1
|
Xie J, Zheng Z, Wang B, Zhang J, Jiang J, Wu F, Zhong X, Chen J. LncRNA HOTAIR promotes aerobic glycolysis by recruiting Lin28 to induce inflammation and apoptosis in acute lung injury. RNA Biol 2025; 22:1-12. [PMID: 40052944 PMCID: PMC11901367 DOI: 10.1080/15476286.2025.2475255] [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] [Revised: 01/24/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Acute lung injury (ALI) is a life-threatening condition with high rates of morbidity and mortality. Recently, there has been growing evidence suggesting a link between lncRNA HOTAIR and ALI. Nonetheless, the precise role and mechanism of lncRNA HOTAIR in ALI remain to be fully elucidated. siHOTAIR transfection, qPCR detection (HOTAIR), ELISA (TNF-α, IL-6, and IL-1β), Lactate detection, Glucose uptake experiment, Cell Apoptosis Analysis, Fluorescence in situ hybridization (FISH) assay. Through siHOTAIR transfection, we discovered that HOTAIR plays a role in the secretion of inflammatory factors in ALI and further regulates glucose uptake and metabolism in lung epithelial cells. Moreover, a comparison between HOTAIR knockdown cells and HOTAIR overexpression cells revealed that HOTAIR promotes cellular aerobic sugar metabolism, leading to increased secretion of inflammatory factors and cell apoptosis. Our in-depth research also identified an interaction between HOTAIR and the LIN28 protein. Knocking down HOTAIR resulted in the downregulation of LIN28 protein expression, which subsequently inhibited the expression of the glucose transporter GLUT1. This indicates that HOTAIR facilitates glucose uptake and boosts cellular aerobic glycolysis by modulating the LIN28 protein, thereby promoting inflammation and apoptosis in acute lung injury. The research findings presented in this article offer significant insights into the function of HOTAIR in ALI and suggest a potential therapeutic target for the treatment of this condition.
Collapse
Affiliation(s)
- Junjie Xie
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Zhicong Zheng
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Bin Wang
- Department of Pediatrics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jianfang Zhang
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Junqi Jiang
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Fengde Wu
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Xiangming Zhong
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Jianfeng Chen
- Department of Pediatrics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Li S, Liu T, Li C, Zhang Z, Zhang J, Sun D. Overcoming immunotherapy resistance in colorectal cancer through nano-selenium probiotic complexes and IL-32 modulation. Biomaterials 2025; 320:123233. [PMID: 40081224 DOI: 10.1016/j.biomaterials.2025.123233] [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: 10/14/2024] [Revised: 02/11/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND AND OBJECTIVE Colorectal cancer (CRC) is a major global health burden, with immunotherapy often limited by immune tolerance and resistance. This study introduces an innovative approach using Selenium Nanoparticles-Loaded Extracellular Vesicles combined with Interleukin-32 and Engineered Probiotic Escherichia coli Nissle 1917 (SeNVs@NE-IL32-EcN) to enhance CD8+ T cell-mediated immune responses and overcome immunotherapy resistance. METHODS Single-cell RNA sequencing (scRNA-seq) and transcriptomic analyses were performed to identify key immune cells and regulators involved in CRC immunotherapy resistance, focusing on CD8+ T cells and the regulatory factor IL32. A humanized xenograft mouse model was used to evaluate the impact of IL32 and SeNVs@NE-IL32-EcN on tumor growth and immune responses. The SeNVs@NE-IL32-EcN complex was synthesized through a reverse micelle method and functionalized using extracellular vesicles. Its morphology, size, antioxidant activity, and safety were characterized using electron microscopy, dynamic light scattering (DLS), and in vitro co-culture assays. RESULTS Single-cell analyses revealed a significant reduction in CD8+ T cell infiltration in immunotherapy-resistant CRC patients. IL32 was identified as a key regulator enhancing CD8+ T cell cytotoxic activity through granzyme B and IFN-γ secretion. Treatment with SeNVs@NE-IL32-EcN significantly improved the proliferation and activity of CD8+ T cells and reduced tumor progression in humanized mouse models. In vitro and in vivo results demonstrated the complex's biocompatibility, antioxidant properties, and ability to enhance CRC immunotherapy while mitigating immune tolerance. CONCLUSION SeNVs@NE-IL32-EcN offers a novel nano-biomaterial strategy that integrates nanotechnology and probiotic therapy to enhance CD8+ T cell-mediated immunity and overcome CRC immunotherapy resistance. This study lays the foundation for future therapeutic applications in cancer treatment by advancing immune-modulating biomaterials.
Collapse
Affiliation(s)
- Shiquan Li
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China
| | - Tao Liu
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China
| | - Chenyao Li
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China
| | - Zhiyuan Zhang
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China
| | - Jiantao Zhang
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China
| | - Di Sun
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
3
|
Wei R, Xie K, Li T, Lin W, Zhao Y, Li J, Lai S, Wei X, Jiang X, Yuan Y, Yang R. Immunity/metabolism dual-regulation via an acidity-triggered bioorthogonal assembly nanoplatform enhances glioblastoma immunotherapy by targeting CXCL12/CXCR4 and adenosine-A2AR pathways. Biomaterials 2025; 319:123216. [PMID: 40037210 DOI: 10.1016/j.biomaterials.2025.123216] [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: 10/15/2024] [Revised: 01/16/2025] [Accepted: 02/24/2025] [Indexed: 03/06/2025]
Abstract
Blocking the C-X-C motif chemokine ligand-12/C-X-C motif chemokine receptor-4 (CXCL12/CXCR4) signal offers the potential to induce immunogenic cell death (ICD) and enhance immunotherapy of glioblastoma (GBM). However, traditional intracellular targeted delivery strategies and adenosine-mediated tumor immunosuppression limit its therapeutic efficacy. Herein, we present an acidity-triggered self-assembly nanoplatform based on bioorthogonal reaction to potentiate GBM immunotherapy through dual regulation of metabolism and immune pathways. AMD3100 (CXCR4 antagonist) and CPI-444 (adenosine 2A receptor inhibitor) were formulated into micelles, denoted as AMD@iNPDBCO and CPI@iNPN3, respectively. Upon administration, the pH-sensitive poly(2-azepane ethyl methacrylate) group of AMD@iNPDBCO responds to the acidic tumor microenvironment, exposing the DBCO moiety, resulting in highly efficient bioorthogonal reaction with azide group on CPI@iNPN3 to form large-sized aggregates, ensuring extracellular drug release. The combination of AMD3100 and CPI-444 contributes to ICD induction, dendritic cell maturation, and immunosuppressive milieu alleviation by reducing tumor-associated macrophages, myeloid-derived suppressor cells, and regulatory T cells, leading to a robust antitumor response, thereby significantly prolonging survival in orthotopic GBM-bearing mice. Furthermore, the nanoplatform remarkably amplifies immuno-radiotherapy by potently evoking cytotoxic CD8+ T cell priming, and synergized with immune checkpoint blockade by delaying CD8+ T cell exhaustion. Our work highlights the potential of the in situ assembly nanoplatform tailored for delivery of extracellular-targeted therapeutic agents for boosting GBM immunotherapy.
Collapse
Affiliation(s)
- Ruili Wei
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China; Department of Radiology, Guangzhou First People's Hospital, Guangzhou 510180, PR China
| | - Kunfeng Xie
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China
| | - Tao Li
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China
| | - Wanxian Lin
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China; Department of Radiology, Guangzhou First People's Hospital, Guangzhou 510180, PR China
| | - Yandong Zhao
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China; Department of Radiology, Guangzhou First People's Hospital, Guangzhou 510180, PR China
| | - Jiamin Li
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China; Department of Radiology, Guangzhou First People's Hospital, Guangzhou 510180, PR China
| | - Shengsheng Lai
- School of Medical Equipment, Guangdong Food and Drug Vocational College, Guangzhou 510520, PR China
| | - Xinhua Wei
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China; Department of Radiology, Guangzhou First People's Hospital, Guangzhou 510180, PR China
| | - Xinqing Jiang
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China; Department of Radiology, Guangzhou First People's Hospital, Guangzhou 510180, PR China
| | - Youyong Yuan
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou 510180, PR China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China.
| | - Ruimeng Yang
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, PR China; Department of Radiology, Guangzhou First People's Hospital, Guangzhou 510180, PR China.
| |
Collapse
|
4
|
Luo R, Liu J, Wang T, Zhao W, Wang Y, Wen J, Wang H, Ding S, Zhou X. The landscape of malignant transition: Unraveling cancer cell-of-origin and heterogeneous tissue microenvironment. Cancer Lett 2025; 621:217591. [PMID: 40054660 PMCID: PMC12040592 DOI: 10.1016/j.canlet.2025.217591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/10/2025] [Accepted: 02/25/2025] [Indexed: 03/12/2025]
Abstract
Understanding disease progression and sophisticated tumor ecosystems is imperative for investigating tumorigenesis mechanisms and developing novel prevention strategies. Here, we dissected heterogeneous microenvironments during malignant transitions by leveraging data from 1396 samples spanning 13 major tissues. Within transitional stem-like subpopulations highly enriched in precancers and cancers, we identified 30 recurring cellular states strongly linked to malignancy, including hypoxia and epithelial senescence, revealing a high degree of plasticity in epithelial stem cells. By characterizing dynamics in stem-cell crosstalk with the microenvironment along the pseudotime axis, we found differential roles of ANXA1 at different stages of tumor development. In precancerous stages, reduced ANXA1 levels promoted monocyte differentiation toward M1 macrophages and inflammatory responses, whereas during malignant progression, upregulated ANXA1 fostered M2 macrophage polarization and cancer-associated fibroblast transformation by increasing TGF-β production. Our spatiotemporal analysis further provided insights into mechanisms responsible for immunosuppression and a potential target to control evolution of precancer and mitigate the risk for cancer development.
Collapse
Affiliation(s)
- Ruihan Luo
- Laboratory of Hepatic AI Translation, Frontier Science Center for Disease-Related Molecular Network and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China; Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA; Med-X Center for Informatics, Sichuan University, Chengdu, 610041, China.
| | - Jiajia Liu
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Tiangang Wang
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Weiling Zhao
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yanfei Wang
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Jianguo Wen
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Hongyu Wang
- Department of Diagnostic and Interventional Imaging, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA; Center for Nursing Research, Cizik School of Nursing, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Shanli Ding
- Graduate School of Biomedical Sciences, The University of MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaobo Zhou
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA; McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
5
|
Zhang J, Bao Y, Yu Z, Lin Y. Application and clinical translational value of a predictive model based on N 7-methylguanosine-related long non-coding RNAs in cervical squamous cell carcinoma. Oncol Lett 2025; 30:341. [PMID: 40438871 PMCID: PMC12117525 DOI: 10.3892/ol.2025.15087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 04/04/2025] [Indexed: 06/01/2025] Open
Abstract
Cervical squamous cell carcinoma (CSCC) is one of the most common gynecological malignancies affecting women globally. The present study aimed to develop a predictive model based on N7-methylguanosine-related long non-coding RNAs (lncRNAs) to evaluate risk stratification, analyze immune infiltration and guide the selection of sensitive drugs in CSCC. Pearson's correlation, univariate Cox and Least Absolute Shrinkage and Selection Operator regression analyses of transcriptome data from The Cancer Genome Atlas and the Genotype-Tissue Expression database were conducted to construct a prognostic risk prediction model for CSCC. The stability of the model was tested before evaluating its prognostic value in CSCC. Further analysis of enrichment, immune infiltration and drug resistance provided directions for clinical translation. The lncRNAs used to construct the model were validated using reverse transcription-quantitative PCR. The developed predictive model was stable and may hold notable clinical translational value for immunotherapy and drug selection in CSCC in the future.
Collapse
Affiliation(s)
- Jun Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
- Department of Radiotherapy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
| | - Yingna Bao
- Department of Radiotherapy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
| | - Zhilong Yu
- Department of Radiotherapy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
| | - Yu Lin
- Department of Radiotherapy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
| |
Collapse
|
6
|
Reader CS, Liao W, Potter-Landua BJ, Veyssier CS, Seal CJ, Brewis N, Morrow M. The tetravalent, bispecific properties of FS118, an anti-LAG-3/PD-L1 antibody, mediate LAG-3 shedding from CD4 + and CD8 + tumor-infiltrating lymphocytes. Anticancer Drugs 2025; 36:447-458. [PMID: 39960386 PMCID: PMC12061380 DOI: 10.1097/cad.0000000000001705] [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: 11/26/2024] [Accepted: 12/06/2024] [Indexed: 03/01/2025]
Abstract
Tumor-infiltrating lymphocytes (TILs) often have upregulated expression of immune checkpoint receptors, such as programmed cell death 1 (PD-1) and lymphocyte-activation gene 3 (LAG-3). Patients treated with antibodies targeting PD-1 or its ligand (PD-L1) can develop resistance or relapse, with LAG-3 upregulation on T cells being one possible mechanism. FS118 is a tetravalent, bispecific antibody comprising a full-length IgG 1 anti-PD-L1 antibody with bivalent LAG-3-binding capability in the fragment crystallizable region. Here we demonstrate how the structure of FS118 is important for its function. We generated variants of FS118 and tested their ability to mediate LAG-3 shedding using staphylococcal enterotoxin B assays, antigen recall assays, and soluble LAG-3 ELISAs. Mediated by metalloproteases ADAM10 and ADAM17, FS118 induced shedding of LAG-3 from the surface of both CD4 + and CD8 + T cells. We also determined the effect of surrogate antibodies on immune cell LAG-3 expression and proliferation in syngeneic mouse models. In vivo , the bivalent LAG-3 binding sites of a mouse surrogate of FS118 and their location in the fragment crystallizable region were important for eliciting maximal reduction in LAG-3 levels on the surface of TILs, as variants with a single LAG-3 binding site in the fragment crystallizable region, or with reversed orientation of the LAG-3 and PD-L1 binding sites, were less efficient at inducing shedding. We also show that PD-L1, not PD-1, binding drives the LAG-3 reduction on TILs. We hypothesize that the LAG-3 bivalency in the fragment crystallizable region of FS118 allows LAG-3 clustering, which optimizes cleavage by ADAM10/ADAM17 and thus shedding.
Collapse
|
7
|
Qu Y, Liang W, Yu M, Wang C, Luo M, Zhong L, Li Z, Wang F. MYO1F in neutrophils is required for the response to immune checkpoint blockade therapy. J Exp Med 2025; 222:e20241957. [PMID: 40202509 PMCID: PMC11980683 DOI: 10.1084/jem.20241957] [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: 10/22/2024] [Revised: 01/29/2025] [Accepted: 02/21/2025] [Indexed: 04/10/2025] Open
Abstract
Tumor-associated neutrophils (TANs) represent a significant barrier to the effectiveness of immune checkpoint blockade (ICB) therapy. A comprehensive understanding of TANs' regulatory mechanisms is therefore essential for predicting ICB efficacy and improving immunotherapy strategies. Our study reveals that MYO1F is selectively downregulated in neutrophils within both human cancers and murine tumor models, showing a negative correlation with ICB response. Mechanistically, MYO1F normally inhibits neutrophil immunosuppression and proliferation by restraining STAT3 activity. However, during tumorigenesis, tumor-derived TGF-β1 disrupts the binding of SPI1 to intron 8 of Myo1f via DNA methylation, thereby suppressing Myo1f transcription. The resultant decrease in MYO1F reprograms neutrophils into an immunosuppressive state through the STAT3-dependent signaling pathways. This immunosuppressive state further contributes to tumor microenvironment (TME) remodeling by inducing CTL exhaustion. These findings establish MYO1F as a critical regulator within TANs, highlighting its significant role in modulating ICB therapy efficacy.
Collapse
Affiliation(s)
- Yingying Qu
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Microbiota and Immunological Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenhua Liang
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingzhu Yu
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenhui Wang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Min Luo
- Institute of Pediatrics of Children’s Hospital of Fudan University, The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Lin Zhong
- Department of Liver Surgery and Organ Transplantation Center, Shenzhen Third People’s Hospital, Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhigang Li
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Wang
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Rojas-Diaz JM, Solorzano-Ibarra F, Garcia-Barrientos NT, Klimov-Kravtchenko K, Cruz-Ramos JA, Guitron-Aviña MS, Urciaga-Gutierrez PI, Ortiz-Lazareno PC, Tellez-Bañuelos MC, Bueno-Topete MR, Haramati J, Del Toro-Arreola S. Beyond Canonical Immune Checkpoints: Overexpression of TNFRSF Members 4-1BB and OX-40 Marks T Cells Exhibiting Phenotypic Features of Exhaustion in Cervical Carcinoma. Immunology 2025. [PMID: 40387515 DOI: 10.1111/imm.13945] [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: 02/09/2025] [Revised: 04/21/2025] [Accepted: 05/06/2025] [Indexed: 05/20/2025] Open
Abstract
T cells are pivotal in combating cancer; however, they can become exhausted during tumour progression, losing their cytotoxic capacity and upregulating inhibitory receptors including PD-1 and TIGIT. While checkpoint blockade has emerged as a potent treatment option for numerous cancers, patient selection, long-term efficacy, and adverse effects still remain an issue. For these reasons, it is important to investigate other pathways that might lead to selective reactivation of the immune system. Co-stimulatory TNFRSF receptors, including 4-1BB and OX-40, have emerged as promising targets for reactivating exhausted T cells. However, their expression on exhausted peripheral and tumour-infiltrating lymphocytes (TILs) is not well characterised, particularly in cervical cancer (CC), which remains the leading cause of gynaecological cancer mortality in low- and middle-income countries. To investigate the expression of these receptors, PBMCs were collected from CC patients and healthy donors, along with TILs from tumour biopsies, and analysed using multiparametric flow cytometry. Our findings revealed an increased population of phenotypically exhausted (PD-1+TIGIT+) CD4+ and CD8+ T cells in TILs, and, to a lesser extent, in peripheral blood and from CC patients. These exhausted T cell subsets exhibited selective overexpression of 4-1BB and OX-40 compared to phenotypically non-exhausted cells (PD-1-TIGIT-). In TILs, 4-1BB was overexpressed 12.7-fold in CD8 cells with the exhausted phenotype, OX-40 was overexpressed 3.3-fold; in CD4 cells with the exhausted phenotype, the overexpression was 7.8× and 3.8× for 4-1BB and OX-40, respectively. CD8 and CD4 T cells that were PD-1 + TIGIT+ 4-1BB+ were 7.3× and 16× more likely to be found in the tumour versus peripheral blood. Additionally, subpopulations of PD-1high T cells were significantly elevated in the tumour-infiltrating T cells and TIGIT expression was positively associated with PD-1 levels in peripheral patient CD8+ and CD4+ T cells, potentially indicating an advanced state of exhaustion. These findings suggest that TNFRSF members, especially 4-1BB, may serve as potential immunotherapeutic targets for reinvigorating exhausted T cells in CC.
Collapse
Affiliation(s)
- Jose Manuel Rojas-Diaz
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Fabiola Solorzano-Ibarra
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Nadia Tatiana Garcia-Barrientos
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Ksenia Klimov-Kravtchenko
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Jose Alfonso Cruz-Ramos
- Coordinación de Investigación, Subdirección de Desarrollo Institucional, Instituto Jalisciense de Cancerología, Guadalajara, Jalisco, Mexico
| | - Marcela Sofia Guitron-Aviña
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
- Laboratorio de Inmunología Traslacional, Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Pedro Ivan Urciaga-Gutierrez
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Pablo Cesar Ortiz-Lazareno
- Centro de Investigación Biomédica de Occidente, División de Inmunología, Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, Mexico
| | - Martha Cecilia Tellez-Bañuelos
- Laboratorio de Inmunología Traslacional, Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Miriam Ruth Bueno-Topete
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Jesse Haramati
- Laboratorio de Inmunología Traslacional, Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Susana Del Toro-Arreola
- Instituto de Investigación en Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
- Laboratorio de Inmunología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| |
Collapse
|
9
|
Zhou M, Chen M, Zheng Z, Li Q, Liao L, Wang Y, Xu Y, Shu G, Luo J, Yang T, Zhang J. CircRNA GRAMD4 induces NBR1 expression to promote autophagy and immune escape in renal cell carcinoma. Autophagy 2025:1-21. [PMID: 40373256 DOI: 10.1080/15548627.2025.2503560] [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: 09/01/2024] [Revised: 04/15/2025] [Accepted: 05/05/2025] [Indexed: 05/17/2025] Open
Abstract
The tumor microenvironment (TME) in renal cell carcinoma (RCC) frequently exhibits significant immune cell infiltration. However, tumor cells often manage to evade immune surveillance. This study revealed the mechanism by which circular RNA circGRAMD4 regulates NBR1. CircGRAMD4 is markedly elevated in RCC, and its high levels are correlated with a poor prognosis. Notably, the absence of circGRAMD4 has been demonstrated to result in a significant inhibition of renal cancer cell growth. This inhibition has been attributed to an enhanced anti-tumor immunity mediated by CD8+ T cells. Mechanistically, circGRAMD4 interacts with the RBM4 protein, stabilizing the autophagic cargo receptor NBR1 mRNA. This interaction promotes NBR1 expression, which in turn leads to the degradation of MHC-I molecules through macroautophagy/autophagy pathways. Consequently, this process affects renal cancer cell antigen presentation, induces CD8+ T cell dysfunction, and contributes to tumor immune escape. Moreover, by inhibiting circGRAMD4 and using immune checkpoint blockers (ICB), the immunosuppressive TME is altered to prevent tumor immune evasion, ultimately increasing the effectiveness of ICB treatment. The discovery highlights the significant impact of circGRAMD4 on RCC immune escape and proposes that blocking circGRAMD4 could serve as a promising immunotherapy strategy when combined with ICB to enhance patient outcomes.
Collapse
Affiliation(s)
- Mi Zhou
- Department of Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Minyu Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Zhousan Zheng
- Department of Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Qihao Li
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Lican Liao
- Department of Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Yunfei Wang
- Department of Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Yi Xu
- Department of Breast and Thyroid Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou Provincial Clinical Research Center for Child Health, Guangdong, PR China
| | - Guannan Shu
- Department of Urology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong Provincial Clinical Research Center for Child Health, Guangdong, PR China
| | - Junhang Luo
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Taowei Yang
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Jiaxing Zhang
- Department of Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| |
Collapse
|
10
|
Dang B, Liang L, Li Z, Luo J, Zhong S. Bolstering CD8 + T Cells' Antitumor Immunity: A Promising Strategy to Improve the Response to Advanced Prostate Cancer Treatment. BIOLOGY 2025; 14:544. [PMID: 40427733 PMCID: PMC12108615 DOI: 10.3390/biology14050544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2025] [Revised: 05/07/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025]
Abstract
Prostate cancer is among the most frequently diagnosed and deadly cancers among men in the Western world. It is typically classified as an immune "cold" tumor due to its sparse immune cell presence and limited immunogenic response. Recent research has revealed the significant role of immune cells, especially CD8+ T cells, in both prostate cancer progression and treatment efficacy. This review integrates recent findings to provide a comprehensive overview of the current understanding of CD8+ T cell dynamics in prostate cancer and discusses emerging strategies to improve treatment outcomes. The ongoing exploration of new molecular targets and the development of innovative immunotherapeutic approaches hold promise for more effective management of prostate cancer, particularly in the context of advanced and resistant forms of the disease.
Collapse
Affiliation(s)
| | | | | | | | - Shangwei Zhong
- The Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang 421009, China; (B.D.); (L.L.); (Z.L.); (J.L.)
| |
Collapse
|
11
|
Gou YK, Zhou J, Liu P, Wang MY. Research progress on monocyte/macrophage in the development of gastric cancer. Future Oncol 2025:1-11. [PMID: 40351251 DOI: 10.1080/14796694.2025.2504334] [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: 03/09/2024] [Accepted: 05/07/2025] [Indexed: 05/14/2025] Open
Abstract
Gastric cancer (GC) is diagnosed more than one million times each year and represents a major cause of cancer-related death worldwide. Although GC presents as a group of different types of disease, chronic inflammation has been strongly associated with tumorigenesis. Monocyte/macrophage play important roles in the development of inflammation and are vital components of the tumor microenvironment (TME). Monocyte/macrophage exert protumor and/or antitumor effects through the release of angiogenic and lymphangiogenic factors. Furthermore, tumor associated macrophages (TAMs) are emerging as key players in GC development. It is necessary to review and elucidate the roles of TAM subsets in GC and their molecular features. In this study, we focused on GC-related subsets of monocytes/macrophages and analyzed signaling related to TAMs in GC as well as the potential roles of these cells as therapeutic targets.
Collapse
Affiliation(s)
- Yuan-Kun Gou
- School of Medical Laboratory, Shandong Second Medical University, Weifang, Shandong, PR China
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, PR China
| | - Jie Zhou
- School of Medical Laboratory, Shandong Second Medical University, Weifang, Shandong, PR China
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, PR China
| | - Peng Liu
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, PR China
| | - Ming-Yi Wang
- School of Medical Laboratory, Shandong Second Medical University, Weifang, Shandong, PR China
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, PR China
| |
Collapse
|
12
|
Li B, Cui H, Liu W, Lan Z, Liu C, Yang Y, Zhao Y, Tian Z, Chen H, Yu G. DDX10 Exacerbates Exosomal PD-L1-Dependent T Cell Exhaustion via Phase Separation of Rab27b in Oral Squamous Cell Carcinoma. RESEARCH (WASHINGTON, D.C.) 2025; 8:0697. [PMID: 40352946 PMCID: PMC12063704 DOI: 10.34133/research.0697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 05/14/2025]
Abstract
DEAD-box ATPase 10 (DDX10), a prominent RNA-binding protein in the DDX family, has a critical function in cancer progression. Nevertheless, its well-defined mechanisms in oral squamous cell carcinoma (OSCC) are still not well understood. Here, we identify that DDX10 is substantially increased in OSCC, which is positively correlated with poor prognosis and malignant behavior. Mechanistically, we found that DDX10 had physical interaction with Rab27b by undergoing phase separation. Knockdown of DDX10 inhibited Rab27b-mediated exosome secretion and the expression of programmed cell death-ligand 1 (PD-L1) within its contents. Furthermore, knocking down DDX10 could restore the function and infiltration of T cells, hence inhibiting the progression of OSCC. These findings highlight that the oncogenic role of DDX10 in promoting exosomal PD-L1 secretion via phase separation with Rab27b has been preliminarily validated in T cell exhaustion in OSCC. A potential strategy for improving OSCC immunotherapy may involve the inhibition of DDX10.
Collapse
Affiliation(s)
- Bowen Li
- Stomatological Hospital, School of Stomatology,
Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Hao Cui
- Stomatological Hospital, School of Stomatology,
Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Wei Liu
- Stomatological Hospital, School of Stomatology,
Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Zhou Lan
- Stomatological Hospital, School of Stomatology,
Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Chang Liu
- Stomatological Hospital, School of Stomatology,
Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yumiao Yang
- Stomatological Hospital, School of Stomatology,
Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yuyue Zhao
- Stomatological Hospital, School of Stomatology,
Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Zhen Tian
- Stomatological Hospital, School of Stomatology,
Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Hao Chen
- Stomatological Hospital, School of Stomatology,
Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Guangtao Yu
- Stomatological Hospital, School of Stomatology,
Southern Medical University, Guangzhou, Guangdong 510280, China
| |
Collapse
|
13
|
Chen Y, Ohara T, Hamada Y, Wang Y, Tian M, Noma K, Tazawa H, Fujisawa M, Yoshimura T, Matsukawa A. HIF-PH inhibitors induce pseudohypoxia in T cells and suppress the growth of microsatellite stable colorectal cancer by enhancing antitumor immune responses. Cancer Immunol Immunother 2025; 74:192. [PMID: 40343532 PMCID: PMC12064516 DOI: 10.1007/s00262-025-04067-3] [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: 09/28/2024] [Accepted: 04/18/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Recent studies have revealed that CD8+ T cells can be activated via genetic upregulation of HIF-1α, thereby augmenting antitumor effector functions. HIF-1α upregulation can be attained by inhibiting HIF-prolyl hydroxylase (HIF-PH) under normoxic conditions, termed pseudohypoxia. This study investigated whether pseudohypoxia induced by HIF-PH inhibitors suppresses Microsatellite stable (MSS) colorectal cancer (CRC) by affecting tumor immune response. METHODS The HIF-PH inhibitors Roxadustat and Vadadustat were utilized in this study. In vitro, we assessed the effects of HIF-PH inhibitors on human and murine colon cancer cell lines (SW480, HT29, Colon26) and murine T cells. In vivo experiments were performed with mice bearing Colon26 tumors to evaluate the effect of these inhibitors on tumor immune responses. Tumor and spleen samples were analyzed using immunohistochemistry, RT-qPCR, and flow cytometry to elucidate potential mechanisms. RESULTS HIF-PH inhibitors demonstrated antitumor effects in vivo but not in vitro. These inhibitors enhanced the tumor immune response by increasing the infiltration of CD8+ and CD4+ tumor-infiltrating lymphocytes (TILs). HIF-PH inhibitors induced IL-2 production in splenic and intratumoral CD4+ T cells, promoting T cell proliferation, differentiation, and immune responses. Roxadustat synergistically enhanced the efficacy of anti-PD-1 antibody for MSS cancer by increasing the recruitment of TILs and augmenting effector-like CD8+ T cells. CONCLUSION Pseudohypoxia induced by HIF-PH inhibitors activates antitumor immune responses, at least in part, through the induction of IL-2 secretion from CD4+ T cells in the spleen and tumor microenvironment, thereby enhancing immune efficacy against MSS CRC.
Collapse
Affiliation(s)
- Yuehua Chen
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Toshiaki Ohara
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| | - Yusuke Hamada
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Yuze Wang
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Miao Tian
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Kazuhiro Noma
- Department of Gastroenterological Surgery, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Hiroshi Tazawa
- Department of Gastroenterological Surgery, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, Japan
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Masayoshi Fujisawa
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Akihiro Matsukawa
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| |
Collapse
|
14
|
Zhang Y, Xu Y, Zhong W, Zhao J, Liu X, Gao X, Chen M, Wang M. Vitamin D and Immune Checkpoint Inhibitors in Lung Cancer: A Synergistic Approach to Enhancing Treatment Efficacy. Int J Mol Sci 2025; 26:4511. [PMID: 40429656 PMCID: PMC12111780 DOI: 10.3390/ijms26104511] [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: 03/15/2025] [Revised: 04/27/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Lung cancer, a malignant neoplasm that is globally prevalent and characterized by high incidence and mortality rates, has seen the rise of immune checkpoint inhibitors (ICIs) as a crucial systemic treatment. However, a subset of patients exhibits suboptimal responses to ICIs. Recently, studies revealed the role of vitamin D in inflammation modulation, cell differentiation, and cancer prevention. Vitamin D precisely modulates immune responses and inflammatory states within the tumor microenvironment (TME) by targeting both innate and adaptive immunity. These effects may reduce immune tolerance to ICIs and synergistically enhance their therapeutic efficacy. Here, we review vitamin D metabolism in lung cancer patients, as well as its anti-tumor mechanisms, immune regulation, and the significant promise of vitamin D in lung cancer immunotherapy and adjuvant therapeutic strategies. Further research is imperative to surmount these challenges and fully realize vitamin D's potential in improving lung cancer immunotherapy outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Minjiang Chen
- Department of Respiratory & Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; (Y.Z.); (Y.X.); (W.Z.); (J.Z.); (X.L.); (X.G.); (M.W.)
| | | |
Collapse
|
15
|
Xin J, Xiang Y, Jiang J, Jiang Z, Yi B. Next-generation probiotics Alistipes onderdonkii enhances the efficacy of anti-PD-1 therapy in colorectal cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167891. [PMID: 40328413 DOI: 10.1016/j.bbadis.2025.167891] [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: 05/09/2024] [Revised: 01/22/2025] [Accepted: 05/02/2025] [Indexed: 05/08/2025]
Abstract
Despite significant breakthroughs of therapeutic antibodies targeting PD-1 in cancer treatment, most colorectal cancer (CRC) patients respond poorly to anti-PD-1 immunotherapy. The combination therapy strategies are used to overcome the limitations mentioned above. Alistipes (A.) onderdonkii has anti-cancer effects. This study aimed to examine A. onderdonkii's effects and the related mechanisms in the anti-PD-1 treatment for CRC. In this study, anti-PD-1 therapy notably affected gut microbiota community composition in mice, particularly upregulating A. onderdonkii abundance. A. onderdonkii supplementation markedly promoted anti-PD-1 therapy's effectiveness on CRC treatment, as manifested by the elevated CD8+ T cell infiltration, promoted intestinal barrier integrity, and affected serum metabolomics. A. onderdonkii showed similar effects to the identified next-generation probiotics (NGP) Akkermansia (A.) muciniphila. Additionally, the combination therapy of anti-PD-1 and probiotics supplementation reduced PD-1 level and elevated IL-2 level, indicating that anti-PD-1 combined with A. onderdonkii efficiently restored T cell functions by inhibiting suppressive checkpoints. In conclusion, anti-PD-1 plus A. onderdonkii supplementation could account for a potential method for CRC therapy. These results provide strong evidence for A. onderdonkii as a potential NGP.
Collapse
Affiliation(s)
- Jiaxuan Xin
- Department of Gastrointestinal Surgery, the Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Yandong Xiang
- Department of Gastrointestinal Surgery, the Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Juan Jiang
- Department of Nephrology, the Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Zhengqi Jiang
- Department of Gastrointestinal Surgery, the Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Bo Yi
- Department of Gastrointestinal Surgery, the Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China.
| |
Collapse
|
16
|
Han Y, Hu M, Wang Y, Xu S, Jiang F, Wang Y, Liu Z. A coagulation-related long non-coding RNA signature to predict prognosis and immune features of breast cancer. Discov Oncol 2025; 16:662. [PMID: 40317354 PMCID: PMC12049355 DOI: 10.1007/s12672-025-02316-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/04/2025] [Indexed: 05/07/2025] Open
Abstract
Breast cancer (BC) remains one of the most common malignancies among women worldwide, with persistently poor prognosis despite advancements in diagnostics and therapies. Long non-coding RNAs (lncRNAs) and coagulation-related genes (CRGs) are increasingly recognized for their roles in prognosis and immune modulation. Using transcriptomic data from 1,045 BC patients in TCGA, we identified CRG-associated lncRNAs via coexpression analysis (Pearson |R|> 0.4, p < 0.001) and constructed a prognostic model through univariate Cox analysis, LASSO regression with tenfold cross-validation (λ = 0.05), and multivariate Cox analysis. The model stratified patients into high- and low-risk groups with distinct overall survival (HR = 3.21, p < 0.001) and demonstrated robust predictive accuracy (AUC = 0.795 at 1 year). Functional enrichment revealed immune-related pathways (e.g., cytokine signaling, PD-L1 regulation), and high-risk patients exhibited elevated tumor mutational burden (TMB) and PD-L1 expression, suggesting enhanced immunotherapy responsiveness. Drug sensitivity analysis identified 5 targeted agents (e.g., BIBW2992) with differential efficacy between risk groups. This CRG-lncRNA signature provides a novel tool for prognosis prediction and personalized immunotherapy in BC, illuminating crosstalk between coagulation and immune pathways.
Collapse
Affiliation(s)
- Yetao Han
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Mengsi Hu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Yanzhong Wang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Shoufang Xu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Feiyu Jiang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Yingjian Wang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Zhiwei Liu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China.
| |
Collapse
|
17
|
Ren C, Li Y, Li M, Wang Y. Unveiling vitamin C: A new hope in the treatment of diffuse large B‑cell lymphoma (Review). Int J Oncol 2025; 66:40. [PMID: 40314093 PMCID: PMC12068847 DOI: 10.3892/ijo.2025.5746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/03/2025] [Indexed: 05/03/2025] Open
Abstract
Lymphoma is a malignancy of the immune system, which originates from lymphatic tissues and lymph nodes. Diffuse large B‑cell lymphoma (DLBCL) is a common type of non‑Hodgkin lymphoma, occurring in 30‑40% of all cases, which has persistent clinical challenges. The treatment of DLBCL is challenging due to its diverse genetic and biological characteristics and complex clinical physiology. Despite advancements in overall prognosis, 20‑25% of patients continue to experience relapse and 10‑15% of patients experience refractory disease. Vitamin C is a water‑soluble vitamin with antioxidant properties and notable pharmacological activity, with potential applications in cancer therapy. Pharmacological doses of vitamin C (1‑4 g/kg) can induce apoptosis in malignant cells by inhibiting and/or reversing gene mutations that are associated with hematological malignancies. For example, 10‑25% of patients with myeloid malignancies have tet methylcytosine dioxygenase 2 (TET2) gene mutations and vitamin C can regulate blood stem cell frequency and leukemia production by enhancing TET2 function. Consequently, pharmacological doses of vitamin C can inhibit the development and progression of hematological malignancies. Therefore, the present review aimed to investigate the role of vitamin C in the pathophysiology and treatment of DLBCL, whilst highlighting the potential challenges and future perspectives.
Collapse
Affiliation(s)
- Chunxiao Ren
- Department of Hematology, Dazhou Central Hospital, Dazhou, Sichuan 635000, P.R. China
| | - Yaqiong Li
- Department of Hematology, Dazhou Central Hospital, Dazhou, Sichuan 635000, P.R. China
| | - Mingrui Li
- Department of Hematology, Dazhou Central Hospital, Dazhou, Sichuan 635000, P.R. China
| | - Yuqun Wang
- Department of Endocrinology and Metabolism, Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, P.R. China
| |
Collapse
|
18
|
Ling X, Chen G, Liu N, Xu W, Ding M. Integration of Single-Cell RNA Sequencing Data and Bulk Sequencing Data to Characterise the CD8+ T-Cell Exhaustion Mediated Immune Microenvironment in CRC. J Cell Mol Med 2025; 29:e70556. [PMID: 40356050 PMCID: PMC12069026 DOI: 10.1111/jcmm.70556] [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: 12/28/2024] [Revised: 03/23/2025] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
CD8+ T cells are crucial for the anti-tumour immune response, and their exhaustion contributes to poor prognosis and limited immunotherapy efficacy in colorectal cancer (CRC). In this study, we examined the immune microenvironment of CRC by integrating single-cell RNA sequencing (scRNA-seq) and bulk sequencing data. T-cell subtypes in tumour tissues were analysed using CellMarker 2.0 and scType, and an intercellular communication network was constructed through CellChat. Our analysis revealed that exhausted CD8+ T cells exhibit strong interactions with epithelial cells, primarily via the MIF-(CD74 + CXCR4), MIF-(CD74 + CD44) and CD99-CD99 pathways. Based on CD8+ T-cell exhaustion markers, we developed a prognostic model using XGBoost, which demonstrated promising predictive capabilities for CRC prognosis and immunotherapy response. Functional assays showed that MIF knock-down significantly inhibited CRC cell proliferation and invasion. Our findings suggest that MIF and CD99 are key regulators of CD8+ T-cell exhaustion in CRC. This study provides novel insights into the mechanisms underlying T-cell exhaustion in CRC and offers potential biomarkers for improving immunotherapy outcomes.
Collapse
Affiliation(s)
- Xiao‐Hua Ling
- Department of GastroenterologyThe Fourth Affiliated Hospital of Harbin Medical UniversityHarbinPeople's Republic of China
| | - Gang Chen
- Department of General SurgeryThe Fourth Hospital of Harbin Medical UniversityHarbinHeilongjiangPeople's Republic of China
| | - Nan‐Nan Liu
- Department of GastroenterologyThe Fourth Affiliated Hospital of Harbin Medical UniversityHarbinPeople's Republic of China
| | - Wen‐Xin Xu
- Department of Intensive Care UnitThe Fourth Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangPeople's Republic of China
| | - Ming‐Feng Ding
- Department of General SurgeryThe Fourth Hospital of Harbin Medical UniversityHarbinHeilongjiangPeople's Republic of China
| |
Collapse
|
19
|
Pham TN, Coupey J, Yger F, Candéias SM, Thariat J, Valable S. Effect of glioblastoma and brain radiotherapy on T-lymphocyte subpopulations in rodents. Radiother Oncol 2025; 206:110801. [PMID: 40081500 DOI: 10.1016/j.radonc.2025.110801] [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: 10/24/2024] [Revised: 02/07/2025] [Accepted: 02/20/2025] [Indexed: 03/16/2025]
Abstract
INTRODUCTION Although lymphopenia is linked to immune suppression favoring tumor growth, the effect of different radiation types on specific T-lymphocyte subsets remains unclear. Among the T-lymphocyte subpopulations, CD8+-lymphocytes serve as key effectors in cancer immunity. This study aimed to examine the changes in T-lymphocyte subpopulations in both tumor-free and glioblastoma-bearing mice following brain-irradiation. METHODS The study was divided into two main phases. First, C57BL/6 mice, with or without glioblastoma (GL261 cells), received hemispheric brain-irradiation or no-treatment. T-lymphocyte subpopulations were analyzed using flow cytometry at various timepoint. The effect of tumor size and brain-irradiation on these cells was assessed using correlation analysis. Next, C57BL/6 mice were subjected to different brain-irradiation conditions. Blood samples were collected during and post-irradiation to analyze T-lymphocyte subpopulations, and tree-based modeling was used to determine radiation parameters impact on naïve CD8+-lymphocyte levels. RESULTS Glioblastoma reduced all T-lymphocyte subpopulations by day 15 post-inoculation. Radiotherapy decreased regulatory and effector CD4+-lymphocytes in both tumor-free and glioblastoma-bearing mice, but not naïve or memory CD4+-lymphocytes, in both tumor-free and glioblastoma-bearing mice. In tumor-free mice, radiotherapy had no effect on CD8+-lymphocytes, but reduced all CD8+-lymphocyte types in glioblastoma-bearing mice. Glioblastoma size negatively affected CD8+-lymphocytes. Brain-irradiation persistently reduced naïve and memory CD8+-lymphocytes, but effector and regulatory T-lymphocytes recovered. Exposure of lymph nodes to radiation worsened CD8+-lymphocyte reduction. CONCLUSION These findings confirm that the presence of glioblastoma and brain-irradiation affect T-lymphocyte subpopulations in mice. The inclusion of lymph nodes in the irradiated area led to a long-term decrease in naïve CD8+-lymphocytes. Further mechanistic studies are needed to understand the molecular basis of radiation impact on T-lymphocyte subpopulations.
Collapse
Affiliation(s)
- Thao-Nguyen Pham
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, F-14000 Caen, France; Laboratoire de Physique Corpusculaire UMR6534 IN2P3/ENSICAEN, France - Normandie Université, France
| | - Julie Coupey
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, F-14000 Caen, France
| | - Florian Yger
- Univ. Paris-Dauphine, PSL Research Univ./CNRS, LAMSADE, Paris, France
| | - Serge M Candéias
- Univ. Grenoble Alpes, CEA, CNRS, IRIG-LCBM-UMR5249, 38054 Grenoble, France
| | - Juliette Thariat
- Laboratoire de Physique Corpusculaire UMR6534 IN2P3/ENSICAEN, France - Normandie Université, France; Department of Radiation Oncology, Centre François Baclesse, Caen, Normandy, France.
| | - Samuel Valable
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, F-14000 Caen, France.
| |
Collapse
|
20
|
Lee SW, Yun JS, Kim YJ, Jeong S, Noh JE, Kim HO, Cho HJ, Park CK, Oh IJ, Cho JH. Progressive accumulation of circulating CD27 -CD28 - effector/memory CD8 + T cells in patients with lung cancer blunts responses to immune checkpoint inhibitor therapy. Exp Mol Med 2025:10.1038/s12276-025-01448-7. [PMID: 40307573 DOI: 10.1038/s12276-025-01448-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 12/30/2024] [Accepted: 03/03/2025] [Indexed: 05/02/2025] Open
Abstract
Suppression of tumor-reactive CD8+ T cells is common within the tumor microenvironment. However, little is known about how tumors systemically affect the overall CD8+ T cell compartment. Here we demonstrate that peripheral blood CD8+ T cells from patients with lung cancer showed altered compositions particularly within CD45RA-CCR7- effector memory subpopulation. Specifically, patients with lung cancer exhibited increased frequency of more differentiated effector memory cells, which are less susceptible to T cell-receptor-induced proliferation. Further analysis using single-cell RNA sequencing revealed that these alterations were correlated with reduced quiescence and increased spontaneous activation at a systemic level, indicative of homeostatic dysregulation of the entire CD8+ T cell population. This phenomenon was found to be correlated with a poor clinical response to immune checkpoint inhibitor therapy across four independent cohorts, consisting of a total of 224 patients with lung cancer. These findings suggest that lung cancers continue to counteract potentially tumor-reactive CD8+ T cells by inducing homeostatic dysregulation of the entire CD8+ T cell compartment systematically.
Collapse
Affiliation(s)
- Sung-Woo Lee
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Ju Sik Yun
- Department of Thoracic and Cardiovascular Surgery, Chonnam National University Medical School, Hwasun Hospital, Gwangju, Republic of Korea
| | - Young Ju Kim
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Gwangju, Republic of Korea
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Gwangju, Republic of Korea
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Saei Jeong
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Gwangju, Republic of Korea
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Gwangju, Republic of Korea
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jeong Eun Noh
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Gwangju, Republic of Korea
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Gwangju, Republic of Korea
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hee-Ok Kim
- Selecxine Inc., Seoul, Republic of Korea
| | - Hyun-Ju Cho
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun Hospital, Gwangju, Republic of Korea
| | - Cheol-Kyu Park
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun Hospital, Gwangju, Republic of Korea
| | - In-Jae Oh
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun Hospital, Gwangju, Republic of Korea.
| | - Jae-Ho Cho
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea.
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Gwangju, Republic of Korea.
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Gwangju, Republic of Korea.
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Gwangju, Republic of Korea.
| |
Collapse
|
21
|
De Backer E, Verdoodt D, Ponsaerts P, Pasciuto E, Van Rompaey V. Cochlear T cells and their role in health and disease: A systematic review. Autoimmun Rev 2025; 24:103814. [PMID: 40221069 DOI: 10.1016/j.autrev.2025.103814] [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: 02/24/2025] [Revised: 03/26/2025] [Accepted: 04/08/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND The role of T cells in health and disease has already been studied extensively in many organs, yet their activity in the cochlea and involvement in hearing loss remains less explored. This review aims to summarize current existing literature on the presence and activity of T cells in the cochlea and the link between T-cell activity and the development of hearing loss. METHODS A systematic review of the literature was performed on PubMed and Web of Science on the 4th of December 2024 using the following search term: ("T-cell" OR "T cells" OR "T-lymphocyte*") AND ("cochlea*" OR "spiral ligament" OR "spiral limbus"). RESULTS The literature search revealed 20 studies that explored the presence and activity of T cells in the cochlea, as well as associations between T cells and hearing loss. The presence of cochlear T cells was compared between steady-state conditions and stimulated environments, which suggested an increase in cochlear T cells post-stimulation. Additionally, the role of T cells in hearing loss, both causal as protective, are described in 12 studies. Finally, three studies introduce cochlin as an inner ear-specific antigen triggering autoimmunity. CONCLUSION This review highlights the critical role of the immune balance in maintaining cochlear homeostasis. Both protective and detrimental T-cell functions have been linked to hearing, reflecting the dual role of T cells in cochlear health. Future therapies for hearing loss should aim to restore the immune balance to support normal hearing functions.
Collapse
Affiliation(s)
- Evi De Backer
- Resonant labs Antwerp, Department of Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Department of Otorhinolaryngology and Head & Neck Surgery, Antwerp University Hospital, Antwerp, Belgium; Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium.
| | - Dorien Verdoodt
- Resonant labs Antwerp, Department of Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Emanuela Pasciuto
- Laboratory of Neuroimmunology, VIB-Center for Molecular Neurology (CMN), Antwerp, Belgium
| | - Vincent Van Rompaey
- Resonant labs Antwerp, Department of Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Department of Otorhinolaryngology and Head & Neck Surgery, Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
22
|
Somay K, Albayrak Ö, Kızılırmak AB, Akan T, Üre ÜB, Akay OM, Ferhanoğlu B, Ateşoğlu EB. T cell subgroup analysis and T cell exhaustion after autologous stem cell transplantation in lymphoma patients. Transfus Apher Sci 2025; 64:104117. [PMID: 40222329 DOI: 10.1016/j.transci.2025.104117] [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: 05/21/2024] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Autologous stem cell transplantation (ASCT) is a common treatment option for relapsed/refractory (R/R) lymphomas and it is considered standard of care as primary consolidation therapy for some types of Non-Hodgkin Lymphomas (NHL). Although ASCT benefits patients by allowing cytoreduction with intensive chemotherapy and reconstituting with stem cells, the effects of immunological changes in T cell subgroups after ASCT are still poorly understood. OBJECTIVES We evaluated changes in frequencies of T cell subsets and T cells expressing some of the exhaustion markers (such as LAG-3 and PD-1) from peripheral blood samples before and after ASCT to investigate bone marrow reconstruction and whether exhaustion predicts relapse. STUDY DESIGN Blood samples were collected on the day before conditioning and at the 1st, 3rd, and 6th months post-ASCT. Flow cytometry analysis was conducted to examine T cell subgroup composition and exhaustion markers, including PD-1 and LAG-3. Additionally, functional analysis was performed using assays for IFN-g and TNF-a production. Furthermore, a CSFE proliferation assay was utilized to assess proliferation capacity. RESULTS In our data set, dominant cells post-transplantation were memory cells, as the naïve cell population did not recover for 6 months. Both single and combined expressions of LAG-3 and PD-1 were found to be high before transplantation, and decreased after transplantation. However, LAG-3 and PD-1 expression increased in the 3rd and 6th month after transplantation respectively. These changes were more evident for the relapsed patients when compared to non-relapsed patients within 3 months follow-up time. Notably, the expression of inhibitory receptors in the relapsed patients was significantly higher at the first month post-transplantation. CD107a+ cytotoxic T lymphocytes (CTL), IFN-g+, TNF-a.+ CTL and T helper lymphocyte (THL) populations significantly decreased in relapsed patients 3rd month after transplantation. Decreased proliferation capacities of CTLs and THLs were also observed in these patients. CONCLUSION These results suggest that increased surface PD-1 and LAG-3 expressions along with functional decline after 3 months of ASCT can be used as prognostic data about the relapse status of transplant patients.
Collapse
Affiliation(s)
- Kayra Somay
- Department of Internal Medicine, Koç University Hospital, Istanbul, Turkey
| | - Özgür Albayrak
- Koç University Research Center for Translational Medicine (KUTTAM) Koç University Hospital, Istanbul, Turkey.
| | - Ali Burak Kızılırmak
- Koç University Research Center for Translational Medicine (KUTTAM) Koç University Hospital, Istanbul, Turkey.
| | - Tuba Akan
- Department of Hematology, Koç University Hospital, Istanbul, Turkey.
| | - Ümit Barbaros Üre
- Department of Hematology, Koç University Hospital, Istanbul, Turkey.
| | - Olga Meltem Akay
- Department of Hematology, Koç University Hospital, Istanbul, Turkey.
| | | | | |
Collapse
|
23
|
Zhang H, Zhou Z, Wang J, Wang S, Ren J, Zhang M, Yang M. Adverse drug reaction assessment of pembrolizumab in cervical cancer treatment: a real-world pharmacovigilance study using the FAERS database. Front Immunol 2025; 16:1582050. [PMID: 40264768 PMCID: PMC12011867 DOI: 10.3389/fimmu.2025.1582050] [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: 02/23/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Objective Advanced cervical cancer remains associated with high mortality rates. While pembrolizumab has improved clinical outcomes in cervical cancer, the therapeutic efficacy in advanced stages is often compromised by immune-related adverse events (irAEs). This study aimed to systematically analyze pembrolizumab-associated adverse events (AEs) in cervical cancer using the FDA Adverse Event Reporting System (FAERS) database, providing new insights for optimizing clinical practice. Methods AE reports related to pembrolizumab in cervical cancer were extracted from the FAERS database (Q1 2016 to Q4 2024). Disproportionality analyses were performed using multiple algorithms, including the reporting odds ratio (ROR), proportional reporting ratio (PRR), Bayesian confidence propagation neural network (BCPNN), and multi-item gamma Poisson shrinker (MGPS). AEs were classified by system organ class (SOC) and preferred term (PT) based on the Medical Dictionary for Regulatory Activities (MedDRA), then ranked by frequency and signal strength. Results A total of 646 pembrolizumab-related AE reports in cervical cancer were identified. Age distribution peaked at 45-65 years cohort (32.75%), followed by 18-44 years (12.85%), 66-75 years (11.76%), and >75 years (4.64%). Among 270 AE reports with documented onset timelines, events predominantly occurred 3-6 months after pembrolizumab initiation (n=114, 41.36%). Clinical outcomes were categorized as other (52.80%), hospitalization (27.00%), death (10.25%), unknown (6.06%), life-threatening (2.77%), and disability (1.12%). Predominant AEs involved hematologic, endocrine, dermatologic, neurologic, gastrointestinal, urinary, and reproductive systems. Conclusion This real-world pharmacovigilance study systematically characterizes pembrolizumab-associated AEs in cervical cancer, identifying high-signal events such as hematologic disorders, endocrine dysfunction, and dermatologic toxicities. These findings provide critical evidence for risk stratification and safety monitoring in clinical practice, emphasizing the need for organ-specific vigilance during the 3-6 months treatment window.
Collapse
Affiliation(s)
- Huiping Zhang
- Department of Obstetrics and Gynecology, Northwest University First Hospital, Xi’an, Shaanxi, China
| | - Zhuo Zhou
- Department of Obstetrics and Gynecology, Northwest University First Hospital, Xi’an, Shaanxi, China
| | - Juan Wang
- Department of Obstetrics and Gynecology, Northwest University First Hospital, Xi’an, Shaanxi, China
| | - Shan Wang
- Department of Obstetrics and Gynecology, Northwest University First Hospital, Xi’an, Shaanxi, China
| | - Jie Ren
- Department of Obstetrics and Gynecology, Northwest University First Hospital, Xi’an, Shaanxi, China
| | - Ming Zhang
- Department of General Practice, Honghui Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Mingyi Yang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
24
|
Jiang K, Liu H, Chen X, Wang Z, Wang X, Gu X, Tong Y, Ba X, He Y, Wu J, Deng W, Wang Q, Tang K. Reprogramming of Glucose Metabolism by Nanocarriers to Improve Cancer Immunotherapy: Recent Advances and Applications. Int J Nanomedicine 2025; 20:4201-4234. [PMID: 40207307 PMCID: PMC11980946 DOI: 10.2147/ijn.s513207] [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: 12/19/2024] [Accepted: 03/20/2025] [Indexed: 04/11/2025] Open
Abstract
Although immunotherapy has made significant progress in cancer treatment, its limited responsiveness has greatly hindered widespread clinical application. The Warburg effect in tumor cells creates a tumor microenvironment (TME) characterized by hypoxia, low glucose levels, and high lactate levels, which severely inhibits the antitumor immune response. Consequently, targeting glucose metabolism to reprogram the TME is considered an effective strategy for reversing immunosuppression and immune evasion. Numerous studies have been conducted on enhancing cancer immunotherapy efficacy through the delivery of glucose metabolism modulators via nanocarriers. This review provides a comprehensive overview of the glucose metabolic characteristics of tumors and their impacts on the immune system, as well as nanodelivery strategies targeting glucose metabolism to enhance immunotherapy. These strategies include inhibiting key glycolytic enzymes, blocking glucose and lactate transporters, and utilizing glucose oxidase and lactate oxidase. Furthermore, this article reviews recent advancements in synergistic antitumor therapy involving glucose metabolism-targeted therapy combined with other treatments, such as chemotherapy, radiotherapy (RT), phototherapy, and immunotherapy. Finally, we discuss the limitations and future prospects of nanotechnology targeting glucose metabolism therapy, hoping to provide new directions and ideas to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Kehua Jiang
- Department of Urology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, People’s Republic of China
| | - Hongming Liu
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Xiaolong Chen
- Department of Urology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, People’s Republic of China
| | - Zhen Wang
- Department of Urology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, People’s Republic of China
| | - Xiaodong Wang
- Department of Urology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, People’s Republic of China
| | - Xiaoya Gu
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, People’s Republic of China
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| | - Jian Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| | - Wen Deng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| | - Qing Wang
- Department of Urology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, People’s Republic of China
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, People’s Republic of China
| |
Collapse
|
25
|
Arai Y, Miyai K, Hamamoto K, Furukawa Y, Asano T, Kobayashi H, Shinchi M, Tsujita Y, Kuroda K, Horiguchi A, Tsuda H, Ito K. Impact of tumor-infiltrating immune cells expressing PD-1 and those expressing PD-L1 on recurrence and prognosis in pathological T1b clear cell renal cell carcinoma. Jpn J Clin Oncol 2025:hyaf054. [PMID: 40183516 DOI: 10.1093/jjco/hyaf054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/21/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND The numbers of tumor-infiltrating immune cells (TIICs) expressing programmed death (PD)-1 or PD-ligand 1 (PD-L1) reportedly predict prognosis and resistance to targeted drugs in clear cell renal cell carcinoma (ccRCC). The impact of local tumor microenvironment based on immunosuppressive TIICs on recurrence and prognosis has not been fully investigated in localized ccRCC. METHODS A total of 105 patients with pT1b ccRCC were included. Immunostaining for PD-1 and PD-L1 were performed. PD-1-positive TIICs and PD-L1-positive TIICs were counted in the tumor periphery (TP) and the tumor nest (TN). RESULTS Patients with elevated PD-1-positive TIIC scores and those with elevated PD-L1-positive TIIC scores had significantly lower recurrence-free survival (RFS) rates than their counterparts (3-year RFS rates; patients with high vs. low PD-1-positive TIIC score of TN = 73.9% vs. 95.0%, those with high vs. low PD-1-positive TIIC score of TP = 73.8% vs. 93.8%, those with high vs. low PD-L1-positive TIIC score of TN = 70.9% vs. 93.0%, and those with high vs. low PD-L1-positive TIIC score of TP = 80.3% vs. 92.6%). Univariate analysis showed that high PD-1-positive scores, high PD-L1-positive scores, high PD-L1-positive tumor cell score, high-grade tumor, tumor necrosis, and lymphovascular invasion were significantly associated with RFS. Multivariate analysis revealed that tumor necrosis [hazard ratio (HR) = 2.841, P = .0269] and PD-1-positive TIIC score of TN (HR = 6.135, P = .0023) were independent risk factors for RFS. Risk stratification using the two factors efficiently predicts recurrence (3-year RFS rates: 96.4% with 0 factor, 83.8% with 1 factor, and 61.4% with 2 factors). CONCLUSION PD-1-positive TIIC score of TN and tumor necrosis may efficiently predict recurrence in pT1b ccRCC.
Collapse
Affiliation(s)
- Yuichi Arai
- Department of Urology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Kosuke Miyai
- Department of Basic Pathology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
- Department of Laboratory Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Koetsu Hamamoto
- Department of Urology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Yoshiyuki Furukawa
- Department of Urology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Takako Asano
- Department of Urology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Hiroaki Kobayashi
- Department of Urology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Masayuki Shinchi
- Department of Urology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Yujiro Tsujita
- Department of Urology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Kenji Kuroda
- Department of Urology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Akio Horiguchi
- Department of Urology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Hitoshi Tsuda
- Department of Basic Pathology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Keiichi Ito
- Department of Urology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| |
Collapse
|
26
|
Lai N, Farman A, Byrne HM. The Impact of T-cell Exhaustion Dynamics on Tumour-Immune Interactions and Tumour Growth. Bull Math Biol 2025; 87:61. [PMID: 40172752 PMCID: PMC11965189 DOI: 10.1007/s11538-025-01433-1] [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: 09/05/2024] [Accepted: 03/03/2025] [Indexed: 04/04/2025]
Abstract
Tumours evade immune surveillance through a number of different immunosuppressive mechanisms. One such mechanism causes cytotoxic T-cells, a major driving force of the immune system, to differentiate to a state of 'exhaustion', rendering them less effective at killing tumour cells. We present a structured mathematical model that focuses on T-cell exhaustion and its effect on tumour growth. We compartmentalise cytotoxic T-cells into discrete subgroups based on their exhaustion level, which affects their ability to kill tumour cells. We show that the model reduces to a simpler system of ordinary differential equations (ODEs) that describes the time evolution of the total number of T-cells, their mean exhaustion level and the total number of tumour cells. Numerical simulations of the model equations reveal how the exhaustion distribution of T-cells changes over time and how it influences the tumour's growth dynamics. Complementary bifurcation analysis shows how altering key parameters significantly reduces the tumour burden, highlighting exhaustion as a promising target for immunotherapy. Finally, we derive a continuum approximation of the discrete ODE model, which admits analytical solutions that provide complementary insight into T-cell exhaustion dynamics and their effect on tumour growth.
Collapse
Affiliation(s)
- Nicholas Lai
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, OX2 6GG, UK.
| | - Alexis Farman
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, OX2 6GG, UK
- Department of Mathematics, University College London, London, WC1E 6BT, UK
| | - Helen M Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, OX2 6GG, UK
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, OX3 7DQ, UK
| |
Collapse
|
27
|
Cao C, Lai H, Shi Y. MYCN/MNX1 Axis Drives NSCLC Progression by Inducing Macrophage M2 Polarization and CD8 + T Cell Apoptosis via the Wnt/β-Catenin Pathway. J Biochem Mol Toxicol 2025; 39:e70251. [PMID: 40226978 DOI: 10.1002/jbt.70251] [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] [Revised: 02/28/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025]
Abstract
Enhanced macrophage M2 polarization and CD8+ T cell dysfunction contribute to the pathophysiology of non-small cell lung cancer (NSCLC). Motor neuron and pancreatic homeobox 1 (MNX1) has emerged as a potential tumor-promoting player. Here, we clarified the activity of MNX1 in NSCLC. PMA-induced THP-1 M0-like macrophages or CD8+ T cells were co-cultured with NSCLC cells. Cell colony formation, migration, proliferation, apoptosis, and invasiveness were assessed by colony formation, wound healing, CCK-8, flow cytometry, and transwell assays, respectively. The ratio of CD206+ macrophages was analyzed by flow cytometry. Ki-67 expression was tested by immunofluorescence. ChIP and luciferase assays were used to evaluate the relationship between MYCN and MNX1. MNX1 was highly expressed in NSCLC, and its loss-of-function suppressed cell growth, motility, and invasiveness in NSCLC cells. MNX1 depletion also diminished macrophage M2 polarization and CD8+ T cell apoptosis. Mechanistically, MYCN increased MNX1 expression at the transcriptional level. MNX1 increase reversed the impact of MYCN depletion on NSCLC cell malignant behaviors, macrophage M2 polarization, and CD8+ T cell viability. MYCN depletion diminished the in vivo growth of A549 subcutaneous xenografts. Additionally, MNX1 increase counteracted the impact of MYCN depletion on the Wnt/β-catenin pathway. Our findings elucidate the oncogenic role of the MYCN/MNX1/Wnt/β-catenin pathway in NSCLC by driving macrophage M2 polarization and diminishing CD8+ T cell viability. Our study thus uncovers a novel mechanism underlying NSCLC development and highlights potential targets for combating NSCLC.
Collapse
Affiliation(s)
- Chengzhang Cao
- Department of Thoracic Surgery, Longyan First Affiliated Hospital of Fujian Medical Univensity, Longyan, China
| | - Haiyin Lai
- Department of Thoracic Surgery, Longyan First Affiliated Hospital of Fujian Medical Univensity, Longyan, China
| | - Yuzhen Shi
- Department of Critical Care Medicine, Longyan First Affiliated Hospital of Fujian Medical Univensity, Longyan, China
| |
Collapse
|
28
|
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
|
29
|
Otero JG, Belmonte-Beitia J, Jiménez-Sánchez J. Exploring neuroblastoma's cellular microenvironment: A novel approach using cellular automata to model Celyvir treatment. Comput Biol Med 2025; 188:109782. [PMID: 39946782 DOI: 10.1016/j.compbiomed.2025.109782] [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: 10/15/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 03/05/2025]
Abstract
Neuroblastoma is a significant health concern in children, as it is one of the most common types of cancer among this age group and is associated with poor survival rates. Currently, there are no effective therapies that significantly improve outcomes for these patients. This study explores the efficacy of Celyvir - an advanced therapy comprising mesenchymal stem cells (MSCs) carrying the oncolytic virus ICOVIR 5 - against neuroblastoma, by means of an individual-based model. A probabilistic cellular automaton was developed to implement the dynamic interactions between neuroblastoma cells, T lymphocytes, and the therapeutic agent Celyvir. The model examines various sizes, shapes, and positions of the tumour within a lattice, along with different infection probabilities associated with the action of Celyvir and various treatment schedules. This analysis identifies the most influential infection probabilities according to the cellular automaton model, and demonstrates that different treatment regimens can effectively eradicate the tumour, in contrast to standard clinical approaches. Additionally, Kaplan-Meier curves have been generated to assess different treatment schedules under specific tumour scenarios, highlighting the importance of precise treatment scheduling to optimise therapeutic outcomes. This study provides insights into the potential of Celyvir in neuroblastoma treatment, emphasising the need to understand tumour dynamics and strategically implement treatment schemes to improve clinical outcomes.
Collapse
Affiliation(s)
- José García Otero
- Mathematical Oncology Laboratory (MOLAB), University Castilla-La Mancha, Avda. Camilo José Cela s/n, Ciudad Real, 13071, Castilla-La Mancha, Spain.
| | - Juan Belmonte-Beitia
- Mathematical Oncology Laboratory (MOLAB), University Castilla-La Mancha, Avda. Camilo José Cela s/n, Ciudad Real, 13071, Castilla-La Mancha, Spain.
| | - Juan Jiménez-Sánchez
- Mathematical Oncology Laboratory (MOLAB), University Castilla-La Mancha, Avda. Camilo José Cela s/n, Ciudad Real, 13071, Castilla-La Mancha, Spain; Dipartimento di Scienze Matematiche Giuseppe Luigi Lagrange, Politecnico di Torino, Corso Duca degli Abruzzi, 24, 10129, Turin, Italy.
| |
Collapse
|
30
|
Lu X, Jin P, Tang Q, Zhou M, Xu H, Su C, Wang L, Xu F, Zhao M, Yin Y, Zhang J, Jia Z, Peng X, Zhou J, Wang L, Chen Y, Wang M, Yang M, Chen D, Chen Y. NAD + Metabolism Reprogramming Drives SIRT1-Dependent Deacetylation Inducing PD-L1 Nuclear Localization in Cervical Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412109. [PMID: 39988985 PMCID: PMC12005810 DOI: 10.1002/advs.202412109] [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: 09/29/2024] [Revised: 01/23/2025] [Indexed: 02/25/2025]
Abstract
Cervical cancer (CC) is a major health threat to women, with immunotherapies targeting the programmed death receptor 1/programmed death ligand 1(PD-1/PD-L1) axis showing promise but encountering resistance in a significant patient population. This resistance has driven a critical quest to uncover the underlying mechanisms. This study uncovers a novel metabolic axis involving the nicotinamide adenine dinucleotide (NAD+) salvage pathway enzyme nicotinamide phosphoribosyltransferase (NAMPT) and the deacetylase Sirtuin 1 (SIRT1), which regulates PD-L1 expression and nuclear localization in CC. This axis may be a key factor contributing to the resistance observed in immunotherapy. This study reveals that PD-L1 overexpression in cancers is regulated by both transcriptional and post-transcriptional processes. Acetyl-proteomic analysis pinpoints SIRT1 as a central regulator in the deacetylation of histone H3 at lysines 27, which may influence PD-L1 subcellular distribution. This finding reveals the epigenetic control of immune checkpoint proteins by metabolic pathways, offering a new perspective on the regulation of PD-L1. The identification of the NAMPT/SIRT1 metabolic axis as a critical factor suggests that targeting this axis may enhance therapeutic responses.
Collapse
Affiliation(s)
- Xinyi Lu
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
- Wuxi Medical CenterNanjing Medical UniversityJiangsu214023China
| | - Pingping Jin
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Qianyun Tang
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Min Zhou
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Hanjie Xu
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Chen Su
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Lei Wang
- Wuxi Medical CenterNanjing Medical UniversityJiangsu214023China
- Department of Hepatopancreatobiliary SurgeryJiangnan University Medical CenterJiangsu214002China
| | - Feng Xu
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Min Zhao
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Yongxiang Yin
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Jinqiu Zhang
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Zhen Jia
- Department of LaboratoryHaidong Second People's HospitalHaidong810699China
| | - Xinrui Peng
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Jie Zhou
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Lu Wang
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Yan Chen
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
| | - Min Wang
- Wuxi Medical CenterNanjing Medical UniversityJiangsu214023China
| | - Min Yang
- Molecular Imaging CentreJiangsu Institute of Nuclear MedicineJiangsu214063China
| | - Daozhen Chen
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
- Wuxi Medical CenterNanjing Medical UniversityJiangsu214023China
- Department of LaboratoryHaidong Second People's HospitalHaidong810699China
| | - Yu Chen
- Affiliated Women's Hospital of Jiangnan UniversityJiangnan UniversityJiangsu214002China
- Wuxi Medical CenterNanjing Medical UniversityJiangsu214023China
| |
Collapse
|
31
|
Ma Y, Wang Z, Qiu C, Xiao M, Wu S, Han K, Xu H, Wang H. Nomogram based on CT imaging and clinical data to predict the efficacy of PD-1 inhibitors combined with chemotherapy in advanced gastric cancer. Front Immunol 2025; 16:1504387. [PMID: 40230847 PMCID: PMC11994692 DOI: 10.3389/fimmu.2025.1504387] [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: 09/30/2024] [Accepted: 03/12/2025] [Indexed: 04/16/2025] Open
Abstract
Background PD-1 inhibitors, in combination with chemotherapy, have become the first-line treatment option for patients with advanced metastatic gastric cancer. However, some patients still do not benefit from this treatment, highlighting an urgent need for simple and reliable markers to predict the efficacy of immunotherapy. Methods Immunotherapy efficacy was evaluated using RECIST 1.1 and categorized into complete remission (CR), partial remission (PR), stable disease (SD), and disease progression (PD). Patients with CR, PR, and SD were classified as non-PD responders, while PD patients were categorized as PD responders. Clinical characteristics and CT imaging features of gastric cancer patients from two centers, before receiving PD-1 inhibitor combination chemotherapy, were retrospectively analyzed. A univariate logistic regression analysis was performed for each variable, and separate models for clinical and imaging characteristics, as well as a nomogram, were developed. Area under the curve (AUC), accuracy, sensitivity, specificity, and decision curve analysis (DCA) were used to evaluate all models. Results Data from 272 patients (non-PD responders = 206, PD responders = 66) from Center 1 were collected for this study. Data from 76 patients (non-PD responders = 54, PD responders = 22) from Center 2 were used as an external validation cohort to verify the robustness of the models. We developed a clinical model, an imaging features model, and a nomogram. The nomogram, combining clinical and imaging features, demonstrated superior performance with an AUC of 0.904 (95% CI: 0.862-0.947) in the training set and an AUC of 0.801 (95% CI: 0.683-0.918) in the validation set, with sensitivity, specificity, and accuracy of 0.889, 0.682, and 0.829, respectively. Calibration curves further confirmed the agreement between actual results and predictions. Conclusions A nomogram combining clinical features and CT imaging features before treatment was developed, which can effectively and simply predict the efficacy response of advanced gastric cancer patients treated with PD-1 inhibitors combined with chemotherapy. This tool can aid in optimizing treatment strategies in clinical practice.
Collapse
Affiliation(s)
- Yinchao Ma
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Radiology, Shandong First Medical University, Taian, Shandong, China
| | - Zhipeng Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Radiology, Shandong First Medical University, Taian, Shandong, China
| | - Chenyang Qiu
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Mengjun Xiao
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shuzhen Wu
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Kun Han
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Hui Xu
- Department of General Education, Shandong First Medical University, Jinan, Shandong, China
| | - Haiyan Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
32
|
Di Spirito A, Balkhi S, Vivona V, Mortara L. Key immune cells and their crosstalk in the tumor microenvironment of bladder cancer: insights for innovative therapies. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002304. [PMID: 40177538 PMCID: PMC11964778 DOI: 10.37349/etat.2025.1002304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Bladder cancer (BC) is a heterogeneous disease associated with high mortality if not diagnosed early. BC is classified into non-muscle-invasive BC (NMIBC) and muscle-invasive BC (MIBC), with MIBC linked to poor systemic therapy response and high recurrence rates. Current treatments include transurethral resection with Bacillus Calmette-Guérin (BCG) therapy for NMIBC and radical cystectomy with chemotherapy and/or immunotherapy for MIBC. The tumor microenvironment (TME) plays a critical role in cancer progression, metastasis, and therapeutic efficacy. A comprehensive understanding of the TME's complex interactions holds substantial translational significance for developing innovative treatments. The TME can contribute to therapeutic resistance, particularly in immune checkpoint inhibitor (ICI) therapies, where resistance arises from tumor-intrinsic changes or extrinsic TME factors. Recent advancements in immunotherapy highlight the importance of translational research to address these challenges. Strategies to overcome resistance focus on remodeling the TME to transform immunologically "cold" tumors, which lack immune cell infiltration, into "hot" tumors that respond better to immunotherapy. These strategies involve disrupting cancer-microenvironment interactions, inhibiting angiogenesis, and modulating immune components to enhance anti-tumor responses. Key mechanisms include cytokine involvement [e.g., interleukin-6 (IL-6)], phenotypic alterations in macrophages and natural killer (NK) cells, and the plasticity of cancer-associated fibroblasts (CAFs). Identifying potential therapeutic targets within the TME can improve outcomes for MIBC patients. This review emphasizes the TME's complexity and its impact on guiding novel therapeutic approaches, offering hope for better survival in MIBC.
Collapse
Affiliation(s)
- Anna Di Spirito
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Sahar Balkhi
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Veronica Vivona
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| |
Collapse
|
33
|
Zhou J, Zhang Y, Liu Y, Li J, Zhang W, Wang J, Yao X, Feng H, Zheng J, Li Y. Integrative analysis of bulk and single-cell sequencing reveals TNFSF9 as a potential regulator in microsatellite instability stomach adenocarcinoma. Eur J Med Res 2025; 30:214. [PMID: 40148957 PMCID: PMC11951761 DOI: 10.1186/s40001-025-02471-0] [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: 05/21/2024] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Stomach adenocarcinoma (STAD) with microsatellite instability (MSI) is associated with a better prognosis compared to Non-MSI. This study aims to elucidate the differences in the tumor microenvironment (TME) of MSI and explore its underlying mechanisms in STAD. METHODS TME differences between MSI and Non-MSI were analyzed using single-cell RNA sequencing (MSI = 7, Non-MSI = 19) and bulk RNA sequencing (MSI = 39, Non-MSI = 198). Differentially expressed genes (DEGs) were used to identify enriched pathways and hub genes. TNFSF9 expression was validated by immunohistochemistry (IHC) on 23 STAD sections (MSI = 13, Non-MSI = 10) and confirmed in tumor epithelial cells using SNU-1 (MSI) and AGS (Non-MSI) cell lines through quantitative polymerase chain reaction (qPCR) and Western blot (WB). RESULTS The results showed MSI was significantly associated with a better prognosis (P < 0.05). Within the TME, MSI was associated with a higher abundance of antigen-presenting cells, including M1 macrophages (40.1% vs. 27.9%) and activated dendritic cells (22.1% vs. 10.5%), as well as pro-inflammatory Th1-like CD4⁺ T cells (15% vs. 11%). However, MSI also showed an increase in exhausted T cells, indicating a complex immune landscape. Signaling pathway and cell communication analyses revealed an enrichment of cytokine-related pathways in MSI. Hub gene analysis revealed that TNFSF9 was predominantly expressed in stromal cells and partially in tumor epithelial cells in MSI, with its upregulation further confirmed through IHC, qPCR, and WB. Correlation analysis demonstrated a positive relationship between TNFSF9 expression and the abundance of M1 macrophages. CONCLUSIONS These findings provide new insights into the TME of MSI in STAD, emphasizing the significant role of TNFSF9 in shaping MSI-specific TME, enhancing immunotherapy efficacy, and improving patient survival.
Collapse
Affiliation(s)
- Jianlong Zhou
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Yucheng Zhang
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Yongfeng Liu
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Jiehui Li
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Wenxing Zhang
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Junjiang Wang
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Xueqing Yao
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
- Department of General Surgery, Guangdong Provincial People's Hospital Ganzhou Hospital (Ganzhou Municipal Hospital), Ganzhou, 341000, China
| | - Huolun Feng
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China.
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China.
| | - Jiabin Zheng
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China.
| | - Yong Li
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China.
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China.
| |
Collapse
|
34
|
Xu Q, Li L, Zhu R. T Cell Exhaustion in Allergic Diseases and Allergen Immunotherapy: A Novel Biomarker? Curr Allergy Asthma Rep 2025; 25:18. [PMID: 40091122 DOI: 10.1007/s11882-025-01199-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE OF REVIEW This review explores the emerging role of T cell exhaustion in allergic diseases and allergen immunotherapy (AIT). It aims to synthesize current knowledge on the mechanisms of T cell exhaustion, evaluate its potential involvement in allergic inflammation, and assess its implications as a novel biomarker for predicting and monitoring AIT efficacy. RECENT FINDINGS Recent studies highlight that T cell exhaustion, characterized by co-expression of inhibitory receptors (e.g., PD-1, CTLA-4, TIM-3), diminished cytokine production, and altered transcriptional profiles, may suppress type 2 inflammation in allergic diseases. In allergic asthma, exhausted CD4 + T cells exhibit upregulated inhibitory receptors, correlating with reduced IgE levels and airway hyperreactivity. During AIT, prolonged high-dose allergen exposure drives allergen-specific Th2 and T follicular helper (Tfh) cell exhaustion, potentially contributing to immune tolerance. Notably, clinical improvements in AIT correlate with depletion of allergen-specific Th2 cells and persistent expression of exhaustion markers (e.g., PD-1, CTLA-4) during maintenance phases. Blockade of inhibitory receptors (e.g., PD-1) enhances T cell activation, underscoring their dual regulatory role in allergy. T cell exhaustion represents a double-edged sword in allergy: it may dampen pathological inflammation in allergic diseases while serving as a mechanism for AIT-induced tolerance. The co-expression of inhibitory receptors on allergen-specific T cells emerges as a promising biomarker for AIT efficacy. Future research should clarify the transcriptional and metabolic drivers of exhaustion in allergy, validate its role across diverse allergic conditions, and optimize strategies to harness T cell exhaustion for durable immune tolerance. These insights could revolutionize therapeutic approaches and biomarker development in allergy management.
Collapse
Affiliation(s)
- Qingxiu Xu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Le Li
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rongfei Zhu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
35
|
Zhuang W, Pan K, Wu J, Liu L, Lv S, Hu J, Shi F, Zhao W, Yu D. Harnessing the power of traceable system C-GAP: homologous-targeting to fire up T-cell immune responses with low-dose irradiation. J Nanobiotechnology 2025; 23:207. [PMID: 40075499 PMCID: PMC11905511 DOI: 10.1186/s12951-025-03281-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
While radiotherapy-induced immunogenic cell death (ICD) holds potential for enhancing cancer immunotherapy, the conventional high-dose irradiation often leads to an immunosuppressive microenvironment and systemic toxicity. Therefore, a biomimetic nanoplatform cell membrane coated-nitrogen-doped graphene quantum dots combined with Au nanoparticles (C-GAP) was developed in this study. Firstly, homologous and traceable targeting features of C-GAP enables tumor-selective accumulation, providing reference for the selection of the timing of radiotherapy. Secondly, radiosensitization by C-GAP with Low-dose irradiation (LDI) amplifies reactive oxygen species (ROS) generation to trigger potent ICD. Thirdly, remarkable immune remodeling induced by C-GAP enhances CD8+ T cell infiltration and effector function. Single-cell RNA sequencing revealed that C-GAP-LDI combination upregulates TNF and CCL signaling pathway expression in tumor-infiltrating CD8+ T cells which potentiates tumor eradication. Our findings present a novel approach for safe and effective radioimmunotherapy, where C-GAP sensitized LDI achieves therapeutic enhancement through precise ICD induction and systemic immune activation.
Collapse
Affiliation(s)
- Weijie Zhuang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China
| | - Kuangwu Pan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China
- Department of Stomatology, The Third People's Hospital of Chengdu, Sichuan, China
| | - Jie Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China
| | - Leyi Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China
| | - Shiyu Lv
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China
| | - Jiajun Hu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China
| | - Fangyang Shi
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China
| | - Wei Zhao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China.
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China.
| | - Dongsheng Yu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China.
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510050, China.
| |
Collapse
|
36
|
Engelbrecht E, Stamp BF, Chew L, Sarkar OS, Harter P, Waigel SJ, Rouchka EC, Chariker J, Smolenkov A, Chesney J, McMasters K, Watson CT, Yaddanapudi K. Single-cell transcriptomics of melanoma sentinel lymph nodes identifies immune cell signatures associated with metastasis. JCI Insight 2025; 10:e183080. [PMID: 40048259 PMCID: PMC11981627 DOI: 10.1172/jci.insight.183080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 02/25/2025] [Indexed: 04/09/2025] Open
Abstract
The sentinel lymph node (SLN) is the first lymph node encountered by a metastatic cancer cell and serves as a predictor of poor prognosis, as cases with clinically occult SLN metastases are classified as stage III with elevated rates of recurrence and diminished overall survival. However, the dynamics of immune infiltrates in SLNs remain poorly characterized. Here, using an unbiased cellular indexing of transcriptomes and epitopes by sequencing technique, we profiled 97,777 cells from SLN tissues obtained from patients with stages I/II and III cutaneous melanoma. We described the transcriptional programs of a multitude of T, B, and myeloid cell subtypes in SLNs. Based on the proportions of cell types, we determined that SLN subtypes stratified along a naive → activated axis; patients with a "high activated" signature score appeared to be undergoing a robust melanoma antigen-driven adaptive immune response and, thus, could be responsive to immunotherapy. Additionally, we identified transcriptomic signatures of SLN-infiltrating dendritic cell subsets that compromise antitumor immune responses. Our analyses provide valuable insights into tumor-driven immune changes in the SLN tissue, offering a powerful tool for the informed design of immune therapies for patients with high-risk melanoma.
Collapse
Affiliation(s)
| | | | - Lewis Chew
- Immuno-Oncology Group, UofL-Health Brown Cancer Center
- Department of Microbiology/Immunology
| | - Omar Sadi Sarkar
- Immuno-Oncology Group, UofL-Health Brown Cancer Center
- Department of Microbiology/Immunology
| | - Phillip Harter
- Immuno-Oncology Group, UofL-Health Brown Cancer Center
- Department of Microbiology/Immunology
| | | | - Eric C. Rouchka
- Department of Biochemistry and Molecular Genetics
- Department of Computer Science and Engineering
| | | | | | - Jason Chesney
- Immuno-Oncology Group, UofL-Health Brown Cancer Center
- UofL-Health Brown Cancer Center
| | | | | | - Kavitha Yaddanapudi
- Immuno-Oncology Group, UofL-Health Brown Cancer Center
- Department of Microbiology/Immunology
- Division of Immunotherapy, Department of Surgery, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
37
|
Pretto S, Yu Q, Bourdely P, Trusso Cafarello S, Van Acker HH, Verelst J, Richiardone E, Vanheer L, Roshanzadeh A, Schneppenheim F, Cresens C, Sassano ML, Dehairs J, Carion M, Ismail S, Agostinis P, Rocha S, Bald T, Swinnen J, Corbet C, Lunt SY, Thienpont B, Di Matteo M, Mazzone M. A functional single-cell metabolic survey identifies Elovl1 as a target to enhance CD8 + T cell fitness in solid tumours. Nat Metab 2025; 7:508-530. [PMID: 40065102 PMCID: PMC11946891 DOI: 10.1038/s42255-025-01233-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 02/04/2025] [Indexed: 03/28/2025]
Abstract
Reprogramming T cell metabolism can improve intratumoural fitness. By performing a CRISPR/Cas9 metabolic survey in CD8+ T cells, we identified 83 targets and we applied single-cell RNA sequencing to disclose transcriptome changes associated with each metabolic perturbation in the context of pancreatic cancer. This revealed elongation of very long-chain fatty acids protein 1 (Elovl1) as a metabolic target to sustain effector functions and memory phenotypes in CD8+ T cells. Accordingly, Elovl1 inactivation in adoptively transferred T cells combined with anti-PD-1 showed therapeutic efficacy in resistant pancreatic and melanoma tumours. The accumulation of saturated long-chain fatty acids in Elovl1-deficient T cells destabilized INSIG1, leading to SREBP2 activation, increased plasma membrane cholesterol and stronger T cell receptor signalling. Elovl1-deficient T cells increased mitochondrial fitness and fatty acid oxidation, thus withstanding the metabolic stress imposed by the tumour microenvironment. Finally, ELOVL1 in CD8+ T cells correlated with anti-PD-1 response in patients with melanoma. Altogether, Elovl1 targeting synergizes with anti-PD-1 to promote effective T cell responses.
Collapse
Affiliation(s)
- Samantha Pretto
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Qian Yu
- Laboratory for Functional Epigenetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Pierre Bourdely
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Sarah Trusso Cafarello
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Heleen H Van Acker
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Joren Verelst
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Elena Richiardone
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Lotte Vanheer
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Amir Roshanzadeh
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Franziska Schneppenheim
- Institute of Experimental Oncology (IEO), University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Charlotte Cresens
- Molecular Imaging and Photonics Division, Chemistry Department, Faculty of Sciences, KU Leuven, Heverlee, Belgium
- VIB BioImaging Core, Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Maria Livia Sassano
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Martin Carion
- Department of Chemistry, KU Leuven, Heverlee, Belgium
| | - Shehab Ismail
- Department of Chemistry, KU Leuven, Heverlee, Belgium
| | - Patrizia Agostinis
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Susana Rocha
- Molecular Imaging and Photonics Division, Chemistry Department, Faculty of Sciences, KU Leuven, Heverlee, Belgium
| | - Tobias Bald
- Institute of Experimental Oncology (IEO), University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Johan Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Sophia Y Lunt
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, USA
| | - Bernard Thienpont
- Laboratory for Functional Epigenetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Mario Di Matteo
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium.
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
38
|
Wang Z, Dai Y, Zhou Y, Wang Y, Chen P, Li Y, Zhang Y, Wang X, Hu Y, Li H, Li G, Jing Y. Research progress of T cells in cholangiocarcinoma. Front Immunol 2025; 16:1453344. [PMID: 40070825 PMCID: PMC11893616 DOI: 10.3389/fimmu.2025.1453344] [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: 06/23/2024] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Cholangiocarcinoma (CCA), a malignant tumor, is typically challenging to detect early and often results in a poor prognosis. In recent years, research interest has grown in the potential application of immunotherapy for CCA treatment. T cells, as a crucial component of the immune system, play a significant role in immune surveillance and therapy for cholangiocarcinoma. This article provides a review of the research advancements concerning T cells in cholangiocarcinoma patients, including their distribution, functional status, and correlation with patient prognosis within the tumor microenvironment. It further discusses the potential applications and challenges of immunotherapy strategies targeting T cells in CCA treatment and anticipates future research directions. A more profound understanding of T cells' role in cholangiocarcinoma can guide the development of clinical treatment strategies, thereby enhancing patient survival rates and quality of life. Finally, we explored the potential risks and side effects of immunotherapy for T-cell cholangiocarcinoma.
Collapse
Affiliation(s)
- Zhiming Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yunyan Dai
- 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
| | - Pinggui Chen
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yaoxuan Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yunfei Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Xiaocui Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Ying Hu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Haonan Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Gaopeng Li
- Department of Hepatobiliary Surgery, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yukai Jing
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
39
|
LI W, ZU L, XU S. [FCN3 Can Serve as A Potential Biomarker for Prognosis and
Immunotherapy of Lung Squamous Cell Carcinoma]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2025; 28:114-130. [PMID: 40114488 PMCID: PMC11931238 DOI: 10.3779/j.issn.1009-3419.2025.105.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Indexed: 03/22/2025]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths worldwide. Lung squamous cell carcinoma (LUSC) is an important pathological subtype of NSCLC. The complex immune escape mechanism limits the effectiveness of immunotherapy. Ficolin-3 (FCN3) is a crucial immunomodulatory molecule that regulates immune escape by remodeling the tumor microenvironment. However, the role of FCN3 in LUSC remains unclear. This study employed bioinformatics methods to analyze LUSC samples from The Cancer Genome Atlas (TCGA) database. The aim of this study was to explore the potential biological functions and prognostic significance of FCN3 in LUSC. METHODS A pan-cancer analysis characterized the expression patterns and prognostic value of FCN3 across various cancer types. Simultaneously, the expression patterns of FCN3 in LUSC samples from the TCGA database and its relationship with prognosis were analyzed. The Nomogram model and somatic mutation analysis, differential expression analysis, correlation analysis, as well as Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) were constructed to explore the potential mechanisms of FCN3. Additionally, immune infiltration analysis, immune escape score (TIDE), and correlation analysis of immune-related molecules were used to reveal the regulatory role of high FCN3 levels on immunity in LUSC. Furthermore, the correlation between FCN3 expression characteristics and drug sensitivity was evaluated. Finally, in vitro experiments verified the expression characteristics of FCN3 in LUSC. RESULTS The expression level of FCN3 in LUSC tissues was significantly lower than that in normal tissues. Patients with high FCN3 expression in LUSC had a poorer prognosis compared to those with low expression. Different expression levels of FCN3 were associated with the abundance of immune cell infiltration and immune cell dysfunction. It was also linked to the expression of immune checkpoints, immune stimulatory molecules, major histocompatibility complex (MHC) class molecules, and chemotherapy drug sensitivity. CONCLUSIONS High expression of FCN3 in LUSC is associated with poor prognosis and is linked to immune cell infiltration, immune-related pathways, and immune-related molecules. FCN3 may be a potential prognostic marker and a new target for immunotherapy in LUSC.
Collapse
|
40
|
Shi Y, Zhang J, Li Y, Feng C, Shao C, Shi Y, Fang J. Engineered mesenchymal stem/stromal cells against cancer. Cell Death Dis 2025; 16:113. [PMID: 39971901 PMCID: PMC11839947 DOI: 10.1038/s41419-025-07443-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/03/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025]
Abstract
Mesenchymal stem/stromal cells (MSCs) have garnered attention for their potential in cancer therapy due to their ability to home to tumor sites. Engineered MSCs have been developed to deliver therapeutic proteins, microRNAs, prodrugs, chemotherapy drugs, and oncolytic viruses directly to the tumor microenvironment, with the goal of enhancing therapeutic efficacy while minimizing off-target effects. Despite promising results in preclinical studies and clinical trials, challenges such as variability in delivery efficiency and safety concerns persist. Ongoing research aims to optimize MSC-based cancer eradication and immunotherapy, enhancing their specificity and efficacy in cancer treatment. This review focuses on advancements in engineering MSCs for tumor-targeted therapy.
Collapse
Affiliation(s)
- Yuzhu Shi
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jia Zhang
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
- Department of Basic Medical Sciences, Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Yanan Li
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Chao Feng
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Changshun Shao
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Yufang Shi
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China.
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200025, China.
| | - Jiankai Fang
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
41
|
Zhang L, Guo X, Sun X, Liao J, Liu Q, Ye Y, Yang Z, Cressey R, He Q, Yuan Q. Analysis of tumor-infiltrating exhausted T cells highlights IL-6 and PD1 blockade as a combined immunotherapy strategy for non-small cell lung cancer. Front Immunol 2025; 16:1486329. [PMID: 40040705 PMCID: PMC11876966 DOI: 10.3389/fimmu.2025.1486329] [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: 08/26/2024] [Accepted: 01/20/2025] [Indexed: 03/06/2025] Open
Abstract
Objective Given the limitations of immunotherapy for treating non-small cell lung cancer (NSCLC), we investigated the phenotype and function of exhausted CD8+T cells and analyzed a novel combination immunotherapy to restore the effector killing function of tumor-infiltrating CD8+T lymphocyte (TIL). Methods We examined the expression and function of immunosuppressive molecules on CD8+T cells of peripheral blood mononuclear cells (PBMCs) and TILs by using prospectively collected peripheral blood, pleural effusions, and tumor tissues from patients with NSCLC and correlated the results with clinical data. We then evaluated the effect of interleukin 6 (IL-6) stimulation on CD8+T cells. Finally, we assessed the effects of combined blockade of PD1 and IL-6 on macrophage recruitment in a zebrafish macrophage model and CD8+ T cell function and tumor growth in PBMC humanized mouse model. Results The expression of exhaustion markers on CD8+ T cells was found to be notably higher in both tumor and paraneoplastic tissues compared to peripheral blood. Furthermore, the degree of CD8+ T cell exhaustion exhibited a progressive increase with proximity to the tumor. When CD8+ T cells from peripheral blood and tumor tissues of NSCLC patients were stimulated with IL-6, the expression level of exhaustion markers, especially PD1, was further elevated. In the in vitro experiment, the combined inhibition of IL-6 and PD1 substantially enhanced the effector killing function of CD8+ T cells in NSCLC pleural effusion samples. In a macrophage-labeled zebrafish model, combined blockade of IL-6 and PD1 enhanced the recruitment of macrophages. In PBMC humanized mouse model, combined blockade of IL-6 and PD1 enhanced the inhibition of tumor growth. Conclusion Our data suggest that CD8+ T cells in NSCLC patients were in a state of exhaustion and combined blockade of IL-6 and PD1 to restore CD8+ T cell function to inhibit tumor growth may be an effective clinical strategy for the treatment of NSCLC.
Collapse
Affiliation(s)
- Lulu Zhang
- Public Center of Experimental Technology, The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
- Blood Distribution Department Nanjing Red Cross Blood Center, Nanjing, Jiangsu, China
| | - Xiyuan Guo
- Public Center of Experimental Technology, The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
- Division of Clinical Chemistry, Department of Medical Technology, Faculty of Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Xiaoke Sun
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Jue Liao
- Public Center of Experimental Technology, The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Qin Liu
- Public Center of Experimental Technology, The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Yingchun Ye
- Public Center of Experimental Technology, The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhihui Yang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ratchada Cressey
- Division of Clinical Chemistry, Department of Medical Technology, Faculty of Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Qing He
- Department of Head and Neck Oncology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Qing Yuan
- Public Center of Experimental Technology, The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
- Institute of Nuclear Medicine, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
42
|
Chen MH, Jiang J, Chen H, Wu RH, Xie W, Dai SZ, Zheng WP, Tan GH, Huang FY. Reinforcing cancer immunotherapy with engineered porous hollow mycobacterium tuberculosis loaded with tumor neoantigens. J Immunother Cancer 2025; 13:e010150. [PMID: 39915006 PMCID: PMC11804190 DOI: 10.1136/jitc-2024-010150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 01/20/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Enhancing antigen cross-presentation is essential for the development of a tumor neoantigen vaccine. One approach is to stimulate antigen-presenting cells (APCs) to uptake neoantigens. Mycobacterium tuberculosis (MTb) contains pathogen-associated molecular patterns (PAMPs) recognized by APCs and adhesion molecules that facilitate MTb invasion of APCs. Therefore, we suggest using MTb as a carrier to enhance APC phagocytosis of neoantigens, thereby promoting antigen cross-presentation. METHODS The successful preparation of the MTb carrier (phMTb) was confirmed through electron and confocal microscopy. Fluorescence microscopy was used to detect PAMPs and adhesion molecules on phMTb as well as to observe its role in aiding dendritic cells (DCs) in antigen uptake into endosomes or lysosomes. Flow cytometry was used to assess the retention of PAMPs and adhesion molecules on phMTb, investigate antigen uptake by DCs, evaluate their activation and maturation status, examine the presentation of tumor neoantigens, and analyze immune cells in draining lymph nodes and tumor tissues. The efficacy of phMTb vaccine formulations in combination with anti-programmed cell death protein 1 (PD-1) antibody therapy was assessed using the MC38 mouse tumor models. Adverse effects were evaluated through H&E staining of major organs, assessment of reproductive capability and detection of biochemical indices. RESULTS The engineered porous hollow phMTb carrier successfully encapsulated model tumor neoantigens, with or without the adjuvant CpG. The phMTb retained PAMPs and adhesion molecules on its surface, similar to the parental MTb, thereby enhancing DC uptake of phMTb and its formulations containing tumor neoantigens and CpG. Vaccines formulated with phMTb facilitated DC maturation, activation, cross-presentation of tumor neoantigens, and promoted migration of phMTb-laden DCs to lymph nodes, enhancing effector and memory CD8+ T lymphocyte function. In murine tumor models, immunization with phMTb-formulated neoantigen vaccines elicited a robust tumor-specific cytotoxic T lymphocyte immune response with minimal adverse effects. Additionally, vaccination with phMTb-formulated neoantigen vaccines effectively reversed the tumor's immune-suppressive microenvironment. Concurrent administration of the PD-1 antibody with the phMTb-formulated neoantigen vaccine exhibited significant synergistic therapeutic effects. CONCLUSIONS The results of our study highlight the potential clinical translation of personalized tumor neoantigen vaccines using the phMTb carrier.
Collapse
Affiliation(s)
- Ming-Hui Chen
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Jie Jiang
- Public Research Center, Hainan Medical University, Haikou, Hainan, China
| | - Hengyu Chen
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Ri-Hong Wu
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Weijing Xie
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Shu-Zhen Dai
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Wu-Ping Zheng
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Guang-Hong Tan
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Feng-Ying Huang
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
43
|
Ishiguro S, Devader S, Blake C, Glover L, Upreti D, Nakashima A, Suzuki K, Comer J, Tamura M. A combination treatment with a water extract from Euglena gracilis and anti-PD-1 antibody strongly inhibits growth of lung cancer in mice through stimulating tumor-infiltrating lymphocytes. Int Immunopharmacol 2025; 147:113953. [PMID: 39809104 DOI: 10.1016/j.intimp.2024.113953] [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: 06/20/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025]
Abstract
Here, we investigated the relationship between the attenuation of lung cancer growth due to oral administration of Euglena gracilis water extract (EWE) and T cell stimulation. Orally administered EWE was revealed to increase PD-1 and PD-L1 mRNA and proteins primarily in tumor-infiltrating lymphocytes (TILs), which was correlated with a significant decrease in the tumor weights in mice. A combination treatment with EWE and anti-PD-1 antibody significantly decreased the growth of murine lung tumors more than treatment with either alone by increasing the number of TILs and attenuating T cell exhaustion. Short-chain fatty acids, which were previously shown to be increased in intestines of mice treated with oral EWE, increased both PD-1 and PD-L1 expression in splenocytes, but not in lung cancer cells in cell culture. These results suggest there is a close relationship between the EWE-induced increase of short-chain fatty acids, the increase of PD-1 expression in TILs, and the attenuation of lung tumor growth. Furthermore, EWE enhances the efficacy of anti-PD-1 antibody-based immune checkpoint blockade therapy against non-small cell lung cancer.
Collapse
Affiliation(s)
- Susumu Ishiguro
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, USA
| | - Sarah Devader
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, USA
| | - Caden Blake
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, USA
| | - Logan Glover
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, USA
| | - Deepa Upreti
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, USA
| | | | - Kengo Suzuki
- Euglena Co. Ltd., Minato-ku, Tokyo 108-0014, Japan
| | - Jeffrey Comer
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, USA
| | - Masaaki Tamura
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, USA.
| |
Collapse
|
44
|
Zeng Z, Zhang T, Zhang J, Li S, Connor S, Zhang B, Zhao Y, Wilson J, Singh D, Kulikauskas R, Church CD, Pulliam TH, Jani S, Nghiem P, Topalian SL, Forde PM, Pardoll DM, Ji H, Smith KN. A minimal gene set characterizes TIL specific for diverse tumor antigens across different cancer types. Nat Commun 2025; 16:1070. [PMID: 39900903 PMCID: PMC11791090 DOI: 10.1038/s41467-024-55059-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 11/27/2024] [Indexed: 02/05/2025] Open
Abstract
Identifying tumor-specific T cell clones that mediate immunotherapy responses remains challenging. Mutation-associated neoantigen (MANA) -specific CD8+ tumor-infiltrating lymphocytes (TIL) have been shown to express high levels of CXCL13 and CD39 (ENTPD1), and low IL-7 receptor (IL7R) levels in many cancer types, but their collective relevance to T cell functionality has not been established. Here we present an integrative tool to identify MANA-specific TIL using weighted expression levels of these three genes in lung cancer and melanoma single-cell RNAseq datasets. Our three-gene "MANAscore" algorithm outperforms other RNAseq-based algorithms in identifying validated neoantigen-specific CD8+ clones, and accurately identifies TILs that recognize other classes of tumor antigens, including cancer testis antigens, endogenous retroviruses and viral oncogenes. Most of these TIL are characterized by a tissue resident memory gene expression program. Putative tumor-reactive cells (pTRC) identified via MANAscore in anti-PD-1-treated lung tumors had higher expression of checkpoint and cytotoxicity-related genes relative to putative non-tumor-reactive cells. pTRC in pathologically responding tumors showed distinguished gene expression patterns and trajectories. Collectively, we show that MANAscore is a robust tool that can greatly enrich candidate tumor-specific T cells and be used to understand the functional programming of tumor-reactive TIL.
Collapse
Affiliation(s)
- Zhen Zeng
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, US
- Mark Center for Advanced Genomics and Imaging, Baltimore, MD, US
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, US
| | - Tianbei Zhang
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, US
- Mark Center for Advanced Genomics and Imaging, Baltimore, MD, US
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, US
| | - Jiajia Zhang
- David Geffen School of Medicine, University of California, Los Angeles, CA, US
| | - Shuai Li
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, US
| | - Sydney Connor
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, US
- Mark Center for Advanced Genomics and Imaging, Baltimore, MD, US
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, US
| | - Boyang Zhang
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, US
| | - Yimin Zhao
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, US
- Department of Biostatistics, University of Washington, Seattle, WA, US
| | - Jordan Wilson
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, US
| | - Dipika Singh
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, US
- Mark Center for Advanced Genomics and Imaging, Baltimore, MD, US
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, US
| | - Rima Kulikauskas
- Fred Hutchinson Cancer Center, Seattle, WA, US
- Department of Medicine, University of Washington, Seattle, WA, US
| | - Candice D Church
- Fred Hutchinson Cancer Center, Seattle, WA, US
- Department of Medicine, University of Washington, Seattle, WA, US
| | - Thomas H Pulliam
- Fred Hutchinson Cancer Center, Seattle, WA, US
- Department of Medicine, University of Washington, Seattle, WA, US
| | - Saumya Jani
- Fred Hutchinson Cancer Center, Seattle, WA, US
- Department of Medicine, University of Washington, Seattle, WA, US
| | - Paul Nghiem
- Fred Hutchinson Cancer Center, Seattle, WA, US
- Department of Medicine, University of Washington, Seattle, WA, US
| | - Suzanne L Topalian
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, US
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, US
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, US
| | - Patrick M Forde
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, US
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, US
| | - Drew M Pardoll
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, US
- Mark Center for Advanced Genomics and Imaging, Baltimore, MD, US
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, US
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, US
| | - Kellie N Smith
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, US.
- Mark Center for Advanced Genomics and Imaging, Baltimore, MD, US.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, US.
| |
Collapse
|
45
|
Dong C, Zhao Y, Han Y, Li M, Wang G. Targeting glutamine metabolism crosstalk with tumor immune response. Biochim Biophys Acta Rev Cancer 2025; 1880:189257. [PMID: 39746457 DOI: 10.1016/j.bbcan.2024.189257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 12/23/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Glutamine, akin to glucose, is a fundamental nutrient for human physiology. Tumor progression is often accompanied by elevated glutamine consumption, resulting in a disrupted nutritional balance and metabolic reprogramming within the tumor microenvironment. Furthermore, immune cells, which depend on glutamine for metabolic support, may experience functional impairments and dysregulation. Although the role of glutamine in tumors has been extensively studied, the specific impact of glutamine competition on immune responses, as well as the precise cellular alterations within immune cells, remains incompletely understood. In this review, we summarize the consequences of glutamine deprivation induced by tumor-driven glutamine uptake on immune cells, assessing the underlying mechanisms from the perspective of various components of the immune microenvironment. Additionally, we discuss the potential synergistic effects of glutamine supplementation and immunotherapy, offering insights into future research directions. This review provides compelling evidence for the integration of glutamine metabolism and immunotherapy as a promising strategy in cancer therapy.
Collapse
Affiliation(s)
- Chenshuang Dong
- Key Laboratory of Cell Biology, Department of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, China
| | - Yan Zhao
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yecheng Han
- Key Laboratory of Cell Biology, Department of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, China
| | - Ming Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| | - Guiling Wang
- Key Laboratory of Cell Biology, Department of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
46
|
Escalera-Anzola S, Rosado M, Yang Y, Parra-Sanchez D, Pedro-Liberal CS, Acedo P. Breakthroughs in nanoparticle-based strategies for pancreatic cancer therapy. Biochem Pharmacol 2025; 232:116685. [PMID: 39613113 DOI: 10.1016/j.bcp.2024.116685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/31/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers worldwide, mainly due to its high heterogeneity, resistance to therapy and late diagnosis, with a 5-year survival rate of less than 10%. This dismal prognosis has promoted strategies to develop more effective treatments. Nanoparticle-based strategies have emerged, in the last decades, as a great opportunity because they can enhance drug delivery and promote controlled release, presenting lower side effects than conventional therapeutic regimens. Moreover, nanoparticles can often be modified to target specific cells or to achieve a sustained release of the drugs into the tumor. However, very few nanoparticle-based therapies are clinically approved. Concretely for pancreatic cancer treatment only two nanoformulations have been approved by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) so far. Clinical translation of nanoparticles remains a challenge for modern medicine, and in particular for pancreatic cancer therapy, because of the complexity of the disease, and a lack of studies been performed in clinically relevant in vitro and in vivo models. In this review, we have summarized the most recent clinical trials using nanoparticle-based formulations in PDAC, giving a small context of the diverse types of nanoparticles employed and the most recent advancements in the field.
Collapse
Affiliation(s)
- Sara Escalera-Anzola
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom; Smart Devices for Nano Medicine Group, Unidad Excelencia Instituto de BioMedicina y Genética Molecular (IBGM) de Valladolid, University of Valladolid and CSIC, Valladolid, Spain
| | - Maria Rosado
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Yuchen Yang
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Daniel Parra-Sanchez
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Carolina San Pedro-Liberal
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Pilar Acedo
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom.
| |
Collapse
|
47
|
Li G, Wen Z, Xiong S. Microenvironmental β-TrCP negates amino acid transport to trigger CD8 + T cell exhaustion in human non-small cell lung cancer. Cell Rep 2025; 44:115128. [PMID: 39754718 DOI: 10.1016/j.celrep.2024.115128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 09/05/2024] [Accepted: 12/10/2024] [Indexed: 01/06/2025] Open
Abstract
CD8+ T cell exhaustion (Tex) has been widely acknowledged in human cancer, while the underlying mechanisms remain unclear. Here, we demonstrate that reduced amino acid (aa) metabolism and mTOR inactivation are accountable for Tex in human non-small cell lung cancer (NSCLC). NSCLC cells impede the T cell-intrinsic transcription of SLC7A5 and SLC38A1, disrupting aa transport and consequently leading to mTOR inactivation. Further, the ubiquitination of YAP1 protein is the basis for NSCLC-mediated transcriptional inhibition of aa transporters. Mechanistically, NSCLC cells transfer β-TrCP-containing exosomes into T cells, inducing YAP1 ubiquitination and Tex. Consequently, inhibiting cancer-associated β-TrCP effectively restores the anti-tumor immune response of CD8+ T cells and curtails tumor growth in NSCLC patient-derived organoids. Together, our findings highlight a β-TrCP-dependent mechanism in steering intrinsic metabolic adaptation and CD8+ Tex, emphasizing microenvironmental β-TrCP as an immune checkpoint for therapeutic exploration against human NSCLC.
Collapse
Affiliation(s)
- Ge Li
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Zhenke Wen
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China.
| | - Sidong Xiong
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China.
| |
Collapse
|
48
|
Park SY, Ju S, Lee J, Kim HR, Sub Y, Park DJ, Park S, Kwon D, Kang HG, Shin JE, Kim DH, Paik JE, Cho SC, Shim H, Kim YJ, Guan KL, Chun KH, Choi J, Ha SJ, Gee HY, Roe JS, Lee HW, Park SY, Park HW. Noncanonical role of Golgi-associated macrophage TAZ in chronic inflammation and tumorigenesis. SCIENCE ADVANCES 2025; 11:eadq2395. [PMID: 39841821 PMCID: PMC11753377 DOI: 10.1126/sciadv.adq2395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025]
Abstract
Until now, Hippo pathway-mediated nucleocytoplasmic translocation has been considered the primary mechanism by which yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) transcriptional coactivators regulate cell proliferation and differentiation via transcriptional enhanced associate domain (TEAD)-mediated target gene expression. In this study, however, we found that TAZ, but not YAP, is associated with the Golgi apparatus in macrophages activated via Toll-like receptor ligands during the resolution phase of inflammation. Golgi-associated TAZ enhanced vesicle trafficking and secretion of proinflammatory cytokines in M1 macrophage independent of the Hippo pathway. Depletion of TAZ in tumor-associated macrophages promoted tumor growth by suppressing the recruitment of tumor-infiltrating lymphocytes. Moreover, in a diet-induced metabolic dysfunction-associated steatohepatitis model, macrophage-specific deletion of TAZ ameliorated liver inflammation and hepatic fibrosis. Thus, targeted therapies being developed against YAP/TAZ-TEAD are ineffective in macrophages. Together, our results introduce Golgi-associated TAZ as a potential molecular target for therapeutic intervention to treat tumor progression and chronic inflammatory diseases.
Collapse
Affiliation(s)
- So Yeon Park
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul 03722, Republic of Korea
| | - Sungeun Ju
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Jaehoon Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul 03722, Republic of Korea
- Gemcro Inc., Seoul 03722, Republic of Korea
| | - Hwa-Ryeon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul 03722, Republic of Korea
| | - Yujin Sub
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Dong Jin Park
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul 03722, Republic of Korea
| | - Seyeon Park
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul 03722, Republic of Korea
| | - Doru Kwon
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Republic of Korea
| | - Hyeok Gu Kang
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji Eun Shin
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul 03722, Republic of Korea
| | - Dong Hyeon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul 03722, Republic of Korea
| | - Ji Eun Paik
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul 03722, Republic of Korea
| | - Seok Chan Cho
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul 03722, Republic of Korea
| | - Hyeran Shim
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul 03722, Republic of Korea
| | - Young-Joon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul 03722, Republic of Korea
| | - Kun-Liang Guan
- School of Life Sciences, Westlake University, Hangzhou 310030, China
| | - Kyung-Hee Chun
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Junjeong Choi
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Republic of Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul 03722, Republic of Korea
| | - Heon Yung Gee
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jae-Seok Roe
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul 03722, Republic of Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul 03722, Republic of Korea
- Gemcro Inc., Seoul 03722, Republic of Korea
| | - Seung-Yeol Park
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
49
|
Lian H, Zhang J, Hou S, Ma S, Yu J, Zhao W, Zhao D, Zhang Z. Immunotherapy of osteosarcoma based on immune microenvironment modulation. Front Immunol 2025; 15:1498060. [PMID: 39916962 PMCID: PMC11799554 DOI: 10.3389/fimmu.2024.1498060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/30/2024] [Indexed: 02/09/2025] Open
Abstract
Osteosarcoma is a highly malignant tumor with unsatisfactory therapeutic outcomes achieved by chemotherapy, radiotherapy, and surgery. As an emerging oncological treatment, immunotherapy has shown potential in the clinical management of many tumors but has a poor response rate in osteosarcoma. The immunosuppressive microenvironment in osteosarcoma is the main reason for the ineffectiveness of immunotherapy, in which the low immune response rate of immune effector cells and the high activation of immunosuppressive cells contribute to this outcome. Therefore, modulating the function of the immune microenvironment in osteosarcoma is expected to remodel the immunosuppressive microenvironment of osteosarcoma and enhance the efficacy of immunotherapy. This article reviews the role of immune cells in the progression of osteosarcoma, describes the corresponding regulatory tools for the characteristics of different cells to enhance the efficacy of osteosarcoma immunotherapy, and concludes the prospects and future challenges of osteosarcoma immunotherapy.
Collapse
Affiliation(s)
- Heping Lian
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, China
| | - Jiakui Zhang
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Shuna Hou
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Shuang Ma
- Nursing Department, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jiachen Yu
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Wei Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Duoyi Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhiyu Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
50
|
Lefèvre A, Parra-Guillen ZP, Trocóniz IF, Boetsch C, Frances N. Mechanistic PKPD modeling to describe cytokine release associated with CD3 T-cell engager therapies. Front Immunol 2025; 15:1463915. [PMID: 39896804 PMCID: PMC11782561 DOI: 10.3389/fimmu.2024.1463915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 12/19/2024] [Indexed: 02/04/2025] Open
Abstract
Introduction T-cell engagers (TCE), a therapeutic class of cancer immunotherapy (CIT), offer a novel approach to cancer treatment by harnessing and reactivating the patient's immune system to eradicate tumor cells. However, the use of TCE in the clinic can lead to severe side effects, including cytokine release syndrome (CRS). Therefore, innovative dosing strategies need to be implemented to mitigate the risk of developing CRS. Method In the presented work, a mechanistic pharmacokinetics/pharmacodynamics (PKPD) model describing cytokine release following TCE therapy has been developed combining literature knowledge and preclinical data. The model was developed to explore and test hypotheses regarding the mechanisms behind the decrease of cytokine release following two repeated TCE administrations. Results The model is able to successfully reproduce the observed dynamics of cytokine levels associated with the initial and subsequent TCE doses, accounting for different dosing intervals. In addition, the model suggests a mechanism of action that uncouples cytokine release from tumor cell killing. Discussion This model provides an initial mechanistic framework to support the design of experiments and paves the way for the application of mathematical modeling to support clinical dosing regimen selection of any TCE.
Collapse
Affiliation(s)
- Apolline Lefèvre
- Roche Pharma Research and Early Development (pRED), Pharmaceutical Sciences PS, Roche Innovation Center Basel, Basel, Switzerland
- Pharmacometrics & Systems Pharmacology, Department of Pharmaceutical Science, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Zinnia P. Parra-Guillen
- Pharmacometrics & Systems Pharmacology, Department of Pharmaceutical Science, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Iñaki F. Trocóniz
- Pharmacometrics & Systems Pharmacology, Department of Pharmaceutical Science, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Institute of Data Science and Artificial Intelligence (DATAI), University of Navarra, Pamplona, Spain
| | - Christophe Boetsch
- Roche Pharma Research and Early Development (pRED), Pharmaceutical Sciences PS, Roche Innovation Center Basel, Basel, Switzerland
| | - Nicolas Frances
- Roche Pharma Research and Early Development (pRED), Pharmaceutical Sciences PS, Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|