1
|
Liu Q, Yu M, Lin Z, Wu L, Xia P, Zhu M, Huang B, Wu W, Zhang R, Li K, Zhu L, Wang Q. COL1A1-positive endothelial cells promote gastric cancer progression via the ANGPTL4-SDC4 axis driven by endothelial-to-mesenchymal transition. Cancer Lett 2025; 623:217731. [PMID: 40254092 DOI: 10.1016/j.canlet.2025.217731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/19/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Gastric cancer (GC) is an aggressive and heterogeneous disease with poor survival outcomes. The progression of GC involves complex, multi-step processes. Endothelial cells (ECs) play a crucial role in tumor angiogenesis, proliferation, invasion, and metastasis, particularly through the process of endothelial-to-mesenchymal transition (EndoMT). However, the specific role and mechanisms of EndoMT in gastric cancer remain unclear. Based on 6 GC single-cell RNA-sequencing (scRNA-seq) cohorts (samples = 97), we established an EndoMT-related gene signature, termed EdMTS. Leveraging this gene signature, ssGSEA was applied to calculate sample scores across multiple bulk RNA-seq datasets, which include information on immunotherapy, metastasis, GC progression, and survival. Moreover, we applied the Monocle2 method to calculate cell pseudotime and used CellChat to analyze interactions between malignant and EC cells. We verified the molecular mechanism by multiple immunofluorescence and cell function experiments. Findings In this study, we established a single-cell atlas of ECs in GC and identified a subpopulation of COL1A1+ ECs that play a critical role in tumor progression and metastasis. These COL1A1+ ECs were significantly associated with worse clinical outcomes in GC patients. Further analysis revealed that COL1A1+ ECs originated from lymphatic ECs and underwent EndoMT through the upregulation of CEBPB, driving tumor invasiveness. Moreover, COL1A1+ ECs interacted with malignant cells via ANGPTL4-SDC4 axis, enhancing invasion and migration. These findings provide a deeper understanding of the role of COL1A1+ ECs in GC progression and highlight potential therapeutic targets for disrupting the EndoMT process in these cells to provide a benefit for GC patients.
Collapse
Affiliation(s)
- Quanzhong Liu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Miao Yu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Zihan Lin
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Lingxiang Wu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Peng Xia
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Mengyan Zhu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Bin Huang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Wei Wu
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Ruohan Zhang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Kening Li
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Lingjun Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Qianghu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China.
| |
Collapse
|
2
|
Gou M, Zhang H, Qian N, Zhang Y, Sun Z, Li G, Wang Z, Dai G. Deep learning radiomics analysis for prediction of survival in patients with unresectable gastric cancer receiving immunotherapy. Eur J Radiol Open 2025; 14:100626. [PMID: 39807092 PMCID: PMC11728962 DOI: 10.1016/j.ejro.2024.100626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/03/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
Objective Immunotherapy has become an option for the first-line therapy of advanced gastric cancer (GC), with improved survival. Our study aimed to investigate unresectable GC from an imaging perspective combined with clinicopathological variables to identify patients who were most likely to benefit from immunotherapy. Method Patients with unresectable GC who were consecutively treated with immunotherapy at two different medical centers of Chinese PLA General Hospital were included and divided into the training and validation cohorts, respectively. A deep learning neural network, using a multimodal ensemble approach based on CT imaging data before immunotherapy, was trained in the training cohort to predict survival, and an internal validation cohort was constructed to select the optimal ensemble model. Data from another cohort were used for external validation. The area under the receiver operating characteristic curve was analyzed to evaluate performance in predicting survival. Detailed clinicopathological data and peripheral blood prior to immunotherapy were collected for each patient. Univariate and multivariable logistic regression analysis of imaging models and clinicopathological variables was also applied to identify the independent predictors of survival. A nomogram based on multivariable logistic regression was constructed. Result A total of 79 GC patients in the training cohort and 97 patients in the external validation cohort were enrolled in this study. A multi-model ensemble approach was applied to train a model to predict the 1-year survival of GC patients. Compared to individual models, the ensemble model showed improvement in performance metrics in both the internal and external validation cohorts. There was a significant difference in overall survival (OS) among patients with different imaging models based on the optimum cutoff score of 0.5 (HR = 0.20, 95 % CI: 0.10-0.37, P < 0.001). Multivariate Cox regression analysis revealed that the imaging models, PD-L1 expression, and lung immune prognostic index were independent prognostic factors for OS. We combined these variables and built a nomogram. The calibration curves showed that the C-index of the nomogram was 0.85 and 0.78 in the training and validation cohorts. Conclusion The deep learning model in combination with several clinical factors showed predictive value for survival in patients with unresectable GC receiving immunotherapy.
Collapse
Affiliation(s)
- Miaomiao Gou
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Hongtao Zhang
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Niansong Qian
- Department of Thoracic Oncology, The Eighth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Yong Zhang
- Department of Medical Oncology, The Second Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Zeyu Sun
- R&D Center, Keya Medical Technology Co., Ltd, Beijing, PR China
| | - Guang Li
- R&D Center, Keya Medical Technology Co., Ltd, Beijing, PR China
| | - Zhikuan Wang
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Guanghai Dai
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| |
Collapse
|
3
|
Jiang Y, Immadi MS, Wang D, Zeng S, On Chan Y, Zhou J, Xu D, Joshi T. IRnet: Immunotherapy response prediction using pathway knowledge-informed graph neural network. J Adv Res 2025; 72:319-331. [PMID: 39097091 DOI: 10.1016/j.jare.2024.07.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/10/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024] Open
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) are potent and precise therapies for various cancer types, significantly improving survival rates in patients who respond positively to them. However, only a minority of patients benefit from ICI treatments. OBJECTIVES Identifying ICI responders before treatment could greatly conserve medical resources, minimize potential drug side effects, and expedite the search for alternative therapies. Our goal is to introduce a novel deep-learning method to predict ICI treatment responses in cancer patients. METHODS The proposed deep-learning framework leverages graph neural network and biological pathway knowledge. We trained and tested our method using ICI-treated patients' data from several clinical trials covering melanoma, gastric cancer, and bladder cancer. RESULTS Our results demonstrate that this predictive model outperforms current state-of-the-art methods and tumor microenvironment-based predictors. Additionally, the model quantifies the importance of pathways, pathway interactions, and genes in its predictions. A web server for IRnet has been developed and deployed, providing broad accessibility to users at https://irnet.missouri.edu. CONCLUSION IRnet is a competitive tool for predicting patient responses to immunotherapy, specifically ICIs. Its interpretability also offers valuable insights into the mechanisms underlying ICI treatments.
Collapse
Affiliation(s)
- Yuexu Jiang
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri-Columbia, Columbia, MO, USA
| | - Manish Sridhar Immadi
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, Columbia, MO, USA
| | - Duolin Wang
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri-Columbia, Columbia, MO, USA
| | - Shuai Zeng
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri-Columbia, Columbia, MO, USA
| | - Yen On Chan
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, Columbia, MO, USA; MU Institute for Data Science and Informatics, University of Missouri-Columbia, Columbia, MO, USA
| | - Jing Zhou
- Department of Surgery, University of Missouri-Columbia, Columbia, MO, USA
| | - Dong Xu
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri-Columbia, Columbia, MO, USA; MU Institute for Data Science and Informatics, University of Missouri-Columbia, Columbia, MO, USA
| | - Trupti Joshi
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri-Columbia, Columbia, MO, USA; MU Institute for Data Science and Informatics, University of Missouri-Columbia, Columbia, MO, USA; Department of Biomedical Informatics, Biostatistics and Medical Epidemiology, University of Missouri-Columbia, Columbia, MO, USA.
| |
Collapse
|
4
|
Kim YS. Gastric Carcinoma. Curr Top Microbiol Immunol 2025. [PMID: 40423781 DOI: 10.1007/82_2025_303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Epstein-Barr virus (EBV)-associated gastric cancers (EBVaGCs) account for about 10% of gastric cancers globally, with higher prevalence in East Asia and Latin America. These cancers develop through a "gastritis-infection-cancer sequence" and are characterized by unique molecular signatures, including CpG island methylator phenotype and mutations in ARID1A and PIK3CA genes. EBVaGCs typically present in the proximal stomach with diffuse-type histology and dense lymphocytic infiltration. Key viral proteins EBNA1 and LMP2A drive oncogenesis by altering cellular processes and immune responses. The IFN-γ signature and extensive epigenetic modifications contribute to their distinct profile. Despite often presenting at advanced stages, EBVaGCs generally have a more favorable prognosis. EBV employs sophisticated strategies to evade immune detection, utilizing latent proteins and noncoding RNAs. Paradoxically, despite an immune-hot environment, EBVaGCs demonstrate effective immune evasion, partly due to the expression of immune checkpoint molecules like PD-L1 and LAG3. Treatment approaches vary based on disease stage, from endoscopic resection for early-stage cancers to systemic therapies for advanced cases. Immunotherapy, particularly PD-1/PD-L1 inhibitors, shows promising results. Emerging research suggests combining these with LAG3 inhibitors may enhance efficacy. Ongoing research and advanced genomic techniques continue to reveal new insights, paving the way for personalized therapies and novel diagnostic approaches.
Collapse
Affiliation(s)
- Young-Sik Kim
- Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Li J, Xu N, Hu L, Xu J, Huang Y, Wang D, Chen F, Wang Y, Jiang J, Hong Y, Ye H. Chaperonin containing TCP1 subunit 5 as a novel pan-cancer prognostic biomarker for tumor stemness and immunotherapy response: insights from multi-omics data, integrated machine learning, and experimental validation. Cancer Immunol Immunother 2025; 74:224. [PMID: 40423850 DOI: 10.1007/s00262-025-04071-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Accepted: 04/28/2025] [Indexed: 05/28/2025]
Abstract
BACKGROUND Chaperonin containing TCP1 subunit 5 (CCT5), a vital component of the molecular chaperonin complex, has been implicated in tumorigenesis, cancer stemness maintenance, and therapeutic resistance. Nevertheless, its comprehensive roles in pan-cancer progression, underlying biological functions, and potential as a predictor of immunotherapy response remains poorly understood. METHODS We performed a comprehensive multi-omics pan-cancer analysis of CCT5 across 33 cancer types, integrating bulk RNA-seq, single-cell RNA-seq (scRNA-seq), and spatial transcriptomics data. CCT5 expression patterns, prognostic relevance, stemness association, and immune microenvironment relationships were evaluated. A novel CCT5-based signature (CCT5.Sig) was developed using machine learning on 23 immune checkpoint blockade (ICB) cohorts (n = 1394) spanning eight cancer types. Model performance was assessed using AUC metrics and survival analyses. RESULTS CCT5 was significantly overexpressed in tumor tissues and primarily localized to malignant and cycling cells. High CCT5 expression correlated with poor prognosis in multiple cancers and was enriched in oncogenic, cell cycle, and DNA damage repair pathways. CCT5 expression was positively associated with mRNAsi, mDNAsi, and CytoTRACE scores, indicating a role in stemness maintenance. Furthermore, CCT5-high tumors exhibited immune-cold phenotypes, with reduced TILs and CD8⁺ T cell activity. The CCT5.Sig model, based on genes co-expressed with CCT5, achieved superior predictive accuracy for ICB response (AUC = 0.82 in validation and 0.76 in independent testing), outperforming existing pan-cancer signatures. CONCLUSION This study reveals the multifaceted oncogenic roles of CCT5 and highlights its potential as a pan-cancer biomarker for prognosis and immunotherapy response. The machine learning-derived CCT5.Sig model provides a robust tool for patient stratification and may inform personalized immunotherapy strategies.
Collapse
Affiliation(s)
- Jiajun Li
- The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute and Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Nuo Xu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute and Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Leyin Hu
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, 305000, Zhejiang, China
| | - Jiayue Xu
- The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yifan Huang
- The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Deqi Wang
- Department of Gastroenterology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Feng Chen
- The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yi Wang
- Department of Gastroenterology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Jiani Jiang
- Department of Gastroenterology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yanggang Hong
- The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Huajun Ye
- Department of Gastroenterology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
6
|
Yu K, Cao Y, Zhang Z, Wang L, Gu Y, Xu T, Zhang X, Guo X, Shen Z, Qin J. Blockade of CLEVER-1 restrains immune evasion and enhances anti-PD-1 immunotherapy in gastric cancer. J Immunother Cancer 2025; 13:e011080. [PMID: 40404204 PMCID: PMC12096977 DOI: 10.1136/jitc-2024-011080] [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: 11/16/2024] [Accepted: 04/16/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) remains a major global health burden. Despite the advancements in immunotherapy for patients with GC, the heterogeneity of GC limits response rates, especially in immune "cold" subtypes, including genomically stable and epithelial-mesenchymal transition GC. Common lymphatic endothelial and vascular endothelial receptor-1 (CLEVER-1), a newly recognized immune checkpoint molecule predominantly expressed on tumor-associated macrophages (TAMs), remains poorly understood in GC. This study aims to explore the clinical significance of CLEVER-1+TAM infiltration, elucidate its role in modulating the tumor immune landscape, and investigate the therapeutic potential of CLEVER-1 blockade in enhancing immunotherapy. METHODS This study analyzed two independent GC cohorts and single-cell RNA sequencing data (GSE183904). CLEVER-1 expression in TAMs was assessed via multiplex immunofluorescence, flow cytometry, and RNA sequencing. The clinical relevance of CLEVER-1+TAM infiltration was evaluated in relation to tumor, node, metastases staging, molecular subtypes, prognosis, and immunochemotherapy response. Transcriptomic and pathway analyses characterized the immunophenotype of CLEVER-1+TAMs. Functional assays examined their suppression on CD8+T cells, while interventional experiments assessed the impact of CLEVER-1 blockade alone or with programmed cell death protein-1 (PD-1) inhibition. RESULTS CLEVER-1 was predominantly expressed on TAMs in GC and was associated with worse clinical outcomes. Transcriptomic and phenotypic analyses revealed that CLEVER-1+TAMs display a dynamic immunophenotype and critically suppress T-cell function, fostering an immunosuppressive microenvironment. High CLEVER-1+TAM infiltration was linked to poor response or adaptive resistance to PD-1 blockade therapy. CLEVER-1 blockade reprogrammed TAMs toward a pro-inflammatory phenotype, resulting in enhanced CD8+T cell cytotoxicity and proliferation. Co-targeting CLEVER-1 and PD-1 synergistically enhanced antitumor responses. CONCLUSIONS High infiltration of CLEVER-1+TAMs indicates immune suppression and poor prognosis in GC. The combination of CLEVER-1 and PD-1 blockade emerges as a dual-pronged strategy to boost immune-mediated tumor control and prevent treatment relapse in GC.
Collapse
Affiliation(s)
- Kuan Yu
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yifan Cao
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital Fudan University, Shanghai, China
| | - Zihao Zhang
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital Fudan University, Shanghai, China
| | - Leihao Wang
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yichao Gu
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital Fudan University, Shanghai, China
| | - Tianwei Xu
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
| | - Xiaolei Zhang
- Department of Pathology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Xinxin Guo
- Department of Pathology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Zhenbin Shen
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital Fudan University, Shanghai, China
| | - Jing Qin
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital Fudan University, Shanghai, China
| |
Collapse
|
7
|
Yang L, Li H, Xia M, Pu X. Novel Composite Scoring System for Predicting Prognosis in Stage IV Gastric Cancer Patients Treated with Immune Checkpoint Inhibitors. J Inflamm Res 2025; 18:6491-6504. [PMID: 40421267 PMCID: PMC12105669 DOI: 10.2147/jir.s519724] [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: 02/17/2025] [Accepted: 04/25/2025] [Indexed: 05/28/2025] Open
Abstract
Background Gastric cancer (GC) with distant metastases has a poor prognosis, and immune checkpoint inhibitors (ICIs) effectively improve the survival time of patients with this disease. This study aimed to identify effective prognostic markers that can predict the treatment effect of ICIs in patients with stage IV GC. Methods This study included 256 patients with GC with distant metastases who had received treatment with ICIs. A receiver operating characteristic (ROC) curve was used to analyze the predictive ability and optimal cutoff values of immune-inflammatory markers. Kaplan‒Meier survival curves were used to analyze the differences in progression-free survival (PFS) and overall survival (OS) among patients. Cox proportional hazard regression analysis was used to identify independent prognostic factors for PFS and OS. Results By comparing the area under the ROC curve (AUC) of immune-inflammatory markers, we selected the preoperative platelet count/(lymphocyte count × prealbumin count) ratio and fibrinogen/albumin ratio to form a combined score (PLPR-FAR score). The ROC curve revealed that when the PLPR-FAR score was used to predict patient PFS and OS, the AUC were 0.614 and 0.672, respectively. The Kaplan‒Meier survival curve revealed that patients with higher PLPR-FAR scores had significantly shorter PFS and OS than those with lower PLPR-FAR scores. Cox proportional hazard regression analysis revealed that the PLPR-FAR score was an independent risk factor for PFS and OS in stage IV GC patients. Conclusion The PLPR-FAR score may help identify which patients are more likely to benefit from ICIs treatment, and could serve as a novel and promising prognostic biomarker.
Collapse
Affiliation(s)
- Lingbing Yang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Hongwei Li
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Mingyu Xia
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Xiaomeng Pu
- School of Stomatology, Gansu Health Vocational College, Lanzhou, People’s Republic of China
| |
Collapse
|
8
|
Lee KW, Zang DY, Kim HD, Kim JW, Kim BJ, Kang YK, Ryu MH, Kim HK. Multicenter phase Ib/II study of second-line durvalumab and tremelimumab in combination with paclitaxel in patients with biomarker-selected metastatic gastric cancer. Br J Cancer 2025:10.1038/s41416-025-03052-y. [PMID: 40399487 DOI: 10.1038/s41416-025-03052-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 04/24/2025] [Accepted: 05/01/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND This multicenter phase Ib/II trial aimed to evaluate the safety and efficacy of combining durvalumab, tremelimumab, and paclitaxel as second-line treatment for biomarker-selected patients with metastatic gastric cancer. METHODS In phase Ib, the standard 3 + 3 dose escalation method was used. Durvalumab and tremelimumab were administered every 4 weeks for 13 and 4 cycles, respectively, combining paclitaxel 80 mg/m2 (dose level 2) or 60 mg/m2 (dose level 1) on days 1, 8, and 15. The primary outcome for phase II was the objective response rate (ORR). RESULTS In phase Ib (n = 7), dose level-1 was selected as the recommended phase II dose. In phase II, 48 patients were enrolled: microsatellite instability-high or deficient mismatch repair protein tumors (n = 16); EBV-positive tumors (n = 15); high tumor mutation burden ( ≥ 5/Mb) (n = 11); CD274 amplification (n = 5); and POLD1 mutation (n = 1). The ORR was 52.1%, meeting the primary endpoint. The median progression-free survival and overall survival were 5.3 and 13.1 months, respectively. The most common any-grade and grade 3-4 adverse events were anemia (41.7%) and neutropenia (10.4%), respectively. CONCLUSIONS Durvalumab-tremelimumab with paclitaxel was tolerable and efficacious in biomarker-selected gastric cancer patients as a second-line treatment, highlighting the importance of biomarker-based approaches for immunotherapy in gastric cancer. CLINICAL TRIAL REGISTRATION NCT03751761.
Collapse
Affiliation(s)
- Keun Wook Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Da Young Zang
- Division of Hematology-Oncology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Republic of Korea
| | - Hyung-Don Kim
- Departmentof Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin-Won Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Bum Jun Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Republic of Korea
| | - Yoon-Koo Kang
- Departmentof Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Min-Hee Ryu
- Departmentof Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Hark Kyun Kim
- Center for Gastric Cancer, National Cancer Center, Goyang, Republic of Korea.
| |
Collapse
|
9
|
Wu Y, Liu J, Yin T, Li X, Liu X, Peng X, Zhan X. SELP can affect the immune microenvironment of gastric cancer and is associated with poor prognosis. Discov Oncol 2025; 16:846. [PMID: 40397261 PMCID: PMC12095770 DOI: 10.1007/s12672-025-02629-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 05/08/2025] [Indexed: 05/22/2025] Open
Abstract
The tumor microenvironment (TME) plays a crucial role in the occurrence and progression of gastric cancer. Yet, we still don't understand how immune and stromal components of TMEs are modulated. In this study, we applied the ESTIMATE algorithm to calculate the number of immune and stromal components in 410 STAD cases in the Cancer Genome Atlas (TCGA) database. COX regression analysis and protein-protein interaction (PPI) network construction were used to analyze differentially expressed genes (DEGs). Then, P-selectin (SELP) was identified as a predictor by cross-analysis of univariate COX and PPI. After verifying the clinical significance of SELP for study, we performed an immune infiltration analysis and identified 54 immunomodulators associated with SELP through public data. Immunomodulation associated with gastric cancer prognosis was then confirmed by LASSO regression, and the previous results were further validated with single-cell data. Finally, we verified that SELP can promote EMT on gastric cancer cells. In conclusion, we validated that SELP may affect the biological phenotype of gastric cancer with the immune microenvironment alteration of gastric cancer.
Collapse
Affiliation(s)
- Yue Wu
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Jingyu Liu
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Tong Yin
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Xiaoxiao Li
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Xian Liu
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Xiaobo Peng
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China.
| | - Xianbao Zhan
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
10
|
Li S, Zhou X, Feng H, Huang K, Chen M, Lin M, Lin H, Deng Z, Chen Y, Liao W, Zhang Z, Chen J, Guan B, Su T, Feng Z, Shu G, Yu A, Pan Y, Fu L. Deciphering the Immunomodulatory Function of GSN + Inflammatory Cancer-Associated Fibroblasts in Renal Cell Carcinoma Immunotherapy: Insights From Pan-Cancer Single-Cell Landscape and Spatial Transcriptomics Analysis. Cell Prolif 2025:e70062. [PMID: 40375605 DOI: 10.1111/cpr.70062] [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: 03/04/2025] [Revised: 04/13/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025] Open
Abstract
The heterogeneity of cancer-associated fibroblasts (CAFs) could affect the response to immune checkpoint inhibitor (ICI) therapy. However, limited studies have investigated the role of inflammatory CAFs (iCAFs) in ICI therapy using pan-cancer single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics sequencing (ST-seq) analysis. We performed pan-cancer scRNA-seq and ST-seq analyses to identify the subtype of GSN+ iCAFs, exploring its spatial distribution characteristics in the context of ICI therapy. The pan-cancer scRNA-seq and bulk RNA-seq data are incorporated to develop the Caf.Sig model, which predicts ICI response based on CAF gene signatures and machine learning approaches. Comprehensive scRNA-seq analysis, along with in vivo and in vitro experiments, investigates the mechanisms by which GSN+ iCAFs influence ICI efficacy. The Caf.Sig model demonstrates well performances in predicting ICI therapy response in pan-cancer patients. A higher proportion of GSN+ iCAFs is observed in ICI non-responders compared to responders in the pan-cancer landscape and clear cell renal cell carcinoma (ccRCC). Using real-world immunotherapy data, the Caf.Sig model accurately predicts ICI response in pan-cancer, potentially linked to interactions between GSN+ iCAFs and CD8+ Tex cells. ST-seq analysis confirms that interactions and cellular distances between GSN+ iCAFs and CD8+ exhausted T (Tex) cells impact ICI efficacy. In a co-culture system of primary CAFs, primary tumour cells and CD8+ T cells, downregulation of GSN on CAFs drives CD8+ T cells towards a dysfunctional state in ccRCC. In a subcutaneously tumour-grafted mouse model, combining GSN overexpression with ICI treatment achieves optimal efficacy in ccRCC. Our study provides the Caf.Sig model as an outperforming approach for patient selection of ICI therapy, and advances our understanding of CAF biology and suggests potential therapeutic strategies for upregulating GSN in CAFs in cancer immunotherapy.
Collapse
Affiliation(s)
- Shan Li
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Uro-Oncology Institute of Central South University, Changsha, Hunan, China
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xinwei Zhou
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Haoqian Feng
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Kangbo Huang
- Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Minyu Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Mingjie Lin
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hansen Lin
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zebing Deng
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Uro-Oncology Institute of Central South University, Changsha, Hunan, China
| | - Yuhang Chen
- Department of Geniturinary Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wuyuan Liao
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhengkun Zhang
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jinwei Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Bohong Guan
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Tian Su
- Department of Pediatric Intensive Care Unit (PICU), Guangdong Provincial People's Hospital Heyuan Hospital, Heyuan, Guangdong, China
| | - Zihao Feng
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Guannan Shu
- Department of Urology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou Institute of Pediatrics, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Anze Yu
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yihui Pan
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Liangmin Fu
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Uro-Oncology Institute of Central South University, Changsha, Hunan, China
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Disease, Changsha, Hunan, China
| |
Collapse
|
11
|
He M, Ji C, Li Z, Chen S, Gao J, Shen L, Zhang C. Circulating tumor DNA predicts clinical benefits of immune checkpoint blockade in HER2-negative patients with advanced gastric cancer. Gastric Cancer 2025:10.1007/s10120-025-01621-x. [PMID: 40372586 DOI: 10.1007/s10120-025-01621-x] [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: 09/25/2024] [Accepted: 04/29/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are becoming more prominent in the treatment of gastric cancer (GC). However, predictive biomarkers of response to ICIs in HER2-negative patients remain incompletely understood. METHODS A total of 47 patients diagnosed with HER2-negative advanced GC who underwent ICI regimens were eligible for this study. Plasma samples with paired white blood cells prior to treatments were collected from these 47 patients. Variations of circulating tumor DNA (ctDNA) was evaluated by next-generation sequencing followed by its significance analysis. RESULTS A total of 658 somatic mutations involving 203 genes were identified in all ctDNA. Mutations in MEN1, MLH1, CEBPA, ATR, GNAQ, and FOXL2 genes were more frequent in responders (P < 0.05). Compared with wild-type patients, patients with CEBPA or IRS2 mutations had prolonged median progression-free survival (mPFS, P = 0.0056). Patients with co-occurring mutations in IRS2/CEBPA, IRS2/POLD1, TP53/PIK3CA, or POLD1/CEBPA had longer mPFS compared with others (P = 0.003; 0.006; 0.0166; 0.0315; respectively). Both alteration of CDKN2A alone and co-mutations with MSH6 were significantly associated with superior overall survival (OS, P = 0.0289; 0.0355; respectively). In addition, higher on-treatment ctDNA concentration or variant allele frequency (VAF) were associated with poorer response (P < 0.05). Additionally, the increased molecular alterations of POLE, FGFR2 and MDC1 seemed to indicate the acquired resistance to ICIs. CONCLUSIONS Variation signatures captured by ctDNA as well as the kinetics of ctDNA could predict the clinical benefits of ICB in HER2-negative GC patients, which was worth further validated in large cohort.
Collapse
Affiliation(s)
- Mei He
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Congcong Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zhiwei Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Shiqing Chen
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, 201114, China
| | - Jing Gao
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Cheng Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
12
|
Walch HS, Borpatragohain R, Jee J, Chatila W, Fong C, Maron SB, Ku GY, Ilson DH, Janjigian YY, Wu AJ, Shah P, Coit DG, Bains MS, Rusch VW, Park BJ, Bott MJ, Gray K, Jones DR, Berger M, Schultz N, Strong VE, Molena D, Sihag S. Clinical Implications of The Cancer Genome Atlas Molecular Classification System in Esophagogastric Cancer. Clin Cancer Res 2025; 31:1912-1921. [PMID: 40299774 DOI: 10.1158/1078-0432.ccr-24-3473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/03/2024] [Accepted: 03/12/2025] [Indexed: 05/01/2025]
Abstract
PURPOSE The Cancer Genome Atlas (TCGA) project defined four distinct molecular subtypes of esophagogastric adenocarcinoma: microsatellite instable (MSI), Epstein-Barr virus (EBV)-associated, genomically stable (GS), and chromosomally instable (CIN). However, an association between molecular subtypes and clinical outcomes has not been clearly demonstrated. Given few actionable biomarkers, we investigated the clinical relevance of TCGA classification system. EXPERIMENTAL DESIGN We identified all patients with esophagogastric adenocarcinoma whose tumors underwent prospective next-generation sequencing using the Memorial Sloan Kettering-IMPACT assay from 2014 to 2023. We classified all tumors in accordance with TCGA methodology and correlated molecular subtypes with high-quality clinicopathologic data. RESULTS Among 1,438 included patients, 941 had CIN, 344 had GS, 103 had MSI, and 50 had EBV tumors. Accounting for the clinical stage and tumor grade, molecular classification was independently associated with overall cancer-specific survival (P < 0.001) on Cox multivariable analysis. Furthermore, genomic signatures, patient demographics, pathologic responses to neoadjuvant therapy, patterns of recurrence, and metastatic organotropism differed significantly by molecular subtype. Although most distal esophageal and gastroesophageal junction tumors were CIN, up to 25% of these included GS, MSI, or EBV subtypes in contrast to TCGA. Random forest machine learning demonstrated that the molecular subtype is more influential in predicting response to treatment than tumor location. CONCLUSIONS Molecular classification is independently prognostic and may warrant inclusion in future staging and treatment guidelines. Routine molecular profiling is clinically feasible and may play a role in the management of patients to help guide appropriate treatment selection and clinical trial enrollment in the place of tumor location.
Collapse
Affiliation(s)
- Henry S Walch
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Raktim Borpatragohain
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Justin Jee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Waleed Chatila
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christopher Fong
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Steven B Maron
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Geoffrey Y Ku
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David H Ilson
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yelena Y Janjigian
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Abraham J Wu
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pari Shah
- Gastroenterology, Hepatology, and Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniel G Coit
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Manjit S Bains
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Valerie W Rusch
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bernard J Park
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Matthew J Bott
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Katherine Gray
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David R Jones
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael Berger
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nikolaus Schultz
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vivian E Strong
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniela Molena
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Smita Sihag
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
13
|
Xiao Q, Liu Y, Li T, Wang C, He S, Zhai L, Yang Z, Zhang X, Wu Y, Liu Y. Viral oncogenesis in cancer: from mechanisms to therapeutics. Signal Transduct Target Ther 2025; 10:151. [PMID: 40350456 PMCID: PMC12066790 DOI: 10.1038/s41392-025-02197-9] [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: 08/16/2024] [Revised: 01/22/2025] [Accepted: 03/03/2025] [Indexed: 05/14/2025] Open
Abstract
The year 2024 marks the 60th anniversary of the discovery of the Epstein-Barr virus (EBV), the first virus confirmed to cause human cancer. Viral infections significantly contribute to the global cancer burden, with seven known Group 1 oncogenic viruses, including hepatitis B virus (HBV), human papillomavirus (HPV), EBV, Kaposi sarcoma-associated herpesvirus (KSHV), hepatitis C virus (HCV), human T-cell leukemia virus type 1 (HTLV-1), and human immunodeficiency virus (HIV). These oncogenic viruses induce cellular transformation and cancer development by altering various biological processes within host cells, particularly under immunosuppression or co-carcinogenic exposures. These viruses are primarily associated with hepatocellular carcinoma, gastric cancer, cervical cancer, nasopharyngeal carcinoma, Kaposi sarcoma, lymphoma, and adult T-cell leukemia/lymphoma. Understanding the mechanisms of viral oncogenesis is crucial for identifying and characterizing the early biological processes of virus-related cancers, providing new targets and strategies for treatment or prevention. This review first outlines the global epidemiology of virus-related tumors, milestone events in research, and the process by which oncogenic viruses infect target cells. It then focuses on the molecular mechanisms by which these viruses induce tumors directly or indirectly, including the regulation of oncogenes or tumor suppressor genes, induction of genomic instability, disruption of regular life cycle of cells, immune suppression, chronic inflammation, and inducing angiogenesis. Finally, current therapeutic strategies for virus-related tumors and recent advances in preclinical and clinical research are discussed.
Collapse
Affiliation(s)
- Qing Xiao
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yi Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Tingting Li
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Chaoyu Wang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Sanxiu He
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Liuyue Zhai
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Zailin Yang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaomei Zhang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| | - Yongzhong Wu
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| | - Yao Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
14
|
Sun F, Gao X, Li T, Zhao X, Zhu Y. Tumor immune microenvironment remodeling after neoadjuvant therapy in gastric cancer: Update and new challenges. Biochim Biophys Acta Rev Cancer 2025; 1880:189350. [PMID: 40355011 DOI: 10.1016/j.bbcan.2025.189350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Gastric cancer (GC) is a malignant tumor with one of the highest morbidity and death rates in the world. Neoadjuvant therapy, including neoadjuvant chemotherapy (NAC) and NAC combined with immunotherapy, can improve the resection and long-term survival rates. However, not all patients respond well to neoadjuvant therapy. It has been confirmed that immune cells in the tumor immune microenvironment, including T cells, B cells, and natural killer cells, can affect the efficacy of neoadjuvant therapy. This paper summarizes current preclinical and clinical evidence to more fully describe the effects of neoadjuvant therapy on the immune microenvironment of GC, to provide the impetus to identify biomarkers to predict the potency of neoadjuvant therapy, and to identify the mechanisms of drug resistance, which should promote the development of individualized and accurate treatments for GC patients.
Collapse
Affiliation(s)
- Fujing Sun
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Xiaozhuo Gao
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Tianming Li
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Xiaoyan Zhao
- Graduate School, Dalian Medical University, Dalian, China
| | - Yanmei Zhu
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China.
| |
Collapse
|
15
|
Xu D, Li J, Zhou L, Jin J. Identify Modules Associated with Immunotherapy Response from Mouse Tumor Profiles for Stratifying Cancer Patients. Interdiscip Sci 2025:10.1007/s12539-025-00719-1. [PMID: 40346402 DOI: 10.1007/s12539-025-00719-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 04/12/2025] [Accepted: 04/18/2025] [Indexed: 05/11/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have demonstrated significant clinical benefits in cancer treatment, but only a minority of patients exhibit favorable response, highlighting the importance of determining patients who will benefit from immunotherapy. Currently, patient datasets regarding immunotherapy response are scarce, while ample experiments can be performed on syngeneic mouse tumor models to generate valuable data. Therefore, how to effectively utilize mouse data to identify predictors of immunotherapy response and subsequently transfer relevant knowledge to predict human response to ICIs is a question worth studying. In this study, we propose a novel methodology to address this issue. Firstly, we identify gene modules associated with immunotherapy response from mouse tumor profiles based on cancer gene panels. Subsequently, these identified modules are employed to build prediction models for immunotherapy response based on mouse data. Furthermore, we transfer these models to predict ICIs responses of human cancer patients. Experimental results demonstrate that the gene modules identified from mouse data are reliable predictors of immunotherapy response. The mouse-based models built on these modules could be transferred to humans, effectively predicting drug responses and survival outcomes for cancer patients. Compared to conventional cancer biomarkers and existing prediction models based on mouse data, our method exhibits superior performance. These findings provide a valuable reference for further in-depth research on immunotherapy response prediction model based on mouse tumor profiles, with the potential for transfer applications in human cancer therapy.
Collapse
Affiliation(s)
- Dechen Xu
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Jie Li
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, 150001, China.
- National Key Laboratory of Smart Farm Technologies and Systems, Harbin, 150001, China.
| | - Li Zhou
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Jiahuan Jin
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| |
Collapse
|
16
|
Li BY, Li HL, Zeng FE, Luan XY, Liu BQ, Wang ZZ, Zhang L, Dong XZ. Identification of PD-L1-related biomarkers for selecting gastric adenocarcinoma patients for PD-1/PD-L1 inhibitor therapy. Discov Oncol 2025; 16:689. [PMID: 40338384 PMCID: PMC12061829 DOI: 10.1007/s12672-025-02515-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 04/25/2025] [Indexed: 05/09/2025] Open
Abstract
PD-1/PD-L1 inhibitors have been used to treat gastric cancer, and PD-L1 expression has been identified as a biomarker for predicting the effectiveness of immunotherapy in the treatment of gastric cancer. However, PD-L1 expression prediction for immunotherapy response is inaccurate, and improved response biomarkers are required. Thus, it is important to identify additional biomarkers that can predict the responses to PD-1/PD-L1 monoclonal antibodies in gastric cancer. In this study, GO and KEGG enrichment analysis of 142 DEGs co-expressed with PD-L1 were performed, and 41 genes were identified based on the intersection of the mRNA-significant GO term network and the mRNA-significant signalling pathway network. Further intersection analysis of the 41 candidate genes and 137 positive immunotherapy response genes indicated that BATF2 significantly affects the overall survival of GC patients. The transcription factor prediction for BATF2 identified additional potential predictors and therapeutic targets for GC. STAT and IRF family members were predicted to be transcription factors for BATF2. In addition, BATF2 knockdown significantly promoted GC cell growth, and PD-L1 expression was upregulated in si-BATF2-treated MKN-45 cells. Thus, BATF2 may serve as a biomarker for predicting the efficacy of PD-L1 blockade therapy in GC. BATF2 acts as a tumour suppressor gene during the development of GC. BATF2 is closely related to PD-L1 expression in GC, and high BATF2 expression positively correlates with low PD-L1 expression. BATF2 can be used as a potential biomarker and therapeutic target for responding to anti-PD-1 and anti-PD-L1 immunotherapies in GC.
Collapse
Affiliation(s)
- Bo-Ya Li
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing, China
| | - Hui-Ling Li
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing, China
| | - Fei-Er Zeng
- Department of Genetics and Genome Biology, Leicester Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, UK
| | - Xuan-Yu Luan
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bi-Qing Liu
- Department of Pharmacy, Children's Hospital Affiliated to Capital Institute of Paediatrics, Beijing, China
| | - Zhi-Zhou Wang
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing, China
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing, China.
| | - Xian-Zhe Dong
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing, China.
| |
Collapse
|
17
|
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
|
18
|
Shen W, Nguyen TH, Li MM, Huang Y, Moon I, Nair N, Marbach D, Zitnik M. Generalizable AI predicts immunotherapy outcomes across cancers and treatments. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.05.01.25326820. [PMID: 40385399 PMCID: PMC12083594 DOI: 10.1101/2025.05.01.25326820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Immune checkpoint inhibitors have become standard care across many cancers, but most patients do not respond. Predicting response remains challenging due to complex tumor-immune interactions and the poor generalizability of current biomarkers and models. Predictors such as tumor mutational burden, PD-L1 expression, and transcriptomic signatures often fail across cancer types, therapies, and clinical settings. There is a clear need for a robust, interpretable model that captures shared immune response principles and adapts to diverse clinical contexts. We present Compass, a foundation model for predicting immunotherapy response from pan-cancer transcriptomic data using a concept bottleneck architecture. Compass encodes tumor gene expression through 44 biologically grounded immune concepts representing immune cell states, tumor-microenvironment interactions, and signaling pathways. Trained on 10,184 tumors across 33 cancer types, Compass outperforms 22 baseline methods in 16 independent clinical cohorts spanning seven cancers and six immune checkpoint inhibitors, increasing precision by 8.5%, Matthews correlation coefficient by 12.3%, and area under the precision-recall curve by 15.7%, with minimal or no additional training. The model generalizes to unseen cancer types and treatments, supporting indication selection and patient stratification in early-phase clinical trials. Survival analysis shows that Compass-stratified responders have significantly longer overall survival (hazard ratio = 4.7, p < 0.0001). Personalized response maps link gene expression to immune concepts, revealing distinct mechanisms of response and resistance. For example, among immune-inflamed non-responders, Compass identifies distinct resistance programs involving TGF- β signaling, endothelial exclusion, CD4+ T cell dysfunction, and B cell deficiency. By combining mechanistic interpretability with transfer learning, Compass provides mechanistic insights into treatment response variability, supports clinical decision-making, and informs trial design.
Collapse
Affiliation(s)
- Wanxiang Shen
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Thinh H. Nguyen
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Michelle M. Li
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Yepeng Huang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Intae Moon
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Nitya Nair
- Roche Pharma Research and Early Development, Oncology Early Clinical Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Daniel Marbach
- Roche Pharma Research and Early Development, Data & Analytics, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Marinka Zitnik
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Data Science Initiative, Cambridge, MA, USA
| |
Collapse
|
19
|
Lee D, Ahn J, Choi J. PathNetDRP: a novel biomarker discovery framework using pathway and protein-protein interaction networks for immune checkpoint inhibitor response prediction. BMC Bioinformatics 2025; 26:119. [PMID: 40325361 PMCID: PMC12051301 DOI: 10.1186/s12859-025-06125-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/31/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Predicting immune checkpoint inhibitor (ICI) response remains a significant challenge in cancer immunotherapy. Many existing approaches rely on differential gene expression analysis or predefined immune signatures, which may fail to capture the complex regulatory mechanisms underlying immune response. Network-based models attempt to integrate biological interactions, but they often lack a quantitative framework to assess how individual genes contribute within pathways, limiting the specificity and interpretability of biomarkers. Given these limitations, we developed PathNetDRP, a framework that integrates biological pathways, protein-protein interaction networks, and machine learning to identify functionally relevant biomarkers for ICI response prediction. RESULTS We introduce PathNetDRP, a novel biomarker discovery approach that applies the PageRank algorithm to prioritize ICI-associated genes, maps them to relevant biological pathways, and calculates PathNetGene scores to quantify their contribution to immune response. Unlike conventional methods that focus solely on gene expression differences, PathNetDRP systematically incorporates biological context to improve biomarker selection. Validation across multiple independent cancer cohorts showed that PathNetDRP achieved strong predictive performance, with cross-validation the area under the receiver operating characteristic curves increasing from 0.780 to 0.940. Interestingly, PathNetDRP did not merely improve predictive accuracy; it also provided insights into key immune-related pathways, reinforcing its potential for identifying clinically relevant biomarkers. CONCLUSION The biomarkers identified by PathNetDRP demonstrated robust predictive performance across cross-validation and independent validation datasets, suggesting their potential utility in clinical applications. Furthermore, enrichment analysis highlighted key immune-related pathways, providing a deeper understanding of their role in ICI response regulation. While these findings underscore the promise of PathNetDRP, future work will explore the integration of additional predictive features, such as tumor mutational burden and microsatellite instability, to further refine its applicability.
Collapse
Affiliation(s)
- Dohee Lee
- Department of Computer Science and Engineering, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon, 22012, Republic of Korea
| | - Jaegyoon Ahn
- Department of Computer Science and Engineering, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon, 22012, Republic of Korea.
| | - Jonghwan Choi
- Division of Software, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea.
| |
Collapse
|
20
|
Zhang R, Zhang X, Lau HCH, Yu J. Gut microbiota in cancer initiation, development and therapy. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1283-1308. [PMID: 39821827 DOI: 10.1007/s11427-024-2831-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/12/2024] [Indexed: 01/19/2025]
Abstract
Cancer has long been associated with genetic and environmental factors, but recent studies reveal the important role of gut microbiota in its initiation and progression. Around 13% of cancers are linked to infectious agents, highlighting the need to identify the specific microorganisms involved. Gut microbiota can either promote or inhibit cancer growth by influencing oncogenic signaling pathways and altering immune responses. Dysbiosis can lead to cancer, while certain probiotics and their metabolites may help reestablish micro-ecological balance and improve anti-tumor immune responses. Research into targeted approaches that enhance therapy with probiotics is promising. However, the effects of probiotics in humans are complex and not yet fully understood. Additionally, methods to counteract harmful bacteria are still in development. Early clinical trials also indicate that modifying gut microbiota may help manage side effects of cancer treatments. Ongoing research is crucial to understand better how gut microbiota can be used to improve cancer prevention and treatment outcomes.
Collapse
Affiliation(s)
- Ruyi Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiang Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Harry Cheuk Hay Lau
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
21
|
Wang B, Gu B, Gao L, Ma C, Li X, Wang Y, Hu J, Wang N, Xiang L, Yu Y, Deng J, Wang X, He P, Zou D, Tao P, Ma Y, Song K, Han Z, Zhang T, Chen H. SERPINE1 Facilitates Metastasis in Gastric Cancer Through Anoikis Resistance and Tumor Microenvironment Remodeling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2500136. [PMID: 40207795 DOI: 10.1002/smll.202500136] [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: 01/04/2025] [Revised: 02/19/2025] [Indexed: 04/11/2025]
Abstract
SERPINE1 is a serine protease inhibitor upregulated in various malignancies and pivotal in gastric cancer (GC) metastasis and the tumor microenvironment (TME). This study elucidates the mechanisms by which SERPINE1 mediates anoikis resistance and fosters an immunosuppressive TME in advanced GC. SERPINE1 is highly expressed in GC tissues and metastatic lesions and serves as an independent risk factor for poor prognosis. The transcriptional activation of SERPINE1 by CEBPB triggers the PI3K/AKT and EMT signaling pathway via autocrine mechanisms, enhancing anoikis resistance and metastatic potential in GC cells. Furthermore, SERPINE1 facilitates M2 macrophage polarization by binding to lipoprotein receptor-related protein 1 (LRP1) in a paracrine manner, suppressing CD8+ T-cell infiltration and functionality in the TME. Therapeutic intervention combining SERPINE1 inhibition with PD-1 blockade exhibits synergistic antitumor effects. Clinically, high SERPINE1 expression is associated with an increased risk of recurrence following immune checkpoint inhibitor therapy in patients with advanced GC. These findings suggest that SERPINE1 is a critical driver of GC progression through anoikis resistance and TME remodeling. Hence, SERPINE1 can offer a promising therapeutic target and represent a predictive biomarker for immunotherapy outcomes in GC.
Collapse
Affiliation(s)
- Bofang Wang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Baohong Gu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Surgical Oncology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Lei Gao
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Chenhui Ma
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Xuemei Li
- Gansu Provincial Key Laboratory of Environmental Oncology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Yunpeng Wang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Jike Hu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Na Wang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Lin Xiang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Pathology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Yang Yu
- Department of Thyroid Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junge Deng
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Surgical Oncology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Xueyan Wang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Puyi He
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Dan Zou
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Pengxian Tao
- Cadre Ward of General Surgery Department, Gansu Provincial Hospital, Lanzhou, China
| | - Yanling Ma
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Surgical Oncology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Kewei Song
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Zhijian Han
- Gansu Provincial Key Laboratory of Environmental Oncology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Tao Zhang
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Hao Chen
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Surgical Oncology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Key Laboratory of Environmental Oncology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- The Key Laboratory of Humanized Animal Models, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| |
Collapse
|
22
|
Peter SA, Cristescu R, Peña C, Watkins A, Espenschied CR, Zhang N, Liao J. Assessment of Circulating Tumor DNA Burden in Patients With Metastatic Gastric Cancer Using Real-World Data. JCO Precis Oncol 2025; 9:e2400582. [PMID: 40324115 DOI: 10.1200/po-24-00582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/19/2024] [Accepted: 02/21/2025] [Indexed: 05/07/2025] Open
Abstract
PURPOSE Circulating tumor DNA (ctDNA) has emerged as a promising biomarker with prognostic and potentially predictive value for several tumor types, including gastric cancer (GC). This study uses real-world data (RWD) to investigate the association of pretreatment ctDNA burden with clinical outcomes among patients with first-line (1L)-treated metastatic gastric cancer (mGC) in the United States. METHODS Patients were identified from the GuardantINFORM real-world clinical-genomic database. Adult patients with mGC who underwent testing with the Guardant360 assay from June 2014 to March 2022 and within 60 days before 1L treatment were included. The median of the maximum variant allele fraction (MVAF) was used to classify patients into high or low ctDNA burden groups, with undetectable ctDNA burden included in the low group. Associations with real-world outcome variables derived from claims, including time to treatment discontinuation (rwTTD), time to next treatment (rwTTNT), and overall survival (rwOS), were assessed using log-rank tests and multivariable Cox models. RESULTS A cohort of 824 patients with mGC was identified. Median MVAF was 2.9%, with 91% having detectable ctDNA. Among patients receiving chemo-based treatment (n = 537), rwTTDs were similar in low and high ctDNA burden groups, while those with high ctDNA burden showed significantly shorter rwTTNT and rwOS (median rwTTNT = 4.8 months v 7.4 months, P < .001; median rwOS = 13.2 months v 19.1 months, P < .001). Multivariable Cox analyses showed similar results. ctDNA burden in immunotherapy (n = 100) and trastuzumab-based (n = 99) treatment groups did not have significant associations with outcomes. CONCLUSION We used RWD to demonstrate that high pretreatment ctDNA burden was associated with worse clinical outcomes in a mGC population receiving 1L chemotherapy-based treatments. Our analysis suggests ctDNA burden could be used as a prognostic biomarker for mGC.
Collapse
|
23
|
Wonglhow J, Tantipisit J, Wetwittayakhlang P, Sunpaweravong P, Sathitruangsak C, Kanjanapradit K, Thongwatchara P, Dechaphunkul A. Association Between Epstein-Barr Virus Infection and PD-L1 Expression in Gastric Cancer: Prevalence, Clinicopathological Features, and Prognostic Implications. Cancers (Basel) 2025; 17:1492. [PMID: 40361419 PMCID: PMC12070931 DOI: 10.3390/cancers17091492] [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: 03/07/2025] [Revised: 04/12/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Epstein-Barr virus-associated gastric cancer (EBVaGC) represents a distinct molecular subgroup with potential responsiveness to immunotherapy approved for programmed death-ligand 1 (PD-L1)-positive gastric cancer. This retrospective study aimed to assess the prevalence and association between EBVaGC and PD-L1 positivity among patients with gastric adenocarcinoma treated at a university hospital in Southern Thailand from January 2017 to October 2023. METHODS The EBV status of the patients and PD-L1 expression were determined using in situ hybridization and immunohistochemistry, respectively. RESULTS The prevalence of EBVaGC was 4.5% among 132 patients, whereas 9.1% of patients exhibited a PD-L1 combined positive score (CPS) of ≥1, with no significant association observed between them. EBVaGC was more prevalent in males, non-antral tumors, diffuse/mixed histologic subtypes, and poorly differentiated tumors. Median overall survival for patients with EBVaGC and PD-L1 CPS ≥ 1 was 9.48 and 14.19 months, respectively, compared with 10.32 and 9.79 months for those with non-EBVaGC (hazard ratio: 1.24; 95% CI: 0.50-3.04; p = 0.645) and PD-L1 CPS < 1 (hazard ratio: 0.82; 95% CI: 0.40-1.69; p = 0.590), respectively. CONCLUSIONS Our findings revealed a low prevalence of EBVaGC and PD-L1 positivity in Thailand, with no significant association or survival impact observed. These findings highlight the regional variation in these biomarkers and support EBV as an independent biomarker from PD-L1. However, further research, particularly studies evaluating immunotherapy outcomes, is warranted to clarify the predictive and clinical significance of EBV in gastric cancer.
Collapse
Affiliation(s)
- Jirapat Wonglhow
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (J.W.); (P.S.); (C.S.); (P.T.)
| | - Jarukit Tantipisit
- Department of Pathology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (J.T.); (K.K.)
| | - Panu Wetwittayakhlang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand;
| | - Patrapim Sunpaweravong
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (J.W.); (P.S.); (C.S.); (P.T.)
| | - Chirawadee Sathitruangsak
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (J.W.); (P.S.); (C.S.); (P.T.)
| | - Kanet Kanjanapradit
- Department of Pathology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (J.T.); (K.K.)
| | - Phatcharaporn Thongwatchara
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (J.W.); (P.S.); (C.S.); (P.T.)
| | - Arunee Dechaphunkul
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (J.W.); (P.S.); (C.S.); (P.T.)
| |
Collapse
|
24
|
Kim HD, Kim SY, Lee H, Lee Y, Hyung J, Moon M, Shin J, Park YS, Ryu MH. Predictive value of EBV-positivity in patients with gastric cancer treated with first-line nivolumab plus chemotherapy. Gastric Cancer 2025:10.1007/s10120-025-01618-6. [PMID: 40295365 DOI: 10.1007/s10120-025-01618-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 04/11/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Epstein-Barr virus (EBV) positivity is a potential predictive biomarker for immune checkpoint inhibitors (ICIs) in gastric cancer patients, but its value in first-line ICI-based chemotherapy remains unclear. This study aimed to evaluate the predictive value of EBV positivity in patients treated with first-line nivolumab plus chemotherapy. METHODS This single-center study included advanced gastric cancer patients treated with first-line nivolumab plus chemotherapy (n = 293). Patients with EBV positivity treated with chemotherapy alone (n = 12) served as the control group. EBV positivity was confirmed by in situ hybridization. RESULTS Among patients treated with nivolumab plus chemotherapy, 18 (6.1%) had EBV-positive tumors, and these were associated with high PD-L1 combined positive score (CPS) expression levels. Progression-free survival (PFS) and overall survival (OS) tended to be more favorable in those with EBV-positive tumors. Multivariate analysis of patients treated with nivolumab-chemotherapy revealed that EBV positivity, combined with PD-L1 CPS ≥ 5, was an independent factor for PFS. In patients with EBV-positive tumors, nivolumab-chemotherapy was associated with significantly favorable PFS and OS compared to chemotherapy alone. Similar results were observed in the subgroup with PD-L1 CPS ≥ 5. However, survival outcomes did not differ between patients treated with nivolumab plus chemotherapy versus chemotherapy alone in the subgroup with PD-L1 CPS < 5. CONCLUSION EBV positivity predicts favorable survival outcomes in patients with gastric cancer treated with nivolumab plus chemotherapy. The benefit of nivolumab plus chemotherapy over chemotherapy alone for patients with EBV positivity appears to be associated with high PD-L1 expression levels.
Collapse
Affiliation(s)
- Hyung-Don Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - So-Yeon Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Hyungeun Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Yuna Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Jaewon Hyung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Meesun Moon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Jinho Shin
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young Soo Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Min-Hee Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
25
|
Wang M, Guo Y, Xu Y, Yu Y, Lin J, Lin Y, Ge L, Zhang Y, Chi L, Xue F, Wang Q. Unraveling the Role of Programmed Cell Death Gene Signature and THBS1 in Gastric Cancer Progression and Therapy Response. J Gastroenterol Hepatol 2025. [PMID: 40294913 DOI: 10.1111/jgh.16987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/12/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Programmed cell death (PCD) genes play crucial roles in cancer progression and response to therapies, yet their impact on gastric cancer remains inadequately elucidated. This study aimed to create a prognostic cell death signature (PCDs) for gastric cancer, providing insights into potential therapeutic targets and survival predictors. METHODS We utilized TCGA-STAD and five GEO datasets, representing thousands of gastric cancer samples, for a comprehensive analysis of PCD genes. Differential gene expression, functional enrichment, survival, and machine learning analyses were conducted to construct a PCD-based prognostic model. RESULTS A total of 249 differentially expressed PCD genes were identified between cancerous and noncancerous gastric tissues. Subsequently, a PCD signature based on seven genes was developed and cross-validated across multiple cohorts. The high-PCD subtype correlated with poorer survival outcomes, lower tumor mutational burden, higher infiltration of M2 macrophages, lower levels of immune checkpoint expression, and decreased response to immunotherapy. A nomogram incorporating the PCDs provided accurate survival rate predictions. Additionally, nine machine learning algorithms were implemented for recurrence prediction, with the random forest model displaying high effectiveness. In this model, thrombospondin 1 (THBS1) showed the highest weight, and its knockdown significantly reduced gastric cancer cell proliferation and invasion. CONCLUSION This study underscores the significance of PCD genes, particularly THBS1, in gastric cancer progression and highlights their value as potential therapeutic targets. The predictive models developed here can aid in assessing patient prognosis and tailoring personalized treatment strategies.
Collapse
Affiliation(s)
- Min Wang
- Jiangsu Province Engineering Research Center of Traditional Chinese Medicine Health Preservation, Nanjing, Jiangsu Province, China
| | - YinChao Guo
- Jiangsu Province Engineering Research Center of Traditional Chinese Medicine Health Preservation, Nanjing, Jiangsu Province, China
| | - YiNing Xu
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Yan Yu
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Jia Lin
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - Yao Lin
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| | - LiLin Ge
- Jiangsu Province Engineering Research Center of Traditional Chinese Medicine Health Preservation, Nanjing, Jiangsu Province, China
| | - Yitong Zhang
- University of Newcastle, Callaghan, New South Wales, Australia
| | - LiangJie Chi
- Department of Gastrointestinal Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian Province, China
| | - FangQin Xue
- Department of Gastrointestinal Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian Province, China
| | - QingShui Wang
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, China
| |
Collapse
|
26
|
Kan L, Yu Y, Wang Y, Shi L, Fan T, Chen H, Ren C. The application of organoids in investigating immune evasion in the microenvironment of gastric cancer and screening novel drug candidates. Mol Cancer 2025; 24:125. [PMID: 40287758 PMCID: PMC12032790 DOI: 10.1186/s12943-025-02328-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Gastric cancer (GC) is a prevalent digestive system tumor, the fifth most diagnosed cancer worldwide, and a leading cause of cancer deaths. GC is distinguished by its pronounced heterogeneity and a dynamically evolving tumor microenvironment (TME). The lack of accurate disease models complicates the understanding of its mechanisms and impedes the discovery of novel drugs. A growing body of evidence suggests that GC organoids, developed using organoid culture technology, preserve the genetic, phenotypic, and behavioral characteristics. GC organoids hold significant potential for predicting treatment responses in individual patients. This review provides a comprehensive overview of the current clinical treatment strategies for GC, as well as the history, construction and clinical applications of organoids. The focus is on the role of organoids in simulating the TME to explore mechanisms of immune evasion and intratumoral microbiota in GC, as well as their applications in guiding clinical drug therapy and facilitating novel drug screening. Furthermore, we summarize the limitations of GC organoid models and underscore the need for continued technological advancements to benefit both basic and translational oncological research.
Collapse
Affiliation(s)
- Liuyue Kan
- Department of Laboratory Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Ying Yu
- Department of Laboratory Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Yaxue Wang
- Department of Laboratory Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Lei Shi
- Department of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, No. 98 Western Nantong Road, Yangzhou, 225001, China
| | - Tingyuan Fan
- Department of Laboratory Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Hui Chen
- Department of Geriatrics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, No. 98 Western Nantong Road, Yangzhou, 225001, China.
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, No. 98, Western Nantong Road, Yangzhou, 225001, China.
| | - Chuanli Ren
- Department of Laboratory Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China.
- Department of Laboratory Medicine, The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, China.
- The Yangzhou Clinical Medical College of Xuzhou Medical University, No. 98, Western Nantong Road, Yangzhou, 225001, China.
| |
Collapse
|
27
|
Yamazawa S, Fukasawa-Hokazono M, Takase A, Kondo A, Matsubara J, Shinozaki-Ushiku A, Seto Y, Ushiku T. Immune evasion strategies in AFP-producing gastric carcinoma: characterized by HLA-G expression and HLA class I deficiency. Virchows Arch 2025:10.1007/s00428-025-04108-3. [PMID: 40278871 DOI: 10.1007/s00428-025-04108-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/28/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025]
Abstract
Alpha-fetoprotein-producing gastric carcinoma (AFPGC) is an aggressive subtype of gastric cancer characterized by a primitive cellular phenotype and poor prognosis. The tumor immunology of AFPGC remains largely unexplored. Given its embryonic-like properties, AFPGC is hypothesized to employ distinct immune evasion strategies, with the oncofetal protein human leukocyte antigen (HLA)-G-a key mediator of maternal-fetal immune tolerance-likely playing a pivotal role. To test this, we assessed the expression of HLA-G, along with other key immune evasion markers, including HLA class I (HLA-I) deficiency and PD-L1 expression, in 39 cases of AFPGC, and compared them with those of 44 Epstein-Barr virus (EBV)-positive, 57 microsatellite instability (MSI), 54 intestinal-type, and 45 diffuse-type gastric carcinomas. HLA-G expression was significantly higher in AFPGCs (71%) than in other subtypes (7-28%; P < 0.001). HLA-I deficiency (≥ 1% of tumor cells) was most prevalent in AFPGC (69%), followed by MSI tumors (56%), with lower rates in other subtypes (22-29%). PD-L1 positivity (combined positive score ≥ 5) was observed in 41% of AFPGCs, lower than in EBV-positive (77%) and MSI tumors (44%), but higher than in intestinal-type (13%) and diffuse-type (9%) carcinomas. Furthermore, CD8-positive T-cell infiltration was found to be lowest in AFPGC compared to the other subtypes. These findings suggest that AFPGC employs multiple immune evasion mechanisms, notably through increased HLA-G expression and HLA-I deficiency, likely linked to its primitive cellular phenotype and reactivation of immunogenic oncofetal antigens. Such immune evasion features may underlie the aggressiveness of AFPGC and present promising targets for immunotherapeutic interventions.
Collapse
Affiliation(s)
- Sho Yamazawa
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Akiko Takase
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Atsushi Kondo
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Joji Matsubara
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Aya Shinozaki-Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Division of Integrative Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
28
|
Tang S, Che X, Wang J, Li C, He X, Hou K, Zhang X, Guo J, Yang B, Li D, Cao L, Qu X, Wang Z, Liu Y. T-bet +CD8 + T cells govern anti-PD-1 responses in microsatellite-stable gastric cancers. Nat Commun 2025; 16:3905. [PMID: 40280928 PMCID: PMC12032036 DOI: 10.1038/s41467-025-58958-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
More than 90% of advanced gastric cancers (GC) are microsatellite-stable (MSS). Compared to the high response rate of immune checkpoint inhibitors (ICI) in microsatellite-instability-high (MSI-H) GCs, only 10% of unstratified MSS GCs respond to ICIs. In this study, we apply semi-supervised learning to stratify potential ICI responders in MSS GCs, achieving high accuracy, quantified by an area under the curve of 0.924. Spatial analysis of the tumor microenvironment of ICI-sensitive GCs reveals a high level of T-bet+ CD8 + T cell infiltration in their tumor compartments. T-bet+ CD8 + T cells exhibit superior anti-tumor activity due to their increased ability to infiltrate tumors and secrete cytotoxic molecules. Adoptive transfer of T-bet+ CD8 + T cells boosts anti-tumor immunity and confers susceptibility to ICIs in immune-ignorant MSS GCs in a humanized mouse model. Spatial RNA sequencing suggests a positive-feedback loop between T-bet+ T cells and PD-L1+ tumor cells, which eventually drives T cell exhaustion and can therefore be leveraged for ICI therapy. In summary, our research provides insights into the underlying mechanism of anti-tumor immunity and deepens our understanding of varied ICI responses in MSS GCs.
Collapse
Affiliation(s)
- Shiying Tang
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Xiaofang Che
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Jinyan Wang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, No. 77, Puhe Road, Shenyang, Liaoning, China
| | - Ce Li
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Xin He
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Kezuo Hou
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Xiaojie Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Jia Guo
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Bowen Yang
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Danni Li
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Lili Cao
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China.
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China.
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China.
| | - Zhenning Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, No.155, Nanjing Street, Shenyang, Liaoning, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, China Medical University, Shenyang, Liaoning, China.
- Institute of Health Sciences, China Medical University, Shenyang, Liaoning, China.
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, No. 155, Nanjing Street, Shenyang, Liaoning, China.
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China.
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumours, Ministry of Education, Shenyang, Liaoning, China.
| |
Collapse
|
29
|
Luo SQ, Dai L, Zhou YJ, He T, Wang FJ, Jin XR, Wang Q. Multiple primary tumors patient developed microsatellite stable gastric cancer after cadonilimab treatment for liver cancer: A case report. World J Clin Oncol 2025; 16:102418. [PMID: 40290697 PMCID: PMC12019263 DOI: 10.5306/wjco.v16.i4.102418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/22/2025] [Accepted: 02/27/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Multiple primary malignant tumors refer to the occurrence of two or more primary malignant tumors in the same organ or multiple organs or tissues at the same time or successively in the same patient, and can occur anywhere in the body. The treatment guidelines for patients with multiple primary malignant tumors are currently controversial. CASE SUMMARY A 51-year-old male patient with liver cancer and portal hypertension received 42 months of co-treatment with atezolizumab and bevacizumab. After that, the disease was rated stable disease. The patient was then diagnosed with gastric cancer. Since the patient was not sensitive to anti-programmed death ligand 1 immunosuppressive agents, a co-treatment with oxaliplatin, tegafur, apatinib, and cadonilimab was selected after multidisciplinary consultation and the patient's agreement. After four cycles of treatment, partial response and stable disease were observed in gastric and liver cancers, respectively. Surgical treatment was performed considering the high-risk factors of gastrointestinal bleeding in patients with gastroesophageal varices. Postoperative pathology showed that the Tumor Regression Grade was 1. Moreover, the genetic testing of postoperative tumor specimens indicated negative programmed death ligand 1 and microsatellite stability. In addition, the latest follow-up indicated an 8 and 40-month progression-free survival in gastric and liver cancer patients, respectively. Currently, the patient is receiving postoperative immunotherapy with cadonilimab. CONCLUSION Cadonilimab not only treats microsatellite stability gastric cancer patients but can also be used for liver cancer treatment.
Collapse
Affiliation(s)
- Si-Qi Luo
- Department of Clinical Medicine, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Li Dai
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Yong-Jin Zhou
- Department of Clinical Medicine, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Tong He
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Fang-Jie Wang
- Department of Emergency, The People’s Hospital of Xishui, Zunyi 564600, Guizhou Province, China
| | - Xiang-Ren Jin
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Qian Wang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| |
Collapse
|
30
|
Tougeron D, Louvet C, Desramé J, Evesque L, Angelergues A, Carnot A, Breysacher G, Zaanan A, Etchepare N, Mabro M, Kaluzinski L, Petorin C, Chibaudel B, Aparicio T, Bodere A, Rinaldi Y, Le Malicot K, Emile JF, Lepage C, Baures A, Djamai H, Taly V, Laurent-Puig P. Circulating tumor DNA strongly predicts efficacy of chemotherapy plus immune checkpoint inhibitors in patients with advanced gastro-esophageal adenocarcinoma. COMMUNICATIONS MEDICINE 2025; 5:136. [PMID: 40275077 PMCID: PMC12022060 DOI: 10.1038/s43856-025-00867-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Efficacy of 2nd line treatment in advanced gastric or gastro-esophageal junction (GEJ) adenocarcinoma remains limited with no identified strong predictor of treatment efficacy. We evaluated the prognostic value of circulating tumor DNA (ctDNA) in predicting the efficacy of immune checkpoint inhibitors (ICI) plus chemotherapy in the randomized PRODIGE 59-FFCD 1707-DURIGAST trial. METHODS ctDNA was evaluated before treatment (baseline) and at 4 weeks (before the third cycle of treatment, C3) using droplet-digital PCR assays based on the detection of CpG methylation. RESULTS Progression-free survival (PFS) and overall survival (OS) were shorter in patients with a high (>1.1 ng/mL) versus low (<1.1 ng/mL) ctDNA concentration at baseline (2.3 vs. 5.8 months; HR = 2.19; 95% CI, 1.09-4.41; p = 0.03 and 4.5 vs. 12.9 months; HR = 2.73; 95% CI, 1.29-5.75; p < 0.01), respectively, after adjustment for identified prognostic variables. Patients with a ctDNA decrease ≤75% between baseline and C3 versus a ctDNA decrease >75% had a worse objective response rate (p = 0.007), shorter PFS (2.2 vs. 7.4 months, HR = 1.90; 95% CI, 1.03-3.51; p = 0.04) and OS (6.6 vs 16.0 months; HR = 2.18; 95% CI, 1.09-4.37; p = 0.03). CONCLUSIONS An early decrease in ctDNA concentration is a strong predictor of the therapeutic efficacy of ICI plus chemotherapy in advanced gastric/GEJ adenocarcinoma. Clinical Trial Information NCT03959293 (DURIGAST).
Collapse
Affiliation(s)
- David Tougeron
- Department of Gastroenterology and Hepatology, Poitiers University Hospital, Poitiers, France.
| | - Christophe Louvet
- Department of Medical Oncology, Institute Mutualiste Montsouris, Paris, France
| | - Jérôme Desramé
- Department of Gastroenterology, Mermoz Hospital, Lyon, France
| | - Ludovic Evesque
- Department of Medical Oncology, Centre Antoine Lacassagne, Nice, France
| | | | - Aurélien Carnot
- Department of Gastroenterology and Digestive Oncology, Oscar Lambret Centre, Lille, France
| | - Gilles Breysacher
- Department of Gastroenterology and Hepatology, Colmar Hospital, Colmar, France
| | - Aziz Zaanan
- Department of Digestive Oncology, Georges Pompidou European Hospital, AP-HP, Université Paris Cité, Paris Cancer Institute CARPEM, Paris, France
| | | | - May Mabro
- Department of Oncology, Foch Hospital, Suresnes, France
| | - Laure Kaluzinski
- Department of Oncology, Cherbourg-en-Cotentin Hospital, Cherbourg-en-Cotentin, France
| | - Caroline Petorin
- Department of Oncology, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | - Benoist Chibaudel
- Department of Oncology, Franco-Britannique Hospital - Fondation Cognacq-Jay, Levallois, France
| | - Thomas Aparicio
- Department of Gastroenterology and Digestive Oncology, Saint Louis Hospital, Paris, France
| | | | - Yves Rinaldi
- Department of Gastroenterology, Marseille European Hospital, Marseille, France
| | - Karine Le Malicot
- Fédération Francophone de Cancérologie Digestive, EPICAD INSERM LNC-UMR 1231, Bourgogne Franche-Comté University, Dijon, France
| | - Jean-François Emile
- Pathology Department, Paris-Saclay University, Versailles SQY University, EA4340-BECCOH, Assistance Publique-Hôpitaux de Paris (APHP), Ambroise-Paré Hospital, Boulogne, France
| | - Côme Lepage
- Fédération Francophone de Cancérologie Digestive, EPICAD INSERM LNC-UMR 1231, Bourgogne Franche-Comté University, Dijon, France
| | - Aurélia Baures
- Centre de recherche des cordeliers, Université Paris Cité, Sorbonne Université, UMR-S1138, CNRS SNC5096, Équipe Labélisée Ligue Nationale Contre le Cancer, Paris, France
| | - Hanane Djamai
- Centre de recherche des cordeliers, Université Paris Cité, Sorbonne Université, UMR-S1138, CNRS SNC5096, Équipe Labélisée Ligue Nationale Contre le Cancer, Paris, France
| | - Valérie Taly
- Centre de recherche des cordeliers, Université Paris Cité, Sorbonne Université, UMR-S1138, CNRS SNC5096, Équipe Labélisée Ligue Nationale Contre le Cancer, Paris, France
- METHYS Dx, Paris, France
| | - Pierre Laurent-Puig
- Centre de recherche des cordeliers, Université Paris Cité, Sorbonne Université, UMR-S1138, CNRS SNC5096, Équipe Labélisée Ligue Nationale Contre le Cancer, Paris, France
- Department of Genomic Medicine of Tumors and Cancers APHP, Institut Cancer Paris Carpem, APHP, Paris, France
| |
Collapse
|
31
|
Hu C, Liu H, Hong B, Wang L, Wu Z, Xie W, Luo B, Cao D, Zhong Y, Liu Y, Gong W. Helicobacter pylori reversing the landscape of neoadjuvant immunotherapy for microsatellite stable gastric cancer: a multicenter cohort study. BMC Med 2025; 23:230. [PMID: 40264112 PMCID: PMC12016324 DOI: 10.1186/s12916-025-04047-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 04/01/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Microsatellite stable (MSS) gastric cancer (GC) is largely unresponsive to immunotherapy, presenting a persistent and formidable challenge in the field. Patients with advanced GC and Helicobacter pylori (H. pylori) infection have shown benefits from immunotherapy. However, it remains unreported whether neoadjuvant immunotherapy is beneficial for H. pylori-positive MSS GC patients. METHODS This retrospective cohort study analyzed data from GC patients treated at three medical centers in China between January 1, 2014, and July 1, 2024. Patients with gastric adenocarcinoma or adenocarcinoma of the gastroesophageal junction underwent testing for H. pylori infection prior to receiving neoadjuvant therapy. RESULTS In this retrospective analysis, those positive for H. pylori had a higher objective response rate of 63.77% (95% CI, 51.98-74.11%) compared to 47.73% (95% CI, 39.39-56.19%) in H. pylori-negative patients. Pathological complete remission was higher in H. pylori-positive patients at 17.39% (95% CI, 10.24-27.98%) versus 15.91% (95% CI, 10.65-23.10%). Logistic regression analysis revealed a strong correlation between H. pylori positivity and increased objective remission rate (P = 0.031, OR = 1.928, 95% CI 1.06-3.51). In H. pylori-positive MSS GC patients receiving neoadjuvant immunotherapy pCR rates can reach 27.27% (95% CI, 15.07-44.21%), much higher than the 8.33% (95% CI, 2.87-21.82%) in neoadjuvant chemotherapy patients. Survival analysis showed a 3-year OS rate of 74.2% (95% CI, 56.75-86.30%) in the H. pylori-positive group and 64.3% (95% CI, 51.20-75.55%) in the H. pylori-negative group, and the hazard ratio (HR) of these two groups was 0.50 (95% CI, 0.28-0.87; P <.001). Multivariable analysis for OS further showed the survival benefit of H. pylori, with HRs of 0.51 (95% CI, 0.29-0.91; P = 0.02). CONCLUSIONS H. pylori infection has emerged as a favorable factor for neoadjuvant immunotherapy in MSS GC, underscoring the importance of considering H. pylori status in preoperative treatment strategies.
Collapse
Affiliation(s)
- Chengyu Hu
- Department of Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Hongming Liu
- Department of Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Bo Hong
- Department of Pathology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Li Wang
- Department of Emergency Medicine, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Zelai Wu
- Department of Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Weixun Xie
- Department of Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Bixian Luo
- Department of Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Dong Cao
- Department of Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Department of Gastrointestinal Surgery, Affiliated Hospital of Shaoxing University, Shaoxing City, China
| | - Yuxin Zhong
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yong Liu
- Department of Gastric Surgery, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China.
| | - Weihua Gong
- Department of Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
32
|
Zhang Y, Peng W, Yang W, Zhang W, Fan Y. Efficacy and safety of programmed cell death protein-1 inhibitor for first-line therapy of advanced gastric or gastroesophageal junction cancer: a network meta-analysis. Front Immunol 2025; 16:1500954. [PMID: 40264761 PMCID: PMC12011870 DOI: 10.3389/fimmu.2025.1500954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/12/2025] [Indexed: 04/24/2025] Open
Abstract
Background This study conducted a network meta-analysis to evaluate and rank the safety and efficacy of programmed cell death protein-1 (PD-1) inhibitors for patients with advanced gastric or gastroesophageal junction cancer (GC/GEJC). Methods A systematic search was conducted in PubMed, Embase, and Cochrane Library databases to compare the efficacy and safety of different treatment regimens, including overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and treatment-related adverse events (TRAEs) in patients with advanced GC/GEJC. Results A total of six RCT studies were ultimately included in the analysis, involving 6,294 patients. Among them, 256 patients received PD-1 inhibitor monotherapy (pembrolizumab), 3,029 patients received a PD-1 inhibitor plus chemotherapy (1,047 with pembrolizumab, 1,154 with nivolumab, 327 with sintilimab, and 501 with tislelizumab), and 3,009 received either chemotherapy or chemotherapy plus placebo. Sintilimab plus chemotherapy had the highest SUCRA value for OS (85.2%), while nivolumab plus chemotherapy had the highest SUCRA values for both PFS and ORR (96.8% and 82.9%). Four PD-1 inhibitors plus chemotherapy significantly improved median OS and ORR compared with chemotherapy. Sintilimab plus chemotherapy, pembrolizumab plus chemotherapy, and nivolumab plus chemotherapy significantly improved median PFS compared with chemotherapy. For TRAEs of grade 3 or worse, pembrolizumab monotherapy had the highest SUCRA value. Tislelizumab plus chemotherapy, as well as sintilimab plus chemotherapy, did not increase the overall incidence of TRAEs and the incidence of grade 3 or worse TRAEs. Conclusions In the first-line treatment of advanced GC/GEJC, PD-1 inhibitors plus chemotherapy have been demonstrated to significantly improve OS, PFS, and ORR compared with chemotherapy. Among them, sintilimab plus chemotherapy achieved the highest SUCRA value for OS, and nivolumab plus chemotherapy achieved the highest SUCRA values for PFS and ORR. Regarding safety, tislelizumab plus chemotherapy and sintilimab plus chemotherapy did not increase the overall incidence of TRAEs and the incidence of grade 3 or worse TRAEs, with good tolerability and safety.
Collapse
Affiliation(s)
- Yunnan Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Wenxing Peng
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Wei Yang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Wenzhou Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Yannan Fan
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
33
|
Wang X, Zhang Y, Fan G, Wu H, Qi X, Cui X, Zhou C. Case Report: A case of synchronous multiple early gastric cancer with a microsatellite instability-high phenotype. Front Oncol 2025; 15:1527495. [PMID: 40248200 PMCID: PMC12003148 DOI: 10.3389/fonc.2025.1527495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/04/2025] [Indexed: 04/19/2025] Open
Abstract
Synchronous multiple early gastric cancer (SMEGC) is a relatively uncommon variant of early gastric cancer (EGC). In this report, we present a case of SMEGC accompanied by a microsatellite instability-high (MSI-H) phenotype. The patient was a 69-year-old man who presented to our hospital with abdominal pain. The endoscopic examination revealed two lesions. Both lesions were pathologically confirmed as EGC, then the patient subsequently underwent endoscopic submucosal dissection (ESD). Nine months post-procedure, the patient returned with recurrent abdominal pain, leading to the diagnosis of a new EGC. Immunohistochemical analysis demonstrated that all lesions exhibited an MSI-H phenotype and BRAF mutant expression, suggesting that these lesions are not associated with Lynch syndrome-related EGC. The case was ultimately diagnosed as SMEGC with an MSI-H phenotype. The current evidence and clinical experience suggest that patients with advanced MSI-H are likely to benefit from immunotherapy and should be considered for early systemic treatment with immunotherapy as a central component. At present, research studies on the molecular characteristics of SMEGC are limited, underscoring the importance of conducting comprehensive molecular diagnostics of each EGC patient, which could help clinicians thoroughly understand the lesion's characteristics.
Collapse
Affiliation(s)
- Xinshuo Wang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yifan Zhang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Guangyan Fan
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Honglei Wu
- Department of Gastroenterology, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xing Qi
- Department of Gastroenterology, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiujie Cui
- Department of Pathology, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Chengjun Zhou
- Department of Pathology, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
34
|
Abate M, Stroobant E, Fei T, Lin YH, Shimada S, Drebin H, Chen E, Tang LH, Shah SP, Wolchok JD, Janjigian YY, Strong VE, Vardhana SA. Host Tissue Factors Predict Immune Surveillance and Therapeutic Outcomes in Gastric Cancer. Cancer Immunol Res 2025; 13:591-601. [PMID: 39786344 PMCID: PMC11964842 DOI: 10.1158/2326-6066.cir-23-0563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/25/2024] [Accepted: 01/07/2025] [Indexed: 01/12/2025]
Abstract
The immune composition of solid tumors is typically inferred from biomarkers, such as histologic and molecular classifications, somatic mutational burden, and PD-L1 expression. However, the extent to which these biomarkers predict the immune landscape in gastric adenocarcinoma-an aggressive cancer often linked to chronic inflammation-remains poorly understood. We leveraged high-dimensional spectral cytometry to generate a comprehensive single-cell immune landscape of tumors, normal tissue, and lymph nodes from patients in the Western Hemisphere with gastric adenocarcinoma. The immune composition of gastric tumors could not be predicted by traditional metrics such as tumor histology, molecular classification, mutational burden, or PD-L1 expression via IHC. Instead, our findings revealed that innate immune surveillance within tumors could be anticipated by the immune profile of the normal gastric mucosa. Additionally, distinct T-cell states in the lymph nodes were linked to the accumulation of activated and memory-like CD8+ tumor-infiltrating lymphocytes. Unbiased reclassification of patients based on tumor-specific immune infiltrate generated four distinct subtypes with varying immune compositions. Tumors with a T cell-dominant immune subtype, which spanned The Cancer Genome Atlas molecular subtypes, were exclusively associated with superior responses to immunotherapy. Parallel analysis of metastatic gastric cancer patients treated with immune checkpoint blockade showed that patients who responded to immunotherapy had a pretreatment tumor composition that corresponded to a T cell-dominant immune subtype from our analysis. Taken together, this work identifies key host-specific factors associated with intratumoral immune composition in gastric cancer and offers an immunological classification system that can effectively identify patients likely to benefit from immune-based therapies.
Collapse
Affiliation(s)
- Miseker Abate
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Surgery, New York-Presbyterian Hospital/Weill Cornell Medicine, New York, NY, USA
| | - Emily Stroobant
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Teng Fei
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ya-Hui Lin
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shoji Shimada
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Digestive Disease Center, Showa University, Northern Yokohama Hospital, Yokohama, Japan
| | - Harrison Drebin
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Eunise Chen
- Department of Surgery, University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, USA
| | - Laura H. Tang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sohrab P. Shah
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jedd D. Wolchok
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Yelena Y. Janjigian
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vivian E. Strong
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Santosha A. Vardhana
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
35
|
Tao Y, Tian C, Qi S, Jia Z, Xu Z, Meng J, Xu G, Hu H, Wang X, Zhang T, You H, Lan X, Lin X, Yu G, Zhou H, Liu J, Zheng H. Targeting both death and paracaspase domains of MALT1 with antisense oligonucleotides overcomes resistance to immune-checkpoint inhibitors. NATURE CANCER 2025; 6:702-717. [PMID: 40075237 DOI: 10.1038/s43018-025-00930-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 02/13/2025] [Indexed: 03/14/2025]
Abstract
Targeting MALT1's paracaspase activity has been explored for B cell lymphoma and solid tumors. While the role of MALT1 in promoting cancer cell proliferation has been investigated, its involvement in immune evasion is unclear. Here we report that MALT1 promotes immune evasion through its paracaspase and death domain. In a paracaspase-dependent manner, MALT1 protects CD274 mRNA from degradation by its cleavage of ROQUIN1 and ROQUIN2. In a death-domain-dependent manner, MALT1 promotes the proliferation and polarization of tumor-associated macrophages to generate an immunosuppressive tumor microenvironment. Targeting MALT1 with antisense oligonucleotides inhibits PD-L1 expression in patient-derived tumor cells and suppresses the proliferation and M2-like polarization of tumor-associated macrophages isolated from patients with cancer. In preclinical models of solid tumors in female mice, treatment with MALT1 antisense oligonucleotides overcomes resistance to immune-checkpoint inhibitors. Together, our study demonstrates that targeting MALT1 is a potential strategy to overcome immune-checkpoint inhibitor resistance.
Collapse
Affiliation(s)
- Yuwei Tao
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Chen Tian
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Shaolong Qi
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Ziqi Jia
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Xu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingjing Meng
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Guoyuan Xu
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Haitian Hu
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Xuxiang Wang
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Tengjiang Zhang
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Huiwen You
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Xun Lan
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Xin Lin
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Haitao Zhou
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaqi Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hanqiu Zheng
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China.
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
36
|
Song J, Zhu J, Jiang Y, Guo Y, Liu S, Qiao Y, Du Y, Li J. Advancements in immunotherapy for gastric cancer: Unveiling the potential of immune checkpoint inhibitors and emerging strategies. Biochim Biophys Acta Rev Cancer 2025; 1880:189277. [PMID: 39938663 DOI: 10.1016/j.bbcan.2025.189277] [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: 06/10/2024] [Revised: 01/08/2025] [Accepted: 02/04/2025] [Indexed: 02/14/2025]
Abstract
Gastric cancer (GC) is linked to high morbidity and mortality rates. Approximately two-thirds of GC patients are diagnosed at an advanced or metastatic stage. Conventional treatments for GC, including surgery, radiotherapy, and chemotherapy, offer limited prognostic improvement. Recently, immunotherapy has gained attention for its promising therapeutic effects in various tumors. Immunotherapy functions by activating and regulating the patient's immune cells to target and eliminate tumor cells, thereby reducing the tumor burden in the body. Among immunotherapies, immune checkpoint inhibitors (ICIs) are the most advanced. ICIs disrupt the inhibitory protein-small molecule (PD-L1, CTLA4, VISTA, TIM-3 and LAG3) interactions produced by immune cells, reactivating these cells to recognize and attack tumor cells. However, adverse reactions and resistance to ICIs hinder their further clinical and experimental development. Therefore, a comprehensive understanding of the advancements in ICIs for GC is crucial. This article discusses the latest developments in clinical trials of ICIs for GC and examines combination therapies involving ICIs (targeted therapy, chemotherapy, radiotherapy), alongside ongoing clinical trials. Additionally, the review investigates the tumor immune microenvironment and its role in non-responsiveness to ICIs, highlighting the function of tumor immune cells in ICI efficacy. Finally, the article explores the prospects and limitations of new immunotherapy-related technologies, such as tumor vaccines, nanotechnologies, and emerging therapeutic strategies, aiming to advance research into personalized and optimized immunotherapy for patients with locally advanced gastric cancer.
Collapse
Affiliation(s)
- Jiawei Song
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China; Department of Experimental Surgery, Xijing Hospital, Xi'an 710038, China
| | - Jun Zhu
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Yu Jiang
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Yajie Guo
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Shuai Liu
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Yihuan Qiao
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Yongtao Du
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Jipeng Li
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China; Department of Experimental Surgery, Xijing Hospital, Xi'an 710038, China.
| |
Collapse
|
37
|
Yao Z, Fan J, Bai Y, He J, Zhang X, Zhang R, Xue L. Unravelling Cancer Immunity: Coagulation.Sig and BIRC2 as Predictive Immunotherapeutic Architects. J Cell Mol Med 2025; 29:e70525. [PMID: 40159652 PMCID: PMC11955421 DOI: 10.1111/jcmm.70525] [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: 02/04/2025] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) represent a groundbreaking advancement in cancer therapy, substantially improving patient survival rates. Our comprehensive research reveals a significant positive correlation between coagulation scores and immune-related gene expression across 30 diverse cancer types. Notably, tumours exhibiting high coagulation scores demonstrated enhanced infiltration of cytotoxic immune cells, including CD8+ T cells, natural killer (NK) cells, and macrophages. Leveraging the TCGA pan-cancer database, we developed the Coagulation.Sig model, a sophisticated predictive framework utilising a coagulation-related genes (CRGs) to forecast immunotherapy outcomes. Through rigorous analysis of ten ICI-treated cohorts, we identified and validated seven critical CRGs: BIRC2, HMGB1, STAT2, IFNAR1, BID, SPATA2, IL33 and IFNG, which form the foundation of our predictive model. Functional analyses revealed that low-risk tumours characterised by higher immune cell populations, particularly CD8+ T cells, demonstrated superior ICI responses. These tumours also exhibited increased mutation rates, elevated neoantigen loads, and greater TCR/BCR diversity. Conversely, high-risk tumours displayed pronounced intratumor heterogeneity (ITH) and elevated NRF2 pathway activity, mechanisms strongly associated with immune evasion. Experimental validation highlighted BIRC2 as a promising therapeutic target. Targeted BIRC2 knockdown, when combined with anti-PD-1 therapy, significantly suppressed tumour growth, enhanced CD8+ T cell infiltration, and amplified IFN-γ and TNF-α secretion in tumour models. Our findings position the Coagulation.Sig model as a novel, comprehensive approach to personalised cancer treatment, with BIRC2 emerging as both a predictive biomarker and a potential therapeutic intervention point.
Collapse
Affiliation(s)
- Ziang Yao
- Department of Traditional Chinese MedicinePeking University People's HospitalBeijingChina
| | - Jun Fan
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yucheng Bai
- Department of Thoracic SurgeryFirst Affiliated Hospital, Anhui Medical UniversityHefeiChina
| | - Jiakai He
- Department of Traditional Chinese MedicinePeking University People's HospitalBeijingChina
| | - Xiang Zhang
- Department of Respiratory and Critical Care MedicineThe Affiliated Huai'an Hospital of Xuzhou Medical University, the Second People's Hospital of Huai'anHuai'anJiangsuChina
| | - Renquan Zhang
- Department of Thoracic SurgeryFirst Affiliated Hospital, Anhui Medical UniversityHefeiChina
| | - Lei Xue
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
38
|
Kus ME, Sahin C, Kilic E, Askin A, Ozgur MM, Karahanogullari G, Aksit A, O'Connell RM, Ekiz HA. TCGEx: a powerful visual interface for exploring and analyzing cancer gene expression data. EMBO Rep 2025; 26:1863-1890. [PMID: 40033050 PMCID: PMC11976970 DOI: 10.1038/s44319-025-00407-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 03/05/2025] Open
Abstract
Analyzing gene expression data from the Cancer Genome Atlas (TCGA) and similar repositories often requires advanced coding skills, creating a barrier for many researchers. To address this challenge, we developed The Cancer Genome Explorer (TCGEx), a user-friendly, web-based platform for conducting sophisticated analyses such as survival modeling, gene set enrichment analysis, unsupervised clustering, and linear regression-based machine learning. TCGEx provides access to preprocessed TCGA data and immune checkpoint inhibition studies while allowing integration of user-uploaded data sets. Using TCGEx, we explore molecular subsets of human melanoma and identify microRNAs associated with intratumoral immunity. These findings are validated with independent clinical trial data on immune checkpoint inhibitors for melanoma and other cancers. In addition, we identify cytokine genes that can be used to predict treatment responses to various immune checkpoint inhibitors prior to treatment. Built on the R/Shiny framework, TCGEx offers customizable features to adapt analyses for diverse research contexts and generate publication-ready visualizations. TCGEx is freely available at https://tcgex.iyte.edu.tr , providing an accessible tool to extract insights from cancer transcriptomics data.
Collapse
Affiliation(s)
- M Emre Kus
- The Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - Cagatay Sahin
- The Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - Emre Kilic
- The Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - Arda Askin
- The Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - M Mert Ozgur
- The Department of Molecular Biology and Genetics, Bilkent University, 06800, Cankaya, Ankara, Turkey
| | - Gokhan Karahanogullari
- The Department of Mathematics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - Ahmet Aksit
- The Department of Information Technologies, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - Ryan M O'Connell
- The Department of Pathology, University of Utah, Salt Lake City, UT, 84112, USA
| | - H Atakan Ekiz
- The Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey.
| |
Collapse
|
39
|
Yang Z, Yu B, Hu J, Jiang L, Jian M. LRRC25 Is a Potential Biomarker for Predicting Immunotherapy Response in Patients with Gastric Cancer. Dig Dis Sci 2025; 70:1395-1410. [PMID: 39961962 PMCID: PMC11972229 DOI: 10.1007/s10620-025-08882-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 01/17/2025] [Indexed: 04/06/2025]
Abstract
BACKGROUND Leucine-rich repeat containing 25 (LRRC25) is distinguishingly expressed in different tumor types, but the relationship with immune cell infiltration in gastric cancer stills unclear. We analyzed LRRC25 expression using pan-cancer data from The Cancer Genome Atlas and gene data from Gene Expression Omnibus. The clinical significance was further evaluated using gastric cancer tissues derived from clinical trials (no. NCT04208347). METHOD Through bioinformatics analysis of TCGA database and the UCSC Xena database, we found the correlation between LRRC15, gastric cancer, and immune cell infiltration. Further, multiplex immunohistochemistry/immunofluorescence (mIHC/IF), tissue microarray, and image acquisition and quantitative analysis confirmed our theory. RESULTS It was discovered that LRRC25 was highly expressed in gastric cancer. Further analysis revealed that the expression of LRRC25 associated with gene sets implicated in immunity, including those in innate immunity, adaptive immunity, and chemokine signaling pathways. The result of (mIHC/IF) suggests a negative relevance between LRRC25 and response of anti-PD-1 treatment and reveals a trend of consistent change on LRRC25 + cells and CD16 expression. We also discovered that LRRC25 expression significantly associated with immune cell infiltration level. In particular, there is a relationship between LRRC25 and the phenotype and function of NK cells. CONCLUSION High LRRC25 expression contributes to immunosuppressive microenvironment by influencing chemokine axis in gastric cancer. LRRC25 may serve as a clinically useful biomarker for predicting neoadjuvant immunotherapeutic response in patients with gastric cancer. LRRC25 can affect the phenotype and function of NK cells.
Collapse
Affiliation(s)
- Zhensong Yang
- Department of Gastrointestinal Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China
| | - Bin Yu
- Department of Gastrointestinal Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China
| | - Jinchen Hu
- Department of Gastrointestinal Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China
| | - Lixin Jiang
- Department of Gastrointestinal Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China
- Department of Surgical Department, The Yeda Hospital of Yantai City, Yantai, Shandong, China
| | - Mi Jian
- Department of Gastrointestinal Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China.
| |
Collapse
|
40
|
Ye B, Fan J, Xue L, Zhuang Y, Luo P, Jiang A, Xie J, Li Q, Liang X, Tan J, Zhao S, Zhou W, Ren C, Lin H, Zhang P. iMLGAM: Integrated Machine Learning and Genetic Algorithm-driven Multiomics analysis for pan-cancer immunotherapy response prediction. IMETA 2025; 4:e70011. [PMID: 40236779 PMCID: PMC11995183 DOI: 10.1002/imt2.70011] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 04/17/2025]
Abstract
To address the substantial variability in immune checkpoint blockade (ICB) therapy effectiveness, we developed an innovative R package called integrated Machine Learning and Genetic Algorithm-driven Multiomics analysis (iMLGAM), which establishes a comprehensive scoring system for predicting treatment outcomes through advanced multi-omics data integration. Our research demonstrates that iMLGAM scores exhibit superior predictive performance across independent cohorts, with lower scores correlating significantly with enhanced therapeutic responses and outperforming existing clinical biomarkers. Detailed analysis revealed that tumors with low iMLGAM scores display distinctive immune microenvironment characteristics, including increased immune cell infiltration and amplified antitumor immune responses. Critically, through clustered regularly interspaced short palindromic repeats screening, we identified Centrosomal Protein 55 (CEP55) as a key molecule modulating tumor immune evasion, mechanistically confirming its role in regulating T cell-mediated antitumor immune responses. These findings not only validate iMLGAM as a powerful prognostic tool but also propose CEP55 as a promising therapeutic target, offering novel strategies to enhance ICB treatment efficacy. The iMLGAM package is freely available on GitHub (https://github.com/Yelab1994/iMLGAM), providing researchers with an innovative approach to personalized cancer immunotherapy prediction.
Collapse
Affiliation(s)
- Bicheng Ye
- Liver Disease Center of Integrated Traditional Chinese and Western Medicine, Department of Radiology, Zhongda Hospital, Medical SchoolSoutheast University, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University)NanjingChina
| | - Jun Fan
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Lei Xue
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yu Zhuang
- Department of Thoracic Surgery, Nanjing Chest HospitalNanjingChina
- Afliated Nanjing Brain HospitalNanjing Medical UniversityNanjingChina
| | - Peng Luo
- Department of Oncology, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Aimin Jiang
- Department of Urology, Changhai HospitalNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Jiaheng Xie
- Department of Plastic Surgery, Xiangya HospitalCentral South UniversityChangshaChina
| | - Qifan Li
- Department of Thoracic SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xiaoqing Liang
- Chongqing Key Laboratory of Molecular Oncology and EpigeneticsThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Jiaxiong Tan
- Department of Lung Cancer, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Songyun Zhao
- Department of Plastic SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Wenhang Zhou
- Department of OncologyThe Affiliated Huai'an Hospital of Xuzhou Medical University, the Second People's Hospital of Huai'anHuai'anChina
| | - Chuanli Ren
- Department of Laboratory MedicineNorthern Jiangsu People's Hospital Affiliated to Yangzhou UniversityYangzhouChina
| | - Haoran Lin
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Pengpeng Zhang
- Department of Thoracic SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Department of Lung Cancer, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjinChina
| |
Collapse
|
41
|
Wang X, Hui H, Han J, Guo T, Wang Y, Meng L, Chen C, He J, Guo X, Zhong F, Du H, Tian J, Xing X, Du Y, Ji J. A CLDN18.2-Targeted Nanoplatform Manipulates Magnetic Hyperthermia Spatiotemporally for Synergistic Immunotherapy in Gastric Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413913. [PMID: 40019387 PMCID: PMC12021038 DOI: 10.1002/advs.202413913] [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: 10/29/2024] [Revised: 01/06/2025] [Indexed: 03/01/2025]
Abstract
Precision treatment of gastric cancer requires specific biomarkers, and CLDN18.2 emerges as a promising target for patients' stratification and therapeutic guidance. In 563 cases, 54.4% of patients are identified as CLDN18.2-positive, with CLDN18.2 expression negatively correlated with immune-related factors like PD-L1, indicating a "cold" tumor microenvironment. Here, a novel CLDN18.2 monoclonal antibody 1D5 is created with superior high specificity and affinity, and the antibody-dependent fluorescence-magnetic nanoparticle is developed for specific detection and magnetic hyperthermia (MHT). Under the assistance of sensitive fluorescence and deep-penetrating magnetic particle imaging for tracing and timing the optimal nanoparticle dosage, MHT induces robust immunogenic response via DNA mismatch repair and tumor-associated antigen release. It recruits CD11c+ dendritic cells, compensates PD-1 in CD8+ T cells, and enhances CD86+ macrophage polarization. The combination of anti-PD-1 therapy increased TNF-α and IFN-γ secretion and further boosted the cytotoxic efficacy of CD8+ T cells. Excellent therapeutic efficacy is found simultaneously on cell-derived allografts and patient-derived xenografts based on this spatiotemporally manipulated strategy, presenting a therapeutic option for enhancing responsiveness to immunotherapy for CLDN18.2-positive individuals.
Collapse
Affiliation(s)
- Xueying Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Gastrointestinal Cancer Translational ResearchPeking University Cancer Hospital & InstituteBeijing100142China
- CAS Key Laboratory of Molecular ImagingInstitute of AutomationChinese Academy of SciencesBeijing100190China
| | - Hui Hui
- CAS Key Laboratory of Molecular ImagingInstitute of AutomationChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100080China
| | - Jing Han
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Gastrointestinal Cancer Translational ResearchPeking University Cancer Hospital & InstituteBeijing100142China
- CAS Key Laboratory of Molecular ImagingInstitute of AutomationChinese Academy of SciencesBeijing100190China
| | - Ting Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Gastrointestinal Cancer Translational ResearchPeking University Cancer Hospital & InstituteBeijing100142China
| | - Yiding Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Gastrointestinal Cancer Translational ResearchPeking University Cancer Hospital & InstituteBeijing100142China
| | - Lin Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Department of Biochemistry and Molecular BiologyPeking University Cancer Hospital & InstituteBeijing100142China
| | - Cong Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Gastrointestinal Cancer Translational ResearchPeking University Cancer Hospital & InstituteBeijing100142China
| | - Jie He
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Gastrointestinal Cancer Translational ResearchPeking University Cancer Hospital & InstituteBeijing100142China
- School of Engineering Medicine & School of Biological Science and Medical EngineeringBeihang UniversityBeijing100191China
| | - Xiaoyong Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Gastrointestinal Cancer Translational ResearchPeking University Cancer Hospital & InstituteBeijing100142China
| | - Fuyu Zhong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Gastrointestinal Cancer Translational ResearchPeking University Cancer Hospital & InstituteBeijing100142China
| | - Hong Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Gastrointestinal Cancer Translational ResearchPeking University Cancer Hospital & InstituteBeijing100142China
| | - Jie Tian
- School of Engineering Medicine & School of Biological Science and Medical EngineeringBeihang UniversityBeijing100191China
- Key Laboratory of Big Data‐Based Precision Medicine (Beihang University)Ministry of Industry and Information Technology of ChinaBeijing100191China
| | - Xiaofang Xing
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersBeijing Key Laboratory of Carcinogenesis and Translational ResearchGastrointestinal Cancer CentrePeking University Cancer Hospital & InstituteBeijing100142China
| | - Yang Du
- CAS Key Laboratory of Molecular ImagingInstitute of AutomationChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100080China
| | - Jiafu Ji
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersBeijing Key Laboratory of Carcinogenesis and Translational ResearchGastrointestinal Cancer CentrePeking University Cancer Hospital & InstituteBeijing100142China
| |
Collapse
|
42
|
Pu W, Li S, Zhang J, Huang J, Li J, Jiang Y, Xu Z, Yi F, Lan Y, Xiao Q, Chen W, Jin J. The efficacy and safety of PD-1/PD-L1 inhibitors in combination with chemotherapy as a first-line treatment for unresectable, locally advanced, HER2-negative gastric or gastroesophageal junction cancer: a meta-analysis of randomized controlled trials. Front Immunol 2025; 16:1566939. [PMID: 40207218 PMCID: PMC11979168 DOI: 10.3389/fimmu.2025.1566939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/11/2025] [Indexed: 04/11/2025] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) plus fluorouracil-based chemotherapy (Chemo) have been approved as an initial treatment strategy for metastatic or recurrent human epidermal growth factor receptor 2 (HER2)-negative gastric cancer (GC) or gastroesophageal junction cancer (GEJC). However, since programmed cell death protein-1 (PD-1) or its ligand 1 (PD-L1) inhibitors have just recently been investigated for the treatment of unresectable GC/GEJC, there is ongoing debate regarding their safety and effectiveness for prespecified subgroups. The purpose of this research is to establish a foundation toward stratified decision-making by methodically assessing the merits and drawbacks of PD-1/PD-L1 inhibitors combined with chemo in the clinical utilization of advanced HER2-negative GC/GEJC according to certain prominent large-scale randomized controlled trials (RCTs). In addition, we limitedly explored the favorable short-term efficacy of PD-1/CTLA-4 bispecific antibodies for the above-mentioned tumors. Methods The researchers retrieved several databases, including PubMed, Embase, Web of Science, ClinicalTrials.gov, and the Cochrane Library, to collect all the relevant literature published since the establishment of the databases until October 30, 2024, and then screened to determine the qualified literature and extracted the relevant information. We only included RCTs for PD-1/PD-L1 inhibitors with or without chemo in advanced GC or GEJC. The primary endpoints were overall survival (OS), progression-free survival (PFS), and objective response rate (ORR). A subgroup analysis for the median overall survival (mOS) was conducted for the following variables: microsatellite instability (MSI) status, PD-L1 expression, combined positive scores (CPS), metastasis status, and primary tumor location. When moderate heterogeneity was found, a random-effect model was applied. The outcome indicators were then statistically analyzed, taking advantage of Review Manager 5.4. Hazard ratio (HR) and risk ratio (RR) were selected as the effect values for statistical analysis. Results A total of 7 eligible RCTs and 6537 participants were included in this meta-analysis. Combining PD-1/PD-L1 inhibitors with chemo significantly improved patients' OS compared with chemo alone, especially in the tumor cell PD-L1 expression ≥ 1% [HR = 0.62, 95% CI (0.48, 0.81); a p-value = 0.0004], PD-L1 CPS ≥ 10 [HR = 0.66, 95% CI (0.57, 0.77); a p-value < 0.00001], and MSI-H subgroups [HR = 0.40, 95% CI (0.28, 0.59); a p-value < 0.00001]. Moreover, distinct primary tumor location (GC or GEJC) and the presence of liver metastases could also benefit from the additive or sustained effect of anti-cancer chemo-immunotherapy. Conclusion For patients with advanced HER2-negative GC/GEJC, PD-1/PD-L1 inhibitors in combination with chemo have almost demonstrated consistent synergistic anti-tumor benefits to survival outcomes when compared to chemo alone. However, the subgroup analysis in this meta-study revealed that neither PD-L1 expression level nor MSI status could fully predict the efficacy of the dual treatment model but faced a higher possibility of serious treatment-related adverse events (sTRAEs), particularly in the synchronous therapy arm. Therefore, urging the need for more investigations into the development of collaborative prognostic forecasting models for achieving precise stratification, established harmonized testing standards and methods for PD-L1 expression and positivity, optimal CPS threshold for benefits, as well as alternative molecular biomarkers for the reason that certain indicators alone may not discriminate responders clearly. Lastly, dual anti-therapy might be a useful tactic for the population with low PD-L1 expression in the future.
Collapse
Affiliation(s)
- Wenji Pu
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
- Medical Department of Shenzhen University/General Hospital of Shenzhen University/Academy of Clinical Medicine of Shenzhen University, Shenzhen, China
| | - Shasha Li
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jinliang Zhang
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jijie Huang
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jishi Li
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yong Jiang
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zhiyuan Xu
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Fan Yi
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yuling Lan
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Qin Xiao
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Wenqi Chen
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jing Jin
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
43
|
Fauvre A, Ursino C, Garambois V, Culerier E, Milazzo LA, Vezzio-Vié N, Jeanson L, Marchive C, Andrade AF, Combes E, Atis S, Lossaint G, Quenet F, Michaud HA, Khellaf L, Corbeau I, Tosi D, Houede N, Bonnefoy N, Sgarbura O, Gongora C, Faget J. Oxaliplatin, ATR inhibitor and anti-PD-1 antibody combination therapy controls colon carcinoma growth, induces local and systemic changes in the immune compartment, and protects against tumor rechallenge in mice. J Immunother Cancer 2025; 13:e010791. [PMID: 40139833 PMCID: PMC11950992 DOI: 10.1136/jitc-2024-010791] [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: 10/21/2024] [Accepted: 03/01/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most common cancer type and one of the leading causes of cancer-related death worldwide. The treatment of advanced metastatic CRC relies on classical chemotherapy combinations (5-fluorouracil, oxaliplatin or irinotecan). However, their use is limited by the emergence of resistance mechanisms, including to oxaliplatin. In this context, we recently showed that the combination of oxaliplatin and ataxia telangiectasia and Rad3-related protein inhibition (VE-822) is synergistic and may have a potential therapeutic effect in metastatic CRC management. METHODS In this study, we investigated the role of the VE-822+oxaliplatin (Vox) combination on the immune response and its potential synergy with an anti-programmed-cell Death receptor-1 (PD-1) antibody. We used cell lines and organoids from metastatic CRC to investigate in vitro Vox efficacy and orthotopic syngeneic mouse models of metastatic CRC to assess the efficacy of Vox+anti-PD-1 antibody and identify the involved immune cells. RESULTS The Vox+anti-PD-1 antibody combination completely cured tumor-bearing mice and protected them from a rechallenge. Vox was associated with a reduction of tumor-infiltrated neutrophils, CD206+ macrophages and regulatory T cells. Vox also induced a deep depletion of blood neutrophils. The increased bone marrow granulopoiesis failed to compensate for the Vox-mediated mature neutrophil depletion. Neutrophil depletion using a mouse recombinant anti-Ly6G antibody partially mimicked the Vox effect on the tumor microenvironment, but to a lower extent compared with the Vox+anti-PD-1 antibody combination. Vox, but not neutrophil depletion, led to the emergence of an Ly6C+ PD-1+ CD8+ T-cell population in the blood and spleen of tumor-harboring mice. These cells were proliferating, and expressed IFN-γ, CD62L, CXCR3 and Eomes. Moreover, the proportion of tumor antigen-specific T cells and of CD122+ BCL6+ T cells, which shared phenotypic characteristics with stem-like CD8+ T cells, was increased in treated mice. CONCLUSIONS Our work strongly suggests that the Vox+anti-PD-1 antibody combination might significantly improve survival in patients with metastatic and treatment-refractory CRC by acting both on cancer cells and CD8+ T cells.
Collapse
Affiliation(s)
- Alexandra Fauvre
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Chiara Ursino
- Immunity and Cancer Team, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Veronique Garambois
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Elodie Culerier
- Immunity and Cancer Team, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Louis-Antoine Milazzo
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Nadia Vezzio-Vié
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Laura Jeanson
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Candice Marchive
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Augusto Faria Andrade
- McGill University/Research Institute of McGill University, Nada Jabado Lab, Montreal, Quebec, Canada
| | - Eve Combes
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Salima Atis
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Gérald Lossaint
- Institut regional du Cancer de Montpellier, Montpellier, France
| | - François Quenet
- Institut regional du Cancer de Montpellier, Montpellier, France
| | - Henri-Alexandre Michaud
- Immunity and Cancer Team, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Lakhdar Khellaf
- Department of Pathology, Montpellier University, Montpellier, France
| | - Ileana Corbeau
- Institut regional du Cancer de Montpellier, Montpellier, France
| | - Diego Tosi
- Medical Oncology Department, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Nadine Houede
- Department of Oncology, University Hospital of Nimes, Nîmes, France
| | - Nathalie Bonnefoy
- Immunity and Cancer Team, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Olivia Sgarbura
- Institut regional du Cancer de Montpellier, Montpellier, France
| | - Céline Gongora
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
- CNRS, Paris, France
| | - Julien Faget
- Immunity and Cancer Team, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| |
Collapse
|
44
|
Wang T, Cheng Y, Hu F, Wang Q. Residual gastric cancer with a mixed small cell neuroendocrine and keratinizing squamous cell carcinoma: A case report. World J Clin Oncol 2025; 16:102301. [PMID: 40130043 PMCID: PMC11866079 DOI: 10.5306/wjco.v16.i3.102301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/19/2024] [Accepted: 12/09/2024] [Indexed: 01/21/2025] Open
Abstract
BACKGROUND Despite advancements in early detection and treatment, the prognosis and histological types for residual gastric cancer (GC) remains poor. CASE SUMMARY This case report presents a rare occurrence of residual GC featuring a combination of small cell neuroendocrine carcinoma (SCNEC) and squamous cell carcinoma (SCC) in a 60-year-old male patient. The patient, with a history of Billroth II gastrectomy for duodenal ulcer bleeding, presented with gastrointestinal bleeding. Preoperative computed tomography and positron emission tomography-computed tomography indicated adenocarcinoma with tumor and abdominal lymph node metastasis. The patient underwent laparoscopic total gastrectomy and lymph node dissection for residual GC. Histological examination of the resected tumor confirmed the presence of both SCNEC and SCC. Postoperatively, the patient underwent adjuvant chemotherapy four times. Two years later, the patient was found to occur esophageal cancer and was performed a small bowel stoma and radical esophagectomy. CONCLUSION In this case report, we detail a rare instance of residual GC with mixed SCNEC and SCC, emphasizing the complexity of diagnosis and treatment, and the need for ongoing research.
Collapse
Affiliation(s)
- Tian Wang
- Department of Gastroenterology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330000, Jiangxi Province, China
| | - Yang Cheng
- Department of Pathology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330000, Jiangxi Province, China
| | - Fan Hu
- Department of Pathology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330000, Jiangxi Province, China
| | - Qiang Wang
- Department of Gastroenterology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330000, Jiangxi Province, China
| |
Collapse
|
45
|
Yu S, Gao Y, Zhao F, Zhou J, Zhang J. Metabolites and metabolic pathway reactions links to sensitization of immunotherapy in pan-cancer. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200933. [PMID: 39968095 PMCID: PMC11834090 DOI: 10.1016/j.omton.2025.200933] [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] [Received: 08/29/2024] [Revised: 11/29/2024] [Accepted: 01/10/2025] [Indexed: 02/20/2025]
Abstract
Metabolic features are crucial in tumor immune interactions, but their relationship with antitumor immune responses is not yet fully understood. This study used Mendelian randomization analysis to identify the causal relationships between blood metabolites and immune cells and to evaluate the effects of metabolic pathways and reactions on antitumor immune responses in various cancers. Levels of 156 metabolites exhibited significant associations with selected immune cells. Metabolic enrichment analysis indicated laurate, propionyl-carnitine, carnitine and l-acetylcarnitine are enriched in fatty acid (FA) metabolism pathways. These enriched pathways are significantly correlated to CD8+ T cell function signatures in tumor environment and favor better prognostic outcomes. Metabolic reactions contributing to better immunotherapy responses were identified and used to establish the immuno-metabolic reaction score (IMRS). IMRS were significantly correlated to CD8+ T cell infiltration levels and CD8+ T cell signature scores in either 10× Visium spatial transcriptomic or RNA-seq samples. Finally, IMRS could significantly predict favorable survival outcomes in different cancer patients treated with immunotherapy. Our study revealed a link between certain metabolites and their related metabolic pathways to tumor immune landscape and immune functions. These results could promote the accurate stratification of patients before treatment and improve the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Shaobo Yu
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou 310016, Zhejiang, China
| | - Yuzhen Gao
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou 310016, Zhejiang, China
| | - Feng Zhao
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou 310016, Zhejiang, China
| | - Jiaqiang Zhou
- Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
| | - Jun Zhang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou 310016, Zhejiang, China
| |
Collapse
|
46
|
Wu X, Hou S, Ye Y, Gao Z. CXCR2P1 enhances the response of gastric cancer to PD-1 inhibitors through increasing the immune infiltration of tumors. Front Immunol 2025; 16:1545605. [PMID: 40176817 PMCID: PMC11961440 DOI: 10.3389/fimmu.2025.1545605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 03/03/2025] [Indexed: 04/04/2025] Open
Abstract
Background Recent years, immunotherapy has emerged as a pivotal approach in cancer treatment. However, the response of gastric cancer to immunotherapy exhibits significant heterogeneity. Therefore, the early identification of gastric cancer patients who are likely to benefit from immunotherapy and the discovery of novel therapeutic targets are of critical importance. Materials and methods We collected data from European Nucleotide Archive (ENA) and Gene Expression Omnibus (GEO) databases. In project PRJEB25780, we performed WGCNA analysis and Lasso regression and chose CXCR2P1 for the subsequent analysis. Then, we compared the expression difference of CXCR2P1 among different groups. Kaplan-Meier curve was used to analyze the prognostic value of CXCR2P1, which was validated by project IMvigor210 and GEO datasets. ESTIMATE and CIBERSORT algorithm were used to evaluate the reshaping effect of CXCR2P1 to immune microenvironment of tumor. Differentially expressed genes (DEG) analysis, enrichGO analysis, Gene Set Enrichment Analysis (GSEA) and co-expression analysis were used to explore the cell biological function and signaling pathway involved in CXCR2P1. Results WGCNA identified CXCR2P1 as a hub gene significantly associated with immune response to PD-1 inhibitors in gastric cancer. CXCR2P1 expression was elevated in responders and correlated with better prognosis. Functional analysis revealed its role in reshaping the tumor immune microenvironment by promoting immune cell infiltration, including M1 macrophages, activated CD4+ T cells, and follicular helper T cells. CXCR2P1 enhanced antigen presentation via the MHC-II complex, influenced key immune pathways, such as Toll-like receptor signaling and T-cell activation, which led to the up-regulation of expression of PD-L1. GSEA showed CXCR2P1 were correlated with microRNAs. Through DEG analysis and expression analysis, MIR215 was identified as a potential direct target of CXCR2P1. Conclusion High expression of CXCR2P1 is correlated with better response to PD-1 inhibitor. It reshapes the immune microenvironment by increasing immune infiltration and changing the fraction of immune cells. In tumor immune microenvironment, CXCR2P1 can promote inflammation, enhance antigen presentation and activate the PD-1/PD-L1-related signaling pathway, which might be achieved by CXCR2P1-MIR215 axis.
Collapse
Affiliation(s)
- Xinchun Wu
- Department of Gastrointestinal Surgery, Peking University People`s Hospital, Beijing, China
- Laboratory of Surgical Oncology, Peking University People`s Hospital, Beijing, China
| | - Sen Hou
- Department of Gastrointestinal Surgery, Peking University People`s Hospital, Beijing, China
- Laboratory of Surgical Oncology, Peking University People`s Hospital, Beijing, China
| | - Yingjiang Ye
- Department of Gastrointestinal Surgery, Peking University People`s Hospital, Beijing, China
- Laboratory of Surgical Oncology, Peking University People`s Hospital, Beijing, China
- Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People’s Hospital, Beijing, China
| | - Zhidong Gao
- Department of Gastrointestinal Surgery, Peking University People`s Hospital, Beijing, China
- Laboratory of Surgical Oncology, Peking University People`s Hospital, Beijing, China
- Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
47
|
Veas Rodriguez J, Piñol M, Sorolla MA, Parisi E, Sorolla A, Santacana M, Ruiz M, Parra G, Bernabeu M, Iglesias M, Aracil C, Escartin A, Vilardell F, Matias-Guiu X, Salud A, Montal R. Comprehensive immunophenotyping of gastric adenocarcinoma identifies an inflamed class of tumors amenable to immunotherapies. J Immunother Cancer 2025; 13:e010024. [PMID: 40102027 PMCID: PMC11927434 DOI: 10.1136/jitc-2024-010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 02/22/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Gastric adenocarcinoma (GAC) imposes a considerable global health burden. Molecular profiling of GAC from the tumor microenvironment perspective through a multi-omics approach is eagerly awaited in order to allow a more precise application of novel therapies in the near future. METHODS To better understand the tumor-immune interface of GAC, we identified an internal cohort of 82 patients that allowed an integrative molecular analysis including mutational profiling by whole-exome sequencing, RNA gene expression of 770 genes associated with immune response, and multiplex protein expression at spatial resolution of 34 immuno-oncology targets at different compartments (tumorous cells and immune cells). Molecular findings were validated in 595 GAC from the TCGA and ACRG external cohorts with available multiomics data. Prediction of response to immunotherapies of the discovered immunophenotypes was assessed in 1039 patients with cancer from external cohorts with available transcriptome data. RESULTS Unsupervised clustering by gene expression identified a subgroup of GAC that includes 52% of the tumors, the so-called Inflamed class, characterized by high tumor immunogenicity and cytotoxicity, particularly in the tumor center at protein level, with enrichment of PIK3CA and ARID1A mutations and increased presence of exhausted CD8+ T cells as well as co-inhibitory receptors such as PD1, CTLA4, LAG3, and TIGIT. The remaining 48% of tumors were called non-inflamed based on the observed exclusion of T cell infiltration, with an overexpression of VEGFA and higher presence of TP53 mutations, resulting in a worse clinical outcome. A 10-gene RNA signature was developed for the identification of tumors belonging to these classes, demonstrating in evaluated datasets comparable clinical utility in predicting response to current immunotherapies when tested against other published gene signatures. CONCLUSIONS Comprehensive immunophenotyping of GAC identifies an inflamed class of tumors that complements previously proposed tumor-based molecular clusters. Such findings may provide the rationale for exploring novel immunotherapeutic approaches for biomarker-enriched populations in order to improve GAC patient's survival.
Collapse
Affiliation(s)
- Joel Veas Rodriguez
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Miquel Piñol
- Department of Pathology, Oncological Pathology Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Maria Alba Sorolla
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Eva Parisi
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Anabel Sorolla
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Maria Santacana
- Scientific and Technical Service of Immunohistochemistry, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Maria Ruiz
- Scientific and Technical Service of Biobank, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Genís Parra
- CNAG-Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mario Bernabeu
- CNAG-Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mar Iglesias
- Department of Pathology, Hospital del Mar, University Pompeu Fabra, Hospital del Mar Research Institute, CIBERONC, Barcelona, Spain
| | - Carles Aracil
- Department of Gastroenterology, Clinical and Experimental Research in Digestive and Hematological Pathology Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Alfredo Escartin
- Department of Surgery, Experimental Surgery Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Felip Vilardell
- Department of Pathology, Oncological Pathology Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Xavier Matias-Guiu
- Department of Pathology, Oncological Pathology Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Antonieta Salud
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| | - Robert Montal
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova - IRBLleida, Lleida, Spain
| |
Collapse
|
48
|
Zhou X, Xu T, Li C, He Y, Hu Y, Gong H, Li J, Jiang H, Wen L, Fu Y, Zeng Z, Pan D. Potentiating anti-tumor immunity by re-engaging immune synapse molecules. Cell Rep Med 2025; 6:101975. [PMID: 39999838 PMCID: PMC11970328 DOI: 10.1016/j.xcrm.2025.101975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/22/2024] [Accepted: 01/28/2025] [Indexed: 02/27/2025]
Abstract
The formation of immune synapses (ISs) between cytotoxic T cells and tumor cells is crucial for effective tumor elimination. However, the role of ISs in immune evasion and resistance to immune checkpoint blockades (ICBs) remains unclear. We demonstrate that ICAM-1, a key IS molecule activating LFA-1 signaling in T and natural killer (NK) cells, is often expressed at low levels in cancers. The absence of ICAM-1 leads to significant resistance to T and NK cell-mediated anti-tumor immunity. Using a CRISPR screen, we show that ICAM-1 is epigenetically regulated by the DNA methylation pathway involving UHRF1 and DNMT1. Furthermore, we engineer an antibody-based therapeutic agent, "LFA-1 engager," to enhance T cell-mediated anti-tumor immunity by reconstituting LFA-1 signaling. Treatment with LFA-1 engagers substantially enhances immune-mediated cytotoxicity, potentiates anti-tumor immunity, and synergizes with ICB in mouse models of ICAM-1-deficient tumors. Our data provide promising therapeutic strategies for re-engaging immune stimulatory signals in cancer immunotherapy.
Collapse
Affiliation(s)
- Xindi Zhou
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Tian Xu
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Changhe Li
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Yufeng He
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Yuanzhi Hu
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Hao Gong
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Jiahui Li
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Haitao Jiang
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Liang Wen
- Chinese People's Liberation Army (PLA) Medical School, Beijing 100850, China
| | - Yangxin Fu
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Zexian Zeng
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China.
| | - Deng Pan
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Science (CLS), Beijing 100084, China.
| |
Collapse
|
49
|
Chen P, Chen Z, Sui W, Han W. Recent advances in the mechanisms of PD-L1 expression in gastric cancer: a review. Biol Res 2025; 58:16. [PMID: 40091086 PMCID: PMC11912799 DOI: 10.1186/s40659-025-00597-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 03/07/2025] [Indexed: 03/19/2025] Open
Abstract
In the progression of gastric cancer (GC), various cell types in the tumor microenvironment (TME) exhibit upregulated expression of programmed death ligand 1 (PD-L1), leading to impaired T-cell function and evasion of immune surveillance. Infection with H. pylori and EBV leads to increased PD-L1 expression in various cell types within TME, resulting in immune suppression and facilitating immune escape of GC cells. In the TME, mesenchymal stem cells (MSCs), M1-like tumor-associated macrophages (MI-like TAM), and myeloid-derived suppressor cells (MDSCs) contribute to the upregulation of PD-L1 expression in GC cells. Conversely, mast cells, M2-like tumor-associated macrophages (M2-like TAM), and tumor-associated neutrophils (TANs) exhibit elevated levels of PD-L1 expression in response to the influence of GC cells. Together, these factors collectively contribute to the upregulation of PD-L1 expression in GC. This review aims to provide a comprehensive summary of the cellular expression patterns of PD-L1 in GC and the underlying molecular mechanisms. Understanding the complex regulatory pathways governing PD-L1 expression may offer novel insights for the development of effective immunotherapeutic interventions.
Collapse
Affiliation(s)
- Peifeng Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Avenue, Shushan District, Hefei, Anhui Province, 230022, China
| | - Zhangming Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Avenue, Shushan District, Hefei, Anhui Province, 230022, China
| | - Wannian Sui
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Avenue, Shushan District, Hefei, Anhui Province, 230022, China
| | - Wenxiu Han
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Avenue, Shushan District, Hefei, Anhui Province, 230022, China.
| |
Collapse
|
50
|
Guo Y, Wan R, Duan J, Yuan L, Wang Z, Zhong J, Zhang X, Ma Z, Bai H, Wang J. Targeting tumor-intrinsic S100 calcium-binding protein A1 augments antitumor immunity and potentiates immunotherapy efficacy. Signal Transduct Target Ther 2025; 10:99. [PMID: 40090947 PMCID: PMC11911448 DOI: 10.1038/s41392-025-02190-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/16/2025] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Immune checkpoint blockade (ICB) has revolutionized cancer treatment, but the therapeutic response is highly heterogeneous, which highlights the necessity for developing predictive biomarkers and overcoming ICB resistance. Cancer cell-intrinsic features, especially those that can be dynamically monitored via liquid biopsy, represent a broader scope for biomarker development. In addition, a potential mode of ICB resistance is tumor-intrinsic mechanisms leading to an immunosuppressive tumor microenvironment (TME). However, the underlying interactive network remains elusive, and the generalizable biomarkers and targeting strategies are still lacking. Here, we uncovered the potential of plasma S100 calcium-binding protein A1 (S100A1) for determining ICB efficacy via liquid biopsy of patients with lung cancer. Multiomics and functional studies have suggested that tumor-intrinsic S100A1 expression correlated with an immunologically "cold" TME and resistance to ICB in multiple syngeneic murine tumors and tissue samples from patients with lung cancer. Mechanistic investigations demonstrated that interfering with the tumor-intrinsic S100A1/ubiquitin-specific protease 7/p65/granulocyte-macrophage colony-stimulating factor (GM-CSF) modulatory axis could potentiate an inflamed TME by promoting M1-like macrophage polarization and T cell function. GM-CSF priming was sufficient to enhance the ICB response in tumors with high S100A1 expression in preclinical models. These findings define S100A1 as a potential blood-based biomarker and a novel synergistic target for cancer immunotherapy.
Collapse
Affiliation(s)
- Yufeng Guo
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Rui Wan
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianchun Duan
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Li Yuan
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhijie Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jia Zhong
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xue Zhang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zixiao Ma
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hua Bai
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|