1
|
Zhang J, Huang X, Li M, Zhang W, Yang H. CSF1R inhibition agents protect against cisplatin ototoxicity and synergize with immunotherapy for Head and Neck Squamous Cell Carcinoma. Int Immunopharmacol 2025; 152:114428. [PMID: 40073814 DOI: 10.1016/j.intimp.2025.114428] [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: 10/18/2024] [Revised: 01/25/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
Immunotherapy has emerged as a promising therapeutic approach. However, limited research exists on combining cisplatin with CSF1/CSF1R immunotherapy in Head and Neck Squamous Cell Carcinoma. Furthermore, few studies have investigated concurrent immunotherapeutic strategies to mitigate cisplatin-induced ototoxicity.Developing otoprotective agents that simultaneously reduce cisplatin resistance and enhance therapeutic efficacy holds significant implications for future treatment modalities. In this investigation, we evaluated the safety and efficacy profile of CSF1R inhibitor (PLX3397). Our findings demonstrate that PLX3397 confers otoprotection in cisplatin-induced ototoxicity through cochlear macrophage depletion, synergizes with cisplatin inhibited tumor cell survival, migration, and invasion in vitro. Additionally, it significantly suppressed xenograft tumor lesion growth and angiogenesis in zebrafish models while modulating the polarization state of tumor-associated macrophages in vitro and inducing tumor immune activation. Our findings suggest that PLX3397 represents a promising immunotherapeutic agent, and its combination with cisplatin may constitute a novel therapeutic strategy for attenuating cisplatin-induced ototoxicity while synergistically enhancing immunotherapy for Head and Neck Squamous Cell Carcinoma.
Collapse
Affiliation(s)
- Jiahui Zhang
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xiaotong Huang
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Moyang Li
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Weijian Zhang
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Haidi Yang
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| |
Collapse
|
2
|
Wang L, ChenLiu Z, Wang D, Tang D. Cross-talks of GSH, mitochondria, RNA m6A modification, NRF2, and p53 between ferroptosis and cuproptosis in HCC: A review. Int J Biol Macromol 2025; 302:140523. [PMID: 39894098 DOI: 10.1016/j.ijbiomac.2025.140523] [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: 11/11/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumor with high morbidity and mortality, as well as poor prognosis. Therefore, it is imperative to explore alternative therapeutic targets for HCC treatment. Ferroptosis and cuproptosis have recently been identified as metal-dependent cell death mechanisms that play significant roles in HCC treatment. This study identified potential cross-talk between ferroptosis and cuproptosis, including the common hub glutathione, common site of occurrence, mitochondria, shared epigenetic modification mode, RNA N6 methyladenosine modification, mutual inhibitor, nuclear factor erythroid 2-related factor 2, and dual regulator, p53. These findings provide a theoretical foundation for the joint induction of HCC cell death and effective inhibition of HCC progression. However, some immune cells are susceptible to ferroptosis or cuproptosis, which may impair or enhance anti-cancer immune function. We propose strategies to target specific targets molecules such as tripartite motif containing 25, ferroptosis suppressor protein 1, and peroxisome proliferator-activated receptor gamma or exploit the unique acidic environment surrounding cancer cells to precisely induce ferroptosis in cancer cells. This approach aims to advance the development of precision medicine for HCC treatment.
Collapse
Affiliation(s)
- Leihan Wang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, People's Republic of China
| | - Zhenni ChenLiu
- Clinical Medical College, Yangzhou University, Yangzhou 225000, People's Republic of China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, The Yangzhou Clinical Medical College of Xuzhou Medical University, The Yangzhou School of Clinical Medicine of Dalian Medical University, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou 225000, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, The Yangzhou Clinical Medical College of Xuzhou Medical University, The Yangzhou School of Clinical Medicine of Dalian Medical University, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou 225000, China.
| |
Collapse
|
3
|
Liu K, Yu J, Huang X, Gao H, Wang J. WGCNA and ferroptosis genes in OSCC: unraveling prognostic biomarkers and therapeutic targets. Discov Oncol 2025; 16:379. [PMID: 40126728 PMCID: PMC11933588 DOI: 10.1007/s12672-025-02151-9] [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: 12/28/2024] [Accepted: 03/14/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is the predominant type of oral cancer, with over 370,000 new cases and approximately 170,000 deaths annually worldwide. Despite therapeutic advancements, OSCC mortality rates have been increasing, underscoring the need for improved prognostic models and therapeutic targets. METHODS We integrated transcriptomic and clinical survival data from the TCGA-OSCC dataset to identify ferroptosis-related prognostic genes. Using weighted gene co-expression network analysis (WGCNA), we selected genes associated with OSCC prognosis and applied Lasso regression analysis to pinpoint key genes. A prognostic model was constructed and validated through survival analysis and receiver operating characteristic (ROC) curve analysis. RESULTS WGCNA identified modules significantly correlated with ferroptosis, yielding 321 genes associated with OSCC prognosis. Univariate Cox analysis identified 13 genes affecting OSCC prognosis. Lasso regression and multivariate Cox regression narrowed down the gene set to a final set of 7 genes, which were used to construct the risk model. The model stratified patients into high- and low-risk groups with significant survival differences (P < 0.001). The model's predictive accuracy was validated, with AUC values ranging from 0.565 to 0.733 for 1-, 3-, and 5-year survival predictions. Immune-related analysis revealed that low-risk patients exhibited higher immune cell infiltration and were more likely to benefit from immunotherapy. CONCLUSION Our study presents a novel prognostic model for OSCC patients based on ferroptosis-related genes, which not only predicts survival but also identifies potential therapeutic targets. The model's predictive accuracy and clinical relevance were validated, offering a new strategy for OSCC treatment.
Collapse
Affiliation(s)
- Ke Liu
- Department of Stomatology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Jiannan Yu
- Department of Pediatric Dental Prevention, The Afiliated Stomatological Hospial of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Xuanxi Huang
- The Ninth Outpatient Department, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hongyan Gao
- Department of Health Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jing Wang
- Department of Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
4
|
Gao X, Liu S, Cao Y, Shi L, Yin Y. The controversial role of CD151 in different solid tumors: promoter or suppressor? Cancer Cell Int 2025; 25:110. [PMID: 40119419 PMCID: PMC11927130 DOI: 10.1186/s12935-025-03751-9] [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: 01/13/2025] [Accepted: 03/13/2025] [Indexed: 03/24/2025] Open
Abstract
As a member of the tetraspanin superfamily, CD151 plays a pivotal role in tumorigenesis, progression, and metastasis. CD151 is involved in various cellular processes, including cell-cell junction, signal transduction, epithelial-mesenchymal transition (EMT), cancer stem cell maintenance (CSCs), angiogenesis, and exosome regulation. Therefore, CD151 is a potential target for tumor therapy and may be valuable in tumor diagnosis and prognosis. Notably, while CD151 predominantly functions as an oncogene in most cancers, it can also exhibit tumor-suppressive roles in specific contexts, indicating that its function is context-dependent. Additionally, CD151 plays a significant role in modulating the immune microenvironment. For instance, CD151 supports the proliferation, activation, and migration of T cells. The CD151 peptide may function as a tumor vaccination by stimulating CD8 + IFNγ + lymphocytes and inducing cytotoxic effects. Thus, the function of CD151 in tumors is intricate and warrants further investigation. In this review, we discuss the diagnostic and prognostic potential of CD151, as well as its regulatory roles in solid cancers, including those of the digestive system, lung, breast, prostate, and gynecological tissues. Basic experiments and clinical data demonstrate the beneficial and detrimental effects of CD151 in malignancies and offer a path forward for future investigation.
Collapse
Affiliation(s)
- Xue Gao
- Medical Oncology, Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Sa Liu
- Medical Oncology, Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yubo Cao
- Medical Oncology, Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lei Shi
- The Shengjing hospital of China Medical University, Shenyang, Liaoning, China.
| | - Yuanqin Yin
- Cancer Institute, First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
5
|
Li H, Hu X, Wang L, Gu X, Chen S, Tang Y, Chen Y, Chen J, Yuan Z, Wang Y. The Expression of Ferroptosis-Related Genes in Hepatocellular Carcinoma and Their Relationships With Prognosis. J Hepatocell Carcinoma 2025; 12:629-648. [PMID: 40130081 PMCID: PMC11932120 DOI: 10.2147/jhc.s500394] [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: 10/11/2024] [Accepted: 02/05/2025] [Indexed: 03/26/2025] Open
Abstract
Background Ferroptosis, a form of cell death discovered in recent years, is expected to provide new targets for the diagnosis and treatment of hepatocellular carcinoma (HCC) through further research. Methods Based on data from The Cancer Genome Atlas (TCGA), we screened HCC-associated genes from 259 candidate genes in the FerrDb database. The screened genes were subjected to differential expression analysis, survival analysis, correlation analysis with clinical data, and univariate and multivariate Cox regression analysis. The results were validated with the Gene Expression Profiling Interactive Analysis 2 (GEPIA2) database and the Human Protein Atlas (HPA) database, and signaling pathways were analyzed with the Gene Set Enrichment Analysis (GSEA) enrichment analysis. Human normal hepatocytes and different liver cancer cell lines were used to verify the expression levels of genes, using quantitative reverse transcription PCR (RT-qPCR). Results Eight ferroptosis-related genes were finally selected, including ACSL3, ASNS, CHMP5, MYB, PCK2, PGD, SLC38A1, and YY1AP1. The expression of eight genes except PCK2 was significantly correlated with a lower survival rate of HCC, and the expression of PCK2 showed a correlation with a higher survival rate of HCC. The expression of all eight genes was also correlated with clinical traits. GSEA enrichment analysis obtained many pathways such as apoptosis, endocytosis, pathways in cancer, Wnt signaling pathway, primary bile acid biosynthesis, and fatty acid metabolism pathway. Conclusion The ACSL3, ASNS, CHMP5, MYB, PCK2, PGD, SLC38A1, and YY1AP1 genes may become markers and new targets for early diagnosis and prognostic assessment of HCC.
Collapse
Affiliation(s)
- Hongxu Li
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, People’s Republic of China
| | - Xinyue Hu
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, People’s Republic of China
| | - Li Wang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Xiangran Gu
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, People’s Republic of China
| | - Shibin Chen
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, People’s Republic of China
| | - Yixuan Tang
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, People’s Republic of China
| | - Yuan Chen
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jin Chen
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Zhengrong Yuan
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, People’s Republic of China
| | - Yajie Wang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
6
|
Luo Y, Yuan Y, Liu D, Peng H, Shen L, Chen Y. Targeting novel immune checkpoints in the B7-H family: advancing cancer immunotherapy from bench to bedside. Trends Cancer 2025:S2405-8033(25)00055-X. [PMID: 40113530 DOI: 10.1016/j.trecan.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025]
Abstract
The B7-H family of immune checkpoint molecules is a crucial component of the immune regulatory network for tumors, offering new opportunities to modulate the tumor microenvironment (TME). The B7-H family - which includes B7-H2 (inducible T cell costimulatory ligand, ICOSL), B7-H3, B7-H4, B7-H5 (V-domain immunoglobulin suppressor of T cell activation, VISTA), B7-H6, and B7-H7 (HHLA2) - is known for its diverse roles in regulating innate and adaptive immunity. These molecules can exhibit co-stimulatory or co-inhibitory effects on T cells, influencing processes such as T cell activation, differentiation, and effector functions, and they are involved in the recruitment and polarization of various immune cells. This review explores the structural characteristics, receptor-ligand interactions, and signaling pathways associated with each B7-H family member. We also discuss the family's impact on tumor immunity and potential therapeutic strategies.
Collapse
Affiliation(s)
- Yiming Luo
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Ye Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Dan Liu
- Early Drug Development Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Haoxin Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China.
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China; Department of Gastrointestinal Cancer, Beijing GoBroad Hospital, Beijing 102200, China.
| |
Collapse
|
7
|
Gao H, Zhang T, Li K, Li X. CD73: a new immune checkpoint for leukemia treatment. Front Immunol 2025; 16:1486868. [PMID: 40114928 PMCID: PMC11922907 DOI: 10.3389/fimmu.2025.1486868] [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: 08/27/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
Recent studies on the pathogenesis of leukemia have led to remarkable advances in disease treatment. Numerous studies have shown the potential and viability of immune responses against leukemia. In the classical pathway, this process is often initiated by the upstream activity of CD39, which hydrolyzes extracellular adenosine triphosphate (ATP) and adenosine diphosphate (ADP) to AMP. Subsequently, CD73 acts on AMP to generate adenosine, contributing to an immunosuppressive microenvironment. However, CD73 can also utilize substrates derived from other molecules through the non-canonical NAD+ pathway, specifically via the CD38/CD203a/CD73 axis, further enhancing adenosine production and facilitating immune escape. Targeting CD73 has shown potential in disrupting these immunosuppressive pathways, thereby enhancing anti-leukemic immune responses and improving patient outcomes. Inhibiting CD73 not only reduces the levels of immunosuppressive adenosine but also increases the efficacy of existing immunotherapies, such as PD-1/PD-L1 inhibitors, making it a versatile therapeutic target in leukemia treatment. This review discusses the potential of CD73 as a therapeutic target and emphasizes its unique position in the immune escape mechanism of leukemia. Moreover, this review provides an overview of the current research progress and future trends, emphasizing the clinical significance of targeting CD73 and other potential therapeutic strategies in leukemia.
Collapse
Affiliation(s)
- Huan Gao
- Marine College, Shandong University, Weihai, China
| | - Tingting Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ke Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xia Li
- Marine College, Shandong University, Weihai, China
| |
Collapse
|
8
|
Xiong Y, Lei J, Wen M, Ma Y, Zhao J, Tian Y, Wan Z, Li X, Zhu J, Wang W, Ji X, Sun Y, Yang J, Zhang J, Xin S, Liu Y, Jia L, Han Y, Jiang T. CENPF (+) cancer cells promote malignant progression of early-stage TP53 mutant lung adenocarcinoma. Oncogenesis 2025; 14:5. [PMID: 40044674 PMCID: PMC11882812 DOI: 10.1038/s41389-025-00546-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/18/2024] [Accepted: 01/23/2025] [Indexed: 03/09/2025] Open
Abstract
The prevention and precise treatment of early-stage lung adenocarcinoma (LUAD) characterized by small nodules (stage IA) remains a significant challenge for clinicians, which is due largely to the limited understanding of the oncogenic mechanisms spanning from preneoplasia to invasive adenocarcinoma. Our study highlights the pivotal role of cancer cells exhibiting high expression of centromere protein F (CENPF), driven by TP53 mutations, which become increasingly prevalent during the transition from preneoplasia to invasive LUAD. Biologically, cancer cells (CENPF+) exhibited robust proliferative and stem-like capabilities, thereby propelling the malignant progression of early-stage LUAD. Clinically, autoantibodies against CENPF in the serum and elevated cancer cells (CENPF+) in tissue correlated positively with the progression of early-stage LUAD, especially those in stage IA. Our findings suggest that cancer cells (CENPF+) play a central role in orchestrating the malignant evolution of LUAD and hold potential as a novel biomarker for early-stage detection and management of the disease.
Collapse
Affiliation(s)
- Yanlu Xiong
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
- Innovation Center for Advanced Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
- Department of Thoracic Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jie Lei
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Miaomiao Wen
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yongfu Ma
- Department of Thoracic Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jinbo Zhao
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yahui Tian
- Department of Thoracic Surgery, Air Force Medical Center, PLA, Beijing, China
| | - Zitong Wan
- College of Life Sciences, Northwestern University, Xi'an, China
| | - Xiaoyan Li
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Jianfei Zhu
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Wenchen Wang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaohong Ji
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Ying Sun
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jie Yang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jiao Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Shaowei Xin
- Department of Thoracic Surgery, Air Force Medical Center, PLA, Beijing, China
| | - Yang Liu
- Department of Thoracic Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Lintao Jia
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, China
| | - Yong Han
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
- Department of Thoracic Surgery, Air Force Medical Center, PLA, Beijing, China.
| | - Tao Jiang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
9
|
Nie XH, Li TZ, Peng CM. ATP ion channel-type P2X purinergic receptors as a key regulatory molecule in breast cancer progression. Pathol Res Pract 2025; 267:155844. [PMID: 39965402 DOI: 10.1016/j.prp.2025.155844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/31/2025] [Accepted: 02/13/2025] [Indexed: 02/20/2025]
Abstract
Studies have confirmed that ATP ion channel P2X purinergic receptors play a key role in tumor growth and metastasis. Similarly, P2X purinergic receptors can be used as a favorable regulatory molecule of breast cancer cells to participate in the progression of breast cancer. There are abundant ATP and its cleavage products in breast cancer microenvironment, which can be used as natural activators of P2X purinergic receptors. P2X purinergic receptors play a role in regulating the growth and metastasis of breast cancer cells by mediating signal transduction, growth regulation and immune cell activity in microenvironment. However, the application of P2X purinergic receptors antagonist has the pharmacological characteristics of inhibiting the progression of breast cancer cells. Among P2X purinergic receptors, there is a close relationship between P2X7 receptor and breast cancer patients. P2X purinergic receptors can be used as a biological marker for breast cancer patients. In this paper, we discuss the functional role and regulatory molecular mechanism of P2X purinergic receptors in the progression of breast cancer. The pharmacological effects of P2X purinergic receptors selective antagonist on the growth, metastasis and invasion of breast cancer cells were further discussed. Therefore, P2X purinergic receptors can be used as a key regulatory molecule of breast cancer and a pharmacological target for potential therapy.
Collapse
Affiliation(s)
- Xin-Hua Nie
- Department of Gastroenterology, The second affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Teng-Zheng Li
- Department of Gastroenterology, The second affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Cheng-Ming Peng
- Department of Gastroenterology, The second affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China.
| |
Collapse
|
10
|
Putatunda V, Jusakul A, Roberts L, Wang XW. Genetic, Epigenetic, and Microenvironmental Drivers of Cholangiocarcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:362-377. [PMID: 39532242 PMCID: PMC11841490 DOI: 10.1016/j.ajpath.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
Cholangiocarcinoma (CCA) is an aggressive and heterogeneous malignancy of the biliary tree that carries a poor prognosis. Multiple features at the genetic, epigenetic, and microenvironmental levels have been identified to better characterize CCA carcinogenesis. Genetic alterations, such as mutations in IDH1/2, BAP1, ARID1A, and FGFR2, play significant roles in CCA pathogenesis, with variations across different subtypes, races/ethnicities, and causes. Epigenetic dysregulation, characterized by DNA methylation and histone modifications, further contributes to the complexity of CCA, influencing gene expression and tumor behavior. Furthermore, CCA cells exchange autocrine and paracrine signals with other cancer cells and the infiltrating cell types that populate the microenvironment, including cancer-associated fibroblasts and tumor-associated macrophages, further contributing to an immunosuppressive niche that supports tumorigenesis. This review explores the multifaceted genetic, epigenetic, and microenvironmental drivers of CCA. Understanding these diverse mechanisms is essential for characterizing the complex pathways of CCA carcinogenesis and developing targeted therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Vijay Putatunda
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland; Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| | - Apinya Jusakul
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand; Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Lewis Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Xin Wei Wang
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland; Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
11
|
Cheng B, Chen J, Katalina V, Long G, Wei C, Niu Z, Chen C, Wang P, Yu Q, Wang W. Targeting PKC as a Therapeutic Strategy to Overcome Chemoresistance in TNBC by Restoring Aurora Kinase B Expression. J Cell Mol Med 2025; 29:e70464. [PMID: 40099930 PMCID: PMC11915661 DOI: 10.1111/jcmm.70464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 12/18/2024] [Accepted: 02/19/2025] [Indexed: 03/20/2025] Open
Abstract
Triple-negative breast cancer (TNBC) poses a significant challenge due to its high mortality rates, primarily attributed to resistance against chemotherapy regimens containing taxanes like paclitaxel. Thus, developing combinatorial strategies to override resistance is a pressing need. By taking advantage of a library screening with various kinase inhibitors, we found that the small-molecule inhibitor enzastaurin targeting protein kinase C (PKC) could overcome resistance in TNBC cells. Mechanistically, dual treatment with paclitaxel and enzastaurin resulted in efficient mitotic arrest and subsequent cell death by restoring AURKB expression. Further analysis revealed that the GCN2-p-eIF2α axis was responsible for the posttranscriptional accumulation of AURKB upon combinatorial treatment. Finally, we confirmed that combinatorial regimens synergistically suppressed tumour growth in vivo in mouse models. Moreover, the efficiency of dual treatment was largely determined by AURKB, implying that AURKB could be a potential predictive marker for stratifying patients who may benefit from the combinatorial treatment. Collectively, our study not only unravels a novel underlying mechanism for paclitaxel resistance in TNBC but also provides a new potential combinatorial therapeutic strategy in the clinic.
Collapse
Affiliation(s)
- Bing Cheng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jinxin Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Vera Katalina
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Guojie Long
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of General Surgery (Department of Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chaoying Wei
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhitong Niu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chen Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Panpan Wang
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Qiang Yu
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer and Stem Cell Biology, DUKE-NUS Medical School, Singapore, Singapore
- Tianfu Jincheng Laboratory, Chengdu, China
| | - Wenyu Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
12
|
Li J, Zhang M, Sun Q, Li X, Du F, Cheng Y, Li S, Zhang J. CENPF interaction with PLA2G4A promotes glioma growth by modulating mTORC1 and NF-κB pathways. Cancer Cell Int 2025; 25:73. [PMID: 40025532 PMCID: PMC11871623 DOI: 10.1186/s12935-025-03700-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 02/17/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Glioma is the most common primary malignant tumor of the central nervous system, and due to the limited effectiveness of traditional single-target therapies, there is an urgent need for new therapeutic targets. Centromere protein F (CENPF) belongs to the centromere protein family and is mainly involved in the regulation of the cell cycle. CENPF has recently been found to play a key role in tumorigenesis and tumor progression, but its role in gliomas has not been well studied. METHODS The expression level and clinical information of CENPF were obtained by analyzing the TCGA, CGGA and GEO databases. Immunohistochemistry and western blot analysis were used to quantitatively detect the expression of CENPF in glioma tissues and cell lines. Gene set enrichment analysis (GSEA) of TCGA and GSE16011 datasets was used to explore the molecular mechanism of the CENPF. CENPF-interacting proteins were detected by molecular docking and co-immunoprecipitation (Co-IP). After silencing CENPF, CCK-8 assay, Transwell assay and flow cytometry were used to detect changes in cell proliferation, invasion, cell cycle and apoptosis, and Western blot was used to detect changes in signaling pathway protein levels. RESULTS Bioinformatics analysis showed that CENPF was generally highly expressed in gliomas and was associated with poor prognosis. This result was confirmed in glioma samples from our hospital. Multivariate Cox regression analysis showed that CENPF was an independent prognostic marker for gliomas. Western blot analysis in vitro showed that CENPF was overexpressed in the U251 and LN229 cell lines; therefore, these two cell lines were selected for subsequent experiments. GSEA analysis showed that CENPF was mainly involved in the G2/M phase-mediated cell cycle and P53 signaling pathway. Flow cytometry analysis confirmed that silencing CENPF induced G2/M phase arrest and increased apoptosis in glioma cells. Subsequent experiments confirmed that CENPF influences the epithelial-mesenchymal transition (EMT) process through the mTORC1 signaling pathway. Molecular docking and Co-IP assay revealed that CENPF exerts its effects by interacting with PLA2G4A promoting the downstream signaling pathway. Finally, we found that silencing CENPF combined with a PLA2G4A inhibitor (AACOCF3) induced glioma cell apoptosis and exhibited anti-glioma effects. CONCLUSIONS This study found that CENPF plays a key role in promoting tumorigenesis through its interaction with PLA2G4A. This study provides a theoretical foundation for advancing multi-targeted therapies in glioma and for developing strategies to overcome tumor drug resistance.
Collapse
Affiliation(s)
- Junhong Li
- Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province, 276000, China
| | - Moxuan Zhang
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
| | - Qiang Sun
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
| | - Xinglan Li
- Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province, 276000, China
| | - Fei Du
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
| | - Yanhao Cheng
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
| | - Shuzhi Li
- Department of Neurosurgery, Gaomi Traditional Chinese Medicine Hospital, Weifang, Shandong Province, 261500, China.
| | - Jian Zhang
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong Province, 276000, China.
| |
Collapse
|
13
|
Guo D, Cai S, Deng L, Xu W, Fu S, Lin Y, Jiang T, Li Q, Shen Z, Zhang J, Luo P, Tang B, Wang L. Ferroptosis in Pulmonary Disease and Lung Cancer: Molecular Mechanisms, Crosstalk Regulation, and Therapeutic Strategies. MedComm (Beijing) 2025; 6:e70116. [PMID: 39991627 PMCID: PMC11847630 DOI: 10.1002/mco2.70116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 01/12/2025] [Accepted: 01/15/2025] [Indexed: 02/25/2025] Open
Abstract
Ferroptosis is a distinct form of iron-dependent programmed cell death characterized primarily by intracellular iron accumulation and lipid peroxidation. Multiple cellular processes, including amino acid metabolism, iron metabolism, lipid metabolism, various signaling pathways, and autophagy, have been demonstrated to influence the induction and progression of ferroptosis. Recent investigations have elucidated that ferroptosis plays a crucial role in the pathogenesis of various pulmonary disorders, including lung injury, chronic obstructive pulmonary disease, pulmonary fibrosis, and asthma. Ferroptosis is increasingly recognized as a promising novel strategy for cancer treatment. Various immune cells within the tumor microenvironment, including CD8+ T cells, macrophages, regulatory T cells, natural killer cells, and dendritic cells, have been shown to induce ferroptosis in tumor cells and modulate the process through the regulation of iron and lipid metabolism pathways. Conversely, ferroptosis can reciprocally alter the metabolic environment, leading to the activation or inhibition of immune cell functions, thereby modulating immune responses. This paper reviews the molecular mechanism of ferroptosis and describes the tumor immune microenvironment, discusses the connection between ferroptosis and the tumor microenvironment in lung cancer and pulmonary diseases, and discusses the development prospect of their interaction in the treatment of lung cancer and pulmonary diseases.
Collapse
Affiliation(s)
- Dandan Guo
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Songhua Cai
- Department of Thoracic SurgeryNational Cancer CenterNational Clinical Research Center for CancerCancer Hospital & Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenGuangdongChina
| | - Lvdan Deng
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Wangting Xu
- Department of RespiratoryFirst Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Sentao Fu
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Yaling Lin
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Tong Jiang
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Qing Li
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Zhijun Shen
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Jian Zhang
- The Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Peng Luo
- The Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Bufu Tang
- Department of Radiation OncologyZhongshan HospitalFudan UniversityShanghaiShanghaiChina
| | - Ling Wang
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| |
Collapse
|
14
|
Zhang Q, Zhang Y, Guo S, Wang H. Emerging insights into the role of microRNAs regulation of ferroptosis in hepatocellular carcinoma. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167642. [PMID: 39734007 DOI: 10.1016/j.bbadis.2024.167642] [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: 09/08/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Hepatocellular carcinoma (HCC) is a major type of liver cancer and an important cause of cancer death. It has been reported that the hepatocyte death plays an important role in HCC. Ferroptosis is an iron-dependent programmed cell death characterized by the accumulation of free iron and lipid peroxidation. A series of studies have shown that ferroptosis contributes to the occurrence and development of HCC. MicroRNAs (miRNAs) are non-coding RNAs with a length of approximately 222 nt. In recent years, miRNAs have been shown to participate in regulating ferroptosis to play a vital role in HCC, but the related mechanisms are not fully understood. This review summarized the current understanding of ferroptosis, as well as the biogenesis and function of miRNAs, and focused on the role of miRNAs regulation of ferroptosis in HCC, with the hope of providing new targets and ideas for the treatment of HCC.
Collapse
Affiliation(s)
- Qi Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yingdan Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Shiyun Guo
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Honggang Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
15
|
Fang Z, Shao Y, Hu M, Yan J, Ye G. Biological roles and molecular mechanism of circular RNAs in epithelial-mesenchymal transition of gastrointestinal malignancies. Oncol Res 2025; 33:549-566. [PMID: 40109856 PMCID: PMC11915071 DOI: 10.32604/or.2024.051589] [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: 03/09/2024] [Accepted: 06/13/2024] [Indexed: 03/22/2025] Open
Abstract
Circular RNAs (circRNAs) are formed by splicing of precursor RNAs and covalently linked at the 5' and 3' ends. Dysregulated circRNAs are closely related to the epithelial-mesenchymal transition (EMT) of gastrointestinal malignancies. CircRNAs, including circRNA_0008717, circGOT1, circ-DOCK5, circVPS33B, circPVT1, circMET, circ-OXCT1, circ_67835, circRTN4, circ_0087502, circFNDC38, circ_PTEN1, circPGPEP1, and circ-E-Cad are involved in the EMT process of gastrointestinal malignancies through a variety of mechanisms, such as regulating EMT-inducing transcription factors, signaling pathways, and tumor microenvironments. Gastrointestinal (GI) malignancies are common malignant tumors worldwide, and the heterogeneity and easy metastasis of gastrointestinal malignancies limit the effectiveness of medical treatments. Therefore, investigating the molecular mechanisms involved in the pathogenesis of gastrointestinal malignancies is essential for clinical treatment. This article summarizes the biological roles and molecular mechanism of circRNAs in EMT of gastrointestinal malignancies, providing a theoretical basis for applying EMT-related circRNAs in targeted therapy.
Collapse
Affiliation(s)
- Ziyi Fang
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315020, China
| | - Yongfu Shao
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315020, China
| | - Meng Hu
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Jianing Yan
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315020, China
| | - Guoliang Ye
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315020, China
| |
Collapse
|
16
|
Ge Y, Jiang L, Dong Q, Xu Y, Yam JWP, Zhong X. Exosome-mediated Crosstalk in the Tumor Immune Microenvironment: Critical Drivers of Hepatocellular Carcinoma Progression. J Clin Transl Hepatol 2025; 13:143-161. [PMID: 39917466 PMCID: PMC11797817 DOI: 10.14218/jcth.2024.00302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 02/09/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a significant global health issue, ranking as the sixth most prevalent malignancy and the fourth leading cause of cancer-related mortality worldwide. Despite advancements in therapeutic strategies, mortality rates for HCC remain high. The tumor immune microenvironment (TIME) plays a vital role in HCC progression by influencing tumor cell survival and growth. Recent studies highlight the essential role of exosomes in mediating intercellular communication within the TIME, particularly in interactions among tumor cells, immune cells, and fibroblasts. These interactions drive critical aspects of tumor development, including immune escape, angiogenesis, drug resistance, and metastasis. A detailed understanding of the molecular mechanisms by which exosomes modulate the TIME is essential for developing targeted therapies. This review systematically evaluated the roles and regulatory mechanisms of exosomes within the TIME of HCC, examining the impact of both HCC-derived and non-HCC-derived exosomes on various cellular components within the TIME. It emphasized their regulatory effects on cell phenotypes and functions, as well as their roles in HCC progression. The review also explored the potential applications of exosome-based immunotherapies, offering new insights into improving therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Yifei Ge
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lixue Jiang
- Department of Breast Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Qingfu Dong
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xiangyu Zhong
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
17
|
Zhang K, Guo L, Li X, Hu Y, Luo N. Cancer-associated fibroblasts promote doxorubicin resistance in triple-negative breast cancer through enhancing ZFP64 histone lactylation to regulate ferroptosis. J Transl Med 2025; 23:247. [PMID: 40022222 PMCID: PMC11871786 DOI: 10.1186/s12967-025-06246-3] [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: 09/28/2024] [Accepted: 02/11/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) have been identified to drive chemotherapy resistance in triple-negative breast cancer (TNBC). This study evaluated the functions of CAFs-mediated suppressive ferroptosis in doxorubicin (DOX) resistance in TNBC and its detailed molecular mechanisms. METHODS TNBC cell lines were co-cultured with CAFs isolated from DOX-sensitive (CAF/S) or DOX-resistant (CAF/R) breast cancer tissues. Cell viability and death were assessed by cell counting Kit-8 (CCK-8) and propidium iodide (PI) staining. Ferroptosis was evaluated by detection of Fe2+, malondialdehyde (MDA), glutathione (GSH), and lipid reactive oxygen species (ROS) levels. Histone lactylation was determined by lactate production, pan-Kla and H3K18la expression. Molecular mechanism was determined by chromatin immunoprecipitation (ChIP) and dual luciferase reporter system. Molecule and protein expression was detected by quantitative Real-Time PCR (RT-qPCR), Western blotting, immunofluorescence and immunohistochemical staining. TNBC cells were injected into the mammary fat pad of nude mice to investigate DOX sensitivity in vivo. RESULTS CAFs-derived lactate repressed ferroptosis to confer resistance of TNBC cells to DOX. Moreover, zinc finger protein 64 (ZFP64) expression was elevated in DOX-resistant TNBC and was associated with high histone lactylation level. CAFs facilitated histone lactylation to enhance ZFP64 expression, which triggered ferroptosis inhibition and DOX resistance. In addition, ZFP64 bound to the promoters of GTP cyclohydrolase-1 (GCH1) and ferritin heavy chain 1 (FTH1), thereby promoting their expression. Rescue experiments indicated that ZFP64 silencing-induced ferroptosis and high sensitivity of TNBC cells to DOX could be counteracted by GCH1 or FTH1 overexpression. CONCLUSION CAFs acted as a ferroptosis inhibitor to cause DOX resistance of TNBC via histone lactylation-mediated ZFP64 up-regulation and subsequent promotion of GCH1-induced lipid peroxidation inhibition and FTH1-induced intracellular Fe2+ consumption.
Collapse
Affiliation(s)
- KeJing Zhang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, P.R. China
- Clinical Research Center For Breast Cancer Control and Prevention In Hunan Province, Changsha, Hunan Province, 410008, P.R. China
| | - Lei Guo
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, P.R. China
- Clinical Research Center For Breast Cancer Control and Prevention In Hunan Province, Changsha, Hunan Province, 410008, P.R. China
| | - Xin Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, P.R. China
- Clinical Research Center For Breast Cancer Control and Prevention In Hunan Province, Changsha, Hunan Province, 410008, P.R. China
| | - Yu Hu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, P.R. China
- Clinical Research Center For Breast Cancer Control and Prevention In Hunan Province, Changsha, Hunan Province, 410008, P.R. China
| | - Na Luo
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, P.R. China.
- Clinical Research Center For Breast Cancer Control and Prevention In Hunan Province, Changsha, Hunan Province, 410008, P.R. China.
- Department of General Surgery, Xiangya Hospital, Central South University & Clinical Research Center For Breast Cancer Control and Prevention In Hunan Province, No. 87, Xiangya Road, Changsha, Hunan Province, 410008, P.R. China.
| |
Collapse
|
18
|
Wang R, Li W, Cao H, Zhang L. Decoding the Tumor-Associated Microbiota: From Origins to Nanomedicine Applications in Cancer Therapy. BIOLOGY 2025; 14:243. [PMID: 40136500 PMCID: PMC11940167 DOI: 10.3390/biology14030243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
Growing evidence reveals that the tumor microbiome-comprising distinct microbial communities within neoplastic tissues-exerts a profound influence on cancer initiation, progression, and therapeutic response. These microbes actively reshape the tumor microenvironment (TME) through metabolite secretion, the modulation of immune pathways, and direct interactions with host cells, thereby affecting tumor biology and therapeutic outcomes. Despite substantial heterogeneity among cancer types, recent insights underscore the tumor microbiome's potential as both a diagnostic/prognostic biomarker and a targetable component for innovative treatments. In this review, we synthesize emerging knowledge on the mechanistic roles of tumor-associated microbiota in shaping the TME, with a focus on how these discoveries can guide novel therapeutic strategies. We further explore interdisciplinary advances, including the convergence of microbiomics and nanotechnology, to enhance drug delivery, circumvent resistance, and foster TME remodeling. By highlighting these cutting-edge developments, our review underscores the transformative potential of integrating tumor microbiome research into precision oncology and advancing more personalized cancer therapies.
Collapse
Affiliation(s)
- Ruiqi Wang
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (R.W.); (W.L.)
| | - Weizheng Li
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (R.W.); (W.L.)
| | - Hongqian Cao
- Department of Health Inspection and Quarantine, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lei Zhang
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (R.W.); (W.L.)
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| |
Collapse
|
19
|
Easwaran VB, Pai KMS, Pai KSR. Mesenchymal Stem Cell-Derived Exosomes in Cancer Resistance Against Therapeutics. Cancers (Basel) 2025; 17:831. [PMID: 40075675 PMCID: PMC11898417 DOI: 10.3390/cancers17050831] [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: 01/20/2025] [Revised: 02/10/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are specialized cells that can differentiate into various types of cells. MSCs can be utilized to treat cancer. However, a MSC is considered a double-edged sword, because it can promote tumor progression and support cancer cell growth. Likewise, MSC-derived exosomes (MSC-Exos) carry various intracellular materials and transfer them to other cells. MSC-Exos could also cause tumor progression, including brain cancer, breast cancer, hepatic cancer, lung cancer, and colorectal cancer, and develop resistance against therapies, mainly chemotherapy, radiotherapy, and immunotherapy. An MSC-Exo promotes tumor development and causes drug resistance in various cancer types. The mechanisms involved in cancer drug resistance vary depending on the cancer cell heterogeneity and complexity. In this article, we have explained the various biomarkers and mechanisms involved in the tumor and resistance development through MSC-Exos in different cancer types.
Collapse
Affiliation(s)
- Vignesh Balaji Easwaran
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India;
| | - K Maya S Pai
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India;
| | - K. Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India;
| |
Collapse
|
20
|
Zhang YZ, Ma Y, Ma E, Chen X, Zhang Y, Yin B, Zhao J. Sophisticated roles of tumor microenvironment in resistance to immune checkpoint blockade therapy in hepatocellular carcinoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:10. [PMID: 40051497 PMCID: PMC11883234 DOI: 10.20517/cdr.2024.165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/13/2025] [Accepted: 02/21/2025] [Indexed: 03/09/2025]
Abstract
Hepatocellular carcinoma (HCC) remains a serious threat to global health, with rising incidence and mortality rates. Therapeutic options for advanced HCC are quite limited, and the overall prognosis remains poor. Recent advancements in immunotherapy, particularly immune-checkpoint blockade (ICB) targeting anti-PD1/PD-L1 and anti-CTLA4, have facilitated a paradigm shift in cancer treatment, demonstrating substantial survival benefits across various cancer types, including HCC. However, only a subset of HCC patients exhibit a favorable response to ICB therapy, and its efficacy is often hindered by the development of resistance. There are many studies to explore the underlying mechanisms of ICB response. In this review, we compiled the latest progression in immunotherapies for HCC and systematically summarized the sophisticated mechanisms by which components of the tumor microenvironment (TME) regulate resistance to ICB therapy. Additionally, we also outlined some scientific rationale strategies to boost antitumor immunity and enhance the efficacy of ICB in HCC. These insights may serve as a roadmap for future research and help improve outcomes for HCC patients.
Collapse
Affiliation(s)
- Yi-Zhe Zhang
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Authors contributed equally
| | - Yunshu Ma
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Authors contributed equally
| | - Ensi Ma
- Liver Transplantation Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Institute of Organ Transplantation, Fudan University, Shanghai 200040, China
| | - Xizhi Chen
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yue Zhang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Baobing Yin
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Hepatobiliary surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, Fujian, China
| | - Jing Zhao
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Hepatobiliary surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, Fujian, China
- Cancer Metastasis Institute, Fudan University, Shanghai 201206, China
| |
Collapse
|
21
|
Chen Y, Jia H, Zhang X, Zhao H, Xiao Y, Li N, Yao Y, Xing X. Disruption of GPSM1/CSF1 signaling reprograms tumor-associated macrophages to overcome anti-PD-1 resistance in colorectal cancer. J Immunother Cancer 2025; 13:e010826. [PMID: 40010765 DOI: 10.1136/jitc-2024-010826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Immune checkpoint blockade (ICB) therapies, particularly anti-PD-1, benefit only a limited subset of colorectal cancer (CRC) patients. G-protein signaling modulator 1 (GPSM1) is implicated in immunity and oncology, yet its role in regulating the CRC tumor microenvironment (TME) and contributing to anti-PD-1 resistance remains poorly understood. METHODS We employed single-cell RNA sequencing and multiplex immunofluorescence on tumor samples from anti-PD-1-resistant CRC patients to evaluate GPSM1 expression and its impact on macrophage polarization. An orthotopic CRC xenograft model in C57BL/6 mice was used to assess the role of GPSM1 in vivo. An in vitro co-culture system, alongside mass cytometry and flow cytometry, explored GPSM1's biological functions within the TME. We further used ChIP-PCR, mass spectrometry, and co-immunoprecipitation to elucidate the mechanisms regulating GPSM1 activity. RESULTS GPSM1 expression was significantly elevated in anti-PD-1-resistant CRC tissues. Enhanced GPSM1 levels promoted anti-PD-1 resistance by driving macrophage polarization toward an immunosuppressive M2 phenotype, facilitating their infiltration into the TME. We identified the deubiquitinase USP9X as a key factor preventing GPSM1 degradation through K63-polyubiquitination. This stabilization of GPSM1 led to MEIS3 nuclear translocation, activating macrophage colony-stimulating factor expression. Importantly, ruxolitinib emerged as a promising GPSM1-targeting candidate, demonstrating improved efficacy in combination with anti-PD-1 therapy in both microsatellite instability-high and microsatellite stable CRC models. CONCLUSIONS Our findings highlight the pivotal role of GPSM1-driven M2 macrophage infiltration in mediating anti-PD-1 resistance in CRC. Targeting GPSM1 offers a novel therapeutic strategy to enhance ICB efficacy, potentially broadening the patient population that may benefit from these therapies.
Collapse
Affiliation(s)
- Yang Chen
- The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Huiqing Jia
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiangyan Zhang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Han Zhao
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yujing Xiao
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Na Li
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yifan Yao
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaoming Xing
- The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
22
|
Liu W, Niu J, Huo Y, Zhang L, Han L, Zhang N, Yang M. Role of circular RNAs in cancer therapy resistance. Mol Cancer 2025; 24:55. [PMID: 39994791 PMCID: PMC11854110 DOI: 10.1186/s12943-025-02254-5] [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: 01/06/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
Over the past decade, circular RNAs (circRNAs) have gained recognition as a novel class of genetic molecules, many of which are implicated in cancer pathogenesis via different mechanisms, including drug resistance, immune escape, and radio-resistance. ExosomalcircRNAs, in particular, facilitatecommunication between tumour cells and micro-environmental cells, including immune cells, fibroblasts, and other components. Notably, micro-environmental cells can reportedly influence tumour progression and treatment resistance by releasing exosomalcircRNAs. circRNAs often exhibit tissue- and cancer-specific expression patterns, and growing evidence highlights their potential clinical relevance and utility. These molecules show strong promise as potential biomarkers and therapeutic targets for cancer diagnosis and treatment. Therefore, this review aimed to briefly discuss the latest findings on the roles and resistance mechanisms of key circRNAs in the treatment of various malignancies, including lung, breast, liver, colorectal, and gastric cancers, as well as haematological malignancies and neuroblastoma.This review will contribute to the identification of new circRNA biomarkers for the early diagnosis as well as therapeutic targets for the treatment of cancer.
Collapse
Affiliation(s)
- Wenjuan Liu
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Jiling Niu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Yanfei Huo
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Long Zhang
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Linyu Han
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Nasha Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Ming Yang
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China.
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong Province, China.
| |
Collapse
|
23
|
Fang P, Yang J, Zhang H, Shuai D, Li M, Chen L, Liu L. Emerging roles of intratumoral microbiota: a key to novel cancer therapies. Front Oncol 2025; 15:1506577. [PMID: 40071093 PMCID: PMC11893407 DOI: 10.3389/fonc.2025.1506577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Microorganisms, including bacteria, viruses, and fungi, have been found to play critical roles in tumor microenvironments. Due to their low biomass and other obstacles, the presence of intratumor microbes has been challenging to definitively establish. However, advances in biotechnology have enabled researchers to reveal the association between intratumor microbiota and cancer. Recent studies have shown that tumor tissues, once thought to be sterile, actually contain various microorganisms. Disrupted mucosal barriers and adjacent normal tissues are important sources of intratumor microbiota. Additionally, microbes can invade tumors by traveling through the bloodstream to the tumor site and infiltrating through damaged blood vessels. These intratumor microbiota may promote the initiation and progression of cancers by inducing genomic instability and mutations, affecting epigenetic modifications, activating oncogenic pathways, and promoting inflammatory responses. This review summarizes the latest advancements in this field, including techniques and methods for identifying and culturing intratumor microbiota, their potential sources, functions, and roles in the efficacy of immunotherapy. It explores the relationship between gut microbiota and intratumor microbiota in cancer patients, and whether altering gut microbiota might influence the characteristics of intratumor microbiota and the host immune microenvironment. Additionally, the review discusses the prospects and limitations of utilizing intratumor microbiota in antitumor immunotherapy.
Collapse
Affiliation(s)
- Pengzhong Fang
- Departments of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Jing Yang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Huiyun Zhang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Diankui Shuai
- Departments of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Min Li
- Departments of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Lin Chen
- Departments of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Liping Liu
- Departments of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
24
|
Li J, Huang W, Kuang J, Zhou S, Li Y, Xia Y. Integrated multiomics analysis highlights the immunosuppressive role of granulin precursor positive macrophages in hepatocellular carcinoma. PeerJ 2025; 13:e18879. [PMID: 39995994 PMCID: PMC11849516 DOI: 10.7717/peerj.18879] [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: 09/19/2024] [Accepted: 12/28/2024] [Indexed: 02/26/2025] Open
Abstract
It has been reported that tumor-associated macrophages (TAMs) play a complicated role in cancer occurrence and development, immune escape, and immune checkpoint blockade (ICB) resistance. However, the role of granulin precursor (GRN) highly expressed macrophages (hereafter refer to GRN+ macrophages) in hepatocellular carcinoma (HCC) remains poorly understood. Herein, we systematically integrated multiomics analysis of human tumor tissues to illustrate the functional role of GRN+ macrophages in HCC. GRN is selectively expressed by TAMs in different type of cancers including HCC, and was significantly associated with poor prognosis in several type of cancer. GRN was closely correlated with infiltration levels of most immune cells, especially the M2 macrophage cells in various cancers. In particular, both mRNA and protein expression level of GRN was significantly upregulated in HCC. Compared with tumor tissue, GRN was more significantly expressed in the stroma area between HCC tissues and adjacent non-tumor tissues. High expression of GRN was significantly correlated with M2-polarization of macrophages and T-cell exhaustion in HCC. GRN+ macrophages communicated with intratumoral immune cells, especially CD8+ T cells. Functionally, GRN+ macrophages contacted with CD8+ T cells, which inducing T-cell exhaustion. Our study offers a comprehensive understanding of the clinical relevance and immunological role of GRN+ macrophages in HCC, indicating its potential role as a promising target for immunotherapeutic strategies.
Collapse
Affiliation(s)
- Jun Li
- Shantou University Medical College, Shantou, China
| | - Weizhen Huang
- The Second Department of Oncology, The First Huizhou Affiliated Hospital of Guangdong Medical University, Huizhou, China
| | - Junjie Kuang
- The Second Department of Oncology, The First Huizhou Affiliated Hospital of Guangdong Medical University, Huizhou, China
| | - Siwei Zhou
- The Second Department of Oncology, The First Huizhou Affiliated Hospital of Guangdong Medical University, Huizhou, China
| | - Yi Li
- The Second Department of Oncology, The First Huizhou Affiliated Hospital of Guangdong Medical University, Huizhou, China
| | - Yuan Xia
- The Second Department of Oncology, The First Huizhou Affiliated Hospital of Guangdong Medical University, Huizhou, China
| |
Collapse
|
25
|
Wang N, Wu S, Huang L, Hu Y, He X, He J, Hu B, Xu Y, Rong Y, Yuan C, Zeng X, Wang F. Intratumoral microbiome: implications for immune modulation and innovative therapeutic strategies in cancer. J Biomed Sci 2025; 32:23. [PMID: 39966840 PMCID: PMC11837407 DOI: 10.1186/s12929-025-01117-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 01/09/2025] [Indexed: 02/20/2025] Open
Abstract
Recent advancements have revealed the presence of a microbiome within tumor tissues, underscoring the crucial role of the tumor microbiome in the tumor ecosystem. This review delves into the characteristics of the intratumoral microbiome, underscoring its dual role in modulating immune responses and its potential to both suppress and promote tumor growth. We examine state-of-the-art techniques for detecting and analyzing intratumoral bacteria, with a particular focus on their interactions with the immune system and the resulting implications for cancer prognosis and treatment. By elucidating the intricate crosstalk between the intratumoral microbiome and the host immune system, we aim to uncover novel therapeutic strategies that enhance the efficacy of cancer treatments. Additionally, this review addresses the existing challenges and future prospects within this burgeoning field, advocating for the integration of microbiome research into comprehensive cancer therapy frameworks.
Collapse
Affiliation(s)
- Na Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Si Wu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Lanxiang Huang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yue Hu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xin He
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jourong He
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ben Hu
- Center for Tumor Precision Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yaqi Xu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yuan Rong
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chunhui Yuan
- Department of Laboratory Medicine, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430016, China.
| | - Xiantao Zeng
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Fubing Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, 430071, China.
| |
Collapse
|
26
|
Ye C, Zhang B, Lin Y, Han F, Shi H, Dong C, Zhou W. Characteristics of gut microbiota and metabolites in extrahepatic cholangiocarcinoma and their prognostic value for resectable lesions. Front Cell Infect Microbiol 2025; 15:1523863. [PMID: 40028184 PMCID: PMC11868125 DOI: 10.3389/fcimb.2025.1523863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
This study aimed to investigate the relationship between gut microbiota composition, fecal metabolites, and postoperative prognosis in patients with extrahepatic cholangiocarcinoma (eCCA). A total of 53 patients with resectable eCCA and 21 healthy volunteers as a control group were included. 16S rRNA gene sequencing and metabolomic analyses revealed significant differences in the gut microbial community structure and altered fecal metabolites profiles between eCCA patients and healthy controls. Univariate and multivariate Cox regression analyses indicated that factors such as preoperative total bilirubin, indirect bilirubin, and specific metabolites were closely associated with overall survival in patients with eCCA post-surgery. The constructed nomogram model further demonstrated the predictive value of these factors, achieving a C-index of 0.718, with calibration curves confirming its strong predictive performance. In conclusion, gut microbiota composition and fecal metabolites play a crucial role in the surgical prognosis of eCCA patients, providing new insights for clinical prognostic assessment.
Collapse
Affiliation(s)
- Cheng Ye
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Bo Zhang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yanyan Lin
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Fangfang Han
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Huaqing Shi
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Chunlu Dong
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Wence Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
27
|
Yang H, Li J, Niu Y, Zhou T, Zhang P, Liu Y, Li Y. Interactions between the metabolic reprogramming of liver cancer and tumor microenvironment. Front Immunol 2025; 16:1494788. [PMID: 40028341 PMCID: PMC11868052 DOI: 10.3389/fimmu.2025.1494788] [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: 09/11/2024] [Accepted: 01/29/2025] [Indexed: 03/05/2025] Open
Abstract
Metabolic reprogramming is one of the major biological features of malignant tumors, playing a crucial role in the initiation and progression of cancer. The tumor microenvironment consists of various non-cancer cells, such as hepatic stellate cells, cancer-associated fibroblasts (CAFs), immune cells, as well as extracellular matrix and soluble substances. In liver cancer, metabolic reprogramming not only affects its own growth and survival but also interacts with other non-cancer cells by influencing the expression and release of metabolites and cytokines (such as lactate, PGE2, arginine). This interaction leads to acidification of the microenvironment and restricts the uptake of nutrients by other non-cancer cells, resulting in metabolic competition and symbiosis. At the same time, metabolic reprogramming in neighboring cells during proliferation and differentiation processes also impacts tumor immunity. This article provides a comprehensive overview of the metabolic crosstalk between liver cancer cells and their tumor microenvironment, deepening our understanding of relevant findings and pathways. This contributes to further understanding the regulation of cancer development and immune evasion mechanisms while providing assistance in advancing personalized therapies targeting metabolic pathways for anti-cancer treatment.
Collapse
Affiliation(s)
- Haoqiang Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Jinghui Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yiting Niu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Tao Zhou
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Pengyu Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yang Liu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yanjun Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, TongjiShanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
28
|
Kuang L, Wu L, Li Y. Extracellular vesicles in tumor immunity: mechanisms and novel insights. Mol Cancer 2025; 24:45. [PMID: 39953480 PMCID: PMC11829561 DOI: 10.1186/s12943-025-02233-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/14/2025] [Indexed: 02/17/2025] Open
Abstract
Extracellular vesicles (EVs), nanoscale vesicles secreted by cells, have attracted considerable attention in recent years due to their role in tumor immunomodulation. These vesicles facilitate intercellular communication by transporting proteins, nucleic acids, and other biologically active substances, and they exhibit a dual role in tumor development and immune evasion mechanisms. Specifically, EVs can assist tumor cells in evading immune surveillance and attack by impairing immune cell function or modulating immunosuppressive pathways, thereby promoting tumor progression and metastasis. Conversely, they can also transport and release immunomodulatory factors that stimulate the activation and regulation of the immune system, enhancing the body's capacity to combat malignant diseases. This dual functionality of EVs presents promising avenues and targets for tumor immunotherapy. By examining the biological characteristics of EVs and their influence on tumor immunity, novel therapeutic strategies can be developed to improve the efficacy and relevance of cancer treatment. This review delineates the complex role of EVs in tumor immunomodulation and explores their potential implications for cancer therapeutic approaches, aiming to establish a theoretical foundation and provide practical insights for the advancement of future EVs-based cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Liwen Kuang
- School of Medicine, Chongqing University, Chongqing, China
| | - Lei Wu
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yongsheng Li
- School of Medicine, Chongqing University, Chongqing, China.
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
29
|
Zhang X, Shi C, Liu Q, Zhong Y, Zhu L, Zhao Y. Combination of adenosine blockade and ferroptosis for photo-immunotherapy of triple negative breast cancer with aptamer-modified copper sulfide. J Mater Chem B 2025; 13:2504-2519. [PMID: 39834279 DOI: 10.1039/d4tb02125h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Combination of immunotherapy and photothermal therapy (PTT) provides a promising therapeutic performance for tumors. However, it still faces negative feedback from suppressive factors such as adenosine. Herein, we developed a new nanodrug that can combine adenosine blockade and ferroptosis to promote the photoimmunotherapy of triple negative breast cancer (TNBC). The nanodrug, named CuS-PEG@Apt, was constructed via the modification of copper sulfide (CuS) nanoparticles with adenosine aptamer and PEG. CuS-PEG@Apt could be effectively enriched in the tumor site and locally generate a strong photothermal effect, directly ablating tumors and inducing immunogenic death (ICD). On the other hand, the aptamers could block the adenosine pathway to inhibit the immune suppression by adenosine, which further promoted the anti-tumor immunity. Moreover, the CuS nanoparticles could consume GSH and inhibit GPX4 to cause the ferroptosis of tumor cells. Collectively, CuS-PEG@Apt achieved potent efficacy of tumor suppression via the combination of PTT, immune activation and ferroptosis, representing an appealing platform for TNBC treatment.
Collapse
Affiliation(s)
- Xingyu Zhang
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Chengyu Shi
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Qiao Liu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Yuting Zhong
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Lipeng Zhu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Yuetao Zhao
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| |
Collapse
|
30
|
Yang Z, Chen W, Liu Y, Niu Y. Recent updates of centromere proteins in hepatocellular carcinoma: a review. Infect Agent Cancer 2025; 20:7. [PMID: 39915786 PMCID: PMC11800463 DOI: 10.1186/s13027-024-00630-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/16/2024] [Indexed: 02/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related death worldwide, with approximately 800,000 deaths worldwide each year. Owing to the atypical early symptoms and characteristics of HCC, over 80% of HCC patients cannot receive curative treatment. The treatment of HCC is facing a bottleneck, and new treatment methods are urgently needed. Since the pathogenesis of HCC is not yet clear, identifying the molecular mechanisms and therapeutic targets related to it is crucial. Centromeres are considered special deoxyribonucleic acid (DNA) sequences with highly repetitive sequences that are physically connected to the spindle during cell division, ensuring equal division of genetic material between daughter cells. The numerous proteins that aggregate on this sequence during cell division are called centromere proteins (CENPs). Currently, numerous studies have shown that CENPs are abnormally expressed in tumor cells and are associated with patient prognosis. The abnormal expression of CENPs is a key cause of chromosomal instability. Furthermore, chromosomal instability is a common characteristic of the majority of tumors. Chromosomal instability can lead to uncontrolled and sustained division and proliferation of malignant tumors. Therapeutic plans targeting CENPs play important roles in the treatment of HCC. For example, small ribonucleic acid (RNA) can silence CENP expression and prevent the occurrence and development of liver cancer. In recent years, studies of HCC-targeting CENPs have gradually increased but are still relatively novel, requiring further systematic elaboration. In this review, we provide a detailed introduction to the characteristics of CENPs and discuss their roles in HCC. In addition, we discuss their application prospects in future clinical practice.
Collapse
Affiliation(s)
- Zhongyuan Yang
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, 1095, Jiefang Avenue, Wuhan, 430030, Hubei, China.
| | - Wenjiao Chen
- Department of Dermatology, Wuhan Hankou Hospital, Wuhan, Hubei, China
| | - Yunhui Liu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, 1095, Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Yuxin Niu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, 1095, Jiefang Avenue, Wuhan, 430030, Hubei, China
| |
Collapse
|
31
|
Davis RJ, Talatala ERR, Wu H, Zhang Y, Park JS, Gelbard A. PD-L1 Acts Independently of PD-1 as a Marker of Pathologic Fibroblasts in Laryngotracheal Stenosis. Otolaryngol Head Neck Surg 2025; 172:563-570. [PMID: 39441651 PMCID: PMC11773441 DOI: 10.1002/ohn.1034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/19/2024] [Accepted: 10/12/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVE Laryngotracheal stenosis (LTS) describes fibrotic airway obstruction that is life-threatening without treatment. Targeted therapies are needed as an adjunct to surgical management. We have previously observed the upregulation of immune checkpoint programmed cell death (PD)-1 and its ligand, PD-L1, in patients with LTS. This study aims to determine whether PD-1 and PD-L1 play a role in the pathophysiology of LTS. STUDY DESIGN Basic science. SETTING Laboratory. METHODS Fibroblasts derived from the subglottic scar of 5 iSGS patients were cultured ex vivo with transforming growth factor β (TGFβ), PD-L1 agonist (PD-1), and PD-L1 blockade (anti-PD-L1). PD-L1, TGFβ receptor II (TGFβRII), and Collagen-1 expression were quantified by flow cytometry. A validated chemomechanical injury model of subglottic stenosis was applied in PD-1 knockout and wild-type (WT) mice, and subglottic thickening was assessed by histologic analysis. RESULTS TGFβ significantly increased the expression of PD-L1 and Collagen-1 in human airway scar fibroblasts (P < .05). PD-1 knockout mice demonstrated no significant difference in subglottic airway fibrosis compared to WT mice. Ex vivo PD-L1 modulation had no impact on fibroblast Collagen-1 expression. PD-L1 high-intensity fibroblasts expressed greater Collagen-1 and TGFβRII compared to PD-L1 low-intensity fibroblasts. CONCLUSION PD-1 knockout does not protect mice from the development of laryngotracheal fibrosis. However, its ligand, PD-L1 is highly expressed on pathologic fibroblasts unique to scar, characterized by high Collagen-1 and TGFβRII expression. PD-L1 is also upregulated in conjunction with Collagen-1 by TGFβ stimulation. PD-L1 may act independently of PD-1 to sensitize fibroblasts to TGFβ, suggesting direct targeting of PD-L1 may have therapeutic potential in LTS.
Collapse
Affiliation(s)
- Ruth J. Davis
- Division of Otolaryngology–Head and Neck Surgery, Department of SurgeryUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - Edward Ryan R. Talatala
- Department of Otolaryngology–Head and Neck SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Hongmei Wu
- Department of Otolaryngology–Head and Neck SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Yueli Zhang
- Department of Otolaryngology–Head and Neck SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jason S. Park
- Department of Otolaryngology–Head and Neck SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Alexander Gelbard
- Department of Otolaryngology–Head and Neck SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
32
|
Yan C, Du Y, Cui L, Bao H, Li H. CircPTK2 as a Valuable Biomarker and Treatment Target in Cancer. J Biochem Mol Toxicol 2025; 39:e70161. [PMID: 39887513 DOI: 10.1002/jbt.70161] [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: 01/06/2025] [Accepted: 01/19/2025] [Indexed: 02/01/2025]
Abstract
Circular RNA (CircRNA)s, a newly discovered type of noncoding RNAs, have been found to play a role in controlling the development and aggressiveness of tumors. Abnormal control of circRNA has been observed in various types of human cancers, including bladder cancer, hepatocellular carcinoma (HCC), breast cancer, and gastric cancer (GC). CircRNAs possess binding sites for microRNAs (miRNAs) and function as miRNA sponges in posttranscriptional regulation. This mechanism has been documented to influence the course of cancer. Significantly, among these putative circRNAs, circular RNA protein tyrosine kinase 2 (circPTK2) exhibited increased expression and displayed a substantial association with adverse clinical characteristics and a negative prognosis. The production of these transcripts occurs via a back-splicing mechanism. The enclosed conformation of circRNAs shields them from destruction and enhances their potential as biomarkers. Gaining insight into the molecular mechanisms involved in these processes would aid in the development of treatment approaches and the discovery of new tumor markers. This article provides a comprehensive assessment of the latest research on the biosynthesis and features of circRNAs. It examines the role of circPTK2 in the diagnosis, treatment, and prognosis evaluation of cancer.
Collapse
Affiliation(s)
- Chengqiu Yan
- Department of Anorectal Center, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Yu Du
- Department of Anorectal Center, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Lihong Cui
- Department of Anorectal Center, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Han Bao
- Department of Anorectal, Changchun Hospital of Traditional Chinese Medicine, Changchun, China
| | - Hui Li
- Department of Anorectal Center, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
33
|
Wu J, Zhang C, Zhang Y, He R, Wang Q, Zhang L, Hu J, Wan R. Prediction model establishment of prognosis factors for distant metastasis of hepatocellular carcinoma based on the SEER database. Cancer Epidemiol 2025; 94:102729. [PMID: 39675259 DOI: 10.1016/j.canep.2024.102729] [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: 08/25/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Distant metastasis in hepatocellular carcinoma (HCC) is an important indicator of poor patient prognosis. Identifying patients who are at high risk of metastasis early on is essential for creating personalized treatment plans, yet currently, there is a scarcity of effective predictive tools. OBJECTIVE To investigate the effects of different factors on distant metastasis in HCC patients and to establish a clinical prediction model for predicting distant metastasis in HCC patients. METHODS Our study retrospectively examined 22,318 patients diagnosed with confirmed HCC from the SEER database. Prognostic factors for developing distant metastases in HCC patients were identified by univariate and multivariate logistic regression analyses. Utilizing data from a multivariate logistic regression analysis, we created a nomogram. Its predictive precision was evaluated by analyzing the calibration curve, the area under the curve (AUC) of the receiver operating characteristic curve, decision curve assessment (DCA), and Kaplan-Meier (KM) curve analysis of overall survival. Finally,the nomogram was visualized with an online calculator. RESULTS We identified six independent prognostic factors: ethnicity, marital status, tumor size, survival time, surgery, and radiotherapy. The nomogram constructed from these six factors showed good calibration, discrimination, and clinical application value after calibration curve analysis, receiver operating characteristic curve analysis and DCA curve analysis. Besides, KaplanMeier survival curves also demonstrated that this nomogram had predictive accuracy. CONCLUSION In this research, a nomogram model was created to accurately predict distant metastasis risk in patients with HCC. This study provides guidance for optimizing individual therapies and making better clinical decisions.
Collapse
Affiliation(s)
- Jixuan Wu
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; Key Laboratory of Luzhou City for Aging Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Chun Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; Key Laboratory of Luzhou City for Aging Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Youjia Zhang
- School of Public Health, Southwest Medical University, Luzhou 646000, China
| | - Rui He
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Qin Wang
- Dazhou Vocational College of Chinese Medicine, Dazhou, Dazhou 635000, China
| | - Lei Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jing Hu
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Runlan Wan
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; Key Laboratory of Luzhou City for Aging Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
34
|
Song JB, Guo SS, Gao WJ, Yang ZP, Tian ZL. Cellular Membrane Protein GRINA is Highly Expressed and Associated with Survival Outcomes in Liver Cancer Patients. Curr Med Sci 2025; 45:122-136. [PMID: 40011365 DOI: 10.1007/s11596-025-00025-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/23/2025] [Accepted: 01/26/2025] [Indexed: 02/28/2025]
Abstract
OBJECTIVE Hepatocellular carcinoma (HCC), a lethal cancer with high global mortality, may be targeted through ferroptosis, an iron-dependent form of cell death. Despite its potential, the prognostic value of ferroptosis in HCC is underexplored. METHODS Our study leveraged single-cell and bulk sequencing datasets to identify ferroptosis-related genes and developed a prognostic model via Cox and LASSO regression analyses. Survival and mutation analyses led to the creation of a nomogram for predicting patient prognosis. Furthermore, we investigated the role of GRINA, a ferroptosis-related gene, through functional assays, including cell proliferation, colony formation, and metastatic potential analyses. We also assessed mitochondrial abnormalities, intracellular iron, and ROS levels in GRINA-knockdown cells. RESULTS The developed ferroptosis-related model classified HCC patients into risk groups, revealing notable survival disparities. High-risk patients presented increased immune checkpoint gene expression. The nomogram revealed robust prognostic accuracy. Additionally, we found that GRINA suppression reduced HCC cell proliferation, colony formation, and metastatic potential. Cells with GRINA knockdown presented mitochondrial abnormalities and increased intracellular iron and ROS levels. CONCLUSIONS By analysing multiomics sequencing data, we established a connection between ferroptosis-related risk groups and the tumor immune microenvironment. These findings provide novel insights into the role of ferroptosis in HCC and suggest that GRINA inhibition is a potential therapeutic strategy, leading to mitochondrial damage and the induction of ferroptosis in HCC cell lines.
Collapse
Affiliation(s)
- Jun-Bo Song
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Shan-Shan Guo
- Department of Physiology and Pathophysiology, Air Force Medical University, Xi'an, 710032, China
| | - Wen-Jie Gao
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Zhi-Peng Yang
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Ze-Lin Tian
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China.
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
35
|
Wang D, Yu X, Yang Y. Investigating SNHG3 as a potential therapeutic approach for HCC stem cells. Gene 2025; 935:149022. [PMID: 39427830 DOI: 10.1016/j.gene.2024.149022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/13/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION Hepatocellular Carcinoma (HCC) is a common malignant tumor worldwide. Long Non-Coding RNA (lncRNA) has gained attention in tumor biology, and this study aims to investigate the role of lncRNA SNHG3 in HCC, specifically in the self-renewal and maintenance of liver cancer stem cells. METHODS The expression of lncRNA SNHG3 was analyzed in HCC and adjacent normal tissue using the TCGA database. The expression levels of SNHG3 in HCC cell lines (Hep3B, HepG2, Huh7) were detected using qRT-PCR and Western blot techniques. Functional assays, including CCK-8, soft agar colony formation, and tumor sphere formation, were performed to evaluate the impact of SNHG3 on HCC stem cell functionality. MeRIP-qPCR was also used to investigate the regulatory role of SNHG3 in m6A modification of ITGA6 mRNA mediated by METTL3. RESULTS The study found that SNHG3 was significantly upregulated in HCC tissue and cell lines compared to normal liver tissue. SNHG3 expression correlated with the pathological stage, metastasis status, and tumor size of liver cancer. Inhibiting SNHG3 reduced proliferation, colony formation, and tumor sphere formation ability in HCC stem cells. SNHG3 also played a role in regulating the m6A modification and expression of ITGA6 through METTL3. CONCLUSION This study emphasizes the upregulation of lncRNA SNHG3 and its role in HCC stem cell self-renewal. SNHG3 may regulate the m6A modification of ITGA6 mRNA through its interaction with METTL3, impacting the function of liver cancer stem cells. These findings support the potential of targeting SNHG3 as a therapeutic approach for HCC.
Collapse
Affiliation(s)
- Dingmao Wang
- Department of Hepatobiliary Surgery, Haikou People's Hospital, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, PR China
| | - Xiao Yu
- The 2nd Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, PR China.
| | - Yijun Yang
- Department of Hepatobiliary Surgery, Haikou People's Hospital, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, PR China.
| |
Collapse
|
36
|
Liu H, Zhang C, Peng S, Yin Y, Xu Y, Wu S, Wang L, Fu Y. Prognostic models of immune-related cell death and stress unveil mechanisms driving macrophage phenotypic evolution in colorectal cancer. J Transl Med 2025; 23:127. [PMID: 39875913 PMCID: PMC11776142 DOI: 10.1186/s12967-025-06143-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/14/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Tumor microenvironment (TME), particularly immune cell infiltration, programmed cell death (PCD) and stress, has increasingly become a focal point in colorectal cancer (CRC) treatment. Uncovering the intricate crosstalk between these factors can enhance our understanding of CRC, guide therapeutic strategies, and improve patient prognosis. METHODS We constructed an immune-related cell death and stress (ICDS) prognostic model utilizing machine learning methodologies. Furthermore, we performed enrichment analyses and deconvolution algorithms to elucidate the complex interactions between immune cell infiltration and the processes of PCD and stress within a substantial array of transcriptomic data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus data base (GEO) related to CRC. Single-cell sequencing and biochemical experiments were used to validate the interaction between the model genes and programmed cell death in tumor cells. RESULTS The ICDS prognostic model exhibited robust predictive performance in seven independent cohorts, revealing an inverse correlation between model scores and patient prognosis. Meanwhile, the ICDS index was positively correlated with clinical stage. Model analysis indicated that patient subgroups with low ICDS index exhibited heightened immune activation features and elevated activity in PCD and stress pathways. Single-cell analysis further revealed that macrophages were the central drivers of immune characteristics underlying prognostic differences within the ICDS prognostic model. Pseudotime analysis and cellular experiments indicated that the model gene GAL3ST4 promotes the transition of macrophages toward an M2 pro-tumor phenotype. Furthermore, cell communication analysis and experimental validation revealed that the cuproptosis in tumor cells suppress GAL3ST4 expression, thereby inhibiting M2-like macrophage polarization. CONCLUSION In summary, we constructed the ICDS prognostic model and uncovered the mechanism by which tumor cells downregulate GAL3ST4 expression via cuproptosis to inhibit M2-like macrophage polarization, providing new targets and biomarkers for CRC treatment and prognosis evaluation.
Collapse
Affiliation(s)
- Hao Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Chuhan Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Sanfei Peng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yuhan Yin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yishi Xu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Sihan Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Liping Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
37
|
Cheng C, Zha Q, Sun L, Cui T, Guo X, Xing C, Chen Z, Ji C, Liang S, Tao S, Chu J, Wu C, Chu Q, Gu X, Zhang N, Fu Y, Deng S, Zhu Y, Wang J, Liu Y, Liu L. VCP downstream metabolite glycerol-3-phosphate (G3P) inhibits CD8 +T cells function in the HCC microenvironment. Signal Transduct Target Ther 2025; 10:26. [PMID: 39848960 PMCID: PMC11758394 DOI: 10.1038/s41392-024-02120-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/25/2025] Open
Abstract
CD8+T cells within the tumor microenvironment (TME) are often functionally impaired, which limits their ability to mount effective anti-tumor responses. However, the molecular mechanisms behind this dysfunction remain incompletely understood. Here, we identified valosin-containing protein (VCP) as a key regulator of CD8+T cells suppression in hepatocellular carcinoma (HCC). Our findings reveal that VCP suppresses the activation, expansion, and cytotoxic capacity of CD8+T cells both in vitro and in vivo, significantly contributing to the immunosuppressive nature of the TME. Mechanistically, VCP stabilizes the expression of glycerol-3-phosphate dehydrogenase 1-like protein (GPD1L), leading to the accumulation of glycerol-3-phosphate (G3P), a downstream metabolite of GPD1L. The accumulated G3P diffuses into the TME and directly interacts with SRC-family tyrosine kinase LCK, a critical component of the T-cell receptor (TCR) signaling pathway in CD8+T cells. This interaction heightens the phosphorylation of Tyr505, a key inhibitory residue, ultimately reducing LCK activity and impairing downstream TCR signaling. Consequently, CD8+T cells lose their functional capacity, diminishing their ability to fight against HCC. Importantly, we demonstrated that targeting VCP in combination with anti-PD1 therapy significantly suppresses HCC tumor growth and restores the anti-tumor function of CD8+T cells, suggesting synergistic therapeutic potential. These findings highlight a previously unrecognized mechanism involving VCP and G3P in suppressing T-cell-mediated immunity in the TME, positioning VCP as a promising upstream target for enhancing immunotherapy in HCC.
Collapse
Affiliation(s)
- Cheng Cheng
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Qingrui Zha
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Linmao Sun
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Tianming Cui
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Xinyu Guo
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Changjian Xing
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Zhengxiang Chen
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Changyong Ji
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Shuhang Liang
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
- Department of Gastrointestinal Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Shengwei Tao
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Junhui Chu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Chenghui Wu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Qi Chu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Xuetian Gu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Ning Zhang
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Yumin Fu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Shumin Deng
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Yitong Zhu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Jiabei Wang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China.
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China.
| | - Yao Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China.
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China.
| | - Lianxin Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China.
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China.
| |
Collapse
|
38
|
de la Cruz-Ojeda P, Parras-Martínez E, Rey-Pérez R, Muntané J. In silico analysis of lncRNA-miRNA-mRNA signatures related to Sorafenib effectiveness in liver cancer cells. World J Gastroenterol 2025; 31:95207. [PMID: 39839902 PMCID: PMC11684161 DOI: 10.3748/wjg.v31.i3.95207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/30/2024] [Accepted: 09/12/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the most common subtype of primary liver cancer with varied incidence and epidemiology worldwide. Sorafenib is still a recommended treatment for a large proportion of patients with advanced HCC. Different patterns of treatment responsiveness have been identified in differentiated hepatoblastoma HepG2 cells and metastatic HCC SNU449 cells. AIM To define the long non-codingRNA-microRNA-mRNA (lncRNA-miRNA-mRNA) predicted signatures related to selected hallmarks of cancer (apoptosis, autophagy, cell stress, cell dedifferentiation and invasiveness) in RNAseq studies using Sorafenib-treated HepG2 and SNU449 cells. Various available software analyses allowed us to establish the lncRNA-miRNA-mRNA regulatory axes following treatment in HepG2 and SNU449 cells. METHODS HepG2 and SNU449 cells were treated with Sorafenib (10 μmol/L) for 24 hours. Total RNA, including small and long RNA, was extracted with a commercial miRNeasy kit. RNAseq was carried out for the identification of changes in lncRNA-miRNA-mRNA regulatory axes. RESULTS MALAT, THAP9-AS1 and SNGH17 appeared to coordinately regulate miR-374b-3p and miR-769-5p that led to upregulation of SMAD7, TIRARP, TFAP4 and FAXDC2 in HepG2 cells. SNHG12, EPB41 L4A-AS1, LINC01578, SNHG12 and GAS5 interacted with let-7b-3p, miR-195-5p and VEGFA in SNU449 cells. The axes MALAT1/hsa-mir-374b-3p/SMAD7 and MALAT1/hsa-mir-769-5p/TFAP4 were of high relevance for Sorafenib response in HepG2 cells, whereas PVT1/hsa-miR-195-5p/VEGFA was responsible for the differential response of SNU449 cells to Sorafenib treatment. CONCLUSION Critical lncRNAs acting as sponges of miRNA were identified that regulated mRNA expression, whose proteins mainly increased the antitumor effectiveness of the treatment (SMAD7, TIRARP, TFAP4, FAXDC2 and ADRB2). However, the broad regulatory axis leading to increased VEGFA expression may be related to the side effect of Sorafenib in SNU449 cells.
Collapse
Affiliation(s)
- Patricia de la Cruz-Ojeda
- Functional Genomics of Solid Tumors Laboratory, Centre de Recherche des Cordeliers, Paris 75006, France
- Department of Oncology Surgery, Cell Therapy and Organ Transplantation, Institute of Biomedicine of Seville, Virgen del Rocio University Hospital, Seville 41013, Spain
- Biomedical Research Center for Hepatic and Digestive Diseases, CIBERehd, Madrid 28029, Spain
| | - Ester Parras-Martínez
- Department of Oncology Surgery, Cell Therapy and Organ Transplantation, Institute of Biomedicine of Seville, Virgen del Rocio University Hospital, Seville 41013, Spain
| | - Raquel Rey-Pérez
- Department of Oncology Surgery, Cell Therapy and Organ Transplantation, Institute of Biomedicine of Seville, Virgen del Rocio University Hospital, Seville 41013, Spain
| | - Jordi Muntané
- Department of Oncology Surgery, Cell Therapy and Organ Transplantation, Institute of Biomedicine of Seville, Virgen del Rocio University Hospital, Seville 41013, Spain
- Biomedical Research Center for Hepatic and Digestive Diseases, CIBERehd, Madrid 28029, Spain
- Department of Medical Physiology and Biophysics, University of Seville, Seville 41009, Spain
| |
Collapse
|
39
|
Jiang F, Dang Y, Zhang Z, Yan Y, Wang Y, Chen Y, Chen L, Zhang J, Liu J, Wang J. Association of intratumoral microbiome diversity with hepatocellular carcinoma prognosis. mSystems 2025; 10:e0076524. [PMID: 39660866 PMCID: PMC11748501 DOI: 10.1128/msystems.00765-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 11/03/2024] [Indexed: 12/12/2024] Open
Abstract
The evidence that intratumoral microbiomes, as a rising hallmark of cancer, have a profound impact on cancer phenotypes is increasingly compelling. However, the impact of the composition and diversity of the intratumoral microbiome on the prognosis of patients undergoing surgical resection for hepatocellular carcinoma (HCC) remains incompletely understood. In this study, we revealed a high abundance of bacteria in the neoplastic tissues. The presence of bacterial lipopolysaccharide and lipoteichoic acid was detected alongside tumor-associated immune cells. By utilizing 16S rRNA gene sequencing, we identified a specific intratumoral microbiome signature that was highly predictive of the prognosis for HCC patients who underwent surgical resection. Specifically, the presence of Intestinimonas, Brachybacterium, and Rothia were identified as independent risk factors for the overall survival of HCC patients who underwent surgical resection.IMPORTANCEAlthough some studies have shown an abundance of bacteria in hepatocellular carcinoma (HCC), there is still limited understanding of the composition and diversity of the intratumoral microbiome that is favorable or adverse to the prognosis of HCC patients. Our results indicated that a greater abundance of bacteria could be observed in the neoplastic tissues than in nonneoplastic tissues. Bacterial cell wall components largely coincided with tumor-associated immune cells. The bacteria in the long overall survival (LOS) group were associated with metabolism and cytokine‒cytokine receptor interaction pathways, while bacteria in the short overall survival (SOS) group were associated with proinflammatory and cell proliferation pathways. Notably, specific taxa could independently predict HCC prognosis. Based on these findings, intratumoral microbiomes facilitate the use of precision medicine in clinical practice.
Collapse
Affiliation(s)
- Fengle Jiang
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Yuan Dang
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Zheting Zhang
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Yanan Yan
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Yingchao Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Yi Chen
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Lihong Chen
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Jialiang Zhang
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Jingfeng Liu
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
- Department of Hepatopancreatobiliary Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Jianmin Wang
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| |
Collapse
|
40
|
Song Y, Tian S, Li Z, Miao J, Wu M, Xu T, Wu X, Qiao J, Zhang X, Zhao H, Kang L, Cao L, Zhu P, Miao M. Progress in the Study of Intratumoral Microorganisms in Hepatocellular Carcinoma. J Hepatocell Carcinoma 2025; 12:59-76. [PMID: 39845367 PMCID: PMC11752873 DOI: 10.2147/jhc.s496964] [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: 09/19/2024] [Accepted: 01/01/2025] [Indexed: 01/24/2025] Open
Abstract
The intratumoral microbiota, an integral part of liver tumors, has garnered significant attention from researchers due to its role in tumor development regulation and impact on cancer treatment. Intratumoral microorganism not only influences tumorigenesis and progression, but also serves as potential biomarkers and targets for tumor therapy. Targeted manipulation of these microorganisms holds great promise for personalized liver cancer treatment. However, there is a lack of systematic summaries and reports on the study of intratumoral microorganism in hepatocellular carcinoma. This comprehensive review aims to address this gap by summarizing research progress related to in the field of hepatocellular carcinoma intratumoral bacteria, including their sources, types, distribution characteristics within tumors, impact on tumor development, underlying mechanisms, and application prospects. Through the analysis, it is proposed that intratumor organisms can be used as markers for liver cancer diagnosis and treatment, drug carrier materials for targeting liver cancer tissues, and the research prospects of developing new combination therapies based on the in-depth understanding of the interactions between intratumor microorganisms and the tumor microenvironment, immune cells, liver cancer cells, etc. as well as exploring the prospects of developing new combination therapies based on these interactions. It is hoped that from the perspective of intratumoral microbiota, potential theoretical support can be provided for future research on targeted cancer therapy for liver cancer intratumoral microbiota, and new insights and ideas can be provided for targeting points and research methods in tumor research.
Collapse
Affiliation(s)
- Yagang Song
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
| | - Shuo Tian
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
| | - Zhanzhan Li
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
| | - Jinxin Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
| | - Mingming Wu
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
| | - Tingli Xu
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
| | - Xiangxiang Wu
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
| | - Jingyi Qiao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
| | - Xialei Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
| | - Hui Zhao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
| | - Le Kang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
| | - Lihua Cao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
| | - Pingsheng Zhu
- College of Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
| | - Mingsan Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, People’s Republic of China
| |
Collapse
|
41
|
Wang W, Hashimi B, Wang P. Targeting ferroptosis: the role of non-coding RNAs in hepatocellular carcinoma progression and therapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03791-y. [PMID: 39820644 DOI: 10.1007/s00210-025-03791-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/06/2025] [Indexed: 01/19/2025]
Abstract
One of the most common tumors is hepatocellular carcinoma (HCC), and the prognosis for late-stage HCC is still not good. It is anticipated that improved outcomes would result from a deeper comprehension of the pathophysiology of HCC. Ferroptosis as a new discovered cell death type is linked to the progression of HCC and may be crucial for its detection, prevention, therapy, and prognosis. Numerous studies suggest that epigenetic alterations mediated by non-coding RNAs (ncRNA) might influence cancer cell susceptibility to ferroptosis. This study elucidates the processes of ferroptosis and delineates the paths by which ncRNAs influence HCC by modulating ferroptosis. Furthermore, it offers significant insights into ferroptosis-associated ncRNAs, intending to discover novel therapeutic approaches for HCC. It also explores innovative concepts for the future use of ncRNA-based ferroptosis-targeted therapeutics.
Collapse
Affiliation(s)
- Weijia Wang
- Department of Oncology, Qilu Hospital of Shandong University Dezhou Hospital (Dezhou People's Hospital), Shandong Province, China
| | - Behishta Hashimi
- Department of Midwifery, Jahan Institute of Health Sciences, Kabul, Afghanistan
| | - Ping Wang
- Department of Oncology, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, Shandong Province, China.
| |
Collapse
|
42
|
Sun L, Ma Y, Geng C, Gao X, Li X, Ru Q, Zhu S, Zhang P. DPP4, a potential tumor biomarker, and tumor therapeutic target: review. Mol Biol Rep 2025; 52:126. [PMID: 39821530 DOI: 10.1007/s11033-025-10235-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025]
Abstract
Dipeptidyl peptidase 4 (DPP4) is a serine protease widely distributed in membrane-bound and soluble forms in various tissues and organs throughout the body. DPP4 plays a role in inflammation, immune regulation, cell growth, migration and differentiation. The role of DPP4 in tumors has garnered increasing attention. Previous research has demonstrated that DPP4 contributes to the promotion of cancer in most cancers, and it may play a specific biological function through the variation in tumor cell types and expression forms. However, the expression of DDP4 in different tumor types and its specific mechanism remains unclear. In this review, we describe the structure of DPP4, summarize the recent research progress of its expression and potential mechanisms in common tumors, and discuss the development prospects of DPP4 inhibitors in tumor therapy. Although current research emphasizes the potential of DPP4 as a drug target, the incomplete understanding of its regulatory mechanisms impedes the discovery and development of new therapies against it. Further research on DPP4-related tumors is anticipated to promote its clinical application as a potential therapeutic target.
Collapse
Affiliation(s)
- Lu Sun
- Department of Ultrasound, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266035, China
| | - Yuhui Ma
- Department of Ultrasound, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266035, China
| | - Chenchen Geng
- Department of Ultrasound, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266035, China
| | - Xiaoqian Gao
- Department of Ultrasound, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266035, China
| | - Xinbing Li
- Department of Ultrasound, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266035, China
| | - Qi Ru
- Department of Ultrasound, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266035, China
| | - Shuzhen Zhu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
- Department of Clinical Laboratory, Qilu Hospital of Shandong University (Qingdao), Qingdao, Shandong, 266035, China.
| | - Ping Zhang
- Department of Ultrasound, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266035, China.
- Health Management Center, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266035, China.
| |
Collapse
|
43
|
Shu F, Yu J, Liu Y, Wang F, Gou G, Wen M, Luo C, Lu X, Hu Y, Du Q, Xu J, Xie R. Mast cells: key players in digestive system tumors and their interactions with immune cells. Cell Death Discov 2025; 11:8. [PMID: 39814702 PMCID: PMC11735678 DOI: 10.1038/s41420-024-02258-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/22/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025] Open
Abstract
Mast cells (MCs) are critical components of both innate and adaptive immune processes. They play a significant role in protecting human health and in the pathophysiology of various illnesses, including allergies, cardiovascular diseases and autoimmune diseases. Recent studies in tumor-related research have demonstrated that mast cells exert a substantial influence on tumor cell behavior and the tumor microenvironment, exhibiting both pro- and anti-tumor effects. Specifically, mast cells not only secrete mediators related to pro-tumor function such as trypsin-like enzymes, chymotrypsin, vascular endothelial cell growth factor and histamine, but also mediators related to anti-tumor progression such as cystatin C and IL-17F. This dual role of mast cells renders them an under-recognized but very promising target for tumor immunotherapy. Digestive system tumors, characterized by high morbidity and associated mortality rates globally, are increasingly recognized as a significant healthcare burden. This paper examines the influence of mast cell-derived mediators on the development of tumors in the digestive system. It also explores the prognostic significance of mast cells in patients with various gastrointestinal cancers at different stages of the disease. Additionally, the article investigates the interactions between mast cells and immune cells, as well as the potential relationships among intratumoral bacteria, immune cells, and mast cell within digestive system microenvironment. The aim is to propose new strategies for the immunotherapy of digestive system tumors by targeting mast cells.
Collapse
Affiliation(s)
- Feihong Shu
- Department of Endoscopy and Digestive System, Guizhou Provincial People's Hospital, Guiyang, China
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Jie Yu
- Department of Endoscopy and Digestive System, Guizhou Provincial People's Hospital, Guiyang, China
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Youjia Liu
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Fang Wang
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Guoyou Gou
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Min Wen
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Chen Luo
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Xianmin Lu
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Yanxia Hu
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Qian Du
- Department of Endoscopy and Digestive System, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jingyu Xu
- Guizhou Medical University, Guiyang, Guizhou, China
| | - Rui Xie
- Department of Endoscopy and Digestive System, Guizhou Provincial People's Hospital, Guiyang, China.
| |
Collapse
|
44
|
Lederer AK, Görrissen N, Nguyen TT, Kreutz C, Rasel H, Bartsch F, Lang H, Endres K. Exploring the effects of gut microbiota on cholangiocarcinoma progression by patient-derived organoids. J Transl Med 2025; 23:34. [PMID: 39789543 PMCID: PMC11716211 DOI: 10.1186/s12967-024-06012-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Recent research indicates a role of gut microbiota in development and progression of life-threatening diseases such as cancer. Carcinomas of the biliary ducts, the so-called cholangiocarcinomas, are known for their aggressive tumor biology, implying poor prognosis of affected patients. An impact of the gut microbiota on cholangiocarcinoma development and progression is plausible due to the enterohepatic circulation and is therefore the subject of scientific debate, however evidence is still lacking. This review aimed to discuss the suitability of complex cell culture models to investigate the role of gut microbiota in cholangiocarcinoma progression. MAIN BODY Clinical research in this area is challenging due to poor comparability of patients and feasibility reasons, which is why translational models are needed to understand the basis of tumor progression in cholangiocarcinoma. A promising approach to investigate the influence of gut microbiota could be an organoid model. Organoids are 3D cell models cultivated in a modifiable and controlled condition, which can be grown from tumor tissue. 3D cell models are able to imitate physiological and pathological processes in the human body and thus contribute to a better understanding of health and disease. CONCLUSION The use of complex cell cultures such as organoids and organoid co-cultures might be powerful and valuable tools to study not only the growth behavior and growth of cholangiocarcinoma cells, but also the interaction with the tumor microenvironment and with components of the gut microbiota.
Collapse
Affiliation(s)
- Ann-Kathrin Lederer
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, 55131, Mainz, Germany.
- Center for Complementary Medicine, Department of Medicine II, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, 79106, Freiburg, Germany.
| | - Nele Görrissen
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, 55131, Mainz, Germany
| | - Tinh Thi Nguyen
- Department of Psychiatry and Psychotherapy, University Medical Center Mainz, 55131, Mainz, Germany
- Institute of Molecular Biology (IMB), 55128, Mainz, Germany
| | - Clemens Kreutz
- Institute of Medical Biometry and Statistics (IMBI), Faculty of Medicine and Medical Center, 79106, Freiburg, Germany
| | - Hannah Rasel
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, 55131, Mainz, Germany
| | - Fabian Bartsch
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, 55131, Mainz, Germany
| | - Hauke Lang
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, 55131, Mainz, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center Mainz, 55131, Mainz, Germany
- Faculty of Computer Sciences and Microsystems Technology, University of Applied Sciences Kaiserslautern, 66482, Zweibrücken, Germany
| |
Collapse
|
45
|
Mai Z, Fu L, Su J, To KKW, Yang C, Xia C. Intra-tumoral sphingobacterium multivorum promotes triple-negative breast cancer progression by suppressing tumor immunosurveillance. Mol Cancer 2025; 24:6. [PMID: 39780177 PMCID: PMC11708301 DOI: 10.1186/s12943-024-02202-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Intratumor-resident bacteria represent an integral component of the tumor microenvironment (TME). Microbial dysbiosis, which refers to an imbalance in the bacterial composition and bacterial metabolic activities, plays an important role in regulating breast cancer development and progression. However, the impact of specific intratumor-resident bacteria on tumor progression and their underlying mechanisms remain elusive. METHODS 16S rDNA gene sequencing was used to analyze the cancerous and paracancerous tissues from breast cancer patients. The mouse models of bearing 4T1 cell tumors were employed to assess the influence of bacterial colonization on tumor growth. Tissue infiltration of regulatory T (Treg) cells and CD8+ T cells was evaluated through immunohistochemistry and flow cytometric analysis. Comparative metabolite profiling in mice tumors was conducted using targeted metabolomics. Differential genes of tumor cells stimulated by bacteria were analyzed by transcriptomics and validated by qPCR assay. RESULTS We found that Sphingobacterium displayed high abundance in cancerous tissues. Intra-tumoral colonization of Sphingobacterium multivorum (S. multivorum) promoted tumor progression in 4T1 tumor-bearing mice. Moreover, S. multivorum diminished the therapeutic efficacy of αPD-1 mAb, which was associated with the increase of regulatory T cell (Treg) infiltration, and decrese of the CD8+ T cell infiltration. Targeted metabolomics revealed a conspicuous reduction of propionylcarnitine in tumors colonized by S. multivorum Furthermore, the combination of metabolite propionylcarnitine and S. multivorum shown to suppress tumor growth compared that in S. multivorum alone in vivo. Mechanistically, S. multivorum promoted the secretion of chemokines CCL20 and CXCL8 from tumor cells. CCL20 secreted into the TME facilitated the recruitment of Treg cells and reduced CD8+ T cell infiltration, thus promoting tumor immune escape. CONCLUSIONS This study reveals S. multivorum suppresses immune surveillance within the TME, thereby promoting breast cancer progression.
Collapse
Affiliation(s)
- Zhikai Mai
- Foshan Maternity and Child Healthcare Hospital; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 515150, China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Jiyan Su
- Foshan Maternity and Child Healthcare Hospital; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 515150, China
| | - Kenneth K W To
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Chuansheng Yang
- Department of Breast, Thyroid and Head-Neck Surgery, Yuebei People's Hospital of Shantou University, Shaoguan, China
| | - Chenglai Xia
- Foshan Maternity and Child Healthcare Hospital; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 515150, China.
| |
Collapse
|
46
|
Xi C, Zhou J, Zheng X, Fu X, Xie M. Sodium aescinate-induced hepatotoxicity via ATF4/GSH/GPX4 axis-mediated ferroptosis. Sci Rep 2025; 15:1141. [PMID: 39774712 PMCID: PMC11706965 DOI: 10.1038/s41598-024-79723-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/12/2024] [Indexed: 01/11/2025] Open
Abstract
Sodium aescinate (SA), a natural plant extract with various bioactivities, is widely used to treat oedema and inflammation in clinics. However, adverse events, including liver injury, kidney injury, and phlebitis, have been reported in patients with SA in recent years. In this study, we used BALB/c mice and L02 cells to evaluate the role of ferroptosis in SA-induced liver injury. SA significantly increased AST, ALT, MDA and Fe2+, decreased GSH levels, and induced pathological changes in the liver in vivo. SA also reduced the viability of L02 cells and induced LDH release, intracellular cysteine reduction, GSH depletion, iron accumulation, ROS production, and lipid peroxidation, indicating that SA causes ferroptosis. In addition, SA inhibited transcriptional activity of activating transcription factor 4 (ATF4) and subsequently reduced the expression of the downstream genes xCT (solute carrier family 7a member 11, SLC7A11) and Cystathionine gamma-lyase (CTH) which play vital roles in GSH biosynthesis. Interestingly, the cytotoxic effects of SA were effectively attenuated by ATF4 overexpression, while they were significantly aggravated by ATF4 silencing. These results revealed that SA triggers hepatocyte ferroptosis by inhibiting the activity of ATF4, which causes an oxidative imbalance.
Collapse
Affiliation(s)
- Chen Xi
- Pharmaceutical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China
| | - Jie Zhou
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, Jiangxi, P.R. China.
| | - Xin Zheng
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, Jiangxi, P.R. China
| | - Xiaoyi Fu
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, Jiangxi, P.R. China
| | - Minjuan Xie
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, Jiangxi, P.R. China
| |
Collapse
|
47
|
Jing R, Wu N, Zhang Q, Liu J, Zhao Y, Zeng S, Wu S, Wu Y, Yi S. DPP4 promotes an immunoenhancing tumor microenvironment through exhausted CD8+ T cells with activating IL13-IL13RA2 axis in papillary thyroid cancer. Int Immunopharmacol 2025; 145:113760. [PMID: 39662266 DOI: 10.1016/j.intimp.2024.113760] [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: 10/11/2024] [Revised: 11/17/2024] [Accepted: 11/29/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Papillary thyroid cancer (PTC) is among the most prevalent forms of endocrine malignancy with a rapid rise in incidence rates worldwide; however, the composition and characteristics of its immune microenvironment is poorly understand. Here, this work investigated the precise function of Dipeptidyl peptidase 4 (DPP4) in tumor-infiltrated T cells within PTC by investigating its role in cytokine-mediated signaling pathways. METHODS TCGA and GEO data as well as human PTC specimens confirmed the expression of DPP4 in PTC. The CIBERSORT and TIMER tool were used to analyze the distribution of tumor-infiltrating immune cells in PTC. CD8+ T cells from PTC patient's peripheral blood were cultured and used in a three-dimensional model for direct co-culture with PTC tumors to investigate DPP4 function. RESULTS Bioinformatic analyses has uncovered a significant upregulation of DPP4, which enhances the survival and migration of PTC cells in vitro. DPP4 upregulation significantly correlated with advanced grades, stages, and poor progression-free survival. DPP4 influences immune function and the exhaustion of CD8+ T cells through the IL13-IL13RA2 axis. The inhibition of DPP4 reduces CD8+ T cell exhaustion and IL13 secretion, while also blocking the IL13-IL13RA2 axis, thereby promoting the mesenchymal-to-epithelial transition of PTC cells. CONCLUSION Blocking DPP4 leads to the conversion of exhausted CD8+ T cells with decreased IL13 level, resulting in downregulation of IL13RA2 to promote mesenchymal-to-epithelial transition of PTC cells. This highlights DPP4 as a potential therapeutic target, particularly between CD8+ T cells and PTC cells via IL13-IL13RA2 axis, and represents a novel avenue for combined immunotherapy in PTC.
Collapse
Affiliation(s)
- Ren Jing
- Department of Breast and Thyroid Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen 518116, PR China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, PR China
| | - Nan Wu
- Department of Breast and Thyroid Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen 518116, PR China
| | - Qian Zhang
- Respiratory Medicine, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen 518118, PR China
| | - Jinlin Liu
- Department of Clinical Laboratory, South China Hospital, Medical School, Shenzhen University, Shenzhen 518116, PR China
| | - Ying Zhao
- Department of Clinical Laboratory, South China Hospital, Medical School, Shenzhen University, Shenzhen 518116, PR China
| | - Shan Zeng
- Department of Pathology, South China Hospital, Medical School, Shenzhen University, Shenzhen 518116, PR China
| | - Shaojie Wu
- Department of Breast and Thyroid Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen 518116, PR China
| | - Yang Wu
- Department of Breast and Thyroid Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen 518116, PR China.
| | - Shijian Yi
- Department of Breast and Thyroid Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen 518116, PR China.
| |
Collapse
|
48
|
Lan T, Gao F, Cai Y, Lv Y, Zhu J, Liu H, Xie S, Wan H, He H, Xie K, Liu C, Wu H. The protein circPETH-147aa regulates metabolic reprogramming in hepatocellular carcinoma cells to remodel immunosuppressive microenvironment. Nat Commun 2025; 16:333. [PMID: 39747873 PMCID: PMC11696079 DOI: 10.1038/s41467-024-55577-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Metabolic reprogramming fuels cancer cell metastasis and remodels the immunosuppressive tumor microenvironment (TME). We report here that circPETH, a circular RNA (circRNA) transported via extracellular vesicles (EVs) from tumor-associated macrophages (TAMs) to hepatocellular carcinoma (HCC) cells, facilitates glycolysis and metastasis in recipient HCC cells. Mechanistically, circPETH-147aa, encoded by circPETH in an m6A-driven manner, promotes PKM2-catalyzed ALDOA-S36 phosphorylation via the MEG pocket. Furthermore, circPETH-147aa impairs anti-HCC immunity by increasing HuR-dependent SLC43A2 mRNA stability and driving methionine and leucine deficiency in cytotoxic CD8+ T cells. Importantly, through virtual and experimental screening, we find that a small molecule, Norathyriol, is an effective inhibitor that targets the MEG pocket on the circPETH-147aa surface. Norathyriol reverses circPETH-147aa-facilitated acquisition of metabolic and metastatic phenotypes by HCC cells, increases anti-PD1 efficacy, and enhances cytotoxic CD8+ T-cell function. Here we show that Norathyriol is a promising anti-HCC agent that contributes to attenuating the resistance of advanced HCC to immune checkpoint blocker (ICB) therapies.
Collapse
Affiliation(s)
- Tian Lan
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China.
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China.
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| | - Fengwei Gao
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yunshi Cai
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yinghao Lv
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jiang Zhu
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hu Liu
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Sinan Xie
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Haifeng Wan
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Haorong He
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Kunlin Xie
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Chang Liu
- Department of Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Wu
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China.
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China.
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
49
|
Zhang M, Guo R, Yuan Z, Wang H. Lipid Nanoparticle (LNP) -A Vector Suitable for Evolving Therapies for Advanced Hepatocellular Carcinoma (HCC). GLOBAL CHALLENGES (HOBOKEN, NJ) 2025; 9:2400217. [PMID: 39802046 PMCID: PMC11717671 DOI: 10.1002/gch2.202400217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/07/2024] [Indexed: 01/16/2025]
Abstract
Hepatocellular carcinoma (HCC) stands as the predominant form of primary liver cancer, characterized by a dismal prognosis. Therapeutic options for advanced HCC remain sparse, with efficacy significantly hampered by the emergence of drug resistance. In parallel with research into novel pharmacological agents, advances in drug delivery systems represent a promising avenue for overcoming resistance. Lipid nanoparticles (LNPs) have demonstrated considerable efficacy in the delivery of nucleic acid-based therapeutics and hold potential for broader applications in drug delivery. This review describes the development of LNPs tailored for HCC treatment and consolidates recent investigations using LNPs to target HCC.
Collapse
Affiliation(s)
- Mingxuan Zhang
- Department of Radiation OncologyCancer Center of Peking University Third HospitalPeking University Third HospitalHaidian, 49 Huayuan North RoadBeijing100191China
| | - Ruiping Guo
- Department of Radiation OncologyCancer Center of Peking University Third HospitalPeking University Third HospitalHaidian, 49 Huayuan North RoadBeijing100191China
| | - Zhuhui Yuan
- Department of Radiation OncologyCancer Center of Peking University Third HospitalPeking University Third HospitalHaidian, 49 Huayuan North RoadBeijing100191China
| | - Hao Wang
- Department of Radiation OncologyCancer Center of Peking University Third HospitalPeking University Third HospitalHaidian, 49 Huayuan North RoadBeijing100191China
| |
Collapse
|
50
|
Wu Y, Jiang X, Yu Z, Xing Z, Ma Y, Qing H. Mechanisms of Anti-PD Therapy Resistance in Digestive System Neoplasms. Recent Pat Anticancer Drug Discov 2025; 20:1-25. [PMID: 38305306 DOI: 10.2174/0115748928269276231120103256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 02/03/2024]
Abstract
Digestive system neoplasms are highly heterogeneous and exhibit complex resistance mechanisms that render anti-programmed cell death protein (PD) therapies poorly effective. The tumor microenvironment (TME) plays a pivotal role in tumor development, apart from supplying energy for tumor proliferation and impeding the body's anti-tumor immune response, the TME actively facilitates tumor progression and immune escape via diverse pathways, which include the modulation of heritable gene expression alterations and the intricate interplay with the gut microbiota. In this review, we aim to elucidate the mechanisms underlying drug resistance in digestive tumors, focusing on immune-mediated resistance, microbial crosstalk, metabolism, and epigenetics. We will highlight the unique characteristics of each digestive tumor and emphasize the significance of the tumor immune microenvironment (TIME). Furthermore, we will discuss the current therapeutic strategies that hold promise for combination with cancer immune normalization therapies. This review aims to provide a thorough understanding of the resistance mechanisms in digestive tumors and offer insights into potential therapeutic interventions.
Collapse
Affiliation(s)
- Yuxia Wu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Xiangyan Jiang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Zeyuan Yu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Zongrui Xing
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Yong Ma
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Huiguo Qing
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|