1
|
Liu J, Fan J, Duan H, Chen G, Zhang W, Wang P. Identification and validation of susceptibility modules and hub genes in polyarticular juvenile idiopathic arthritis using WGCNA and machine learning. Autoimmunity 2025; 58:2437239. [PMID: 39699225 DOI: 10.1080/08916934.2024.2437239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Juvenile idiopathic arthritis (JIA), superseding juvenile rheumatoid arthritis (JRA), is a chronic autoimmune disease affecting children and characterized by various types of childhood arthritis. JIA manifests clinically with joint inflammation, swelling, pain, and limited mobility, potentially leading to long-term joint damage if untreated. This study aimed to identify genes associated with the progression and prognosis of JIA polyarticular to enhance clinical diagnosis and treatment. METHODS We analyzed the gene expression omnibus (GEO) dataset GSE1402 to screen for differentially expressed genes (DEGs) in peripheral blood single nucleated cells (PBMCs) of JIA polyarticular patients. Weighted gene co-expression network analysis (WGCNA) was applied to identify key gene modules, and protein-protein interaction networks (PPIs) were constructed to select hub genes. The random forest model was employed for biomarker gene screening. Functional enrichment analysis was conducted using David's online database, gene ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis to annotate and identify potential JIA pathways. Hub genes were validated using the receiver operating characteristic (ROC) curve. RESULTS PHLDA1, EGR3, CXCL2, and PF4V1 were identified as significantly associated with the progression and prognosis of JIA polyarticular phenotype, demonstrating high diagnostic and prognostic assessment value. CONCLUSION These genes can be utilized as potential molecular biomarkers, offering valuable insights for the early diagnosis and personalized treatment of JIA polyarticular patients.
Collapse
Affiliation(s)
- Junfeng Liu
- Department of Orthopedics, Dazhou Central Hospital, Dazhou, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianhui Fan
- North Sichuan Medical College, Nanchong, China
| | - Hongxiang Duan
- College of Public Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Guoming Chen
- Department of Orthopedics, Dazhou Central Hospital, Dazhou, China
| | - Weihua Zhang
- Division of Rheumatology, Dazhou Central Hospital, Dazhou, China
| | - Pingxi Wang
- Department of Orthopedics, Dazhou Central Hospital, Dazhou, China
| |
Collapse
|
2
|
Dong H, Wang X, Zheng Y, Li J, Liu Z, Wang A, Shen Y, Wu D, Cui H. Mapping the rapid growth of multi-omics in tumor immunotherapy: Bibliometric evidence of technology convergence and paradigm shifts. Hum Vaccin Immunother 2025; 21:2493539. [PMID: 40275437 PMCID: PMC12026087 DOI: 10.1080/21645515.2025.2493539] [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: 11/08/2024] [Revised: 04/01/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
This study aims to fill the knowledge gap in systematically mapping the evolution of omics-driven tumor immunotherapy research through a bibliometric lens. While omics technologies (genomics, transcriptomics, proteomics, metabolomics)provide multidimensional molecular profiling, their synergistic potential with immunotherapy remains underexplored in large-scale trend analyses. A comprehensive search was conducted using the Web of Science Core Collection for literature related to omics in tumor immunotherapy, up to August 2024. Bibliometric analyses, conducted using R version 4.3.3, VOSviewer 1.6.20, and Citespace 6.2, examined publication trends, country and institutional contributions, journal distributions, keyword co-occurrence, and citation bursts. This analysis of 9,494 publications demonstrates rapid growth in omics-driven tumor immunotherapy research since 2019, with China leading in output (63% of articles) yet exhibiting limited multinational collaboration (7.9% vs. the UK's 61.8%). Keyword co-occurrence and citation burst analyses reveal evolving frontiers: early emphasis on "PD-1/CTLA-4 blockade" has transitioned toward "machine learning," "multi-omics," and "lncRNA," reflecting a shift to predictive modeling and biomarker discovery. Multi-omics integration has facilitated the development of immune infiltration-based prognostic models, such as TIME subtypes, which have been validated across multiple tumor types, which inform clinical trial design (e.g. NCT06833723). Additionally, proteomic analysis of melanoma patients suggests that metabolic biomarkers, particularly oxidative phosphorylation and lipid metabolism, may stratify responders to PD-1 blockade therapy. Moreover, spatial omics has confirmed ENPP1 as a potential novel therapeutic target in Ewing sarcoma. Citation trends underscore clinical translation, particularly mutation-guided therapies. Omics technologies are transforming tumor immunotherapy by enhancing biomarker discovery and improving therapeutic predictions. Future advancements will necessitate longitudinal omics monitoring, AI-driven multi-omics integration, and international collaboration to accelerate clinical translation. This study presents a systematic framework for exploring emerging research frontiers and offers insights for optimizing precision-driven immunotherapy.
Collapse
Affiliation(s)
- Huijing Dong
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Xinmeng Wang
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Yumin Zheng
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Jia Li
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Zhening Liu
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Aolin Wang
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Yulei Shen
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Daixi Wu
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Huijuan Cui
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
3
|
Imtiaz S, Ferdous UT, Nizela A, Hasan A, Shakoor A, Zia AW, Uddin S. Mechanistic study of cancer drug delivery: Current techniques, limitations, and future prospects. Eur J Med Chem 2025; 290:117535. [PMID: 40132495 DOI: 10.1016/j.ejmech.2025.117535] [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: 02/26/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
Cancer drug delivery remains a critical challenge with systemic toxicity, poor drug bioavailability, and a lack of effective targeting. Overcoming these barriers is essential for improving treatment efficacy and patient outcomes. This review discusses current drug delivery techniques that reshape cancer therapy by offering precise, controlled-release tailored to tumor-specific features. Innovations in nanotechnology, immunotherapy, and gene therapy enable interventions at molecular and cellular levels. Radiomics and pathomics integrate high-dimensional data to optimize diagnostics and treatment planning. Combination therapy addresses the complexities of tumor heterogeneity by synergizing multiple agents within a single therapeutic framework, while peptide-drug conjugates enhance specificity and potency. Hydrogel-based systems and microneedle arrays offer localized, sustained release, significantly improving therapeutic outcomes. However, clinical translation of these advancements faces significant barriers such as drug resistance, off-target effects, scalability, cost, and ethical concerns. Moreover, regulatory complexities and the economic feasibility of these therapies highlight the need for innovative frameworks to make them accessible globally. Therefore, there is a need for innovation in gene and cell therapy, next-generation drug delivery platforms, and personalized medicine. This review focuses on recent advancements in drug delivery techniques over the past decade, evaluating their limitations and exploring potential future directions for transforming cancer treatment.
Collapse
Affiliation(s)
- Saiqa Imtiaz
- Department of Bioengineering, King Fahd University of Petroleum & Minerals, Dhahran, 31261, Saudi Arabia
| | - Umme Tamanna Ferdous
- Center for Biosystems and Machines, King Fahd University of Petroleum & Minerals, Dhahran, 31261, Saudi Arabia
| | - Alexis Nizela
- Department of Bioengineering, King Fahd University of Petroleum & Minerals, Dhahran, 31261, Saudi Arabia; Center for Membranes and Water Security, King Fahd University of Petroleum and Minerals, Dhahran, 31261, Saudi Arabia
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha, 2713, Qatar; Biomedical Research Center, Qatar University, Doha, 2713, Qatar
| | - Adnan Shakoor
- Center for Biosystems and Machines, King Fahd University of Petroleum & Minerals, Dhahran, 31261, Saudi Arabia; Department of Control & Instrumentation Engineering, King Fahd University of Petroleum & Minerals, Dhahran, 31261, Saudi Arabia
| | - Abdul Wasy Zia
- Institute of Mechanical, Process, and Energy Engineering (IMPEE), School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS, United Kingdom.
| | - Shihab Uddin
- Department of Bioengineering, King Fahd University of Petroleum & Minerals, Dhahran, 31261, Saudi Arabia; Center for Biosystems and Machines, King Fahd University of Petroleum & Minerals, Dhahran, 31261, Saudi Arabia.
| |
Collapse
|
4
|
Sun L, Liu Y, Sun Q, Wang G, Du B, Liu B, Gao T, Zhao P, Yang Y, Rong R. Polysaccharides from traditional Chinese medicine and their nano-formulated delivery systems for cancer immunotherapy. Carbohydr Polym 2025; 357:123416. [PMID: 40158963 DOI: 10.1016/j.carbpol.2025.123416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 04/02/2025]
Abstract
Cancer immunotherapy has evolved into a new generation strategy in the field of anti-tumor treatment. Polysaccharides derived from Traditional Chinese Medicine (TCM) are gaining recognition as powerful immunomodulators in cancer therapy, noted for their multi-target and multi-pathway actions. Owing to their beneficial properties such as water solubility, biocompatibility, and chemical structure modifiability, TCM polysaccharides can also serve as carriers for hydrophobic drugs in the development of innovative drug delivery systems, enhancing synergistic antitumor effects. In this article, we summarize the diverse mechanisms of immunoregulation by TCM polysaccharides in tumor therapy. The applications of these polysaccharides as both active ingredients and drug carriers within nanodelivery systems for cancer immunotherapy are also introduced. Additionally, extensive research on TCM polysaccharides in clinical settings has been collected. Furthermore, discussions are presented on the development prospects and challenges faced by these polysaccharides in the field of tumor immunotherapy. Our goal is to improve researchers' comprehension of TCM polysaccharides in cancer immunotherapy, providing promising strategies to optimize cancer treatment and benefit diverse patient populations.
Collapse
Affiliation(s)
- Linlin Sun
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yuting Liu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Qihui Sun
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Guimei Wang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Baoxiang Du
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Bodong Liu
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Tian Gao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Pan Zhao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yong Yang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China; Collaborative Innovation Center for Antiviral Traditional Chinese Medicine in Shandong Province, Jinan 250355, PR China; Shandong Antiviral Engineering Research Center of Traditional Chinese Medicine, Jinan 250355, PR China.
| | - Rong Rong
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| |
Collapse
|
5
|
Xie Y, Guo J, Hu J, Li Y, Zhang Z, Zhu Y, Deng F, Qi J, Zhou Y, Chen W. A factorial design-optimized microfluidic LNP vaccine elicits potent magnesium-adjuvating cancer immunotherapy. Mater Today Bio 2025; 32:101703. [PMID: 40230646 PMCID: PMC11994397 DOI: 10.1016/j.mtbio.2025.101703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/19/2025] [Accepted: 03/23/2025] [Indexed: 04/16/2025] Open
Abstract
Human papillomavirus (HPV)-associated cancers remain a critical health challenge, prompting the development of effective therapeutic vaccines. This study presents a lipid nanoparticle (LNP)-based vaccine co-loading E7 antigen peptide and magnesium ions as the adjuvant. Microfluidic technology was employed to optimize LNP preparation and formulation, ensuring efficient co-delivery of antigen and adjuvant. Magnesium ions were chosen over conventional aluminum-based adjuvants, which often suffer from limited efficacy and adverse effects, particularly for cancer immunotherapy. Compared to aluminum, magnesium ions exhibited superior capabilities in enhancing T-cell activation and promoting cellular immune response. Mechanistic insights suggest that magnesium ions facilitate dendritic cell maturation and antigen presentation via a collagen-CD36 axis, contributing to the adjuvant activity of magnesium. Through design of experiments (DoE) optimization, the LNP formulation was tailored for enhanced encapsulation and stability, positioning it as a targeted system for immune activation. These findings support the promise of magnesium ions as effective and safer adjuvants in LNP-based vaccines, marking a potential advancement for therapeutic cancer vaccination.
Collapse
Affiliation(s)
- Yongyi Xie
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Jiaxin Guo
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Jialin Hu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yuan Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Zhongqian Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yongcheng Zhu
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Fei Deng
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, UNSW Sydney, NSW, 2052, Australia
| | - Jialong Qi
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, PR China
| | - You Zhou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Wenjie Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| |
Collapse
|
6
|
Zhang Y, Chen Y, Guo C, Li S, Huang C. Systemic immune-inflammation index as a predictor of survival in non-small cell lung cancer patients undergoing immune checkpoint inhibition: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2025; 210:104669. [PMID: 39978427 DOI: 10.1016/j.critrevonc.2025.104669] [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: 10/31/2024] [Revised: 02/06/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND This meta-analysis aims to evaluate the association between pretreatment systemic immune-inflammation index (SII) levels and progression-free survival (PFS) and overall survival (OS) in NSCLC patients receiving immune checkpoint inhibitors (ICIs). METHODS A systematic search was conducted across PubMed, Embase, and Web of Science. Hazard ratios (HRs) with 95 % confidence intervals (CIs) for PFS and OS were extracted or calculated. Random-effects models were employed to pool the results and subgroup analyses were performed based on study characteristics, treatment regimens, and analytical methods. RESULTS Two prospective and 11 retrospective studies involving 2342 NSCLC patients treated with ICIs were included. A high pretreatment SII was significantly associated with poor PFS (HR: 2.05, 95 % CI: 1.59-2.64, p < 0.001; I2 = 42 %) and poor OS (HR: 1.54, 95 % CI: 1.29-1.82, p < 0.001; I2 = 22 %). Subgroup analyses according to the country of the study, lines of treatment, cancer stage, methods for determining the cutoffs of SII, and the analytic models showed consistent results (p for subgroup difference all > 0.05). Interestingly, the subgroup analyses indicated a stronger association in patients receiving ICIs alone versus those receiving concurrent chemotherapy (p for subgroup difference = 0.04). CONCLUSIONS High pretreatment SII is associated with worse PFS and OS in NSCLC patients treated with ICIs, particularly for the patients receiving ICIs alone without concurrent chemotherapy.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yeye Chen
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Chao Guo
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Cheng Huang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
7
|
Wu C, Gao M, Xiao W, Huang X, Yang X, Wu Z, Yu X, Mo B, Du Z, Shang Z, Liu J, Shi C, Li R, Luo S, Wang W. Light-activatable manganese carbonate nanocubes elicit robust immunotherapy by amplifying endoplasmic reticulum stress-meditated pyroptotic cell death. J Exp Clin Cancer Res 2025; 44:147. [PMID: 40380194 DOI: 10.1186/s13046-025-03408-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025] Open
Abstract
Although tumor immunotherapy has emerged as a promising treatment modality, it faces significant challenges stemming from the immunosuppressive characteristics of the tumor microenvironment (TME), the low immunogenicity of tumors, and the poor specificity of immunoactivation. These factors can hinder the efficacy of immunotherapeutic approaches and lead to immune-related adverse events. This study reports a multifunctional nanocube (Mn-ER-Cy) that integrates Mn carbonate (MnCO3) and a photosensitizer (ER-Cy) by targeting tumor-cell endoplasmic reticulum (ER). The results demonstrate that Mn-ER-Cy preferentially accumulates in tumor tissues and is retained within ER organelles, facilitating photothermal therapy (PTT) and photodynamic therapy (PDT) upon exposure to 808 nm light irradiation. Triggered by acidic TME and light irradiation, MnCO3 is rapidly degraded to Mn2+, which in turn promotes the generation of reactive oxygen species through the Mn2+-mimic Fenton reaction, enabling chemical dynamics therapy (CDT). Triple-modal synergistic therapy simultaneously happens in ER to induce excessive ER stress, which subsequently amplify highly immunogenic pyroptotic cell death through activating NLRP3 inflammasome, caspase-1, and gasdermin D (GSDMD) pathway. Meanwhile, the decomposition of MnCO3 consumes H+ and contributes to an increased intracellular pH by regulating lactic acid levels, thereby counteracting the immunosuppressive acidic TME. Furthermore, Mn-ER-Cy serves as an inherent dual-modality imaging contrast agent for near-infrared fluorescence and photoacoustic imaging, facilitating imaging-guided precision therapy. These findings underscore the potential of Mn-ER-Cy to substantially enhance the efficacy and specificity of tumor immunotherapy, portraying a bright prospect to improve the clinical outcomes of patients with cancer.
Collapse
Affiliation(s)
- Chuan Wu
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Mingquan Gao
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Weidong Xiao
- Department of Pharmacy, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Xie Huang
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xinrui Yang
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zifei Wu
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xudong Yu
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Banghui Mo
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zaizhi Du
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ziqian Shang
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jing Liu
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Can Shi
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Rong Li
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Shenglin Luo
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Weidong Wang
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China.
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, 610041, China.
| |
Collapse
|
8
|
Zhao L, Zeng J, Wen J, Li Z, Xu J, Wang J, Tang X, Hou L. Global trends and research hotspots in autophagy and tumor drug resistance: a bibliometric analysis. Discov Oncol 2025; 16:734. [PMID: 40354002 PMCID: PMC12069191 DOI: 10.1007/s12672-025-02379-5] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
Autophagy plays a crucial role in tumor drug resistance by enabling cancer cells to survive under stress conditions, including chemotherapy. It helps tumor cells maintain homeostasis, resist cell death, and contribute to therapy failure. This study analyzed the literature related to autophagy and tumor drug resistance based on the Web of Science Core Collection (WoSCC) database. The results revealed that there are 9284 relevant articles published to date, covering 103 countries and regions, with contributions from 5964 institutions and 37,240 researchers. The annual number of publications has steadily increased since 2004, especially after 2019, indicating the growing importance of autophagy in tumor drug resistance research. China leads globally in terms of publication output, accounting for nearly 50% of the total publications. Additionally, international collaboration and cross-country research have become increasingly prominent, particularly collaborations between China and countries like South Korea and Japan. Journal analysis showed that the International Journal of Molecular Sciences and Oncotarget are the most productive journals, while Autophagy stands out with a higher impact factor. Author, citation, and keyword analyses revealed research hotspots and future trends in the field of autophagy and tumor drug resistance, including chemotherapy resistance, cell death mechanisms, and immunotherapy. This study provides a systematic academic perspective for future research in the field of autophagy and tumor drug resistance and emphasizes the importance of strengthening international cooperation.
Collapse
Affiliation(s)
- Long Zhao
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, No. 1 South Maoyuan Road, Shunqing District, Nanchong, 637000, Sichuan, People's Republic of China
| | - Jiao Zeng
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Junfeng Wen
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Zhaoyang Li
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Jianguo Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Jinxiang Wang
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, People's Republic of China.
| | - Xiaoping Tang
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, No. 1 South Maoyuan Road, Shunqing District, Nanchong, 637000, Sichuan, People's Republic of China.
| | - Lingmi Hou
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, Renmin South Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
9
|
Yao J, Gan W, Sun J, Han Z, Li D, Cao L, Zhu L. APOL6 as a potential biomarker of immuno-correlation and therapeutic prediction in cancer immunotherapy. Medicine (Baltimore) 2025; 104:e42406. [PMID: 40355224 PMCID: PMC12073870 DOI: 10.1097/md.0000000000042406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 04/22/2025] [Indexed: 05/14/2025] Open
Abstract
The emergence of immune checkpoint inhibitors (ICIs) has significantly revolutionized the approach to treating advanced cancers. Despite their remarkable efficacy, not all patients exhibit favorable responses to ICI therapy. Hence, more biomarkers for therapeutic prediction need to be discovered. In this study, we utilized public cohorts to investigate the predictive significance and immunological associations of apolipoprotein L6 (APOL6) in cancers. The expression of APOL6 was found to be enhanced in tumors of patients who exhibited strong immunotherapeutic responses across various types of cancer. Furthermore, APOL6 showed immune correlations in pan-cancer and was confirmed by the tissue microarray cohort and in vitro experiments. Overall, this study highlights that APOL6 serves as a beneficial biomarker for immune checkpoint inhibitors in patients with cancer. Additional research involving larger numbers of patients and the underlying mechanism is necessary to determine its effectiveness as a biomarker for predicting the benefits of ICIs.
Collapse
Affiliation(s)
- Jialin Yao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenyuan Gan
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiukang Sun
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhihang Han
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dongqing Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Cao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Oncology, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
| | - Lingjun Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Wang X, Wang X, Wang X, Xu K, Long J, Chen Y, Liu Y, Niu H, Zeng B, Fan Y. Discovery of (E)-2-cyano-N-(2,3-dihydrobenzo[b][1,4] dioxin-6-yl)-3-(1H-indol-3-yl)acrylamide as a novel TDO inhibitor for cancer treatment. Bioorg Chem 2025; 161:108555. [PMID: 40359840 DOI: 10.1016/j.bioorg.2025.108555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/19/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025]
Abstract
Tryptophan 2,3- oxygenase (TDO), an immune-suppressing enzyme, catalyzes the catabolism of tryptophan to N-formylkynurenine. Overexpression of TDO in multiple malignancies associates with immune evasion mechanisms. Therefore, we aimed to discover novel inhibitors targeting TDO as a potential strategy for cancer treatment. In this study, we designed and synthesized a series of (E)-2-cyano-3-(1H-indol-3-yl) acrylamide derivatives. A lead compound, 5c, was identified and exhibited inhibitory activity against TDO, with an IC50 value of 1.25 ± 0.04 μM. 5c demonstrated significant tumor growth suppression with a favorable safety profile in Hepa1-6 hepatocellular carcinoma allograft model. Notably, 5c synergized with the PD-1/PD-L1 inhibitor BMS-202, both in vitro and in vivo. These findings suggested compound 5c could serve as a promising candidate for targeting TDO to address tumor immune tolerance in cancer therapy.
Collapse
Affiliation(s)
- Xiang Wang
- Xingzhi College, Zhejiang Normal University, Lanxi, 321004, China
| | - Xiaoshuang Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xiaoxue Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Kejia Xu
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Jie Long
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Yanan Chen
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Yanhua Liu
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Haiying Niu
- Department of Gynecology, Tianjin First Central Hospital, Tianjin, China..
| | - Beilei Zeng
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China..
| | - Yan Fan
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| |
Collapse
|
11
|
Li Y, Sun Y, Yu K, Li Z, Miao H, Xiao W. Keratin: A potential driver of tumor metastasis. Int J Biol Macromol 2025; 307:141752. [PMID: 40049479 DOI: 10.1016/j.ijbiomac.2025.141752] [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: 10/08/2024] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
Keratins, as essential components of intermediate filaments in epithelial cells, play a crucial role in maintaining cell structure and function. In various malignant epithelial tumors, abnormal keratin expression is frequently observed and serves not only as a diagnostic marker but also closely correlates with tumor progression. Extensive research has demonstrated that keratins are pivotal in multiple stages of tumor metastasis, including responding to mechanical forces, evading the immune system, reprogramming metabolism, promoting angiogenesis, and resisting apoptosis. Here we emphasize that keratins significantly enhance the migratory and invasive capabilities of tumor cells, making them critical drivers of tumor metastasis. These findings highlight the importance of targeting keratins as a strategic approach to combat tumor metastasis, thereby advancing our understanding of their role in cancer progression and offering new therapeutic opportunities.
Collapse
Affiliation(s)
- Yuening Li
- Army Medical University, Chongqing, China
| | - Yiming Sun
- Department of General Surgery, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Kun Yu
- Department of General Surgery, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Zhixi Li
- Department of General Surgery, the Second Affiliated Hospital of Army Medical University, Chongqing, China.
| | - Hongming Miao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China; Jinfeng Laboratory, Chongqing, China.
| | - Weidong Xiao
- Department of General Surgery, the Second Affiliated Hospital of Army Medical University, Chongqing, China.
| |
Collapse
|
12
|
Xu P, Zhang Q, Zhai J, Chen P, Deng X, Miao L, Zhang X. APOA1 promotes tumor proliferation and migration and may be a potential pan-cancer biomarker and immunotherapy target. Transl Oncol 2025; 55:102344. [PMID: 40088749 PMCID: PMC11957500 DOI: 10.1016/j.tranon.2025.102344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/26/2025] [Accepted: 03/02/2025] [Indexed: 03/17/2025] Open
Abstract
INTRODUCTION Aberrant expression of APOA1 has been reported in various cancers. However, a comprehensive investigation into its role in cancer is currently lacking. METHODS Online websites and databases such as TIMER2.0, GEPIA2, UALCAN and GSCA were used to investigate the relationship between APOA1 expression and prognostic value, immune infiltration, gene mutations, and drug sensitivity. In addition, in vitro CCK-8 and transwell migration and invasion assays were performed to determine the biological functions of APOA1 in gastric cancer (GC) cells. RESULTS The pan-cancer analysis showed that APOA1 is differentially expressed in different cancer types and significantly correlated with tumor stages. A survival analysis revealed that APOA1 predicted a poor prognosis in ACC, KIRC, STAD, and a good prognosis in BRCA, OV, and UCEC. We also found that the most common genetic alteration type of APOA1 was deep deletion, and the DNA methylation level of APOA1 decreased in various cancers. Furthermore, APOA1 expression negatively correlated with immune cells infiltration in cancers, including CD4+ T, CD8+ T, and myeloid dendritic cells. For STAD, GO/KEGG enrichment analysis revealed the possible involvement of APOA1 in cholesterol metabolism and PPAR signaling pathway. Finally, we further performed in vitro experiments to verify that overexpression of APOA1 could promote the proliferation, migration and invasion of GC cells. CONCLUSION The results of this study indicate that APOA1 is a potential tumor prognostic biomarker and immunotherapy target. In addition, APOA1 plays an essential role in the proliferation, migration, and invasion of GC cells by vitro experiments.
Collapse
Affiliation(s)
- Peiyi Xu
- Department of Gastroenterology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Qiuyan Zhang
- Department of Gastroenterology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jing Zhai
- Department of Gastroenterology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Pu Chen
- Department of Gastroenterology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Xueting Deng
- Department of Gastroenterology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Lin Miao
- Department of Gastroenterology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Xiuhua Zhang
- Department of Gastroenterology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
13
|
Wang T, Bai Y, Dong Y, Qin J, Zhou X, Wang A, Liu D, Li X, Ma Z, Hu Y. A comprehensive analysis of deubiquitinase USP20 on prognosis and immunity in pan-cancer. FASEB J 2025; 39:e70499. [PMID: 40270318 DOI: 10.1096/fj.202402603r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 02/05/2025] [Accepted: 03/20/2025] [Indexed: 04/25/2025]
Abstract
USP20 is a deubiquitinase enzyme in the ubiquitin-proteasome system that plays a role in the development and progression of tumors. However, the relationships between USP20 expression and clinical prognosis and tumor immunity remain unclear. In this study, the USP20 expression and its relationships with potential prognostic value, the tumor microenvironment (TME), immune-related genes, the tumor mutational burden (TMB), microsatellite instability (MSI), homologous recombination deficiency, cancer stemness, and correlated signaling pathways were investigated via The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), Cancer Cell Line Encyclopedia (CCLE), STRING, Gene Expression Profiling Interactive Analysis (GEPIA2), and the Human Protein Atlas (HPA). Moreover, we explored the oncogenic capability of USP20 in breast cancer. Data analysis was performed via GraphPad Prism and the R package. The results indicated that the expression of USP20 was upregulated in most cancers and was associated with survival in 17 tumor types. Furthermore, USP20 expression was strongly correlated with immune infiltration and the expression of immunomodulatory genes. We also verified the correlations between USP20 expression and tumor heterogeneity, cancer stemness, and the corresponding signaling pathways. Moreover, our work revealed that USP20 was highly expressed and predicted a poor outcome in patients with breast cancer. Basic experiments verified that USP20 overexpression promoted both the proliferation and migration of breast cancer cells. This study comprehensively investigated the expression of USP20 and its correlation with clinical prognostic assessment and tumor immune modulation across cancers, indicating that USP20 might have utility as a biomarker associated with prognosis and cancer immunotherapy.
Collapse
Affiliation(s)
- Ting Wang
- School of Medicine, Nankai University, Tianjin, China
- Department of Oncology, The First Medical Center of PLA General Hospital, Beijing, China
| | - Yibing Bai
- Department of Oncology, The First Medical Center of PLA General Hospital, Beijing, China
- Graduate School, Medical School of Chinese PLA, Beijing, China
| | - Yi Dong
- Department of Oncology, The First Medical Center of PLA General Hospital, Beijing, China
- Graduate School, Medical School of Chinese PLA, Beijing, China
| | - Jiapei Qin
- Department of Oncology, The First Medical Center of PLA General Hospital, Beijing, China
- Graduate School, Medical School of Chinese PLA, Beijing, China
| | - Xin Zhou
- Department of Oncology, The First Medical Center of PLA General Hospital, Beijing, China
- Graduate School, Medical School of Chinese PLA, Beijing, China
| | - An Wang
- Department of Oncology, The First Medical Center of PLA General Hospital, Beijing, China
- Graduate School, Medical School of Chinese PLA, Beijing, China
| | - Dong Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiaoyan Li
- Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Zhiqiang Ma
- Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Yi Hu
- School of Medicine, Nankai University, Tianjin, China
- Department of Oncology, The First Medical Center of PLA General Hospital, Beijing, China
- Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
14
|
Tao E, Song Q, Tang J, Xin W, Xiao Z, Liu Z, Xie G. Prognostic biomarker RIMS1 and its association with immune infiltration in glioblastoma. Sci Rep 2025; 15:15240. [PMID: 40307326 PMCID: PMC12043866 DOI: 10.1038/s41598-025-99499-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 04/21/2025] [Indexed: 05/02/2025] Open
Abstract
Glioblastoma (GBM) is the most common and deadly malignant tumor of the nervous system. RIMS1, a member of the RAS gene superfamily, plays a critical role in signaling pathways regulating cell growth and differentiation. However, the prognostic value of RIMS1, particularly its relationship with immune cell infiltration in gliomas, has not been fully explored. RIMS1 expression in glioblastoma cells and tissues was assessed using bioinformatics platforms. The association between RIMS1 expression levels and overall survival was analyzed using Kaplan-Meier analysis and Cox regression model. To evaluate the proliferative and migratory capacity of GBM cells, we conducted CCK-8, colony formation, transwell, and scratch assays. With data from The Cancer Genome Atlas (TCGA), we investigated the correlation between RIMS1 expression and immune cell infiltration levels and assessed the prognostic impact of RIMS1 on GBM patient survival, focusing on its potential involvement in immune pathways. Lower RIMS1 expression was associated with poorer overall survival and was linked to patient age, gender, and tumor grade. Importantly, RIMS1 expression showed a significant correlation with immune cell infiltration levels, suggesting that RIMS1 influences glioblastoma survival, at least in part, through immune-related mechanisms. In glioblastoma patients, elevated RIMS1 expression may serve as an independent prognostic biomarker, potentially through its impact on immune cell infiltration.
Collapse
Affiliation(s)
- Erxing Tao
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qimin Song
- Queen Mary School, Nanchang University, Nanchang, 330006, China
| | - Jialin Tang
- Department of Transplantation, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Wenqiang Xin
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhipeng Xiao
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhixin Liu
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Guangbin Xie
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
15
|
Sonar PV, Singh AK, Mandadi S, Sharma NK. Expanding horizons of cancer immunotherapy: hopes and hurdles. Front Oncol 2025; 15:1511560. [PMID: 40352591 PMCID: PMC12061710 DOI: 10.3389/fonc.2025.1511560] [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: 10/15/2024] [Accepted: 03/31/2025] [Indexed: 05/14/2025] Open
Abstract
Background Tumor displays various forms of tumor heterogeneity including immune heterogeneity that allow cancer cells to survive during conventional anticancer drug interventions. Thus, there is a strong rationale for overcoming anticancer drug resistance by employing the components of immune cells. Using the immune system to target tumor cells has revolutionized treatment. Recently, significant progress has been achieved at preclinical and clinical levels to benefit cancer patients. Approach A review of literature from the past ten years across PubMed, Scopus, and Web of Science focused on immunotherapy strategies. These include immune checkpoint inhibitors (ICIs), tumor-infiltrating lymphocyte therapy, antibody-drug conjugates (ADCs), cancer vaccines, CAR T-cell therapy, and the role of the gut microbiome. Conclusion While immunotherapy outcomes have improved, particularly for tumor types such as melanoma and non-small cell lung cancer (NSCLC), challenges persist regarding predictive biomarker identification and better management. Ongoing research on modifiers of immune function like gut microbiome-derived metabolites, next-generation ADCs, and new classes of biologics is warranted. Overall, continued investigation toward optimizing synergistic immunotherapeutic combinations through strategic drug delivery systems is imperative for preclinical and clinical success in cancer patients.
Collapse
Affiliation(s)
- Priyanka Vijay Sonar
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Anuj Kumar Singh
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
- Ichnos Glenmark Innovation, Glenmark Pharmaceuticals Limited, Navi Mumbai, Maharashtra, India
| | - Sravan Mandadi
- Ichnos Glenmark Innovation, Glenmark Pharmaceuticals Limited, Navi Mumbai, Maharashtra, India
| | - Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| |
Collapse
|
16
|
Wang LJ, Lei CL, Wang TA, Lin ZF, Feng SJ, Wei T, Li YQ, Shen MR, Li Y, Liao LF. Prognostic value of the preoperative systemic immune-inflammation nutritional index in patients with gastric cancer. World J Clin Oncol 2025; 16:102294. [PMID: 40290682 PMCID: PMC12019271 DOI: 10.5306/wjco.v16.i4.102294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/04/2024] [Accepted: 01/21/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is the fifth most common cancer and the third leading cause of cancer-related deaths in China. Many patients with GC frequently experience symptoms related to the disease, including anorexia, nausea, vomiting, and other discomforts, and often suffer from malnutrition, which in turn negatively affects perioperative safety, prognosis, and the effectiveness of adjuvant therapeutic measures. Consequently, some nutritional indicators such as nutritional risk index (NRI), prognostic nutritional index (PNI), and systemic immune-inflammatory-nutritional index (SIINI) can be used as predictors of the prognosis of GC patients. AIM To examine the prognostic significance of PNI, NRI, and SIINI in postoperative patients with GC. METHODS A retrospective analysis was conducted on the clinical data of patients with GC who underwent surgical treatment at the Guangxi Medical University Cancer Hospital between January 2010 and December 2018. The area under the receiver operating characteristic (ROC) curve was assessed using ROC curve analysis, and the optimal cutoff values for NRI, PNI, and SIINI were identified using the You-Review-HTMLden index. Survival analysis was performed using the Kaplan-Meier method. In addition, univariate and multivariate analyses were conducted using the Cox proportional hazards regression model. RESULTS This study included a total of 803 patients. ROC curves were used to evaluate the prognostic ability of NRI, PNI, and SIINI. The results revealed that SIINI had superior predictive accuracy. Survival analysis indicated that patients with GC in the low SIINI group had a significantly better survival rate than those in the high SIINI group (P < 0.05). Univariate analysis identified NRI [hazard ratio (HR) = 0.68, 95% confidence interval (CI): 0.52-0.89, P = 0.05], PNI (HR = 0.60, 95%CI: 0.46-0.79, P < 0.001), and SIINI (HR = 2.10, 95%CI: 1.64-2.69, P < 0.001) as prognostic risk factors for patients with GC. However, multifactorial analysis indicated that SIINI was an independent risk factor for the prognosis of patients with GC (HR = 1.65, 95%CI: 1.26-2.16, P < 0.001). CONCLUSION Analysis of clinical retrospective data revealed that SIINI is a valuable indicator for predicting the prognosis of patients with GC. Compared with NRI and PNI, SIINI may offer greater application for prognostic assessment.
Collapse
Affiliation(s)
- Li-Jing Wang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Cai-Lu Lei
- School of Pharmaceutical Science, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Ting-An Wang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Zhi-Feng Lin
- School of Pharmaceutical Science, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Shi-Jie Feng
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Tao Wei
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yan-Qin Li
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Meng-Ru Shen
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yan Li
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Liu-Feng Liao
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
17
|
Srinivasan G, Le MK, Azher Z, Liu X, Vaickus L, Kaur H, Kolling F, Palisoul S, Perreard L, Lau KS, Yao K, Levy J. Histology-Based Virtual RNA Inference Identifies Pathways Associated with Metastasis Risk in Colorectal Cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.22.25326170. [PMID: 40313260 PMCID: PMC12045403 DOI: 10.1101/2025.04.22.25326170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Colorectal cancer (CRC) remains a major health concern, with over 150,000 new diagnoses and more than 50,000 deaths annually in the United States, underscoring an urgent need for improved screening, prognostication, disease management, and therapeutic approaches. The tumor microenvironment (TME)-comprising cancerous and immune cells interacting within the tumor's spatial architecture-plays a critical role in disease progression and treatment outcomes, reinforcing its importance as a prognostic marker for metastasis and recurrence risk. However, traditional methods for TME characterization, such as bulk transcriptomics and multiplex protein assays, lack sufficient spatial resolution. Although spatial transcriptomics (ST) allows for the high-resolution mapping of whole transcriptomes at near-cellular resolution, current ST technologies (e.g., Visium, Xenium) are limited by high costs, low throughput, and issues with reproducibility, preventing their widespread application in large-scale molecular epidemiology studies. In this study, we refined and implemented Virtual RNA Inference (VRI) to derive ST-level molecular information directly from hematoxylin and eosin (H&E)-stained tissue images. Our VRI models were trained on the largest matched CRC ST dataset to date, comprising 45 patients and more than 300,000 Visium spots from primary tumors. Using state-of-the-art architectures (UNI, ResNet-50, ViT, and VMamba), we achieved a median Spearman's correlation coefficient of 0.546 between predicted and measured spot-level expression. As validation, VRI-derived gene signatures linked to specific tissue regions (tumor, interface, submucosa, stroma, serosa, muscularis, inflammation) showed strong concordance with signatures generated via direct ST, and VRI performed accurately in estimating cell-type proportions spatially from H&E slides. In an expanded CRC cohort controlling for tumor invasiveness and clinical factors, we further identified VRI-derived gene signatures significantly associated with key prognostic outcomes, including metastasis status. Although certain tumor-related pathways are not fully captured by histology alone, our findings highlight the ability of VRI to infer a wide range of "histology-associated" biological pathways at near-cellular resolution without requiring ST profiling. Future efforts will extend this framework to expand TME phenotyping from standard H&E tissue images, with the potential to accelerate translational CRC research at scale.
Collapse
Affiliation(s)
- Gokul Srinivasan
- Departments of Pathology and Laboratory Medicine and Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Minh-Khang Le
- Departments of Pathology and Laboratory Medicine and Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Zarif Azher
- Departments of Pathology and Laboratory Medicine and Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- California Institute of Technology, Pasadena, CA, 91125
| | - Xiaoying Liu
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center and Geisel School of Medicine at Dartmouth, Lebanon, NH 03766
| | - Louis Vaickus
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center and Geisel School of Medicine at Dartmouth, Lebanon, NH 03766
| | - Harsimran Kaur
- Center for Computational Systems Biology, Department of Cell and Developmental Biology, Chemical and Physical Biology Program, Vanderbilt University School of Medicine, Nashville TN 37232
| | | | - Scott Palisoul
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center and Geisel School of Medicine at Dartmouth, Lebanon, NH 03766
| | | | - Ken S. Lau
- Center for Computational Systems Biology, Department of Cell and Developmental Biology, Chemical and Physical Biology Program, Vanderbilt University School of Medicine, Nashville TN 37232
| | - Keluo Yao
- Departments of Pathology and Laboratory Medicine and Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Joshua Levy
- Departments of Pathology and Laboratory Medicine and Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center and Geisel School of Medicine at Dartmouth, Lebanon, NH 03766
| |
Collapse
|
18
|
Luan X, Peng X, Hou Q, Liu J. LINC00892 as a Prognostic Biomarker in Lung Adenocarcinoma: Role in Immune Infiltration and EMT Suppression. J Immunol Res 2025; 2025:4341348. [PMID: 40308809 PMCID: PMC12041620 DOI: 10.1155/jimr/4341348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 03/10/2025] [Indexed: 05/02/2025] Open
Abstract
Lung adenocarcinoma (LUAD) is a prevalent and aggressive form of lung cancer with poor prognosis, largely due to late-stage diagnosis and limited therapeutic options. Recent studies suggest that long noncoding RNAs (lncRNAs) play critical roles in cancer progression and immune modulation, emerging as potential therapeutic targets. In this study, we investigated the expression and functional role of LINC00892 in LUAD using RNA sequencing data from The Cancer Genome Atlas (TCGA) and functional assays in vitro and in vivo. We found that LINC00892 is significantly downregulated in LUAD tissues compared to normal tissues, and lower LINC00892 expression correlates with poorer overall survival (OS), disease-specific survival (DSS), and progression-free interval (PFI), particularly in younger patients and those with early-stage disease. Bioinformatic analyses revealed that LINC00892 expression is positively correlated with immune cell infiltration, including CD4+ and CD8+ T cells, and negatively correlated with tumor-promoting Th2 cells, suggesting its role in shaping the tumor immune microenvironment. In vitro functional assays showed that LINC00892 overexpression inhibits LUAD cell proliferation, migration, and invasion while promoting apoptosis. Mechanistically, LINC00892 upregulation was found to suppress epithelial-mesenchymal transition (EMT) by increasing E-cadherin expression and decreasing levels of N-cadherin, vimentin, and slug. Additionally, in an in vivo mouse xenograft model, LINC00892 overexpression suppressed tumor growth and metastasis, accompanied by enhanced immune cell infiltration such as CD4+ and CD8+ T cells. Collectively, these findings suggest that LINC00892 acts as a tumor suppressor in LUAD by modulating immune infiltration and EMT, highlighting its potential as a prognostic biomarker and therapeutic target.
Collapse
Affiliation(s)
- Xinyu Luan
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Xuxing Peng
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Qinghua Hou
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Jixian Liu
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
19
|
Liu Z, Zhou X, Kuang L, Chen Q, Zhao J, Yin H, Zhou Z, Liu X, Liu D, Wu S, Wu L. Novel insights into immune-gut microbiota interactions in colorectal cancer: a Mendelian randomization study. Infect Agent Cancer 2025; 20:27. [PMID: 40251662 PMCID: PMC12008918 DOI: 10.1186/s13027-025-00653-3] [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: 12/27/2024] [Accepted: 03/25/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND The relationship between immune cells and colorectal cancer (CRC) development has been extensively studied; however, the mediating role of gut microbiota in this relationship remains poorly understood. METHODS We utilized summary data from genome-wide association studies (GWAS) to analyze 731 immune cell phenotypes, 473 gut microbiota, and CRC-related data. A two-step mediation analysis was employed to identify mediating gut microbiota. The primary analysis method was inverse variance weighting (IVW), supplemented by MR-Egger, simple mode, weighted median, and weighted mode analyses. Robustness of the results was ensured through systematic sensitivity analyses. RESULTS Our analysis identified 13 immune cell phenotypes significantly associated with CRC, including 10 protective factors and 3 risk factors. Additionally, 13 gut microbiota showed significant associations with CRC, comprising 8 protective factors and 5 risk factors. Mediation analysis revealed that 4-gut microbiota (1 order, 1 family, 1 genus, and 1 unclassified) mediated the relationship between immune cells and CRC. For instance, unclassified CAG - 977 mediated the effects of FSC-A on NK and NKT %lymphocyte on CRC risk, with mediation proportions of 11% and 12.3%, respectively. Notably, 22.3% of the protective effect of EM CD8br %CD8br on CRC was mediated through order Francisellales. CONCLUSION This study provides evidence for a potential causal relationship between immune cells, gut microbiota, and CRC, highlighting the mediating role of specific gut microbiota. These findings offer new insights into the pathogenesis of CRC and may inform future therapeutic strategies.
Collapse
Affiliation(s)
- Zenghui Liu
- Department of Clinical Laboratory, The Affiliated Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Xiaohui Zhou
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Lu Kuang
- Department of Clinical Laboratory, The Affiliated Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qijun Chen
- Department of Clinical Laboratory, The Affiliated Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiaxing Zhao
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Huayu Yin
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Zeyu Zhou
- Department of Clinical Laboratory, The Affiliated Hospital of Chengde Medical College, Cengde,, Hebei,, China
| | - Xuehui Liu
- Department of Clinical Laboratory, The Affiliated Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dabin Liu
- Department of Clinical Laboratory, The Affiliated Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shaoguo Wu
- Department of Clinical Laboratory, The Affiliated Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Limei Wu
- Department of Clinical Laboratory, The Affiliated Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China.
| |
Collapse
|
20
|
Eissa MM, Allam SRA, Ismail CA, Ghazala RA, El Skhawy N, Zaki IIA, Ibrahim EIES. Unveiling the anti-neoplastic potential of Schistosoma mansoni-derived antigen against breast cancer: a pre-clinical study. Eur J Med Res 2025; 30:304. [PMID: 40247360 PMCID: PMC12007238 DOI: 10.1186/s40001-025-02531-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/28/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Cancer is a global health concern, with millions of new cases and deaths annually. Recently, immunotherapy has strengthened cancer treatment by harnessing the body's immune system to fight cancer. The search for advanced cancer immunotherapies has expanded to explore pathogens like parasites for their potential anti-neoplastic effects. While some parasites have shown promising results, the role of Schistosoma mansoni in breast cancer remains unexplored. METHODS This pre-clinical study investigated the anti-neoplastic potential of autoclaved Schistosoma mansoni antigen against breast cancer. In vitro, autoclaved Schistosoma mansoni antigen was evaluated on the MCF-7 human breast cancer cell line, while in vivo experiments used a chemically induced breast cancer rat model to evaluate tumour growth, liver enzyme levels, and immune response. Histopathological and immunohistochemical analyses assessed changes in tumour tissue, cell proliferation (Ki-67), angiogenesis (CD31), immune cell infiltration (CD8+ T cells), regulatory T cells (FoxP3+), and programmed death ligand 1 (PD-L1) expression. RESULTS In vitro, autoclaved Schistosoma mansoni antigen significantly reduced MCF-7 cell viability in a dose- and time-dependent manner. In vivo, autoclaved Schistosoma mansoni antigen treatment significantly reduced tumour weight and volume, improved liver enzyme levels, increased tumour necrosis, and decreased fibrosis. Immunohistochemical analysis revealed decreased Ki-67 and CD31 expression, indicating reduced cell proliferation and angiogenesis, respectively. Autoclaved Schistosoma mansoni antigen also enhanced immune responses by increasing CD8+ T cells infiltration and decreasing FoxP3+ expression, resulting in a higher CD8+ T cells/FoxP3+ ratio within the tumour microenvironment. Notably, PD-L1 expression was also downregulated, suggesting potential immune checkpoint inhibition. CONCLUSIONS Autoclaved Schistosoma mansoni antigen demonstrated potent anti-neoplastic activity, significantly reducing tumour growth and modulating the immune response within the tumour microenvironment. These results highlight autoclaved Schistosoma mansoni antigen's potential as a novel immunotherapy for breast cancer.
Collapse
Affiliation(s)
- Maha Mohamed Eissa
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Al-Moassat Medical Campus, Alexandria, Egypt.
| | - Sonia Rifaat Ahmed Allam
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Al-Moassat Medical Campus, Alexandria, Egypt
| | - Cherine Adel Ismail
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Rasha Abdelmawla Ghazala
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Nahla El Skhawy
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Al-Moassat Medical Campus, Alexandria, Egypt
| | | | - Eman Ibrahim El-Said Ibrahim
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Al-Moassat Medical Campus, Alexandria, Egypt
| |
Collapse
|
21
|
Timnik VR, Zoeschg A, Diederich S, Nefzger SM, Huang Z, Schmid NA, Giller M, Steiger K, Combs SE, Kroemer G, Schmid TE, Fischer JC. Experimental Investigation of Hematological Toxicity After Radiation Therapy Combined With Immune Checkpoint Inhibitors. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00372-4. [PMID: 40250771 DOI: 10.1016/j.ijrobp.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/28/2025] [Accepted: 04/05/2025] [Indexed: 04/20/2025]
Abstract
PURPOSE Combining immune checkpoint inhibitors (ICIs) with radiation therapy (RT) has led to significant advancements in cancer treatment. However, evidence from clinical and experimental studies suggests that this combination may increase hematopoietic and lymphatic toxicity. This study aims to investigate the effects of the concurrent application of ICIs (anti-PD-1 and anti-CTLA-4) on radiation-induced hematopoietic and lymphatic injuries under standardized and controlled experimental conditions. METHODS AND MATERIALS We used various experimental models in C57BL/6 and BALB/c mice to evaluate the impact of ICIs combined with RT on the hematopoietic system. These models involved different RT doses, regimens, and target sites in both healthy and tumor-bearing mice. RESULTS Our findings showed that the concurrent use of ICIs did not meaningfully affect post-RT pancytopenia kinetics or the regeneration of specific blood cell lineages over time. Consistently, combining RT with ICIs did not significantly enhance DNA damage in immune cells within the bloodstream. This outcome was comparable across different RT doses, regimens, and target sites and was reproducible in both tumor-bearing and nontumor-bearing mice. Additionally, there were no significant increases in late side effects, including reductions in bone marrow cell counts or megakaryocyte numbers, after combined radioimmunotherapy. CONCLUSIONS These findings suggest that combining ICIs with RT does not exacerbate hematological toxicity. This information is valuable for interpreting adverse events in clinical trials involving radioimmunotherapy and for predicting potential hematological side effects in cancer patients receiving these treatments.
Collapse
Affiliation(s)
- Vincent R Timnik
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Andreas Zoeschg
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Sarah Diederich
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Sophie M Nefzger
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Ziyi Huang
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Nicole A Schmid
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Maximilian Giller
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Katja Steiger
- Comparative Experimental Pathology (CEP), Institute of Pathology, School of Medicine, Technical University of Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany; Helmholtz Zentrum München, Institute of Radiation Medicine, Neuherberg, Germany
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, France-HP, Paris, France
| | - Thomas E Schmid
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany; Helmholtz Zentrum München, Institute of Radiation Medicine, Neuherberg, Germany
| | - Julius C Fischer
- Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France.
| |
Collapse
|
22
|
Li G, Guo Y, Zhang Y. Role of necroptosis and immune infiltration in essential thrombocytosis. Hereditas 2025; 162:62. [PMID: 40229837 PMCID: PMC11995491 DOI: 10.1186/s41065-025-00428-1] [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: 01/17/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Necroptosis, a recently identified form of programmed cell death involved in the pathogenesis of a variety of tumor and non-tumor diseases. Nevertheless, the function of necroptosis in essential thrombocytosis (ET) remains unclear, which is a classic myeloproliferative tumor. MATERIALS AND METHODS The role of necroptosis in ET was determined via bioinformatics combined with qRT-PCR analysis of clinical samples. GSE57793 and GSE26049 datasets were recruited to identify necroptosis differentially expressed genes based on differential gene identification, necroptosis gene sets and data machine learning. Enrichment analysis (GSEA) was used to evaluate the gene enrichment signaling pathway of ET, immune infiltration analysis was used to explore the abundance of immune cell infiltration in ET, and the correlation between necroptosis differential genes and immune cell infiltration was studied. RESULTS Five necroptosis genes were recognized to be remarkably enriched in the necroptosis pathway, including CHMP1B, FTH1, HSP90AB1, IL1A, and RBCK1. The imbalance of invasion of Th1/Th17 cells was identified in ET, and the differential necroptosis gene was positively correlated with the infiltration of multiple immune cells. There is significant necroptosis in ET, which is enriched in the necrotizing apoptotic pathway, and is associated with immune infiltration. CONCLUSIONS Necroptosis might drive the progression of ET via stimulating immune infiltration and immune responses. The findings bring new insights into the treatment mechanism and treatment strategy of ET in the future.
Collapse
Affiliation(s)
- Guangming Li
- Surgery Base Training, Shanghai Fengxian District Central Hospital, Shanghai, 201499, China
| | - Ying Guo
- Department of Hematology, Daqing Oilfield General Hospital, Daqing City, Heilongjiang Province, 163001, China
| | - Yuanyuan Zhang
- Internal Medicine Base Training, Shanghai Fengxian District Central Hospital, No.6600, Nanfeng Highway, Nanqiao Town, Fengxian District, Shanghai, 201499, China.
| |
Collapse
|
23
|
Zhu X, Li B, Qin L, Liang T, Hu W, Li J, Wang J. Development and Validation of a Prognostic Model for Lung Adenocarcinoma Based on CAF-Related Genes: Unveiling the Role of COX6A1 in Cancer Progression and CAF Infiltration. Int J Mol Sci 2025; 26:3478. [PMID: 40331946 PMCID: PMC12026577 DOI: 10.3390/ijms26083478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
Lung adenocarcinoma (LUAD), the predominant subtype of non-small cell lung cancer (NSCLC), presents significant challenges in early diagnosis and personalized treatment. Recent research has focused on the role of the tumor microenvironment, particularly tumor-associated fibroblasts (CAFs), in tumor progression. This study systematically analyzed CAF immune infiltration-related genes to construct a prognostic model for LUAD, confirming its predictive value for patient outcomes. The risk score derived from CAF-related genes (CAFRGs) was negatively correlated with immune microenvironment scores and linked to the expression of immune checkpoint genes, indicating that high-risk patients may exhibit immune escape characteristics. Analysis via the TIDE tool revealed that low-risk patients had more active T-cell immune responses. The risk score also correlated with anti-tumor drug sensitivity, particularly to doramapimod. Notably, COX6A1 emerged as a key gene in the model, with its upregulation associated with immune cell infiltration and immune escape. Further in vitro experiments demonstrated that COX6A1 regulates LUAD cell migration, proliferation, and senescence, suggesting its role in tumor immune evasion. Additionally, further co-culture studies of lung cancer cells and fibroblasts revealed that COX6A1 knockdown promotes the expression of CAF-related cytokines, enhancing CAF infiltration. Overall, this study provides a foundation for personalized treatment of LUAD and highlights COX6A1 as a promising therapeutic target within the tumor immune microenvironment, guiding future clinical research.
Collapse
Affiliation(s)
| | | | | | | | | | - Jianxiang Li
- School of Public Health, Suzhou Medicine College of Soochow University, Suzhou 215123, China; (X.Z.); (B.L.); (L.Q.); (T.L.); (W.H.)
| | - Jin Wang
- School of Public Health, Suzhou Medicine College of Soochow University, Suzhou 215123, China; (X.Z.); (B.L.); (L.Q.); (T.L.); (W.H.)
| |
Collapse
|
24
|
Liu L, Li X, Yang H, Xu F, Dong X. Bioinformatic Analysis of Apoptosis-Related Genes in Preeclampsia Using Public Transcriptomic and Single-Cell RNA Sequencing Datasets. J Inflamm Res 2025; 18:4785-4812. [PMID: 40224388 PMCID: PMC11992479 DOI: 10.2147/jir.s507660] [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: 11/25/2024] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
Purpose Apoptosis, which is crucial in preeclampsia (PE), affects trophoblast survival and placental function. We used transcriptomics and single-cell RNA sequencing (scRNA-seq) to explore apoptosis-related genes (ARGs) and their cellular mechanisms as potential PE biomarkers. Patients and Methods All the data included in this study were sourced from public databases. We used scRNA-seq and differential expression analysis, combined with five algorithms from the CytoHubba plugin, to identify ARGs as PE biomarkers. These were integrated into diagnostic nomograms. Mechanistic studies involved enrichment analysis and immune profiling. Biomarker expression was examined at the single-cell level, and verified in clinical samples by RT-qPCR. Results Cluster of Differentiation 44 (CD44), Macrophage migration inhibitory factor (MIF), PIK3R1, and Toll-like receptor 4 (TLR4) were identified as PE biomarkers. CD44 and TLR4 were down-regulated, while MIF and PIK3R1 were up-regulated. When integrated into the diagnostic nomogram, they showed clinical utility and affected cell functions. In the immune profile of PE, monocytes decreased, resting NK cells increased, and the activities of APC, checkpoint, T-cell co-stimulation, and MHC class I pathways reduced. ScRNA-seq identified 11 cell types, 10 of which were significantly different. Endothelial cell communication with other cell types decreased, while the interaction between common myeloid progenitors (CMP) and villous cytotrophoblasts (VCT) enhanced. The expression levels of CD44, MIF, and PIK3R1 in VCT were significantly different and key to PE. Their decrease in early PE and increase in late PE reflected the placenta's adaptation to adverse pregnancy conditions. Conclusion Four ARGs, CD44, MIF, PIK3R1, and TLR4, identified through comprehensive analyses, served as significant biomarkers for PE and offered insights into PE's cellular mechanisms of PE, providing valuable references for further research.
Collapse
Affiliation(s)
- Lingyan Liu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Department of Obstetrics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
| | - Xiuling Li
- Department of Obstetrics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
| | - Hongfen Yang
- Department of Obstetrics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
| | - Fei Xu
- Medical School, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Department of Pain Management, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
| | - Xudong Dong
- Department of Obstetrics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, People’s Republic of China
| |
Collapse
|
25
|
Kalemoglu E, Jani Y, Canaslan K, Bilen MA. The role of immunotherapy in targeting tumor microenvironment in genitourinary cancers. Front Immunol 2025; 16:1506278. [PMID: 40260236 PMCID: PMC12009843 DOI: 10.3389/fimmu.2025.1506278] [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: 10/04/2024] [Accepted: 03/19/2025] [Indexed: 04/23/2025] Open
Abstract
Genitourinary (GU) cancers, including renal cell carcinoma, prostate cancer, bladder cancer, and testicular cancer, represent a significant health burden and are among the leading causes of cancer-related mortality worldwide. Despite advancements in traditional treatment modalities such as chemotherapy, radiotherapy, and surgery, the complex interplay within the tumor microenvironment (TME) poses substantial hurdles to achieving durable remission and cure. The TME, characterized by its dynamic and multifaceted nature, comprises various cell types, signaling molecules, and the extracellular matrix, all of which are instrumental in cancer progression, metastasis, and therapy resistance. Recent breakthroughs in immunotherapy (IO) have opened a new era in the management of GU cancers, offering renewed hope by leveraging the body's immune system to combat cancer more selectively and effectively. This approach, distinct from conventional therapies, aims to disrupt cancer's ability to evade immune detection through mechanisms such as checkpoint inhibition, therapeutic vaccines, and adoptive cell transfer therapies. These strategies highlight the shift towards personalized medicine, emphasizing the importance of understanding the intricate dynamics within the TME for the development of targeted treatments. This article provides an in-depth overview of the current landscape of treatment strategies for GU cancers, with a focus on IO targeting the specific cell types of TME. By exploring the roles of various cell types within the TME and their impact on cancer progression, this review aims to underscore the transformative potential of IO strategies in TME targeting, offering more effective and personalized treatment options for patients with GU cancers, thereby improving outcomes and quality of life.
Collapse
Affiliation(s)
- Ecem Kalemoglu
- Department of Internal Medicine, Rutgers-Jersey City Medical Center, Jersey City, NJ, United States
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Türkiye
| | - Yash Jani
- Medical College of Georgia, Augusta, GA, United States
| | - Kubra Canaslan
- Department of Medical Oncology, Dokuz Eylul University, Izmir, Türkiye
| | - Mehmet Asim Bilen
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, United States
- Department of Urology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
26
|
Song Z, Sun Q, Yang W, Li Y, Hu C, Chen C, Liu K, Shen W, Yang Y, Yin D. Inflammation-targeted nanomedicine prevents tumor metastasis following photodynamic therapy by reversing epithelial-mesenchymal transition and ROS-mediated immunosuppression. J Nanobiotechnology 2025; 23:271. [PMID: 40186261 PMCID: PMC11969706 DOI: 10.1186/s12951-025-03332-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/14/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Prolonging the duration of photodynamic therapy (PDT) enhances the level of reactive oxygen species (ROS), thereby facilitating tumor ablation. However, our findings indicated that excessive ROS not only induces epithelial-mesenchymal transition (EMT) but also creates an immunosuppressive microenvironment in tumor, thereby triggering tumor metastasis. METHODS We initially developed neutrophil membrane hybrid liposomes (NLs) that can specifically target inflamed tumor tissues following PDT. Then, we utilized NLs to encapsulate the antioxidant nanozyme FeGA and the antiplatelet drug Lysine Acetylsalicylate (LAS), resulting in the formulation NLASF. RESULTS Experimental results demonstrated that FeGA effectively scavenges ROS, thereby reversing the immunosuppressive microenvironment induced by prolonged PDT. Furthermore, the incorporation of LAS effectively disrupts the interaction between tumor cells and platelets, mitigating tumor EMT and inhibiting hematogenous tumor metastasis. In a breast cancer mouse model, we observed that treatment with NLASF led to a near-complete suppression of tumor lung metastasis following the prolonged PDT. Additionally, the in vivo application of NLASF did not result in any blood toxicity or organ toxicity, highlighting its significant advantages over the free drugs group. CONCLUSIONS This study provides a novel approach to enhance the efficacy of PDT and successfully suppress PDT-mediated tumor metastasis.
Collapse
Affiliation(s)
- Zhengwei Song
- School of Pharmacy, Anhui University of Chinese Medicine, #1 Qian Jiang Road, Hefei, 230031, Anhui, PR China
| | - Quanwei Sun
- School of Pharmacy, Anhui University of Chinese Medicine, #1 Qian Jiang Road, Hefei, 230031, Anhui, PR China
| | - Wenshuo Yang
- School of Pharmacy, Anhui University of Chinese Medicine, #1 Qian Jiang Road, Hefei, 230031, Anhui, PR China
| | - Yunlong Li
- School of Pharmacy, Anhui University of Chinese Medicine, #1 Qian Jiang Road, Hefei, 230031, Anhui, PR China
| | - Chaoyu Hu
- School of Pharmacy, Anhui University of Chinese Medicine, #1 Qian Jiang Road, Hefei, 230031, Anhui, PR China
| | - Chen Chen
- School of Pharmacy, Anhui University of Chinese Medicine, #1 Qian Jiang Road, Hefei, 230031, Anhui, PR China
| | - Kang Liu
- School of Pharmacy, Anhui University of Chinese Medicine, #1 Qian Jiang Road, Hefei, 230031, Anhui, PR China
| | - Wei Shen
- School of Pharmacy, Anhui University of Chinese Medicine, #1 Qian Jiang Road, Hefei, 230031, Anhui, PR China.
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, 230031, China.
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, China.
| | - Ye Yang
- School of Pharmacy, Anhui University of Chinese Medicine, #1 Qian Jiang Road, Hefei, 230031, Anhui, PR China.
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, 230031, China.
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, China.
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, #1 Qian Jiang Road, Hefei, 230031, Anhui, PR China.
- Anhui Provincial Key Laboratory of Chinese Medicinal Formula, Hefei, 230021, China.
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, China.
| |
Collapse
|
27
|
Martel Matos AA, Scheff NN. Sensory neurotransmission and pain in solid tumor progression. Trends Cancer 2025; 11:309-320. [PMID: 39884880 DOI: 10.1016/j.trecan.2025.01.003] [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: 10/09/2024] [Revised: 12/17/2024] [Accepted: 01/06/2025] [Indexed: 02/01/2025]
Abstract
Sensory nerves form a crucial component of the tumor microenvironment (TME) that relays vital information to the central nervous system and modulates tumor progression via immunosurveillance. Afferent activity processed by the brain can sensitize brain circuitry and influence host behaviors. Peripheral sensory signaling (e.g., release of neuropeptides in the TME) can drive phenotypic changes in the tumor immune response, such as increased exhaustion markers and inhibited effector cell activity, which promote cancer progression. In this review we highlight the most recent evidence demonstrating the pivotal role of the sensory nervous system in cancer, with a focus on primary tumor pain, and we discuss the extent to which pain can influence cancer progression and treatment response, including immunotherapeutic strategies.
Collapse
Affiliation(s)
- Andre A Martel Matos
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA; Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Nicole N Scheff
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA; Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
28
|
Kang Y, Meng Y, Jin J, Dai Y, Li F, Chen N, Xie H, Cui Y. Mitochondrial metabolism-related features guiding precision subtyping and prognosis in breast cancer, revealing FADS2 as a novel therapeutic target. Transl Oncol 2025; 54:102330. [PMID: 39986190 PMCID: PMC11904520 DOI: 10.1016/j.tranon.2025.102330] [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: 10/26/2024] [Revised: 01/27/2025] [Accepted: 02/13/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Breast cancer is one of the most prevalent malignant tumors in women. Mitochondria, essential for cellular function, have altered metabolic activity in cancer cells, influencing tumor regulation and clinical outcomes. The connection between mitochondrial metabolism-related genes and breast cancer prognosis remains underexplored. This study aims to investigate the role of these genes in breast cancer by constructing risk models. METHODS Breast cancer transcriptome data were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), and mitochondrial gene data were sourced from the MitoCarta3.01 database. Clustering analysis was conducted using the "ConsensusClusterPlus" package, followed by Gene Set Enrichment Analysis (GSEA), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. A prognostic model was built using Cox regression and Least Absolute Shrinkage and Selection Operator (LASSO) algorithms. Immune cell infiltration levels were assessed via CIBERSORT and MCPcounter algorithms. Validation of key gene expression was performed on breast cancer tissue specimens and cell models to explore their biological functions in breast cancer cells. RESULTS The LASSO regression analysis of the TCGA BRCA dataset identified four prognosis-related mitochondrial metabolism genes: MYH11, LTF, FADS2, and PSPHP1. Validation using the GEO dataset confirmed that patients with high-risk scores (based on these four genes) had shorter overall survival compared to those with lower risk scores. Immunological analysis revealed that high-risk patients were less responsive to immunotherapy but more sensitive to conventional chemotherapies. This suggests that combining chemotherapy with immunotherapy might enhance T cell-based treatments. Univariate and multivariate Cox regression confirmed that the mitochondrial gene model was an independent predictor of overall survival, and a nomogram was developed to predict patient prognosis. Tissue validation showed consistent expression patterns with bioinformatic predictions. Functional assays confirmed that FADS2 was highly expressed in breast cancer cells, and its knockout significantly reduced cell invasion, migration, and colony formation. CONCLUSION This study reveals that mitochondrial metabolism-related genes are closely associated with breast cancer progression, clinical outcomes, and genetic alterations. The findings may offer new avenues for treatment strategies, early intervention, and prognosis prediction in breast cancer.
Collapse
Affiliation(s)
- Yakun Kang
- Department of Breast Surgery, The First Hospital Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China; Affiliated Zhongda Hospital of Southeast University, Nanjing 210009, China
| | - You Meng
- Department of Thyroid and Breast Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215002, China
| | - Jiangdong Jin
- Department of Breast Surgery, The First Hospital Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Yuhan Dai
- Department of Breast Surgery, The First Hospital Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Fei Li
- Nanjing Medical University, Nanjing 211166, China
| | - Nuo Chen
- Nanjing Medical University, Nanjing 211166, China
| | - Hui Xie
- Department of Breast Surgery, The First Hospital Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.
| | - Yangyang Cui
- Department of Breast Surgery, The First Hospital Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
29
|
Zhang H, Fan MD, Hu Y, Yang Q, Jiang JW, Xu M. Prognostic Assessment and Analysis of Underlying Biological Mechanisms of Prostate Cancer Based on Estrogen-Related Genes. Horm Metab Res 2025; 57:273-285. [PMID: 40209748 DOI: 10.1055/a-2548-1568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Abstract
Prostate cancer (PCa) ranks among the most prevalent cancers in men, noted for its high mortality rate and unfavorable prognosis. Estrogen-related genes (ERGs) are significantly associated with the progression of PCa. This investigation aims to comprehensively assess the prognosis of PCa based on ERGs and explore its underlying biological mechanisms. Univariate, multivariate, and Least Absolute Shrinkage and Selection Operator (LASSO) regression analyses were conducted to identify prognostic signature genes and build a prognostic model. The model's predictive performance was assessed using Receiver Operating Characteristic (ROC) curve analysis. Gene Set Enrichment Analysis (GSEA), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were employed to investigate the underlying molecular mechanisms of PCa. Antitumor drugs with high sensitivity were predicted using the CellMiner database and the pRRophitic package. Additionally, miRNAs targeting the identified signature genes were predicted using the miRNet database. This study identified six ERGs as prognostic biomarkers for PCa: POU4F1, BMP2, PGF, GAS1, GNAZ, and FGF11. The findings indicated that individuals in the low-risk category exhibited improved prognostic results. Notably, PCa progression may be closely linked to the cell adhesion molecule pathway and epigenetic regulation. Additionally, hsa-let-7a-5p and hsa-miR-34a-5p were identified as potential therapeutic regulators for PCa treatment. In conclusion, this research offers novel perspectives into the progression of PCa, providing robust scientific support for the development of personalized treatment strategies for PCa patients.
Collapse
Affiliation(s)
- Heng Zhang
- Urology Department, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Meng-Die Fan
- Dental Department, Hubei Medical College Affiliated Shiyan People's Hospital, Hubei, China
| | - Yang Hu
- Urology Department, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Qing Yang
- Urology Department, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jia-Wei Jiang
- Urology Department, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Min Xu
- Urology Department, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
30
|
Zou Y, Que T, Sun P, Xie J, Shi D, Wu H, Tan JE. A novel PET tracer for noninvasive imaging the checkpoints expression of innate and adaptive immunity in tumors by simultaneously targeting CD24 and PD-L1. Bioorg Chem 2025; 157:108260. [PMID: 39952064 DOI: 10.1016/j.bioorg.2025.108260] [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: 12/24/2024] [Revised: 01/24/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
The success of tumor immunotherapy depends on the innate and adaptive immune responses, with CD24 and PD-L1 being key targets. DBP1 peptide is a novel bispecific D-peptide, targeting both CD24 and PD-L1 simultaneously. In this study, by radiolabeling DBP1 peptide, we developed a novel PET modality to noninvasively evaluate CD24 and PD-L1 expressions in tumors. To enhance the solubility of DBP1, a hydrophilic lysine was added into C-terminal residue of the peptide, which was then modified with a chelator NOTA to produce the radiotracer precursor NOTA-DBP1k. NOTA-DBP1k showed high affinity for CD24 (KD = 10.70 ± 0.70 nM) and PD-L1 (KD = 5.40 ± 0.61 nM). [68Ga]Ga-NOTA-DBP1k was synthesized with a high radiochemical yield (71 ± 3.0 %) and exhibited high hydrophilicity and stability. [68Ga]Ga-NOTA-DBP1k showed higher uptake in high CD24/PD-L1 expressed MCF-7 cells than that in low CD24/PD-L1 expressed U-87MG cells in vitro. In vivo, [68Ga]Ga-NOTA-DBP1k showed high uptake in MCF-7 tumors and had favorable tumor-to-background ratios by microPET imaging. On the contrary, low uptake was found in U-87MG tumors, which was significantly lower than that in MCF-7 tumors (0.42 ± 0.02 %ID/g vs. 1.01 ± 0.06 %ID/g, p < 0.05). The biodistribution study was consistent with the findings of microPET imaging results. These results demonstrated that [68Ga]Ga-NOTA-DBP1k can noninvasively image the CD24 and PD-L1 checkpoint expression of innate and adaptive immunity in tumors and may be helpful for guiding the CD24/PD-L1 dual-checkpoints blockage immunotherapy.
Collapse
Affiliation(s)
- Yijin Zou
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515 Guangdong Province, China
| | - Tianshi Que
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515 Guangdong Province, China
| | - Penghui Sun
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515 Guangdong Province, China
| | - Jiahao Xie
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515 Guangdong Province, China
| | - Dazhi Shi
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515 Guangdong Province, China
| | - Hubing Wu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515 Guangdong Province, China.
| | - Jian-Er Tan
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515 Guangdong Province, China.
| |
Collapse
|
31
|
Wang Z, Wang M, Wang M, Zhou R, Deng X, Ouyang X, Chu M, Wei X, Yang L, Liu J, Xu Y. From molecular design to clinical translation: dual-targeted CAR-T strategies in cancer immunotherapy. Int J Biol Sci 2025; 21:2676-2691. [PMID: 40303292 PMCID: PMC12035882 DOI: 10.7150/ijbs.108036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/10/2025] [Indexed: 05/02/2025] Open
Abstract
The pathogenesis of tumors involves various abnormalities at both the cellular and genetic levels. Chimeric antigen receptor (CAR)-T cell immunotherapy has emerged as a transformative treatment strategy that effectively addresses these challenges. While CAR-T therapy has shown remarkable success in treating hematological malignancies, limitations have been identified, particularly in single antigen-targeting CAR-T therapies. These limitations include antigenic mutation or loss, reduced efficacy against leukemia, and poor results in solid tumors due to factors like low CAR-T cell persistence, limited tumor infiltration, rapid cell exhaustion, the suppressive tumor microenvironment, and heterogeneous tumor antigen expression. In recent years, multi-antigen targeted CAR-T therapies have garnered significant attention for their potential to prevent tumor relapse and progression. This review outlines the fundamental design of dual CAR structures and summarizes the major advancements in both preclinical studies and clinical trials of dual-targeted CAR-T cell therapy, categorized by cancer type. Additionally, it discusses the challenges associated with dual-targeted CAR-T therapy and the strategies to enhance its efficacy and applicability in treating both hematologic and solid tumors. In conclusion, the progress in dual-targeted CAR-T cell therapy presents a promising therapeutic avenue for multiple malignancies, offering insights into future modifications of immunotherapy to advance the field.
Collapse
Affiliation(s)
- Zhenrong Wang
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Mengyi Wang
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Mengting Wang
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Ruijie Zhou
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Xiaotong Deng
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Xin Ouyang
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Minghui Chu
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Xinyu Wei
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Lei Yang
- People's Hospital of Jingyang County, Xianyang, Shaanxi, 713700, China
| | - Jinbiao Liu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, Hubei, 430068, China
- Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
| | - Yao Xu
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| |
Collapse
|
32
|
Yang C, Du J, Qiu X, Jia C, Ding C, Wu Y, Gao C, Wang W, Wang X, Liu SB. ERCC3 serves as a prognostic biomarker for hepatocellular carcinoma and positively regulates cell proliferation and migration. Discov Oncol 2025; 16:419. [PMID: 40155569 PMCID: PMC11953519 DOI: 10.1007/s12672-025-02194-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/20/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND ERCC3, a crucial component of the nucleotide excision repair pathway, is implicated in the development and progression of various cancers and is a potential indicator of poor prognosis. However, the expression and function of ERCC3 in hepatocellular carcinoma (HCC) remain unclear. This study aimed to investigate the expression of ERCC3 in HCC tissues and its clinical significance, focusing on elucidating its potential mechanisms and therapeutic value in immunotherapy. METHODS The differential expression and genetic variation characteristics of ERCC3 across various cancers were evaluated using the TCGA database. The expression and prognostic value of ERCC3 in HCC were analyzed by integrating TCGA, GEO, and ICGC datasets. Independent prognostic value of ERCC3 expression levels in HCC was assessed using Cox regression analysis, Kaplan-Meier survival analysis, receiver operating characteristic curves, and nomograms. Pathway association scores were determined using ssGSEA to reveal the biological functions of ERCC3 in HCC and its potential clinical efficacy in immunotherapy. Stable transient cell lines were established by infecting HepG2 cells with lentivirus overexpressing ERCC3. The effects of ERCC3 on HCC cell biological phenotypes were evaluated using RTCA, wound healing, and Transwell assays. Cell cycle distribution and apoptosis were detected by flow cytometry. Transcriptome sequencing was performed to explore the impact of ERCC3 overexpression on the expression of signaling pathway-related genes in HCC. RESULTS The study revealed that ERCC3 is aberrantly expressed in various tumors, with significantly higher mRNA and protein levels in HCC tissues compared to normal tissues. High ERCC3 expression was significantly correlated with poor survival outcomes in HCC patients. Multivariate Cox regression analysis revealed that ERCC3 expression level is an independent prognostic factor for overall survival (P = 0.014). Gene sets associated with the high ERCC3 group were significantly involved in multiple immune pathways and tumor progression-related pathways, and ERCC3 expression was significantly correlated with immune checkpoints in HCC. Overexpression of ERCC3 promoted the proliferation and migration of HCC cells and influenced cell cycle progression. Transcriptome sequencing analysis indicated that ERCC3 overexpression regulated the proliferation of HCC cells, participated in multiple pro-inflammatory pathways, induced the formation of an inflammatory tumor microenvironment, and promoted HCC progression. CONCLUSION This study is the first to reveal the association between high ERCC3 expression and poor prognosis in HCC and to elucidate its immunomodulatory role in HCC. Unlike previous studies, we found that ERCC3 promotes HCC progression by regulating the inflammatory microenvironment and immune checkpoints. These findings establish a novel theoretical foundation for the development of targeted immunotherapies for HCC and provide new insights into the molecular mechanisms underlying ERCC3's role in HCC.
Collapse
Affiliation(s)
- Chen Yang
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan, 063210, China
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Kehua Road 28, Suzhou, 215009, China
| | - Jiahui Du
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Kehua Road 28, Suzhou, 215009, China
| | - Xiuqin Qiu
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Kehua Road 28, Suzhou, 215009, China
| | - Changhong Jia
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan, 063210, China
| | - Cunbao Ding
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan, 063210, China
| | - Yijie Wu
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan, 063210, China
| | - Chaoxu Gao
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan, 063210, China
| | - Weijie Wang
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan, 063210, China.
| | - Xiaojun Wang
- Department of Laboratory Medicine, Suzhou Wuzhong People's Hospital, Suzhou, 215128, Jiangsu, China.
| | - Song-Bai Liu
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan, 063210, China.
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Kehua Road 28, Suzhou, 215009, China.
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
33
|
Gao H, Sun F, Zhang X, Qiao X, Guo Y. The role and application of Coronin family in human tumorigenesis and immunomodulation. Biochim Biophys Acta Rev Cancer 2025; 1880:189304. [PMID: 40154644 DOI: 10.1016/j.bbcan.2025.189304] [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: 10/16/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
The Coronin family, a class of actin-binding proteins involved in the formation and maintenance of cytoskeleton structural stability, is aberrantly expressed in various tumors, including lung, gastric and head and neck cancers. They can regulate tumor cell metabolism and proliferation through RAC-1 and Wnt/β-Catenin signaling pathways and regulate invasion by influencing the PI3K, PAK4, and MT1-MMP signaling pathways and impacting the actin-network dynamics. In recent years, an increasing number of studies have highlighted the crucial roles of the cytoskeleton and immune modulation in the occurrence and development of tumors. The article delves into the Coronin family's pivotal role in tumor immune evasion, highlighting its modulation of neutrophil, T cell, and vesicular transport functions, as well as its interactions with tumorigenesis related organelles such as the endoplasmic reticulum, Golgi apparatus, mitochondria, and lysosomes. It also summarizes the potential therapeutic applications of the Coronin family in oncology. This review provides valuable insights into the mechanisms through which the Coronin family is implicated in the onset and progression of tumors. It also provides more theoretical foundation for tumor immunotherapy and combination drug therapy.
Collapse
Affiliation(s)
- Huimeng Gao
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China
| | - Fuli Sun
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China; Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Diseases, Shenyang, Liaoning 110002, China
| | - Xuanyu Zhang
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China
| | - Xue Qiao
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China; Department of Central Laboratory, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China.
| | - Yan Guo
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China; Department of Central Laboratory, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China.
| |
Collapse
|
34
|
Ingelshed K, Melssen MM, Spiegelberg D. Protocol for in vivo immune cell analysis in subcutaneous murine tumor models using advanced flow cytometry. STAR Protoc 2025; 6:103505. [PMID: 39823235 PMCID: PMC11786769 DOI: 10.1016/j.xpro.2024.103505] [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: 08/31/2024] [Revised: 10/22/2024] [Accepted: 11/14/2024] [Indexed: 01/19/2025] Open
Abstract
Here, we present a protocol for guiding tissue preparation and flow cytometric analysis in subcutaneous murine tumor models and secondary lymphoid organs. We describe steps for dissociating tumors, spleens, and lymph nodes to obtain single-cell suspensions. We then detail procedures for immune cell staining and analysis and gating strategies including the use of fluorescence-minus-one controls (FMOs). This approach provides valuable insights into the impact of cancer therapies on the tumor and systemic immune response. For complete details on the use and execution of this protocol, please refer to Ingelshed et al.1.
Collapse
Affiliation(s)
- Katrine Ingelshed
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden.
| | - Marit M Melssen
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden.
| | - Diana Spiegelberg
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden; Department of Surgical Sciences, Uppsala University, 75185 Uppsala, Sweden.
| |
Collapse
|
35
|
Narote S, Desai SA, Patel VP, Deshmukh R, Raut N, Dapse S. Identification of new immune target and signaling for cancer immunotherapy. Cancer Genet 2025; 294-295:57-75. [PMID: 40154216 DOI: 10.1016/j.cancergen.2025.03.004] [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: 01/01/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
Immunotherapy has become one of the innovative treatments in malignancy as it activates the immune system to find and eliminate malignant cells. The tumor immunology interface has become increasingly intricate, making the identification of new immune targets and signalling pathways on which to base improved therapeutic strategies an ongoing process. This review, we goal to clarify the contacts between cancer and immune system with a focus on immune surveillance as well as immune evasion mechanisms. Comprehensive immunotherapeutic therapies are overviewed with ICI (CTLA-4, PD-1, PD-L1), CAR-T cell therapy, and cancer vaccines whereas, advanced therapies targeting new immune checkpoints are also elucidated including TIM-3, LAG-3, and TIGIT. The JAK/STAT, MAPK and PI3K-AKT-mTOR pathways are reviewed with regards to cancer progression and immunotherapeutic resistance. The dysregulation of these pathways gives hope for the identification of fresh targets for therapy. Genomics, proteomics, immunopeptidomics, single cell mass spectrometry, CRISPR-based functional genomics and bioinformatics are described as essential for immune target identification and for mapping of cancer relevant signaling pathways. This review also considers some emerging issues in the subject area like the tumor heterogeneity, immune-related adverse events (irAEs), and personalized treatment. These barriers are described to facilitate the understanding of ways to overcome them and increase the efficacy of immunotherapies through combination therapies. This means that by developing new knowledge of immunological targets and pathways, immunoprecision medicine for cancer could greatly enhance outcomes.
Collapse
Affiliation(s)
- Sakshi Narote
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Sharav A Desai
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India.
| | - Vipul P Patel
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Rutuja Deshmukh
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Nikita Raut
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Sejal Dapse
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| |
Collapse
|
36
|
Chi X, Wang SH, Gao J, Su J, Du YZ, Xu XL. Carrier-Free Nanocombo-Sensitized Photoimmunotherapy via Activation of α2-Adrenergic Receptors. ACS APPLIED MATERIALS & INTERFACES 2025; 17:16437-16452. [PMID: 40040324 PMCID: PMC11931486 DOI: 10.1021/acsami.4c18052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 12/10/2024] [Accepted: 12/10/2024] [Indexed: 03/06/2025]
Abstract
Photodynamic therapy (PDT)-based photoimmunotherapy has attracted increasing attention in the field of cancer immunotherapy. Nonetheless, monotherapy alone proves insufficient in eliciting robust and enduring tumor immunogenicity within the "cold" microenvironment of triple-negative breast cancer. Therefore, it is imperative to integrate phototherapy and immunostimulation strategies to achieve synergistic effects. Here, we developed a carrier-free nanocombo comprising a photosensitizer (chlorin e6, Ce6) and an α2-adrenergic receptor (α2-AR) agonist (guanfacine, GFC) to enhance photoimmunotherapy through α2-AR activation. Ce6 and GFC possessed the ability to self-assemble into spherical nanoparticles, with the resulting Ce6-GFC (CeG) exhibiting exceptional drug loading efficiency (approaching 100%) and long-lasting colloidal stability, along with effective in vivo tumor-targeting capabilities. Following near-infrared laser irradiation, CeG-mediated phototherapy instigated a rapid generation of reactive oxygen species, leading to membrane disruption and the release of tumor-associated antigens, thereby facilitating dendritic cell maturation. Furthermore, α2-AR agonists served to repolarize M2 tumor-associated macrophages toward the M1 phenotype via adenylyl cyclase-mediated activation of α2-AR, thereby promoting the recruitment and activation of cytotoxic T lymphocytes. As a result, the carrier-free nanocombo significantly enhanced the efficacy of photoimmunotherapy in combatting poorly immunogenic breast tumors in female mice. Our findings showcase a "killing two birds with one stone" approach that boosts tumor immunogenicity, mitigates tumor immunosuppression, and advances the field of photoimmunotherapy.
Collapse
Affiliation(s)
- XiaoKai Chi
- Shulan International
Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
- College of
Pharmacy, Jiamusi University, Jiamusi 154007, PR China
- Institute
of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Si-Hui Wang
- Shulan International
Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - JingJing Gao
- Shulan International
Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - Jin Su
- College of
Pharmacy, Jiamusi University, Jiamusi 154007, PR China
| | - Yong-Zhong Du
- Institute
of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Xiao-Ling Xu
- Shulan International
Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| |
Collapse
|
37
|
ZHAO SHUANG, WEN HONGYONG, WANG BAIQI, XIONG QINGLIN, LI LANXIN, CHENG AILAN. p53: A player in the tumor microenvironment. Oncol Res 2025; 33:795-810. [PMID: 40191727 PMCID: PMC11964878 DOI: 10.32604/or.2025.057317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/09/2024] [Indexed: 04/09/2025] Open
Abstract
Approximately half of all cancers have p53 inactivating mutations, in addition to which most malignancies inactivate the p53 pathway by increasing p53 inhibitors, decreasing p53 activators, or inactivating p53 downstream targets. A growing number of researches have demonstrated that p53 can influence tumor progression through the tumor microenvironment (TME). TME is involved in the process of tumor development and metastasis and affects the clinical prognosis of patients. p53 participates in host immunity and engages in the immune landscape of the TME, but the specific mechanisms remain to be investigated. This review briefly explores the interactions between different states of p53 and TME components and their mechanisms, as well as their effects on tumor progression. To understand the progress of drug development and clinical studies related to p53 and tumor microenvironment.
Collapse
Affiliation(s)
- SHUANG ZHAO
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - HONGYONG WEN
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - BAIQI WANG
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - QINGLIN XIONG
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - LANXIN LI
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - AILAN CHENG
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, China
| |
Collapse
|
38
|
Zhang Y, Yan L, Sun H, Zhang Z, Shen F, Sun L. Targeted Delivery of Personalized Cancer Vaccines Based on Antibody-Antigen Complexes. Vaccines (Basel) 2025; 13:324. [PMID: 40266219 PMCID: PMC11946472 DOI: 10.3390/vaccines13030324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/15/2025] [Accepted: 03/16/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Personalized cancer vaccines based on tumor neoantigens show great potential in cancer immunotherapy due to their high safety and specificity. However, it is inherently difficult to realize the efficiently targeted delivery of personalized cancer vaccines to antigen-presenting cells (APCs). METHODS This study aimed to address these challenges by developing and evaluating a personalized cancer vaccine based on antibody-antigen complexes, which was designed to enhance antitumor effects by increasing the utilization of tumor neoantigens by APCs. Mice were immunized with a carrier protein, keyhole limpet hemocyanin (KLH), to induce the production of antibodies against KLH. Subsequently, mice were immunized with KLH loaded with tumor neoantigens and the immunoadjuvant CpG ODN and underwent immunological analysis to evaluate the immune and antitumor effects. RESULTS The results showed that preimmunization with KLH could promote the uptake of the personalized KLH-based tumor vaccine, which was enhanced by dendritic cells (DCs) and macrophages (Mφs), by strengthening the T-cell immune responses to tumors. CONCLUSIONS Collectively, this work provides a new idea for the targeted delivery of personalized cancer vaccines.
Collapse
Affiliation(s)
- Yaling Zhang
- School of Life Sciences, Shanghai University, Shanghai 200444, China (L.Y.)
| | - Lingling Yan
- School of Life Sciences, Shanghai University, Shanghai 200444, China (L.Y.)
| | - He Sun
- School of Life Sciences, Shanghai University, Shanghai 200444, China (L.Y.)
| | - Ziyi Zhang
- School of Life Sciences, Shanghai University, Shanghai 200444, China (L.Y.)
| | - Fengyun Shen
- Institute of Materiobiology, Department of Chemistry, College of Science, Shanghai University, Shanghai 200444, China
| | - Lele Sun
- Institute of Materiobiology, Department of Chemistry, College of Science, Shanghai University, Shanghai 200444, China
| |
Collapse
|
39
|
Cheng B, Li H, Hong Y, Zhou Y, Chen J, Shao C, Kong Z. Research progress in bifunctional small molecules for cancer immunotherapy. Eur J Med Chem 2025; 286:117289. [PMID: 39919914 DOI: 10.1016/j.ejmech.2025.117289] [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: 11/21/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 02/09/2025]
Abstract
Immunotherapy has become one of the most revolutionary modalities for cancer treatment with the approval of many anti-PD-L1 (programmed cell death-ligand 1)/PD-1 (programmed cell death-1) monoclonal antibodies (mAbs). However, anti-PD-L1/PD-1 mAbs suffer from several drawbacks including limited clinical efficacy (∼20 %), poor pharmacokinetics, and the development of immune resistance. Hence, the search for PD-1/PD-L1-based combination therapies and other PD-L1-based bifunctional small molecule modulators [e.g. PD-L1/HDAC (Histone Deacetylase), PD-L1/CXCL12 (C-X-C chemokine ligand 12), PD-L1/Tubulin, PD-L1/IDO1 (Indoleamine 2,3 dioxygenase 1), PD-L1/PARP (Poly(ADP-ribose) polymerase), PD-L1/STING (Stimulator of interferon genes), and PD-L1/NAMPT (Nicotinamide phosphoribosyltransferase)-targeting dual inhibitors] has been intensified with considerable strides achieved in the past couple of years. Herein, we summarize the latest development of bifunctional small molecules as immunotherapy for tumor treatment, including those PD-L1-based, A2AR (Adenosine 2A receptor)-based, IDO1-based, Toll-like receptor (TLR)-based, SHP2 (Src homology 2 domain-containing phosphatase 2)-based, and HPK1 (Hematopoietic progenitor kinase 1)-based dual-acting compounds. In addition, we also summarize the tumorigenesis and synergy mechanism of various targets. Finally, the challenges and future directions for bifunctional small molecules for cancer immunotherapy are also discussed in detail.
Collapse
Affiliation(s)
- Binbin Cheng
- Hubei Polytechnic University, Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, 435003, China; Central Laboratory, Wenzhou Medical University Lishui Hospital, Lishui People's Hospital, Lishui, Zhejiang, 323000, China
| | - Hongqiao Li
- The Central Hospital of Huangshi, Huangshi, 435000, China
| | - Yimeng Hong
- Hubei Polytechnic University, Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, 435003, China
| | - Yingxing Zhou
- Hubei Polytechnic University, Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, 435003, China; Huangshi Key Laboratory of Molecular Diagnosis and Individualized Treatment, Huangshi Love&health Hospital Affiliated of Hubei Polytechnic University, China.
| | - Jianjun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Chuxiao Shao
- Central Laboratory, Wenzhou Medical University Lishui Hospital, Lishui People's Hospital, Lishui, Zhejiang, 323000, China.
| | - Zhihua Kong
- Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, FoShan, 528200, China.
| |
Collapse
|
40
|
Arnet L, Emilius L, Hamann A, Carmo-Fonseca M, Berking C, Dörrie J, Schaft N. The Influence of Indisulam on Human Immune Effector Cells: Is a Combination with Immunotherapy Feasible? Pharmaceutics 2025; 17:368. [PMID: 40143032 PMCID: PMC11945250 DOI: 10.3390/pharmaceutics17030368] [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: 07/31/2024] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Background: As a modulator of pre-mRNA splicing, the anti-cancer agent indisulam can induce aberrantly spliced neoantigens, enabling immunologic anti-tumor activity. Consequently, combining indisulam with immunotherapy is expected to be a promising novel approach in cancer therapy. However, a prerequisite for such a combination is that immune effector cells remain functional and unharmed by the chemical. Methods: To ensure the immunocompetence of human immune effector cells is maintained, we investigated the influence of indisulam on ex vivo-isolated T cells and monocyte-derived dendritic cells (moDCs) from healthy donors. We used indisulam concentrations from 0.625 µM to 160 µM and examined the impact on the following: (i) the activation of CD4+ and CD8+ T cells by CD3-crosslinking and via a high-affinity TCR, (ii) the cytotoxicity of CD8+ T cells, (iii) the maturation process of moDCs, and (iv) antigen-specific CD8+ T cell priming. Results: We observed dose-dependent inhibitory effects of indisulam, and substantial inhibition occurred at concentrations around 10 µM, but the various functions of the immune system exhibited different sensitivities. The weaker activation of T cells via CD3-crosslinking was more sensitive than the stronger activation via the high-affinity TCR. T cells remained capable of killing tumor cells after treatment with indisulam up to 40 µM, but T cell cytotoxicity was impaired at 160 µM indisulam. While moDC maturation was also rather resistant, T cell priming was almost completely abolished at a concentration of 10 µM. Conclusions: These effects should be considered in possible future combinations of immunotherapy with the mRNA splicing inhibitor indisulam.
Collapse
Affiliation(s)
- Lisa Arnet
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Lisabeth Emilius
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Annett Hamann
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Maria Carmo-Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal;
| | - Carola Berking
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Jan Dörrie
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Niels Schaft
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| |
Collapse
|
41
|
Qian Y, Yang D, Lin X, Shen C, Zhang J, Xu J, Zhao Y, Zhu L, Kong H, Zhang M, Zhu Y, Zhou C, He J. Dendrimer-Derived Mimics of Host Defense Peptides Selectively Disrupt Cancer Cell Membranes for Melanoma Therapy. Pharmaceutics 2025; 17:361. [PMID: 40143025 PMCID: PMC11946345 DOI: 10.3390/pharmaceutics17030361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/18/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Melanoma is one of the most common malignancies, posing a significant health threat to patients, particularly in advanced stages due to its high aggressiveness. Chemotherapy agents with biocompatibility and low susceptibility to induce resistance are required for systematic management. Methods: Dendrimer-derived mimics (DMs) of host defense peptides (HDPs), which were constructed by a dendrimer core and optimized ratios of the hydrophobic arm, were used to treat A375 cells and HaCaT cells as the control. Live/dead staining, flow cytometry, and scanning electron microscopy (SEM) were conducted to analyze the anticancer mechanism. Mice with subcutaneous tumors were used to test the antitumor activity and toxicity in vivo. Results: DMs exhibited enhanced activity against A375 cells with remarkable selectivity, which mimics the action of natural HDPs and can cause damage to cell membranes. DMs can effectively inhibit solid tumor growth with minimal systemic toxicity and no adverse effects on healthy tissues. Conclusion: All the findings highlight DMs as promising anticancer candidates with significant potential for systemic melanoma therapy.
Collapse
Affiliation(s)
- Yusheng Qian
- Translational Medical Center for Stem Cell Therapy, Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Q.); (D.Y.); (X.L.); (C.S.); (J.Z.); (H.K.); (M.Z.)
- School of Material Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Danjing Yang
- Translational Medical Center for Stem Cell Therapy, Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Q.); (D.Y.); (X.L.); (C.S.); (J.Z.); (H.K.); (M.Z.)
| | - Xiangyu Lin
- Translational Medical Center for Stem Cell Therapy, Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Q.); (D.Y.); (X.L.); (C.S.); (J.Z.); (H.K.); (M.Z.)
| | - Chenyun Shen
- Translational Medical Center for Stem Cell Therapy, Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Q.); (D.Y.); (X.L.); (C.S.); (J.Z.); (H.K.); (M.Z.)
| | - Jieping Zhang
- Translational Medical Center for Stem Cell Therapy, Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Q.); (D.Y.); (X.L.); (C.S.); (J.Z.); (H.K.); (M.Z.)
| | - Jin Xu
- Laboratory Animal Center of Tongji University, Tongji University, Shanghai 200092, China;
| | - Yan Zhao
- Experimental Teaching Center for Medicine and Life Science, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Z.); (L.Z.)
| | - Ling Zhu
- Experimental Teaching Center for Medicine and Life Science, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Z.); (L.Z.)
| | - Haoran Kong
- Translational Medical Center for Stem Cell Therapy, Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Q.); (D.Y.); (X.L.); (C.S.); (J.Z.); (H.K.); (M.Z.)
| | - Mingyu Zhang
- Translational Medical Center for Stem Cell Therapy, Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Q.); (D.Y.); (X.L.); (C.S.); (J.Z.); (H.K.); (M.Z.)
| | - Yueqian Zhu
- Department of Dermatology, The First Affiliated Hospital of Soochow University, Suzhou 215031, China;
| | - Chuncai Zhou
- School of Material Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Jing He
- Translational Medical Center for Stem Cell Therapy, Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Q.); (D.Y.); (X.L.); (C.S.); (J.Z.); (H.K.); (M.Z.)
| |
Collapse
|
42
|
Lang S, Zhu Y, Tan Z, Zhang Y, Liang R, Ren J, Li P, Pan F, Cai L, Chen YH. Cancer immunotherapy by silencing transcription factor c-Rel using peptide-based nanoparticles. Front Immunol 2025; 16:1554496. [PMID: 40134427 PMCID: PMC11933105 DOI: 10.3389/fimmu.2025.1554496] [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: 01/02/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
Background Cancer immunotherapy has shown promising results in the clinic, but it faces great challenges such as low response rates and low efficacy in solid tumors. c-Rel, a member of the nuclear factor (NF)-κB family, is a newly described immune checkpoint for myeloid-derived suppressor cells (MDSCs), which contribute to the formation of immune-suppressive tumor microenvironment and resistance to cancer immunotherapy. How to selectively target myeloid c-Rel for the treatment of cancer is not well established. In this study, we investigated the feasibility and efficacy of knocking down myeloid c-Rel with siRNA-loaded peptide-based nanoparticles as a new cancer immunotherapy strategy. Methods The knockdown of c-Rel gene by the siRNA-loaded peptide nanoparticles was confirmed on MDSCs in vitro and in vivo. The effects of c-Rel silencing on cell number and immune suppressive function of the murine bone marrow-derived MDSCs were then investigated. To evaluate the anti-tumor efficacy of the c-Rel siRNA loaded nanoparticles, female C57BL/6 mice with subcutaneous B16 tumor were treated with PBS, c-Rel siRNA loaded nanoparticles, control siRNA loaded nanoparticles or empty nanoparticles. The tumor growth and body weight of mice were monitored, and the numbers and immune activities of tumor infiltrated immune cells in different groups were analyzed at the end of the experiment. The immune function of MDSCs isolated from tumor bearing mice received different treatments were further investigated ex vivo by T cell proliferation assays. Results The c-Rel siRNA nanoparticles significantly reduced c-Rel expression in MDSCs, diminished both the number and immune suppressive function of MDSCs, and enhanced intratumor CD8+ T cell responses. Significantly reduced tumor growth was observed in mice treated with the c-Rel siRNA nanoparticles compared to control mice. Conclusion Our data indicates that peptide-based nanoparticles can be successfully utilized to target the myeloid immune checkpoint c-Rel for the treatment of cancer.
Collapse
Affiliation(s)
- Shuyao Lang
- Center for Cancer Immunology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Key Laboratory of Cellular and Gene Therapy of Guangdong Province, Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
| | - Yuxuan Zhu
- Center for Cancer Immunology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zibin Tan
- Center for Cancer Immunology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Key Laboratory of Cellular and Gene Therapy of Guangdong Province, Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
| | - Yu Zhang
- Center for Cancer Immunology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ruijing Liang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jian Ren
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ping Li
- Center for Cancer Immunology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Key Laboratory of Cellular and Gene Therapy of Guangdong Province, Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
- Sino-European Center of Biomedicine and Health, Shenzhen, China
| | - Fan Pan
- Center for Cancer Immunology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Key Laboratory of Cellular and Gene Therapy of Guangdong Province, Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Sino-European Center of Biomedicine and Health, Shenzhen, China
| | - Youhai H. Chen
- Center for Cancer Immunology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Key Laboratory of Cellular and Gene Therapy of Guangdong Province, Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
- Sino-European Center of Biomedicine and Health, Shenzhen, China
| |
Collapse
|
43
|
Arroyo-Nogales A, Plaza-Palomo G, González-Larre J, Jiménez-Falcao S, Baeza A. Silicasomes in Oncology: From Conventional Chemotherapy to Combined Immunotherapy. Molecules 2025; 30:1257. [PMID: 40142031 PMCID: PMC11945772 DOI: 10.3390/molecules30061257] [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: 01/30/2025] [Revised: 02/21/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
The use of nanoparticles as drug carriers in oncology has evolved from their traditional role as chemotherapy carriers to their application in immunotherapy, exploiting not only their passive accumulation in solid tumors but also their ability to interact with immune cells. Silicasomes are highly versatile nanoplatforms composed of a mesoporous silica core whose external surface is coated with a lipid bilayer that allows the co-delivery of therapeutic agents having different chemical natures (small molecules, proteins, enzymes, or oligonucleotides, among others). Herein, cutting-edge advances carried out in the development and application of silicasomes are presented, providing a general description of the performance of these nanotransporters. Additionally, the specific load of chemotherapeutic drugs is explored, followed by a discussion of the immunotherapeutic application of silicasomes and the combination of different therapeutic strategies, including theragnosis, in a single silicasome platform, highlighting the enormous potential of these nanosystems.
Collapse
Affiliation(s)
| | | | | | | | - Alejandro Baeza
- Materials and Aerospace Production Department, Superior Technic School of Aeronautics and Space Engineering, Politechnic University of Madrid Department Materiales y Producción Aeroespacial, ETSI Aeronáutica y del Espacio, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (A.A.-N.); (G.P.-P.); (J.G.-L.); (S.J.-F.)
| |
Collapse
|
44
|
Chen M, Zhou X, Fan Y, Wang C. Identification and validation of prognostic biomarkers related to tumor immune invasion in pancreatic cancer. Front Genet 2025; 16:1556544. [PMID: 40129606 PMCID: PMC11931078 DOI: 10.3389/fgene.2025.1556544] [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: 01/10/2025] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
Background The diagnosis and treatment of pancreatic adenocarcinoma (PAAD) remain clinically challenging, and new molecular markers for prognostic assessment and targeted therapy are urgently needed. The tumor microenvironment (TME) and immune invasion play an important role in pancreatic cancer development and progression. Therefore, immunotherapeutic strategies based on the TME and immune invasion may have important clinical value. Methods In this study, we extracted transcriptome and clinicopathological data for 179 PAAD samples from the TCGA database and evaluated the immune composition, stromal composition, and infiltrating immune cell landscape in the tumor samples. Then, we identified relevant differentially expressed genes (DEGs) and performed functional annotation and prognostic correlation analysis to identify prognostic biomarkers for pancreatic cancer, the correlation between biomarkers and tumor immune invasion was analyzed to reveal the molecular immune mechanism of pancreatic cancer. Finally, GEO databases (GES71729), GEPIA, TISIDB, TIMER databases and RT-PCR were used for further analysis. Results CXCL10 and CXCL11 were highly expressed in pancreatic cancer and associated with poor prognosis of patients through cell adhesion molecules chemokine signaling, cytokine-cytokine receptor interaction, natural killer cell-mediated cytotoxicity, and Toll-like receptor signaling pathways. Finally, the correlation between CXCL10 and CXCL11 and tumor immune invasion was analyzed. The results confirmed that the expression levels of CXCL10 and CXCL11 were positively correlated with the contents of CD8+ T cells. Activated memory CD4+ T cells, M1 macrophages and resting mast cells. The levels of CXCL10 and CXCL11 were related to but negatively correlated with the contents of memory B cells, Tregs and M0 macrophages. Conclusion Our study demonstrates that CXCL10 and CXCL11 are novel biomarkers of TME and immune cell infiltration in pancreatic cancer by affecting the distribution of immune cells. CXCL10 and CXCL11 may be new targets for molecular targeted therapy and immunotherapy of pancreatic cancer.
Collapse
Affiliation(s)
| | | | | | - Chen Wang
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
45
|
Guo S, Lv G, Zhu H, Guo Y, Yin K, Yu H, Zhang H. Disulfidptosis related immune genes drive prognostic model development and tumor microenvironment characterization in bladder urothelial carcinoma. Sci Rep 2025; 15:8130. [PMID: 40057601 PMCID: PMC11890603 DOI: 10.1038/s41598-025-92297-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/26/2025] [Indexed: 05/13/2025] Open
Abstract
The intricate nature and varied forms of bladder urothelial carcinoma (BLCA) highlight the need for new indicators to define tumor prognosis. Disulfidptosis, a novel form of cell death, is closely linked to BLCA progression, prognosis, and treatment outcomes. Our current goal is to develop a novel disulfidptosis-related immune prognostic model to enhance BLCA treatment strategies. Utilizing RNA-seq data from The Cancer Genome Atlas (TCGA) , which included 419 patients (19 normal, 400 tumor), we performed weighted gene co-expression network analysis (WGCNA) to identify disulfidptosis-associated immune genes. Through multivariate Cox regression, and the least absolute shrinkage and selection operator (LASSO) regularization, we established a disulfidptosis-related immune risk scoring system. A nomogram combining risk score and clinical features predicted prognosis. Model performance was validated through survival curve analysis and independent validation cohort. Immune checkpoints, cell infiltration, and tumor mutation load were assessed. Differential gene enrichment analysis was conducted. Prognostic genes were validated via in vitro experiments. Eight immune genes related to disulfidptosis were identified and verified in BLCA prognosis. A prognostic model outperformed previous ones in predicting overall survival (OS) for high- and low-risk groups. Patients with low-risk scores had higher OS rates and tumor mutation burden (TMB) compared to high-risk score patients. CD4 memory T cells, CD8 T cells, M1 macrophages, and resting NK cells were found to be higher in the low-risk group. Immune checkpoint inhibitor (ICI) treatment may be more effective for the low-risk score group. High-risk score group exhibited stronger correlation with cancer malignant pathways. Knocking out tumor necrosis factor receptor superfamily member 12 A (TNFRSF12A) inhibits BLCA cell proliferation and invasion while overexpressing it has the opposite effect. We constructed a novel risk score model that combines disulfidptosis and immune genes, demonstrating good prognostic prediction performance. We discovered and verified that the TNFRSF12A gene is an oncogene in BLCA, which may help provide personalized guidance for individualized treatment and immunotherapy selection for BLCA patients to a certain extent.
Collapse
Affiliation(s)
- Shenchao Guo
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Guangjia Lv
- College of Life Sciences, Northeast Forestry University, Harbin, 150000, China
| | - Hengyue Zhu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yangyang Guo
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Ke Yin
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Haibo Yu
- Department of Hepatobiliary and Pancreatic Surgery, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, 325000, China, No. 252, Baili East Road, Zhejiang
| | - Hewei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, 325000, China, No. 252, Baili East Road, Zhejiang.
| |
Collapse
|
46
|
Tang Z, Chen C, Zhou C, Liu Z, Li T, Zhang Y, Feng Y, Gu C, Li S, Chen J. Insights into tumor-derived exosome inhibition in cancer therapy. Eur J Med Chem 2025; 285:117278. [PMID: 39823808 DOI: 10.1016/j.ejmech.2025.117278] [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: 10/02/2024] [Revised: 01/11/2025] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
Exosomes are critical mediators of cell-to-cell communication in physiological and pathological processes, due to their ability to deliver a variety of bioactive molecules. Tumor-derived exosomes (TDEs), in particular, carry carcinogenic molecules that contribute to tumor progression, metastasis, immune escape, and drug resistance. Thus, TDE inhibition has emerged as a promising strategy to combat cancer. In this review, we discuss the key mechanisms of TDE biogenesis and secretion, emphasizing their implications in tumorigenesis and cancer progression. Moreover, we provide an overview of small-molecule TDE inhibitors that target specific biogenesis and/or secretion pathways, highlighting their potential use in cancer treatment. Lastly, we present the existing obstacles and propose corresponding remedies for the future development of TDE inhibitors.
Collapse
Affiliation(s)
- Ziwei Tang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Cheng Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chen Zhou
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, United States
| | - Zhouyan Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tong Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ye Zhang
- School of Petrochemical Engineering, Changzhou University, Changzhou, 213164, China.
| | - Yanyan Feng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chenglei Gu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shijia Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jichao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
47
|
Ma C, Wu W, Zhang P, Xie J. Editorial: Unveiling biomarkers and mechanisms in the tumor-immune nexus. Front Immunol 2025; 16:1581492. [PMID: 40109343 PMCID: PMC11920117 DOI: 10.3389/fimmu.2025.1581492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/22/2025] Open
Affiliation(s)
- Chenfeng Ma
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, Jiangsu, China
| | - Wantao Wu
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Pengpeng Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jiaheng Xie
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
48
|
Ma J, Hu G, Kuang L, Zhu Z. Identification of the Molecular Subtype and Prognostic Characteristics of Breast Cancer Based on Tumor-Infiltrating Regulatory T Cells. Breast J 2025; 2025:6913291. [PMID: 40224950 PMCID: PMC11991805 DOI: 10.1155/tbj/6913291] [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: 10/08/2024] [Accepted: 02/19/2025] [Indexed: 04/15/2025]
Abstract
Background: T regulatory cells (Tregs) are essential for preserving immune tolerance. They are present in large numbers in many tumors, hindering potentially beneficial antitumor responses. However, their predictive significance for breast cancer (BC) remains ambiguous. This study aimed to explore genes associated with Tregs and develop a prognostic signature associated with Tregs. Methods: The gene expression and clinical data on BC were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. The integration of CIBERSORT and weighted correlation network analysis (WGCNA) algorithms was utilized to identify modules associated with Tregs. The consensus cluster algorithm was utilized to create molecular subtypes determined by genes associated with Tregs. Then, a prognostic signature associated with Tregs was constructed and its relationship to tumor immunity and the prognosis was evaluated. Results: The blue module genes exhibited the most significant correlation with Tregs, and 1080 genes related to Tregs were acquired. A total of 93 genes from the TCGA dataset were found to have a significant impact on patient prognosis. Samples from BC were categorized into two clusters by consensus cluster analysis. The overall survival, immune checkpoint genes, molecular subtype, and biological behaviors varied significantly between these two subtypes. A 10-gene signature developed from differentially expressed genes between two subtypes demonstrated consistent prognostic accuracy in both TCGA and GEO datasets. It functioned as a standalone prognostic marker for individuals with BC. In addition, patients with low risk are more inclined to exhibit increased immune cell infiltration, TME score, and tumor mutation burden (TMB). Meanwhile, Individuals classified within the low-risk group showed better responses to immunotherapies compared to their counterparts in the high-risk group. Conclusions: The prognostic model derived from Tregs-related genes could aid in assessing the prognosis, guiding personalized treatment, and potentially enhancing the clinical outcomes for patients with BC.
Collapse
Affiliation(s)
- Jianying Ma
- Department of Breast Surgery, Thyroid Surgery, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, Hubei, China
| | - Gang Hu
- Department of Breast Surgery, Thyroid Surgery, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lianghong Kuang
- Department of Neurology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, Hubei, China
| | - Zhongzhong Zhu
- Department of Gastroentero Rectal Surgery, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, Hubei, China
| |
Collapse
|
49
|
Zhang J, Wang F, Sun Z, Ye J, Chu H. Multidimensional applications of prussian blue-based nanoparticles in cancer immunotherapy. J Nanobiotechnology 2025; 23:161. [PMID: 40033359 PMCID: PMC11874808 DOI: 10.1186/s12951-025-03236-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/16/2025] [Indexed: 03/05/2025] Open
Abstract
Immunotherapy holds notable progress in the treatment of cancer. However, the clinical therapeutic effect remains a significant challenge due to immune-related side effects, poor immunogenicity, and immunosuppressive microenvironment. Nanoparticles have emerged as a revolutionary tool to surmount these obstacles and amplify the potency of immunotherapeutic agents. Prussian blue nanoparticles (PBNPs) exhibit multi-dimensional immune function in cancer immunotherapy, including acting as a nanocarrier to deliver immunotherapeutic agents, as a photothermal agent to improve the efficacy of immunotherapy through photothermal therapy, as a nanozyme to regulate tumor microenvironment, and as an iron donor to induce immune events related to ferroptosis and tumor-associated macrophages polarization. This review focuses on the advances and applications of PBNPs in cancer immunotherapy. First, the biomedical functions of PBNPs are introduced. Then, based on the immune function of PBNPs, we systematically reviewed the multidimensional application of PBNPs in cancer immunotherapy. Finally, the challenges and future developments of PBNPs-based cancer immunotherapy are highlighted.
Collapse
Affiliation(s)
- Jiayi Zhang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Fang Wang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Zhaogang Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China.
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
| |
Collapse
|
50
|
Nie X, Zhu Z, Liu Y, Zhang X, Chen J, Zhang F, Guo B. Fibroblast Growth Factor 11 Promotes Immune Escape of Cervical Cancer Cells by Promoting Infiltration of CD4 + T Cells, Particularly Regulatory T Cells. Genet Test Mol Biomarkers 2025; 29:85-98. [PMID: 40101238 DOI: 10.1089/gtmb.2024.0478] [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: 03/20/2025] Open
Abstract
Background: Cervical cancer (CC) is one of the leading gynecological malignancies. Immunotherapy has shown limited efficacy, particularly for advanced, recurrent CC. Consequently, dependable prognostic biomarkers and treatment targets are needed. Methods and Results: In this study, we aimed to determine the association of fibroblast growth factor 11 (FGF11) with prognosis. FGF11 expression was assessed in both tissues and cells through immunohistochemical and immunocytochemical staining. Immune cell infiltration was predicted using Tumor Immune Estimation Resource (TIMER) and TIMER2.0. FGF11 was significantly correlated with prognosis. FGF11 expression was significantly elevated in CC tissues. Moreover, FGF11 expression was significantly higher in SiHa and HeLa cancer cells than in normal H8 cells, particularly SiHa cells. Enrichment analyses suggested that FGF11 may be involved in arachidonic acid and linoleic acid metabolism, indicating roles in epithelial adhesion and cell differentiation. FGF11 correlated positively with CD4+ T, regulatory T, and dendritic cells but negatively with CD8+ T cells. FGF11 also correlated positively with Cluster of Differentiation 4 (CD4), CD25, Forkhead box P3 (FOXP3), and transforming growth factor β but negatively with human leukocyte antigens. Conclusions: FGF11 may enhance the immune escape abilities of CC cells by promoting CD4+ T cell infiltration (particularly regulatory T cells) into the tumor microenvironment, leading to poor prognosis. These findings provide a reference for the exploration of FGF11 as a prognostic biomarker and treatment target in CC.
Collapse
Affiliation(s)
- Xinyi Nie
- Hebei North University, Zhangjiakou, China
| | - Ziyan Zhu
- The First Hospital Affiliated to Hebei North University, Zhangjiakou, China
| | | | | | - Jiangping Chen
- The First Hospital Affiliated to Hebei North University, Zhangjiakou, China
| | - Fan Zhang
- The First Hospital Affiliated to Hebei North University, Zhangjiakou, China
| | - Bowei Guo
- The First Hospital Affiliated to Hebei North University, Zhangjiakou, China
| |
Collapse
|