1
|
Li H, Chen H, Zhao T, Zhang W, Deng J, Xie W, Fan J, Lou H, Dong P, Han Z, Xing D, Mao S, Shen X, Xue X, Lu M. CD2AP shapes a stromal reduced tumor microenvironment and contributes to immunotherapy in gastric cancer. BMC Cancer 2025; 25:910. [PMID: 40399857 DOI: 10.1186/s12885-025-14248-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/29/2025] [Indexed: 05/23/2025] Open
Abstract
Gastric cancer (GC) ranks as the fifth most prevalent malignant tumor and stands as the fourth leading contributor to cancer-related fatalities on a global scale. The specific link between CD2 Associated Protein (CD2AP) expression and the tumor microenvironment (TME) remains unclear, and further exploration is needed to understand its potential role in immune response and as a target for immunotherapy in GC. Utilizing RNA sequencing data acquired from The Cancer Genome Atlas (TCGA) for a pan-cancer analysis, a comprehensive evaluation was carried out to determine the expression pattern and immunological involvement of CD2AP. Systematic association of CD2AP with immunological features within the stomach adenocarcinoma (STAD) TME was subsequently performed, encompassing factors like cancer immunity cycles, immune checkpoints, immunomodulators, tumor-infiltrating immune cells (TIICs). We found that CD2AP was enhanced expression in the TME of a variety of malignancies. CD2AP contributes to forming a stromal reduced TME in GC and improve the efficacy of immunotherapy. It was observed that patients with elevated levels of CD2AP, along with high scores on their CD4, CD20, and CD57 immune markers, tended to experience the most favorable prognosis. Furthermore, an IRS was constructed to accurately assess the prognosis of STAD patients. Since CD2AP was associated with the formation of stromal reduced TME in STAD, the expression of CD2AP can improve the effect of immunotherapy of STAD. CD2AP could emerge as a novel prognostic biomarker for STAD, offering a fresh avenue for molecular targeted therapy.
Collapse
Affiliation(s)
- Haoliang Li
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hua Chen
- Department of Radiation and Medical Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ting Zhao
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenqi Zhang
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jing Deng
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wangkai Xie
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jianing Fan
- School of Second Clinical Medical, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Han Lou
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Pingping Dong
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zheng Han
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Dong Xing
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Sunzhong Mao
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Xian Shen
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Xiangyang Xue
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Mingdong Lu
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
2
|
Gou YK, Zhou J, Liu P, Wang MY. Research progress on monocyte/macrophage in the development of gastric cancer. Future Oncol 2025:1-11. [PMID: 40351251 DOI: 10.1080/14796694.2025.2504334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 05/07/2025] [Indexed: 05/14/2025] Open
Abstract
Gastric cancer (GC) is diagnosed more than one million times each year and represents a major cause of cancer-related death worldwide. Although GC presents as a group of different types of disease, chronic inflammation has been strongly associated with tumorigenesis. Monocyte/macrophage play important roles in the development of inflammation and are vital components of the tumor microenvironment (TME). Monocyte/macrophage exert protumor and/or antitumor effects through the release of angiogenic and lymphangiogenic factors. Furthermore, tumor associated macrophages (TAMs) are emerging as key players in GC development. It is necessary to review and elucidate the roles of TAM subsets in GC and their molecular features. In this study, we focused on GC-related subsets of monocytes/macrophages and analyzed signaling related to TAMs in GC as well as the potential roles of these cells as therapeutic targets.
Collapse
Affiliation(s)
- Yuan-Kun Gou
- School of Medical Laboratory, Shandong Second Medical University, Weifang, Shandong, PR China
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, PR China
| | - Jie Zhou
- School of Medical Laboratory, Shandong Second Medical University, Weifang, Shandong, PR China
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, PR China
| | - Peng Liu
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, PR China
| | - Ming-Yi Wang
- School of Medical Laboratory, Shandong Second Medical University, Weifang, Shandong, PR China
- Department of Central Lab, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, PR China
| |
Collapse
|
3
|
Wu C, Sun L, Zhu W, Huang C, Zhu Z, Liu Z. A Golgi apparatus-related signature predicts the immune microenvironment and prognosis of gastric cancer. Genes Immun 2025:10.1038/s41435-025-00332-8. [PMID: 40335646 DOI: 10.1038/s41435-025-00332-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 01/01/2025] [Accepted: 04/25/2025] [Indexed: 05/09/2025]
Abstract
In recent years, numerous studies have provided evidence of the involvement of the Golgi apparatus (GA) in various stages of cancer development. Nonetheless, the specific impact of GA-related characteristics on gastric cancer (GC) progression remains ambiguous. We utilized LASSO and multivariate COX regression methods to develop a GA-associated risk score (GARS). The GARS is constructed from seven signature genes, which are highly expressed in tumors. In our research, we have found that GARS is an effective indicator for predicting the prognosis of GC, chemotherapy sensitivity, and immune therapy response. Patients in the low GARS group exhibit characteristics such as a good prognosis, increased sensitivity to immune therapy, 5-fluorouracil, and paclitaxel. Finally, our experimental results confirm that knocking down F2R significantly reduces the proliferation and migration abilities of GC cells. This study highlights the importance of GA characteristics in predicting the prognosis of GC and in developing personalized treatment strategies. The experimental findings on F2R offer valuable theoretical insights for the diagnosis and management of GC.
Collapse
Affiliation(s)
- Changlei Wu
- Department of Gastrointestinal Surgery, The Second Afffliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Liang Sun
- Department of Gastrointestinal Surgery, The Second Afffliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Wenjie Zhu
- Department of Gastrointestinal Surgery, The Second Afffliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Chao Huang
- Department of Gastrointestinal Surgery, The Second Afffliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhengming Zhu
- Department of Gastrointestinal Surgery, The Second Afffliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China.
| | - Zitao Liu
- Department of Gastrointestinal Surgery, The Second Afffliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China.
| |
Collapse
|
4
|
Liu M, Zou G, Lu M, Fu J, Chen H, Pan C, Liu HM, Fu L. Mechanism of Rabdosia rubescens extract against gastric cancer microenvironment by SIRT1/NF-κB/p53 pathway and promoting tumor-associated macrophage polarization. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119935. [PMID: 40345273 DOI: 10.1016/j.jep.2025.119935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/23/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The traditional action of Rabdosia rubescens (Hemsl.) H. Hara is heat-clearing and detoxifying, relieve sore throat, dissipate binds and disperse swelling. DLC, as an extract prepared from Rabdosiae Rubescentis Herba, could regulate the polarization of tumor associated macrophages (TAMs). For TAMs play an important role in the tumor microenvironment. It is worthy to further explore the mechanism of DLC on the polarized function of macrophages. AIM OF THE STUDY The aim of this study is to investigate the activity and molecular mechanisms of DLC on dissipating binds and dispersing swelling by modulating the gastric cancer microenvironment and macrophage polarization. MATERIALS AND METHODS We conducted comprehensive qualitative and quantitative chromatographic analyses to characterize the main components of DLC. To evaluate its anti-tumor effects, immunofluorescence, MTT assay, plate cloning, transcriptomics analysis, western blotting, and siRNA knockdown experiments were performed to assess DLC's action on gastric cancer cell proliferation. Additionally, we utilized Trypan blue staining, a THP-1 and MGC-803 co-culture model, flow cytometry, enzyme-linked immunosorbent assay (ELISA), and a mouse xenograft model with five distinct dosage groups to systematically investigate DLC's effects on macrophage polarization. RESULTS Key compounds in DLC were identified. The vivo tests demonstrated the tumor inhibition rate of the 5 g/kg DLC group reached 66.99 %, surpassing that of the 5-fluorouracil group (59.94 %). Mechanistically, DLC upregulated SIRT1 expression and suppressed NF-κB pathway, thereby preventing p65 from translocating into nuclear and modulating downstream p53/MDM2/USP7 signaling. Moreover, DLC enhanced M1 macrophage factors such as TNF-α, IL-6 while inhibiting M2 marker TGF-β, effectively repolarizing M2 TAMs toward an M1 phenotype. This effect was associated with suppressed protein expression of HIF-1α, p-p65, and p-PI3K. CONCLUSION This study provides insights into DLC's mechanisms in regulating tumor microenvironment remodeling and promoting macrophage polarization toward an anti-tumor phenotype. These results provide a solid basis for DLC's potential clinical treament in gastric cancer, highlighting its promise as a natural therapeutic agent.
Collapse
Affiliation(s)
- Mengran Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Guona Zou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Mengyao Lu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Jiayue Fu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Han Chen
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Chengxue Pan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Hong-Min Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China.
| | - Ling Fu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
5
|
Kirsche L, He J, Müller A, Leary P. MARMOT: A multifaceted R pipeline for analysing spectral flow cytometry data from subcutaneously growing murine gastric organoids. J Immunol Methods 2025; 540:113854. [PMID: 40122453 DOI: 10.1016/j.jim.2025.113854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
The analysis of murine immune cell types is a critical component of immunological research, necessitating precise and reproducible methodologies. Here, we present a comprehensive protocol and pipeline designed to streamline the process from murine gastric organoid transplant sample preparation to figure generation. This pipeline includes a detailed staining panel tailored for murine immune cells, ensuring accurate and comprehensive identification of various cell types. Additionally, it integrates an R-based analysis script (MARMOT Pipeline), encompassing data processing and visualisation. A key feature of this pipeline is its ability to produce publication-quality figures with minimal direct R coding, thus making advanced data analysis accessible to researchers with limited programming experience. Additionally, figures can be customised using a provided Shiny application. This approach both enhances the efficiency of data analysis and enables the reproducibility required for high-quality scientific research.
Collapse
Affiliation(s)
- Lydia Kirsche
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Jiazhuo He
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland; Comprehensive Cancer Center Zürich, Zürich, Switzerland
| | - Peter Leary
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland; Functional Genomics Center Zürich, University of Zürich/ETHZ, Zürich, Switzerland.
| |
Collapse
|
6
|
Zhang F, Zhou C, Wang X, Liu Y, Hou Y, Niu L. INHBA, transcriptionally activated by SPI1, facilitates gastric cancer progression by inducing macrophage recruitment and M2 polarization via activating the TGF-β signaling to increase CCL2. Pathol Res Pract 2025; 269:155920. [PMID: 40132395 DOI: 10.1016/j.prp.2025.155920] [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: 12/26/2024] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025]
Abstract
Tumor-associated macrophages (TAMs) are associated with the occurrence, development, and poor prognosis of human cancers. Inhibin beta A subunit (INHBA) is found to be aberrantly upregulated in gastric cancer (GC). However, whether INHBA is involved in macrophage recruitment and M2 polarization is unclear. Herein, INHBA expression was increased in GC tumor tissues and cells. INHBA expression was positively correlated with macrophage infiltration and M2 macrophage markers. Knockdown of INHBA in GC cells suppressed macrophage recruitment and M2 polarization by downregulaitng CCL2 expression and secretion. Mechanistic assays showed that SPI1 could bind to INHBA and transcriptionally activate its expression. SPI1 promoted macrophage recruitment and M2 polarization by upregulating INHBA expression. Moreover, SPI1 induced CCL2 expression by regulating INHBA in GC cells. INHBA upregulated CCL2 expression by activating the TGF-β signaling. Furthermore, SPI1-induced macrophages facilitated cell proliferation, migration, and invasion by increasing INHBA expression. INHBA-induced macrophages promoted cell proliferation, migration, and invasion by inducing CCL2 expression. Additionally, knockdown of INHBA inhibited tumor growth in vivo. In conclusion, SPI1 induces the macrophage recruitment and M2 polarization by transcriptionally regulating INHBA to activating the TGF-β signaling, thereby upregulating CCL2 expression and then contributing to GC cell malignant progression. Targeting SPI1/INHBA/CCL2 axis might be a promising therapeutic strategy for GC and potentially used for cancer immunotherapy.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Congya Zhou
- Department of Radiotherapy, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Xifang Wang
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Ying Liu
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Yinyin Hou
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Lu Niu
- Department of Gastroenterology, Shaanxi Provincial People's Hospital, Xi'an 710068, China.
| |
Collapse
|
7
|
Saadh MJ, Ahmed HH, Kareem RA, Bishoyi AK, Roopashree R, Shit D, Arya R, Joshi KK, Sameer HN, Yaseen A, Athab ZH, Adil M. The hidden messengers: Tumor microenvironment-derived exosomal ceRNAs in gastric cancer progression. Pathol Res Pract 2025; 269:155905. [PMID: 40073646 DOI: 10.1016/j.prp.2025.155905] [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: 11/26/2024] [Revised: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
The tumor microenvironment (TME) plays a crucial role in the development and progression of gastric cancer (GC). The TME comprises a network of cancer cells, immune cells, fibroblasts, endothelial cells, and extracellular matrix components, which provide a supportive niche for cancer cells. This study investigates the role of TME-derived exosomal competitive endogenous RNAs (ceRNAs), particularly long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), as major regulating agents in GC development. Exosomal ceRNAs control gene expression across several TME components, amplifying cancer hallmarks like cell proliferation, invasion, metastases, and chemoresistance. They promote dynamic interplay between cancer cells and adjacent stromal cells, enabling tumor development through immune suppression, angiogenesis, and epithelial-mesenchymal transition (EMT). Exosomal ceRNAs can modify the TME, creating a pro-tumorigenic milieu and preparing cancer cells to avoid immunological responses, defy death, and adapt to therapeutic pressures. This review highlights the understudied interactions between the TME and exosomal ceRNAs in gastric cancer and emphasizes their potential utility as diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan
| | | | | | - Ashok Kumar Bishoyi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat 360003, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Debasish Shit
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab 140401, India
| | - Renu Arya
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, Uttarakhand 248002, India; Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | | |
Collapse
|
8
|
Li J, Li Z, Wang K. Targeting angiogenesis in gastrointestinal tumors: strategies from vascular disruption to vascular normalization and promotion strategies angiogenesis strategies in GI tumor therapy. Front Immunol 2025; 16:1550752. [PMID: 40330478 PMCID: PMC12052729 DOI: 10.3389/fimmu.2025.1550752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/12/2025] [Indexed: 05/08/2025] Open
Abstract
Angiogenesis plays a critical role in the progression of gastrointestinal (GI) tumors, making it an important therapeutic target. This review explores recent advancements in targeting angiogenesis for GI tumor therapy, highlighting strategies that range from vascular disruption to vascular promotion. The biological foundation of tumor angiogenesis is discussed, with a focus on the molecular mechanisms that regulate this process, including key players such as VEGF, HIFs, and non-coding RNAs. Current therapeutic strategies, including anti-angiogenic agents, vascular normalization approaches, and emerging vascular promotion therapies, are analyzed for their clinical applications and limitations. Additionally, the review examines combination strategies that integrate anti-angiogenic therapy with chemotherapy, immunotherapy, and other modalities to enhance efficacy and overcome resistance. Despite significant progress, challenges such as drug resistance, tumor heterogeneity, and adverse effects remain. Future research directions emphasize the discovery of novel molecular targets, development of personalized treatments, and innovative combination therapies to optimize outcomes for patients with GI tumors. This comprehensive review provides a foundation for advancing angiogenesis-targeted therapies in GI cancer treatment.
Collapse
Affiliation(s)
- Jiajia Li
- School of Rehabilitation Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Keliang Wang
- Department of Gastroenterology, Ningbo No.2 Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
9
|
Huang H, Fang L, Zhu C, Lv J, Xu P, Chen Z, Zhang Z, Wang J, Wang W, Xu Z. YBX1 promotes 5-Fluorouracil resistance in gastric cancer via m5C-dependent ATG9A mRNA stabilization through autophagy. Oncogene 2025:10.1038/s41388-025-03411-2. [PMID: 40251390 DOI: 10.1038/s41388-025-03411-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/20/2025]
Abstract
5-Fluorouracil (5-FU) is a first-line chemotherapeutic agent for advanced gastric cancer (GC). However, its clinical efficacy is often undermined by the development of chemoresistance. Aberrant activation of oncogenic pathways, including autophagy, has been implicated in 5-FU resistance. Epigenetic modifications, such as 5-methylcytosine (m5C), are also recognized to modulate autophagy and contribute to chemoresistance, though the underlying molecular mechanisms remain poorly understood. In this study, we discovered that YBX1, an m5C reader protein, was significantly upregulated in 5-FU-resistant GC cell lines and patient tissues. Both in vitro and in vivo experiments demonstrated that YBX1 promoted autophagy in GC cells, thereby enhancing 5-FU resistance. Mechanistically, the transcription factor MAZ was found to bind to the YBX1 promoter, driving its transcriptional upregulation. YBX1, in turn, stabilized ATG9A mRNA via NSUN2-mediated m5C modification, thereby enhancing autophagic activity and conferring chemoresistance. Clinically, elevated YBX1 expression correlated with poor prognosis in patients with advanced GC undergoing 5-FU-based chemotherapy. These findings establish YBX1 as a key regulator of autophagy and 5-FU resistance in GC and highlight its potential as a novel therapeutic target for overcoming 5-FU resistance.
Collapse
Affiliation(s)
- Hongxin Huang
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Lang Fang
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Chuming Zhu
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Jialun Lv
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Penghui Xu
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Zetian Chen
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Zhijun Zhang
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Jihuan Wang
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Weizhi Wang
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| | - Zekuan Xu
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- Institute for Gastric Cancer Research, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
10
|
Fan S, Tang Y, Cao J, Peng R, Su B, Tu D, Yu W, Chen C, Wang S, Jin S, Jiang G, Zhang C, Bai D. Integrative Analysis the Role of ENG as a Metabolic and Macrophage-Related Gene in Hepatocellular Carcinoma. Biochem Genet 2025:10.1007/s10528-025-11098-z. [PMID: 40244558 DOI: 10.1007/s10528-025-11098-z] [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: 10/05/2024] [Accepted: 04/03/2025] [Indexed: 04/18/2025]
Abstract
High levels of M2 macrophages often correlate with poor prognosis. Endoglin (ENG) is a potential target for anti-angiogenesis therapy in various cancers, but the link between M2 macrophages and metabolism-related genes (MRGs) in hepatocellular carcinoma (HCC) is unclear. We employed cibersort analysis to identify genes associated with M2 macrophages and metabolic reprogramming in HCC, utilizing data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. TCGA data were stratified basis on ENG expression levels, and the relationships between ENG and relevant genes were assessed alongside clinical features. Furthermore, we validated ENG expression in HCC tissues and its correlation with M2 macrophages via qRT-PCR, Western blotting (WB), and immunohistochemistry (IHC). Patients with high ENG expression presented superior overall survival (OS) and longer progression-free survival (PFS). Univariate and multivariate regression analyses identified ENG as an independent prognostic predictor. Moreover, GSEA, GO, and KEGG analyses suggested a correlation between ENG-related gene expression and immunity, particularly TAMs. Additionally, ENG was found to reshape the tumor microenvironment (TME) of HCC and influence the response to immunotherapy. Single-cell analysis revealed the differential expression and distribution of ENG in the TME. In vitro experiments demonstrated lower ENG expression in HCC tissues than in paracancerous tissues, with a concomitant correlation with M2 macrophages. ENG emerges as a novel predictive marker for HCC, could reshap the TME and impacts the response to immunotherapy and provides a fresh perspective for investigating combined immunotherapy targeting MRGs in HCC.
Collapse
Affiliation(s)
- Songsong Fan
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
| | - Yuhong Tang
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
| | - Jun Cao
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
| | - Rui Peng
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
| | - Bingbing Su
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
| | - Daoyuan Tu
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
| | - Weidi Yu
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
| | - Chen Chen
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
| | - Shunyi Wang
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
| | - Shengjie Jin
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
| | - Guoqing Jiang
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
| | - Chi Zhang
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
| | - Dousheng Bai
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China.
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225000, China.
| |
Collapse
|
11
|
Zhao LL, Liu YJ, Guo QJ, Yan N, Yang J, Han JQ, Xie XH, Luo YS. TPM4 influences the initiation and progression of gastric cancer by modulating ferroptosis via SCD1. Clin Exp Med 2025; 25:115. [PMID: 40214825 PMCID: PMC11991984 DOI: 10.1007/s10238-025-01629-8] [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: 01/31/2025] [Accepted: 03/11/2025] [Indexed: 04/14/2025]
Abstract
Gastric cancer (GC) is a deadly disease with poor prognosis and few treatment options. Tropomyosin 4 (TPM4) is an actin-binding protein that stabilizes the cytoskeleton of cells and has an unclear role in GC. This study aimed to elucidate the role and underlying mechanisms of TPM4 in GC pathogenesis. The expression and diagnostic and prognostic value of TPM4 in GC were analyzed using bioinformatics. A nomogram based on TPM4 expression was created and validated with an external cohort. TPM4-knockdown GC cells and xenograft models in nude mice were used to study the function of TPM4 in vitro and in vivo. Proteomic and rescue experiments confirmed the regulatory effect of TPM4 on stearoyl-CoA desaturase 1 (SCD1) in GC. Immunohistochemistry verified the expression and correlation of the TPM4 and SCD1 proteins in GC tissues. Our study identified TPM4 as an oncogene in GC, suggesting its potential diagnostic and prognostic value. The TPM4-based nomogram showed potential prognostic value for clinical use. TPM4 knockdown inhibited GC cell proliferation, induced ferroptosis, and slowed tumor growth in vivo, which is achieved by inhibiting SCD1 expression. Immunohistochemical analysis of GC tissues revealed elevated expression levels of both TPM4 and SCD1 proteins, with a positive correlation observed between their expression. TPM4 is a promising target for new diagnostic, prognostic, and therapeutic strategies for GC. Downregulation of TPM4 inhibits GC cell growth and induces ferroptosis by suppressing SCD1 expression.
Collapse
Affiliation(s)
- Ling-Lin Zhao
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China
- Qinghai Provincial People's Hospital, Xining, 810000, China
| | - Yu-Jun Liu
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, 810001, China
| | - Qi-Jing Guo
- Department of Oncology, Air Force Medical Center, PLA, Beijing, 100142, China
| | - Nan Yan
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China
| | - Jie Yang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China
| | - Jing-Qi Han
- Department of Pathology, Affiliated Hospital of Qinghai University, Xining, 810001, China
| | - Xiao-Hong Xie
- Qinghai Provincial People's Hospital, Xining, 810000, China
| | - Yu-Shuang Luo
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China.
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, 810001, China.
| |
Collapse
|
12
|
Chu C, Liu B, Zhang Y, Xu Z, Wang B, Chin KL. MicroRNA-668-3p Mediates Macrophage M2 Polarization by Targeting NFKBIA to Affect Gastric Cancer Cell Proliferation and Migration. TOHOKU J EXP MED 2025; 265:173-182. [PMID: 39505541 DOI: 10.1620/tjem.2024.j115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Macrophage polarization is implicated in the pathological mechanism of gastric cancer (GC). This study investigated how the miR-668-3p/ Nuclear factor kappa B inhibitor alpha (NFKBIA) axis drives macrophage polarization to contribute to GC progression. Inhibitors or shRNA were used to interfere with the expression of miR-668-3p or NFKBIA in the GC cell line. Subsequently, CCK-8, EdU, wound healing, and transwell assays were used to assess the biological behavior of the GC cells. Bioinformatics analysis predicted the target connection between miR-668-3p and NFKBIA, and a dual luciferase reporter gene experiment confirmed this relationship. After THP-1 macrophages were co-cultured with the supernatant of transfected GC cells, the M1 and M2 macrophage phenotypes were determined. Subsequently, these THP-1 macrophages were co-cultured with GC cells using the Transwell, and the biological behaviors of the GC cells were determiend. miR-668-3p inhibitor suppressed proliferation, invasion and migration of GC cells. The phenotype of M1 macrophage (IL-1β, TNF-α and IL-6) was boosted yet the phenotype of M2 macrophage (CD206, Fizz1 and IL-10) was declined by miR-668-3p inhibitor. NFKBIA was the target gene of miR-668-3p and it reversed the effects of miR-668-3p inhibitor on macrophage polarization and biological behaviors of the GC cells.miR-668-3p suppressed NFKBIA in GC cells to mediate M2 polarization of macrophages, thereby facilitating the tumorigenesis of GC.
Collapse
Affiliation(s)
- Chenghao Chu
- Department of General Surgery, Anqing First People's Hospital
- Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah
| | - Bin Liu
- Department of General Surgery, Anqing First People's Hospital
- Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah
| | - Yongwei Zhang
- Department of General Surgery, Anqing First People's Hospital
- Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah
| | - Zhangxuan Xu
- Department of General Surgery, Anqing First People's Hospital
| | - Bin Wang
- Department of General Surgery, Anqing First People's Hospital
| | - Kai Ling Chin
- Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah
- Borneo Medical and Health Research Centre, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah
| |
Collapse
|
13
|
Santos-Sousa DC, da Rosa S, Filippi-Chiela E. Molecular signatures of cellular senescence in cancer: a critical review of prognostic implications and therapeutic opportunities. Mech Ageing Dev 2025; 225:112052. [PMID: 40120861 DOI: 10.1016/j.mad.2025.112052] [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: 12/16/2024] [Revised: 03/01/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Cellular senescence is a state of permanent loss of proliferative capacity. Therefore, cells that reach a senescent state prevent tumor initiation, acting as an anti-tumor mechanism. However, despite not being proliferative, senescent cells have high secretory activity, constituting the Senescence-Associated Secretory Phenotype (SASP). SASP includes thousands of soluble molecules and extracellular vesicles, through which senescent cells can affect other cells and the extracellular matrix. In advanced tumors, the enrichment of senescent cells can have anti- or pro-tumor effects depending on features like SASP composition, tumor microenvironment (TME) composition, the anatomic site, histopathologic characteristics of malignancy, and tumor molecular background. We reviewed the studies assessing the impact of the senescence status, measured by mRNA or lncRNA molecular signatures, in the prognosis and other clinically relevant information in cancer, including anti-tumor immunity and response to therapy. We discussed the pros and cons of different strategies to define those molecular signatures and the main limitations of the studies. Finally, we also raised clinical challenges regarding the crossroad between cellular senescence and cancer prognosis, including some therapeutic opportunities in the field.
Collapse
Affiliation(s)
- Débora C Santos-Sousa
- Center of Biotechnology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 91501-970, Brazil; Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul 90035-903, Brazil.
| | - Solon da Rosa
- Center of Biotechnology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 91501-970, Brazil; Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul 90035-903, Brazil.
| | - Eduardo Filippi-Chiela
- Center of Biotechnology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 91501-970, Brazil; Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul 90035-903, Brazil; Department of Morphological Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90050-170, Brazil.
| |
Collapse
|
14
|
Zhang Y, Wang B, Chen J, Li T. Role of exosomal miRNAs and macrophage polarization in gastric cancer: A novel therapeutic strategy. Eur J Pharmacol 2025; 990:177268. [PMID: 39805486 DOI: 10.1016/j.ejphar.2025.177268] [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: 10/04/2024] [Revised: 01/09/2025] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
Gastric cancer (GC) is one of the most common gastrointestinal cancers worldwide, with consistently high morbidity and mortality rates and poor prognosis. Most patients are diagnosed at an advanced stage due to the lack of specific presentation in the early stages. Exosomes are a class of extracellular vesicles (EVs) widely found in body fluids and can release genetic material or multiple proteins to facilitate intercellular communication. In recent years, exosomal miRNAs have gained attention for their role in various cancers. These exosomal miRNAs can impact GC development and progression by targeting specific genes or influencing signaling pathways and cytokines involved in Angiogenesis, epithelial-mesenchymal transition (EMT), drug resistance, and immune regulation. They show great potential in terms of diagnosis, prognosis, and treatment of GC. Notably, the gastrointestinal tract has the largest number of macrophages, which play a significant role in GC progression. Tumor-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment (TME) and can influence macrophage programming through various mediators, including macrophage polarization. Macrophage polarization is involved in inflammatory responses and significantly impacts the GC process.
Collapse
Affiliation(s)
- Yun Zhang
- School of Clinical Medicine, Ningxia Medical University, Ningxia, China; General Hospital of Ningxia Medical University, Ningxia, China
| | - Baozhen Wang
- School of Clinical Medicine, Ningxia Medical University, Ningxia, China; General Hospital of Ningxia Medical University, Ningxia, China
| | - Jing Chen
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China.
| | - Tao Li
- Department of Surgical Oncology, Tumor Hospital, The General Hospital of Ningxia Medical University, Ningxia, China.
| |
Collapse
|
15
|
He Y, Hong Q, Chen S, Zhou J, Qiu S. Reprogramming tumor-associated macrophages in gastric cancer: a pathway to enhanced immunotherapy. Front Immunol 2025; 16:1558091. [PMID: 40098971 PMCID: PMC11911521 DOI: 10.3389/fimmu.2025.1558091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Gastric cancer (GC) remains a significant global health concern due to its poor prognosis and limited therapeutic options, particularly in advanced stages. Tumor microenvironment (TME), particularly tumor-associated macrophages (TAMs), plays a key role in tumor progression, immune evasion, and therapy resistance. TAMs exhibit plasticity, shifting between pro-inflammatory M1 and immunosuppressive M2 phenotypes, with the latter predominating in GC and contributing to poor outcomes. Recent therapeutic advancements focus on targeting TAMs, including inhibiting M2 polarization, reprogramming TAMs to M1 phenotypes, and combining TAM-targeted approaches with immune checkpoint inhibitors. Innovations in nanotechnology, metabolic reprogramming, and targeting key pathways such as interleukin-6 and C-C motif ligand 2/C-C motif chemokine receptor 2 further enhance these strategies. However, challenges remain, including the spatial and functional heterogeneity of TAMs within the TME and the need for selective targeting to avoid disrupting immune homeostasis. Ongoing research on TAM origins, functions, and interactions within the TME is crucial for developing precise and effective therapies. These advances hold promise not only for improving outcomes in GC but also for addressing other cancers with similarly complex microenvironments.
Collapse
Affiliation(s)
| | | | | | | | - Shengliang Qiu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang
Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
16
|
Handschin C, Shalhoub H, Mazet A, Guyon C, Dusserre N, Boutet-Robinet E, Oliveira H, Guillermet-Guibert J. Biotechnological advances in 3D modeling of cancer initiation. Examples from pancreatic cancer research and beyond. Biofabrication 2025; 17:022008. [PMID: 40018875 DOI: 10.1088/1758-5090/adb51c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 02/12/2025] [Indexed: 03/01/2025]
Abstract
In recent years, biofabrication technologies have garnered significant attention within the scientific community for their potential to create advancedin vitrocancer models. While these technologies have been predominantly applied to model advanced stages of cancer, there exists a pressing need to develop pertinent, reproducible, and sensitive 3D models that mimic cancer initiation lesions within their native tissue microenvironment. Such models hold profound relevance for comprehending the intricacies of cancer initiation, to devise novel strategies for early intervention, and/or to conduct sophisticated toxicology assessments of putative carcinogens. Here, we will explain the pivotal factors that must be faithfully recapitulated when constructing these models, with a specific focus on early pancreatic cancer lesions. By synthesizing the current state of research in this field, we will provide insights into recent advances and breakthroughs. Additionally, we will delineate the key technological and biological challenges that necessitate resolution in future endeavors, thereby paving the way for more accurate and insightfulin vitrocancer initiation models.
Collapse
Affiliation(s)
- C Handschin
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - H Shalhoub
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
| | - A Mazet
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - C Guyon
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - N Dusserre
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - E Boutet-Robinet
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - H Oliveira
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - J Guillermet-Guibert
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| |
Collapse
|
17
|
Khizar H, Ali K, Wang J. From silent partners to potential therapeutic targets: macrophages in colorectal cancer. Cancer Immunol Immunother 2025; 74:121. [PMID: 39998578 PMCID: PMC11861851 DOI: 10.1007/s00262-025-03965-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/30/2025] [Indexed: 02/27/2025]
Abstract
Cancer cells grow and survive in the tumor microenvironment, which is a complicated process. As a key part of how colorectal cancer (CRC) progresses, tumor-associated macrophages (TAMs) exhibit a double role. Through angiogenesis, this TAM can promote the growth of cancers. Although being able to modify and adjust immune cells is a great advantage, these cells can also exhibit anti-cancer properties including direct killing of cancer cells, presenting antigens, and aiding T cell-mediated responses. The delicate regulatory mechanisms between the immune system and tumors are composed of a complex network of pathways regulated by several factors including hypoxia, metabolic reprogramming, cytokine/chemokine signaling, and cell interactions. Decoding and figuring out these complex systems become significant in building targeted treatment programs. Targeting TAMs in CRC involves disrupting chemokine signaling or adhesion molecules, reprogramming them to an anti-tumor phenotype using TLR agonists, CD40 agonists, or metabolic modulation, and selectively removing TAM subsets that promote tumor growth. Multi-drug resistance, the absence of an accurate biomarker, and drug non-specificity are also major problems. Combining macrophage-targeted therapies with chemotherapy and immunotherapy may revolutionize treatment. Macrophage studies will advance with new technology and multi-omics methodologies to help us understand CRC and build specific and efficient treatments.
Collapse
Affiliation(s)
- Hayat Khizar
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Kamran Ali
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jianwei Wang
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, 2nd Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310009, China.
| |
Collapse
|
18
|
Hu Y, Du Y, Qiu Z, Mao P, Da M. Identification and validation VAT1 in gastric cancer through bioinformatics and experimental analysis. Int Immunopharmacol 2025; 148:114047. [PMID: 39832459 DOI: 10.1016/j.intimp.2025.114047] [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: 09/15/2024] [Revised: 12/22/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
This study investigated the expression pattern of Vesicular Amine Transporter 1 (VAT1) in gastric cancer (GC) and its impact on prognosis, alongside evaluating its potential as a biomarker for immunotherapy and chemotherapy. Analysis of transcriptomic data, supported by experimental validation, revealed that VAT1 is highly expressed in GC and is associated with poor prognosis. Kaplan-Meier and ROC analyses demonstrated VAT1's potential in GC diagnosis, while multivariate analysis confirmed its role as an independent risk factor. Gene set enrichment analysis indicated that VAT1 plays a role in regulating the MAPK signaling pathway and epithelial-mesenchymal transition (EMT) in GC. Immune infiltration analysis showed a positive correlation between VAT1 and immune cells, particularly macrophages, and a negative correlation with chemotherapy sensitivity. In vitro and in vivo experiments further confirmed VAT1's critical role in promoting GC cell proliferation and inhibiting apoptosis. Overall, VAT1 holds significant value not only in GC diagnosis and prognosis but also as a potential target for immunotherapy and overcoming drug resistance.
Collapse
Affiliation(s)
- Yongli Hu
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou 730000, China; Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, China.
| | - Yan Du
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou 730000, China.
| | - Zhisheng Qiu
- Department of Oncology Surgery, Gansu Provincial Hospital, Lanzhou 730000, China.
| | - Pengxue Mao
- Department of General Surgery, Minle County People's Hospital, Gansu Province 734500, China.
| | - Mingxu Da
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou 730000, China; Department of Oncology Surgery, Gansu Provincial Hospital, Lanzhou 730000, China.
| |
Collapse
|
19
|
Chen J, Zeng A, Yu Y, Liao L, Huang S, Sun S, Wu W. Deciphering the risk of developing liver cancer following gastric cancer diagnosis with genetic evidence: a Mendelian randomization analysis in an East Asian population. Discov Oncol 2025; 16:166. [PMID: 39937324 PMCID: PMC11822172 DOI: 10.1007/s12672-025-01938-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 02/05/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Liver cancer is a common second primary cancer in gastric cancer patients, but whether a gastric cancer diagnosis contributes to the development of second primary liver cancer remains contentious. This study aims to utilize Mendelian randomization (MR) analysis to investigate the potential causal relationship between gastric cancer and second primary liver cancer from a genetic perspective. METHODS We extracted single nucleotide polymorphism for gastric cancer and liver cancer in the East Asian population from the Genome-Wide Association Studies database as instrumental variables and employed univariate and multivariate MR analysis to evaluate the causal relationship between gastric cancer and liver cancer. The robustness of the findings was ensured through heterogeneity and sensitivity analyses. RESULTS Univariate MR analysis revealed that genetic susceptibility to gastric cancer in the East Asian population was significantly associated with an increased risk of liver cancer [Inverse-variance weighted (IVW): OR = 1.252, 95% CI 1.076-1.457, P = 0.004]. Multivariate MR analysis indicated that after adjusting for confounding factors, the significant positive causal relationship between gastric cancer and liver cancer remained robust (all P < 0.05). Furthermore, no causal relationship was observed between liver cancer diagnosis and the development of gastric cancer in the East Asian population (IVW: OR = 1.111, 95% CI 0.936-1.318, P = 0.228). CONCLUSION Genetic prediction results suggest that gastric cancer survivors might face an increased risk of developing second primary liver cancer, implying the potential value of routine liver cancer screening for gastric cancer survivors.
Collapse
Affiliation(s)
- Jiansheng Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
- National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Aiming Zeng
- Department of Clinical Laboratory, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Yunzhe Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
| | - Liqun Liao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
| | - Siwei Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
| | - Sida Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China.
| | - Weijie Wu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China.
- Department of Digestive Endoscopy Center, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, 134 East Street, Fuzhou, 350001, China.
| |
Collapse
|
20
|
Liu R, Liu Y, Huang W, Chen P, Cheng Y. An anoikis-related signature predicts prognosis and immunotherapy response in gastrointestinal cancers. Front Immunol 2025; 16:1477913. [PMID: 39981252 PMCID: PMC11839610 DOI: 10.3389/fimmu.2025.1477913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 01/21/2025] [Indexed: 02/22/2025] Open
Abstract
Background Gastrointestinal (GI) cancers have high incidence rates and mortality rates. Anoikis is a special type of cell apoptosis, and anoikis resistance has been reported to be associated with tumor malignancy. We aimed to explore the roles of anoikis-related genes (ARGs) in the GI cancer prognosis. Methods We extracted RNA sequencing and clinical data from The Cancer Genome Atlas and Gene Expression Omnibu databases for patients with esophageal cancer, gastric cancer, colon cancer and rectal cancer and identified ARGs from GeneCards and Harmonizome. Anoikis-related patterns were identified via unsupervised clustering analysis. We constructed a prognostic signature (Anoscore) based on prognostic ARGs through univariate, LASSO, and multivariate Cox regression analyses. The model was validated and evaluated using Kaplan-Meier analysis, receiver operating characteristic curves, univariate Cox regression analysis, multivariate Cox regression analysis, column charts, and calibration curves. We also performed a single-cell sequencing analysis of candidate genes via TISCH2. A correlation analysis between the Anoscore, the tumor microenvironment and drug sensitivity was conducted in GI cancers. The expression and function of some candidate genes were validated in vitro. Results In terms of prognostic ARGs, two anoikis-related patterns, ARG clusters A and B, were identified. ARG cluster B had a worse prognosis than did ARG cluster A. Subsequently, the Anoscore was developed as an independent prognostic factor. It demonstrated the robust predictive capability for the prognosis of patients with GI cancers. Notably, patients with high Anoscores exhibited poor outcomes. In addition, we established a nomogram (Ano-nomogram) based on the Anoscore and clinicopathological factors of patients to predict the 3-year and 5-year survival probabilities. Moreover, patients with high Anoscores had higher levels of immune cell infiltration and higher immune checkpoint expression. The drug sensitivity analysis revealed that patients with high or low Anoscores were sensitive to different chemotherapies and targeted drugs. S100A11 and TLR3, representative candidate genes, exhibited different expression patterns and biological functions. Conclusion This study highlighted the significant potential of the Anoscore in predicting prognosis and guiding the selection of personalized therapeutic regimens for patients with GI cancers.
Collapse
Affiliation(s)
- Ruyi Liu
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Laboratory of Malignant Tumor Precision Treatment, Jinan, China
- Shandong Provincial Engineering Research Center for Tumor Precision Treatment, Jinan, China
- Cancer Institute of Shandong University, Jinan, China
- Neutron Medical Center, Qilu Hospital of Shandong University, Jinan, China
| | - Yuchen Liu
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Laboratory of Malignant Tumor Precision Treatment, Jinan, China
- Shandong Provincial Engineering Research Center for Tumor Precision Treatment, Jinan, China
- Cancer Institute of Shandong University, Jinan, China
- Neutron Medical Center, Qilu Hospital of Shandong University, Jinan, China
| | - Weicheng Huang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Laboratory of Malignant Tumor Precision Treatment, Jinan, China
- Shandong Provincial Engineering Research Center for Tumor Precision Treatment, Jinan, China
- Cancer Institute of Shandong University, Jinan, China
- Neutron Medical Center, Qilu Hospital of Shandong University, Jinan, China
| | - Pengxiang Chen
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Laboratory of Malignant Tumor Precision Treatment, Jinan, China
- Shandong Provincial Engineering Research Center for Tumor Precision Treatment, Jinan, China
- Cancer Institute of Shandong University, Jinan, China
- Neutron Medical Center, Qilu Hospital of Shandong University, Jinan, China
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Laboratory of Malignant Tumor Precision Treatment, Jinan, China
- Shandong Provincial Engineering Research Center for Tumor Precision Treatment, Jinan, China
- Cancer Institute of Shandong University, Jinan, China
- Neutron Medical Center, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
21
|
Liao W, Wang Y, Wang R, Fu B, Chen X, Ouyang Y, Bai B, Jin Y, Lu Y, Liu F, Zhang Y, Shi D, Zhang D. Signature Construction Associated with Tumor-Infiltrating Macrophages Identifies IRF8 as a Novel Biomarker for Immunotherapy in Advanced Gastric Cancer. Int J Mol Sci 2025; 26:1089. [PMID: 39940857 PMCID: PMC11817691 DOI: 10.3390/ijms26031089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Advanced gastric cancer (AGC) is characterized by poor prognosis and limited responsiveness to immunotherapy. Tumor-associated macrophages (TAMs) play a pivotal role in cancer progression and therapeutic outcomes. In this study, we developed a novel gene signature associated with M1-like TAMs using data from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) to predict prognosis and immunotherapy response. This gene signature was determined as an independent prognostic indicator for AGC, with high-risk patients exhibiting an immunosuppressive tumor immune microenvironment (TIME) and poorer survival outcomes. Furthermore, Interferon regulatory factor 8 (IRF8) was identified as a key gene and validated through in vitro and in vivo experiments. IRF8 overexpression reshaped the suppressive TIME, leading to an increased presence of M1-like TAMs, IFN-γ+ CD8+ T cells, and Granzyme B+ CD8+ T cells. Notably, the combination of IRF8 overexpression and anti-PD-1 therapy significantly inhibited tumor growth in syngeneic mouse models. AGC patients with elevated IRF8 expression were found to be more responsive to anti-PD-1 treatment. These findings highlight potential biomarkers for prognostic evaluation and immunotherapy in AGC, offering insights that could guide personalized treatment strategies.
Collapse
Affiliation(s)
- Wanqian Liao
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (W.L.); (Y.W.); (B.F.); (B.B.); (Y.J.); (Y.L.); (F.L.); (Y.Z.)
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (R.W.); (X.C.); (Y.O.)
| | - Yu Wang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (W.L.); (Y.W.); (B.F.); (B.B.); (Y.J.); (Y.L.); (F.L.); (Y.Z.)
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (R.W.); (X.C.); (Y.O.)
| | - Rui Wang
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (R.W.); (X.C.); (Y.O.)
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Bibo Fu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (W.L.); (Y.W.); (B.F.); (B.B.); (Y.J.); (Y.L.); (F.L.); (Y.Z.)
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (R.W.); (X.C.); (Y.O.)
| | - Xiangfu Chen
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (R.W.); (X.C.); (Y.O.)
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ying Ouyang
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (R.W.); (X.C.); (Y.O.)
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Bing Bai
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (W.L.); (Y.W.); (B.F.); (B.B.); (Y.J.); (Y.L.); (F.L.); (Y.Z.)
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (R.W.); (X.C.); (Y.O.)
| | - Ying Jin
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (W.L.); (Y.W.); (B.F.); (B.B.); (Y.J.); (Y.L.); (F.L.); (Y.Z.)
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (R.W.); (X.C.); (Y.O.)
| | - Yunxin Lu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (W.L.); (Y.W.); (B.F.); (B.B.); (Y.J.); (Y.L.); (F.L.); (Y.Z.)
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (R.W.); (X.C.); (Y.O.)
| | - Furong Liu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (W.L.); (Y.W.); (B.F.); (B.B.); (Y.J.); (Y.L.); (F.L.); (Y.Z.)
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (R.W.); (X.C.); (Y.O.)
| | - Yang Zhang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (W.L.); (Y.W.); (B.F.); (B.B.); (Y.J.); (Y.L.); (F.L.); (Y.Z.)
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (R.W.); (X.C.); (Y.O.)
| | - Dongni Shi
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (R.W.); (X.C.); (Y.O.)
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Dongsheng Zhang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (W.L.); (Y.W.); (B.F.); (B.B.); (Y.J.); (Y.L.); (F.L.); (Y.Z.)
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; (R.W.); (X.C.); (Y.O.)
| |
Collapse
|
22
|
Panahizadeh R, Panahi P, Asghariazar V, Makaremi S, Noorkhajavi G, Safarzadeh E. A literature review of recent advances in gastric cancer treatment: exploring the cross-talk between targeted therapies. Cancer Cell Int 2025; 25:23. [PMID: 39856676 PMCID: PMC11762578 DOI: 10.1186/s12935-025-03655-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) ranks fourth in global mortality rates and fifth in prevalence, making it one of the most common cancers worldwide. Recent clinical studies have highlighted the potential of immunotherapies as a promising approach to treating GC. This study aims to shed light on the most impactful therapeutic strategies in the context of GC immunotherapy, highlighting both established and emerging approaches. MAIN BODY This review examines over 160 clinical studies conducted globally, focusing on the effectiveness of various immunotherapy modalities, including cancer vaccines, adoptive cell therapy, immune checkpoint inhibitors (ICIs), and monoclonal antibodies (mAbs). A comprehensive search of peer-reviewed literature was performed using databases such as Web of Science, PubMed, and Scopus. The selection criteria included peer-reviewed articles published primarily within the last 10 years, with a focus on studies that provided insights into targeted therapies and their mechanisms of action, clinical efficacy, and safety profiles. The findings indicate that these immunotherapy strategies can enhance treatment outcomes for GC, aligning with current treatment guidelines. ICIs like pembrolizumab and nivolumab have shown significant survival benefits in specific GC subgroups. Cancer vaccines and CAR-T cell therapies demonstrate potential, while mAbs targeting HER2 and VEGFR pathways enhance outcomes in combination regimens. We discuss the latest advancements and challenges in targeted therapy and immunotherapy for GC. Given the evolving nature of this field, this research emphasizes significant evidence-based therapies and those currently under evaluation rather than providing an exhaustive overview. Challenges include resistance mechanisms, immunosuppressive tumor environments, and inconsistent results from combination therapies. Biomarker-driven approaches and further research into emerging modalities like CAR-T cells and cancer vaccines are critical for optimizing treatments. CONCLUSIONS Immunotherapy is reshaping GC management by improving survival and quality of life. Ongoing research and clinical evaluations are crucial for refining personalized and effective therapies.
Collapse
Affiliation(s)
- Reza Panahizadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Padideh Panahi
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Asghariazar
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Shima Makaremi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ghasem Noorkhajavi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Microbiology, Parasitology and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, 85991-56189, Iran.
| |
Collapse
|
23
|
Xin Y, Liu N, Peng G, Huang X, Cao X, Zhou X. The Prognostic Value of Peripheral Blood Inflammatory Markers in Hepatocellular Carcinoma Treated with Lenvatinib Combined with PD-1 Inhibitors. J Hepatocell Carcinoma 2025; 12:135-147. [PMID: 39877858 PMCID: PMC11774115 DOI: 10.2147/jhc.s486910] [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: 08/27/2024] [Accepted: 11/15/2024] [Indexed: 01/31/2025] Open
Abstract
Purpose To investigate the prognostic value of inflammatory indexes based on peripheral blood cells in unresectable hepatocellular carcinoma (HCC) patients treated with Lenvatinib combined with PD-1 inhibitors. Methods This study retrospectively collected baseline inflammatory indexes from HCC patients received Lenvatinib and PD-1 inhibitor-based combination therapy at the Cancer Hospital of the Chinese Academy of Medical Sciences between October 2018 and October 2021. The optimal threshold values for inflammatory indexes determined using X-tile. The factors related to treatment response and survival outcomes were analyzed through logistic regression and Cox regression, respectively. A novel preoperative prognostic nomogram was constructed based on inflammatory indexes, and the predictive efficacy of the nomogram and BCLC staging was compared by the area under the ROC curve. Results 156 eligible patients with unresectable HCC were included, with median OS and PFS of 23.8 and 11.5 months, respectively, and ORR of 48.7%. The baseline SIRI was an independent factor of treatment response, with a significantly higher ORR for patients with a SIRI <0.8 than for patients with a SIRI ≥0.8 (59.7% vs 41.5%, P=0.03). SIRI and PNI were independent prognostic factors of PFS, and SIRI was an independent prognostic factor of OS. The AUC value of nomogram based on baseline SIRI, PNI, and tumor distribution in predicting the 6-,12- and 18-month PFS of patients was significantly higher than that of traditional BCLC stage, and its prediction performance was substantially better than that of BCLC stage system (C-index, 0.730 vs 0.535). Conclusion The baseline SIRI could be used as a potential non-invasive biomarker to predict the efficacy and survival benefit of immune combination therapy for HCC. The nomogram based on inflammation indexes could achieve better prediction performance and help clinicians to identify high-risk patients and formulate treatment plans.
Collapse
Affiliation(s)
- Yujing Xin
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
| | - Ning Liu
- School of Software, Shandong University, Jinan, Shandong, 250101, People’s Republic of China
| | - Gang Peng
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Xiaoyu Huang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Xiaojing Cao
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Xiang Zhou
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China
| |
Collapse
|
24
|
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
|
25
|
Wang B, Yin Y, Wang A, Liu W, Chen J, Li T. SMR-guided molecular subtyping and machine learning model reveals novel prognostic biomarkers and therapeutic targets in non-small cell lung adenocarcinoma. Sci Rep 2025; 15:1640. [PMID: 39794414 PMCID: PMC11723915 DOI: 10.1038/s41598-025-85471-8] [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: 09/20/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Non-small cell lung adenocarcinoma (LUAD) is a markedly heterogeneous disease, with its underlying molecular mechanisms and prognosis prediction presenting ongoing challenges. In this study, we integrated data from multiple public datasets, including TCGA, GSE31210, and GSE13213, encompassing a total of 867 tumor samples. By employing Mendelian randomization (MR) analysis, machine learning techniques, and comprehensive bioinformatics approaches, we conducted an in-depth investigation into the molecular characteristics, prognostic markers, and potential therapeutic targets of LUAD. Our analysis identified 321 genes significantly associated with LUAD, with CENP-A, MCM7, and DLGAP5 emerging as highly connected nodes in network analyses. By performing correlation analysis and Cox regression analysis, we identified 26 prognostic genes and classified LUAD samples into two molecular subtypes with significantly distinct survival outcomes. The Random Survival Forest (RSF) model exhibited robust prognostic predictive capabilities across multiple independent cohorts (AUC > 0.75). Beyond merely predicting patient outcomes, this model also captures key features of the tumor immune microenvironment and potential therapeutic responses. Functional enrichment analysis revealed the complex interplay of cell cycle regulation, DNA repair, immune response, and metabolic reprogramming in the progression of LUAD. Furthermore, we observed a strong correlation between risk scores and the expression of specific cytokines, such as CCL17, CCR2, and CCL20, suggesting novel avenues for developing cytokine network-based therapeutic strategies. This study offers fresh insights into the molecular subtyping, prognostic prediction, and personalized therapeutic decision-making in LUAD, laying a critical foundation for future clinical applications and targeted therapy research.
Collapse
Affiliation(s)
- Baozhen Wang
- School of Clinical Medicine, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China
- Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China
- Department of Surgical Oncology II, The General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, Ningxia, 750004, China
| | - Yichen Yin
- School of Clinical Medicine, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China
- Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China
- Department of Surgical Oncology II, The General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, Ningxia, 750004, China
| | - Anqi Wang
- Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China
- School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China
| | - Weidi Liu
- School of Clinical Medicine, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China
- Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China
- Department of Surgical Oncology II, The General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, Ningxia, 750004, China
| | - Jing Chen
- Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China.
- School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China.
| | - Tao Li
- Department of Surgical Oncology II, The General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, Ningxia, 750004, China.
| |
Collapse
|
26
|
Li H, Zheng L, Zhang X, Yu X, Zhong G, Chen X, Chen X, Chen L. SH3 domain‑binding glutamic acid‑rich protein‑like 3 is associated with hyperglycemia and a poor outcome in Epstein‑Barr virus‑negative gastric carcinoma. Oncol Lett 2025; 29:8. [PMID: 39492939 PMCID: PMC11526421 DOI: 10.3892/ol.2024.14754] [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: 06/02/2024] [Accepted: 09/03/2024] [Indexed: 11/05/2024] Open
Abstract
SH3 domain-binding glutamic acid-rich protein-like 3 (SH3BGRL3) is involved in several human cancers. However, its relationship with gastric cancer (GC) remains elusive. Multiple online bioinformatic tools were used to evaluate the messenger (m)RNA expression levels of SH3BGRL3 in GC using data from The Cancer Genome Atlas and Gene Expression Omnibus databases. Reverse transcription-quantitative PCR and tissue microarray-based immunohistochemistry were performed to assess SH3BGRL3 expression in relation to clinicopathological parameters and outcomes in patients with GC. Significant differentially expressed genes (DEGs) of SH3BGRL3 were enriched and visualized. Furthermore, associations between the expression of SH3BGRL3 and the infiltration of immune cells were explored. SH3BGRL3 exhibited aberrant expression in tumor tissues compared with adjacent normal tissues at the mRNA and protein expression levels, especially in Epstein-Barr virus-negative GC (EBVnGC). Higher SH3BGRL3 expression was significantly associated with increasing tumor-node-metastasis staging, tumor budding, perineural invasion, EGFR expression, and a notably higher preoperative blood glucose concentration in clinical specimens. Multivariate analysis revealed that higher SH3BGRL3 expression was an independent adverse prognostic factor for the overall survival of patients with EBVnGC (hazard ratio, 1.666; P=0.018). Furthermore, the stratified analysis revealed that the SH3BGRL3 phenotype could help to refine prognosis in patients. The C-index of the nomogram was 0.740 when combining SH3BGRL3 with other clinicopathological parameters, which indicated a good model for clinical follow-up decisions. Gene functional enrichment analysis also revealed that the DEGs of SH3BGRL3 were mainly enriched in regulating ATP metabolism, ATP synthesis, oxidative phosphorylation and the electron transport chain in GC. Moreover, a higher SH3BGRL3 expression was significantly positively correlated with the infiltrating macrophages in GC. In conclusion, SH3BGRL3 is upregulated in GC, particularly in EBVnGC. Higher SH3BGRL3 expression is closely associated with hyperglycemia and poor outcomes in patients with EBVnGC, suggesting its potential as a biomarker and prognostic predictor.
Collapse
Affiliation(s)
- Houqiang Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Lanqing Zheng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
- Nursing Department, Fujian Provincial Hospital, Fuzhou, Fujian 35001, P.R. China
| | - Xia Zhang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Xunbin Yu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Guodong Zhong
- Department of Pathology, The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, Fujian 350003, P.R. China
| | - Xiaoyan Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Xin Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
- Department of Pathology, Fujian Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Linying Chen
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| |
Collapse
|
27
|
Yun H, Dong F, Wei X, Yan X, Zhang R, Zhang X, Wang Y. Role and value of the tumor microenvironment in the progression and treatment resistance of gastric cancer (Review). Oncol Rep 2025; 53:14. [PMID: 39611496 PMCID: PMC11622107 DOI: 10.3892/or.2024.8847] [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: 08/08/2024] [Accepted: 10/08/2024] [Indexed: 11/30/2024] Open
Abstract
Gastric cancer (GC) is characterized by a complex and heterogeneous tumor microenvironment (TME) that significantly influences disease progression and treatment outcomes. The tumor stroma, which is composed of a variety of cell types such as cancer‑associated fibroblasts, immune cells and vascular components, displays significant spatial and temporal diversity. These stromal elements engage in dynamic crosstalk with cancer cells, shaping their proliferative, invasive and metastatic potential. Furthermore, the TME is instrumental in facilitating resistance to traditional chemotherapy, specific treatments and immunotherapy strategies. Understanding the underlying mechanisms by which the GC microenvironment evolves and supports tumor growth and therapeutic resistance is critical for developing effective treatment strategies. The present review explores the latest progress in understanding the intricate interactions between cancer cells and their immediate environment in GC, highlighting the implications for disease pathogenesis and therapeutic interventions.
Collapse
Affiliation(s)
- Heng Yun
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Fangde Dong
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Xiaoqin Wei
- Department of Pain, The Second People's Hospital of Baiyin, Baiyin, Gansu 730900, P.R. China
| | - Xinyong Yan
- Department of Proctology, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Ronglong Zhang
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Xiuyu Zhang
- Department of Gastroenterology, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Yulin Wang
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| |
Collapse
|
28
|
Buzaglo GBB, Telles GD, Araújo RB, Junior GDS, Ruberti OM, Ferreira MLV, Derchain SFM, Vechin FC, Conceição MS. The Therapeutic Potential of Physical Exercise in Cancer: The Role of Chemokines. Int J Mol Sci 2024; 25:13740. [PMID: 39769501 PMCID: PMC11678861 DOI: 10.3390/ijms252413740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 01/11/2025] Open
Abstract
The global increase in cancer cases and mortality has been associated with inflammatory processes, in which chemokines play crucial roles. These molecules, a subfamily of cytokines, are essential for the migration, adhesion, interaction, and positioning of immune cells throughout the body. Chemokines primarily originate in response to pathogenic stimuli and inflammatory cytokines. They are expressed by lymphocytes in the bloodstream and are divided into four classes (CC, CXC, XC, and CX3C), playing multifaceted roles in the tumor environment (TME). In the TME, chemokines regulate immune behavior by recruiting cells such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), which promote tumor survival. Additionally, they directly influence tumor behavior, promoting pathological angiogenesis, invasion, and metastasis. On the other hand, chemokines can also induce antitumor responses by mobilizing CD8+ T cells and natural killer (NK) cells to the tumor, reducing pro-inflammatory chemokines and enhancing essential antitumor responses. Given the complex interaction between chemokines, the immune system, angiogenic factors, and metastasis, it becomes evident how important it is to target these pathways in therapeutic interventions to counteract cancer progression. In this context, physical exercise emerges as a promising strategy due to its role modulating the expression of anti-inflammatory chemokines and enhancing the antitumor response. Aerobic and resistance exercises have been associated with a beneficial inflammatory profile in cancer, increased infiltration of CD8+ T cells in the TME, and improvement of intratumoral vasculature. This creates an environment less favorable to tumor growth and supports the circulation of antitumor immune cells and chemokines. Therefore, understanding the impact of exercise on the expression of chemokines can provide valuable insights for therapeutic interventions in cancer treatment and prevention.
Collapse
Affiliation(s)
- Glenda B. B. Buzaglo
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Guilherme D. Telles
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (G.D.T.); (F.C.V.)
| | - Rafaela B. Araújo
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Gilmar D. S. Junior
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Olivia M. Ruberti
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Marina L. V. Ferreira
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Sophie F. M. Derchain
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Campinas, Campinas, Sao Paulo 13083-881, Brazil;
| | - Felipe C. Vechin
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (G.D.T.); (F.C.V.)
| | - Miguel S. Conceição
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| |
Collapse
|
29
|
Chen S, Liu J, He G, Tang N, Zeng Y. Research Hotspots and Trends in Global Cancer immunometabolism:A Bibliometric Analysis from 2000 to 2023. J Multidiscip Healthc 2024; 17:5117-5137. [PMID: 39553266 PMCID: PMC11568773 DOI: 10.2147/jmdh.s495330] [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: 09/18/2024] [Accepted: 10/30/2024] [Indexed: 11/19/2024] Open
Abstract
Background Cancer poses a major global health challenge, and immunotherapy, known as the third revolution in cancer treatment, has brought new hope to patients. The emerging field of immunometabolism has further enhanced the safety and efficacy of immunotherapy. Over the past two decades, this field has rapidly evolved in oncology, leading to numerous significant findings. This review systematically examines the literature on immunometabolism in cancer, visualizing research trends and identifying future directions. Methods A comprehensive literature search was conducted in the Web of Science, PubMed, and Scopus databases, covering publications from January 2000 to December 2023. We employed tools like Citespace, VOSviewer, and RStudio for visual analysis of publication trends, regional contributions, institutions, authors, journals, and keywords. Results A total of 3320 articles were published by 8090 authors across 1738 institutions, involving 71 countries. Leading contributors were China (n=469), the United States (n=361), and Germany (n=82). Harvard University was the most influential institution, while Frontiers in Immunology had the highest number of publications. The top research areas included glucose, lipid, and amino acid metabolism, the tumor microenvironment, and immune cell regulation. Conclusion International collaboration and interdisciplinary efforts are advancing the field of cancer immunometabolism. Future research will likely focus on the interplay between metabolism and immunity, metabolic markers, immune cell reprogramming, and tumor-immune metabolic competition.
Collapse
Affiliation(s)
- Shupeng Chen
- School of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Jie Liu
- School of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Guilian He
- School of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, People’s Republic of China
| | - Nana Tang
- Hematology Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330006, People’s Republic of China
| | - Yingjian Zeng
- Hematology Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330006, People’s Republic of China
| |
Collapse
|
30
|
Li Z, Li X, Lu Y, Zhu X, Zheng W, Chen K, Wang X, Wang T, Guan W, Su Z, Liu S, Wu J. Novel Photo-STING Agonists Delivered by Erythrocyte Efferocytosis-Mimicking Pattern to Repolarize Tumor-Associated Macrophages for Boosting Anticancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2410937. [PMID: 39380354 DOI: 10.1002/adma.202410937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/22/2024] [Indexed: 10/10/2024]
Abstract
Immunotherapy has emerged as a highly effective therapeutic strategy for cancer treatment. Cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon gene (STING) pathway activation facilitates tumor-associated macrophage (TAM) polarization toward M1 phenotype, and Mn2+ are effective agents for this pathway activation. However, the high in vivo degradation rate and toxicity of Mn2+ hamper clinical application of immunotherapy. Here, this work has newly synthesized and screened manganese porphyrins for Mn2+ transport, referred to as photo-STING agonists (PSAs), and further encapsulate them into core-shell nanoparticles named Rm@PP-GA with dual specificity for tumor tissue and TAMs. Not only do PSAs achieve higher Mn2+ delivery efficiency compared to Mn2+, but they also generate reactive oxygen species under light exposure, promoting mitochondrial DNA release for cGAS-STING pathway activation. In Rm@PP-GA, globin and red blood cell membranes (Rm) are used for erythrocyte efferocytosis-mimicking delivery. Rm can effectively prolong the in vivo circulation period while globin enables PSAs to be taken up by TAMs via CD163 receptors. After Rm rupture mediated by perfluorohexane in nanoparticles under ultrasonication, drugs are specifically released for TAM repolarization. Further, dendritic cells mature, as well as T lymphocyte infiltrate, both of which favor tumor eradication. Therefore, cancer immunotherapy is optimized by novel PSAs delivered by erythrocyte efferocytosis-mimicking delivery pattern.
Collapse
Affiliation(s)
- Zhiyan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Xianghui Li
- First Affiliated Hospital of Guangxi Medical University, Department of Dermatology, Nanning, 530021, China
| | - Yanjun Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
| | - Xudong Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Wenxuan Zheng
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Kai Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Xingzhou Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Tao Wang
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Wenxian Guan
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Zhi Su
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Song Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
31
|
Kim YJ, Nanda SS, Jiang F, Pyo SY, Han JY, Koh SS, Kang TH. Pancreatic Adenocarcinoma Up-Regulated Factor (PAUF) Transforms Human Monocytes into Alternative M2 Macrophages with Immunosuppressive Action. Int J Mol Sci 2024; 25:11545. [PMID: 39519098 PMCID: PMC11547018 DOI: 10.3390/ijms252111545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Tumor-associated macrophages (TAMs) in the tumor microenvironment (TME) promote immune evasion, cancer cell proliferation, and metastasis. Ongoing research is focused on finding ways to prevent tumor growth by inhibiting TAM polarization, which has shown a correlation with unfavorable prognosis in clinical studies. Pancreatic adenocarcinoma up-regulated factor (PAUF) is a protein secreted from pancreatic cancer (PC) and acts as a TME modulator that affects the TME by acting on not only cancer cells but also stromal cells and immune cells. Tumor cells can evade the immune system by PAUF binding to Toll-like receptor (TLR) in monocytes, as this research shows. In this study, the examination centered around the recruitment of human monocytes by PAUF and the subsequent differentiation into macrophages. In an in vitro chemotaxis assay, PAUF induced chemotactic migration of TLR2-mediated monocytes. In addition, PAUF induced differentiation of monocytes into M2 macrophages, which was verified based on expressing surface markers and cytokines and morphological analysis. The inhibition of T cell proliferation and function was observed in differentiated M2 macrophages. To conclude, these findings indicate that PAUF functions as a promoter of cancer progression by regulating the recruitment and differentiation of macrophages within TMEs, ultimately causing immunosuppression.
Collapse
Affiliation(s)
- Yeon Jeong Kim
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
- Department of Biomedical Sciences, Dong-A University, Busan 49315, Republic of Korea
| | - Sitansu Sekhar Nanda
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| | - Fen Jiang
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| | - Seung Yeon Pyo
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| | - Jin-Yeong Han
- Department of Laboratory Medicine, College of Medicine, Dong-A University, Busan 49201, Republic of Korea;
| | - Sang Seok Koh
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| | - Tae Heung Kang
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| |
Collapse
|
32
|
Liu C, Fu L, Wang Y, Yang W. Influence of the gut microbiota on immune cell interactions and cancer treatment. J Transl Med 2024; 22:939. [PMID: 39407240 PMCID: PMC11476117 DOI: 10.1186/s12967-024-05709-3] [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: 07/16/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
The tumour microenvironment represents a novel frontier in oncological research. Over the past decade, accumulating evidence has underscored the importance of the tumour microenvironment (TME), including tumour cells, stromal cells, immune cells, and various secreted factors, which collectively influence tumour growth, invasion, and responses to therapeutic agents. Immune cells within the TME are now widely acknowledged to play pivotal roles in tumour development and treatment. While some perspectives have posited that immune cells within the TME facilitate tumour progression and confer resistance to therapeutic interventions, contrasting conclusions also exist. Affirmative and negative conclusions appear to be context dependent, and a unified consensus has yet to be reached. The burgeoning body of research on the relationship between the gut microbiota and tumours in recent years has led to a growing understanding. Most studies have indicated that specific components of the gut microbiota, such as unique bacterial communities or specific secretory factors, play diverse roles in regulating immune cells within the TME, thereby influencing the prognosis and outcomes of cancer treatments. A detailed understanding of these factors could provide novel insights into the TME and cancer therapy. In this study, we aimed to synthesise information on the interactions between the gut microbiota and immune cells within the TME, providing an in-depth exploration of the potential guiding implications for future cancer therapies.
Collapse
Affiliation(s)
- Chunxiao Liu
- Department of Gastroenterological Surgery, Hengqin Hospital, First Affiliated Hospital of Guangzhou Medical University, No. 118 Baoxing Road, Hengqin, Guangdong, 519031, China
| | - Lingfeng Fu
- Department of Gastroenterological Surgery, Hengqin Hospital, First Affiliated Hospital of Guangzhou Medical University, No. 118 Baoxing Road, Hengqin, Guangdong, 519031, China
| | - Yuxin Wang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, No. 1838, North Guangzhou Avenue, Guangzhou, Guangdong, 510515, China.
- Central Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Weijun Yang
- Department of Gastroenterological Surgery, Hengqin Hospital, First Affiliated Hospital of Guangzhou Medical University, No. 118 Baoxing Road, Hengqin, Guangdong, 519031, China.
| |
Collapse
|
33
|
Wang Y, Ding G, Chu C, Cheng XD, Qin JJ. Genomic biology and therapeutic strategies of liver metastasis from gastric cancer. Crit Rev Oncol Hematol 2024; 202:104470. [PMID: 39111457 DOI: 10.1016/j.critrevonc.2024.104470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
The liver is a frequent site of metastasis in advanced gastric cancer (GC). Despite significant advancements in diagnostic and therapeutic techniques, the overall survival rate for patients afflicted with gastric cancer liver metastasis (GCLM) remains dismally low. Precision oncology has made significant progress in identifying therapeutic targets and enhancing our understanding of metastasis mechanisms through genome sequencing and molecular characterization. Therefore, it is crucial to have a comprehensive understanding of the various molecular processes involved in GCLM and the fundamental principles of systemic therapy to develop new treatment approaches. This paper aims to review recent findings on the diagnosis, potential biomarkers, and therapies targeting the multiple molecular processes of GCLM, with the goal of improving treatment strategies for patients with GCLM.
Collapse
Affiliation(s)
- Yichao Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 313200, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Guangyu Ding
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Chu Chu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 313200, China
| | - Xiang-Dong Cheng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China.
| | - Jiang-Jiang Qin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Key Laboratory for Molecular Medicine and Chinese Medicine Preparations, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China.
| |
Collapse
|
34
|
Chen X, Peng H, Zhang Z, Yang C, Liu Y, Chen Y, Yu F, Wu S, Cao L. SPDYC serves as a prognostic biomarker related to lipid metabolism and the immune microenvironment in breast cancer. Immunol Res 2024; 72:1030-1050. [PMID: 38890248 DOI: 10.1007/s12026-024-09505-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024]
Abstract
Breast cancer remains the most common malignant carcinoma among women globally and is resistant to several therapeutic agents. There is a need for novel targets to improve the prognosis of patients with breast cancer. Bioinformatics analyses were conducted to explore potentially relevant prognostic genes in breast cancer using The Cancer Genome Atlas (TCGA) and The Gene Expression Omnibus (GEO) databases. Gene subtypes were categorized by machine learning algorithms. The machine learning-related breast cancer (MLBC) score was evaluated through principal component analysis (PCA) of clinical patients' pathological statuses and subtypes. Immune cell infiltration was analyzed using the xCell and CIBERSORT algorithms. Kyoto Encyclopedia of Genes and Genomes enrichment analysis elucidated regulatory pathways related to speedy/RINGO cell cycle regulator family member C (SPDYC) in breast cancer. The biological functions and lipid metabolic status of breast cancer cell lines were validated via quantitative real-time polymerase chain reaction (RT‒qPCR) assays, western blotting, CCK-8 assays, PI‒Annexin V fluorescence staining, transwell assays, wound healing assays, and Oil Red O staining. Key differentially expressed genes (DEGs) in breast cancer from the TCGA and GEO databases were screened and utilized to establish the MLBC score. Moreover, the MLBC score we established was negatively correlated with poor prognosis in breast cancer patients. Furthermore, the impacts of SPDYC on the tumor immune microenvironment and lipid metabolism in breast cancer were revealed and validated. SPDYC is closely related to activated dendritic cells and macrophages and is simultaneously correlated with the immune checkpoints CD47, cytotoxic T lymphocyte antigen-4 (CTLA-4), and poliovirus receptor (PVR). SPDYC strongly correlated with C-C motif chemokine ligand 7 (CCL7), a chemokine that influences breast cancer patient prognosis. A significant relationship was discovered between key genes involved in lipid metabolism and SPDYC, such as ELOVL fatty acid elongase 2 (ELOVL2), malic enzyme 1 (ME1), and squalene epoxidase (SQLE). Potent inhibitors targeting SPDYC in breast cancer were also discovered, including JNK inhibitor VIII, AICAR, and JW-7-52-1. Downregulation of SPDYC expression in vitro decreased proliferation, increased the apoptotic rate, decreased migration, and reduced lipid droplets. SPDYC possibly influences the tumor immune microenvironment and regulates lipid metabolism in breast cancer. Hence, this study identified SPDYC as a pivotal biomarker for developing therapeutic strategies for breast cancer.
Collapse
Affiliation(s)
- Xinxin Chen
- Department of Breast Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Haojie Peng
- Department of Breast Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhentao Zhang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Changnian Yang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yingqi Liu
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yanzhen Chen
- Department of Gynecology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Fei Yu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Shanshan Wu
- Department of Biology, School of Basic Medical Science, Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Lixue Cao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
35
|
Li W, Wei J, Cheng M, Liu M. Unveiling promising targets in gastric cancer therapy: A comprehensive review. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200857. [PMID: 39280587 PMCID: PMC11396074 DOI: 10.1016/j.omton.2024.200857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Gastric cancer (GC) poses a significant global health challenge, ranking fifth in incidence and third in mortality among all malignancies worldwide. Its insidious onset, aggressive growth, proclivity for metastasis, and limited treatment options have contributed to its high fatality rate. Traditional approaches for GC treatment primarily involve surgery and chemotherapy. However, there is growing interest in targeted therapies and immunotherapies. This comprehensive review highlights recent advancements in GC targeted therapy and immunotherapy. It delves into the mechanisms of various strategies, underscoring their potential in GC treatment. Additionally, the review evaluates the efficacy and safety of relevant clinical trials. Despite the benefits observed in numerous advanced GC patients with targeted therapies and immunotherapies, challenges persist. We discuss pertinent strategies to overcome these challenges, thereby providing a solid foundation for enhancing the clinical effectiveness of targeted therapies and immunotherapies.
Collapse
Affiliation(s)
- Wenke Li
- Gastric Cancer Center/Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Jing Wei
- Gastric Cancer Center/Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Mo Cheng
- Gastric Cancer Center/Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Ming Liu
- Gastric Cancer Center/Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| |
Collapse
|
36
|
Su G, Xu Z, Liu S, Hao D, Li Y, Pan G. Investigation of the Mechanism of SEMA5A and Its Associated Autophagy-Related Genes in Gastric Cancer. Int J Gen Med 2024; 17:4101-4117. [PMID: 39295854 PMCID: PMC11409931 DOI: 10.2147/ijgm.s471370] [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: 06/04/2024] [Accepted: 08/27/2024] [Indexed: 09/21/2024] Open
Abstract
Purpose Semaphorin 5A (SEMA5A) and autophagy-related genes (ARGs) are pivotal in the pathogenesis of gastric cancer (GC). However, the potential regulatory role of SEMA5A in autophagy via its associated ARGs and the underlying molecular mechanisms remain unresolved. Patients and Methods GC-related datasets from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) were analyzed to identify differentially expressed genes (DEGs) between GC and control samples. The intersection of DEGs with ARGs produced candidate genes, which were further analyzed using Spearman correlation with SEMA5A to identify signature genes. Stratification of GC samples based on signature gene expression, followed by Kaplan-Meier survival analysis, identified key genes. Subsequent analyses, including gene set enrichment analysis (GSEA), immune infiltration, and immune checkpoint evaluation, were conducted on the key genes and SEMA5A. The mRNA expression level was quantified using real-time quantitative polymerase chain reaction (RT-qPCR). Results Ninety candidate genes were identified for Spearman correlation with SEMA5A, revealing TNFSF11, BMP6, ITPR1, and DLC1 with correlation coefficients exceeding 0.3. Survival analysis underscored DLC1 and BMP6 as key genes due to significant prognostic differences. GSEA implicated SEMA5A, BMP6, and DLC1 in the ECM receptor interaction pathway. Immune infiltration analysis indicated a negative correlation of SEMA5A and BMP6 with M1 macrophages, while DLC1 exhibited the strongest association with the immune checkpoint PDCD1LG2 (p < 0.05, cor = 0.43). The mRNA expression level of SEMA5A was significantly upregulated in AGS parental cells compared to GES-1 cells (p < 0.01), whereas DLC1 and BMP6 mRNA levels were markedly downregulated in AGS parental cells relative to GES-1 (p < 0.0001). Conclusion ARGs BMP6 and DLC1, associated with SEMA5A, were identified, and their prognostic significance in GC was demonstrated. Additionally, their regulatory mechanisms were further elucidated through immune infiltration analysis and molecular network construction, providing a theoretical foundation for future research on the molecular mechanisms in patients with GC.
Collapse
Affiliation(s)
- Guomiao Su
- Department of Pathology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Zifan Xu
- Department of Pathology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Shiyue Liu
- Department of Pathology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Dou Hao
- Department of Pathology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Yanxi Li
- Department of Pathology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Guoqing Pan
- Department of Pathology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| |
Collapse
|
37
|
Liu X, Wu F, Pan W, Liu G, Zhang H, Yan D, Zheng S, Ma Z, Ren X. Tumor-associated exosomes in cancer progression and therapeutic targets. MedComm (Beijing) 2024; 5:e709. [PMID: 39247621 PMCID: PMC11380050 DOI: 10.1002/mco2.709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/10/2024] Open
Abstract
Exosomes are small membrane vesicles that are released by cells into the extracellular environment. Tumor-associated exosomes (TAEs) are extracellular vesicles that play a significant role in cancer progression by mediating intercellular communication and contributing to various hallmarks of cancer. These vesicles carry a cargo of proteins, lipids, nucleic acids, and other biomolecules that can be transferred to recipient cells, modifying their behavior and promoting tumor growth, angiogenesis, immune modulation, and drug resistance. Several potential therapeutic targets within the TAEs cargo have been identified, including oncogenic proteins, miRNAs, tumor-associated antigens, immune checkpoint proteins, drug resistance proteins, and tissue factor. In this review, we will systematically summarize the biogenesis, composition, and function of TAEs in cancer progression and highlight potential therapeutic targets. Considering the complexity of exosome-mediated signaling and the pleiotropic effects of exosome cargoes has challenge in developing effective therapeutic strategies. Further research is needed to fully understand the role of TAEs in cancer and to develop effective therapies that target them. In particular, the development of strategies to block TAEs release, target TAEs cargo, inhibit TAEs uptake, and modulate TAEs content could provide novel approaches to cancer treatment.
Collapse
Affiliation(s)
- Xiaomin Liu
- Lab for Noncoding RNA & Cancer School of Life Sciences Shanghai University Shanghai China
- Shanghai New Tobacco Product Research Institute Co., Ltd. Shanghai China
| | - Fan Wu
- Lab for Noncoding RNA & Cancer School of Life Sciences Shanghai University Shanghai China
| | - Wei Pan
- Lab for Noncoding RNA & Cancer School of Life Sciences Shanghai University Shanghai China
| | - Guangchao Liu
- Shanghai New Tobacco Product Research Institute Co., Ltd. Shanghai China
| | - Hui Zhang
- Shanghai New Tobacco Product Research Institute Co., Ltd. Shanghai China
| | - Dawei Yan
- Shanghai New Tobacco Product Research Institute Co., Ltd. Shanghai China
| | - Saijing Zheng
- Shanghai New Tobacco Product Research Institute Co., Ltd. Shanghai China
| | - Zhongliang Ma
- Lab for Noncoding RNA & Cancer School of Life Sciences Shanghai University Shanghai China
| | - Xiaojun Ren
- Department of Chemistry College of Chemistry and Life Sciences Beijing University of Technology Beijing China
| |
Collapse
|
38
|
Zhao Q, Yu H, Shi M, Wang X, Fan Z, Wang Z. Tumor microenvironment characteristics of lipid metabolism reprogramming related to ferroptosis and EndMT influencing prognosis in gastric cancer. Int Immunopharmacol 2024; 137:112433. [PMID: 38870879 DOI: 10.1016/j.intimp.2024.112433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Gastric cancer (GC) is a refractory malignant tumor with high tumor heterogeneity, a low rate of early diagnosis, and poor patient prognosis. Lipid metabolism reprogramming plays a critical role in tumorigenesis and progression, but its prognostic role and regulatory mechanism in GC are rarely studied. Thus, the identification of signatures related to lipid metabolism is necessary and may present a new avenue for improving the overall prognosis of GC. METHODS Lipid metabolism-associated genes (LMAGs) with differential expression in tumor and tumor-adjacent tissue were acquired to identify lipid metabolism-associated subtypes. The differentially expressed genes (DEGs) between the two clusters were then utilized for prognostic analysis and signature construction. Additionally, pathway enrichment analysis and immune cell infiltration analysis were employed to identify the characteristics of the prognostic model. Further analyses were conducted at the single-cell level to better understand the model's prognostic mechanism. Finally, the prediction of immunotherapy response was used to suggest potential treatments. RESULTS Two lipid metabolism-associated subtypes were identified and 9 prognosis-related genes from the DEGs between the two clusters were collected for the construction of the prognostic model named lipid metabolism-associated signature (LMAS). Then we found the low LMAS patients with favorable prognoses were more sensitive to ferroptosis in the Cancer Genome Atlas of Stomach Adenocarcinoma (TCGA-STAD). Meanwhile, the tumor cells exhibiting high levels of lipid peroxidation and accumulation of reactive oxygen species (ROS) in single-cell levels were primarily enriched in the low LMAS group, which was more likely to induce ferroptosis. In addition, endothelial cells and cancer-associated fibroblasts (CAFs) facilitated tumor angiogenesis, proliferation, invasion, and metastasis through endothelial-mesenchymal transition (EndMT), affecting the prognosis of the patients with high LMAS scores. Moreover, CD1C- CD141- dendritic cells (DCs) also secreted pro-tumorigenic cytokines to regulate the function of endothelial cells and CAFs. Finally, the patients with low LMAS scores might have better efficacy in immunotherapy. CONCLUSIONS A LMAS was constructed to guide GC prognosis and therapy. Meanwhile, a novel anti-tumor effect was found in lipid metabolism reprogramming of GC which improved patients' prognosis by regulating the sensitivity of tumor cells to ferroptosis. Moreover, EndMT may have a negative impact on GC prognosis.
Collapse
Affiliation(s)
- Qian Zhao
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou 014030, China; School of Basic Medicine, Baotou Medical College, Baotou 014040, China
| | - Hui Yu
- Translational Medicine Center, Baotou Medical College, Baotou 014040, China
| | - Mengqi Shi
- School of Basic Medicine, Baotou Medical College, Baotou 014040, China
| | - Xujie Wang
- School of Basic Medicine, Baotou Medical College, Baotou 014040, China
| | - Zixu Fan
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou 014030, China
| | - Zhanli Wang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou 014030, China.
| |
Collapse
|
39
|
Li X, Qu X, Wang N, Li S, Zhao X, Lin K, Shi Y. A novel M2-like tumor associated macrophages-related gene signature for predicting the prognosis and immunotherapy efficacy in gastric cancer. Discov Oncol 2024; 15:353. [PMID: 39150637 PMCID: PMC11329457 DOI: 10.1007/s12672-024-01221-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 08/05/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND M2-like tumor-associated macrophages (M2-like TAMs) play key roles in tumor progression and the immune response. However, the clinical significance and prognostic value of M2-like TAMs-associated regulatory genes in gastric cancer (GC) have not been clarified. METHODS Herein, we identified M2-like TAM-related genes by weighted gene coexpression network analysis of TCGA-STAD and GSE84437 cohort. Lasso-Cox regression analyses were then performed to screen for signature genes, and a novel signature was constructed to quantify the risk score for each patient. Tumor mutation burden (TMB), survival outcomes, immune cells, and immune function were analyzed in the risk groups to further reveal the immune status of GC patients. A gene-drug correlation analysis and sensitivity analysis of anticancer drugs were used to identify potential therapeutic agents. Finally, we verified the mRNA expression of signature genes in patient tissues by qRT-PCR, and analyzed the expression distribution of these genes by IHC. RESULTS A 4-gene (SERPINE1, MATN3, CD36, and CNTN1) signature was developed and validated, and the risk score was shown to be an independent prognostic factor for GC patients. Further analyses revealed that GC patients in the high-risk group had a worse prognosis than those in the low-risk group, with significant differences in TMB, clinical features, enriched pathways, TIDE score, and tumor microenvironment features. Finally, we used qRT-PCR and IHC analysis to verify mRNA and protein level expression of signature genes. CONCLUSION These findings highlight the importance of M2-like TAMs, provide a new perspective on individualized immunotherapy for GC patients.
Collapse
Affiliation(s)
- Xuezhi Li
- State key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Xiaodong Qu
- State key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Na Wang
- State key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Songbo Li
- State key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Xingyu Zhao
- State key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Kexin Lin
- State key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Yongquan Shi
- State key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
40
|
Liu Z, Sun L, Zhu W, Zhu J, Wu C, Peng X, Tian H, Huang C, Zhu Z. Disulfidptosis signature predicts immune microenvironment and prognosis of gastric cancer. Biol Direct 2024; 19:65. [PMID: 39148138 PMCID: PMC11325698 DOI: 10.1186/s13062-024-00518-6] [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: 03/04/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Disulfidptosis is a newly identified mechanism of cell death triggered by disulfide stress. Thus, gaining a comprehensive understanding of the disulfidptosis signature present in gastric cancer (GC) could greatly enhance the development of personalized treatment strategies for this disease. METHODS We employed consensus clustering to identify various subtypes of disulfidptosis and examined the distinct tumor microenvironment (TME) associated with each subtype. The Disulfidptosis (Dis) score was used to quantify the subtype of disulfidptosis in each patient. Subsequently, we assessed the predictive value of Dis score in terms of GC prognosis and immune efficacy. Finally, we conducted in vitro experiments to explore the impact of Collagen X (COL10A1) on the progression of GC. RESULTS Two disulfidptosis-associated molecular subtypes (Discluster A and B) were identified, each with distinct prognosis, tumor microenvironment (TME), immune cell infiltration, and biological pathways. Discluster A, characterized by high expression of disulfidptosis genes, exhibited a high immune score but poor prognosis. Furthermore, the Dis score proved useful in predicting the prognosis and immune response in GC patients. Those in the low Dis score group showed better prognosis and increased sensitivity to immunotherapy. Finally, our experimental findings validated that downregulation of COL10A1 expression attenuates the proliferation and migration capabilities of GC cells while promoting apoptosis. CONCLUSIONS This study demonstrates that the disulfidptosis signature can assist in risk stratification and personalized treatment for patients with GC. The results offer valuable theoretical support for anti-tumor strategies.
Collapse
Affiliation(s)
- Zitao Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, MinDe Road, Nanchang, Jiangxi, P. R. China
| | - Liang Sun
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, MinDe Road, Nanchang, Jiangxi, P. R. China.
| | - Wenjie Zhu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, MinDe Road, Nanchang, Jiangxi, P. R. China
| | - Jinfeng Zhu
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan, P. R. China
| | - Changlei Wu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, MinDe Road, Nanchang, Jiangxi, P. R. China
| | - Xingyu Peng
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, MinDe Road, Nanchang, Jiangxi, P. R. China
| | - Huakai Tian
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P. R. China
| | - Chao Huang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, MinDe Road, Nanchang, Jiangxi, P. R. China
| | - Zhengming Zhu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, MinDe Road, Nanchang, Jiangxi, P. R. China.
| |
Collapse
|
41
|
Gu W, Guo W, Ren Z, Zhang Y, Han M, Zhao Q, Gao Y, Mao Y, Wang S. A bioactive nanocomposite integrated specific TAMs target and synergistic TAMs repolarization for effective cancer immunotherapy. Bioact Mater 2024; 38:472-485. [PMID: 38779591 PMCID: PMC11109736 DOI: 10.1016/j.bioactmat.2024.04.029] [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: 12/07/2023] [Revised: 04/10/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Reactive oxygen species (ROS) generated from photosensitizers exhibit great potential for repolarizing immunosuppressive tumor-associated macrophages (TAMs) toward the anti-tumor M1 phenotype, representing a promising cancer immunotherapy strategy. Nevertheless, their effectiveness in eliminating solid tumors is generally limited by the instability and inadequate TAMs-specific targeting of photosensitizers. Here, a novel core-shell integrated nano platform is proposed to achieve a coordinated strategy of repolarizing TAMs for potentiating cancer immunotherapy. Colloidal mesoporous silica nanoparticles (CMSN) are fabricated to encapsulate photosensitizer-Indocyanine Green (ICG) to improve their stability. Then ginseng-derived exosome (GsE) was coated on the surface of ICG/CMSN for targeting TAMs, as well as repolarizing TAMs concurrently, named ICG/CMSN@GsE. As expected, with the synergism of ICG and GsE, ICG/CMSN@GsE exhibited better stability, mild generation of ROS, favorable specificity toward M2-like macrophages, enhancing drug retention in tumors and superior TAMs repolarization potency, then exerted a potent antitumor effect. In vivo, experiment results also confirm the synergistic suppression of tumor growth accompanied by the increased presence of anti-tumor M1-like macrophages and maximal tumor damage. Taken together, by integrating the superiorities of TAMs targeting specificity and synergistic TAMs repolarization effect into a single nanoplatform, ICG/CMSN@GsE can readily serve as a safe and high-performance nanoplatform for enhanced cancer immunotherapy.
Collapse
Affiliation(s)
- Wei Gu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Wen Guo
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Zhishuang Ren
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Yimeng Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Meiqi Han
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Yikun Gao
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Yuling Mao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| |
Collapse
|
42
|
Shang JR, Zhu J, Bai L, Kulabiek D, Zhai XX, Zheng X, Qian J. Adipocytes impact on gastric cancer progression: Prognostic insights and molecular features. World J Gastrointest Oncol 2024; 16:3011-3031. [PMID: 39072151 PMCID: PMC11271780 DOI: 10.4251/wjgo.v16.i7.3011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/17/2024] [Accepted: 05/28/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Adipocytes, especially adipocytes within tumor tissue known as cancer-associated adipocytes, have been increasingly recognized for their pivotal role in the tumor microenvironment of gastric cancer (GC). Their influence on tumor progression and patient prognosis has sparked significant interest in recent research. The main objectives of this study were to investigate adipocyte infiltration, assess its correlation with clinical pathological features, develop a prognostic prediction model based on independent prognostic factors, evaluate the impact of adipocytes on immune cell infiltration and tumor invasiveness in GC, and identify and validate genes associated with high adipocyte expression, exploring their potential diagnostic and prognostic value. AIM To explore the relationship between increased adipocytes within tumor tissue and prognosis in GC patients as well as the associated mechanisms and potential biomarkers, using public databases and clinical data. METHODS Using mRNA microarray datasets from the Gene Expression Omnibus database and clinical samples from Jiangsu Provincial Hospital, survival and regression analyses were conducted to determine the relevant prognostic factors in GC. Feature gene selection was performed using least absolute shrinkage and selection operator and support vector machine recursive feature elimination algorithms, followed by differential gene expression analysis, gene ontology, pathway analysis, and Gene Set Enrichment Analysis. Immune cell infiltration was analyzed using the CIBERSORT algorithm. RESULTS Tumor adipocyte infiltration correlated with poor prognosis in GC, leading to the development of a highly accurate and discriminative prognostic prediction model. Key genes, ADH1B, SFRP1, PLAC9, and FABP4, were identified as associated with high adipocyte expression in GC. The diagnostic and prognostic potential of these identified genes was validated using independent datasets. Downregulation of immune cells was observed in GC with high adipocyte expression. CONCLUSION GC with high intratumoral adipocyte expression demonstrated aggressive tumor biology and a poorer prognosis. The genes ADH1B, SFRP1, PLAC9, and FABP4 have been identified as holding diagnostic and prognostic significance in GC. These findings strongly support the use of adipocyte expression as a valuable indicator of tumor invasiveness and anticipated patient outcomes in GC.
Collapse
Affiliation(s)
- Jia-Rong Shang
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Jin Zhu
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Lu Bai
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Delida Kulabiek
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Xiao-Xue Zhai
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Xia Zheng
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Jun Qian
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| |
Collapse
|
43
|
Salmaninejad A, Layeghi SM, Falakian Z, Golestani S, Kobravi S, Talebi S, Yousefi M. An update to experimental and clinical aspects of tumor-associated macrophages in cancer development: hopes and pitfalls. Clin Exp Med 2024; 24:156. [PMID: 39003350 PMCID: PMC11246281 DOI: 10.1007/s10238-024-01417-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024]
Abstract
Tumor-associated macrophages (TAMs) represent one of the most abundant tumor-infiltrating stromal cells, and their normal function in tumor microenvironment (TME) is to suppress tumor cells by producing cytokines which trigger both direct cell cytotoxicity and antibody-mediated immune response. However, upon prolonged exposure to TME, the classical function of these so-called M1-type TAMs can be converted to another type, "M2-type," which are recruited by tumor cells so that they promote tumor growth and metastasis. This is the reason why the accumulation of TAMs in TME is correlated with poor prognosis in cancer patients. Both M1- and M2-types have high degree of plasticity, and M2-type cells can be reprogrammed to M1-type for therapeutic purposes. This characteristic introduces TAMs as promising target for developing novel cancer treatments. In addition, inhibition of M2-type cells and blocking their recruitment in TME, as well as their depletion by inducing apoptosis, are other approaches for effective immunotherapy of cancer. In this review, we summarize the potential of TAMs to be targeted for cancer immunotherapy and provide an up-to-date about novel strategies for targeting TAMs.
Collapse
Affiliation(s)
- Arash Salmaninejad
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Pediatric Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| | - Sepideh Mehrpour Layeghi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeinab Falakian
- Department of Laboratory Science, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Shahin Golestani
- Department of Ophthalmology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepehr Kobravi
- Department of Oral and Maxillofacial Surgery, Tehran Azad University, Tehran, Iran
| | - Samaneh Talebi
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Yousefi
- Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
44
|
Sun J, Feng Q, He Y, Wang M, Wu Y. Lactate activates CCL18 expression via H3K18 lactylation in macrophages to promote tumorigenesis of ovarian cancer. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1373-1386. [PMID: 39010846 PMCID: PMC11543520 DOI: 10.3724/abbs.2024111] [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: 09/10/2023] [Accepted: 02/02/2024] [Indexed: 07/17/2024] Open
Abstract
This study investigates the role of lactate in the genesis and progression of ovarian cancer (OV) and explores the underlying mechanisms. Serum lactate levels show a positive correlation with tumor grade and poor prognosis in patients with OV. Bioinformatics analysis identifies CCL18 as a lactate-related gene in OV. CCL18 is up-regulated in cancerous tissues and positively related to serum lactate levels in OV patients. THP-1 cells are exposed to phorbol-12-myristate-13-acetate for M0 macrophage induction. The results of RT-qPCR and ELISA for M1/M2 macrophage-related markers and inflammatory cytokines show that the exposure of lactate to macrophages induces M2 polarization. Based on the coculture of OV cells with macrophages, lactate-treated macrophages induces a significant increase in the proliferation and migration of OV cells. However, these effects can be reversed by silencing of Gpr132 in macrophages or treatment with anti-CCL18 antibody. Experiments using the xenograft model verify that the oncogenic role of lactate in tumor growth and metastasis relies on Gpr132 and CCL18. ChIP-qPCR and luciferase reporter assays reveal that lactate regulates CCL18 expression via H3K18 lactylation. In conclusion, lactate is a potential therapeutic target for OV. It is involved in tumorigenesis by activating CCL18 expression via H3K18 lactylation in macrophages.
Collapse
Affiliation(s)
- Jinrui Sun
- Department of GynecologyShanxi Provincial People’s HospitalTaiyuan030001China
- Department of Gynecologic OncologyBeijing Obstetrics and Gynecology HospitalCapital Medical UniversityBeijing Maternal and Child Health Care HospitalBeijing100006China
| | - Qinmei Feng
- Department of GynecologyShanxi Provincial People’s HospitalTaiyuan030001China
| | - Yue He
- Department of Gynecologic OncologyBeijing Obstetrics and Gynecology HospitalCapital Medical UniversityBeijing Maternal and Child Health Care HospitalBeijing100006China
| | - Ming Wang
- Department of Gynecologic OncologyBeijing Obstetrics and Gynecology HospitalCapital Medical UniversityBeijing Maternal and Child Health Care HospitalBeijing100006China
| | - Yumei Wu
- Department of Gynecologic OncologyBeijing Obstetrics and Gynecology HospitalCapital Medical UniversityBeijing Maternal and Child Health Care HospitalBeijing100006China
| |
Collapse
|
45
|
Jiang X, Zhu Z, Ding L, Du W, Pei D. ALKBH4 impedes 5-FU Sensitivity through suppressing GSDME induced pyroptosis in gastric cancer. Cell Death Dis 2024; 15:435. [PMID: 38902235 PMCID: PMC11189908 DOI: 10.1038/s41419-024-06832-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024]
Abstract
5-Fluorouracil (5-FU) is the primary treatment option for advanced gastric cancer. However, the current challenge lies in the absence of validated biomarkers to accurately predict the efficacy and sensitivity of 5-FU in individual patients. It has been confirmed that 5-FU can regulate tumor progression by promoting gasdermin E (GSDME, encoded by DFNA5) cleavage to induce pyroptosis. Lysine demethylase ALKBH4 has been shown to be upregulated in a variety of tumors to promote tumor progression. However, its role in gastric cancer is not clear. In this study, we observed a significant upregulation of ALKBH4 expression in gastric cancer tissues compared to adjacent normal tissues, indicating its potential as a predictor for the poor prognosis of gastric cancer patients. On the contrary, GSDME exhibits low expression levels in gastric cancer and demonstrates a negative correlation with poor prognosis among patients diagnosed with gastric cancer. In addition, we also found that high expression of ALKBH4 can inhibit pyroptosis and promote the proliferation of gastric cancer cells. Mechanistically, ALKBH4 inhibits GSDME activation at the transcriptional level by inhibiting H3K4me3 histone modification in the GSDME promoter region, thereby reducing the sensitivity of gastric cancer cells to 5-FU treatment. These findings provide further insight into the regulatory mechanisms of ALKBH4 in the progression of gastric cancer and underscore its potential as a prognostic marker for predicting the sensitivity of gastric cancer cells to 5-FU treatment.
Collapse
Affiliation(s)
- Xin Jiang
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, 221004, China
| | - Zhiman Zhu
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, 221004, China
| | - Lina Ding
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, 221004, China
| | - Wenqi Du
- Department of Human Anatomy, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Dongsheng Pei
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, 221004, China.
| |
Collapse
|
46
|
Jiang YK, Li W, Qiu YY, Yue M. Advances in targeted therapy for human epidermal growth factor receptor 2 positive in advanced gastric cancer. World J Gastrointest Oncol 2024; 16:2318-2334. [PMID: 38994153 PMCID: PMC11236256 DOI: 10.4251/wjgo.v16.i6.2318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 04/04/2024] [Accepted: 04/18/2024] [Indexed: 06/13/2024] Open
Abstract
Emerging therapeutic methods represented by targeted therapy are effective supplements to traditional first-line chemoradiotherapy resistance. Human epidermal growth factor receptor 2 (HER2) is one of the most important targets in targeted therapy for gastric cancer. Trastuzumab combined with chemotherapy has been used as the first-line treatment for advanced gastric cancer. The safety and efficacy of pertuzumab and margetuximab in the treatment of gastric cancer have been verified. However, monoclonal antibodies, due to their large molecular weight, inability to penetrate the blood-brain barrier, and drug resistance, lead to decreased therapeutic efficacy, so it is necessary to explore the efficacy of other HER2-targeting therapies in gastric cancer. Small-molecule tyrosine kinase inhibitors, such as lapatinib and pyrrotinib, have the advantages of small molecular weight, penetrating the blood-brain barrier and high oral bioavailability, and are expected to become the drugs of choice for perioperative treatment and neoadjuvant therapy of gastric cancer after validation by large-scale clinical trials in the future. Antibo-drug conjugate, such as T-DM1 and T-DXd, can overcome the resistance of monoclonal antibodies despite their different mechanisms of tumor killing, and are a supplement for the treatment of patients who have failed the treatment of monoclonal antibodies such as trastuzumab. Therefore, after more detailed stratification of gastric cancer patients, various gastric cancer drugs targeting HER2 are expected to play a more significant role.
Collapse
Affiliation(s)
- Ya-Kun Jiang
- Department of Gastroenterology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, Shandong Province, China
| | - Wei Li
- Health Management Center, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, Shandong Province, China
| | - Ying-Yang Qiu
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Meng Yue
- Department of Gastroenterology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, Shandong Province, China
| |
Collapse
|
47
|
Li K, Xie T, Li Y, Huang X. LncRNAs act as modulators of macrophages within the tumor microenvironment. Carcinogenesis 2024; 45:363-377. [PMID: 38459912 DOI: 10.1093/carcin/bgae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/21/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024] Open
Abstract
Long non-coding RNAs (lncRNAs) have been established as pivotal players in various cellular processes, encompassing the regulation of transcription, translation and post-translational modulation of proteins, thereby influencing cellular functions. Notably, lncRNAs exert a regulatory influence on diverse biological processes, particularly in the context of tumor development. Tumor-associated macrophages (TAMs) exhibit the M2 phenotype, exerting significant impact on crucial processes such as tumor initiation, angiogenesis, metastasis and immune evasion. Elevated infiltration of TAMs into the tumor microenvironment (TME) is closely associated with a poor prognosis in various cancers. LncRNAs within TAMs play a direct role in regulating cellular processes. Functioning as integral components of tumor-derived exosomes, lncRNAs prompt the M2-like polarization of macrophages. Concurrently, reports indicate that lncRNAs in tumor cells contribute to the expression and release of molecules that modulate TAMs within the TME. These actions of lncRNAs induce the recruitment, infiltration and M2 polarization of TAMs, thereby providing critical support for tumor development. In this review, we survey recent studies elucidating the impact of lncRNAs on macrophage recruitment, polarization and function across different types of cancers.
Collapse
Affiliation(s)
- Kangning Li
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
- HuanKui Academy, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Tao Xie
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yong Li
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xuan Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
48
|
Syrnioti A, Petousis S, Newman LA, Margioula-Siarkou C, Papamitsou T, Dinas K, Koletsa T. Triple Negative Breast Cancer: Molecular Subtype-Specific Immune Landscapes with Therapeutic Implications. Cancers (Basel) 2024; 16:2094. [PMID: 38893213 PMCID: PMC11171372 DOI: 10.3390/cancers16112094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Triple Negative Breast Cancer (TNBC) is characterized by distinct molecular subtypes with unique biological and clinical features. This systematic review aimed to identify articles examining the differences in the tumor immune microenvironment (TIME) across different TNBC molecular subtypes. Six studies meeting inclusion criteria were analyzed, utilizing gene expression profiling and bioinformatic analyses to classify TNBC samples into molecular subtypes, as well as immunohistochemistry and cell deconvolution methods to characterize the TIME. Results revealed significant heterogeneity in immune cell composition among TNBC subtypes, with the immunomodulatory (IM) subtype demonstrating robust immune infiltration, composed mainly of adaptive immune cells along with an increased density of CTLA-4+ and PD-1+ TILs, high PD-L1 tumor cell expression, and upregulation of FOXP3+ Tregs. A more immunosuppressive TIME with a predominance of innate immune cells and lower levels of tumor-infiltrating lymphocytes (TILs) was observed in luminal androgen receptor (LAR) tumors. In mesenchymal stem-like (MSL) tumors, the TIME was mainly composed of innate immune cells, with a high number of M2 tumor-associated macrophages (TAMs), while the BL and M tumors displayed poor adaptive and innate immune responses, indicating an "immune-cold" phenotype. Differential activation of signaling pathways, genomic diversity, and metabolic reprogramming were identified as contributors to TIME heterogeneity. Understanding this interplay is crucial for tailoring therapeutic strategies, especially regarding immunotherapy.
Collapse
Affiliation(s)
- Antonia Syrnioti
- Department of Pathology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Stamatios Petousis
- 2nd Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (S.P.); (K.D.)
| | - Lisa A. Newman
- Department of Breast Surgery, New York Presbyterian-Weill Cornell Medicine, New York, NY 10065, USA;
| | - Chrysoula Margioula-Siarkou
- MSc Program in Gynaecologic Oncology and Breast Oncology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Theodora Papamitsou
- Laboratory of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Konstantinos Dinas
- 2nd Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (S.P.); (K.D.)
| | - Triantafyllia Koletsa
- Department of Pathology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| |
Collapse
|
49
|
Zheng Y, Yao J, Liu J, Zhao F. An unusual cause of significantly elevated blood alpha-fetoprotein levels: a case report and literature review. Front Oncol 2024; 14:1393074. [PMID: 38812781 PMCID: PMC11133681 DOI: 10.3389/fonc.2024.1393074] [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: 02/28/2024] [Accepted: 04/18/2024] [Indexed: 05/31/2024] Open
Abstract
Alpha-fetoprotein (AFP) serves as a crucial diagnostic marker for primary hepatocellular carcinoma (HCC) and germ cell tumors (GCTs), with rare instances of significantly elevated levels in other diseases. In this study, we present a case of an elderly patient who was diagnosed with AFP-producing gastric cancer (AFPGC) following an elevated AFP result during physical examination. In investigating liver cancer at an early stage, the diagnosis was missed because of failure in detecting the lesion, resulting in delayed treatment initiation. AFPGC is a rare aggressive tumor that demands heightened awareness among clinicians to foster early detection, diagnosis, and treatment for improved prognosis.
Collapse
Affiliation(s)
- Yue Zheng
- Department of General Medicine, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Jianping Yao
- Department of Endocrinology, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Jin Liu
- Department of Pathology, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Feimin Zhao
- Department of General Medicine, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, China
| |
Collapse
|
50
|
Wei R, Song J, Pan H, Liu X, Gao J. CPT1C-positive cancer-associated fibroblast facilitates immunosuppression through promoting IL-6-induced M2-like phenotype of macrophage. Oncoimmunology 2024; 13:2352179. [PMID: 38746869 PMCID: PMC11093039 DOI: 10.1080/2162402x.2024.2352179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/02/2024] [Indexed: 06/05/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) exhibit remarkable phenotypic heterogeneity, with specific subsets implicated in immunosuppression in various malignancies. However, whether and how they attenuate anti-tumor immunity in gastric cancer (GC) remains elusive. CPT1C, a unique isoform of carnitine palmitoyltransferase pivotal in regulating fatty acid oxidation, is briefly indicated as a protumoral metabolic mediator in the tumor microenvironment (TME) of GC. In the present study, we initially identified specific subsets of fibroblasts exclusively overexpressing CPT1C, hereby termed them as CPT1C+CAFs. Subsequent findings indicated that CPT1C+CAFs fostered a stroma-enriched and immunosuppressive TME as they correlated with extracellular matrix-related molecular features and enrichment of both immunosuppressive subsets, especially M2-like macrophages, and multiple immune-related pathways. Next, we identified that CPT1C+CAFs promoted the M2-like phenotype of macrophage in vitro. Bioinformatic analyses unveiled the robust IL-6 signaling between CPT1C+CAFs and M2-like phenotype of macrophage and identified CPT1C+CAFs as the primary source of IL-6. Meanwhile, suppressing CPT1C expression in CAFs significantly decreased IL-6 secretion in vitro. Lastly, we demonstrated the association of CPT1C+CAFs with therapeutic resistance. Notably, GC patients with high CPT1C+CAFs infiltration responded poorly to immunotherapy in clinical cohort. Collectively, our data not only present the novel identification of CPT1C+CAFs as immunosuppressive subsets in TME of GC, but also reveal the underlying mechanism that CPT1C+CAFs impair tumor immunity by secreting IL-6 to induce the immunosuppressive M2-like phenotype of macrophage in GC.
Collapse
Affiliation(s)
- Rongyuan Wei
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Junquan Song
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongda Pan
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaowen Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianpeng Gao
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|