1
|
Zhao R, Zhu X, Wei W, Zhen L. The role of HSPA14 in breast cancer: implications for tumorigenesis, immune response modulation, and personalized therapies. Int J Hyperthermia 2025; 42:2452922. [PMID: 39828281 DOI: 10.1080/02656736.2025.2452922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/26/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Heat shock proteins have been implicated in the process of carcinogenesis. HSPA14, a member of the heat shock protein family, remains poorly understood in terms of its significance and pathomechanisms in breast cancer. METHODS We analyzed the expression levels of HSPA14 and its prognostic significance in breast cancer using TCGA data. TCGA data was used to investigate the association between HSPA14 expression and clinicopathological features in breast cancer patients. GSEA analysis was conducted to identify the biological function of HSPA14. Spearman's correlation analysis was performed to examine the correlation between HSPA14 expression and immune cell infiltration, as well as immune checkpoint genes. Single cell transcriptomic data from GSE114727 was utilized to calculate the expression of HSPA14 in different cell subpopulations. The data on HSPA14 levels and drug sensitivity were extracted from the CellMiner dataset. The mRNA expression of HSPA14 was validated through cell experiments. RESULTS HSPA14 expression is elevated in breast cancer, which is associated with poor overall survival. It can serve as a diagnostic biomarker for breast cancer patients. Pathway analysis revealed that HSPA14-associated differential genes are involved in cell cycle, apoptosis, cellular response to heat stress, and more. Additionally, HSPA14 expression is significantly correlated with the immune microenvironment. The expression of HSPA14 may also indicate drug sensitivity. CONCLUSION Our study elucidates the involvement of HSPA14 in tumorigenesis, particularly in modulating the immune response, shaping the immune microenvironment, and contributing to drug resistance, which are pivotal for the development of personalized breast cancer therapies.
Collapse
Affiliation(s)
- Ruipeng Zhao
- Department of Thyroid and Breast Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Xiaocun Zhu
- Department of Thyroid and Breast Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Wan Wei
- Department of Thyroid and Breast Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Linlin Zhen
- Department of Thyroid and Breast Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| |
Collapse
|
2
|
Gao D. The role of non-malignant B cells in malignant hematologic diseases. Hematology 2025; 30:2466261. [PMID: 39964954 DOI: 10.1080/16078454.2025.2466261] [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: 06/02/2024] [Accepted: 02/08/2025] [Indexed: 02/20/2025] Open
Abstract
The tumor microenvironment (TME) represents a heterogeneous, complicated ecosystem characterized by intricate interactions between tumor cells and immune cells. During the past decade, immune cells especially T cells were found to play an important role in the progression of tumor and many related immune checkpoints drugs were created. In recent years, more and more scientists revealed the critical role of B-cells within the TME, particularly various populations of non-malignant B cells. Some studies indicated that non-malignant B cells may exert a 'double-edged sword' role in solid tumors. However, there has been comparatively less focus on the role of non-malignant B cells in hematologic malignancies. In this review, we characterized the development of B cells and summarized its functions of antitumor immunity within TME, with an emphasis on elucidating the roles and potential mechanisms of non-malignant B cells in the progression of hematologic diseases including classical Hodgkin's lymphoma, non-Hodgkin's B-cell lymphoma, non-Hodgkin's T-cell lymphoma, leukemia and multiple myeloma.
Collapse
Affiliation(s)
- Daquan Gao
- Department of Hematology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, People's Republic of China
| |
Collapse
|
3
|
Quan Z, Fan S, Zheng H, Ning Y, Yang Y. A pan-cancer analysis of MARCH8: molecular characteristics, clinical relevance, and immuno-oncology features. Cancer Biol Ther 2025; 26:2458773. [PMID: 39881438 PMCID: PMC11784653 DOI: 10.1080/15384047.2025.2458773] [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: 10/16/2022] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025] Open
Abstract
Membrane-associated RING-CH8 (MARCH8) is a member of the recently discovered MARCH family of ubiquitin ligases. MARCH8 has been shown to participate in immune responses. However, the role of MARCH8 in prognosis and immunology in human cancers remains largely unknown. The expression of MARCH8 protein was detected via immunohistochemistry in non-small cell lung cancer (NSCLC) and non-cancerous lung tissues. The study investigated the role of MARCH8 in tumor immunity through pan-cancer analysis of multiple databases. MARCH8 genetic alternations and expression were explored with the cBioPortal, GTEx, and TCGA databases. We investigated the role of MARCH8 expression in clinical relevance, prognosis, tumor immune microenvironment, immune checkpoint (ICP) with a series of bioinformatics tools and methods, such as TISIDB database, ESTIMATE, and CIBERSORT method. MARCH8 expression showed cancer-specific dysregulation and was associated with the prognosis of patients in various cancers. MARCH8 was related to the tumor microenvironment and participated in tumor immune regulation. Furthermore, low expression of MARCH8 was associated with poor prognosis and might serve as an independent prognostic biomarker for NSCLC patients. The comprehensive pan-cancer analysis revealed the potential of MARCH8 in tumor-targeted therapy, and suggested MARCH8 as a promising prognostic and immunological pan-cancer biomarker.
Collapse
Affiliation(s)
- Zihan Quan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongmei Zheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yue Ning
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yang Yang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, The Fourth People’s Hospital of Longgang District, Shenzhen, Guangdong, China
| |
Collapse
|
4
|
Chen N, Li Z, Liu H, Jiang A, Zhang L, Yan S, He W, Yang J, Liu T. Enhancing PD-1 blockade in NSCLC: Reprogramming tumor immune microenvironment with albumin-bound statins targeting lipid rafts and mitochondrial respiration. Bioact Mater 2025; 49:140-153. [PMID: 40124597 PMCID: PMC11930202 DOI: 10.1016/j.bioactmat.2025.03.003] [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: 11/03/2024] [Revised: 02/09/2025] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) has shown limited response to immunotherapy, primarily due to an immunosuppressive tumor microenvironment characterized by hypoxia and lipid raft formation, which together inhibit T-cell infiltration and function, impeding effective immune responses. To address these challenges, we developed Abstatin, an albumin-bound fluvastatin formulation that targets lipid raft disruption and mitochondrial respiration inhibition, aiming to reduce hypoxia and destabilize lipid rafts to enhance T-cell activity within the tumor. Using bioinformatics analysis, in vitro assays, and in vivo studies in both murine and humanized PDX models, we demonstrated that Abstatin reprograms the NSCLC microenvironment by concurrently lowering hypoxia levels and lipid raft integrity, thereby restoring T-cell infiltration, enhancing cytotoxic T-cell function, and ultimately improving response to Anti-PD-1 therapy. Results showed that Abstatin significantly amplifies Anti-PD-1 efficacy with minimal toxicity, indicating a favorable safety profile for clinical use. This study highlights Abstatin as a promising immunotherapy adjuvant that addresses critical barriers in NSCLC by modulating metabolic pathways linked to immune resistance. Abstatin's approach, which combines modulation of cellular metabolism with immune sensitization, broadens the potential of immunotherapy and provides a practical, scalable strategy to enhance treatment outcomes in NSCLC and potentially other tumors, offering insights into combinatory cancer therapies.
Collapse
Affiliation(s)
- Na Chen
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Zhanfeng Li
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Heyuan Liu
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
- Department of Tumor and Immunology in Precision Medical Institute, Western China Science and Technology Innovation Port, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
| | - Aimin Jiang
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, PR China
| | - Liqiang Zhang
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
| | - Siqi Yan
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
| | - Wangxiao He
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
| | - Jingyue Yang
- Department of Clinical Oncology, Air Force Medical University, Xi'an, 710032, PR China
| | - Tianya Liu
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
- Department of Tumor and Immunology in Precision Medical Institute, Western China Science and Technology Innovation Port, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China
| |
Collapse
|
5
|
Liu M, Tang Y, Yan M, Zhang J, Chen H, Zhang Q. Self-regulating immunosuppressive tumor microenvironment by NIR-II photothermal agent with anti-inflammatory activity for self-reinforcing immunotherapy synergy with cancer photothermal ablation. Biomaterials 2025; 318:123187. [PMID: 39965425 DOI: 10.1016/j.biomaterials.2025.123187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
Cancer thermal immunotherapeutic strategy has garnered tremendous attention in nanomedicine frontier. Photothermal therapy (PTT) within the second near-infrared (NIR-II) window is popular hyperthermia technique, but the effect of NIR-II PTT on antitumor immunity remains extensive exploration. Here, we first reveal the inflammatory immunosuppressive tumor microenvironment (TME) characterized by high-influx of myeloid-derived suppressor cells (MDSCs) following NIR-II PTT. For this issue, we develop biomineralized copper sulfide nanoparticles (BCS NPs) as NIR-II photothermal agents (PTAs), and found for the first time that they are superior electron-donor antioxidants with pronounced anti-inflammatory activities. Impressively, the excessive inflammation triggered by BCS NPs-mediated NIR-II PTT can be self-alleviated to minimize the high-influx of MDSCs, and the immunosuppression-related reactive oxygen species produced by MDSCs can also be self-scavenged. Such reprogramming of TME facilitates the activation of systemic adaptive antitumor immunity and the strengthened tumour-infiltrating of cytotoxic T lymphocytes, thereby realizing self-reinforcing immunotherapy synergy with cancer NIR-II PTT. More importantly, a robust abscopal effect against distant tumors is also observed in bilateral tumor models. This work provides the first example to underscore the potential of PTAs with antioxidant and anti-inflammatory functions as innovative thermal immuno-nanomedicines.
Collapse
Affiliation(s)
- Min Liu
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Sub-Lane Xiangshan, Hangzhou, 310024, China; State Key Laboratory of High-Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Ding-Xi Road, Shanghai, 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yiwen Tang
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Sub-Lane Xiangshan, Hangzhou, 310024, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mijia Yan
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Sub-Lane Xiangshan, Hangzhou, 310024, China; State Key Laboratory of High-Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Ding-Xi Road, Shanghai, 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiale Zhang
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Sub-Lane Xiangshan, Hangzhou, 310024, China; State Key Laboratory of High-Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Ding-Xi Road, Shanghai, 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hangrong Chen
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Sub-Lane Xiangshan, Hangzhou, 310024, China; State Key Laboratory of High-Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Ding-Xi Road, Shanghai, 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Qiuhong Zhang
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Sub-Lane Xiangshan, Hangzhou, 310024, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
6
|
Hu J, Jia X, Li M, Duan G, Man K, Dai H, Wen L, Geng H. Enhanced Delivery of Photothermal Gelatin Nanoparticle for Redox Balanced Nanocatalytic Tumor Chemotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2411018. [PMID: 40159797 DOI: 10.1002/smll.202411018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/13/2025] [Indexed: 04/02/2025]
Abstract
Nanocatalytic platforms are promising in cancer therapeutics via combining multiple treatments, which can be leveraged through the metabolic dysfunction in cancer progression. However, the lack of effective tumor delivery platforms hampers this approach. Here, a gelatin-based platform is designed that is preloaded with gold nanoparticles and photothermal polypyrrole (GNPs@AuNPs-PPy) with an acid-induced doping enhancement. Benefiting from the tumor associated overexpression of H2O2, peroxidase-like Au nanoparticles induce a burst of oxidative reactive oxygen species in the local tumor microenvironment (TME). Subsequent orchestration of redox surroundings recruits immune cells, showcasing an effective antineoplastic pathway. Under near infrared light (NIR) irradiation, nanohybrids exhibit dual pH/NIR enhanced drug release within the TME, while allowing for multimodal imaging-guided theranostics. Leveraging this modality, GNPs@AuNPs-PPy delivers quercetin (a natural antitumor mediator) in TME, boosting anti-tumor therapy. The gelatin-mediated nanomedicine provides an alternative platform for combinatorial dynamic antitumor treatment.
Collapse
Affiliation(s)
- Jiayi Hu
- Institute of Biomedical and Health Engineering, Shenzhen International Graduate School, Key Laboratory of Active Proteins and Peptides Green Biomanufacturing of Guangdong Higher Education Institutes, Tsinghua University, Shenzhen, Guangdong, 518055, China
| | - Xiaoyu Jia
- School of Environmental and Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212003, China
| | - Manlin Li
- Department of Radiology, The Fourth Affiliated Hospital of Soochow University, Medical Centre of Soochow University, Suzhou, Jiangsu, 215000, China
| | - Guangxin Duan
- Department of Radiology, The Fourth Affiliated Hospital of Soochow University, Medical Centre of Soochow University, Suzhou, Jiangsu, 215000, China
| | - Kwan Man
- Department of Surgery, HKU-SZH & Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hongliang Dai
- School of Environmental and Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212003, China
| | - Ling Wen
- Department of Radiology, The Fourth Affiliated Hospital of Soochow University, Medical Centre of Soochow University, Suzhou, Jiangsu, 215000, China
| | - Hongya Geng
- Institute of Biomedical and Health Engineering, Shenzhen International Graduate School, Key Laboratory of Active Proteins and Peptides Green Biomanufacturing of Guangdong Higher Education Institutes, Tsinghua University, Shenzhen, Guangdong, 518055, China
| |
Collapse
|
7
|
Sakref C, Saby A, Rodriguez C, Ardin M, Moudombi L, Doffin AC, Gobbini E, Voissiere A, Besson L, Laoubi L, Böttcher J, Depil S, Hubert M, Bendriss-Vermare N, Caux C, Valladeau-Guilemond J. Type III interferon primes pDCs for TLR7 activation and antagonizes immune suppression mediated by TGF-β and PGE2. Nat Commun 2025; 16:3045. [PMID: 40155377 PMCID: PMC11953300 DOI: 10.1038/s41467-025-58220-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 03/12/2025] [Indexed: 04/01/2025] Open
Abstract
Conventional dendritic cell and plasmacytoid dendritic cell (pDC) subsets have specialized functions that can be modulated by the tumor microenvironment, and produce different interferons that are central to antitumor immune responses. While the function of type I interferons in tumor immunity is well characterized, that of type III interferons produced by type 1 conventional dendritic cells in the tumor microenvironment remains unclear. Here we demonstrate in vitro that type III interferons orchestrate pDC survival, activation and TLR7 expression in the blood, thereby enhancing pDC responses to a TLR7 ligand. Moreover, we show that tumor-associated pDCs express the highest level of IFNLR1, and that these immune cell subsets are the most responsive to IFN-III. Importantly, type III interferons prevent the inhibition of pDCs induced by TGF-β or PGE2 in tumor soluble milieu from patients to restores production of IFN-α in pDCs. With TGF-β or PGE2 having pleotropic functions in immune regulation, our results thus implicate IFN-III-mediated immune modulation to have broad impact on various pathological situations.
Collapse
Affiliation(s)
- Candice Sakref
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, PLASCAN, INSERM 1052, CNRS, 5286, Lyon, France
- LabEx DEVweCAN, Lyon, France
| | - Alexis Saby
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, PLASCAN, INSERM 1052, CNRS, 5286, Lyon, France
| | - Céline Rodriguez
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, PLASCAN, INSERM 1052, CNRS, 5286, Lyon, France
- Laboratory of Cancer Immunotherapy of Lyon (LICL), Centre Léon Bérard, Lyon, France
| | - Maude Ardin
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, PLASCAN, INSERM 1052, CNRS, 5286, Lyon, France
- Synergie Lyon Cancer, Plateforme de bio-informatique 'Gilles Thomas', Centre Léon Bérard, Lyon, France
| | - Lyvia Moudombi
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, PLASCAN, INSERM 1052, CNRS, 5286, Lyon, France
| | - Anne-Claire Doffin
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, PLASCAN, INSERM 1052, CNRS, 5286, Lyon, France
| | - Elisa Gobbini
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, PLASCAN, INSERM 1052, CNRS, 5286, Lyon, France
| | - Aurélien Voissiere
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, PLASCAN, INSERM 1052, CNRS, 5286, Lyon, France
| | - Laurie Besson
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, PLASCAN, INSERM 1052, CNRS, 5286, Lyon, France
- Centre Léon Bérard, F-, Lyon, France
| | - Léo Laoubi
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, PLASCAN, INSERM 1052, CNRS, 5286, Lyon, France
| | - Jan Böttcher
- Department of Experimental Immunology, Institute of Immunology, University of Tübingen, Tübingen, Germany
- Institute of Molecular Immunology, TUM University Hospital, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Stéphane Depil
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, PLASCAN, INSERM 1052, CNRS, 5286, Lyon, France
- Centre Léon Bérard, F-, Lyon, France
| | - Margaux Hubert
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, PLASCAN, INSERM 1052, CNRS, 5286, Lyon, France
- Synergie Lyon Cancer, Plateforme de bio-informatique 'Gilles Thomas', Centre Léon Bérard, Lyon, France
- Centre Léon Bérard, F-, Lyon, France
| | - Nathalie Bendriss-Vermare
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, PLASCAN, INSERM 1052, CNRS, 5286, Lyon, France
- Laboratory of Cancer Immunotherapy of Lyon (LICL), Centre Léon Bérard, Lyon, France
| | - Christophe Caux
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, PLASCAN, INSERM 1052, CNRS, 5286, Lyon, France
- LabEx DEVweCAN, Lyon, France
- Laboratory of Cancer Immunotherapy of Lyon (LICL), Centre Léon Bérard, Lyon, France
| | - Jenny Valladeau-Guilemond
- Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, PLASCAN, INSERM 1052, CNRS, 5286, Lyon, France.
- LabEx DEVweCAN, Lyon, France.
| |
Collapse
|
8
|
Wu Y, Zhu L, Li S, Liu L, Wang Y, Yang Y, Mu Y, Zhu Q, Jiang Y, Wu C, Xi P, Ma C, Liang L, Gao M, Hu Y, Ding Q, Pan S. DA-DRD5 signaling reprograms B cells to promote CD8 + T cell-mediated antitumor immunity. Cell Rep 2025; 44:115364. [PMID: 40023842 DOI: 10.1016/j.celrep.2025.115364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 12/16/2024] [Accepted: 02/06/2025] [Indexed: 03/04/2025] Open
Abstract
Neuronal signals have emerged as pivotal regulators of B cells that regulate antitumor immunity and tumor progression. However, the functional relevance and mechanistic basis of the effects of the neurotransmitter dopamine (DA) on tumor immunity remain elusive. Here, we discovered that plasma DA levels are positively correlated with circulating B cell numbers and potently activate B cell responses in a manner dependent on the DRD5 receptor. Notably, DRD5 signaling enhanced the Janus kinase 1 (JAK1)-STAT1 signaling in B cell responses, which enhanced B cell activation and increased antigen presentation and co-stimulation, resulting in increased expansion and cytotoxicity in tumor-specific effector of T cells. Our findings demonstrate that DA signaling suppresses tumor progression and highlight DRD5 as a promising target for cancer immunotherapy.
Collapse
Affiliation(s)
- Yuqing Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| | - Lei Zhu
- Jiangsu Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210036, China; Department of Breast Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), Nanjing 210004, China
| | - Sheng Li
- Department of Clinical Laboratory, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Lu Liu
- Department of Immunology, Nanjing Medical University, Nanjing 211166, China
| | - Yaman Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| | - Yongbing Yang
- Department of Medical Laboratory, Affiliated Children's Hospital of Jiangnan University, Wuxi 214000, China
| | - Yuan Mu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| | - Qiuying Zhu
- The First Clinical School of Nanjing Medical University, Nanjing 210029, China
| | - Yuying Jiang
- Department of Immunology, Nanjing Medical University, Nanjing 211166, China
| | - Chunyan Wu
- Department of Pathology, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), Nanjing 210004, China
| | - Peiwen Xi
- Department of Health Management Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chunmei Ma
- Department of Immunology, Nanjing Medical University, Nanjing 211166, China
| | - Lijun Liang
- Department of Thoracic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China
| | - Min Gao
- Department of Nephrology, Tongren Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200336, China
| | - Yingchao Hu
- Department of Immunology, Nanjing Medical University, Nanjing 211166, China.
| | - Qiang Ding
- Jiangsu Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210036, China.
| | - Shiyang Pan
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China.
| |
Collapse
|
9
|
Liu D, Liu L, Zhao X, Zhang X, Chen X, Che X, Wu G. A comprehensive review on targeting diverse immune cells for anticancer therapy: Beyond immune checkpoint inhibitors. Crit Rev Oncol Hematol 2025; 210:104702. [PMID: 40122356 DOI: 10.1016/j.critrevonc.2025.104702] [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: 02/10/2025] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
Although immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, primary resistance and acquired resistance continue to limit their efficacy for many patients. To address resistance and enhance the anti-tumor activity within the tumor immune microenvironment (TIME), numerous therapeutic strategies targeting both innate and adaptive immune cells have emerged. These include combination therapies with ICIs, chimeric antigen receptor T-cell (CAR-T), chimeric antigen receptor macrophages (CAR-Ms) or chimeric antigen receptor natural killer cell (CAR-NK) therapy, colony stimulating factor 1 receptor (CSF1R) inhibitors, dendritic cell (DC) vaccines, toll-like receptor (TLR) agonists, cytokine therapies, and chemokine inhibition. These approaches underscore the significant potential of the TIME in cancer treatment. This article provides a comprehensive and up-to-date review of the mechanisms of action of various innate and adaptive immune cells within the TIME, as well as the therapeutic strategies targeting each immune cell type, aiming to deepen the understanding of their therapeutic potential.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lei Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xinming Zhao
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaoman Zhang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaochi Chen
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Xiangyu Che
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Guangzhen Wu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
10
|
Cannet F, Sequera C, Veloso PM, El Kaoutari A, Methia M, Richelme S, Kaya M, Cherni A, Dupont M, Borg JP, Morel C, Boursier Y, Maina F. Tracing specificity of immune landscape remodeling associated with distinct anticancer treatments. iScience 2025; 28:112071. [PMID: 40124507 PMCID: PMC11930375 DOI: 10.1016/j.isci.2025.112071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 07/18/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Immune cells within the tumor microenvironment impact cancer progression, resistance, response to treatments. Despite remarkable outcomes for some cancer patients, immunotherapies remain unsatisfactory for others. Here, we designed an experimental setting using the Alb-R26 Met "inside-out" mouse model, faithfully recapitulating molecular features of liver cancer patients, to explore the effects of distinct anticancer targeted therapies on the tumor immune landscape. Using two treatments in clinical trials for different cancer types, Decitabine and MEK+BCL-XL blockage, we show their capability to trigger tumor regression in Alb-R26 Met mice and to superimpose distinct profiles of immune cell types and immune-checkpoints, impacting immunotherapy response. A machine learning approach processing tumor imaging and immune profile data identified a putative signature predicting tumor treatment response in mice and patients. Outcomes exemplify how the tumor immune microenvironment is differentially reshaped by distinct anticancer agents and highlight the importance of measuring its modulation during treatment to optimize oncotherapy and immunotherapy combinations.
Collapse
Affiliation(s)
- Floriane Cannet
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| | - Célia Sequera
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| | - Paula Michea Veloso
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Abdessamad El Kaoutari
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Melissa Methia
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Sylvie Richelme
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| | - Muge Kaya
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Afef Cherni
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
| | - Mathieu Dupont
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
| | - Jean-Paul Borg
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Institut Universitaire de France, Paris, France
| | - Christian Morel
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
| | | | - Flavio Maina
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| |
Collapse
|
11
|
Du Q, Meng C, Zhang W, Huang L, Xue C. Establishing a Prognostic Model Correlates to Inflammatory Response Pathways for Prostate Cancer via Multiomic Analysis of Lactylation-Related Genes. Int J Genomics 2025; 2025:6681711. [PMID: 40161494 PMCID: PMC11952923 DOI: 10.1155/ijog/6681711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Prostate cancer (PCa) continues to pose substantial clinical challenges, with molecular heterogeneity significantly impacting therapeutic decision-making and disease trajectories. Emerging evidence implicates protein lactylation-a novel epigenetic regulatory mechanism-in oncogenic processes, though its prognostic relevance in PCa remains underexplored. Through integrative bioinformatics interrogation of lactylation-associated molecular signatures, we established prognostic correlations using multivariable feature selection methodologies. Initial screening via differential expression analysis (limma package) coupled with Cox proportional hazards modeling revealed 11 survival-favorable regulators and 16 hazard-associated elements significantly linked to biochemical recurrence. To enhance predictive precision, ensemble machine learning frameworks were implemented, culminating in a 10-gene lactylation signature demonstrating robust discriminative capacity (concordance index = 0.738) across both primary (TCGA-PRAD) and external validation cohorts (DKFZ). Multivariable regression confirmed the lactylation score's prognostic independence, exhibiting prominent associations with clinicopathological parameters including tumor staging and metastatic potential. The developed clinical-molecular nomogram achieved superior predictive accuracy (C - index > 0.7) through the synergistic integration of biological and clinical covariates. Tumor microenvironment deconvolution uncovered distinct immunological landscapes, with high-risk stratification correlating with enriched stromal infiltration and immunosuppressive phenotypes. Pathway enrichment analyses implicated chromatin remodeling processes and cytokine-mediated inflammatory cascades as potential mechanistic drivers of prognostic divergence. Therapeutic vulnerability profiling demonstrated differential response patterns: low-risk patients exhibited enhanced immune checkpoint inhibitor responsiveness, whereas high-risk subgroups showed selective chemosensitivity to docetaxel and mitoxantrone. Functional validation in PC-3 models revealed AK5 silencing induced proapoptotic effects, suppressed metastatic potential of migration and invasion, and modulated immune checkpoint regulation through CD276 coexpression. These multimodal findings position lactylation dynamics, particularly AK5-mediated pathways, as promising therapeutic targets and stratification biomarkers in PCa management.
Collapse
Affiliation(s)
- Qinglong Du
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - CuiYu Meng
- The Department of EICU, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| | - Wenchao Zhang
- The Department of Urology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| | - Li Huang
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, Guangdong, China
| | - Chunlei Xue
- The Department of Urology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| |
Collapse
|
12
|
Yang Y, Wang H, Xue Q, Peng W, Zhou Q. New advances of natural products in non-small cell lung cancer: From mechanisms to therapies. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119636. [PMID: 40120701 DOI: 10.1016/j.jep.2025.119636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE With the rise of immunotherapy, the treatment approach for non-small cell lung cancer (NSCLC) has undergone revolutionary changes. However, the prognosis for NSCLC patients has not been significantly improved due to the development of acquired drug resistance. Therefore, there is an urgent need to develop new and more effective drugs for treating NSCLC or improving tumor treatment resistance. Traditional Chinese medicine (TCM) has been gradually incorporated into the combined treatment of NSCLC. Its active components (also known as natural products) exhibit novel structures, multi-target effects, diverse pathways, minimal toxicity, and varied biological activities, which play a therapeutic role in various diseases. Thus, natural products hold great potential for future clinical applications. AIM OF THE STUDY Screening main traditional plants widely used in NSCLC and their derived natural products, as well as exploring the mechanisms by which these natural products act on NSCLC-particularly focusing on their applications-can provide valuable insights for the development of therapeutic drugs targeting NSCLC. METHODS A comprehensive, computerized literature search was conducted in PubMed, Embase, Web of Science, Cochrane Library, CNKI Scholar, the American Chemical Abstracts, and Wanfang Database up to June 2024, using the following keywords: "traditional Chinese medicine", "herbal medicine", "medicinal plants", and "herbal", paired with terms such as "non-small cell lung cancer", "therapy", "natural products", and "active ingredient". RESULTS Summarizing current research findings, we discovered eleven medicinal plants containing a total of fourteen natural products. Natural products have a significant impact on tumor progression in NSCLC, including apotosis, autophagy, pyrotosis, cell-cycle arrest and metasis. Moreover, natural products can modulate the activities of various immune cells and reshape the immune microenvironment. Combined with conventional cancer treatments, natural products demonstrate promising therapeutic effects and effectively reverse drug resistance. Furthermore,the use of nano-drug delivery systems to address limitations associated with natural products. CONCLUSIONS This review summarizes eleven medicinal plants containing a total of fourteen natural products that can enhance NSCLC treatment and indicates their action mechanisms. Furthermore, we also discuss limitations of natural products and explore the use of nano-drug delivery systems to address limitations associated with natural products.
Collapse
Affiliation(s)
- Yuening Yang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Haolei Wang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Qianqian Xue
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Wenbei Peng
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Qiong Zhou
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China.
| |
Collapse
|
13
|
Alfawaz Altamimi AS, Arockia Babu M, Afzal M, Bishoyi AK, Roopashree R, Saini S, Sharma RSK, Pathak PK, Chauhan AS, Goyal K, Ali H, Khan NH, Balaraman AK. Exosomes derived from natural killer cells: transforming immunotherapy for aggressive breast cancer. Med Oncol 2025; 42:114. [PMID: 40100465 DOI: 10.1007/s12032-025-02647-y] [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: 12/01/2024] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
Natural killer cell-derived exosomes (NK-Exos) hold great promise as immune modulators and immunotherapeutics against cancer due to their intrinsically latent anti-tumor effects. They use these nanosized vesicles to deliver cytotoxic molecules, such as perforin, granzymes, and miRNAs, directly to cancer cells to kill them, avoiding immune suppression. NK-Exos has particular efficacy for treating aggressive breast cancer by modulating the TME to activate the immune response and suppress immunosuppressive factors. Bioengineering advances have extended the therapeutic potential of NK-Exos, which permits precise tumor cell targeting and efficient delivery of therapeutic payloads, including small RNAs and chemotherapeutic agents. In engineered NK-Exos, sensitization of cancer cells to apoptosis, reduction of tumor growth, and resistance to drugs have been demonstrated to be highly effective. When combined, NK-Exos synergizes with radiotherapy, chemotherapy, or checkpoint inhibitors, enhancing therapeutic efficacy, and minimizing systemic toxicity. This review emphasizes the critical role of NK-Exos in breast cancer treatment, their integration into combination therapies, and the need for further research to overcome existing limitations and fully realize their clinical potential.
Collapse
Affiliation(s)
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Ashok Kumar Bishoyi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Suman Saini
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - R S K Sharma
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Piyus Kumar Pathak
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Ashish Singh Chauhan
- Division of Research and Innovation, Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun, 248002, India
| | - Haider Ali
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, 63000, Cyberjaya, Selangor, Malaysia.
| |
Collapse
|
14
|
Lan X, Johnston E, Ning T, Chen G, Haglund L, Li J. Immunomodulatory bioadhesive technologies. Biomaterials 2025; 321:123274. [PMID: 40156979 DOI: 10.1016/j.biomaterials.2025.123274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/20/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
Bioadhesives have found significant use in medicine and engineering, particularly for wound care, tissue engineering, and surgical applications. Compared to traditional wound closure methods such as sutures and staples, bioadhesives offer advantages, including reduced tissue damage, enhanced healing, and ease of implementation. Recent progress highlights the synergy of bioadhesives and immunoengineering strategies, leading to immunomodulatory bioadhesives capable of modulating immune responses at local sites where bioadhesives are applied. They foster favorable therapeutic outcomes such as reduced inflammation in wounds and implants or enhanced local immune responses to improve cancer therapy efficacy. The dual functionalities of bioadhesion and immunomodulation benefit wound management, tissue regeneration, implantable medical devices, and post-surgical cancer management. This review delves into the interplay between bioadhesion and immunomodulation, highlighting the mechanobiological coupling involved. Key areas of focus include the modulation of immune responses through chemical and physical strategies, as well as the application of these bioadhesives in wound healing and cancer treatment. Discussed are remaining challenges such as achieving long-term stability and effectiveness, necessitating further research to fully harness the clinical potential of immunomodulatory bioadhesives.
Collapse
Affiliation(s)
- Xiaoyi Lan
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A3, Canada; Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada
| | - Evan Johnston
- Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada
| | - Tianqin Ning
- Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada; Department of Biomedical Engineering, McGill University, 3775 Rue University, Montreal, Quebec, H3A 2B4, Canada
| | - Guojun Chen
- Department of Biomedical Engineering, McGill University, 3775 Rue University, Montreal, Quebec, H3A 2B4, Canada; Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Pine Ave W, Montreal, Quebec, H3A 1A3, Canada
| | - Lisbet Haglund
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A3, Canada; Shriners Hospital for Children, 1003 Decarie Blvd, Montreal, Quebec, H4A 0A9, Canada.
| | - Jianyu Li
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A3, Canada; Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada; Department of Biomedical Engineering, McGill University, 3775 Rue University, Montreal, Quebec, H3A 2B4, Canada.
| |
Collapse
|
15
|
Zhang L, Peng Y, Huang S, Zhong L. Integrative analysis of DNA methylation and gene expression in skin cutaneous melanoma by bioinformatic approaches. Arch Dermatol Res 2025; 317:545. [PMID: 40067504 PMCID: PMC11897118 DOI: 10.1007/s00403-025-03863-2] [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: 11/05/2024] [Revised: 01/06/2025] [Accepted: 01/18/2025] [Indexed: 03/15/2025]
Abstract
Skin cutaneous melanoma represents a significant threat among skin cancers. Investigating key methylated genes with prognostic implications remains an area ripe for exploration in this field. This study aims to identify survival-associated methylated genes and their specific methylation sites in skin cutaneous melanoma through integrated bioinformatic analysis. Utilizing data from the Cancer Genome Atlas database, gene methylation and expression files were analyzed. The results indicate that patients with skin cutaneous melanoma exhibiting high expression of hypomethylated HHEX experience better outcomes compared to those with low expression of hypermethylated HHEX. Furthermore, fifteen methylation sites within HHEX were found to significantly correlate with its expression levels. Expression of HHEX demonstrated a downward trend across pathological stages I-IV. The identified driven gene, HHEX, likely plays a crucial role in the survival of skin cutaneous melanoma patients. These findings provide new epigenetic insights and potential targets for early prognosis prediction in skin cutaneous melanoma.
Collapse
Affiliation(s)
- Liming Zhang
- Department of Anesthesiology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Yuchuan Peng
- Department of Anesthesiology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Shan Huang
- Department of Otolaryngology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Liang Zhong
- Department of Anesthesiology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China.
| |
Collapse
|
16
|
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] [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
|
17
|
Zhou Y, Geng S, Tang RC, Yu H, Zhang A, Bai Y, Zhang J. Clinical and functional significance of SPATA2 in cancer particularly in LIHC. Sci Rep 2025; 15:8392. [PMID: 40069269 PMCID: PMC11897323 DOI: 10.1038/s41598-025-91386-1] [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: 11/28/2024] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
Spermatogenesis-associated protein 2 (SPATA2) is primarily named for its important role in spermatogenesis. Its function in tumorigenesis remains elusive. Here, we used various bioinformatic tools to systematically analyze the expression patterns of SPATA2 in cancers, the correlation of SPATA2 expression with clinical parameters, genetic variation, methylation, phosphorylation, immune infiltration and immune therapy. SPATA2 is significantly upregulated in multiple cancers and its expression was associated with tumor stage, grade and serve as a potential prognostic marker in LIHC. Notably, SPATA2 was also linked to immune suppression, exhibiting positive correlations with immune checkpoint genes and immune suppressive cells such as regulatory T cells and MDSCs. Furthermore, SPATA2 interacted and co-expressed with proteins involved in DNA repair mechanisms, indicating its potential role in maintaining genomic stability. Finally, we conducted biological experiments to investigate the role of SPATA2 in LIHC. SPATA2 knockdown enhances the migration and proliferation capabilities of Hep-G2 and HuH7 cell lines. These findings underscore the significance of SPATA2 in cancer biology, suggests its role in both the tumor microenvironment and the tumor cell level and its potential as a prognostic marker and therapeutic target in oncology, particularly in LIHC.
Collapse
Affiliation(s)
- Yunxuan Zhou
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Shijin Geng
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Rong-Chun Tang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Hengxiang Yu
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Ao Zhang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Yuekui Bai
- General Surgery, Haidian Hospital, Beijing, China.
| | - Jun Zhang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China.
| |
Collapse
|
18
|
Zhang Y, Chen H, Mo H, Zhao N, Sun X, Liu B, Gao R, Xu B, Zhang Z, Liu Z, Ma F. Distinct cellular mechanisms underlie chemotherapies and PD-L1 blockade combinations in triple-negative breast cancer. Cancer Cell 2025; 43:446-463.e7. [PMID: 39919737 DOI: 10.1016/j.ccell.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 11/05/2024] [Accepted: 01/13/2025] [Indexed: 02/09/2025]
Abstract
Combining immune checkpoint blockade (ICB) with chemotherapy shows promise for treating triple-negative breast cancer (TNBC), though the mechanisms remain incompletely understood. Here, we integrate published and new single-cell RNA sequencing (scRNA-seq) data to investigate the tumor immune microenvironment (TIME) in TNBC patients treated with paclitaxel (PTX), nab-paclitaxel (Nab-PTX), and their combinations with the anti-PD-L1 antibody atezolizumab (ATZ). Compared to ATZ plus PTX, ATZ plus Nab-PTX rewires TCF7+ stem-like effector memory CD8+ T cells (Tsem) and CD4+ T follicular helper (Tfh) cells. Nab-paclitaxel, unlike PTX, also reshapes the myeloid compartment, expanding mast cells and pro-inflammatory macrophages. Our analyses in human TNBC and murine models underscore the crucial role of mast cells in orchestrating anti-tumor immune responses, likely by promoting the recruitment and activation of T and B cells. In vivo experiments demonstrate that activating mast cells alongside PD-L1 blockade attenuates TNBC progression, suggesting mast cells as a promising adjunct for enhancing ICB therapy efficacy.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing 100871, China.
| | - Hongyan Chen
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hongnan Mo
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ning Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiaoying Sun
- Department of Medical Oncology, Cancer Hospital of HuanXing, ChaoYang District, Beijing 100005, China
| | - Baolin Liu
- BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing 100871, China
| | - Ranran Gao
- BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing 100871, China
| | - Binghe Xu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Zemin Zhang
- BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing 100871, China.
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Fei Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
19
|
Juthi RT, Sazed SA, Mareboina M, Zaravinos A, Georgakopoulos-Soares I. Characterization of Exhausted T Cell Signatures in Pan-Cancer Settings. Int J Mol Sci 2025; 26:2311. [PMID: 40076932 PMCID: PMC11899893 DOI: 10.3390/ijms26052311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
T cells play diverse roles in cancer immunology, acting as tumor suppressors, cytotoxic effectors, enhancers of cytotoxic T lymphocyte responses and immune suppressors; providing memory and surveillance; modulating the tumor microenvironment (TME); or activating innate immune cells. However, cancer cells can disrupt T cell function, leading to T cell exhaustion and a weakened immune response against the tumor. The expression of exhausted T cell (Tex) markers plays a pivotal role in shaping the immune landscape of multiple cancers. Our aim was to systematically investigate the role of known T cell exhaustion (Tex) markers across multiple cancers while exploring their molecular interactions, mutation profiles, and potential implications for immunotherapy. The mRNA expression profile of six Tex markers, LAG-3, PDCD1, TIGIT, HAVCR2, CXCL13, and LAYN was investigated in pan-cancer. Utilizing data from The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), The Cancer Proteome Atlas (TCPA), and other repositories, we characterized the differential expression of the Tex markers, their association with the patients' survival outcome, and their mutation profile in multiple cancers. Additionally, we analyzed the effects on cancer-related pathways and immune infiltration within the TME, offering valuable insights into mechanisms of cancer immune evasion and progression. Finally, the correlation between their expression and sensitivity to multiple anti-cancer drugs was investigated extensively. Differential expression of all six markers was significantly associated with KIRC and poor prognosis in several cancers. They also played a potential activating role in apoptosis, EMT, and hormone ER pathways, as well as a potential inhibitory role in the DNA damage response and RTK oncogenic pathways. Infiltration of different immune cells was also found to be associated with the expression of the Tex-related genes in most cancer types. These findings underline that the reviving of exhausted T cells can be used to enhance the efficacy of immunotherapy in cancer patients.
Collapse
Affiliation(s)
- Rifat Tasnim Juthi
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh;
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (S.A.S.); (M.M.)
| | - Saiful Arefeen Sazed
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (S.A.S.); (M.M.)
| | - Manvita Mareboina
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (S.A.S.); (M.M.)
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, 22006, 1516 Nicosia, Cyprus
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), 22006, 1516 Nicosia, Cyprus
| | - Ilias Georgakopoulos-Soares
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (S.A.S.); (M.M.)
| |
Collapse
|
20
|
Almawash S. Revolutionary Cancer Therapy for Personalization and Improved Efficacy: Strategies to Overcome Resistance to Immune Checkpoint Inhibitor Therapy. Cancers (Basel) 2025; 17:880. [PMID: 40075727 PMCID: PMC11899125 DOI: 10.3390/cancers17050880] [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: 01/06/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a significant public health issue worldwide, standing as a primary contributor to global mortality, accounting for approximately 10 million fatalities in 2020 [...].
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
21
|
Chen L, He Y, Duan M, Yang T, Chen Y, Wang B, Cui D, Li C. Exploring NUP62's role in cancer progression, tumor immunity, and treatment response: insights from multi-omics analysis. Front Immunol 2025; 16:1559396. [PMID: 40098960 PMCID: PMC11911477 DOI: 10.3389/fimmu.2025.1559396] [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: 01/12/2025] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Background NUP62, a key component of the nuclear pore complex, is closely associated with cellular functions and cancer progression. However, its expression patterns, prognostic value, and relationship with tumour immunity and drug sensitivity across multiple cancers have not been systematically studied. This study used multi-omics analyses combined with experimental validation in gastric cancer to investigate the expression, functional characteristics, and clinical relevance of NUP62 in cancer. Methods Data from TCGA, GTEx, and CPTAC databases were used to analyse the expression, mutation characteristics, and clinical associations of NUP62. Tools such as SangerBox, TIMER 2.0, and GSEA were employed to evaluate the relationship between NUP62 and the tumour immune microenvironment, as well as its involvement in signalling pathways. Immunohistochemistry and RT-PCR were used to validate the expression of NUP62 in gastric cancer tissues. PRISM and CTRP databases were utilised to assess the correlation between NUP62 expression and drug sensitivity. Results NUP62 was significantly upregulated in multiple cancers and was associated with poor prognosis in cancers such as clear cell renal carcinoma (KIRC), lower-grade glioma (LGG), and adrenocortical carcinoma (ACC), while playing a protective role in others, such as bladder cancer (BLCA) and stomach cancer (STAD). Functional analyses showed that NUP62 is involved in cell cycle regulation, DNA damage repair, and tumour immunity. High NUP62 expression was significantly correlated with increased infiltration of immune cells, such as macrophages and T cells, and a higher response rate to immunotherapy. Drug sensitivity analysis identified NUP62 as a marker of sensitivity to various chemotherapeutic agents. Validation experiments demonstrated that NUP62 mRNA and protein levels were significantly higher in gastric cancer tissues than in adjacent normal tissues. Conclusions NUP62 plays a critical role in multiple cancers and shows potential as a biomarker for cancer diagnosis, prognosis, and therapeutic response prediction. Its role in tumour immunity and signalling pathways highlights its potential as a target for immunotherapy and precision medicine.
Collapse
Affiliation(s)
- Lihong Chen
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Youfu He
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Menghui Duan
- Department of Critical Care Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Tianwen Yang
- Department of Orthopaedics, Guizhou Second People's Hospital, Guiyang, Guizhou, China
| | - Yin Chen
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Bo Wang
- Department of General Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Dejun Cui
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Chen Li
- Department of Pharmacy, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
22
|
Du Y, Gu B, Shi L, She Y, Zhao Q, Gao S. Data-Driven Molecular Typing: A New Frontier in Esophageal Cancer Management. Cancer Med 2025; 14:e70730. [PMID: 40018789 PMCID: PMC11868787 DOI: 10.1002/cam4.70730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/09/2025] [Accepted: 02/15/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a predominant and highly lethal form of esophageal cancer, with a five-year survival rate below 20%. Despite advancements, most patients are diagnosed at advanced stages, limiting effective treatment options. Multi-omics integration, encompassing somatic genomic alterations, inherited genetic mutations, transcriptomics, proteomics, metabolomics, and single-cell sequencing, has enabled the identification of distinct molecular subtypes of ESCC. METHOD This article systematically reviewed the current status of molecular subtyping of ESCC based on big data, summarized unique subtypes with differing treatment responses and prognostic outcomes. RESULT Key findings included subtype-specific genetic mutations, signaling pathway alterations, and metabolomic profiles, which offer novel biomarkers and therapeutic targets. Furthermore, this review discusses the link between molecular subtypes and immunotherapy efficacy, chemotherapy response, and drug development. CONCLUSION These insights highlight the potential of omics-based molecular typing to transform ESCC management and facilitate personalized treatment strategies.
Collapse
Affiliation(s)
- Yue Du
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital (College of Clinical Medicine) of Henan University of Science and TechnologyCancer HospitalLuoyangHenanChina
| | - Bianli Gu
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital (College of Clinical Medicine) of Henan University of Science and TechnologyCancer HospitalLuoyangHenanChina
| | - Linlin Shi
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital (College of Clinical Medicine) of Henan University of Science and TechnologyCancer HospitalLuoyangHenanChina
| | - Yong She
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouGuangdongChina
| | - Qi Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouGuangdongChina
| | - Shegan Gao
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, Henan Key Laboratory of Cancer Epigenetics, The First Affiliated Hospital (College of Clinical Medicine) of Henan University of Science and TechnologyCancer HospitalLuoyangHenanChina
| |
Collapse
|
23
|
Wang Y, Liu C, Pang J, Li Z, Zhang J, Dong L. The Extra-Tumoral Vaccine Effects of Apoptotic Bodies in the Advancement of Cancer Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410503. [PMID: 39871756 DOI: 10.1002/smll.202410503] [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: 11/11/2024] [Revised: 01/16/2025] [Indexed: 01/29/2025]
Abstract
The induction of apoptosis in tumor cells is a common target for the development of anti-tumor therapies; however, these therapies still leave patients at increased risk of disease recurrence. For example, apoptotic tumor cells can promote tumor growth and immune evasion via the secretion of metabolites, apoptotic extracellular vesicles, and induction of pro-tumorigenic macrophages. This paradox of apoptosis induction and the pro-tumorigenic effects of tumor cell apoptosis has begged the question of whether apoptosis is a suitable cancer therapy, and led to further explorations into other immunogenic cell death-based approaches. However, these strategies still face multiple challenges, the most critical of which is the tumor microenvironment. Contrary to the promotion of immune tolerance mediated by apoptotic tumor cells, apoptotic bodies with enriched tumor-related antigens have demonstrated great immunogenic potential, as evidenced by their ability to initiate systemic T-cell immune responses. These characteristics indicate that apoptotic body-based therapies could be ideal "in situ" extra-tumoral tumor vaccine candidates for the treatment of cancers, and further address the current issues with apoptosis-based or immunotherapy treatments. Although not yet tested clinically, apoptotic body-based vaccines have the potential to better treatment strategies and patient outcomes in the future.
Collapse
Affiliation(s)
- Yulian Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Chunyan Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Jiayun Pang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Zhenjiang Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- Chemistry and Biomedicine Innovative Center, Nanjing University, Nanjing, Jiangsu, 210023, China
| |
Collapse
|
24
|
Fan J, Qin Y, Qiu W, Liang J, Xiao C, Xie Q, Tong C, Yuan L, Long Y, Liu B. Gamabufotalin loaded micro-nanocomposites for multimodal therapy of metastatic TNBC by efficiently inducing ICD. Biomaterials 2025; 314:122851. [PMID: 39366186 DOI: 10.1016/j.biomaterials.2024.122851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/30/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Gamabufotalin (CS-6), a main active compound derived from Chinese medicine Chansu, exhibits a robust inhibitory effect on programmed death-ligand 1 (PD-L1) in triple-negative breast cancer (TNBC) cells. Despite its potential for tumor therapy, the medical application of CS-6 is constrained by its hydrophobic nature, lack of targeting capability, and weak immunogenic cell death (ICD) effect. To address these limitations and improve the therapeutic efficiency of this drug against metastatic TNBC, we designed a new kind of CS-6@CPB-S.lux that integrates carboxy-Prussian blue nanoparticles (CPB NPs), CS-6, and attenuated Salmonella typhimurium (S.lux) for TNBC therapy. In vitro and in vivo results have confirmed that CS-6@CPB NPs were efficiently delivered to neoplastic tissue by the tumor hypoxic chemotaxis property of S.lux, wherein the nanomedicine induced significant tumor cell necroptosis and apoptosis via photothermal therapy (PTT) of CPB NPs and chemotherapy of CS-6, which elicited ICD and inhibited PD-L1 expression, resulting in dendritic cells (DCs) maturation and effector T cells activation to comprehensively eliminate tumors. Additionally, the CS-6@CPB-S.lux + Laser treatment significantly transformed the immunosuppressive tumor microenvironment (TME), enhancing antitumor immunity through promoting the polarization of tumor-associated macrophages into antitumorigenic M1 and reducing Tregs recruitment. Consequently, this comprehensive therapy not only inhibited primary and abscopal tumor progression but also prevented TNBC metastasis, which significantly prolonged survival time in animal models. In summary, these findings indicated an alternative approach for metastatic TNBC therapy.
Collapse
Affiliation(s)
- Jialong Fan
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Yan Qin
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China; TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Wensheng Qiu
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Jiahao Liang
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Chang Xiao
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Qian Xie
- Department of Pharmacy, Maternal and Child Health of Hunan Province, Changsha, 410008, China
| | - Chunyi Tong
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Liqin Yuan
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| | - Ying Long
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China.
| | - Bin Liu
- College of Biology, School of Biomedical Sciences, Hunan University, Changsha, 410082, China; NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
25
|
Zhu C, Zhang S, Wu Q. Prognostic significance of alterations in peripheral cellular and humoral immune biomarkers during radiochemotherapy in head and neck cancer patients. Transl Cancer Res 2025; 14:685-705. [PMID: 40104738 PMCID: PMC11912046 DOI: 10.21037/tcr-24-1510] [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: 08/25/2024] [Accepted: 01/09/2025] [Indexed: 03/20/2025]
Abstract
Background The impacts of radiochemotherapy on peripheral blood cell and lymphocyte cell counts, immunoglobulin (Ig) and complement levels remain unclear. This study aims to investigate the above parameters regulated by radiotherapy (RT), induction chemotherapy (ICT) and concurrent chemotherapy (CCT) in head and neck cancer (HNC) patients. Methods Patients with non-metastatic HNC treated by conventional intensity-modulated radiation therapy (IMRT) were enrolled in this study. Data of peripheral blood cells, lymphocyte subpopulations, complements and immunoglobulins were collected before, during and after IMRT. And conducted regular follow-up on patients. SPSS (IBM, version 26.0), R (MathSoft, 4.0.3) and Graphpad Prism were used to perform statistical analysis and plot figures. Results A total of 126 HNC patients undergoing RT were enrolled in this study. Among them, 44 patients received ICT, 56 patients received CCT, and 123 patients had complete survival information. Number of white blood cells (WBCs), platelets, basophils, total lymphocytes, CD4+ and CD8+ T cells, natural killer (NK) cells, B cells declined significantly during RT. Accordingly, the ratio of help T cells to suppressor T cells (Th/Ts) and the percentages of B cells, CD4+ T cells also declined. There were increased levels of neutrophils and complement 4 (C4) and percentage of NK cells during RT. ICT caused significant reductions of platelets, B cells and immunoglobulin A (IgA). CCT reduced WBCs, red blood cells (RBCs), platelets, hemoglobin (HGB), granulocytes, total lymphocytes, B cells, CD4+ and CD8+ T cells, NK cells and immunoglobulin G (IgG). Generalized linear model (GLM) analysis further confirmed that RT was a risk factor for lower total lymphocytes, B cells, CD4+ and CD8+ T cells, NK cells, Th/Ts ratios, and lower percentages of B cells, CD4+ T cells. ICT contributed to decreased Th/Ts ratios, and immunoglobulin M (IgM) and IgA levels. As for CCT, it was an unfavorable factor for reduced total lymphocytes, B cells, CD4+ and CD8+ T cells, NK cells and IgG. Conversely, complement 3 (C3) or 4 levels were higher in patients treated with RT, ICT or CCT. Importantly, we found that HNC patients with higher lymphocytes or lymphocyte percentages like CD3+, CD4+ and CD8+ T cells before or after RT had a better prognosis. While higher NK cells and NK cell percentage before RT were associated with worse prognosis. In addition, higher levels of C3 and C4 before and after RT were associated with a favorable prognosis. However, higher levels of IgA, immunoglobulin E (IgE), IgG, and IgM before RT were associated with poorer prognosis. Conclusions To sum up, chemoradiotherapy resulted in significant alterations in peripheral immune biomarkers which in return influenced HNC patients' survival.
Collapse
Affiliation(s)
- Chunmei Zhu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Radiation and Medical Oncology, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shuyuan Zhang
- Department of Medical Oncology, Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Radiation and Medical Oncology, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
26
|
Li W, Pan H, Zhou B, Cao Y, Shen B, Li N, Zhang Y, He Y, Jin J, Shi L, Gao J. Exploration of a prognostic signature for mitochondria-related genes and the therapeutic prospects of vorinostat in clear cell renal cell carcinoma. Transl Androl Urol 2025; 14:360-378. [PMID: 40114844 PMCID: PMC11921268 DOI: 10.21037/tau-24-565] [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: 10/10/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is a lethal urological cancer that accounts for a considerable portion of all malignant tumors in adults. Alterations in mitochondrial function and metabolism may influence the onset and progression of ccRCC. This study aims to develop a novel clinical prognostic signature for ccRCC based on mitochondria-associated genes, as well as to identify new potential therapeutic agents for this condition. Methods A total of 539 ccRCC tumors and 72 normal kidney specimens were analyzed from The Cancer Genome Atlas (TCGA). We created a prognostic signature based on univariate, multivariate Cox, and least absolute shrinkage and selection operator (LASSO) Cox regression. A differential expression analysis, functional enrichment, and assessment of tumor immune cell infiltration were performed. Connectivity Map (CMap) and L1000CDS2 dataset were utilized for potential therapeutic drug identification. Vorinostat was examined for its effects on ccRCC cell proliferation, cell death, migration, and invasion using Cell Counting Kit-8 (CCK-8), lactate dehydrogenase (LDH) release, wound healing, and transwell assays. Results The prognostic signature, comprising 16 mitochondria-related genes, demonstrated marked changes in overall survival between high- and low-risk groups. Functional analysis implicated immune-related pathways, indicating potential for immunotherapy strategies. Vorinostat, identified through drug screening, exhibited inhibitory implications on ccRCC cell proliferation, migration, and invasion and induced cell death. Conclusions The constructed prognostic signature provides a valuable tool for patient prognosis prediction. Vorinostat emerges as a promising therapeutic candidate for high-risk ccRCC patients, contributing to a deeper understanding of ccRCC biology and personalized therapeutic interventions.
Collapse
Affiliation(s)
- Wenhui Li
- Department of Pharmacology, College of Medical, Jiaxing University, Jiaxing, China
- College of Life Science, Zhejiang Sci-Tech University, Hangzhou, China
| | - Huan Pan
- Department of Central Laboratory, The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Bangwei Zhou
- Department of Pharmacology, College of Medical, Jiaxing University, Jiaxing, China
| | - Yifang Cao
- Department of Urology, The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Bin Shen
- Department of Pharmacology, College of Medical, Jiaxing University, Jiaxing, China
| | - Nan Li
- Department of Pharmacology, College of Medical, Jiaxing University, Jiaxing, China
| | - Yao Zhang
- Department of Pharmacology, College of Medical, Jiaxing University, Jiaxing, China
| | - Yi He
- Department of Urology, The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jing Jin
- Department of Urology, The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Lili Shi
- Department of Pharmacology, College of Medical, Jiaxing University, Jiaxing, China
| | - Jinlai Gao
- Department of Pharmacology, College of Medical, Jiaxing University, Jiaxing, China
- College of Life Science, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
27
|
Maggi E, Landolina N, Munari E, Mariotti FR, Tumino N, Vacca P, Azzarone B, Moretta L. T cells in the microenvironment of solid pediatric tumors: the case of neuroblastoma. Front Immunol 2025; 16:1544137. [PMID: 40092980 PMCID: PMC11906424 DOI: 10.3389/fimmu.2025.1544137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Neuroblastoma (NB) is an immunologically "cold" tumor with poor or no inflamed substrates as most of solid pediatric tumors (SPT). Consistent data indicate that NB tumor microenvironment (TME) is dominated by myeloid cells, with little (but variable) T cell infiltration. The obstacles to lymphocyte infiltration and to their anti-tumor activity are due to different tumor immune evasion strategies, including loss of HLA Class I molecules, high expression of immune checkpoint molecular ligands leading to exhaustion of T effector (and NK) cells, induction of T regulatory, myeloid and stromal cells and secretion of immunosuppressive mediators. In odds with adult solid tumors, NB displays weak immunogenicity caused by intrinsic low mutational burden and scant expression of neoepitopes in the context of MHC-class I antigens which, in turn, are particularly poorly expressed on NB cells, thus inducing low anti-tumor T cell responses. In addition, NB is generated from embryonal cells and is the result of transcriptional abnormalities and not of the accumulation of genetic mutations over time, thus further explaining the low immunogenicity. The poor expression of immunogenic molecules on tumor cells is associated with the high production of immunosuppressive factors which further downregulate lymphocyte infiltration and activity, thus explaining the limited efficacy of new drugs in NB, as immune checkpoint inhibitors. This review is focused on examining the role of T effector and regulatory cells infiltrating TME of NB, taking into account their repertoire, phenotype, function, plasticity and, importantly, predictive value for defining novel targets for therapy.
Collapse
Affiliation(s)
- Enrico Maggi
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Nadine Landolina
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Enrico Munari
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | | | - Nicola Tumino
- Innate Lymphoid Cells Unit, Immunology Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Paola Vacca
- Innate Lymphoid Cells Unit, Immunology Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Bruno Azzarone
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Lorenzo Moretta
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| |
Collapse
|
28
|
Guan X, Xu L, Liu J, Fei H, Wang C. Single-Cell Sequencing and Transcriptome Analysis Explored Changes in Midnolin-Related Immune Microenvironment and Constructed Combined Prognostic Model for Pancreatic Cancer. J Inflamm Res 2025; 18:2975-2990. [PMID: 40026303 PMCID: PMC11872096 DOI: 10.2147/jir.s503326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/22/2025] [Indexed: 03/05/2025] Open
Abstract
Background Pancreatic cancer has one of the worst prognoses of any malignant tumor. The value of MIDN, midnolin-related genes and midnolin-related immune infiltrating cells (MICs) in the prognosis of pancreatic cancer remains unknown. Methods Single-cell analysis were used to identify midnolin-related genes. Immune cell infiltration was obtained using CIBERSORT. The prognostic midnolin-related genes were identified through the utilization of Cox regression and the least absolute selection operator (LASSO) approach. The combined prognostic model was created using multifactorial Cox regression analysis. Survival analyses, immune microenvironment assessments, drug sensitivity checks were performed to evaluate the combined model performance. Finally, cellular experiments were carried out to confirm MIDN significance in pancreatic cancer. Results The combined model was constructed based on MIDN expression, prognostic model of 10 midnolin-related genes and M1 cell infiltration. Most immune checkpoint-related genes were expressed at greater levels in the low-risk group, suggesting a greater chance of immunotherapy's benefits. The most significant model gene, MIDN, was shown to have a function by cellular tests. In pancreatic cancer, MIDN knockdown drastically decreased pancreatic cancer cell lines' activity, proliferation, and invasive potential. Conclusion The combined model helped assess the prognosis of pancreatic cancer and offered fresh perspectives on immunotherapy in particular.
Collapse
Affiliation(s)
- Xiao Guan
- Department of Pancreatic and Gastric Surgery, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Lei Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Jinsong Liu
- Department of VIP Medical, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - He Fei
- Department of Pancreatic and Gastric Surgery, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Chengfeng Wang
- Department of Pancreatic and Gastric Surgery, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
29
|
Hayashi H, Seki S, Tomita T, Kato M, Ashihara N, Chano T, Sanjo H, Kawade M, Yan C, Sakai H, Tomida H, Tanaka M, Iwaya M, Taki S, Nakazawa Y, Soejima Y, Ueno Y, Hiratsuka S. Synthetic short mRNA prevents metastasis via innate-adaptive immunity. Nat Commun 2025; 16:1925. [PMID: 40000682 PMCID: PMC11862117 DOI: 10.1038/s41467-025-57123-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Although most cancer deaths are caused by metastasis, there are no effective therapeutic approaches. This study describes the efficacy of a short synthetic mRNA (s-mRNA) designed by the sequence of non-vesicular extracellular IL1β-mRNA found in the pre-metastatic lung of tumor-bearing mice. The administration of s-mRNA inhibits murine lung metastasis by inducing the innate and adaptive immune systems. s-mRNA binds to ZC3H12D, an RNA-binding protein on natural killer cells and cytotoxic T lymphocytes. The ZC3H12D-s-mRNA complex translocated to the nucleus without being involved in translation. This process induces cytolytic activity and cell death in cancer cells without inducing a cytokine storm, and immune cells retain their antitumor activity. Although the antitumor activity of cytotoxic lymphocytes declines as the disease progresses in cancer patients, s-mRNA induces sustained high killing capacities of natural killer cells and cytotoxic T lymphocytes from colon cancer patients. Therefore, s-mRNA could be a breakthrough solution to prevent metastasis.
Collapse
Affiliation(s)
- Hikaru Hayashi
- Department of Biochemistry and Molecular Biology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
- Department of Surgery, Division of Gastroenterological, Hepato-Biliary-Pancreatic, Transplantation and Pediatric Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Sayaka Seki
- Department of Biochemistry and Molecular Biology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Takeshi Tomita
- Department of Biochemistry and Molecular Biology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Masayoshi Kato
- Department of Biochemistry and Molecular Biology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Norihiro Ashihara
- Department of Biochemistry and Molecular Biology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Tokuhiro Chano
- Department of Medical Genetics, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, Japan
| | - Hideki Sanjo
- Department of Molecular and Cellular Immunology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Miwa Kawade
- Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu, Japan
| | - Chenhui Yan
- Department of Biochemistry and Molecular Biology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Hiroki Sakai
- Department of Biochemistry and Molecular Biology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
- Department of Surgery, Division of Gastroenterological, Hepato-Biliary-Pancreatic, Transplantation and Pediatric Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Hidenori Tomida
- Department of Biochemistry and Molecular Biology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
- Department of Surgery, Division of Gastroenterological, Hepato-Biliary-Pancreatic, Transplantation and Pediatric Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Miyuki Tanaka
- Department of Pediatrics, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Mai Iwaya
- Department of Laboratory Medicine, Shinshu University Hospital, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Shinsuke Taki
- Department of Molecular and Cellular Immunology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Yozo Nakazawa
- Department of Pediatrics, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Yuji Soejima
- Department of Surgery, Division of Gastroenterological, Hepato-Biliary-Pancreatic, Transplantation and Pediatric Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan
| | - Yoshihito Ueno
- Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Institute for Advanced Study, Gifu University, 1-1 Yanagido, Gifu, Japan
| | - Sachie Hiratsuka
- Department of Biochemistry and Molecular Biology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan.
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, Japan.
| |
Collapse
|
30
|
Li H, Zhou Y, Xiao J, Liu F. A comprehensive prognostic and immunological implications of Gremlin 1 in lung adenocarcinoma. Front Immunol 2025; 16:1529195. [PMID: 40066442 PMCID: PMC11891240 DOI: 10.3389/fimmu.2025.1529195] [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: 11/16/2024] [Accepted: 02/04/2025] [Indexed: 03/15/2025] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a prevalent form of lung cancer globally, known for its high invasiveness, metastatic potential, and notable heterogeneity, particularly in its response to immunotherapy. Gremlin 1 (GREM1) is implicated in tumor progression and poor prognosis in multiple cancers. However, GREM1's specific role in LUAD remains unclear. This study systematically examines GREM1 expression in LUAD and its association with tumor progression, immune microenvironment, and prognosis. Methods Gene expression data from the TCGA and GSE31210 databases were analyzed using Weighted Gene Co-expression Network Analysis (WGCNA), GO and KEGG enrichment analyses. The prognostic value of GREM1 was evaluated through survival analysis, Cox regression, and Kaplan-Meier curves. Additionally, immune microenvironment analysis was conducted to explore the relationship between GREM1 and immune cell infiltration. In vitro experiments, including Western blot and assays for cell proliferation, migration, and invasion, were performed to confirm the specific role of GREM1 in LUAD cells. Results GREM1 was significantly upregulated in tumor tissues and correlated with poor prognosis. Moreover, GREM1 was significantly associated with immune cell infiltration and immunotherapy response within the immune microenvironment. In vitro experiments confirmed that GREM1 overexpression significantly promoted LUAD cell proliferation, migration, and epithelial-mesenchymal transition (EMT), whereas GREM1 knockdown suppressed these functions. Conclusions A comprehensive analysis indicates that GREM1 is crucial in LUAD progression, with its overexpression predicting poor prognosis. GREM1 could be a potential therapeutic target for LUAD, providing insights for personalized therapy optimization.
Collapse
Affiliation(s)
- Hongyan Li
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yang Zhou
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jiaqing Xiao
- Institute of Disinfection and Infection Control, Heilongjiang Provincial Center for Disease Control and Prevention, Harbin, China
| | - Fang Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
31
|
Shi T, Zhang H, Chen Y. The m6A revolution: transforming tumor immunity and enhancing immunotherapy outcomes. Cell Biosci 2025; 15:27. [PMID: 39987091 PMCID: PMC11846233 DOI: 10.1186/s13578-025-01368-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/14/2025] [Indexed: 02/24/2025] Open
Abstract
N6-methyladenosine (m6A), the most prevalent RNA modification in eukaryotes, plays a critical role in the development and progression of various diseases, including cancer, through its regulation of RNA degradation, stabilization, splicing, and cap-independent translation. Emerging evidence underscores the significant role of m6A modifications in both pro-tumorigenic and anti-tumorigenic immune responses. In this review, we provide a comprehensive overview of m6A modifications and examine the relationship between m6A regulators and cancer immune responses. Additionally, we summarize recent advances in understanding how m6A modifications influence tumor immune responses by directly modulating immune cells (e.g., dendritic cells, tumor-associated macrophages, and T cells) and indirectly affecting cancer cells via mechanisms such as cytokine and chemokine regulation, modulation of cell surface molecules, and metabolic reprogramming. Furthermore, we explore the potential synergistic effects of targeting m6A regulators in combination with immune checkpoint inhibitor (ICI) therapies. Together, this review consolidates current knowledge on the role of m6A-mediated regulation in tumor immunity, offering insights into how a deeper understanding of these modifications may identify patients who are most likely to benefit from immunotherapies.
Collapse
Affiliation(s)
- Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China.
| | - Huan Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China
| | - Yueqiu Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China.
| |
Collapse
|
32
|
Gao R, Wu P, Yin X, Zhuang L, Meng X. Deep analysis of the trials and major challenges in the first-line treatment for patients with extensive-stage small cell lung cancer. Int Immunopharmacol 2025; 148:114116. [PMID: 39847950 DOI: 10.1016/j.intimp.2025.114116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 01/25/2025]
Abstract
The median overall survival (OS) is approximately 10 months when chemotherapy alone is the first-line treatment for extensive-stage small cell lung cancer (ES-SCLC). The approval of the two PD-L1 inhibitors, atezolizumab and durvalumab, marked the beginning of the immunotherapy era for ES-SCLC. Serplulimab, as the first PD-1 inhibitor to achieve success in the first-line treatment of ES-SCLC, has not only demonstrated significant improvements in patient survival outcomes but also ushered in a new era for PD-1 inhibitors in the treatment of ES-SCLC. Recently, antiangiogenic agents with chemo-immunotherapy have achieved breakthroughs in first-line ES-SCLC treatment. Improving the clinical benefits of individualized treatment for patients with ES-SCLC remains challenging. Challenges include identifying biomarkers for targeted therapy, exploring new treatments, developing new medicines, and classifying SCLC molecular subtypes. This review provides an in-depth analysis of research on first-line ES-SCLC treatment. Additionally, it discusses advances in ES-SCLC treatment.
Collapse
Affiliation(s)
- Ran Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Peizhu Wu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Xiaoyan Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Lulu Zhuang
- Cheeloo College of Cancer Center, Shandong University, Jinan, Shandong, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China.
| |
Collapse
|
33
|
Fan M, Zhu Y, Qian L, Hu C, Ding H. Association between preoperative inflammatory status via CALLY index and postoperative pneumonia occurrence in resectable esophageal squamous cell carcinoma patients: a retrospective cohort study. Front Oncol 2025; 15:1486983. [PMID: 40034601 PMCID: PMC11872739 DOI: 10.3389/fonc.2025.1486983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 01/29/2025] [Indexed: 03/05/2025] Open
Abstract
Background Postoperative pneumonia significantly affects recovery and prognosis in patients with esophageal squamous cell carcinoma. The CALLY index, derived from preoperative hematological parameters, may serve as a predictive marker for such complications. Objectives To assess the association between preoperative inflammatory status via the CALLY index and the occurrence of postoperative pneumonia in patients with resectable ESCC. Methods A retrospective cohort study was conducted from January 2020 to December 2022 at The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University. A total of 215 patients who met inclusion criteria were analyzed. Clinical data, including CALLY indices calculated preoperatively, were collected. Propensity score matching was applied to minimize confounding biases. The predictive value of the CALLY index was assessed using receiver operating characteristic analysis, and logistic regression was used to identify factors associated with postoperative pneumonia. Results ROC curve analysis demonstrated the CALLY index had an area under the curve of 0.764 for predicting postoperative pneumonia, with a cutoff value of 1.97 achieving 67.69% sensitivity and 84.67% specificity. In multivariate analysis, a lower CALLY index was significantly associated with increased pneumonia risk, independent of other factors (adjusted OR = 0.66, p < 0.001). High CALLY index scores correlated with a decreased likelihood of postoperative pneumonia, reinforcing its utility as a non-invasive prognostic marker. Conclusions The CALLY index is a robust, independent predictor of postoperative pneumonia in patients with resectable ESCC. Preoperative assessment of this index could enhance risk stratification and guide proactive management strategies to improve postoperative outcomes.
Collapse
Affiliation(s)
| | | | | | - Chuanxian Hu
- Department of Cardiothoracic Surgery, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian, China
| | - Hui Ding
- Department of Cardiothoracic Surgery, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian, China
| |
Collapse
|
34
|
Muraoka S, Baba T, Akazawa T, Katayama KI, Kusumoto H, Yamashita S, Kohjimoto Y, Iwabuchi S, Hashimoto S, Hara I, Inoue N. Tumor-derived lactic acid promotes acetylation of histone H3K27 and differentiation of IL-10-producing regulatory B cells through direct and indirect signaling pathways. Int J Cancer 2025; 156:840-852. [PMID: 39482832 DOI: 10.1002/ijc.35229] [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: 07/29/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 11/03/2024]
Abstract
Tumor cells are known to enhance glycolysis, even under normoxic conditions, via the Warburg effect, producing excess lactic acid in the tumor microenvironment. Lactic acid enhances the IL-23/IL-17 pathway and induces chronic inflammation. The acidic microenvironment formed by lactic acid suppresses immune cell proliferation and activation. In the present study, we clarified that lactic acid had two novel activities for immune cells. First, lactic acid specifically enhanced acetylation at lysine 27 of histone H3 (H3K27ac) in splenic B cells and monocytes/macrophages, and this epigenetically up-regulates the expression of genes. Acetylation and methylation of other residues of histone H3 were rarely induced. Second, lactic acid induced a particularly-marked enhancement of Il10 gene expression in B cells, leading to an increase in IL-10-producing regulatory B (Breg) cells. Furthermore, two pathways should be involved in both the enhancement of H3K27ac and the induction of Breg cells by lactic acid: a direct pathway that enhances the CD40 signal in B cells, and an indirect pathway that affects B cells by activating the exchange protein directly activated by cAMP (EPAC) 1/2 in non-B cells. In tumor-bearing mice, the levels of H3K27ac of tumor-infiltrating B cells were significantly higher than splenic B cells and were suppressed by intraperitoneal injection of the EPAC1/2 inhibitor. In conclusion, tumor-derived lactic acid increases H3K27ac and IL-10-producing Breg cells, causing the suppression of anti-tumor immunity.
Collapse
Affiliation(s)
- Satoshi Muraoka
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | - Takashi Baba
- Department of Molecular Genetics, Wakayama Medical University, Wakayama, Japan
| | - Takashi Akazawa
- Department of Cancer Drug Discovery and Development, Research Center, Osaka International Cancer Institute, Chuo-ku, Osaka, Japan
| | - Kei-Ichi Katayama
- Department of Molecular Genetics, Wakayama Medical University, Wakayama, Japan
| | - Hiroki Kusumoto
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | | | - Yasuo Kohjimoto
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | - Sadahiro Iwabuchi
- Department of Molecular Pathophysiology, Wakayama Medical University, Wakayama, Japan
| | - Shinichi Hashimoto
- Department of Molecular Pathophysiology, Wakayama Medical University, Wakayama, Japan
| | - Isao Hara
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | - Norimitsu Inoue
- Department of Molecular Genetics, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
35
|
He X, Guan XY, Li Y. Clinical significance of the tumor microenvironment on immune tolerance in gastric cancer. Front Immunol 2025; 16:1532605. [PMID: 40028336 PMCID: PMC11868122 DOI: 10.3389/fimmu.2025.1532605] [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: 11/22/2024] [Accepted: 01/30/2025] [Indexed: 03/05/2025] Open
Abstract
In the realm of oncology, the tumor microenvironment (TME)-comprising extracellular matrix components, immune cells, fibroblasts, and endothelial cells-plays a pivotal role in tumorigenesis, progression, and response to therapeutic interventions. Initially, the TME exhibits tumor-suppressive properties that can inhibit malignant transformation. However, as the tumor progresses, various factors induce immune tolerance, resulting in TME behaving in a state that promotes tumor growth and metastasis in later stages. This state of immunosuppression is crucial as it enables TME to change from a role of killing tumor cells to a role of promoting tumor progression. Gastric cancer is a common malignant tumor of the gastrointestinal tract with an alarmingly high mortality rate. While chemotherapy has historically been the cornerstone of treatment, its efficacy in prolonging survival remains limited. The emergence of immunotherapy has opened new therapeutic pathways, yet the challenge of immune tolerance driven by the gastric cancer microenvironment complicates these efforts. This review aims to elucidate the intricate role of the TME in mediating immune tolerance in gastric cancer and to spotlight innovative strategies and clinical trials designed to enhance the efficacy of immunotherapeutic approaches. By providing a comprehensive theoretical framework, this review seeks to advance the understanding and application of immunotherapy in the treatment of gastric cancer, ultimately contributing to improved patient outcomes.
Collapse
Affiliation(s)
- Xiangyang He
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin-Yuan Guan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Clinical Oncology, The University of Hongkong, Hong Kong, Hong Kong SAR, China
| | - Yan Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
36
|
Xie B, Xiao Z, Ling J, Peng Y, Chen T. Exploring the application of metal-based photothermal agents in photothermal therapy combined with immune checkpoint therapy. Front Pharmacol 2025; 16:1553158. [PMID: 40017598 PMCID: PMC11865196 DOI: 10.3389/fphar.2025.1553158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 01/23/2025] [Indexed: 03/01/2025] Open
Abstract
Photothermal therapy (PTT), a popular local treatment that uses heat to ablate tumors, has limited efficacy in addressing metastatic and deeply located tumors when used alone. Integrating PTT with immunotherapy not only yields a synergistic effect but also promotes cancer regression and confers the benefit of immune memory, which can surmount the challenges faced by PTT when used in isolation. Metal-based nanomaterials, renowned for their superior photothermal conversion efficiency and distinctive photochemical properties, have been extensively researched and applied in the field of PTT. This review summarizes the latest developments in combination therapies, with a specific focus on the combination of PTT and immune checkpoint therapy (ICT) for cancer treatment, including a comprehensive overview of the recent advancements in noble metal-based and 2D transition metal chalcogenides (TMDCs)-based photothermal agents, and their anticancer effect when combining PTT with immune checkpoint blockades (anti-CTLA-4 and anti-PD-L1) therapy. The goal of this review is to present an overview of the application, current challenges and future prospects of metal-based photothermal agents in PTT combined with ICT for cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Yichao Peng
- Department of Pharmacy and General Surgery of Puning People’s Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), College of Chemistry and Materials Science, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangdong, China
| | - Tianfeng Chen
- Department of Pharmacy and General Surgery of Puning People’s Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), College of Chemistry and Materials Science, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangdong, China
| |
Collapse
|
37
|
Han R, Luo Y, Gao J, Zhou H, Wang Y, Chen J, Zheng G, Ling C. HDAC3: A Multifaceted Modulator in Immunotherapy Sensitization. Vaccines (Basel) 2025; 13:182. [PMID: 40006729 PMCID: PMC11860249 DOI: 10.3390/vaccines13020182] [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: 12/18/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Histone deacetylase 3 (HDAC3) has emerged as a critical epigenetic regulator in tumor progression and immune modulation, positioning it as a promising target for enhancing cancer immunotherapy. This work comprehensively explores HDAC3's multifaceted roles, focusing on its regulation of key immune-modulatory pathways such as cGAS-STING, ferroptosis, and the Nrf2/HO-1 axis. These pathways are central to tumor immune evasion, antigen presentation, and immune cell activation. Additionally, the distinct effects of HDAC3 on various immune cell types-including its role in enhancing T cell activation, restoring NK cell cytotoxicity, promoting dendritic cell maturation, and modulating macrophage polarization-are thoroughly examined. These findings underscore HDAC3's capacity to reshape the tumor immune microenvironment, converting immunologically "cold tumors" into "hot tumors" and thereby increasing their responsiveness to immunotherapy. The therapeutic potential of HDAC3 inhibitors is highlighted, both as standalone agents and in combination with immune checkpoint inhibitors, to overcome resistance and improve treatment efficacy. Innovative strategies, such as the development of selective HDAC3 inhibitors, advanced nano-delivery systems, and integration with photodynamic or photothermal therapies, are proposed to enhance treatment precision and minimize toxicity. By addressing challenges such as toxicity, patient heterogeneity, and resistance mechanisms, this study provides a forward-looking perspective on the clinical application of HDAC3 inhibitors. It highlights its significant potential in personalized cancer immunotherapy, paving the way for more effective treatments and improved outcomes for cancer patients.
Collapse
Affiliation(s)
- Rui Han
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Yujun Luo
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Jingdong Gao
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
- Oncology Department, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine Suzhou, Suzhou 215009, China
| | - Huiling Zhou
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Yuqian Wang
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Jiaojiao Chen
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Guoyin Zheng
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Changquan Ling
- Oncology Department of Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; (R.H.)
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
38
|
Gu T, Qi H, Wang J, Sun L, Su Y, Hu H. Identification of T cell dysfunction molecular subtypes and exploration of potential immunotherapy targets in BRAF V600E-mutant colorectal cancer. Discov Oncol 2025; 16:163. [PMID: 39934467 DOI: 10.1007/s12672-025-01930-8] [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: 11/21/2024] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Immunotherapy is an effective treatment for BRAF V600E-mutant colorectal cancer, but currently, only a few benefit from it. Therefore, exploring new immunotherapy strategies is essential. METHODS We obtained RNA sequencing data and clinical information of colorectal cancer patients from the TCGA and GEO databases. The impact of the BRAF V600E mutation on tumor microenvironment characteristics, gene expression, and signaling pathways was evaluated using bioinformatics approaches. Weighted gene co-expression network analysis (WGCNA) were used to identify core genes associated with T cell dysfunction. Consensus clustering was applied for subtype construction. Least Absolute Shrinkage and Selection Operator (LASSO) and Random Forest (RF) algorithms were employed to filter potential immunotherapy targets. RESULTS We found that BRAF V600E mutation has a complex impact on the immune profile of colorectal cancer. It increases immune cell infiltration and activates immune-related signaling pathways, yet it also severely restricts T cell function. We subsequently identified 39 core genes associated with T cell dysfunction and constructed subtypes of BRAF V600E colorectal cancer based on their expression profiles. Significant heterogeneity was observed between these subtypes in immune signaling pathway activity, immune infiltration patterns, immune phenotype scores, and mechanisms of resistance to immunotherapy. Ultimately, using machine learning algorithms and bioinformatics validation, we identified IDO1 as a potential immunotherapy targets for BRAF V600E-mutant colorectal cancer. CONCLUSION This study constructed novel T cell dysfunction molecular subtypes for BRAF V600E-mutant colorectal cancer and identified IDO1 as a potential immunotherapy target, providing a new strategy for immunotherapy.
Collapse
Affiliation(s)
- Tiefeng Gu
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, China
| | - Haonan Qi
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, China
| | - Jiaqi Wang
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, China
| | - Liangwei Sun
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, China
| | - Yongqi Su
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, China
| | - Hanqing Hu
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, China.
| |
Collapse
|
39
|
Wu C, Fang S, Wu L, Mi Z, Yin Y, Liao Y, Zhao Y, Wang T, Na J. Identification of the entosis-related prognostic signature and tumour microenvironment in hepatocellular carcinoma on the basis of bioinformatics analysis and experimental validation. Clin Exp Med 2025; 25:55. [PMID: 39937284 PMCID: PMC11821697 DOI: 10.1007/s10238-025-01580-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 01/28/2025] [Indexed: 02/13/2025]
Abstract
Liver cancer ranks among the deadliest cancers worldwide. Entosis, a recently uncovered method of cell death, has not yet been fully explored for its relevance to HCC. A bioinformatics analysis was performed to determine the expression and mutational landscapes of Entosis-related genes (ERGs). A subset of differentially expressed Entosis-related genes (DEERGs) was generated. A risk model for entosis was subsequently constructed employing LASSO and Cox regression methodologies. The correlations among ERGs, genes associated with risk, the developed risk model, and the immune context of the tumour were explored. Furthermore, the study investigated the varying drug sensitivities between high-risk and slight-risk patient groups. The expression patterns of four pivotal risk genes were delineated via qRT‒PCR and WB. A prognostic model comprising four DEERGs (KIF18A, SPP1, LCAT and TRIB3) was developed. The ability of this model to predict the survival outcomes of patients with HCC was confirmed through receiver operating characteristic curve analysis. Patients were grouped according to their risk assessments, revealing that the low-risk population demonstrated a more favourable survival outcome than did the high-risk population. The high-risk population presented reduced tumour stroma, immune and ESTIMATE scores, along with an increased proportion of cancer stem cells and tumour mutation burden. Additionally, a connection between the risk model and the responsiveness of various chemotherapy drugs as well as the efficacy of immunotherapies in patients was noted. These findings provide significant guidance for the development of targeted clinical treatment strategies. qRT‒PCR and WB analysis revealed that the gene expression of KIF18A and SPP1 were elevated in HCCLM3 cells compared with that in THLE2 cells; whereas, the expression level of LCAT and TIRB3 was decreased. The four genes KIF18A, SPP1, LCAT and TRIB3 could effectively predict the survival prognosis of patients with liver cancer. KIF18A and SPP1 were elevated in HCC tissues compared with that in THLE2 cells.
Collapse
Affiliation(s)
- Chen Wu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Shixu Fang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Liangliang Wu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Zhengcheng Mi
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yao Yin
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yuan Liao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yongxiang Zhao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Tinghua Wang
- Laboratory Animal Department, Kunming Medical University, Kunming, 650031, Yunnan, China.
| | - Jintong Na
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| |
Collapse
|
40
|
Gao C, Chen L, Zhao L, Su Y, Ma M, Zhang W, Hong X, Xiao L, Xu B, Hu T. Apatinib Degrades PD-L1 and Reconstitutes Colon Cancer Microenvironment via the Regulation of Myoferlin. Cancers (Basel) 2025; 17:524. [PMID: 39941891 PMCID: PMC11816266 DOI: 10.3390/cancers17030524] [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: 12/05/2024] [Revised: 01/20/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND For most colorectal cancer (CRC) patients, expanding the benefits of immunotherapy, particularly through blocking programmed cell death-1 (PD-1) and its ligand (PD-L1), is crucial, especially in cases with limited response to neoadjuvant therapy. This study investigates the role of Myoferlin (MYOF) as a novel target in CRC immunotherapy. METHODS Human CRC cell lines (RKO, HCT116), normal intestinal epithelial cells (HIEC-6), and the murine CRC cell line MC38 were used to study the effects of apatinib and MYOF in CRC cells. RNA sequencing, the CPTAC and TCGA databases, and other molecular and cellular methods were applied to disclose the mechanisms involved. A series of mouse models were established to assess the effects of apatinib and MYOF knockdown on tumor progression, immune cell infiltration, and immune checkpoint protein response. RESULTS We found that MYOF is overexpressed in CRC and linked to immune cell infiltration and checkpoint expression. Suppression of MYOF expression significantly inhibited CRC cell proliferation and migration, as well as reduced PD-L1 protein levels. Integrative analysis showed that apatinib modulates MYOF expression via VEGFR2, resulting in decreased PD-L1 expression, increased CD8+ T cell infiltration, and reduced pro-tumor M2 macrophages. Animal experiments further revealed that apatinib treatment or MYOF knockdown enhanced the efficacy of immune checkpoint blockade (ICB) in CRC. CONCLUSIONS These findings highlight novel antitumor mechanisms of MYOF and suggest that combining apatinib with ICB therapy may improve CRC treatment outcomes, offering a promising strategy to enhance immune responses.
Collapse
Affiliation(s)
- Chunyi Gao
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (C.G.); (L.C.); (Y.S.); (M.M.); (W.Z.); (X.H.)
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, Ganzhou 341000, China
| | - Lu Chen
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (C.G.); (L.C.); (Y.S.); (M.M.); (W.Z.); (X.H.)
| | - Lingying Zhao
- Department of Laboratory Medicine, Shenzhen Children’s Hospital, Shenzhen 518038, China;
| | - Yongcheng Su
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (C.G.); (L.C.); (Y.S.); (M.M.); (W.Z.); (X.H.)
| | - Miaomiao Ma
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (C.G.); (L.C.); (Y.S.); (M.M.); (W.Z.); (X.H.)
| | - Wenqing Zhang
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (C.G.); (L.C.); (Y.S.); (M.M.); (W.Z.); (X.H.)
| | - Xiaoting Hong
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (C.G.); (L.C.); (Y.S.); (M.M.); (W.Z.); (X.H.)
| | - Li Xiao
- Department of Oncology, Zhongshan Hospital of Xiamen University, Xiamen 361004, China;
| | - Beibei Xu
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Tianhui Hu
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (C.G.); (L.C.); (Y.S.); (M.M.); (W.Z.); (X.H.)
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
41
|
Wang TY, Hu HG, Zhao L, Zhuo SH, Su JY, Feng GH, Li YM. EXO TLR1/2-STING: A Dual-Mechanism Stimulator of Interferon Genes Activator for Cancer Immunotherapy. ACS NANO 2025; 19:5017-5028. [PMID: 39846950 DOI: 10.1021/acsnano.4c18056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
As natural agonists of the stimulator of interferon genes (STING) protein, cyclic dinucleotides (CDNs) can activate the STING pathway, leading to the expression of type I interferons and various cytokines. Efficient activation of the STING pathway in antigen-presenting cells (APCs) and tumor cells is crucial for antitumor immune response. Tumor-derived exosomes can be effectively internalized by APCs and tumor cells and have excellent potential to deliver CDNs to the cytoplasm of APCs and tumor cells. Here, we leverage tumor exosomes as a delivery platform, designing an EXOTLR1/2-STING loaded with CDNs. To achieve efficient loading of CDNs onto exosomes, we chemically conjugated CDNs with Pam3CSK4, a compound featuring multiple fatty acid chains, resulting in Pam3CSK4-CDGSF. Utilizing the high lipophilicity of Pam3CSK4, Pam3CSK4-CDGSF could be efficiently loaded onto the exosomes through simple incubation. Moreover, as an agonist for Toll-like receptor 1/2, Pam3CSK4 also exhibits robust immunological synergistic effects in conjunction with CDNs. EXOTLR1/2-STING effectively induced the activation of APCs and triggered tumor cell death, producing a favorable antitumor therapeutic effect. It also demonstrated significant synergistic effects with immune checkpoint therapies.
Collapse
Affiliation(s)
- Tian-Yang Wang
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Hong-Guo Hu
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Lang Zhao
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Shao-Hua Zhuo
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Jing-Yun Su
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Geng-Hui Feng
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Yan-Mei Li
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
- Beijing Institute for Brain Disorders, Beijing 100069, China
- Center for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, China
| |
Collapse
|
42
|
Su X, Zhang M, Zhu H, Cai J, Wang Z, Xu Y, Wang L, Shen C, Cai M. Mechanisms of T-cell Depletion in Tumors and Advances in Clinical Research. Biol Proced Online 2025; 27:5. [PMID: 39905296 PMCID: PMC11792740 DOI: 10.1186/s12575-025-00265-6] [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: 11/10/2024] [Accepted: 01/20/2025] [Indexed: 02/06/2025] Open
Abstract
T lymphocytes (T cells) are essential components of the adaptive immune system that play a vital role in identifying and eliminating infected and tumor cells. In tumor immunotherapy, T cells have emerged as a promising therapeutic strategy due to their high specificity, potent cytotoxic capability, long-lasting immune memory, and adaptability within immunotherapeutic approaches. However, tumors can evade the immune system by depleting T cells through various mechanisms, such as inhibitory receptor signaling, metabolic exhaustion, and physical barriers within the tumor microenvironment. This review provided an overview of the mechanisms underlying T-cell depletion in tumors and discussed recent advances in clinical research related to T-cell immunotherapy for tumors. It highlighted the need for in-depth studies on key issues such as indications, dosage, and sequencing of combined therapeutic strategies tailored to different patients and tumor types, providing practical guidance for individualized treatment. Future research on T-cell depletion would be necessary to uncover the fundamental mechanisms and laws of T-cell depletion, offering both theoretical insights and practical guidance for the selection and optimization of tumor immunotherapy. Furthermore, interdisciplinary, cross-disciplinary, and international collaborative innovations are necessary for developing more effective and safer treatments for tumor patients.
Collapse
Affiliation(s)
- Xiangfei Su
- China Association of Chinese Medicine, Beijing, China
| | - Mi Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, No. 300, Shouchun Road, Hefei, Anhui, 230061, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Hong Zhu
- Tongling People's Hospital, Tongling, Anhui, China
| | - Jingwen Cai
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhen Wang
- Anhui Provincial Children's Hospital, Hefei, Anhui, China
| | - Yuewei Xu
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, No. 300, Shouchun Road, Hefei, Anhui, 230061, China
| | - Li Wang
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, No. 300, Shouchun Road, Hefei, Anhui, 230061, China
| | - Chen Shen
- Key Laboratory of Data Science and Innovation and Development of Traditional Chinese Medicine and Social Sciences of Anhui Province, Anhui University of Chinese Medicine, No. 350, Longzihu Road, Hefei, Anhui, 230012, China.
| | - Ming Cai
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, No. 300, Shouchun Road, Hefei, Anhui, 230061, China.
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China.
| |
Collapse
|
43
|
Qi J, Zhou M, Yang N, Ma H, He M, Wu G, Ge C, Jin L, Cheng L, Liao W, Ren H, Lei C. TUBA1B as a novel prognostic biomarker correlated with immunosuppressive tumor microenvironment and immunotherapy response. Front Pharmacol 2025; 16:1517887. [PMID: 39968182 PMCID: PMC11832512 DOI: 10.3389/fphar.2025.1517887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 01/03/2025] [Indexed: 02/20/2025] Open
Abstract
Background: Tubulin alpha 1b (TUBA1B) is a key microtubule protein essential for maintaining cellular structure and function. This protein contributes significantly to cytoskeletal formation and is implicated in various diseases. Despite its fundamental roles, TUBA1B's impact on tumor prognosis and the tumor immune microenvironment across cancer types remains inadequately understood. Methods To elucidate TUBA1B's role in cancer prognosis and immune response, we conducted a comprehensive analysis, integrating data from established databases such as The Cancer Genome Atlas, Genotype Tissue Expression, Cancer Cell Lineage Encyclopedia, Human Protein Atlas, Kaplan-Meier Plotter, cBioPortal, TIMER, and ImmuCellAI, along with a large-scale clinical study and immunotherapy cohort. We also conducted in vitro functional assays to assess TUBA1B's functional role in tumor cells, allowing for a detailed examination of its relationship with cancer prognosis and immune modulation. Results: Our findings indicate that TUBA1B expression is dysregulated across multiple cancers, correlating strongly with poor survival outcomes and advanced pathological stages. Functional enrichment analyses further revealed that TUBA1B regulates key cell cycle processes, driving tumor proliferation, migration, and invasion. It also influences immune functions within both the innate and adaptive immune systems, affecting immune-related signaling pathways. These insights underscore TUBA1B's multifaceted role in cancer progression and immune response. Conclusion: This study highlights TUBA1B's potential as a human oncogene with substantial roles in tumorigenesis and immune regulation. Elevated TUBA1B levels are associated with an immunosuppressive tumor microenvironment, impacting cancer progression and treatment outcomes. Targeting TUBA1B may offer promising therapeutic avenues for enhancing cancer treatment, offering new perspectives for innovative anti-tumor strategies with high clinical impact.
Collapse
Affiliation(s)
- Juntao Qi
- Rehabilitation Medicine Department, Hunan Aerospace Hospital, Hunan Normal University, Changsha, Hunan, China
- Research Center of Clinical Medicine, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, China
| | - Mingming Zhou
- Department of Critical Care Medicine, Chongqing University Affiliated Cancer Hospital, Chongqing, China
| | - Na Yang
- Laboratory of Oncology and Immunology, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Huiyun Ma
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Min He
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Gujie Wu
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Chang Ge
- School of Medicine, Wuhan University, Wuhan, China
| | - Liuyin Jin
- School of Medicine, Wuhan University, Wuhan, China
| | - Lin Cheng
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Liao
- Department of Otolaryngology and Head and Neck Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Hefei Ren
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Caiyun Lei
- Rehabilitation Medicine Department, Hunan Aerospace Hospital, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
44
|
Liu T, Lu C, Jiang X, Wang Y, Chen Z, Qi C, Xu X, Feng X, Wang Q. Nano-Based Strategies Aiming at Tumor Microenvironment for Improved Cancer Therapy. Mol Pharm 2025; 22:647-677. [PMID: 39818981 DOI: 10.1021/acs.molpharmaceut.4c01267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Malignant tumors pose a considerable threat to human life and health. Traditional treatments, such as radiotherapy and chemotherapy, often lack specificity, leading to collateral damage to normal tissues. Tumor microenvironment (TME) is characterized by hypoxia, acidity, redox imbalances, and elevated ATP levels factors that collectively promote tumor growth and metastasis. This review provides a comprehensive overview of the nanoparticles developed in recent years for TME-responsive strategies or TME-modulating methods for tumor therapy. The TME-responsive strategies focus on designing and synthesizing nanoparticles that can interact with the tumor microenvironment to achieve precisely controlled drug release. These nanoparticles activate drug release under specific conditions within the tumor environment, thereby enhancing the efficacy of the drugs while reducing toxicity to normal cells. Moreover, simply eliminating tumor cells does not fundamentally solve the problem. Only by comprehensively regulating the TME to make it unsuitable for tumor cell survival and proliferation can we achieve more thorough therapeutic effects and reduce the risk of tumor recurrence. TME regulation strategies aim to suppress the growth and metastasis of tumor cells by modulating various components within the TME. These strategies not only improve treatment outcomes but also have the potential to lay the foundation for future personalized cancer therapies.
Collapse
Affiliation(s)
- Tianhui Liu
- College of Life Science and Technology, Changchun University of Science and Technology, 7089 Satellite Road, Changchun 130022, China
| | - Changshun Lu
- College of Life Science and Technology, Changchun University of Science and Technology, 7089 Satellite Road, Changchun 130022, China
| | - Xue Jiang
- College of Life Science and Technology, Changchun University of Science and Technology, 7089 Satellite Road, Changchun 130022, China
| | - Yutong Wang
- College of Life Science and Technology, Changchun University of Science and Technology, 7089 Satellite Road, Changchun 130022, China
| | - Zhengrong Chen
- College of Life Science and Technology, Changchun University of Science and Technology, 7089 Satellite Road, Changchun 130022, China
| | - Chunshuang Qi
- College of Life Science and Technology, Changchun University of Science and Technology, 7089 Satellite Road, Changchun 130022, China
| | - Xiaoru Xu
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun 130117, China
| | - Xiangru Feng
- College of Life Science and Technology, Changchun University of Science and Technology, 7089 Satellite Road, Changchun 130022, China
| | - Qingshuang Wang
- College of Life Science and Technology, Changchun University of Science and Technology, 7089 Satellite Road, Changchun 130022, China
| |
Collapse
|
45
|
Zheng X, Zhang X, Li D, Wang Z, Zhang J, Li J, Li Y. Integrative bioinformatics and experimental analyses identify U2SURP as a novel lactylation-related prognostic signature in esophageal carcinoma. Immunol Res 2025; 73:45. [PMID: 39900790 DOI: 10.1007/s12026-024-09589-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/27/2024] [Indexed: 02/05/2025]
Abstract
The lactylation modification has been implicated in several cancer types; however, the role of lactylation modification-related genes in esophageal carcinoma (EC) remains underexplored. Utilizing a set of 16 lactylation modification-related genes, cohorts of patients with EC were stratified into two distinct clusters, characterized by significant disparities in both survival outcomes and the immune microenvironment. An extensive bioinformatics analysis unveiled 382 differentially expressed genes (DEGs) between these two clusters. A subsequent univariate Cox regression analysis identified 24 DEGs specifically associated with lactylation, forming the basis of a constructed lactylation-related score. The resultant lactylation-related score exhibited notable predictive efficacy for survival and other clinicopathological traits, which was validated through calibration curves, Kaplan-Meier survival curves and the Wilcoxon test. Moreover, the lactylation-related score displayed a close correlation with immune cell infiltration in EC. Notable differential expressions of immune checkpoints and regulators were observed between groups stratified by low and high lactylation scores, with the latter exhibiting a more favorable response to anti-PD-1/PD-L1 therapy. Furthermore, the expression profile of U2 snRNP associated SURP domain containing (U2SURP), a constituent of the lactylation-related score, underwent both ex vivo and in vitro validation. The expression of U2SURP was significantly associated with lactylation levels, histological grade and tumor stage. Notably, knockdown of U2SURP expression inhibited the lactylation levels, immune genes IL-1A and IL-1B, proliferation, migration and invasion of EC cells. In conclusion, the lactylation-related score developed in the present study showed promise in predicting the prognosis and immunotherapeutic responses among patients with EC. Moreover, the identification of U2SUPR as a novel oncogene in EC suggests its potential as a prospective therapeutic target for EC treatment.
Collapse
Affiliation(s)
- Xuan Zheng
- The Cancer Institute, Tangshan People's Hospital, Tangshan, 063001, China
- Hebei Key Laboratory of Molecular Oncology, Tangshan, 063001, China
| | - Xiaoru Zhang
- Nuclear Medicine Laboratory, Tangshan People's Hospital, Tangshan, 063001, China
| | - Dan Li
- The Cancer Institute, Tangshan People's Hospital, Tangshan, 063001, China
- Hebei Key Laboratory of Molecular Oncology, Tangshan, 063001, China
| | - Zhuo Wang
- The Cancer Institute, Tangshan People's Hospital, Tangshan, 063001, China
- Hebei Key Laboratory of Molecular Oncology, Tangshan, 063001, China
| | - Jun Zhang
- The Cancer Institute, Tangshan People's Hospital, Tangshan, 063001, China
- Hebei Key Laboratory of Molecular Oncology, Tangshan, 063001, China
| | - Jingwu Li
- The Cancer Institute, Tangshan People's Hospital, Tangshan, 063001, China.
- Hebei Key Laboratory of Molecular Oncology, Tangshan, 063001, China.
| | - Yufeng Li
- The Cancer Institute, Tangshan People's Hospital, Tangshan, 063001, China.
- Hebei Key Laboratory of Molecular Oncology, Tangshan, 063001, China.
| |
Collapse
|
46
|
Ma K, Wang L, Li W, Tang T, Ma B, Zhang L, Zhang L. Turning cold into hot: emerging strategies to fire up the tumor microenvironment. Trends Cancer 2025; 11:117-134. [PMID: 39730243 DOI: 10.1016/j.trecan.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/29/2024]
Abstract
The tumor microenvironment (TME) is a complex, highly structured, and dynamic ecosystem that plays a pivotal role in the progression of both primary and metastatic tumors. Precise assessment of the dynamic spatiotemporal features of the TME is crucial for understanding cancer evolution and designing effective therapeutic strategies. Cancer is increasingly recognized as a systemic disease, influenced not only by the TME, but also by a multitude of systemic factors, including whole-body metabolism, gut microbiome, endocrine signaling, and circadian rhythm. In this review, we summarize the intrinsic, extrinsic, and systemic factors contributing to the formation of 'cold' tumors within the framework of the cancer-immunity cycle. Correspondingly, we discuss potential strategies for converting 'cold' tumors into 'hot' ones to enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Kaili Ma
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China
| | - Lin Wang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China
| | - Wenhui Li
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China
| | - Tingting Tang
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Bo Ma
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Liyuan Zhang
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, China; PRAG Therapy Center, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, China.
| | - Lianjun Zhang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China; Key Laboratory of Synthetic Biology Regulatory Element, Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
47
|
Han X, Wang X, Yan J, Song P, Wang Y, Kang Y, Rauf A, Zhang H. Multifunctional biosynthesized magnetosome for multimodal imaging and combined therapy of tumor. Mater Today Bio 2025; 30:101429. [PMID: 39839492 PMCID: PMC11750283 DOI: 10.1016/j.mtbio.2024.101429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/23/2025] Open
Abstract
The large recruitment of tumor-associated macrophages and low exposure of tumor-associated antigens in tumor microenvironment have severely suppress the efficacy of anti-tumor immunotherapy. Herein, biosynthesized magnetosome (Mag) from bacteria was loaded with photothermal/photodynamic agent/near infrared (NIR) fluorescence dye (IR780) and further modified with lipid-PEG-c(RGDyK) through biomembrane, forming IMagRGD for fluorescence imaging, magnetic resonance imaging, immunotherapy and photodynamic/photothermal therapy. After intravenous injection into B16F10 tumor-bearing mice, IMagRGD could efficiently accumulate in tumor tissues based on near infrared (NIR) fluorescence and magnetic resonance dual-modality imaging, and repolarize tumor-associated macrophages (TAMs) from M2 phenotype to M1 phenotype, significantly improving the effect of tumor immunotherapy. Moreover, photothermal and photodynamic effect of IR780 could kill tumor cells and elicit immunogenic cell death to mediate anti-tumor immunity, promoting dendritic cells (DCs) maturation and then activating specific effector T cells to further eliminate tumor cells. This study provides a new approach for reversing the activity of tumor immunosuppressive microenvironment and strengthening the efficiency of tumor photoimmunotherapy.
Collapse
Affiliation(s)
- Xiaoqing Han
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, 130024, China
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Xingbo Wang
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jiao Yan
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Panpan Song
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yanjing Wang
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yaqing Kang
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Ambar, 23430, Pakistan
| | - Haiyuan Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| |
Collapse
|
48
|
Estephan H, Tailor A, Parker R, Kreamer M, Papandreou I, Campo L, Easton A, Moon EJ, Denko NC, Ternette N, Hammond EM, Giaccia AJ. Hypoxia promotes tumor immune evasion by suppressing MHC-I expression and antigen presentation. EMBO J 2025; 44:903-922. [PMID: 39753950 PMCID: PMC11790895 DOI: 10.1038/s44318-024-00319-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 09/29/2024] [Accepted: 10/14/2024] [Indexed: 02/05/2025] Open
Abstract
Hypoxia is a common feature of solid tumors that has previously been linked to resistance to radiotherapy and chemotherapy, and more recently to immunotherapy. In particular, hypoxic tumors exclude T cells and inhibit their activity, suggesting that tumor cells acquire a mechanism to evade T-cell recognition and killing. Our analysis of hypoxic tumors indicates that hypoxia downregulates the expression of MHC class I and its bound peptides (i.e., the immunopeptidome). Hypoxia decreases MHC-I expression in an oxygen-dependent manner, via activation of autophagy through the PERK arm of the unfolded protein response. Using an immunopeptidomics-based LC-MS approach, we find a significant reduction of presented antigens under hypoxia. Inhibition of autophagy under hypoxia enhances antigen presentation. In experimental tumors, reducing mitochondrial metabolism through a respiratory complex-I inhibitor increases tumor oxygenation, as well as MHC-I levels and the immunopeptidome. These data explain the molecular basis of tumor immune evasion in hypoxic conditions, and have implications for future therapeutic interventions targeting hypoxia-induced alterations in antigen presentation.
Collapse
Affiliation(s)
- Hala Estephan
- Department of Oncology, The University of Oxford, Oxford, OX3 7DQ, UK
| | - Arun Tailor
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, OX37BN, UK
| | - Robert Parker
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, OX37BN, UK
| | - McKenzie Kreamer
- Department of Radiation Oncology, OSU Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Ohio State University, Columbus, OH, USA
| | - Ioanna Papandreou
- Department of Radiation Oncology, OSU Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Ohio State University, Columbus, OH, USA
| | - Leticia Campo
- Department of Oncology, The University of Oxford, Oxford, OX3 7DQ, UK
| | - Alistair Easton
- Department of Oncology, The University of Oxford, Oxford, OX3 7DQ, UK
| | - Eui Jung Moon
- Department of Oncology, The University of Oxford, Oxford, OX3 7DQ, UK
| | - Nicholas C Denko
- Department of Radiation Oncology, OSU Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Ohio State University, Columbus, OH, USA
| | - Nicola Ternette
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, OX37BN, UK
| | - Ester M Hammond
- Department of Oncology, The University of Oxford, Oxford, OX3 7DQ, UK
| | - Amato J Giaccia
- Department of Oncology, The University of Oxford, Oxford, OX3 7DQ, UK.
| |
Collapse
|
49
|
Melzer YF, Fergen NL, Mess C, Stadler JC, Geidel G, Schwietzer YA, Kött J, Pantel K, Schneider SW, Utikal J, Wladykowski E, Vidal-Y-Sy S, Bauer AT, Gebhardt C. Evaluation of S100A8/A9 and neutrophils as prognostic markers in metastatic melanoma patients under immune-checkpoint inhibition. Transl Oncol 2025; 52:102224. [PMID: 39700646 PMCID: PMC11718343 DOI: 10.1016/j.tranon.2024.102224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 12/21/2024] Open
Abstract
Immune-checkpoint inhibitors (ICIs) have revolutionized melanoma treatment, yet approximately half of patients do not respond to these therapies. Identifying prognostic biomarkers is crucial for treatment decisions. Our retrospective study assessed liquid biopsies and tumor tissue analyses for two potential biomarkers: danger-associated molecular pattern (DAMP) S100A8/A9 and its source, neutrophils. In 43 metastatic unresected stage III/IV melanoma patients, elevated serum levels of S100A8/A9 and neutrophils before and during ICI treatment correlated with worse outcomes. Furthermore, in 113 melanoma patients, neutrophil expression in the tumor microenvironment (TME) was associated with relapse and reduced survival. Measuring S100A8/A9 and neutrophils could enhance immunotherapy monitoring by predicting impaired clinical outcomes and non-response to ICIs. Serum S100A8/A9 levels and neutrophil counts at baseline (T0) and during treatment (T3) correlated with reduced progression-free survival (PFS). Elevated S100A8/A9 levels at T0 and T3 negatively impacted overall survival (OS). Notably, neutrophil infiltration was more prevalent in primary melanomas than in nevi and metastases, and its presence in primary melanomas was linked to poorer survival. S100A8/A9 serum levels, neutrophil counts, and tumor-associated neutrophil infiltration represent promising biomarkers for predicting treatment response and clinical outcomes in melanoma patients receiving ICIs. SIGNIFICANCE: These findings underscore the critical need for reliable biomarkers in melanoma research, particularly for predicting responses to immune-checkpoint inhibitors (ICIs). Identifying S100A8/A9 levels and neutrophil infiltration as potential indicators of treatment outcomes offers valuable insights for personalized therapy decisions. By enhancing monitoring and prognosis assessment, these biomarkers contribute to refining treatment strategies, ultimately improving patient care and outcomes. This research bridges gaps in understanding melanoma response mechanisms and highlights avenues for further investigation into immune-related markers, fostering advancements in precision medicine for melanoma patients.
Collapse
Affiliation(s)
- Yasmin F Melzer
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Nadine L Fergen
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Christian Mess
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Julia-Christina Stadler
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Glenn Geidel
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Ysabel A Schwietzer
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Julian Kött
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Klaus Pantel
- Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Department of Tumor Biology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Stefan W Schneider
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.
| | - Ewa Wladykowski
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Sabine Vidal-Y-Sy
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Alexander T Bauer
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.
| | - Christoffer Gebhardt
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| |
Collapse
|
50
|
Hoang Nguyen KH, Le NV, Nguyen PH, Nguyen HHT, Hoang DM, Huynh CD. Human immune system: Exploring diversity across individuals and populations. Heliyon 2025; 11:e41836. [PMID: 39911431 PMCID: PMC11795082 DOI: 10.1016/j.heliyon.2025.e41836] [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: 07/21/2024] [Revised: 12/23/2024] [Accepted: 01/08/2025] [Indexed: 02/07/2025] Open
Abstract
The immune response is an intricate system that involves the complex connection of cellular and molecular components, each with distinct functional specialisations. It has a distinct capacity to adjust and mould the immune response in accordance with specific stimuli, influenced by both genetic and environmental factors. The presence of genetic diversity, particularly across different ethnic and racial groups, significantly contributes to the impact of incidence of diseases, disease susceptibility, autoimmune disorders, and cancer risks in specific regions and certain populations. Environmental factors, including geography and socioeconomic status, further modulate the variety of the immune system responses. These, in turn, affect the susceptibility to infectious diseases and development of autoimmune disorders. Despite the complexity of the relationship, there remains a gap in understanding the specificity of immune indices across races, immune reference ranges among populations, highlighting the need for deeper understanding of immune diversity for personalized approaches in diagnostics and therapeutics. This review systematically organizes these findings, with the goal of emphasizing the potential of targeted interventions to address health disparities and advance translational research, enabling a more comprehensive strategy. This approach promises significant advancements in identifying specific immunological conditions, focusing on personalized interventions, through both genetic and environmental factors.
Collapse
Affiliation(s)
| | - Nghi Vinh Le
- College of Health Sciences, VinUniversity, Hanoi, Viet Nam
| | | | - Hien Hau Thi Nguyen
- College of Health Sciences, VinUniversity, Hanoi, Viet Nam
- Institute of Research and Development, Duy Tan University, Da Nang, Viet Nam
- School of Medicine and Pharmacy, Duy Tan University, Da Nang, Viet Nam
| | - Duy Mai Hoang
- College of Health Sciences, VinUniversity, Hanoi, Viet Nam
| | | |
Collapse
|