1
|
Niu C, Song Y, Chen Y, Shi Y, Wang H, Wu X, Wang X, Zhao X, Bu Y, Li J, Tao T, Wu J, Xue C, Zhang F, Han C, Yuan J, Zhang Q. Epidemiology of Helicobacter pylori, gastric precancerous lesions and gastric cancer: a multicenter, population-based cross-sectional study in Nanjing. BMC Infect Dis 2025; 25:766. [PMID: 40426075 PMCID: PMC12117699 DOI: 10.1186/s12879-025-11147-3] [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: 09/05/2024] [Accepted: 05/20/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Nanjing City has a high-incidence gastric cancer (GC), but the epidemiology of gastric precancerous lesions (GPLs) remains poorly understood. This study aimed to investigate the epidemiological characteristics of Helicobacter pylori (H. pylori) infection, GPLs, and GCs in patients undergoing endoscopic examination in Lishui District, Nanjing. METHODS This retrospective, population-based, cross-sectional study was conducted collaboratively by the Nanjing Lishui People's Hospital and six medical community units within the county between July 2022 and June 2023. Data on biopsies and 13C urea breath tests (13C-UBT) were collected. RESULTS A total of 15,668 individuals were included, among whom 259 had GPL (1.65%) and 218 had GC (1.39%). The H. pylori infection rate in total patients was 5014 (32.00%) (males: 2684 (34.06%); females: 2335 (29.92%)). The H. pylori infection rate is 31.45% in benign gastric lesions, 44.40% in GPLs, and 55.50% in GC, respectively. The multivariable logistic regression analysis showed that male (OR = 3.156, 95% CI: 2.865-3.376, P < 0.001), age (OR = 1.785, 95% CI: 1.703-1.876, P < 0.001), fresh vegetable, fruit, and white meat intake frequently (OR = 0.865, 95% CI: 0.506-2.061, P = 0.029), high-salt diet and high-fat diet intake frequently (OR = 1.906, 95% CI: 1.101-2.932, P = 0.014), rural residence (OR = 2.682, 95% CI: 1.010-4.754, P = 0.040), H. pylori infection (OR = 2.022, 95% CI: 1.155-2.865, P < 0.001) and atrophic gastritis and/or intestinal metaplasia (OR = 4.875, 95% CI: 2.229-10.663, P < 0.001) were associated with GPLs. Male (OR = 2.021, 95% CI: 1.080-3.780, P = 2.028), age (OR = 1.201, 95% CI: 1.174-1.238, P < 0.001), digestive symptoms (OR = 2.256, 95% CI: 1.548-3.289, P < 0.001), bachelor degree below (OR = 4.792, 95% CI: 3.439-6.837, P < 0.001), farmer (OR = 1.039, 95% CI: 1.026-1.159, P < 0.001), fresh vegetable, fruit, and white meat intake (OR = 0.231, 95% CI: 0.141-0.379, P < 0.001), fried/barbecue/pickled food intake (OR = 6.781, 95% CI: 3.783-12.153, P < 0.001), high-salt diet and high-fat diet intake (OR = 4.374, 95% CI: 2.363-8.097, P < 0.001), rural residence (OR = 1.230, 95% CI: 1.121-1.437, P < 0.001), H. pylori infection (OR = 3.248, 95% CI: 2.357-4.477, P < 0.001) and atrophic gastritis and/or intestinal metaplasia (OR = 4.875, 95% CI: 2.636-9.016, P < 0.001) were associated with GCs. CONCLUSIONS These findings underscore the importance of implementing targeted prevention strategies and early detection programs in high-risk populations to mitigate the burden of GPLs and GCs in Nanjing.
Collapse
Affiliation(s)
- Chunyan Niu
- Department of Gastroenterology, Nanjing Lishui People's Hospital (Zhongda Hospital Lishui Branch, Southeast University), Nanjing, 211200, China.
| | - Yongqiang Song
- Department of Gastroenterology, Nanjing Lishui People's Hospital (Zhongda Hospital Lishui Branch, Southeast University), Nanjing, 211200, China
| | - Yue Chen
- Department of Gastroenterology, Nanjing Lishui People's Hospital (Zhongda Hospital Lishui Branch, Southeast University), Nanjing, 211200, China
| | - Yongqiang Shi
- Department of Gastroenterology, Nanjing Lishui People's Hospital (Zhongda Hospital Lishui Branch, Southeast University), Nanjing, 211200, China
| | - Hui Wang
- Department of Gastroenterology, Nanjing Lishui People's Hospital (Zhongda Hospital Lishui Branch, Southeast University), Nanjing, 211200, China
| | - Xinguo Wu
- Department of Gastroenterology, Nanjing Lishui People's Hospital (Zhongda Hospital Lishui Branch, Southeast University), Nanjing, 211200, China
| | - Xiaoping Wang
- Department of Gastroenterology, Nanjing Lishui People's Hospital (Zhongda Hospital Lishui Branch, Southeast University), Nanjing, 211200, China
| | - Xiangyang Zhao
- Department of Gastroenterology, Nanjing Lishui People's Hospital (Zhongda Hospital Lishui Branch, Southeast University), Nanjing, 211200, China
| | - Yongdan Bu
- Dongping Street Health Center in Nanjing Lishui District, Nanjing, 211212, China
| | - Jijin Li
- Jingqiao Central Health Center in Nanjing Lishui District, Nanjing, 211224, China
| | - Tao Tao
- Honglan Street Health Center in Nanjing Lishui District, Nanjing, 211219, China
| | - Jinhua Wu
- Shiqiu Central Health Center in Nanjing Lishui District, Nanjing, 211222, China
| | - Changlin Xue
- Baima Central Health Center in Nanjing Lishui District, Nanjing, 211225, China
| | - Fuyu Zhang
- Yongyang Street Community Health Service Center in Nanjing Lishui District, Nanjing, 211299, China
| | - Chunrong Han
- Department of Pathology, Nanjing Lishui People's Hospital (Zhongda Hospital Lishui Branch, Southeast University), Nanjing, 211200, China
| | - Juan Yuan
- Department of Pathology, Nanjing Lishui People's Hospital (Zhongda Hospital Lishui Branch, Southeast University), Nanjing, 211200, China
| | - Qiang Zhang
- Department of Gastroenterology, Yancheng Third People's Hospital (The Yancheng School of Clinical Medicine of Nanjing Medical University), Yancheng, 224000, China.
| |
Collapse
|
2
|
Liu L, Xiong L, Peng H, Deng Q, Liu K, Xia S. BNC1 inhibits the development and progression of gastric cancer by regulating the CCL20/JAK-STAT axis. PeerJ 2025; 13:e19477. [PMID: 40444282 PMCID: PMC12121617 DOI: 10.7717/peerj.19477] [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: 12/19/2024] [Accepted: 04/25/2025] [Indexed: 06/02/2025] Open
Abstract
The role of basonuclin 1 (BNC1), a zinc finger protein-specific transcription factor, in gastric cancer remains unclear. In this study, BNC1 was downregulated in gastric cancer and functioned as a tumor suppressor. Through integrative analyses of transcriptome sequencing and functional assays, C-C motif chemokine ligand 20 (CCL20) was identified as a direct downstream target of BNC1. Overexpression of BNC1 inhibited the proliferation, migration, and invasion of gastric cancer cells both in vitro and in vivo. Mechanistically, BNC1 suppresses CCL20 expression by binding to its promoter, leading to reduced activation of the JAK-STAT signaling pathway and promoting apoptosis in gastric cancer cells. These findings highlight the pivotal role of BNC1 in gastric cancer progression and suggest that targeting BNC1 and its downstream pathways could serve as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Lixin Liu
- The Second Department of Gastrointestinal Surgery, The Affiliated Hospital of the North Sichuan Medical College, Nanchong, Sichuan, China
- Institute of Tissue Engineering and Stem Cell, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Li Xiong
- Department of Clinical Laboratory, People’s Hospital of Leshan, Leshan, Sichuan, China
| | - Hong Peng
- Department of Anorectal Surgery, Nanchong Central Hospital, The Second clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Qin Deng
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cell, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Shusen Xia
- The Second Department of Gastrointestinal Surgery, The Affiliated Hospital of the North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Gastrointestinal Surgery, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Hong Q, Lin W, Yan Y, Chen S, Li J, Yu J, Zhu Y, Qiu S. Huashi Jiedu Decoction Enhances 5-Fluorouracil Efficacy in Gastric Cancer via miRNA-21-3p/p53 Pathway. Drug Des Devel Ther 2025; 19:3883-3906. [PMID: 40391175 PMCID: PMC12087606 DOI: 10.2147/dddt.s513371] [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: 01/26/2025] [Accepted: 05/09/2025] [Indexed: 05/21/2025] Open
Abstract
Purpose To explore the mechanism of Huashi Jiedu Decoction (HJD) synergizing with 5-fluorouracil (5-Fu) in gastric cancer (GC) therapy. Methods MicroRNAs (miRNAs) and genes involved in HJD-mediated GC treatment were identified using ultra-high-performance liquid chromatography coupled with quadrupole-Orbitrap mass spectrometry, network pharmacology, Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and molecular docking. The effects of HJD on 5-Fu sensitivity in BGC-823 cells were evaluated with 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays, reverse transcription quantitative polymerase chain reaction (RT-qPCR), and Western blotting. Synergistic effects in vector-transfected and miRNA-21-3p knockdown cells were assessed by colony formation, wound healing, transwell assays, and flow cytometry (FCM). An in vivo study evaluated the impact of HJD on 5-Fu sensitivity, measuring miRNA-21-3p, tumor protein p53 (p53), N-cadherin, vimentin, and E-cadherin in tumors, along with tumor volume and weight. Results miRNA-21-3p and p53 were key targets in HJD's therapeutic effect on GC. RT-qPCR showed that HJD combined with 5-Fu reduced miRNA-21-3p and upregulated p53 in vector cells and increased p53 mRNA (p < 0.01) and protein (p < 0.05) compared to 5-Fu alone. These effects were abolished in miRNA-21-3p knockdown cells. The combination reduced colony formation by 48.92% (p < 0.01), suppressed transwell migration by 28.5% (p < 0.01), and inhibited wound healing by 81.9% compared to 5-Fu monotherapy (p < 0.001), with no effects in knockdown cells. FCM showed a 15.1% increase in G0/G1 phase arrest (p < 0.05). In vivo, the combination significantly reduced tumor volume (p < 0.05) and weight by 18.7%, with concomitant miRNA-21-3p downregulation (p < 0.0001), EMT marker suppression (N-cadherin, vimentin), and tumor suppressor activation (p53, E-cadherin) versus 5-Fu alone. Conclusion HJD enhances 5-Fu's effects on GC by regulating the miRNA-21-3p/p53 pathway and modulating cadherin expression, supporting its potential as an adjunctive treatment in GC.
Collapse
MESH Headings
- Stomach Neoplasms/drug therapy
- Stomach Neoplasms/pathology
- Stomach Neoplasms/metabolism
- MicroRNAs/metabolism
- MicroRNAs/genetics
- MicroRNAs/antagonists & inhibitors
- Fluorouracil/pharmacology
- Fluorouracil/administration & dosage
- Humans
- Tumor Suppressor Protein p53/metabolism
- Tumor Suppressor Protein p53/genetics
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/chemistry
- Drugs, Chinese Herbal/administration & dosage
- Cell Proliferation/drug effects
- Animals
- Drug Screening Assays, Antitumor
- Mice
- Mice, Nude
- Dose-Response Relationship, Drug
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/metabolism
- Mice, Inbred BALB C
- Cell Line, Tumor
- Antimetabolites, Antineoplastic/pharmacology
- Tumor Cells, Cultured
- Male
Collapse
Affiliation(s)
- Qianran Hong
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, People’s Republic of China
| | - Weiye Lin
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, People’s Republic of China
| | - Yici Yan
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, People’s Republic of China
| | - Shuangyu Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, People’s Republic of China
| | - Jiayang Li
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, People’s Republic of China
| | - Jieru Yu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People’s Republic of China
| | - Ying Zhu
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, People’s Republic of China
| | - Shengliang Qiu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, People’s Republic of China
| |
Collapse
|
4
|
Xu L, Li M, Dong X, Wang Z, Tong Y, Feng T, Xu S, Shang H, Zhao B, Lin J, Cao Z, Zheng Y. The value of deep learning and radiomics models in predicting preoperative serosal invasion in gastric cancer: a dual-center study. Abdom Radiol (NY) 2025:10.1007/s00261-025-04949-1. [PMID: 40285792 DOI: 10.1007/s00261-025-04949-1] [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: 02/18/2025] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025]
Abstract
PURPOSE To establish and validate a model based on deep learning (DL), integrating radiomic features with relevant clinical features to generate nomogram, for predicting preoperative serosal invasion in gastric cancer (GC). METHODS This retrospective study included 335 patients from dual centers. T staging (T1-3 or T4) was used to assess serosal invasion. Radiomic features were extracted from primary GC lesions in the venous phase CT, and DL features from 8 transfer learning models were combined to create the Hand-crafted Radiomics and Deep Learning Radiomics (HCR-DLR) model. The Clinical (CL) model was built using clinical features, and both were combined into the Clinical and Radiomics Combined (CRC) model. In total, 15 predictive models were developed using 5 machine learning algorithms. The best-performing models were visualized as nomograms. RESULTS The total of 14 radiomic features, 13 DL features, and 2 clinical features were considered valuable through dimensionality reduction and selection. Among the constructed models: CRC model (AUC, training cohort: 0.9212; internal test cohort: 0.8743; external test cohort: 0.8853) than HCR-DLR model (AUC, training cohort: 0.8607; internal test cohort: 0.8543; external test cohort: 0.8824) and CL model (AUC, training cohort: 0.7632; internal test cohort: 0.7219; external test cohort: 0.7294) showed better performance. A nomogram based on the logistic CL model was drawn to facilitate the usage and showed its excellent predictive performance. CONCLUSION The predictive performance of the CRC Model, which integrates clinical features, radiomic features, and DL features, exhibits robust predictive capability and can serve as a simple, non-invasive, and practical tool for predicting the serosal invasion status of GC.
Collapse
Affiliation(s)
- Lihang Xu
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Mingyu Li
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Xianling Dong
- Hebei International Research Center for Medical-Engineering, Chengde Medical University, Chengde, China
| | - Zhongxiao Wang
- Hebei International Research Center for Medical-Engineering, Chengde Medical University, Chengde, China
| | - Ying Tong
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Tao Feng
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Shuangyan Xu
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Hui Shang
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Bin Zhao
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Jianpeng Lin
- Hebei International Research Center for Medical-Engineering, Chengde Medical University, Chengde, China
| | - Zhendong Cao
- Radiology Department, Affiliated Hospital of Chengde Medical College, Chengde, China.
| | - Yi Zheng
- Radiology department, Chengde Central Hospital, Chengde, China.
| |
Collapse
|
5
|
Chen Y, Jia K, Xie Y, Yuan J, Liu D, Jiang L, Peng H, Zhong J, Li J, Zhang X, Shen L. The current landscape of gastric cancer and gastroesophageal junction cancer diagnosis and treatment in China: a comprehensive nationwide cohort analysis. J Hematol Oncol 2025; 18:42. [PMID: 40234884 PMCID: PMC12001465 DOI: 10.1186/s13045-025-01698-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/07/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Gastric cancer is the fifth most common cancer globally and is associated with significant morbidity and mortality. Despite its alarming prevalence, limited comparative evidence exists on its treatment efficacy and prognosis across diverse China populations. METHODS To address this, our study used a large-scale dataset from the National Cancer Information Database, including data from 220,304 patients from 53 leading hospitals across 27 provinces in China. RESULTS From 2017 to 2023, early-stage (Stages I-II) gastric cancer diagnoses increased to 35.63% of all cancer cases. Our study evaluated the neoadjuvant treatment strategies, adjuvant post-operative therapy, first- and second-line management for progressive stages, alongside current gastric cancer treatment guidelines in China. Notably, immunotherapy accounted for 16.17% and 23.28% of first- and second-line treatments for late-stage gastric cancers, and 14.56% and 5.00% for neoadjuvant and adjuvant therapies, respectively. Analysis of survival rates revealed that the 1-, 2-, 3-, 4-, and 5-year survival rates were 74.07%, 54.89%, 44.21%, 37.97%, and 33.53%, respectively. The 5-year survival rates across stages I-IV were 85.07%, 49.34%, 35.56%, and 13.15%, respectively. CONCLUSIONS These findings offer critical insights into the current state of gastric cancer treatment in China and can inform future initiatives to improve therapeutic outcomes for patients with gastric cancer.
Collapse
Affiliation(s)
- Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| | - Keren Jia
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yi Xie
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Jiajia Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Dan Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Lei Jiang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Haoxin Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | | | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
6
|
Liu Y, Chen Y, Shu J, Zhang Z, You Y, Yue S, Ji Q, Chen K, Liu Y, Duan B, Yu B, Kou S, Pang X, Wang W, Yang L, Zhao Z, Gao J. Dual-energy CT for predicting progression-free survival of locally advanced gastric cancer after gastrectomy: Insights into tumor angiogenesis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:110017. [PMID: 40222263 DOI: 10.1016/j.ejso.2025.110017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/12/2025] [Accepted: 04/03/2025] [Indexed: 04/15/2025]
Abstract
OBJECTIVES To investigate preoperative dual-energy CT (DECT)-derived independent risk factors affecting progression-free survival (PFS) in patients with locally advanced gastric cancer (LAGC) undergoing gastrectomy, and to reveal the underlying histopathologic changes. METHODS This prospective study included patients who underwent preoperative DECT scan and gastrectomy. Clinical data, DECT-derived morphological characteristics and iodine-related parameters were comprehensively collected. Univariate and multivariate analyses were carried out to identify independent risk factors associated with PFS. The prognostic performance of various parameters was evaluated using the bootstrap-based consistency index (C-index) and time-dependent receiver operating characteristic (ROC) analysis. Kaplan-Meier curves were used to assess the differences in survival analysis. The histopathologic underpinnings of the DECT-based combined parameter for evaluating PFS were explored. RESULTS 120 LAGC patients (63.3 ± 10.9 years; 94 men) were analyzed. Age, arterial enhancement fraction (AEF), serosal invasion, and tumor thickness were identified as preoperative independent risk factors affecting PFS (all p < 0.05). The combined parameters based on these risk factors achieved a C-index of 0.75, significantly or slightly superior to that of any single risk factor (all p < 0.05) or postoperative pathological staging (C-index, 0.67; p > 0.05). For predicting the 0.5-, 1- and 2-year PFS, the combined parameter had an area-under-the-curve (AUC) of 0.72, 0.77, and 0.74, respectively. PFS significantly differed between patients of high- and low-risks assessed with the combined parameter (p < 0.001). Histopathologically, the combined parameter was associated with tumor microvessel density (r = 0.31, p < 0.001). CONCLUSION The combination of DECT-derived morphological characteristics, iodine-related parameters, and clinical data helped accurately stratify PFS in LAGC before surgery and is associated with tumor angiogenesis. CLINICAL RELEVANCE STATEMENT Dual-energy CT was promising in the preoperative evaluation of the progression-free survival in LAGC patients after gastrectomy.
Collapse
Affiliation(s)
- Yiyang Liu
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Henan International Joint Laboratory of Medical Imaging, Zhengzhou, China; Henan Key Laboratory of Image Diagnosis and Treatment for Digestive System Tumor, Zhengzhou, China; Henan Key Laboratory of CT Imaging, Zhengzhou, China; The First Clinical School of Medicine, Zhengzhou University, Zhengzhou, 450052, China
| | - Yusong Chen
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Henan International Joint Laboratory of Medical Imaging, Zhengzhou, China; Henan Key Laboratory of Image Diagnosis and Treatment for Digestive System Tumor, Zhengzhou, China; Henan Key Laboratory of CT Imaging, Zhengzhou, China; The First Clinical School of Medicine, Zhengzhou University, Zhengzhou, 450052, China
| | - Jiao Shu
- The First Clinical School of Medicine, Zhengzhou University, Zhengzhou, 450052, China; Department of Pathology, The First Affiliated Hospital of Zhengzhou University, China
| | - Zhe Zhang
- The First Clinical School of Medicine, Zhengzhou University, Zhengzhou, 450052, China; Department of Pathology, The First Affiliated Hospital of Zhengzhou University, China
| | - Yaru You
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Henan International Joint Laboratory of Medical Imaging, Zhengzhou, China; Henan Key Laboratory of Image Diagnosis and Treatment for Digestive System Tumor, Zhengzhou, China; Henan Key Laboratory of CT Imaging, Zhengzhou, China; The First Clinical School of Medicine, Zhengzhou University, Zhengzhou, 450052, China
| | - Songwei Yue
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Henan International Joint Laboratory of Medical Imaging, Zhengzhou, China; Henan Key Laboratory of Image Diagnosis and Treatment for Digestive System Tumor, Zhengzhou, China; Henan Key Laboratory of CT Imaging, Zhengzhou, China
| | - Qingyu Ji
- Department of Radiology, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, 014030, China
| | - Kuisheng Chen
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, China
| | - Yao Liu
- Department of Pathology, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, China
| | - Bo Duan
- Department of Radiology, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, 014030, China
| | - Baiqing Yu
- Department of Medical Oncology, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, China
| | - Songzi Kou
- The First Clinical School of Medicine, Zhengzhou University, Zhengzhou, 450052, China; Department of Pathology, The First Affiliated Hospital of Zhengzhou University, China
| | - Xia Pang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, China
| | - Weitao Wang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Li Yang
- Department of Pathology, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, China.
| | - Zihao Zhao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Henan International Joint Laboratory of Medical Imaging, Zhengzhou, China; Henan Key Laboratory of Image Diagnosis and Treatment for Digestive System Tumor, Zhengzhou, China; Henan Key Laboratory of CT Imaging, Zhengzhou, China.
| | - Jianbo Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Henan International Joint Laboratory of Medical Imaging, Zhengzhou, China; Henan Key Laboratory of Image Diagnosis and Treatment for Digestive System Tumor, Zhengzhou, China; Henan Key Laboratory of CT Imaging, Zhengzhou, China; The First Clinical School of Medicine, Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
7
|
Qin Z, Ye F, Wang J, Jiang J, Zhang X, Li H, Feng L. BUB1B promotes cisplatin resistance in gastric cancer via Rad51-mediated DNA damage repair. Transl Oncol 2025; 54:102334. [PMID: 40056529 PMCID: PMC11930193 DOI: 10.1016/j.tranon.2025.102334] [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: 11/28/2024] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Cisplatin resistance significantly impedes the treatment of gastric cancer (GC). This work examined the possible therapeutic target status and function of BUB1B in controlling cisplatin resistance. METHODS Following the identification of differentially expressed genes (DEGs), protein-protein interaction (PPI) network analysis was conducted using datasets from the Cancer Genome Atlas-stomach adenocarcinoma (TCGA-STAD), GSE51575, and GSE79973. Functional tests assessed the effect of BUB1B overexpression and knockdown on the GC cells. Enrichment analysis and RNA-seq identified pathways linked to BUB1B. Additionally, the function of BUB1B in GC cells resistant to cisplatin in regulating DNA repair was examined, as its relationship with Rad51 inhibitor (B02) in regulating cell cycle, proliferation, and apoptosis. The combined effects of Rad51 suppression and BUB1B overexpression on tumor development in cisplatin-resistant GC cells were further validated in vivo xenograft models. RESULTS Significant overexpression of six critical overlapping genes was seen in GC tissues. The GC cell invasion, migration, and proliferation processes were improved by BUB1B overexpression, whereas BUB1B knockdown prevented these outcomes. Genes involved in DNA repair were downregulated by BUB1B knockdown, according to an RNA-seq study. BUB1B overexpression boosted cell survival via modulating cell cycle proteins, but BUB1B knockdown hampered DNA repair and increased death in cisplatin-resistant GC cells. Overexpression of BUB1B enhanced tumor development in vivo and counteracted the inhibitory effects of B02 on cell growth. CONCLUSION BUB1B enhances cisplatin resistance in gastric cancer by regulating DNA repair and cell cycle progression, suggesting that targeting BUB1B may be a feasible therapeutic strategy.
Collapse
Affiliation(s)
- Zhe Qin
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Fangzhou Ye
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Jiayi Wang
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Jun Jiang
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Xiaohong Zhang
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Huanqing Li
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Li Feng
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China.
| |
Collapse
|
8
|
Li R, Li J, Wang Y, Liu X, Xu W, Sun R, Xue B, Zhang X, Ai Y, Du Y, Jiang J. The artificial intelligence revolution in gastric cancer management: clinical applications. Cancer Cell Int 2025; 25:111. [PMID: 40119433 PMCID: PMC11929235 DOI: 10.1186/s12935-025-03756-4] [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: 08/27/2024] [Accepted: 03/18/2025] [Indexed: 03/24/2025] Open
Abstract
Nowadays, gastric cancer has become a significant issue in the global cancer burden, and its impact cannot be ignored. The rapid development of artificial intelligence technology is attempting to address this situation, aiming to change the clinical management landscape of gastric cancer fundamentally. In this transformative change, machine learning and deep learning, as two core technologies, play a pivotal role, bringing unprecedented innovations and breakthroughs in the diagnosis, treatment, and prognosis evaluation of gastric cancer. This article comprehensively reviews the latest research status and application of artificial intelligence algorithms in gastric cancer, covering multiple dimensions such as image recognition, pathological analysis, personalized treatment, and prognosis risk assessment. These applications not only significantly improve the sensitivity of gastric cancer risk monitoring, the accuracy of diagnosis, and the precision of survival prognosis but also provide robust data support and a scientific basis for clinical decision-making. The integration of artificial intelligence, from optimizing the diagnosis process and enhancing diagnostic efficiency to promoting the practice of precision medicine, demonstrates its promising prospects for reshaping the treatment model of gastric cancer. Although most of the current AI-based models have not been widely used in clinical practice, with the continuous deepening and expansion of precision medicine, we have reason to believe that a new era of AI-driven gastric cancer care is approaching.
Collapse
Affiliation(s)
- Runze Li
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Jingfan Li
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Yuman Wang
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Xiaoyu Liu
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Weichao Xu
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China
| | - Runxue Sun
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China
| | - Binqing Xue
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Xinqian Zhang
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Yikun Ai
- North China University of Science and Technology, Tanshan 063000, China
| | - Yanru Du
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China.
- Hebei Provincial Key Laboratory of Integrated Traditional and Western Medicine Research on Gastroenterology, Hebei, 050011, China.
- Hebei Key Laboratory of Turbidity and Toxicology, Hebei, 050011, China.
| | - Jianming Jiang
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China.
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China.
| |
Collapse
|
9
|
Chen S, Han X, Lu Y, Wang S, Fang Y, Leng C, Sun X, Li X, Qiu W, Qi W. A prognostic model based on autophagy-and senescence-related genes for gastric cancer: implications for immunotherapy and personalized treatment. Front Oncol 2025; 15:1509771. [PMID: 40182050 PMCID: PMC11965130 DOI: 10.3389/fonc.2025.1509771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
Background The process of human aging is accompanied by an increased susceptibility to various cancers, including gastric cancer. This heightened susceptibility is linked to the shared molecular characteristics between aging and tumorigenesis. Autophagy is considered a critical mediator connecting aging and cancer, exerting a dynamic regulatory effect in conjunction with cellular senescence during tumor progression. In this study, a combined analysis of autophagy- and senescence-related genes was employed to comprehensively capture tumor heterogeneity. Methods The gene expression profiles and clinical data for GC samples were acquired from TCGA and GEO databases. Differentially expressed autophagy- and senescence-related genes (DEASRGs) were identified between tumor and normal tissues. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were carried out to provide insights into biological significance. A prognostic signature was established using univariate Cox and LASSO regression analyses. Furthermore, consensus clustering analyses and nomograms were employed for survival prediction. TME and drug sensitivity analyses were conducted to compare differences between the groups. To predict immunotherapy efficacy, the correlations between risk score and immune checkpoints, MSI, TMB, and TIDE scores were investigated. Results A fourteen-gene prognostic signature with superior accuracy was constructed. GC patients were stratified into three distinct clusters, each exhibiting significant variations in their prognosis and immune microenvironments. Drug sensitivity analysis revealed that the low-risk group demonstrated greater responsiveness to several commonly used chemotherapeutic agents for gastric cancer, including oxaliplatin. TME analysis further indicated that the high-risk group exhibited increased immune cell infiltration, upregulated expression of ICs, and a higher stromal score, suggesting a greater capacity for immune evasion. In contrast, the low-risk group was characterized by a higher proportion of microsatellite instability-high (MSI-H) cases, an elevated TIDE score, and a greater TMB, indicating a higher likelihood of benefiting from immunotherapy. In addition, Single-cell sequencing demonstrated that TXNIP was expressed in epithelial cells. Cellular experiments preliminarily verified that TXNIP could promote the proliferation and migration of gastric cancer cells. Conclusion This study presents a robust predictive model for GC prognosis using autophagy- and senescence-related genes, demonstrating its ability to predict immune infiltration, immunotherapy effectiveness, and guide personalized treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Wensheng Qiu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Weiwei Qi
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
10
|
Liu Z, Liu A, Li M, Xiang J, Yu G, Sun P. Efficacy and safety of sintilimab combined with trastuzumab and chemotherapy in HER2-positive advanced gastric or gastroesophageal junction cancer. Front Immunol 2025; 16:1545304. [PMID: 40028325 PMCID: PMC11867958 DOI: 10.3389/fimmu.2025.1545304] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Background To evaluate the efficacy and safety of sintilimab in combination with trastuzumab and chemotherapy for HER2-positive advanced gastric/gastroesophageal junction cancer (GC/GEJC). Methods HER2-positive advanced GC/GEJC patients admitted to our department between January 2018 and October 2024 were included in this study. Patients who received sintilimab in combination with trastuzumab and chemotherapy were assigned to cohort A, while patients who received trastuzumab and chemotherapy alone were assigned to cohort B. The primary endpoints were progression-free survival (PFS) and overall survival (OS), while the secondary endpoints included disease control rate (DCR), objective response rate (ORR), and safety. Results A total of 103 patients were analyzed, with 46 in cohort A and 57 in cohort B. The ORR was 65.2% in cohort A compared to 40.4% in cohort B, while the DCR was 87.0% in cohort A and 70.2% in cohort B. The median follow-up duration was 14.0 months. Median PFS (mPFS) was 9.4 months (95% CI: 5.6-13.2) for cohort A and 7.4 months (95% CI: 6.1-8.7) for cohort B (p = 0.089). Median OS (mOS) was 16.4 months (95% CI: 11.5-21.3) in cohort A versus 14.2 months (95% CI: 11.2-17.2) in cohort B (p = 0.069). Adverse events were predominantly mild, and no treatment-related deaths occurred. Conclusion Sintilimab combined with trastuzumab and chemotherapy showed promising efficacy and acceptable safety in HER2-positive advanced GC/GEJC. However, no statistically significant improvement in survival outcomes was observed compared to trastuzumab and chemotherapy alone.
Collapse
Affiliation(s)
- Zeyu Liu
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Aina Liu
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Ming Li
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Jinyu Xiang
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Guohua Yu
- Department of Pathology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Ping Sun
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| |
Collapse
|
11
|
Liu ZC, Li WQ. Large-scale cluster randomised trial reveals effectiveness of Helicobacter pylori eradication for gastric cancer prevention. Clin Transl Med 2025; 15:e70229. [PMID: 39936539 DOI: 10.1002/ctm2.70229] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 02/02/2025] [Indexed: 02/13/2025] Open
Affiliation(s)
- Zong-Chao Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Haidian District, Beijing, China
| | - Wen-Qing Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Haidian District, Beijing, China
| |
Collapse
|
12
|
Liu J, Gan T, Hu W, Li Y. Does ambient particulate matter 1 increase the risk of gastric cancer in the northwest of China? Int J Cancer 2025; 156:104-113. [PMID: 39177481 DOI: 10.1002/ijc.35144] [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: 03/31/2024] [Revised: 07/21/2024] [Accepted: 08/06/2024] [Indexed: 08/24/2024]
Abstract
Gastric cancer (GC) remains a significant health concern in Gansu province, China, with morbidity and mortality rates surpassing national averages. Despite the recognized health risks associated with ambient particulate matter with an aerodynamic diameter <1 μm (PM1), the relationship between PM1 exposure and GC incidence has not been extensively studied. Data on GC cases from 2013 to 2021 were gathered from 262 hospitals in Gansu, China. Concurrently, data on the normalized vegetation index (NDVI), gross domestic product (GDP), drinking and smoking behavioral index (DSBI), PM1, PM2.5, and PM2.5-1 were collected. Utilizing a Bayesian conditional autoregressive (CAR) combined generalized linear model (GLM) with quasi-Poisson regression, we evaluated the impact of PM1, PM2.5, PM2.5-1, NDVI, DSBI, and GDP on GC morbidity while adjusting for potential confounders. Our analysis indicated that exposure to PM1 (μg/m3) is significantly positively correlated with GC incidence in regions with an overall age-standardized incidence rate (ASIR) >40 (relative risks [RR]: 1.023, 95% confidence intervals [CI, 1.007, 1.039]), male ASIR >50 (RR: 1.014, 95% CI [1.009, 1.019]), and female ASIR >20 (RR: 1.010, 95% CI [1.002, 1.018]). PM2.5, PM2.5-1, DSBI, and GDP were positively correlated with GC incidence, while NDVI was negatively correlated in the study regions. Our findings provided evidence of a positive correlation between PM1 exposure and GC incidence in high-risk areas of GC within arid regions. Further research is warranted to elucidate the complex nonlinear relationships between environmental factors and GC. These insights could inform strategies for improving the control and prevention of GC in Gansu and similar regions.
Collapse
Affiliation(s)
- Jie Liu
- Department of Tumor Surgery, General Surgery, Gansu Provincial Key Laboratory of Gastrointestinal Cancer, Key Laboratory of Environmental Oncology in Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
- Center for Digestive System Tumor Transformation and Innovation Engineering, Environmental Oncology Center, Lanzhou University, Lanzhou, China
- Ecosystem Change and Population Health Research Group, School of Public Health and Social Work, The Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Ting Gan
- Ecosystem Change and Population Health Research Group, School of Public Health and Social Work, The Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Wenbiao Hu
- Ecosystem Change and Population Health Research Group, School of Public Health and Social Work, The Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Yumin Li
- Department of Tumor Surgery, General Surgery, Gansu Provincial Key Laboratory of Gastrointestinal Cancer, Key Laboratory of Environmental Oncology in Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
- Center for Digestive System Tumor Transformation and Innovation Engineering, Environmental Oncology Center, Lanzhou University, Lanzhou, China
| |
Collapse
|
13
|
Zhu M, Zhou H, Zhuo Y, Liu C, Li J, He P, Liu N, Zhao Z, Huafeng P. Pan-cancer multi-omics analysis to identify the potential pro-oncogenic properties of GREM1 as a promising targets for cancer prognosis and therapeutics. Int J Immunopathol Pharmacol 2025; 39:3946320251331850. [PMID: 40231657 PMCID: PMC12033649 DOI: 10.1177/03946320251331850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 03/16/2025] [Indexed: 04/16/2025] Open
Abstract
We aimed to investigate the potential pro-oncogenic properties of GREM1 by Pan-cancer multi-omics analysis. Accumulating evidence has highlighted that GREM1 (Gremlin 1), serves as an inhibitor of BMP (Bone Morphogenetic Protein) family, involve in bone related diseases, carcinogenesis, cell stemness, and cell differentiation. However, the effect and underlying mechanism of GREM1 on the cancer biology remain largely elusive. The mRNA expression of GREM1 were extracted from GTEx (Genotype-Tissue Expression) and TCGA (The Cancer Genome Atlas) database. Analysis of OS (Overall Survival), PFI (Progression Free Interval), DSS (Disease-Specific Survival), and ROC (Receiver Operating Characteristic) were performed to predicted prognostic value of GREM1 in various cancers. The TIMER (Tumor Immune Estimation Resource) online tool was used to investigate the relationship between GREM1 transcriptional level and infiltration of immune cells. KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis and GO (Gene Ontology) analysis were used to investigate the GREM1 related molecular events, and then constructed a PPI (Protein-Protein Interaction) network via the STRING (Search Tool for the Retrieval of Interaction Genes/Proteins) online tool. Western blot was performed to investigate the indicated protein expression. In the present study, our results showed that GREM1 tended to be upregulated in various cancers, which would correlate with the poor prognosis. Mechanistically, our results showed that GREM1 involve in regulating the ECM-receptor interaction pathway, upregulation of MMP activity, angiogenesis, and immune cell infiltration. In vitro studies, our results further showed that BMP agonist significantly decreased the protein level of GREM1 in GES-1 cells and BGC cells, which accompanied by inhibiting migration and proliferation in GES-1 cells and BGC cells. BMP inhibitor significantly promoted GREM1 expression and migration in BGC cells, but not GES-1 cells. GREM1 might serve as a potential and promising prognostic biomarker for drug development and cancer treatment.
Collapse
Affiliation(s)
- Menglu Zhu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hengli Zhou
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yue Zhuo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Changhua Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaxin Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peiyao He
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Naihua Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziming Zhao
- Guangdong Province Engineering Technology Research Institute of T.C.M, Guangzhou, China
| | - Pan Huafeng
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
14
|
Zhang T, Lv H, Li J, Zhang S, Zhang J, Wang S, Wang Y, Guo Z. The impact of immune-related adverse events on the outcome of advanced gastric cancer patients with immune checkpoint inhibitor treatment. Front Immunol 2024; 15:1503316. [PMID: 39776906 PMCID: PMC11703953 DOI: 10.3389/fimmu.2024.1503316] [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: 09/28/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Background The occurrence of immune-related adverse events (irAEs) seemed to be associated with better outcomes in advanced gastric cancer (AGC) patients. However, research focusing on the impact of the single-organ irAE (uni-irAE) or multi-organ irAEs (multi-irAEs) on the AGC outcome is relatively limited. In this study, we investigated individually the impact of the different irAEs on AGC survival as well as the co-occurrence patterns of multi-irAEs. Methods The uni-irAE, multi-irAEs, and non-irAE were identified based on National Comprehensive Cancer Network (NCCN) guidelines. ICI efficacy for the disease control rate (DCR) and the objective response rate (ORR) was assessed based on the Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1. The association for the irAEs with progression-free survival (PFS) or overall survival (OS) was analyzed using the Kaplan-Meier method and Cox regression model. We also performed pairwise correlation analysis to identify co-occurrence patterns of multi-organ irAEs. Results A total of 288 patients including 175 non-irAE, 73 uni-irAE, and 40 multi-irAE patients were evaluated for their association with AGC outcome. The irAEs patients displayed higher DCR (78.8% vs. 67.4%, p=0.037) when compared with those of non-irAE patients, and both uni-irAE patients (82.2% vs. 67.4%, p=0.019) and multi-irAE patients (72.5% vs. 67.4%, p=0.534) showed higher DCR than that of non-irAE patients. The multivariate analyses revealed that multi-irAEs was an independent risk factor for PFS (hazard ratio [HR] of 0.63, 95% confidence interval [CI] 0.41~0.96, p=0.031) and OS (HR 0.47, 95% CI 0.29~0.76, p=0.002), whereas the survival association for uni-irAE was not obtained. The analysis of the co-occurrence patterns for multi-irAEs revealed that the thyroid, adrenal gland, heart, skin, and lung irAEs exhibited a high risk of co-occurrence of multi-irAEs. The multivariate Cox regression analysis for organ-specific irAEs revealed that patients experiencing thyroid, adrenal gland, and skin irAEs had favorable survival outcomes compared with those without these irAEs. Conclusion Multi-irAEs and some organ-specific irAEs can be used as predictive indicators for ICI treatment efficacy in AGC patients. The thyroid, adrenal gland, heart, skin, and lung irAEs are often accompanied by multi-irAE occurrence.
Collapse
Affiliation(s)
- Tianhang Zhang
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Haitao Lv
- Department of Hepatobiliary Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiasong Li
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shasha Zhang
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jingjing Zhang
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Siqi Wang
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yingnan Wang
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Gastroenterology and Hepatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhanjun Guo
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
15
|
Cai X, Cao M, Yang Q, Yu X, Feng XH, Zhao RY. HER2-targeted ADC DX126-262 combined with chemotherapy demonstrates superior antitumor efficacy in HER2-positive gastric cancer. Am J Cancer Res 2024; 14:5752-5768. [PMID: 39803638 PMCID: PMC11711518 DOI: 10.62347/qcdr9612] [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: 09/18/2024] [Accepted: 11/14/2024] [Indexed: 01/16/2025] Open
Abstract
Gastric cancer is a common malignant tumor with high incidence and mortality. The overexpression of Human epidermal growth factor receptor 2 (HER2) is associated with increased metastatic potential and poor clinical outcome in gastric cancer. Despite the proven clinical response rates of approved HER2-targeted therapies, including Trastuzumab combined with chemotherapy, their limited long-term clinical benefits and inevitable disease progression still pose significant challenges to the clinical treatment of gastric cancer. Hence, exploring novel strategies to enhance therapeutic outcomes for HER2-positive patients is extremely crucial and urgent. Here, we reported that DX126-262, a novel HER2-targeted antibody-drug conjugate, generated by conjugating a potent Tubulysin B analogue (Tub-114) to humanized anti-HER2 monoclonal antibody, exhibited a significant synergistic inhibitory effect with both Cisplatin and 5-FU in HER2-positive gastric cancer NCI-N87 cells. Moreover, the triple-drug combination strategy of DX126-262 combined with Cisplatin and 5-FU showed much better in vitro and in vivo therapeutic efficacy than monotherapy or double-drug combination (Cisplatin plus 5-FU) or first-line standard-of-care (SOC, Herceptin plus Cisplatin and 5-FU), and comparable or even superior in vivo efficacy than third-line SOC (DS-8201a) in NCI-N87 cells and xenograft models. Meanwhile, the triple-drug combination therapy did not exhibit superimposed toxicity. Taken together, our findings provide compelling evidence that DX126-262 in combination with Cisplatin and 5-FU exerts synergistic antitumor activity and is a promising strategy to improve the clinical efficacy of HER2-positive advanced or metastatic gastric cancer.
Collapse
Affiliation(s)
- Xiaobo Cai
- Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang UniversityHangzhou 310058, Zhejiang, China
- Hangzhou DAC Biotechnology Co., Ltd.No. 369 Qiaoxin Road, Qiantang District, Hangzhou 310018, Zhejiang, China
| | - Min Cao
- Hangzhou DAC Biotechnology Co., Ltd.No. 369 Qiaoxin Road, Qiantang District, Hangzhou 310018, Zhejiang, China
| | - Qingliang Yang
- Hangzhou DAC Biotechnology Co., Ltd.No. 369 Qiaoxin Road, Qiantang District, Hangzhou 310018, Zhejiang, China
| | - Xiazhen Yu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhou 310003, Zhejiang, China
| | - Xin-Hua Feng
- Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang UniversityHangzhou 310058, Zhejiang, China
| | - Robert Yongxin Zhao
- Hangzhou DAC Biotechnology Co., Ltd.No. 369 Qiaoxin Road, Qiantang District, Hangzhou 310018, Zhejiang, China
| |
Collapse
|
16
|
Xiong X, Huo Z, Zhou Y, Bishai DM, Grépin KA, Clarke PM, Chen C, Luo L, Quan J. Economic costs attributable to modifiable risk factors: an analysis of 24 million urban residents in China. BMC Med 2024; 22:549. [PMID: 39574122 PMCID: PMC11580671 DOI: 10.1186/s12916-024-03772-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/13/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Estimating the economic burden of modifiable risk factors is crucial for allocating scarce healthcare resources to improve population health. We quantified the economic burden attributable to modifiable risk factors in an urban area of China. METHODS Our Shanghai Municipal Health Commission dataset covered 2.2 million inpatient admissions for adults (age ≥ 20) in public and private hospitals in 2015 (1,327,187 admissions) and 2020 (837,482 admissions). We used a prevalence-based cost-of-illness approach by applying population attributable fraction (PAF) estimates for each modifiable risk factor from the Global Burden of Diseases Study (GBD) to estimate attributable costs. We adopted a societal perspective for cost estimates, comprising direct healthcare costs and productivity losses from absenteeism and premature mortality. Future costs were discounted at 3% and adjusted to 2020 prices. RESULTS In 2020, the total societal cost attributable to modifiable risk factors in Shanghai was US$7.9 billion (95% uncertainty interval [UI]: 4.6-12.4b), mostly from productivity losses (67.9%). Two health conditions constituted most of the attributable societal cost: cancer (51.6% [30.2-60.2]) and cardiovascular disease (31.2% [24.6-50.7]). Three modifiable risk factors accounted for half of the total attributable societal cost: tobacco (23.7% [16.4-30.5]), alcohol (13.3% [8.2-19.7]), and dietary risks (12.2% [7.5-17.7]). The economic burden varied by age and sex; most of the societal costs were from males (77.7%), primarily driven by their tobacco and alcohol use. The largest contributor to societal costs was alcohol for age 20-44, and tobacco for age 45 + . Despite the COVID-19 pandemic, the pattern of major modifiable risk factors remained stable from 2015 to 2020 albeit with notable increases in attributable healthcare costs from cancers and productivity losses from cardiovascular diseases. CONCLUSIONS The substantial economic burden of diseases attributable to modifiable risk factors necessitates targeted policy interventions. Priority areas are reducing tobacco and alcohol consumption and improving dietary habits that together constitute half of the total attributable costs. Tailored interventions targeting specific age and sex groups are crucial; namely tobacco in middle-aged/older males and alcohol in younger males.
Collapse
Affiliation(s)
- Xuechen Xiong
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Zhaohua Huo
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yinan Zhou
- School of Public Health, Fudan University, Shanghai, China
| | - David M Bishai
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Karen A Grépin
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Philip M Clarke
- Health Economics Research Centre, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Cynthia Chen
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Li Luo
- School of Public Health, Fudan University, Shanghai, China
| | - Jianchao Quan
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
- HKU Business School, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
17
|
Shi C, Tao R, Wang W, Tang J, Dou Z, Yuan X, Xu G, Liu H, Chen X. Development and validation of a nomogram for obesity and related factors to detect gastric precancerous lesions in the Chinese population: a retrospective cohort study. Front Oncol 2024; 14:1419845. [PMID: 39634264 PMCID: PMC11614725 DOI: 10.3389/fonc.2024.1419845] [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: 05/04/2024] [Accepted: 10/30/2024] [Indexed: 12/07/2024] Open
Abstract
Objectives The purpose of this study was to construct a nomogram to identify patients at high risk of gastric precancerous lesions (GPLs). This identification will facilitate early diagnosis and treatment and ultimately reduce the incidence and mortality of gastric cancer. Methods In this single-center retrospective cohort study, 563 participants were divided into a gastric precancerous lesion (GPL) group (n=322) and a non-atrophic gastritis (NAG) group (n=241) based on gastroscopy and pathology results. Laboratory data and demographic data were collected. A derivation cohort (n=395) was used to identify the factors associated with GPLs to develop a predictive model. Then, internal validation was performed (n=168). We used the area under the receiver operating characteristic curve (AUC) to determine the discriminative ability of the predictive model; we constructed a calibration plot to evaluate the accuracy of the predictive model; and we performed decision curve analysis (DCA) to assess the clinical practicability predictive model. Results Four -predictors (i.e., age, body mass index, smoking status, and -triglycerides) were included in the predictive model. The AUC values of this predictive model were 0.715 (95% CI: 0.665-0.765) and 0.717 (95% CI: 0.640-0.795) in the derivation and internal validation cohorts, respectively. These values indicated that the predictive model had good discrimination ability. The calibration plots and DCA suggested that the predictive model had good accuracy and clinical net benefit. The Hosmer-Lemeshow test results in the derivation and validation cohorts for this predictive model were 0.774 and 0.468, respectively. Conclusion The nomogram constructed herein demonstrated good performance in terms of predicting the risk of GPLs. This nomogram can be beneficial for the early detection of patients at high risk of GPLs, thus facilitating early treatment and ultimately reducing the incidence and mortality of gastric cancer.
Collapse
Affiliation(s)
- Chang’e Shi
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Gastroenterology, Anhui Public Health Clinical Center, Hefei, China
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University North District, Hefei, China
| | - Rui Tao
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, China
- Department of Psychiatry, School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China
- Department of Psychiatry, Anhui Psychiatric Center, Hefei, China
| | - Wensheng Wang
- Department of Gastroenterology, Anhui Public Health Clinical Center, Hefei, China
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University North District, Hefei, China
| | - Jinzhi Tang
- Department of Gastroenterology, Anhui Public Health Clinical Center, Hefei, China
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University North District, Hefei, China
| | - Zhengli Dou
- Department of Gastroenterology, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Xiaoping Yuan
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, China
- Department of Psychiatry, School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China
- Department of Psychiatry, Anhui Psychiatric Center, Hefei, China
| | - Guodong Xu
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, China
- Department of Psychiatry, School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China
- Department of Psychiatry, Anhui Psychiatric Center, Hefei, China
| | - Huanzhong Liu
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, China
- Department of Psychiatry, School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China
- Department of Psychiatry, Anhui Psychiatric Center, Hefei, China
- Department of Psychiatry, Huizhou NO.2 Hospital, Huizhou, China
| | - Xi Chen
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
18
|
Zhang H, Liu Y, Feng L, Wang L, Han J, Zhang X, Wang Y, Li D, Liu J, Liu Y, Jin H, Fan Z. Blood lipid profiles associated with metastatic sites in advanced gastric cancer. BMC Gastroenterol 2024; 24:391. [PMID: 39497073 PMCID: PMC11533295 DOI: 10.1186/s12876-024-03479-2] [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: 05/24/2024] [Accepted: 10/24/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND This study explored the correlation between peripheral blood lipid levels and clinicopathological parameters in patients with advanced gastric cancer (GC), focusing on changes in lipid levels during disease progression. METHODS Pathological features and serum lipid profiles of 179 patients with stage III-IV gastric adenocarcinoma were analyzed. Lipid parameters examined included total cholesterol (TC), triglycerides (TG), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), very low-density lipoprotein cholesterol (VLDL-C), apolipoprotein AI (Apo AI), apolipoprotein B (Apo B), lipoprotein(a) (Lp(a)), among others. The total cholesterol-lymphocyte score (TL score) and BMI were also calculated. The association between lipid parameters and clinicopathological characteristics such as age, gender, family history, and metastasis sites was assessed. RESULTS In GC patients, females had higher TG levels than males. Patients with peritoneal metastasis had significantly lower levels of TC, LDL-C, Apo B, and B/A ratio. Those with lung metastasis exhibited higher LDL-C levels and lower levels of VLDL-C. No significant associations were found between lipid levels and metastasis to distant lymph nodes, liver, or bone. Female patients with ovarian metastasis had significantly lower VLDL-C levels. Multivariate analysis revealed low TC as an independent risk factor for peritoneal metastasis, high LDL-C and low VLDL-C levels for lung metastasis, and younger age and low VLDL-C for ovarian metastasis. CONCLUSION Specific blood lipid levels are significantly associated with metastatic sites in advanced gastric cancer. Lipid profiles could serve as potential biomarkers for predicting metastatic sites in GC patients.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yiming Liu
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Li Feng
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Long Wang
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jing Han
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xue Zhang
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yudong Wang
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Dan Li
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jiayin Liu
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yan Liu
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hui Jin
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhisong Fan
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
19
|
Gao MJ, Li SB, Zhu XJ, Zhang LF, Chen M, Shi YQ. Analysis of Risk Factors for Gastric Cancer and Precancerous Lesions: A Case-Control Study. J Dig Dis 2024; 25:674-684. [PMID: 39865979 PMCID: PMC11877994 DOI: 10.1111/1751-2980.13326] [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: 04/15/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/28/2025]
Abstract
OBJECTIVES To investigate the correlation between gastric xanthoma (GX) and precancerous lesions (PCL) and gastric cancer (GC), and to explore other potential risk factors for PCL and GC in northwest China. METHODS A case-control study was conducted from September 2022 to September 2023 at Xijing Hospital, Air Force Medical University (Xi'an, Shaanxi Province, China). The patients who underwent gastroscopy were enrolled and divided into the chronic gastritis (CG) group (n = 423), PCL group (n = 422), and GC group (n = 415). The variables were selected through univariate analysis, including demographic information, dietary habits, lifestyle, gastroscopic findings, and Helicobacter pylori (H. pylori) infection. Multivariate logistic regression analysis was performed to analyze the factors associated with PCL and GC, and odds ratio (OR) and 95% confidence interval (CI) were calculated. RESULTS GX was more prevalent in the PCL group (14.93%) and the GC group (19.76%) than in the CG group (6.15%). Multivariate analysis revealed that age ≥ 50 years, male gender, rural residence, H. pylori infection, family history of GC, GX, and hypertension were independent risk factors for GC and PCL. Furthermore, a diet high in salt and spice, coupled with daily intake of less than 100 g of fresh fruits, might be associated with the occurrence of GC. CONCLUSION Age ≥ 50 years, male gender, rural residence, family history of GC and H. pylori infection, presence of GX, and a history of hypertension may be risk factors for PCL and GC.
Collapse
Affiliation(s)
- Meng Jie Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive DiseasesXijing Hospital, Air Force Medical UniversityXi'an, Shaanxi ProvinceChina
- Graduate Department, Xi'an Medical UniversityXi'an, Shaanxi ProvinceChina
| | - Song Bo Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive DiseasesXijing Hospital, Air Force Medical UniversityXi'an, Shaanxi ProvinceChina
| | - Xiao Jing Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive DiseasesXijing Hospital, Air Force Medical UniversityXi'an, Shaanxi ProvinceChina
| | - Li Feng Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive DiseasesXijing Hospital, Air Force Medical UniversityXi'an, Shaanxi ProvinceChina
| | - Min Chen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive DiseasesXijing Hospital, Air Force Medical UniversityXi'an, Shaanxi ProvinceChina
| | - Yong Quan Shi
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive DiseasesXijing Hospital, Air Force Medical UniversityXi'an, Shaanxi ProvinceChina
- Graduate Department, Xi'an Medical UniversityXi'an, Shaanxi ProvinceChina
| |
Collapse
|
20
|
Zhang YT, Zhao LJ, Zhou T, Zhao JY, Geng YP, Zhang QR, Sun PC, Chen WC. The lncRNA CADM2-AS1 promotes gastric cancer metastasis by binding with miR-5047 and activating NOTCH4 translation. Front Pharmacol 2024; 15:1439497. [PMID: 39309008 PMCID: PMC11412803 DOI: 10.3389/fphar.2024.1439497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
Background Multi-organ metastasis has been the main cause of death in patients with Gastric cancer (GC). The prognosis for patients with metastasized GC is still very poor. Long noncoding RNAs (lncRNAs) always been reported to be closely related to cancer metastasis. Methods In this paper, the aberrantly expressed lncRNA CADM2-AS1 was identified by lncRNA-sequencing in clinical lymph node metastatic GC tissues. Besides, the role of lncRNA CADM2-AS1 in cancer metastasis was detected by Transwell, Wound healing, Western Blot or other assays in vitro and in vivo. Further mechanism study was performed by RNA FISH, Dual-luciferase reporter assay and RT-qPCR. Finally, the relationship among lncRNA CADM2-AS1, miR-5047 and NOTCH4 in patient tissues was detected by RT-qPCR. Results In this paper, the aberrantly expressed lncRNA CADM2-AS1 was identified by lncRNA-sequencing in clinical lymph node metastatic GC tissues. Besides, the role of lncRNA CADM2-AS1 in cancer metastasis was detected in vitro and in vivo. The results shown that overexpression of the lncRNA CADM2-AS1 promoted GC metastasis, while knockdown inhibited it. Further mechanism study proved that lncRNA CADM2-AS1 could sponge and silence miR-5047, which targeting mRNA was NOTCH4. Elevated expression of lncRNA CADM2-AS1 facilitate GC metastasis by up-regulating NOTCH4 mRNA level consequently. What's more, the relationship among lncRNA CADM2-AS1, miR-5047 and NOTCH4 was further detected and verified in metastatic GC patient tissues. Conclusions LncRNA CADM2-AS1 promoted metastasis in GC by targeting the miR-5047/NOTCH4 signaling axis, which may be a potential target for GC metastasis.
Collapse
Affiliation(s)
- Yu-Tong Zhang
- Department of Gastrointestinal Surgery, Henan Provincial People’s Hospital, Henan University People’s Hospital, Zhengzhou University People’s Hospital, Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Li-Juan Zhao
- Department of Gastrointestinal Surgery, Henan Provincial People’s Hospital, Henan University People’s Hospital, Zhengzhou University People’s Hospital, Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, China
| | - Teng Zhou
- Department of Gastrointestinal Surgery, Henan Provincial People’s Hospital, Henan University People’s Hospital, Zhengzhou University People’s Hospital, Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jin-Yuan Zhao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yin-Ping Geng
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qiu-Rong Zhang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Pei-Chun Sun
- Department of Gastrointestinal Surgery, Henan Provincial People’s Hospital, Henan University People’s Hospital, Zhengzhou University People’s Hospital, Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, China
| | - Wen-Chao Chen
- Department of Gastrointestinal Surgery, Henan Provincial People’s Hospital, Henan University People’s Hospital, Zhengzhou University People’s Hospital, Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
21
|
Lei L, Zhao LY, Cheng R, Zhang H, Xia M, Chen XL, Kudriashov V, Liu K, Zhang WH, Jiang H, Chen Y, Zhu L, Zhou H, Yang K, Hu T, Hu JK. Distinct oral-associated gastric microbiota and Helicobacter pylori communities for spatial microbial heterogeneity in gastric cancer. mSystems 2024; 9:e0008924. [PMID: 38940519 PMCID: PMC11265414 DOI: 10.1128/msystems.00089-24] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 06/03/2024] [Indexed: 06/29/2024] Open
Abstract
The gastric microbial community plays a fundamental role in gastric cancer (GC), and the two main anatomical subtypes of GC, non-cardia and cardia GC, are associated with different risk factors (Helicobacter pylori for non-cardia GC). To decipher the different microbial spatial communities of GC, we performed a multicenter retrospective analysis to characterize the gastric microbiota in 223 GC patients, including H. pylori-positive or -negative patients, with tumors and paired adjacent normal tissues, using third-generation sequencing. In the independent validation cohort, both dental plaque and GC tumoral tissue samples were collected and sequenced. The prevalence of H. pylori and oral-associated bacteria was verified using fluorescence in situ hybridization (FISH) assays in GC tumoral tissues and matched nontumoral tissues. We found that the vertical distribution of the gastric microbiota, at the upper, middle, and lower third sites of GC, was likely an important factor causing microbial diversity in GC tumor tissues. The oral-associated microbiota cluster, which included Veillonella parvula, Streptococcus oralis, and Prevotella intermedia, was more abundant in the upper third of the GC. However, H. pylori was more abundant in the lower third of the GC and exhibited a significantly high degree of microbial correlation. The oral-associated microbiota module was co-exclusive with H. pylori in the lower third site of the GC tumoral tissue. Importantly, H. pylori-negative GC patients with oral-associated gastric microbiota showed worse overall survival, while the increase in microbial abundance in H. pylori-positive GC patients showed no difference in overall survival. The prevalence of V. parvula in both the dental plaque and GC tissue samples was concordant in the independent validation phase. We showed that the oral-associated species V. parvula and S. oralis were correlated with overall survival. Our study highlights the roles of the oral-associated microbiota in the upper third of the GC. In addition, oral-associated species may serve as noninvasive screening tools for the management of GC and an independent prognostic factor for H. pylori-negative GCs. IMPORTANCE Our study highlights the roles of the oral-associated microbiota in the upper third of gastric cancer (GC).We showed that the oral-associated species Veillonella parvula and Streptococcus oralis were correlated with overall survival. In addition, oral-associated species may serve as noninvasive screening tools for the management of GC and an independent prognostic factor for Helicobacter pylori-negative GCs.
Collapse
Affiliation(s)
- Lei Lei
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Lin-Yong Zhao
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Ran Cheng
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hongyu Zhang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mengying Xia
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiao-Long Chen
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Valentin Kudriashov
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Kai Liu
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Wei-Han Zhang
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Han Jiang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Chen
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Infection Control, West China Hospital, Sichuan University, Chengdu, China
| | - Liang Zhu
- Nuffield Department of Medicine, University of Oxford, Target Discovery Institute, Center for Medicines Discovery, Oxford, United Kingdom
- Nuffield Department of Medicine, University of Oxford, Chinese Academy of Medical Sciences (CAMS), CAMS Oxford Institute (COI), Oxford, United Kingdom
| | - Hongmei Zhou
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Kun Yang
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Tao Hu
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian-Kun Hu
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
22
|
Shang JR, Zhu J, Bai L, Kulabiek D, Zhai XX, Zheng X, Qian J. Adipocytes impact on gastric cancer progression: Prognostic insights and molecular features. World J Gastrointest Oncol 2024; 16:3011-3031. [PMID: 39072151 PMCID: PMC11271780 DOI: 10.4251/wjgo.v16.i7.3011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/17/2024] [Accepted: 05/28/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Adipocytes, especially adipocytes within tumor tissue known as cancer-associated adipocytes, have been increasingly recognized for their pivotal role in the tumor microenvironment of gastric cancer (GC). Their influence on tumor progression and patient prognosis has sparked significant interest in recent research. The main objectives of this study were to investigate adipocyte infiltration, assess its correlation with clinical pathological features, develop a prognostic prediction model based on independent prognostic factors, evaluate the impact of adipocytes on immune cell infiltration and tumor invasiveness in GC, and identify and validate genes associated with high adipocyte expression, exploring their potential diagnostic and prognostic value. AIM To explore the relationship between increased adipocytes within tumor tissue and prognosis in GC patients as well as the associated mechanisms and potential biomarkers, using public databases and clinical data. METHODS Using mRNA microarray datasets from the Gene Expression Omnibus database and clinical samples from Jiangsu Provincial Hospital, survival and regression analyses were conducted to determine the relevant prognostic factors in GC. Feature gene selection was performed using least absolute shrinkage and selection operator and support vector machine recursive feature elimination algorithms, followed by differential gene expression analysis, gene ontology, pathway analysis, and Gene Set Enrichment Analysis. Immune cell infiltration was analyzed using the CIBERSORT algorithm. RESULTS Tumor adipocyte infiltration correlated with poor prognosis in GC, leading to the development of a highly accurate and discriminative prognostic prediction model. Key genes, ADH1B, SFRP1, PLAC9, and FABP4, were identified as associated with high adipocyte expression in GC. The diagnostic and prognostic potential of these identified genes was validated using independent datasets. Downregulation of immune cells was observed in GC with high adipocyte expression. CONCLUSION GC with high intratumoral adipocyte expression demonstrated aggressive tumor biology and a poorer prognosis. The genes ADH1B, SFRP1, PLAC9, and FABP4 have been identified as holding diagnostic and prognostic significance in GC. These findings strongly support the use of adipocyte expression as a valuable indicator of tumor invasiveness and anticipated patient outcomes in GC.
Collapse
Affiliation(s)
- Jia-Rong Shang
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Jin Zhu
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Lu Bai
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Delida Kulabiek
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Xiao-Xue Zhai
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Xia Zheng
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Jun Qian
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| |
Collapse
|
23
|
Burz C, Pop V, Silaghi C, Lupan I, Samasca G. Helicobacter pylori Infection in Patients with Gastric Cancer: A 2024 Update. Cancers (Basel) 2024; 16:1958. [PMID: 38893079 PMCID: PMC11171272 DOI: 10.3390/cancers16111958] [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: 04/22/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Numerous studies have been performed on Helicobacter pylori infection because of the high death rate linked to this illness and gastric cancer. An update on the key developments in recent years in the investigation of Helicobacter pylori and gastric cancer is the goal of this review. Using the search term "Helicobacter pylori, gastric cancer", the PubMed database was searched. Only papers published in 2024 fulfilled the inclusion criteria. Because case report papers were not part of our investigation, they satisfied the exclusion criteria. Most of the research on the variable genes of Helicobacter pylori is guided by genetics to determine potential treatments. Studies on clinical treatments for the eradication of H. pylori with promising therapeutic options are needed. We found the fewest studies related to the immunopathology of H. pylori infection, which is still unknown. In conclusion, priority should be given to this kind of research.
Collapse
Affiliation(s)
- Claudia Burz
- Institute of Oncology “Prof. Ion Chiricuta”, 400015 Cluj-Napoca, Romania; (C.B.); (V.P.)
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Vlad Pop
- Institute of Oncology “Prof. Ion Chiricuta”, 400015 Cluj-Napoca, Romania; (C.B.); (V.P.)
| | - Ciprian Silaghi
- Department of Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Iulia Lupan
- Institute for Interdisciplinary Research in Bio-Nanosciences, 400271 Cluj-Napoca, Romania;
- Department of Molecular Biology, Babes-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Gabriel Samasca
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| |
Collapse
|