou
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Guangfa Bao
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Department of Nuclear Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, China
| | - Jun Zhao
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Department of Anatomy, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiaohua Zhu
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| |
Collapse
|
45
|
Hosseinalizadeh H, Wang LS, Mirzaei H, Amoozgar Z, Tian L, Yu J. Emerging combined CAR-NK cell therapies in cancer treatment: Finding a dancing partner. Mol Ther 2025:S1525-0016(24)00895-5. [PMID: 39754357 DOI: 10.1016/j.ymthe.2024.12.057] [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: 03/21/2024] [Revised: 09/21/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025] Open
Abstract
In recent decades, immunotherapy with chimeric antigen receptors (CARs) has revolutionized cancer treatment and given hope where other cancer therapies have failed. CAR-natural killer (NK) cells are NK cells that have been engineered ex vivo with a CAR on the cell membrane with high specificity for specific target antigens of tumor cells. The impressive results of several studies suggest that CAR-NK cell therapy has significant potential and successful performance in cancer treatment. Despite its effectiveness, CAR-NK cell therapy can have significant challenges when it comes to treating cancer. These challenges include tumor heterogeneity, antigen escape, an immunosuppressive tumor microenvironment, limited tissue migration from blood, exhaustion of CAR-NK cells, and inhibition by immunosuppressive checkpoint molecule signaling, etc. In CAR-T cell therapy, the use of combined approaches has shown encouraging outcomes for tumor regression and improved cancer treatment compared to single therapies. Therefore, to overcome these significant challenges in CAR-NK cells, innovative combination therapies of CAR-NK cells with other conventional therapies (e.g., chemotherapy and radiotherapy) or other immunotherapies are needed to counteract the above challenges and thereby increase the activity of CAR-NK cells. This review comprehensively discusses various cancer-treatment approaches in combination with CAR-NK cell therapy in the hope of providing valuable insights that may improve cancer treatment in the near future.
Collapse
Affiliation(s)
- Hamed Hosseinalizadeh
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Li-Shu Wang
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zohreh Amoozgar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lei Tian
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| | - Jianhua Yu
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
46
|
Sun J, Tian Y, Yang C. Target therapy of TIGIT; a novel approach of immunotherapy for the treatment of colorectal cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:231-241. [PMID: 39158733 DOI: 10.1007/s00210-024-03346-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024]
Abstract
The T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT), a newly discovered checkpoint, is characterized by its elevated expression on CD4 + T cells, CD8 + T cells, natural killer (NK) cells, regulatory T cells (Tregs), and tumor-infiltrating lymphocytes (TILs). Research to date has been shown that TIGIT has been linked to exhaustion of NK cell both and T cells in numerous cancers. CD155, being the specific ligand of TIGIT in humans, emerges as a key target for immunotherapy owing to its crucial interaction with TIGIT. Furthermore, numerous studies have demonstrated that the combination of TIGIT with other immune checkpoint inhibitors (ICIs) and/or traditional treatments elicits a potent antitumor response in colorectal cancer (CRC). This review provides an overview of the structure, function, and signaling pathways associated with TIGIT across multiple immune system cell types. Additionally, focusing on the role of TIGIT in the progression of CRC, this study reviewed various studies exploring TIGIT-based immunotherapy in CRC.
Collapse
Affiliation(s)
- Jing Sun
- Department of Gastroenterology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China
| | - Yan Tian
- Department of Gastroenterology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China
| | - Changqing Yang
- Department of Gastroenterology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China.
| |
Collapse
|
47
|
Hua W, Liu J, Li Y, Yin H, Shen HR, Wu JZ, Kong YL, Pan BH, Liang JH, Wang L, Li JY, Gao R, Liang JH, Xu W. Revealing the heterogeneity of treatment resistance in less-defined subtype diffuse large B cell lymphoma patients by integrating programmed cell death patterns and liquid biopsy. Clin Transl Med 2025; 15:e70150. [PMID: 39731274 DOI: 10.1002/ctm2.70150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 12/29/2024] Open
Abstract
Precision medicine in less-defined subtype diffuse large B-cell lymphoma (DLBCL) remains a challenge due to the heterogeneous nature of the disease. Programmed cell death (PCD) pathways are crucial in the advancement of lymphoma and serve as significant prognostic markers for individuals afflicted with lymphoid cancers. To identify robust prognostic biomarkers that can guide personalized management for less-defined subtype DLBCL patients, we integrated multi-omics data derived from 339 standard R-CHOP-treated patients diagnosed with less-defined subtype DLBCL from three independent cohorts. By employing various machine learning algorithms, we pinpointed eight pivotal genes linked to PCD, specifically FLT3, SORL1, CD8A, BCL2L1, COL13A1, MPG, DYRK2 and CAMK2B. Following this, we established a Programmed Cell Death Index (PCDI) utilizing the aforementioned genes and amalgamated it with pertinent clinical characteristics to formulate a predictive nomogram model for prognosis. We observed a significant correlation between the PCDI, pre-treatment circulating tumour DNA (ctDNA) burden, minimal residual disease (MRD) status and immune features. Furthermore, our research indicated that patients with elevated PCDI scores could potentially show resistance to conventional chemotherapy treatments, yet they might derive an advantage from alternative inhibitors targeting specific signalling pathways. Conclusively, leveraging these results, we have created an online analytical tool (https://xulymphoma.shinyapps.io/PCDI_pred/) designed for the prognostic prediction of patients with less-defined subtype DLBCL. This tool facilitates the forecasting of outcomes for these patients, enhancing the precision of their clinical management. KEY POINTS: Developing the Programmed Cell Death Index (PCDI) utilizing multiple machine learning algorithms for patients with less-defined subtype diffuse large B-cell lymphoma. The difference in clinical characteristics, circulating tumour DNA burden and immune profiling between patients with distinct PCDI groups. A potentially effective regimen was speculated for patients with high PCDI scores who tend to exhibit worse progression-free survival.
Collapse
Affiliation(s)
- Wei Hua
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Jie Liu
- Department of Hematology, The Third Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Hua Yin
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Hao-Rui Shen
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Jia-Zhu Wu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Yi-Lin Kong
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Bi-Hui Pan
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Jun-Heng Liang
- Department of Medical Affairs, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Jian-Yong Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Rui Gao
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Jin-Hua Liang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Wei Xu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| |
Collapse
|
48
|
Tao JH, Zhang J, Li HS, Zhou Y, Guan CX. Nature killer cell for solid tumors: Current obstacles and prospective remedies in NK cell therapy and beyond. Crit Rev Oncol Hematol 2025; 205:104553. [PMID: 39515404 DOI: 10.1016/j.critrevonc.2024.104553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
In recent years, cell therapy has emerged as an innovative treatment method for the management of clinical tumors following immunotherapy. Among them, Natural killer (NK) cell therapy has achieved a significant breakthrough in the treatment of hematological tumors. However, the therapeutic effectiveness of NK cells in the treatment of solid tumors remains challenging. With the progress of gene editing and culture techniques and their application to NK cell engineering, it is expected that NK cell therapy will revolutionize the treatment of solid tumors. In this review, we explore the discovery and biological properties of NK cells, their role in the tumor microenvironment, and the therapeutic strategies, clinical trials, challenges, and prospects of NK cells in the treatment of solid tumors.
Collapse
Affiliation(s)
- Jia-Hao Tao
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Jun Zhang
- Ascle Therapeutics, Suzhou, Jiangsu 215000, China
| | - Hua-Shun Li
- Ascle Therapeutics, Suzhou, Jiangsu 215000, China.
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China.
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
49
|
Luo J, Lin M, Chen M, Chen J, Zhou X, Liu K, Liang Y, Chen J, Liang H, Wang Z, Deng Q, Wang J, Jin M, Luo J, Chen W, Cen J. Machine learning-derived natural killer cell signature predicts prognosis and therapeutic response in clear cell renal cell carcinoma. Transl Oncol 2025; 51:102180. [PMID: 39536695 PMCID: PMC11600779 DOI: 10.1016/j.tranon.2024.102180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/11/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Natural killer cells, interconnected with patient prognosis and treatment response, play a pivotal role in the tumor immune microenvironment and may serve as potential novel predictive biomarkers for renal cell carcinoma. METHODS Clear cell renal cell carcinoma transcriptome data and the corresponding clinical data were obtained from the Cancer Genome Atlas (TCGA) database. Single-cell sequencing data were sourced from the Gene Expression Omnibus (GEO) database. A risk model was established by integrating ten different machine learning algorithms, which resulted in 101 combined models. The model with the highest average C-index was selected for further analysis, and was assessed using nomogram, time-dependent receiver operating characteristics (ROC) and Kaplan-Meier survival analysis. The differences in immune infiltration fractions, clinicopathological features, and response to various targeted therapies and immunotherapy between high- and low-risk groups were investigated. Furthermore, qRT-PCR, IHC, colony formation test, CCK8 assay and flow cytometry were conducted to explore the expression pattern and function of ARHGAP9 in our own patient samples and renal cancer cell lines. RESULTS Totally, 156 NK cell-related genes and 5189 prognosis-related genes were identified, and 36 genes of their intersection demonstrated prognostic value. A risk model with 18 genes was established by Coxboost plus plsRcox, which can accurately predict the prognosis of ccRCC patients. Significant correlations were determined between risk score and tumor malignancy and immune cell infiltration. Meanwhile, a combination of tumor mutation burden plus risk score could have higher accuracy of predicting clinical outcomes. Moreover, high-risk group patients were more likely to be responsive to targeted therapy but show no response to immunotherapy. CONCLUSIONS Intricate signaling interactions between NK cells and various cellular subgroups were depicted and the developmental trajectory of NK cells was elucidated. A NK cells-related risk model was established, which can provide reliable prognostic information and identified patients with more probability of benefiting from therapy.
Collapse
Affiliation(s)
- Jinchen Luo
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China
| | - Mingjie Lin
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China
| | - Minyu Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China
| | - Jinwei Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China
| | - Xinwei Zhou
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China
| | - Kezhi Liu
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China
| | - Yanping Liang
- Department of Laboratory Medicine, The First Affiliated Hospital f Sun Yat-sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China
| | - Jiajie Chen
- Department of Pediatrics, The First Affiliated Hospital of Sun Yat-sen University, No.58, Zhongshan Road II, Guangzhou, 510080, China
| | - Hui Liang
- Department of Urology, Affiliated Longhua People's Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Zhu Wang
- Department of Urology, Affiliated Longhua People's Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Qiong Deng
- Department of Urology, Affiliated Longhua People's Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Jieyan Wang
- Department of Urology, Affiliated Longhua People's Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Meiyu Jin
- Department of Urology, Affiliated Longhua People's Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Junhang Luo
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China.
| | - Wei Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China.
| | - Junjie Cen
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Road II, Guangzhou, 510080, China.
| |
Collapse
|
50
|
Dumbrava EE, Ben Haj Frej K, Sharon E, Tawbi H. Application and Expectations for Immune Checkpoint Blockade of LAG3 and TIGIT. Annu Rev Med 2025; 76:189-205. [PMID: 39656959 DOI: 10.1146/annurev-med-080222-100847] [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] [Indexed: 12/17/2024]
Abstract
Immune checkpoint blockade targeting the novel targets of the lymphocyte activation gene 3 (LAG3) and the T cell immunoreceptor with Ig and immunoreceptor tyrosine-based inhibition motif domains (TIGIT) has marked a significant advancement in oncology, offering new therapeutic opportunities to fight diverse malignancies. This review covers the biological basis and clinical application of LAG3 and TIGIT inhibitors, highlighting pivotal trials and therapeutic outcomes. We underscore the use of dual therapy immune checkpoint blockade in enhancing antitumor immunity, particularly in settings where monotherapy has shown limited efficacy. Additionally, we address the emerging challenges such as treatment resistance and adverse effects. We explore the strategic integration of LAG3 and TIGIT blockade within the broader immunotherapy landscape, emphasizing innovative combinations and the quest for predictive biomarkers to optimize patient selection and treatment efficacy.
Collapse
Affiliation(s)
- Ecaterina Elena Dumbrava
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA;
| | | | - Elad Sharon
- Immunotherapy Toxicity Service, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Hussein Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|