1
|
Seisenbekova A, Laryushina Y, Yukhnevich Y, Lavrinenko A, Shkreba A. Prevalence and risk factors of H. pylori infection among outpatient in Karaganda city (Kazakhstan). Future Sci OA 2025; 11:2461429. [PMID: 39927633 PMCID: PMC11812317 DOI: 10.1080/20565623.2025.2461429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/10/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND To better understand the factors associated with Helicobacter pylori infection, it is important to quantify the prevalence of H. pylori and identify the clinical and demographic characteristics of individuals with the infection. METHOD In this cross-sectional study 369 participants underwent a structured questionnaire, urease breath test, and endoscopy to determine their H. pylori status. RESULTS The frequency of H. pylori in the sample was 27.64%, and erosive antral gastritis, gastric ulcers, and duodenal ulcers were found to be significantly associated with infection. However, no differences were found in social status, family size, or shared utensil use between individuals with and without the infection. CONCLUSION These findings suggest that H. pylori is a significant risk factor for gastrointestinal conditions.
Collapse
Affiliation(s)
- Aizhan Seisenbekova
- Department of Internal Diseases, Karaganda Medical University, Karaganda, Kazakhstan
| | - Yelena Laryushina
- Department of Internal Diseases, Karaganda Medical University, Karaganda, Kazakhstan
| | | | | | - Alexey Shkreba
- University Clinic NC JSC “Karaganda Medical University”, Karaganda, Kazakhstan
| |
Collapse
|
2
|
Eid R, Blochouse E, Giese A, Papot S, Jay P, Varon C, Poinot P. Induced volatolomics to uncover new enzymatic hallmarks of precancerous lesions: a proof of concept on gastric preneoplasia in mice. Biochem Biophys Rep 2025; 43:102062. [PMID: 40519703 PMCID: PMC12166735 DOI: 10.1016/j.bbrep.2025.102062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 04/08/2025] [Accepted: 05/20/2025] [Indexed: 06/18/2025] Open
Abstract
Induced volatolomics is an emerging field of research that offers new opportunities in biology by detecting volatile reporters released by activatable probes, enabling the exploration of oncogenic processes. Building on its proven efficiency in exploring the evolution of implanted tumours, we hypothesized that induced volatolomics could extend its application to detect precancerous conditions. As a proof of concept, we performed a longitudinal study and investigated glycosidase activity during the early stages of gastric carcinogenesis development induced by Helicobacter felis infection in mice, mimicking the gastric carcinogenesis cascade induced by chronic Helicobacter pylori infection in humans. We identified upregulated exoglycosidases linked to acute infections or inflammatory processes in tissues infected by Helicobacter. Specifically, α-mannosidase and β-galactosidase activities in stomach tissue were found to be strongly associated with the initial stages of Helicobacter infection. Additionally, the activities of β-Glucuronidase and β-N-acetyl-glucosaminidase increased during the progression to preneoplastic stages, potentially signalling the transition from infection to inflammation-driven carcinogenesis. These enzymes may serve as early biomarkers for detecting gastric carcinogenesis. Our study highlights the potential of VOC-based probes for real-time monitoring of gastric cancer progression through tissue biopsies. Therefore, this study demonstrates the potential of induced volatolomics for investigating biological processes and uncovering new therapeutic strategies.
Collapse
Affiliation(s)
- Rony Eid
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Cedex 9, Poitiers, 86073, France
| | - Estelle Blochouse
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Cedex 9, Poitiers, 86073, France
| | - Alban Giese
- University of Bordeaux, INSERM, UMR1312 BoRdeaux Institute of Oncology, F33000, Bordeaux, France
| | - Sébastien Papot
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Cedex 9, Poitiers, 86073, France
| | - Philippe Jay
- University of Montpellier, CNRS UMR 5203, INSERM, Institute of Functional Genomics, Cedex 5, Montpellier, 34094, France
| | - Christine Varon
- University of Bordeaux, INSERM, UMR1312 BoRdeaux Institute of Oncology, F33000, Bordeaux, France
| | - Pauline Poinot
- University of Poitiers, UMR CNRS 7285, Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Cedex 9, Poitiers, 86073, France
| |
Collapse
|
3
|
Albano F, Severini FL, Calice G, Zoppoli P, Falco G, Notarangelo T. The role of the tumor microenvironment and inflammatory pathways in driving drug resistance in gastric cancer: A systematic review and meta-analysis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167821. [PMID: 40203956 DOI: 10.1016/j.bbadis.2025.167821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/14/2025] [Accepted: 03/26/2025] [Indexed: 04/11/2025]
Abstract
Tumor microenvironment (TME) plays a pivotal role in progression and low responsiveness to chemotherapy of gastric cancer (GC). The cascade of events that culminate with a sustained and chronic activation of inflammatory pathways underlies gastric tumorigenesis. Infiltrating immune cells enrolling in crosstalk with cancer cells that regulate inflammatory and immune status, generating an immunosuppressive TME that influences the response to therapy. Here we discuss the role of TME and the activation of inflammatory pathways to comprehend strategies to improve drug response. Furthermore, we provides systematic insight the role of TME cytotypes and related signatures reinforcing the critical roles of TAMs and Tregs, in promoting GC chemoresistance and tumor progression.
Collapse
Affiliation(s)
- Francesco Albano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Francesca Lospinoso Severini
- Laboratory of Preclinical and Translational Research, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, PZ, Rionero in Vulture, Italy
| | - Giovanni Calice
- Laboratory of Preclinical and Translational Research, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, PZ, Rionero in Vulture, Italy
| | - Pietro Zoppoli
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Geppino Falco
- Department of Biology, University of Naples Federico II, Naples, Italy; Biogem, Istituto di Biologia e Genetica Molecolare, AV, Ariano Irpino, Italy
| | - Tiziana Notarangelo
- Laboratory of Preclinical and Translational Research, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, PZ, Rionero in Vulture, Italy.
| |
Collapse
|
4
|
Lee CK, Park S, Lee Y, Yun C, Hong M, Nam CM, Chung HC, Rha SY. Efficacy of the first-line immune checkpoint inhibitor plus chemotherapy for gastroesophageal cancer: A meta-analysis of phase III trials including unreported PD-L1 subgroups. Cancer Lett 2025; 623:217718. [PMID: 40239914 DOI: 10.1016/j.canlet.2025.217718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 03/03/2025] [Accepted: 04/12/2025] [Indexed: 04/18/2025]
Abstract
The treatment paradigm for gastroesophageal cancers is evolving with immune checkpoint inhibitors (ICIs) as first-line therapy, making it crucial to understand their efficacy across patient subgroups, especially concerning PD-L1 expression. We performed a meta-analysis of Phase III randomized controlled trials targeting the effectiveness of ICIs with or without chemotherapy for advanced/metastatic HER2-negative gastroesophageal adenocarcinoma (GEA) or esophageal squamous cell carcinoma (ESCC). Kaplan-Meier (KM) curves of all-comer populations and subgroups according to reported PD-L1 cut-offs were extracted from published reports. Using KMSubtraction algorithm, unreported PD-L1 subgroup survival data were reconstructed by utilizing published KM survival curves. Thirteen first-line phase III RCTs involving 11,795 patients with GEA or ESCC were included. For GEA, ICI with or without chemotherapy showed longer OS in patients with PD-L1 combined positive score ≥1 (HR 0.77, 95 % confidence intervals [CI] 0.71-0.83 for ICI plus chemotherapy; HR 0.86, 95 %CI 0.75-1.01 for ICI alone) compared to chemotherapy alone, showing less benefits in low PD-L1 subgroups. ICI, with or without chemotherapy displayed survival benefits among PD-L1 tumor proportion score ≥1 % for ESCC (HR 0.62, 95 %CI 0.52-0.74 for ICI plus chemotherapy; HR 0.67, 95 %CI 0.54-0.84 for ICI alone) compared to chemotherapy alone. ICI combinations were similarly beneficial for Asian and global patients with GEA or ESCC. In conclusion, this meta-analysis, which includes unreported PD-L1 subgroups show benefit of ICIs with or without chemotherapy as a first-line treatment for advanced gastroesophageal cancers, particularly among patients with high PD-L1 expression.
Collapse
Affiliation(s)
- Choong-Kun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea; Song-dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Sejung Park
- Song-dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Yaeji Lee
- Department of Biostatistics and Computing, Yonsei University College of Medicine, Seoul, South Korea
| | - Choa Yun
- Department of Biostatistics and Computing, Yonsei University College of Medicine, Seoul, South Korea
| | - Moonki Hong
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Chung Mo Nam
- Department of Biostatistics and Computing, Yonsei University College of Medicine, Seoul, South Korea; Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Cheol Chung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea; Song-dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Sun Young Rha
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea; Song-dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
5
|
Pargali RH, Salehi MB, Bahador N. Molecular epidemiology and antibiotic resistance profiles of Mycobacterium and Nocardia species in patients with cancer: Detection of multidrug-resistant and extensively drug-resistant strains. Microb Pathog 2025; 204:107514. [PMID: 40154851 DOI: 10.1016/j.micpath.2025.107514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Patients with cancer are vulnerable to infections caused by mycobacteria and Nocardia. Antimicrobial agents are necessary to treat these infections, but extensive antibiotic use can lead to drug-resistant strains. Due to this issue, in the present study, we aimed to isolate, molecularly identify, prevalence, and assess the resistance genes in mycobacterial and nocardial isolates from patients with cancer. 79 clinical samples were obtained from patients with cancer which were referred to Mohammad Rasolallah hospitals in Shiraz. Initial identification was conducted through phenotypic and biochemical assays, followed by molecular confirmation using PCR amplification of the 16S rRNA and hsp65 genes. Genus and species determination was achieved through direct sequencing of the 16S rRNA gene. Drug susceptibility testing was carried out using the serial dilution method, adhering to the guidelines set by the Clinical and Laboratory Standards Institute (CLSI) in 2021. Out of 79 clinical samples, 17 isolates (21.51 %) were recovered and identified as Mycobacterium and Nocardia species through molecular and biochemical analyses. In our study, the most prevalent species were M. tuberculosis complex 6 (35.3 %) isolates, M. avium complex 3(17.64 %) isolates, M. terrae 2(11.76 %)isolates, N.cyriacigeorgica 2 (11.76 %) isolates, N. nova, N. kroppenstedtii, M. arupense, and M. canariasense 1 (5/89 %) isolate each. The analysis of the resistance profile of isolates showed that the M. tuberculosis (MTB) isolates HM7 and HM9 showed multidrug-resistant (MDR), and the isolates HM11 and HM12 showed extensively drug-resistant (XDR) profile, M. avium showed MDR profile, while no resistance patterns were observed in the Nocardia isolates. Moreover, the results show that some of the MTB isolates harbored katG, ermA, and rpoB resistance genes and M. avium isolates harbored katG and rpoB resistance genes. In conclusion, our results showed, that patients with cancer are vulnerable to infections caused by mycobacteria and Nocardia, moreover the emergence of antibiotic resistance further complicates the management of these infections, necessitating vigilant surveillance and the development of tailored treatment guidelines.
Collapse
Affiliation(s)
- Razieh Helali Pargali
- Department of Microbiology, Kazeroun Branch, Islamic Azad University, Kazeroun, Iran
| | - Majid Baseri Salehi
- Department of Microbiology, Kazeroun Branch, Islamic Azad University, Kazeroun, Iran.
| | - Nima Bahador
- Department of Microbiology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| |
Collapse
|
6
|
Lin N, Lin L, Huang X, Huang C, Gong J. Label-free quantitative proteomics of gastric high-grade intraepithelial neoplasia. Exp Ther Med 2025; 30:133. [PMID: 40432843 PMCID: PMC12107227 DOI: 10.3892/etm.2025.12883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 05/01/2025] [Indexed: 05/29/2025] Open
Abstract
Early detection and diagnosis are key to improving the survival rate and reducing the fatality rate linked to gastric cancer. The precancerous lesion of gastric cancer is referred to as gastric high-grade intraepithelial neoplasia (HGIN). Both the sensitivity and specificity of current biomarkers that aid in the diagnosis of gastric HGIN are still relatively low. Furthermore, proteomic data on gastric HGIN are still scarce. The present study aimed to explore candidate protein biomarkers for gastric HGIN screening with proteomics and bioinformatics technology. A total of 10 serum samples were collected and categorized into two groups, i.e., the gastric HGIN and the healthy control groups. Label-free quantification in conjunction with liquid chromatography with tandem mass spectrometry was employed to identify the probable biomarkers for gastric HGIN. Furthermore, differentially expressed proteins (DEPs) were quantified by proteomics analysis. In total, 1,192 distinct serum proteins were discovered between the gastric HGIN group and the healthy control group. DEPs were identified in the further analyses, utilizing a threshold of a 1.5-fold difference in expression level (P<0.05) in comparison with the control group. There were 18 upregulated and 12 downregulated proteins in the gastric HGIN group in comparison with the control group. Bioinformatics analyses were performed using Gene Ontology and KEGG pathway enrichment analyses. The GO analysis revealed that the DEPs were enriched in biological processes such as 'cellular', 'biological regulation', 'multicellular organismal', 'developmental' and 'reaction to stimulus processes', localized to 'cell', 'intracellular' and 'protein-containing complex', and involved in molecular functions such as 'molecular function modulator', 'binding' and 'catalytic activity'. The KEGG pathway enrichment analysis manifested that the DEPs were predominantly enriched in 'antigen processing and presentation', 'diabetic cardiomyopathy', 'Epstein-Barr virus infection', 'herpes simplex virus 1 infection', 'human immunodeficiency virus 1 infection' and 'human cytomegalovirus infection'. In conclusion, the present data provide more biological information for the formation of gastric HGIN and clues for further research on the pathogenesis of early gastric cancer.
Collapse
Affiliation(s)
- Nan Lin
- Department of Gastroenterology, The Affiliated Hospital of Putian University, Putian, Fujian 351100, P.R. China
| | - Liping Lin
- Department of Gastroenterology, The Affiliated Hospital of Putian University, Putian, Fujian 351100, P.R. China
| | - Xinxiang Huang
- Department of Gastroenterology, The Affiliated Hospital of Putian University, Putian, Fujian 351100, P.R. China
| | - Chaozhong Huang
- Department of Gastroenterology, The Affiliated Hospital of Putian University, Putian, Fujian 351100, P.R. China
| | - Jinrong Gong
- Department of Gastroenterology, The Affiliated Hospital of Putian University, Putian, Fujian 351100, P.R. China
| |
Collapse
|
7
|
Cruz Correa M, Xu RH, Moehler M, Oh DY, Kato K, Spigel D, Arkenau HT, Tabernero J, Zimina AV, Bai Y, Shi J, Lee KW, Hirano H, Wyrwicz L, Pazo Cid R, Xu H, Sheng T, Barnes G. Tislelizumab plus chemotherapy versus placebo plus chemotherapy as first-line treatment of advanced gastric or gastroesophageal junction adenocarcinoma: patient-reported outcomes in the RATIONALE-305 study. Curr Med Res Opin 2025:1-17. [PMID: 40528576 DOI: 10.1080/03007995.2025.2501588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/17/2025] [Accepted: 04/30/2025] [Indexed: 06/20/2025]
Abstract
OBJECTIVE RATIONALE-305 (NCT03777657) demonstrated that tislelizumab plus chemotherapy statistically improved overall survival versus placebo plus chemotherapy as first-line treatment in patients with advanced gastric/gastroesophageal junction adenocarcinoma (GC/GEJC). This analysis examined patient-reported outcomes (PROs) at final analysis. METHODS Adults with previously untreated, unresectable, or metastatic GC/GEJC were randomized (1:1) to tislelizumab or placebo intravenously once every 3 weeks plus chemotherapy. PROs assessed health-related quality of life (HRQoL) using EORTC QLQ-C30 and EORTC QLQ-STO22. A mixed model for repeated measures was used for PRO endpoints at treatment cycles 4 and 6, and time to deterioration was analyzed. RESULTS Tislelizumab arm had improved outcomes over placebo arm in least-squares (LS) mean change from baseline to cycle 6 for QLQ-C30 global health status/quality of life (GHS/QoL) (LS mean difference, 2.52 [95% CI: 0.29-4.74]), physical functioning (2.46 [0.49-4.43]), fatigue (-3.01 [-5.78 to -0.24]), and STO22 index score (-1.62 [-3.12 to -0.12]) as well as maintenance of upper gastrointestinal symptoms (-1.74 [-3.55 to 0.06]) and pain/discomfort (-1.88 [-4.03 to 0.27]). Patients receiving tislelizumab plus chemotherapy had a lower risk for deterioration of GHS/QoL (hazard ratio 0.77 [95% CI: 0.60-0.98]), physical functioning (0.72 [0.57-0.92]), STO22 index score (0.64 [0.45-0.92]), pain/discomfort (0.74 [0.58-0.96]), and upper gastrointestinal symptoms (0.73 [0.56-0.95]). CONCLUSIONS Advanced GC/GEJC patients treated with tislelizumab plus chemotherapy versus placebo plus chemotherapy in first-line had sustained and improved HRQoL. These results, along with previous efficacy and safety data, support tislelizumab plus chemotherapy as a first-line treatment option for GC/GEJC.Trial registration: The RATIONALE-305 trial is registered on ClinicalTrials.gov (ClinicalTrials.gov identifier: NCT03777657).
Collapse
Affiliation(s)
- Marcia Cruz Correa
- University of Puerto Rico, School of Medicine, San Juan, Puerto Rico and Pan American Center for Oncology Trials, San Juan, Puerto Rico
| | - Rui-Hua Xu
- University Cancer Center State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Department of Medical Oncology, Guangzhou, China
| | - Markus Moehler
- Johannes Gutenberg-University Clinic, Department of Internal Medicine I, Mainz, Germany
| | - Do-Youn Oh
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, South Korea
| | - Ken Kato
- National Cancer Center Hospital, Department of Gastrointestinal Medical Oncology, Tokyo, Japan
| | | | | | - Josep Tabernero
- Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), Department of Medical Oncology, Barcelona, Spain
| | | | - Yuxian Bai
- Harbin Medical University Cancer Hospital, Department of Gastrointestinal Oncology, Harbin, China
| | - Jianhua Shi
- Linyi Cancer Hospital, Department II of Medical Oncology, Linyi, China
| | - Keun-Wook Lee
- Seoul National University College of Medicine, Seoul National University Bundang Hospital, Department of Internal Medicine, Seongnam, South Korea
| | - Hidekazu Hirano
- National Cancer Center Hospital, Department of Gastrointestinal Medical Oncology, Tokyo, Japan
| | - Lucjan Wyrwicz
- Maria Sklodowska-Curie National Cancer Research Institute, Department of Oncology and Radiotherapy, Warsaw, Poland
| | - Roberto Pazo Cid
- Hospital Universitario Miguel Servet, Department of Medical Oncology, Zaragoza, Spain
| | - Hui Xu
- BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Tao Sheng
- BeiGene (Beijing) Co., Ltd., Beijing, China
| | | |
Collapse
|
8
|
Baghbanzadeh A, Rahmani S, Eslami S, Ahmadpour Youshanlui M, Shafiee N, Shafiee A, Khalaji A, Baradaran B. miR-146a-5p suppresses migration and downregulates vimentin and MMP-9 expression in gastric cancer cells. Discov Oncol 2025; 16:1075. [PMID: 40504386 PMCID: PMC12162404 DOI: 10.1007/s12672-025-02929-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 06/05/2025] [Indexed: 06/16/2025] Open
Abstract
Gastric cancer (GC) is a significant global health issue, characterized by poor prognosis due to its metastatic nature. This study investigates the role of microRNA-146a-5p (miR-146a-5p) in regulating migration and invasion of KATO III GC cells by targeting vimentin and matrix metalloproteinase-9 (MMP-9), critical mediators of epithelial-mesenchymal transition (EMT). miR-146a-5p expression was analyzed in GC cell lines, and functional assays were performed after transfection with a miR-146a-5p mimic. Results showed that miR-146a-5p expression was significantly reduced in GC cells, particularly in KATO III. Transfection with the mimic reduced cell viability, increased apoptosis, and inhibited migration, as shown by wound-healing assays. Additionally, miR-146a-5p downregulated vimentin and MMP-9 mRNA expression. These findings suggest that miR-146a-5p may influence metastatic behavior in GC cells through regulation of EMT-related genes. However, further studies, including protein-level validation and direct target confirmation, are needed. These results provide a basis for future investigation into the potential therapeutic role of miR-146a-5p in GC.
Collapse
Affiliation(s)
- Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shima Rahmani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahand Eslami
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Nadia Shafiee
- Bahrami Children's Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Atefe Shafiee
- Rajaie Cardiovascular Medical and Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirreza Khalaji
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
9
|
Zhu Y, Lin X, Wang T, Wang S, Wang W, Ke M, Zhu Y, Zhang B, Ofosuhemaa P, Wang Y, Hu M, Yang W, Hu A, Huang F, Zhao Q. Associated effects of blood metal(loid) exposure and impaired glucose metabolism in patients with gastric precancerous lesions or gastric cancer. Biometals 2025; 38:887-902. [PMID: 40232351 DOI: 10.1007/s10534-025-00684-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 04/04/2025] [Indexed: 04/16/2025]
Abstract
Exposure to metal(loid)s and glucose metabolism may influence the progression of gastric precancerous lesions (GPLs) or gastric cancer (GC), but their combined effects remain unclear. Our study aimed to elucidate the combined impact of metal (including metalloid and trace element) exposure and disturbances in glucose metabolism on the progression of GPLs and GC. From a prospective observational cohort of 1829 individuals, their metal(loid) levels and blood metabolism were analysed via inductively coupled plasma‒mass spectrometry and targeted metabolomics gas chromatography‒mass spectrometry, respectively. From healthy normal controls (NC) or GPLs to GC, we observed that the aluminum and arsenic levels decreased, whereas the vanadium, titanium and rubidium levels increased, but the iron, copper, zinc and barium levels initially decreased but then increased; these changes were not obvious from the NC to GPL group. With respect to glucose homeostasis, most metabolites decreased, except for phosphoenolpyruvate (PEP), which increased. Multiple logistic regression analysis revealed that titanium and phosphoenolpyruvate might be risk factors for GPLs, that barium is a protective factor for GC, and that D-glucaric acid might be a protective factor for GPLs and GC. Selenium, vanadium, titanium, succinate, maleate, isocitrate, PEP, and the tricarboxylic acid cycle (TCA) had good predictive potential for GPL and GC. Additionally, metal(loid)s such as arsenic, titanium, barium, aluminum, and vanadium were significantly correlated with multiple glucose metabolites involved in the TCA cycle in the GPL and GC groups. Our findings imply that metal(loid) exposure disrupts glucose metabolism, jointly influencing GPL and GC progression.
Collapse
Affiliation(s)
- Yuting Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Xiao Lin
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, China
- Department of Tuberculosis Control, Xiangcheng Center for Disease Control and Prevention, Suzhou, 215131, China
| | - Tingting Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, China
- Department of Hospital Nosocomial Infection, Chaohu Hospital of Anhui Medical University, Hefei, 230032, China
| | - Sheng Wang
- Research and Experiment Center, Anhui Medical University, Hefei, 230032, China
| | - Wuqi Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Mengran Ke
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Yan Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Bowen Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Princess Ofosuhemaa
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Yalei Wang
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230011, China
| | - Mingjun Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Wanshui Yang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Anla Hu
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Fen Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, 230032, China.
| | - Qihong Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
10
|
Qin J, Chen B, Sun YH, Wang XX, Wu C, Zhang C. The predictive value of miR-132-3p combined with Prognostic Nutritional Index (PNI) for gastric cancer prognosis. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2025; 117:303-312. [PMID: 39784731 DOI: 10.17235/reed.2024.10882/2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
BACKGROUND previous studies have demonstrated that the Prognostic Nutritional Index (PNI) can predict the prognosis of gastric cancer (GC) patients. However, few studies have focused on the auxiliary role of miRNA in predicting the prognosis of GC. OBJECTIVE this study aimed to clarify the role of the combined use of miR-132-3p and PNI to predict the prognosis of GC patients. METHODS a total of 126 patients with GC who underwent surgical treatment at our institution were included in the study. The qRT-PCR data was used to assess the expression of miR-132-3p in tumor and adjacent normal tissues. The predictive value of miR-132-3p and PNI for postoperative prognosis, and the relationships between miR-132-3p, PNI, and preoperative clinical characteristics, were assessed using ROC, χ², Kaplan-Meier survival analysis, and Cox regression analysis. Bioinformatics analyses and dual-luciferase reporter assay were conducted to elucidate the biological function of miR-132-3p and the underlying molecular mechanisms. RESULTS miR-132-3p was downregulated in GC tumor tissues and significantly and positively correlated with PNI. Both miR-132-3p and PNI were significantly associated with TNM stage and lymph node metastasis. Postoperative GC patients with low miR-132-3p expression and low PNI had lower survival rates, and both were independent risk factors for poor prognosis. The combination of miR-132-3p and PNI demonstrated better sensitivity and specificity to predict postoperative prognosis than either indicator alone. miR-132-3p may be involved in DNA damage response, angiogenesis, and immune response associated with GC development. MAPK1 was identified as a target gene regulated by miR-132-3p in GC. CONCLUSION the combination of miR-132-3p and PNI can effectively improve the predictive value of postoperative prognosis in GC patients. The combination of miR-132-3p and PNI, which are strongly correlated with pathological characteristics and can be readily obtained, has a potential clinical applicability in the prognosis of patients with GC.
Collapse
Affiliation(s)
| | - Bixia Chen
- Gastroenterology, Jiangmen Central Hospital
| | - Yan-Hui Sun
- Nutrition, The Seventh Medical Center of Chinese PLA General Hospital
| | - Xiao-Xiao Wang
- Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital
| | - Cong Wu
- Nutrition, The Seventh Medical Center of Chinese PLA General Hospital, China
| | - Caihua Zhang
- Oncology, People's Hospital Affiliated to Chongqing Three Gorges Medical College
| |
Collapse
|
11
|
Oje A, Galati J, Peek RM. Current Understanding of Optimal Prevention of Helicobacter pylori-Induced Cancer. Gastroenterol Clin North Am 2025; 54:397-413. [PMID: 40348495 DOI: 10.1016/j.gtc.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Gastric cancer is the fifth most common cancer and the fifth most common cause of cancer-related death globally. The key to improving outcomes lies in effective prevention and early detection, which are critical for successful curative interventions. Helicobacter pylori is the strongest known risk factor for gastric cancer, and eradication of this pathogen is critical for reducing cancer risk. By synthesizing current evidence and exploring the advanced therapeutic approaches, this review provides a comprehensive overview of best practices for mitigating gastric cancer through targeted bacterial intervention.
Collapse
Affiliation(s)
- Adesola Oje
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Jonathan Galati
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Richard M Peek
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
12
|
Zheng Y, Chen X, Huang Y, Lin X, Lin J, Mo Y, Gan L, Wei S, Wang Z, Song X, Tu Z. DDX27: An RNA helicase regulating cancer progression and therapeutic prospects. Int J Biol Macromol 2025; 313:144388. [PMID: 40394785 DOI: 10.1016/j.ijbiomac.2025.144388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2025] [Revised: 05/07/2025] [Accepted: 05/18/2025] [Indexed: 05/22/2025]
Abstract
DDX27, a member of the DEAD-box RNA helicase family, plays a crucial role in RNA metabolism, inflammation, and cancer progression. Elevated expression of DDX27 has been observed in multiple cancers, including oral squamous cell carcinoma (OSCC), breast cancer (BC), colorectal cancer (CRC), gastric cancer (GC), and hepatocellular carcinoma (HCC), where it is associated with poor prognosis, tumor growth, metastasis, and chemoresistance. DDX27 regulates the NF-κB signaling pathway, which is central to inflammation and tumor progression, and influences key cellular processes such as cell cycle regulation, apoptosis, migration, and stemness. Additionally, DDX27 promotes epithelial-mesenchymal transition (EMT), further contributing to metastasis. Its interactions with non-coding RNAs and various signaling pathways complicate treatment responses, making DDX27 a promising therapeutic target. This review explores the role of DDX27 as both a biomarker and therapeutic target, with potential strategies including small molecule inhibitors, RNA interference, and combination therapies with existing treatments such as NF-κB inhibitors or chemotherapy. Targeting DDX27 may help overcome resistance, reduce metastasis, and improve cancer treatment outcomes. Further research into its molecular mechanisms and interactions will be crucial for developing effective therapies, particularly for cancers with high metastatic potential.
Collapse
Affiliation(s)
- Yuantong Zheng
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, PR China
| | - Xinyi Chen
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, PR China
| | - Yunfei Huang
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, PR China
| | - Xuanli Lin
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, PR China
| | - Jiaxin Lin
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, PR China
| | - Yuting Mo
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, PR China
| | - Lu Gan
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, PR China
| | - Shuhua Wei
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, PR China
| | - Zhen Wang
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, PR China
| | - Xiaojuan Song
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, PR China
| | - Zhengchao Tu
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, PR China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development (MOE), School of Pharmacy, Jinan University, Guangzhou 510632, PR China.
| |
Collapse
|
13
|
Biswas S, Kanodia R, Seervi S, Kaur R, Shukla S, Singh S, Banerjee J, Banerjee S. Portrayal of the complex molecular landscape of multidrug resistance in gastric cancer: Unveiling the potential targets. Exp Cell Res 2025; 449:114580. [PMID: 40306607 DOI: 10.1016/j.yexcr.2025.114580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/27/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Gastric cancer (GC) is an aggressive malignancy among all Gastrointestinal cancer (GIC) types. Worldwide, among all cancer types, gastric cancer incidence and related mortality remain in fifth position. Multidrug resistance (MDR) in GC presents a major challenge to chemotherapy, and it significantly affects patient survival. A better understanding of the dynamic interaction of cellular factors contributing to MDR phenotype, e.g., the presence and expression of variants of MDR-related genes, including various drug-detoxifying and drug-efflux transporters, and expression of regulatory ncRNAs affecting the expression of MDR-related genes, is required to comprehend the molecular mechanisms for MDR development in GCs. This review article provides a holistic discussion of the cellular factors involved in the MDR development in GC cells, i.e., their roles and cross-talk between specific molecules that give rise to drug-sensitive and drug-resistant phenotypes. Moreover, the pharmacological perspective of drug resistance and the underlying biological processes that allow the escape of GC cells from the cytotoxic effects of drugs have also been discussed. Additionally, this review article provides an in-depth discussion on most potential candidates that can serve as MDR biomarkers in GIC cancer and the growing research interest in non-coding RNAs (ncRNAs) in GC. Notably, the miRNAs, circRNAs, and lncRNAs are not only emerging as crucial prognostic biomarkers of MDR in gastric cancers but also as potential targets for personalized medicine to combat the MDR challenge in GC patients.
Collapse
Affiliation(s)
- Siddhant Biswas
- School of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Koba, Institutional Area, Gandhinagar, Gujarat, 382426, India
| | - Riya Kanodia
- School of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Koba, Institutional Area, Gandhinagar, Gujarat, 382426, India
| | - Suman Seervi
- School of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Koba, Institutional Area, Gandhinagar, Gujarat, 382426, India
| | - Rajinder Kaur
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Sakshi Shukla
- School of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Koba, Institutional Area, Gandhinagar, Gujarat, 382426, India
| | - Samer Singh
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Juni Banerjee
- School of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Koba, Institutional Area, Gandhinagar, Gujarat, 382426, India.
| | - Shuvomoy Banerjee
- School of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Koba, Institutional Area, Gandhinagar, Gujarat, 382426, India.
| |
Collapse
|
14
|
Kim YS. Gastric Carcinoma. Curr Top Microbiol Immunol 2025. [PMID: 40423781 DOI: 10.1007/82_2025_303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Epstein-Barr virus (EBV)-associated gastric cancers (EBVaGCs) account for about 10% of gastric cancers globally, with higher prevalence in East Asia and Latin America. These cancers develop through a "gastritis-infection-cancer sequence" and are characterized by unique molecular signatures, including CpG island methylator phenotype and mutations in ARID1A and PIK3CA genes. EBVaGCs typically present in the proximal stomach with diffuse-type histology and dense lymphocytic infiltration. Key viral proteins EBNA1 and LMP2A drive oncogenesis by altering cellular processes and immune responses. The IFN-γ signature and extensive epigenetic modifications contribute to their distinct profile. Despite often presenting at advanced stages, EBVaGCs generally have a more favorable prognosis. EBV employs sophisticated strategies to evade immune detection, utilizing latent proteins and noncoding RNAs. Paradoxically, despite an immune-hot environment, EBVaGCs demonstrate effective immune evasion, partly due to the expression of immune checkpoint molecules like PD-L1 and LAG3. Treatment approaches vary based on disease stage, from endoscopic resection for early-stage cancers to systemic therapies for advanced cases. Immunotherapy, particularly PD-1/PD-L1 inhibitors, shows promising results. Emerging research suggests combining these with LAG3 inhibitors may enhance efficacy. Ongoing research and advanced genomic techniques continue to reveal new insights, paving the way for personalized therapies and novel diagnostic approaches.
Collapse
Affiliation(s)
- Young-Sik Kim
- Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Yang Z, Niu R, Han J, Guo J, Lv Y. Hedgehog inhibitors exert anti-proliferation effects and synergistically interact with trastuzumab in HER2-positive gastric cancer models. Acta Oncol 2025; 64:715-728. [PMID: 40426308 DOI: 10.2340/1651-226x.2025.42219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 03/17/2025] [Indexed: 05/29/2025]
Abstract
BACKGROUND Gastric cancer (GC) remains a significant health concern with limited therapeutic options. While trastuzumab, a Human Epidermal Growth Factor Receptor 2 (HER2)-targeting antibody, has shown efficacy in HER2-positive GC, its therapeutic response is moderate. Hedgehog (Hh) signalling plays a critical role in the progression of GC. METHODS We evaluated the sensitivity of various GC cell lines to trastuzumab. The HER2-positive HGC-27 cell line was identified as the most sensitive. In addition, the effects of two Hedgehog inhibitors, vismodegib and cyclopamine, were assessed on cell growth by monitoring SMO expression. Both in vitro and in vivo assays were conducted to explore the combination of Hh inhibitors and trastuzumab. RESULTS Both vismodegib and cyclopamine exerted anti-proliferative effects, and synergistically enhanced the anti-tumour activity of trastuzumab in HER2-positive GC models. Mechanistically, Hh inhibitors inhibited the AKT/mTOR signalling pathway through Smoothened (SMO) depletion, contributing to their anti-growth effects. INTERPRETATION This study highlights the potential of combining Hh inhibitors with trastuzumab as a therapeutic strategy for HER2-positive GC by targeting the AKT/mTOR pathway.
Collapse
Affiliation(s)
- Zixin Yang
- The second department of Oncology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Ren Niu
- The second department of Oncology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Jinzhu Han
- The second department of Oncology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Jie Guo
- The second department of Oncology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Yingqian Lv
- The second department of Oncology, The Second Hospital of Hebei Medical University, Hebei, China.
| |
Collapse
|
16
|
Li X, Li Q, Yang Y, Zhu Z, Zhu H. Global, Regional and National Burden of Stomach Cancer and Its Prediction in 25 Years: A Cross-Sectional Study of the Global Burden of Disease Study 2021. Br J Hosp Med (Lond) 2025; 86:1-21. [PMID: 40405840 DOI: 10.12968/hmed.2025.0023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
Aims/Background Given the tremendous threat of stomach cancer (SC) to global public health, detailed information and dynamic updates on the SC health burden are needed to mitigate the repercussions. In this article, we present a systematic analysis of the global burden and trends of SC using data from the Global Burden of Disease (GBD) Study 2021, aiming to provide insights for forming effective global management and prevention strategies. Methods Stomach cancer incidence, prevalence, mortality, disability-adjusted life years (DALYs) and the corresponding age-standardized rates (ASRs) were estimated. Then, trends of the disease burden were analyzed using the estimated annual percentage changes (EAPC). Lastly, we made a unique attempt to use two powerful and versatile techniques, autoregressive integrated moving average (ARIMA) and exponential smoothing (ES) models, to provide more comprehensive and accurate forecasts for future trends in the disease burden. Results Despite the steady decreasing trend in age-standardized rates, the total numbers of incidence, prevalence, and deaths all increased from 1990 to 2021. Subgroup analysis demonstrated great disparities in different age and gender groups, sociodemographic index (SDI) quintiles, GBD regions and countries. Both the ARIMA and ES models demonstrated a persistent rise in SC cases and a concurrent decline in ASRs, with the trend being more pronounced in males. Conclusion Since SC is already a significant health issue globally, it is expected that the estimated disease burden will continue to rise in the future. Therefore, global coordinated efforts are needed to implement effective screening projects, consolidate preventive measures and formulate targeted treatments to alleviate the SC burden.
Collapse
Affiliation(s)
- Xinyan Li
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qinghua Li
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yanqing Yang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhenghui Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hong Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
17
|
Yu K, Cao Y, Zhang Z, Wang L, Gu Y, Xu T, Zhang X, Guo X, Shen Z, Qin J. Blockade of CLEVER-1 restrains immune evasion and enhances anti-PD-1 immunotherapy in gastric cancer. J Immunother Cancer 2025; 13:e011080. [PMID: 40404204 PMCID: PMC12096977 DOI: 10.1136/jitc-2024-011080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 04/16/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) remains a major global health burden. Despite the advancements in immunotherapy for patients with GC, the heterogeneity of GC limits response rates, especially in immune "cold" subtypes, including genomically stable and epithelial-mesenchymal transition GC. Common lymphatic endothelial and vascular endothelial receptor-1 (CLEVER-1), a newly recognized immune checkpoint molecule predominantly expressed on tumor-associated macrophages (TAMs), remains poorly understood in GC. This study aims to explore the clinical significance of CLEVER-1+TAM infiltration, elucidate its role in modulating the tumor immune landscape, and investigate the therapeutic potential of CLEVER-1 blockade in enhancing immunotherapy. METHODS This study analyzed two independent GC cohorts and single-cell RNA sequencing data (GSE183904). CLEVER-1 expression in TAMs was assessed via multiplex immunofluorescence, flow cytometry, and RNA sequencing. The clinical relevance of CLEVER-1+TAM infiltration was evaluated in relation to tumor, node, metastases staging, molecular subtypes, prognosis, and immunochemotherapy response. Transcriptomic and pathway analyses characterized the immunophenotype of CLEVER-1+TAMs. Functional assays examined their suppression on CD8+T cells, while interventional experiments assessed the impact of CLEVER-1 blockade alone or with programmed cell death protein-1 (PD-1) inhibition. RESULTS CLEVER-1 was predominantly expressed on TAMs in GC and was associated with worse clinical outcomes. Transcriptomic and phenotypic analyses revealed that CLEVER-1+TAMs display a dynamic immunophenotype and critically suppress T-cell function, fostering an immunosuppressive microenvironment. High CLEVER-1+TAM infiltration was linked to poor response or adaptive resistance to PD-1 blockade therapy. CLEVER-1 blockade reprogrammed TAMs toward a pro-inflammatory phenotype, resulting in enhanced CD8+T cell cytotoxicity and proliferation. Co-targeting CLEVER-1 and PD-1 synergistically enhanced antitumor responses. CONCLUSIONS High infiltration of CLEVER-1+TAMs indicates immune suppression and poor prognosis in GC. The combination of CLEVER-1 and PD-1 blockade emerges as a dual-pronged strategy to boost immune-mediated tumor control and prevent treatment relapse in GC.
Collapse
Affiliation(s)
- Kuan Yu
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yifan Cao
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital Fudan University, Shanghai, China
| | - Zihao Zhang
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital Fudan University, Shanghai, China
| | - Leihao Wang
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yichao Gu
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital Fudan University, Shanghai, China
| | - Tianwei Xu
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
| | - Xiaolei Zhang
- Department of Pathology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Xinxin Guo
- Department of Pathology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Zhenbin Shen
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital Fudan University, Shanghai, China
| | - Jing Qin
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital Fudan University, Shanghai, China
| |
Collapse
|
18
|
Zhu J, Zhu X, Su T, Zhou H, Wang S, Shi W. The Development and Assessment of a Unique Disulfidptosis-Associated lncRNA Profile for Immune Microenvironment Prediction and Personalized Therapy in Gastric Adenocarcinoma. Biomedicines 2025; 13:1224. [PMID: 40427051 PMCID: PMC12109475 DOI: 10.3390/biomedicines13051224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 05/11/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Long non-coding RNAs (lncRNAs) are crucial factors affecting the occurrence, progression, and prognosis of gastric carcinoma (GC). The accumulation of disulfide bonds to excessive levels in cells expressing high SLC7A11 triggers disulfidptosis, which functions as a regulated form of cellular death. Research has demonstrated that upregulated SLC7A11 is common in human cancers, but the effect of disulfidptosis on GC remains unclear. Identifying lncRNAs associated with disulfidptosis (drlncRNAs) and establishing a prognostic risk profile holds considerable importance for advancing GC research and treatment. Methods: Clinical records and transcriptomic datasets from individuals with GC were acquired from The Cancer Genome Atlas (TCGA) repository. A three-drlncRNA risk model was built using three common regression analysis methods. Then we used receiver operating characteristic (ROC) curves, independent prognostic analysis, and additional statistical approaches to assess the precision of the model. This investigation additionally encompassed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, immune cell infiltration evaluation, and pharmacological sensitivity predictions. To further investigate immunotherapy response disparities between patient cohorts with elevated- and reduced-risk scores, analyses of tumor mutational burden (TMB), tumor immune dysfunction and exclusion (TIDE), and microsatellite instability (MSI) were implemented. Results: We constructed a unique model composed of three drlncRNAs (AC107021.2, AC016394.2, and AC129507.1). Its independent prognostic capability for GC patients was validated through both single-variable and multivariable Cox regression analyses. GO and KEGG pathway assessments revealed predominant enrichment within the elevated-risk cohort, particularly in pathways involving sulfur compound interactions, traditional Wnt signaling mechanisms, cell-substrate adherens junctions, and cAMP signaling cascades, among others. Tumor microenvironment (TME) evaluation demonstrated elevated ImmuneScores, StromalScores, and ESTIMATEScores within the high-risk patient population. Concurrently, this elevated-risk cohort exhibited enhanced immune cell infiltration patterns, whereas the reduced-risk group displayed superior expression of immune checkpoints (ICPs). Additional investigations revealed that patients categorized into the reduced-risk classification possessed greater tumor mutational burden, increased MSI-high proportions, and diminished tumor immune dysfunction and exclusion scores compared to their high-risk counterparts. Pharmacological sensitivity assessments confirmed the superior efficacy of several therapeutic agents, including gemcitabine and veliparib (ABT.888), in patients with lower risk classifications. Conclusions: Our established risk stratification system demonstrates independent prognostic predictive capacity while offering personalized clinical intervention guidance for individuals diagnosed with GC.
Collapse
Affiliation(s)
- Jiyue Zhu
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; (J.Z.); (X.Z.); (T.S.)
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai 200092, China
| | - Xiang Zhu
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; (J.Z.); (X.Z.); (T.S.)
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai 200092, China
| | - Tingting Su
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; (J.Z.); (X.Z.); (T.S.)
| | - Huiqing Zhou
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China;
| | - Shouhua Wang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; (J.Z.); (X.Z.); (T.S.)
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai 200092, China
| | - Weibin Shi
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai 200092, China
| |
Collapse
|
19
|
Abraham H, Sluggett L, Huber D, Olson R. Population-based differences in cancer incidence between immigrants and non-immigrants in Canada between 1992 and 2015. BMC Public Health 2025; 25:1849. [PMID: 40389969 PMCID: PMC12087223 DOI: 10.1186/s12889-025-23117-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 05/09/2025] [Indexed: 05/21/2025] Open
Abstract
OBJECTIVES With increasing immigration in Canada and strained cancer treatment infrastructure, there's a pressing need for long-term data on immigrant health and cancer incidence. This information is crucial for planning future cancer services and to alleviate the burden on both the population and healthcare system. METHODS Statistics Canada data were linked from the 1991 Canadian Census, Canadian Cancer Registry, and Canadian Vital Statistics Database to follow a cohort from 1992 to 2015 and compare cancer incidence between immigrants and the Canadian-born for any cancer and specific types of cancers. Immigrants were further classified based on time spent in Canada. RESULTS Immigrants had lower odds of developing any cancer (OR = 0.92, 95% CI [0.92-0.93], p < 0.001) compared to non-immigrants. However, for stomach cancer and non-cervical gynecological cancers, the odds of cancer incidence were greater for immigrants than for the Canadian-born. Cox regression showed that recent immigrants (0-4 years in Canada) had a lower hazard ratio (HR = 0.77, 95% CI [0.71-0.84], p < 0.001) compared to non-immigrants. Those who lived 5-9 years and 10-19 years in Canada had a higher hazard ratio (HR = 0.82, 95% CI [0.75-0.89], p < 0.001; HR = 0.90, 95% CI [0.82-0.98], p = 0.011), respectively. Immigrants who had been in Canada for 20 years or longer had the highest hazard ratio (HR = 0.98, 95% CI [0.90-1.07], p = 0.632), indicating that the so-called "healthy immigrant effect" lessens over time. CONCLUSION Results demonstrated the healthy immigrant effect lessens over time spent in Canada. However, this effect was not uniform across countries of origin and cancer types. Therefore, this research, provides a deeper understanding of immigrant cancer outcomes and will be useful for cancer planning services and cancer control strategies.
Collapse
Affiliation(s)
| | - Larine Sluggett
- School of Health Sciences, University of Northern, Prince George, BC, Canada
| | - Dezene Huber
- Faculty of Environment, University of Northern, Prince George, BC, Canada
| | - Robert Olson
- BC Cancer- Prince George, Prince George, BC, Canada.
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
20
|
Drapkina OM, Maev IV, Bordin DS, Bakulin IG, Garbuzova EV, Ershova AI, Livzan MA. Key Scientific Achievements in Gastroenterology in 2024: Meeting of the Internal Medicine Sciences’ Council of the Department of Medical Sciences of the Russian Academy of Sciences. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2025; 24:4386. [DOI: 10.15829/1728-8800-2025-4386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2025] Open
Abstract
19.12.2024 состоялось очередное заседание Совета по терапевтическим наукам Секции клинической медицины РАН, посвященное ключевым научным достижениям гастроэнтерологии в 2024 г. В ходе заседания обсуждались актуальные вопросы гастроэнтерологической практики, включая проблемы профилактики рака желудка у пациентов с хроническим гастритом, а также приоритетные направления в диагностике и лечении патологии печени и различных заболеваний кишечника.
Collapse
Affiliation(s)
- O. M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine; Russian University of Medicine
| | | | - D. S. Bordin
- Russian University of Medicine; Tver State Medical University; Loginov Moscow Clinical Scientific Center
| | - I. G. Bakulin
- I.I. Mechnikov North-Western State Medical University
| | - E. V. Garbuzova
- National Medical Research Center for Therapy and Preventive Medicine
| | - A. I. Ershova
- National Medical Research Center for Therapy and Preventive Medicine
| | | |
Collapse
|
21
|
Philip D, Hodgkiss R, Radhakrishnan SK, Sinha A, Acharjee A. Deciphering microbial and metabolic influences in gastrointestinal diseases-unveiling their roles in gastric cancer, colorectal cancer, and inflammatory bowel disease. J Transl Med 2025; 23:549. [PMID: 40380167 DOI: 10.1186/s12967-025-06552-w] [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: 03/16/2025] [Accepted: 05/04/2025] [Indexed: 05/19/2025] Open
Abstract
INTRODUCTION Gastrointestinal disorders (GIDs) affect nearly 40% of the global population, with gut microbiome-metabolome interactions playing a crucial role in gastric cancer (GC), colorectal cancer (CRC), and inflammatory bowel disease (IBD). This study aims to investigate how microbial and metabolic alterations contribute to disease development and assess whether biomarkers identified in one disease could potentially be used to predict another, highlighting cross-disease applicability. METHODS Microbiome and metabolome datasets from Erawijantari et al. (GC: n = 42, Healthy: n = 54), Franzosa et al. (IBD: n = 164, Healthy: n = 56), and Yachida et al. (CRC: n = 150, Healthy: n = 127) were subjected to three machine learning algorithms, eXtreme gradient boosting (XGBoost), Random Forest, and Least Absolute Shrinkage and Selection Operator (LASSO). Feature selection identified microbial and metabolite biomarkers unique to each disease and shared across conditions. A microbial community (MICOM) model simulated gut microbial growth and metabolite fluxes, revealing metabolic differences between healthy and diseased states. Finally, network analysis uncovered metabolite clusters associated with disease traits. RESULTS Combined machine learning models demonstrated strong predictive performance, with Random Forest achieving the highest Area Under the Curve(AUC) scores for GC(0.94[0.83-1.00]), CRC (0.75[0.62-0.86]), and IBD (0.93[0.86-0.98]). These models were then employed for cross-disease analysis, revealing that models trained on GC data successfully predicted IBD biomarkers, while CRC models predicted GC biomarkers with optimal performance scores. CONCLUSION These findings emphasize the potential of microbial and metabolic profiling in cross-disease characterization particularly for GIDs, advancing biomarker discovery for improved diagnostics and targeted therapies.
Collapse
Affiliation(s)
- Daryll Philip
- Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham Dubai, Dubai, UAE
| | - Rebecca Hodgkiss
- Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | | | - Akshat Sinha
- Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Animesh Acharjee
- Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham Dubai, Dubai, UAE.
- Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK.
- Centre for Health Data Research, University of Birmingham, Birmingham, UK.
- Institute of Translational Medicine, University Hospitals Birmingham NHS, Foundation Trust, Birmingham, UK.
| |
Collapse
|
22
|
Yu BM, Lee SD, Hwang BR, Kim JS, Yu S, Nam KT, Lee YC. Application of an organoid-based model to explore Helicobacter pylori-human gastric epithelium interaction in vitro. Front Cell Infect Microbiol 2025; 15:1572244. [PMID: 40444149 PMCID: PMC12119492 DOI: 10.3389/fcimb.2025.1572244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/23/2025] [Indexed: 06/02/2025] Open
Abstract
Helicobacter pylori infection causes histopathologic changes in gastric epithelial cells, resulting in conditions such as gastritis, gastric ulcers, and ultimately, gastric cancer. To date, various experimental models, including cell lines and animal studies, have been employed to investigate these pathological processes. However, each model presents its limitations. This study compared the re-cultured three-dimensional organoids from infected single cells, apical-out, and two-dimensional (2D) organoids models to better understand the mechanisms underlying the epithelial changes caused by H. pylori infection in the human stomach. Thus, we analyzed the epithelial cell responses, inflammatory mediator expressions, apical-junctional complex alterations, and H. pylori infection interactions in these organoid models. Moreover, we revealed that the high accessibility and experimental efficiency of the apical-out and 2D models enable easier manipulation and faster analysis of H. pylori infection compared to the single-cell infection model. These results indicate the potential of various organoid models in H. pylori infection studies to provide essential data that help in understanding the mechanisms of epithelial changes and in developing new therapeutic strategies for gastric pathology.
Collapse
Affiliation(s)
- Byeong Min Yu
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - So Dam Lee
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Bo Ram Hwang
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji Seon Kim
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sungsook Yu
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ki Taek Nam
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong Chan Lee
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
23
|
Yang W, Wu X, Wang J, Ou W, Huang X. Integrated single-cell and transcriptome sequencing data reveal the value of IL1RAP in gastric cancer microenvironment and prognosis. Front Oncol 2025; 15:1584619. [PMID: 40444099 PMCID: PMC12119286 DOI: 10.3389/fonc.2025.1584619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 04/23/2025] [Indexed: 06/02/2025] Open
Abstract
Background Investigating the pivotal role of IL1RAP in the tumor microenvironment of gastric cancer. Method Download and collate transcriptomic and single-cell data from gastric cancer patients. Three machine learning algorithms identified distinct sets of prognostic genes in gastric cancer patients. The CIBERSORT and ssGSEA algorithms elucidated immune infiltration patterns, while TIDE and TCGA predicted immune-related outcomes. Furthermore, single-cell sequencing data confirmed the interaction of IL1RAP within the tumor microenvironment. Finally, differential expression levels of IL1RAP protein and mRNA were validated. Result After machine learning screening and independent dataset validation, high IL1RAP expression was identified as a poor prognostic factor for gastric cancer patients. Immune infiltration analysis indicated that the low IL1RAP expression group was associated with higher infiltration of CD8+ T cells and M1-type macrophages, whereas the high IL1RAP expression group exhibited increased presence of M2-type macrophages. Immunotherapy prediction models suggested a more favorable response to PD-1 treatment in the low IL1RAP expression group. Prognostic models incorporating IL1RAP demonstrated superior predictive performance. Single-cell data analysis revealed that IL1RAP plays a critical role in regulating intercellular communication within the tumor microenvironment. Our findings were further validated by confirming elevated IL1RAP expression levels in gastric cancer tissues. Conclusion IL1RAP plays a critical role in the tumor microenvironment of gastric cancer and serves as a robust predictor of immunotherapy efficacy in gastric cancer.
Collapse
Affiliation(s)
| | | | | | | | - Xing Huang
- Gastrointestinal Surgery, Nanping First Hospital Affiliated to Fujian Medical
University, Nanping, Fujian, China
| |
Collapse
|
24
|
Bertok T, Pinkeova A, Lorencova L, Datkova A, Hires M, Jane E, Tkac J. Glycoproteomics of Gastrointestinal Cancers and Its Use in Clinical Diagnostics. J Proteome Res 2025. [PMID: 40368336 DOI: 10.1021/acs.jproteome.5c00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Cancer is a leading cause of death worldwide, resulting in substantial economic costs. Because cancer is a complex, heterogeneous group of diseases affecting a variety of cells, its detection may sometimes be difficult. Herein we review a large group of the gastrointestinal cancers (oral, esophageal, stomach, pancreatic, liver, and bowel cancers) and the possibility of using glycans conjugated to protein backbones for less-invasive diagnoses than the commonly used endoscopic approaches. The reality of bacterial N-glycosylation and the effect of epithelial mucosa on gut microbiota are discussed. Current advantages, barriers, and advantages in the prospective use of selected glycomic approaches in clinical practice are also detailed.
Collapse
Affiliation(s)
- Tomas Bertok
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovak Republic
| | - Andrea Pinkeova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovak Republic
- Glycanostics, Kudlakova 7, 841 01 Bratislava, Slovak Republic
| | - Lenka Lorencova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovak Republic
| | - Anna Datkova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovak Republic
| | - Michal Hires
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovak Republic
| | - Eduard Jane
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovak Republic
- Glycanostics, Kudlakova 7, 841 01 Bratislava, Slovak Republic
| | - Jan Tkac
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovak Republic
- Glycanostics, Kudlakova 7, 841 01 Bratislava, Slovak Republic
| |
Collapse
|
25
|
Samalin E, Evesque L, Turpin A, De La Fouchardiere C, Khemissa-Akouz F, Bouché O, Muller M, Dermeche S, Botsen D, Tougeron D, Zaanan A, Ben Abdelghani M, Guardiola E, Dubreuil O, Le Brun Ly V, Hennequin A, Watson S, Sefrioui D, Lecomte T, De Sousa Carvalho N, Hulin A, Crapez E, Castan F, Senellart H. Regorafenib combined with irinotecan as second-line treatment in metastatic gastro-oesophageal adenocarcinomas: results of PRODIGE 58-UCGI35-REGIRI Unicancer randomised phase II study. ESMO Open 2025; 10:105096. [PMID: 40359707 DOI: 10.1016/j.esmoop.2025.105096] [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: 11/12/2024] [Revised: 03/13/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Several options have been evaluated in metastatic gastro-oesophageal adenocarcinomas (mGA) after failure of first-line fluoropyrimidine and platinum-based chemotherapy. Regorafenib (REGO), a receptor tyrosine kinase inhibitor, has shown promising activity as second- and third-line treatment of mGA. PATIENTS AND METHODS PRODIGE58-UCGI35-REGIRI was a comparative, prospective, phase II, open-label study evaluating the safety and efficacy of REGO [160 mg/day on day 2 (D2)-D8/D16-D22] plus irinotecan (IRI: 180 mg/m2 intravenously on D1/D15 every 28 days) versus IRI alone in patients with mGA (gastric or gastro-oesophageal junction/tumour Siewert II and III) after failure of first-line fluoropyrimidine and platinum-based chemotherapy. Primary endpoint was overall survival (OS). RESULTS Forty-four patients were included in the REGIRI arm and 45 in the IRI arm, primary tumours (67.4%) were mainly localised in the gastro-oesophageal junction, and 60.7% patients had synchronous metastases. With a median follow-up of 19.4 months [95% confidence interval (CI) 16.8-29.9 months], median OS was 6.3 months (95% CI 5.2-7.1 months) versus 8.2 months (95% CI 5.2-9.7 months) in the REGIRI versus IRI arms (hazard ratio 1.11, 95% CI 0.70-1.74, P = 0.66). Median progression-free survival was 2.2 months versus 1.9 months, objective response rate 15.9% versus 13.3%, and disease control rate 45.5% versus 33.3%. Grade 3 treatment-related adverse events (AEs) were reported for 52.3% of patients in the REGIRI arm versus 23.3% in the IRI arm with four toxic deaths (two homozygous UGT1A1∗28 patients died from sepsis and thrombotic microangiopathy, and two heterozygous UGT1A1∗1/∗28 patients from diarrhoea and pulmonary embolism), versus one (UGT1A1∗1 wild-type patient died from primary tumour perforation). Main grade ≥3 AEs were diarrhoea (18.2% versus 7.0%), hypertension (9.1% versus 0.0%), asthenia (6.8% versus 0.0%), febrile neutropenia (6.8% versus 0.0%), neutropenia (6.8% versus 11.6%), and weight decrease (6.8% versus 0.0%). CONCLUSIONS The study was stopped early because of limited efficacy and increased toxicities in the REGIRI arm, possibly due to drug interactions. No optimal sub-population that could benefit from a REGIRI regimen exposure was identified.
Collapse
Affiliation(s)
- E Samalin
- ICM, Department of Medical Oncology, Université de Montpellier, Montpellier, France.
| | - L Evesque
- Centre Antoine Lacassagne, Nice, France
| | - A Turpin
- Department of Medical Oncology, CHRU Lille, Lille, France; CNRS INSERM UMR9020-U1277, CANTHER Cancer Heterogeneity Plasticity and Resistance to Therapies, Université de Lille, Lille, France
| | | | | | | | - M Muller
- CHRU de Nancy, Vandoeuvre-Les-Nancy, France
| | - S Dermeche
- Institut Paoli Calmettes, Marseille, France
| | - D Botsen
- Department of Medical Oncology, Institut Godinot, Reims, France
| | | | - A Zaanan
- Hôpital Européen George Pompidou, Paris, France
| | | | - E Guardiola
- Centre de Cancérologie du Grand Montpellier, Montpellier, France
| | | | | | - A Hennequin
- Centre Georges François Leclerc, Dijon, France
| | | | | | - T Lecomte
- CHRU Tours-Hôpital Trousseau, Chambray-lès-Tours, France
| | | | - A Hulin
- APHP, GH H Mondor, Créteil, France
| | - E Crapez
- ICM, Translational Research Unit, Montpellier, France
| | - F Castan
- ICM, Department of Medical Oncology, Université de Montpellier, Montpellier, France
| | | |
Collapse
|
26
|
He Z, Dang J, Cui X, Li B, Bao S, Fan J. The distinct role of IL-34 and IL-35 in gastric cancer. Front Immunol 2025; 16:1559508. [PMID: 40416985 PMCID: PMC12098620 DOI: 10.3389/fimmu.2025.1559508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 04/21/2025] [Indexed: 05/27/2025] Open
Abstract
Gastric cancer (GC) remains a major challenge due to its high mortality and morbidity, despite extensive research. Dysregulated host immunity plays a critical role in carcinogenesis, particularly among susceptible cohorts. In the gastric mucosa of GC patients, a reduction in IL-34 and TAM1, accompanied by an increase in TAM2 via M-CSF, enhances Th2 cell function, reduces pro-inflammatory activity, and elevates anti-inflammatory responses. Consequently, TAM2 acts in both paracrine and autocrine manners to polarize and boost TAM2, creating a tumour-favourable microenvironment that supports GC progression. High levels of TAM2, observed during advanced GC stages, suppress gastric IL-34 production, further promoting GC development. In contrast, IL-35, a cytokine involved in immune regulation and suppression, is produced by activated T cells and/or B cells in the affected gastric mucosa. Persistent H. pylori infection in GC tissues is associated with significant infiltration of IL-35-producing B cells and regulatory T cells (Tregs), which enhance the immunosuppressive and pro-tumour microenvironment by disrupting the local immune balance. Upregulated mucosal IL-35 promotes the polarization of TAM2 and Tregs while suppressing TAM1 cells, fostering a tumour-friendly environment that allows transformed gastric mucosal cells to evade immune surveillance, particularly in chronic H. pylori-infected patients. This cascade enhances proliferation and invasion while suppressing differentiation and apoptosis of GC cells. Together, the differential regulation of these cytokines creates an environment that supports cancer progression and resistance to therapy. Targeting the IL-34 and IL-35 pathways may offer a novel therapeutic strategy for improving outcomes in GC patients.
Collapse
Affiliation(s)
- Zhiyun He
- General Surgery Department, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Jie Dang
- Central Sterile Supply Department, Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xiang Cui
- General Surgery Department, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Bo Li
- General Surgery Department, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Shisan Bao
- Scientific Research Division, The Third Affiliated Hospital of Gansu University of Chinese Medicine, Baiyin, China
- Scientific Research Division, The First People’s Hospital of Baiyin, Baiyin, Gansu, China
| | - Jingchun Fan
- School of Public Health, Centre for Evidence-based Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
27
|
Wang Z, Liu P, Wang J, Ma P, Liu X. Causal relationship of garlic or onion with gastric cancer based on a Mendelian randomization study. Medicine (Baltimore) 2025; 104:e41639. [PMID: 40324220 PMCID: PMC12055067 DOI: 10.1097/md.0000000000041639] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 12/02/2024] [Accepted: 02/05/2025] [Indexed: 05/07/2025] Open
Abstract
In observational studies, it has been known that garlic or onions have a negative causal relationship with gastric cancer (GC). In this study, we aim to explore the negative causal relationship between garlic or onion and GC through Mendelian randomization (MR) analysis. The instrumental variable selection for MR analysis is single nucleotide polymorphisms associated with garlic or onion, mainly using the inverse variance weighted method, combined with MR Egger, weighted media, simple mode, and weighted modes to evaluate their causal impact on GC. In addition, sensitivity analysis such as Cochran Q test, pleiotropy test, and leave-one-out method were used to evaluate the robustness of the impact of these single nucleotide polymorphisms on GC. The inverse variance weighted method showed a negative correlation between garlic and GC risk (odds ratio = 0.70; 95% confidence interval 0.49-0.99; P = .046), while there was no relationship between onion and GC, and the sensitivity analysis results showed robustness. The current study has revealed that garlic may be a factor in reducing the risk of GC, providing a strategy for preventing and treating GC.
Collapse
Affiliation(s)
- Zhaoyin Wang
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Pengfei Liu
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Jingbin Wang
- Department of Spleen and Stomach Diseases, Shenzhen Hospital (Fu Tian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Pengli Ma
- Department of Spleen and Stomach Diseases, Shenzhen Hospital (Fu Tian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Xinyao Liu
- Department of Spleen and Stomach Diseases, Shenzhen Hospital (Fu Tian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
28
|
Palominos C, Kirdun M, Nikzad AH, Spilka MJ, Homan P, Sommer IE, Tang SX, Hinzen W. A single composite index of semantic behavior tracks symptoms of psychosis over time. Schizophr Res 2025; 279:116-127. [PMID: 40187184 DOI: 10.1016/j.schres.2025.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/28/2025] [Accepted: 03/29/2025] [Indexed: 04/07/2025]
Abstract
Semantic variables automatically extracted from spontaneous speech characterize anomalous semantic associations generated by groups with schizophrenia spectrum disorders (SSD). However, with the use of different language models and numerous aspects of semantic associations that could be tracked, the semantic space has become very high-dimensional, challenging both theoretical understanding and practical applications. This study aimed to summarize this space into a single composite semantic index and to test whether it can track diagnosis and symptom profiles over time at an individual level. The index was derived from a principal component analysis (PCA) yielding a linear combination of 117 semantic variables. It was tested in discourse samples of English speakers performing a picture description task, involving a total of 103 individuals with SSD and 36 healthy controls (HC) compared across four time points. Results showed that the index distinguished between SSD and HC groups, identified transitions from acute psychosis to remission and stabilization, predicted the sum of scores of the Thought, Language and Communication (TLC) index as well as subscores, capturing 65 % of the variance in the sum of TLC scores. These findings show that a single indicator meaningfully summarizes a shift in semantic associations in psychosis and tracks symptoms over time, while also pointing to variance unexplained, which is likely covered by other semantic and non-semantic factors.
Collapse
Affiliation(s)
- Claudio Palominos
- Department of Translation & Language Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
| | - Maryia Kirdun
- Department of Translation & Language Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Amir H Nikzad
- Zucker Hillside Hospital Northwell Health, New Hyde Park, NY, United States of America; Feinstein Institutes for Medical Research, Institute of Behavioral Science, Manhasset, NY, United States of America
| | | | - Philipp Homan
- Department of Adult Psychiatry and Psychotherapy, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Iris E Sommer
- Department of Neuroscience, University Medical Center Groningen, Antoni Deusinglaan 2, room 117, Groningen, Netherlands
| | - Sunny X Tang
- Zucker Hillside Hospital Northwell Health, New Hyde Park, NY, United States of America; Feinstein Institutes for Medical Research, Institute of Behavioral Science, Manhasset, NY, United States of America; Donald and Barbara Zucker School of Medicine, Department of Psychiatry, Hempstead, NY, United States of America; Linguistic Data Consortium, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Wolfram Hinzen
- Department of Translation & Language Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
29
|
Cao Z, Wang Z, Yang L, Li T, Tao X, Niu X. Reshaping the immune microenvironment and reversing immunosenescence by natural products: Prospects for immunotherapy in gastric cancer. Semin Cancer Biol 2025; 110:1-16. [PMID: 39923925 DOI: 10.1016/j.semcancer.2025.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025]
Abstract
Gastric cancer (GC) represents a global health-care challenge. Recent progress in immunotherapy has elicited attracted considerable attention as a viable treatment option through modulating the host immune system and unleashing pre-existing immunity, which has profoundly revolutionized oncology, especially GC. Nonetheless, low clinical response and intrinsic and acquired resistance remain persistently challenging. The microenvironment of GC comprising multifarious stromal cell types has remarkable immunosuppressive elements that may impact the efficacy of immunotherapy. Immunosenescence is increasingly regarded as a factor that contributes to cancer development, remodels the tumor microenvironment and affects the efficacy of immunotherapy. Natural products are at the forefront of traditional medicine. Senotherapeutics is a class of drugs and natural products capable of delaying, preventing, or reversing the senescence process (i.e., senolytics) or suppressing senescence-associated secretory phenotype (i.e., senomorphics). Emerging evidence supports that natural products can improve the efficacy of existing immunotherapy and expand their indications in GC mainly based upon remodeling the immunosuppressive microenvironment and reversing immunosenescence. The review provides an integrated review of previously reported and ongoing clinical trials with immunotherapeutic regimens in GC and discusses current challenges. Next, we focus on natural compounds that exert anti-GC functions and possess immunomodulatory properties. More attention is paid to the potential of these natural compounds in modulating the immune microenvironment and immunosenescence. Lastly, we discuss the nanomedicine that can overcome the deficiencies of natural products. Altogether, our review suggests the enormous potential of natural compounds in GC immunotherapy, and provides an important direction for future research.
Collapse
Affiliation(s)
- Zhipeng Cao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, China
| | - Zhilin Wang
- Department of Pain Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Li Yang
- Department of Anesthesiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Tian Li
- Tianjin Key Laboratory of Acute Abdomen Disease-Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China.
| | - Xueshu Tao
- Department of Pain Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Xing Niu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
30
|
Lee HJ, Kim J, Yoon SH, Kong SH, Kim WH, Park DJ, Lee HJ, Yang HK. Effectiveness of ERAS program on postoperative recovery after gastric cancer surgery: a randomized clinical trial. Int J Surg 2025; 111:3306-3313. [PMID: 40072360 PMCID: PMC12165489 DOI: 10.1097/js9.0000000000002328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/20/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND Previous studies have reported the effectiveness of the "enhanced recovery after surgery" program in patients who underwent gastric cancer surgery, mostly based on the 2014 gastrectomy guidelines. Therefore, based on subsequent advancements in perioperative management, this randomized, controlled, open-label, single-center study aimed to assess the impact of a recent evidence-based multimodal enhanced recovery after surgery program on the quality of early recovery after gastric cancer surgery. MATERIALS AND METHODS This study included adult patients scheduled to undergo elective laparoscopic or robotic distal gastrectomy for gastric cancer. Patients were randomly assigned to the enhanced recovery after surgery or conventional group. The primary outcome was the total Quality of Recovery-15 score assessed 24, 48, and 72 h postoperatively. Differences between both groups were evaluated using a linear mixed-effects model. We hypothesized that an increase of at least 8 points in the Korean version of Quality of Recovery-15 scores would indicate a clinically significant improvement, consistent with the minimal clinically important difference (≥8) for Quality of Recovery-15. Secondary outcomes included pain scores at rest and during coughing, cumulative fentanyl consumption through intravenous patient-controlled analgesia, postoperative nausea/vomiting incidence, and gastrointestinal dysfunction as measured using the I-FEED score - all assessed 24, 48, and 72 h postoperatively. RESULTS For the 92 patients examined (enhanced recovery after surgery, n = 45; conventional, n = 47), the estimated difference in the postoperative Quality of Recovery-15 total scores between the two groups during the first days was significantly larger than the minimal clinically important difference of Quality of Recovery-15 (mean difference: 16.0, 95% confidence interval: 8.9-23.0, P < 0.001). Furthermore, excluding the incidence of postoperative nausea/vomiting, the enhanced recovery after surgery group demonstrated significant improvements in other secondary outcomes. CONCLUSIONS Our evidence-based multimodal enhanced recovery after surgery program significantly improved the quality of early postoperative recovery after minimally invasive distal gastrectomy.
Collapse
Affiliation(s)
- Ho-Jin Lee
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jeesun Kim
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Soo-Hyuk Yoon
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seong-Ho Kong
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Won Ho Kim
- Department of Anesthesiology and Pain Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Do Joong Park
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyuk-Joon Lee
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Han-Kwang Yang
- Department of Surgery and Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
31
|
Zaman S, Hussain MI, Kausar M, Mostafa OE, Mohamedahmed AY, Hajibandeh S, Hajibandeh S, Camprodon R, Sellahewa C. Intracorporeal versus extracorporeal anastomosis in laparoscopic total gastrectomy: a systematic review and meta-analysis. Int J Surg 2025; 111:3441-3455. [PMID: 40009559 PMCID: PMC12165532 DOI: 10.1097/js9.0000000000002296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/16/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND To evaluate outcomes of intracorporeal (IOJ) versus extracorporeal (EOJ) oesophagojejunostomy following laparoscopic total gastrectomy (LTG) for the treatment of gastric cancer. METHODS A comprehensive search of various electronic databases was conducted. Comparative studies of IOJ versus EOJ following LTG in patients with gastric malignancy were included. Primary outcomes were anastomotic leak, anastomotic bleeding, and anastomotic stricture formation. Secondary outcomes included operative time, length of hospital stay (LOS), volume of intra-operative haemorrhage, number of harvested lymph nodes, time to flatus, time to soft diet, intra-abdominal infection, pulmonary infection, surgical site infection (SSI), duodenal stump leak, pancreatic fistula occurrence, postoperative ileus, re-operation, and mortality. Combined overall effect sizes were calculated using the random-effects model, and the Newcastle-Ottawa Scale was used to assess risk of bias. RESULTS Seventeen non-randomised studies enrolling 2,960 patients divided between an IOJ ( n = 1430) and EOJ ( n = 1530) group were included. IOJ was associated with significantly lower risk of anastomotic stricture ( P = 0.01), volume of intra-operative bleeding ( P = < 0.001), and SSI (P = 0.04) compared to EOJ. No difference was found in anastomotic leak ( P = 0.93); anastomotic bleeding ( P = 0.35); operative time ( P = 0.63); LOS ( P = 0.30); lymph node yield ( P = 0.17); time to first flatus ( P = 0.77); time to resumption of soft diet ( P = 0.32); intra-abdominal infection ( P = 0.22); pulmonary infection ( P = 0.45); duodenal stump leak ( P = 0.46); pancreatic fistula occurrence ( P = 0.16); and paralytic ileus ( P = 0.59), re-operation ( P = 0.50), and mortality ( P = 0.23) between the two groups. CONCLUSIONS LTG for gastric malignancy with IOJ may be associated with lower risk of anastomotic stricture and SSI compared to the extracorporeal approach. However, future adequately powered randomized studies are needed to compare the two techniques.
Collapse
Affiliation(s)
- Shafquat Zaman
- Department of General Surgery, Queen’s Hospital Burton, University Hospitals of Derby and Burton NHS Foundation Trust, Burton on Trent, UK
- College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Mohammad Iqbal Hussain
- Department of General Surgery, Great Western Hospitals NHS Foundation Trust, Swindon, UK
| | - Maria Kausar
- Department of General and Upper Gastrointestinal Surgery, Russells Hall Hospital, Dudley Group NHS Foundation Trust, Dudley, West Midlands, UK
| | - Omar E.S. Mostafa
- Department of General and Upper Gastrointestinal Surgery, Russells Hall Hospital, Dudley Group NHS Foundation Trust, Dudley, West Midlands, UK
| | - Ali Yasen Mohamedahmed
- Department of General Surgery, Queen’s Hospital Burton, University Hospitals of Derby and Burton NHS Foundation Trust, Burton on Trent, UK
| | - Shahab Hajibandeh
- Department of Hepatobiliary and Pancreatic Surgery, Swansea Bay University Health Board, Swansea, UK
| | - Shahin Hajibandeh
- Department of Hepatobiliary and Pancreatic Surgery, Royal Stoke University Hospital, University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, UK
| | - Ricardo Camprodon
- Department of General and Upper Gastrointestinal Surgery, Russells Hall Hospital, Dudley Group NHS Foundation Trust, Dudley, West Midlands, UK
| | - Chaminda Sellahewa
- Department of General and Upper Gastrointestinal Surgery, Russells Hall Hospital, Dudley Group NHS Foundation Trust, Dudley, West Midlands, UK
| |
Collapse
|
32
|
Ying X, Ying Z, Gao X, Wang Y, Lv X. CRISPR-mediated WNK4 point mutation aggravates tumor progression and weakens chemotherapy sensitivity in gastric cancer. Histol Histopathol 2025; 40:711-720. [PMID: 39363579 DOI: 10.14670/hh-18-810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
OBJECTIVE Gastric cancer (GC) is the fifth most common malignancy, the molecular targets of which have been increasingly explored in recent years. As a serine/threonine protein kinase, the role of WNK lysine deficient protein kinase 4 (WNK4) in GC was clarified in this study. METHODS Human GC lines AGS and MKN45 were stably transfected with a WNK4 mutant constructed by the CRISPR/Cas9 method and treated with cis-dichlorodiammine platinum (CDDP, 2 μg/mL) and 5-fluorouracil (5-FU, 5 μg/mL) for 48h. Tumor-bearing mice were established with 5×106 mutant-type AGS cells, and injected with 40 mg/kg WP1066, the inhibitor of signal transducer and activator of transcription 3 (STAT3), for 21 days. Cell malignant potential and tumor growth were assessed. STAT3 activation was identified by western blot and immunohistochemistry. The interaction between WNK4 and STAT3 was determined using co-immunoprecipitation and immunofluorescence co-localization. RESULTS WNK4 mutation promoted proliferation and invasion, and upregulated the p-STAT3/STAT3 value in GC cells with/without 5-FU and CDDP treatments, while inhibiting apoptosis of GC cells without drug treatment. In tumor-bearing mice, WNK4 mutation accelerated tumor growth, increased levels of p-STAT3, STAT3, and p-STAT3/STAT3, and strengthened the co-immunoprecipitation and co-localizing with STAT3; however, these effects were reversed by WP1066 treatment. CONCLUSION Through activating STAT3, WNK4 mutation impacts both the natural and drug-treated growth of GC cells or tumors, suggesting a new avenue for preclinical research.
Collapse
Affiliation(s)
- Xiaojun Ying
- Department of General Surgery, Yongkang First People's Hospital, PR China
| | - Zhen Ying
- Department of Gynecology, Yongkang First People's Hospital, PR China
| | - Xiaobing Gao
- Department of General Surgery, Yongkang First People's Hospital, PR China
| | - Yong Wang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, PR China
| | - Xinting Lv
- Department of General Surgery, Yongkang First People's Hospital, PR China.
| |
Collapse
|
33
|
Maubach G, Kanthasamy AK, Gogia S, Naumann M. The enigma of maladaptation in gastric pathophysiology. Trends Cancer 2025; 11:448-461. [PMID: 39984410 DOI: 10.1016/j.trecan.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/23/2025]
Abstract
Despite a decline in global incidence, gastric cancer (GC) remains a major health concern. The development of GC is a sequential, multistage maladaptive process involving numerous different factors. Understanding the complexity of GC development is crucial for early detection, effective treatment, and, ultimately, prevention. In this respect, identifying the impact of risk factors contributing to the emergence or progression of GC, such as Helicobacter pylori infection, host and bacterial genetics, alcohol consumption, smoking, and preserved foods, will aid in combatting this disease. In this review, we focus on recent developments in understanding the role of the microbiome, dysfunctional molecular pathways, and immune evasion in gastric pathophysiology. We also highlight challenges and advances in treatment of GC.
Collapse
Affiliation(s)
- Gunter Maubach
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Arun K Kanthasamy
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Sandro Gogia
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany.
| |
Collapse
|
34
|
He B, Hu Y, Wu Y, Wang C, Gao L, Gong C, Li Z, Gao N, Yang H, Xiao Y, Yang S. Helicobacter pylori CagA elevates FTO to induce gastric cancer progression via a "hit-and-run" paradigm. Cancer Commun (Lond) 2025; 45:608-631. [PMID: 39960839 PMCID: PMC12067399 DOI: 10.1002/cac2.70004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 01/23/2025] [Accepted: 01/26/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) infection contributes significantly to gastric cancer (GC) progression. The intrinsic mechanisms of H. pylori-host interactions and their role in promoting GC progression need further investigation. In this study, we explored the potential role of fat mass and obesity-associated protein (FTO) in mediating Cytotoxin-associated gene A (CagA)-induced GC progression. METHODS The effects of H. pylori infection on N6-methyladenosine (m6A) modification were evaluated in both human samples and GC cell lines. The function of FTO in the progression of GC was elucidated through in vitro and in vivo studies. A series of techniques, including methylated RNA immunoprecipitation sequencing, RNA sequencing, RNA binding protein immunoprecipitation, and chromatin immunoprecipitation assays, were utilized to investigate the mechanism by which FTO mediates the capacity of cagA-positive H. pylori to promote GC progression. Furthermore, the therapeutic potential of the FTO inhibitor meclofenamic acid (MA) in impeding GC progression was evaluated across GC cells, animal models, and human GC organoids. RESULTS Infection with cagA-positive H. pylori upregulated the expression of FTO, which was essential for CagA-mediated GC metastasis and significantly associated with a poor prognosis in GC patients. Mechanistically, CagA delivered by H. pylori enhanced FTO transcription via Jun proto-oncogene. Elevated FTO induced demethylation of m6A and inhibited the degradation of heparin-binding EGF-like growth factor (HBEGF), thereby facilitating the epithelial-mesenchymal transition (EMT) process in GC cells. Interestingly, eradication of H. pylori did not fully reverse the increases in FTO and HBEGF levels induced by cagA-positive H. pylori. However, treatment with a combination of antibiotics and MA substantially inhibited cagA-positive H. pylori-induced EMT and prevented GC metastasis. CONCLUSION Our study revealed that FTO mediates the "hit-and-run" mechanism of CagA-induced GC progression, which suggests that the therapeutic targeting of FTO could offer a promising approach to the prevention of CagA-induced cancer progression.
Collapse
Affiliation(s)
- Bing He
- Department of GastroenterologyXinqiao HospitalArmy Military Medical UniversityChongqingP. R. China
| | - Yiyang Hu
- Department of OncologyThe General Hospital of Western Theater CommandChengduSichuanP. R. China
| | - Yuyun Wu
- Department of GastroenterologyXinqiao HospitalArmy Military Medical UniversityChongqingP. R. China
| | - Chao Wang
- Department of GastroenterologyXinqiao HospitalArmy Military Medical UniversityChongqingP. R. China
| | - Limin Gao
- Department of GastroenterologyXinqiao HospitalArmy Military Medical UniversityChongqingP. R. China
| | - Chunli Gong
- Department of GastroenterologyXinqiao HospitalArmy Military Medical UniversityChongqingP. R. China
| | - Zhibin Li
- Department of GastroenterologyXinqiao HospitalArmy Military Medical UniversityChongqingP. R. China
| | - Nannan Gao
- Department of GastroenterologyXinqiao HospitalArmy Military Medical UniversityChongqingP. R. China
| | - Huan Yang
- Department of GastroenterologyXinqiao HospitalArmy Military Medical UniversityChongqingP. R. China
| | - Yufeng Xiao
- Department of GastroenterologyXinqiao HospitalArmy Military Medical UniversityChongqingP. R. China
| | - Shiming Yang
- Department of GastroenterologyXinqiao HospitalArmy Military Medical UniversityChongqingP. R. China
- Chongqing Institute for Brain and Intelligence, Guangyang Bay LaboratoryChongqingP. R. China
| |
Collapse
|
35
|
Bahcecioglu IB, Guler S, Morkavuk SB, Turan M, Akgul GG, Erzincan MB, Ozluk KK, Bardakci O, Gulcelik MA. A Novel and Feasible Intracorporeal Esophagojejunostomy Anastomosis in Totally Laparoscopic Total Gastrectomy Surgery: Sutureless L-Shape with Endoscopic Assistance (SLEJ). MEDICINA (KAUNAS, LITHUANIA) 2025; 61:795. [PMID: 40428753 PMCID: PMC12113305 DOI: 10.3390/medicina61050795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/19/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025]
Abstract
Background and Objectives: In contrast to the standardization of laparoscopic gastrectomy techniques, the complexity of intracorporeal anastomosis techniques in totally laparoscopic total gastrectomy, the lack of standardization, the positional challenges posed by working in a confined space, and varying complication rates have prevented a consensus on the optimal intracorporeal digestive tract reconstruction method. Selecting an appropriate reconstruction method for esophagojejunostomy is crucial for a successful surgical outcome. This study aims to define a modified anastomotic technique for TLTG and share our experience with this technique. Materials and Methods: A total of 21 patients who underwent TLTG with D2 LND between July 2024 and December 2024 using the sutureless L-shape esophagojejunostomy (SLEJ) technique at the Surgical Oncology Clinic of Gulhane Training and Research Hospital due to gastric cancer were included in the study. In our technique, gastrectomy, lymph node dissection, anastomosis preparation, esophagojejunostomy anastomosis, and enteroenterostomy anastomosis were all performed laparoscopically and intracorporeally. Results: The mean operative time was 180.48 min, with a mean EJ anastomosis duration of 40.24 min. In the standard technique, two Endo GIA™ staplers were used for pyloric and small bowel transection, two for EJ anastomosis, and one for intracorporeal jejunojejunostomy. In only one patient, three staplers were used for anastomosis. Therefore, the average number of staplers was 5.05, with a mean of 2.05 staplers used for anastomosis. The mean hospital stay was 8.19 days, and there were no mortalities. The number of patients with an anastomotic leakage was 1. Since the patient's general condition remained stable, percutaneous drainage or laparotomy was not planned. The patients' esophagojejunostomy anastomotic leak was classified as Class 1 and Grade 3a according to the Clavien-Dindo classification. The average size of our widest incision was 3.28 cm, and surgical site infections were developed in two patients. Conclusions: Sutureless L-Shape With Endoscopic Assistance (SLEJ) is an easily applicable, technically simpler, shorter-in-duration, easier-to-learn, and safer intracorporeal EJ anastomosis technique with a low rate of postoperative complications.
Collapse
Affiliation(s)
- Ibrahim Burak Bahcecioglu
- Department of Surgical Oncology, Gulhane Research and Training Hospital, Ankara 06010, Turkey; (I.B.B.); (S.G.); (M.B.E.); (K.K.O.); (O.B.)
| | - Sumeyra Guler
- Department of Surgical Oncology, Gulhane Research and Training Hospital, Ankara 06010, Turkey; (I.B.B.); (S.G.); (M.B.E.); (K.K.O.); (O.B.)
| | - Sevket Baris Morkavuk
- Department of Surgical Oncology, Gulhane Faculty of Medicine, Health Sciences University, Ankara Gulhane Research and Training Hospital, Ankara 06010, Turkey; (G.G.A.); (M.A.G.)
| | - Mujdat Turan
- Department of General Surgery, Gulhane Faculty of Medicine, Health Sciences University, Ankara Gulhane Research and Training Hospital, Ankara 06010, Turkey;
| | - Gokhan Giray Akgul
- Department of Surgical Oncology, Gulhane Faculty of Medicine, Health Sciences University, Ankara Gulhane Research and Training Hospital, Ankara 06010, Turkey; (G.G.A.); (M.A.G.)
| | - Mirac Baris Erzincan
- Department of Surgical Oncology, Gulhane Research and Training Hospital, Ankara 06010, Turkey; (I.B.B.); (S.G.); (M.B.E.); (K.K.O.); (O.B.)
| | - Kubilay Kenan Ozluk
- Department of Surgical Oncology, Gulhane Research and Training Hospital, Ankara 06010, Turkey; (I.B.B.); (S.G.); (M.B.E.); (K.K.O.); (O.B.)
| | - Osman Bardakci
- Department of Surgical Oncology, Gulhane Research and Training Hospital, Ankara 06010, Turkey; (I.B.B.); (S.G.); (M.B.E.); (K.K.O.); (O.B.)
| | - Mehmet Ali Gulcelik
- Department of Surgical Oncology, Gulhane Faculty of Medicine, Health Sciences University, Ankara Gulhane Research and Training Hospital, Ankara 06010, Turkey; (G.G.A.); (M.A.G.)
| |
Collapse
|
36
|
In K, Kang S, Lee H, Eun H, Moon H, Lee E, Kim S, Sung J, Lee B. Proportional Correlation Between Systemic Inflammation Response Index and Gastric Cancer Recurrence Time: A Retrospective Study. Cancers (Basel) 2025; 17:1415. [PMID: 40361340 PMCID: PMC12070897 DOI: 10.3390/cancers17091415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Disease recurrence is the primary cause of death in patients with gastric cancer who have undergone complete surgical resection. No prognostic factors for recurrence, other than the Tumor, Node, and Metastasis stage, have been established. However, recurrence rates differ even within the same Tumor, Node, and Metastasis stage. Therefore, we aimed to develop a new prognostic confidence measure for gastric cancer recurrence and demonstrate its practical utility. METHODS This was a retrospective study based on the medical records of the Chungnam National University Hospital, Republic of Korea. We enrolled patients diagnosed with stage II/III gastric cancer who underwent complete surgical resection and adjuvant chemotherapy over the past 12 years. The association between seven variables, including the systemic inflammation response index (SIRI) and gastric cancer recurrence, was analyzed. RESULTS A total of 296 patients were enrolled in this study. Although other factors did not exhibit significant correlations, SIRI showed a significant positive correlation with gastric cancer recurrence risk, confirmed through Cox regression testing (hazard ratio, 1.231; 95% confidence interval, 1.04-1.45). Linear regression analysis revealed a significant association between higher SIRI values and shorter recurrence time (p = 0.044; β = -0.225). CONCLUSIONS In this study, other than SIRI, effective prognostic factors related to gastric cancer recurrence were not verified, thus indicating SIRI as a potential independent prognostic factor.
Collapse
Affiliation(s)
| | - Sunhyung Kang
- Division of Gastroenterology, Department of Internal Medicine, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea; (K.I.); (H.L.); (H.E.); (H.M.); (E.L.); (S.K.); (J.S.); (B.L.)
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Gu Y, Hao J, He D, Sun H, Cui X, Tian W, Zhang Y, Jin C, Wang H. The universal zero markup drug policy and gastric cancer hospitalization expenses: an analysis of trends and influencing factors in Shanghai from 2014 to 2021. BMC Health Serv Res 2025; 25:580. [PMID: 40264071 PMCID: PMC12016107 DOI: 10.1186/s12913-025-12422-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/12/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND The increasing hospitalization expenses for Gastric Cancer (GC) impose a notable economic burden on society. Although the Chinese government has implemented the Universal Zero Markup Drug Policy (UZMDP) to control the growth of hospitalization expenditures, costs have continued to rise. Identifying the factors influencing the hospitalization expenses of GC patients is crucial. This study aimed to analyze the trends and factors influencing hospitalization expenses of GC patients in Shanghai from 2014 to 2021. METHODS Data were sourced from the Health Network of Shanghai Economic Information Center. We employed interrupted time series analysis (ITSA) to analyze the trends in various medical expenditures before and after the implementation of the UZMDP. The degree of association between various medical expenditures and hospitalization expenditures of GC patients was calculated by using the new grey relational analysis (GRA). Furthermore, multiple linear regression was employed to identify the influencing factors. RESULTS A total of 23,335 participants were included in this study. The ITSA results showed an increasing trend in hospitalization expenses following the implementation of UZMDP. Drug expenses decreased immediately post-UZMDP, but subsequently began to rise over time. Post-UZMDP, the expenses of medical consumables, examination, and healthcare services all showed an upward trend. The new GRA indicated that the influencing factors of hospitalization expenses, in order of importance, were expenses for drugs, consumables, healthcare services, and examination. Multivariable linear regression analysis revealed that GC patients aged 60 or below incurred lower hospitalization expenses (Coefficient = -780.06, P = 0.0398). However, factors associated with increased hospitalization expenses included longer length of stay (Coefficient = 1753.01, P < 0.001), surgeries (Coefficient = 29,047.26, P < 0.001), and hospitalization in the tertiary hospitals (Coefficient = 25,485.19, P < 0.001) or secondary hospitals (Coefficient = 17,755.12, P < 0.001). CONCLUSIONS Hospitalization expenses of GC patients in Shanghai have been rising annually from 2014 to 2021. Despite the implementation of the UZMDP, drug expenses remain a major factor in escalating hospitalization expenses. The hospitalization expenses of GC patients are significantly influenced by several factors, including the demographic characteristics of patients, the severity of diseases, and the levels of hospitals. These findings provide a basis for more effective management of the hospitalization expenses for GC patients.
Collapse
Affiliation(s)
- Yichun Gu
- Shanghai Health Development Research Center, Shanghai, China.
| | - Jiajun Hao
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Da He
- Shanghai Health Development Research Center, Shanghai, China
| | - Hui Sun
- Shanghai Health Development Research Center, Shanghai, China
| | - Xin Cui
- Shanghai Health Statistics Center, Shanghai, China
| | - Wenqi Tian
- Shanghai Health Statistics Center, Shanghai, China
| | - Yulin Zhang
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Chunlin Jin
- Shanghai Health Development Research Center, Shanghai, China.
| | - Haiyin Wang
- Shanghai Health Development Research Center, Shanghai, China.
| |
Collapse
|
38
|
Wu Z, Tang Y, Tang M, Wu Z, Xu Y. The relationship between the eradication of Helicobacter pylori and the occurrence of stomach cancer: an updated meta-analysis and systemic review. BMC Gastroenterol 2025; 25:278. [PMID: 40259215 PMCID: PMC12010618 DOI: 10.1186/s12876-025-03886-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 04/11/2025] [Indexed: 04/23/2025] Open
Abstract
OBJECTIVE Helicobacter pylori (H. pylori) are classified as a Group 1 carcinogen by the International Agency for Research on Cancer (IARC), highlighting its well-established role in gastric carcinogenesis. While previous studies and systematic reviews suggest that H. pylori eradication may lower the incidence and mortality of gastric cancer, the evolving body of evidence necessitates continual reassessment. In light of newly available data, we conducted a comprehensive meta-analysis to evaluate the association between H. pylori eradication therapy and gastric cancer risk, aiming to strengthen the evidence base and inform clinical decision-making. METHOD We systematically searched the Cochrane Library, PubMed, Web of Science, and Embase up to December 2024, including only randomized controlled trials (RCTs) while excluding non-RCT studies. The target population comprised adults diagnosed with H. pylori infection who were either healthy or had previously undergone gastrectomy for gastric tumors. Eradication therapy served as the intervention, while placebo was the control. Eligible studies had a treatment duration exceeding seven days and a follow-up period of more than three years. The Cochrane risk-of-bias tool was used to assess methodological quality, and effect estimates were expressed as relative risk (RR) and the number needed to treat (NNT). OUTCOMES A total of 11 RCTs encompassing 104,786 individuals were analyzed. The meta-analysis revealed that H. pylori eradication significantly reduced gastric cancer risk (RR: 0.61; 95% CI: 0.47-0.79; NNT = 332). Subgroup analysis indicated that among healthy adults, the relative risk (RR) for the occurrence of gastric cancer was 0.67 (95% CI: 0.48-0.93; NNT = 476). In individuals who had undergone endoscopic mucosal resection, the reduction was even more pronounced (RR: 0.51; 95% CI: 0.36-0.71; NNT = 21). Although stomach cancer-specific mortality showed a slight decline (RR: 0.84; 95% CI: 0.69-1.01), all-cause mortality remained statistically unchanged (RR: 1.00; 95% CI: 0.89-1.13). CONCLUSION Our findings support H. pylori eradication as an effective strategy for reducing gastric cancer incidence, particularly in East Asian populations. While the effect on overall mortality remains inconclusive, the observed reduction in gastric cancer-related mortality highlights the potential clinical significance of eradication therapy as a preventive measure. Further well-designed, long-term studies are warranted to reinforce the evidence base and optimize clinical recommendations.
Collapse
Affiliation(s)
- Zhouhan Wu
- Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Yi Tang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), 54 Youdian Road, Hangzhou, 310006, Zhejiang, China
| | - Meiwen Tang
- Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Zhoutong Wu
- Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Yonghui Xu
- Guangxi University of Chinese Medicine, Nanning, 530200, China.
| |
Collapse
|
39
|
Park J, Kim DY, Suh M, Kim YH, Won S. Assessing gastric cancer risk through longitudinal health check-up data: Insights from a national cohort study in South Korea. PLoS One 2025; 20:e0312861. [PMID: 40245012 PMCID: PMC12005563 DOI: 10.1371/journal.pone.0312861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 10/14/2024] [Indexed: 04/19/2025] Open
Abstract
Gastric cancer (GC) is the fourth most prevalent cancer and a leading cause of cancer-related fatalities in South Korea. Although periodic screening policies are in place, the early detection and prediction of GC remain challenging. This study evaluated the risk of GC incidence by utilizing longitudinal health check-up data from the National Health Insurance Service-Health Screening Cohort spanning from 2009 to 2019. The criteria selected for this study are general health examination candidates aged 40 or older who have been eligible for health insurance since 2009. The exclusion criteria included individuals diagnosed with cancer prior to 2009 or before their examination date, as well as those who did not complete the examination questionnaire. A time-dependent Cox proportional hazards model was employed to analyze the time from health examination to the first GC diagnosis, comparing our results with previous cohort studies that evaluated the GC risk through general check-up parameters. Significant risk factors for GC incidence in both genders were age, high levels of AST and γ-GTP, low levels of ALT and hemoglobin. Among males, dyslipidemia, smoking and physical activities were also significantly associated with GC risk. Although further evidence is needed, low hemoglobin levels emerged as a promising potential risk factor for GC, ascertainable through routine general health check-ups.
Collapse
Affiliation(s)
- Juwon Park
- Trend Sensing and Risk Modeling Center, Institute of Quality of Life in Cancer, Samsung Medical Center, Seoul, Republic of Korea
- Department of Public Health Sciences, Seoul National University, Seoul, Republic of Korea
| | - Do-young Kim
- Department of Acupuncture & Moxibustion, Jaseng Korean Medicine Hospital, Seoul, Republic of Korea
| | - Mina Suh
- National Cancer Control Institute, National Cancer Center, Goyang, Republic of Korea
| | - Yeong-Hwa Kim
- Department of Applied Statistics, Chung-Ang University, Seoul, Republic of Korea
| | - Sungho Won
- Department of Public Health Sciences, Seoul National University, Seoul, Republic of Korea
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
- RexSoft Corps, Seoul National University Administration Building, Seoul, Republic of Korea
| |
Collapse
|
40
|
Barua A, Masum MHU, Mahdeen AA. A Reverse Vaccinology and Immunoinformatic Approach for the Designing of a Novel mRNA Vaccine Against Stomach Cancer Targeting the Potent Pathogenic Proteins of Helicobacter pylori. Bioinform Biol Insights 2025; 19:11779322251331104. [PMID: 40290636 PMCID: PMC12033411 DOI: 10.1177/11779322251331104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 03/14/2025] [Indexed: 04/30/2025] Open
Abstract
Helicobacter pylori infection of the stomach's epithelial cells is a significant risk factor for stomach cancer. Various H pylori proteins (CagA, GGT, NapA, PatA, urease, and VacA) were targeted to design 2 messenger RNA (mRNA) vaccines, V1 and V2, using bioinformatics tools. Physicochemical parameters, secondary and tertiary structure, molecular docking and dynamic simulation, codon optimization, and RNA structure prediction have also been estimated for these developed vaccines. Physicochemical analyses revealed that these developed vaccines are soluble (GRAVY < 0), basic (pI < 7), and stable (aliphatic index < 80). The secondary and tertiary structure of the vaccines demonstrated robustness. The docking with toll-like receptors (TLRs) revealed that the vaccines have a potential affinity for TLR-2 (V1: -1132.3 kJ/mol, V2: -1093.6 kJ/mol) and TLR-4 (V1: -1042.7 kJ/mol, V2: -1201.2 kJ/mol), and molecular dynamics simulations confirmed their dynamic stability. Structural analyses of V1 (-505.96 kcal/mol) and V2 (-634.92 kcal/mol) mRNA vaccines underscored their stability. In addition, the vaccine showed a considerable rise in the counts of B cells and extended activation of both T cells was also observed for the vaccines, suggesting the potential for long-lasting immunity, and offering enhanced protection against H pylori. These findings not only suggest potential long-lasting immunity against H pylori but also offer hope for the future of stomach cancer prevention. Notably, the study emphasizes the need for subsequent animal and human-based studies to confirm these promising results.
Collapse
Affiliation(s)
- Abanti Barua
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Md. Habib Ullah Masum
- Department of Genomics and Bioinformatics, Faculty of Biotechnology and Genetic Engineering, Chattogram Veterinary and Animal Sciences University, Khulshi, Chattogram, Bangladesh
| | - Ahmad Abdullah Mahdeen
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| |
Collapse
|
41
|
Alghoul I, Hussain T, Nazir S, Tit N. Efficient detection of gastric cancer biomarkers on functionalized carbon nanoribbons using DFT analysis. Sci Rep 2025; 15:13173. [PMID: 40240795 PMCID: PMC12003821 DOI: 10.1038/s41598-025-97518-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
Early diagnosis of gastric cancer (GC) is crucially important to initiate a therapy plan aiming at rescue and cure. In this regard, the detection of volatile organic compounds (VOCs), related to GC in the patient's exhaled breath, is known to be an efficient and cost-effective technique for early diagnosis. The scope of the present study is to develop a nano-biosensor with great sensitivity and suitable selectivity towards specific VOCs related to GC, such as 2-pentanone, butanone, isoprene, methylglyoxal, N-decanal, N-pentanal, and pyridine. We employed van der Waals corrected density functional theory (DFT) to study the adsorption properties of the mentioned VOCs along with interfering air molecules (N2, O2, H2O, CO2) using recently synthesized carbon nanoribbons (CNRs). We found that pristine CNRs weakly adsorbed the VOCs with adsorption energies ([Formula: see text]), which is not suitable for practical sensing applications. However, the incorporation of selected transition metals (Co, Fe, Mn, Ni) in nitrogen-functionalized CNRs (N-CNRs) enhanced the [Formula: see text] values to -0.802, -0.899, -1.566, -1.260, -1.482, -1.057, and - 0.674 eV for 2-pentanone, butanone, isoprene, methylglyoxal, N-decanal, N-pentanal, and pyridine, respectively. Appropriate [Formula: see text] values along with distinct variations in the electronic and magnetic properties, measured through band structures, density of states, work function and charge transfer analysis, validated the potential of TM-doped N-CNRs as efficient biosensors towards GC-related VOCs. Consequently, the TM-doped N-CNRs are proposed as candidates for platforms of nano biosensors to detect GC biomarkers with high selectivity.
Collapse
Affiliation(s)
- Ibrahim Alghoul
- Department of Physics, College of Science, UAE University, P.O. Box 15551, Al-Ain, United Arab Emirates
- National Water and Energy Center, UAE University, P.O. Box 15551, Al-Ain, United Arab Emirates
| | - Tanveer Hussain
- School of Science and Technology, University of New England, Armidale, NSW, 2351, Australia
| | - Shahid Nazir
- School of Science and Technology, University of New England, Armidale, NSW, 2351, Australia
| | - Nacir Tit
- Department of Physics, College of Science, UAE University, P.O. Box 15551, Al-Ain, United Arab Emirates.
- National Water and Energy Center, UAE University, P.O. Box 15551, Al-Ain, United Arab Emirates.
| |
Collapse
|
42
|
Shen Y, Gao XJ, Zhang XX, Zhao JM, Hu FF, Han JL, Tian WY, Yang M, Wang YF, Lv JL, Zhan Q, An FM. Endoscopists and endoscopic assistants' qualifications, but not their biopsy rates, improve gastric precancerous lesions detection rate. World J Gastrointest Endosc 2025; 17:104097. [PMID: 40291134 PMCID: PMC12019122 DOI: 10.4253/wjge.v17.i4.104097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/27/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Detecting gastric precancerous lesions (GPLs) is critical for the early diagnosis and treatment of gastric cancer. Endoscopy combined with tissue examination is an important method for detecting GPLs. However, negative biopsy results often increase patients' risks, economic burdens, and lead to additional healthcare costs. Improving the detection rate of GPLs and reducing the rate of negative biopsies is currently a key focus in endoscopic quality control. AIM To explore the relationships between the endoscopist biopsy rate (EBR), qualifications of endoscopists and endoscopic assistants, and detection rate of GPLs. METHODS EBR, endoscopists, and endoscopic assistants were divided into four groups: Low, moderate, high, and very high levels. Multivariable logistic regression analysis was used to analyze the relationships between EBR and the qualifications of endoscopists with respect to the detection rate of positive lesions. Pearson and Spearman correlation analyses were used to evaluate the correlation between EBR, endoscopist or endoscopic assistant qualifications, and the detection rate of positive lesions. RESULTS Compared with those in the low EBR group, the odds ratio (OR) values for detecting positive lesions in the moderate, high, and very high EBR groups were 1.12 [95% confidence interval (CI): 1.06-1.19, P < 0.001], 1.22 (95%CI: 1.14-1.31, P < 0.001), and 1.38 (95%CI: 1.29-1.47, P < 0.001), respectively. EBR was positively correlated with the detection rate of gastric precancerous conditions (atrophic gastritis/intestinal metaplasia) (ρ = 0.465, P = 0.004). In contrast, the qualifications of the endoscopists were positively correlated with GPLs detection (ρ = 0.448, P = 0.005). Compared to endoscopists with low qualification levels, those with moderate, high, and very high qualification levels endoscopists demonstrated increased detection rates of GPLs by 13% (OR = 1.13, 95%CI: 0.98-1.31), 20% (OR = 1.20, 95%CI: 1.03-1.39), and 32% (OR = 1.32, 95%CI: 1.15-1.52), respectively. Further analysis revealed that the qualifications of endoscopists were positively correlated with the detection rates of GPLs in the cardia (ρ = 0.350, P = 0.034), angularis (ρ = 0.396, P = 0.015) and gastric body (ρ = 0.453, P = 0.005) but not in the antrum (ρ = 0.292, P = 0.079). Moreover, the experience of endoscopic assistants was positively correlated with the detection rate of precancerous lesions by endoscopists with low or moderate qualifications (ρ = 0.427, P = 0.015). CONCLUSION Endoscopists and endoscopic assistants with high/very high qualifications, but not EBR, can improve the detection rate of GPLs. These results provide reliable evidence for the development of gastroscopic quality control indicators.
Collapse
Affiliation(s)
- Yao Shen
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Xiao-Juan Gao
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Xiao-Xue Zhang
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Jia-Min Zhao
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Fei-Fan Hu
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Jing-Lue Han
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Wen-Ying Tian
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Mei Yang
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Yun-Fei Wang
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Jia-Le Lv
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Qiang Zhan
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| | - Fang-Mei An
- Department of Gastroenterology, Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, National Clinical Research Center for Digestive Diseases (Xi’an) Jiangsu Branch, Wuxi 214023, Jiangsu Province, China
| |
Collapse
|
43
|
Kolstad A, Emanuel G, Hjortland GO, Nilssen Y, Ulvestad M, Areffard A, Aahlin EK. Long-term trends in the clinical management and outcomes of patients with gastroesophageal cancer in Norway. Acta Oncol 2025; 64:540-549. [PMID: 40235057 PMCID: PMC12016665 DOI: 10.2340/1651-226x.2025.43167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/02/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND AND PURPOSE Gastroesophageal cancers are highly prevalent internationally, with many patients diagnosed with metastatic disease, leading to challenging treatment and poor survival. This study uses real-world evidence from a population-level database to describe demographics, clinical characteristics, initial treatment patterns, and survival for patients with gastroesophageal cancer in Norway. MATERIAL AND METHODS Individual patient data was sourced from the Cancer Registry of Norway for patients diagnosed with oesophageal squamous cell carcinoma (ESCC), oesophageal adenocarcinoma (EAC), gastroesophageal junction cancer (GEJC), and gastric cancer from 2001 to 2021, with follow-up from diagnosis to death or last follow-up. Treatment patterns were captured from 2010 to 2022, defined as curative or palliative based on surgery, chemotherapy, and radiotherapy. RESULTS AND INTERPRETATION The cohort included 14,334 Norwegian patients with gastroesophageal cancer; predominantly male, mean age 69-73 years, with a median follow-up of 9-11 months across cancer subtypes. Approximately 40% of patients received curative treatment, and multi-modality treatments increased for EAC, GEJC, and ESCC. Median survival ranged from 6 to 11 months for patients treated palliatively, and 17-95 months for those treated with curative intent. Interestingly, median survival was higher for patients with EAC and GEJC treated with neoadjuvant chemotherapy (86.1 and 75.1 months) versus neoadjuvant chemoradiotherapy (49.1 and 42.1 months), which was confirmed by a multivariate Cox regression model adjusted for age, sex, and disease stage. This study demonstrates that multimodal treatment strategies, consisting of chemotherapy and surgery, may be associated with improved survival outcomes for gastroesophageal cancers. Future studies are required to identify optimum treatment strategies for gastroesophageal cancer subtypes.
Collapse
Affiliation(s)
- Alexander Kolstad
- Department of Gastrointestinal Surgery, University Hospital of North Norway, Tromsø, Norway; Institute of Clinical Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | | | | | - Yngvar Nilssen
- Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
| | | | | | - Eirik Kjus Aahlin
- Department of Gastrointestinal Surgery, University Hospital of North Norway, Tromsø, Norway; Institute of Clinical Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway.
| |
Collapse
|
44
|
Du XY, Xia RJ, Shen LW, Ma JG, Yao WQ, Xu W, Lin ZP, Ma LB, Niu GQ, Fan RF, Xu SM, Yan L. Quadruple therapy with immunotherapy and chemotherapy as first-line conversion treatment for unresectable advanced gastric adenocarcinoma: A case report. World J Gastrointest Oncol 2025; 17:102258. [PMID: 40235902 PMCID: PMC11995329 DOI: 10.4251/wjgo.v17.i4.102258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/20/2025] [Accepted: 02/24/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND The treatment of gastric cancer remains highly challenging, particularly in cases of unresectable locally advanced or metastatic disease. Although chemotherapy and immunotherapy have shown some efficacy in such patients, significant limitations persist in extending survival and enhancing safety. To address these challenges, we designed an innovative first-line quadruple conversion therapy regimen that integrates a programmed cell death protein 1 (PD-1) inhibitor with chemotherapy, and we successfully implemented this therapy regimen in the treatment of a patient with unresectable locally advanced gastric adenocarcinoma. CASE SUMMARY We report the case of a 55-year-old male who was diagnosed with unresectable locally advanced gastric adenocarcinoma and presented with intermittent epigastric pain and multiple lymph node metastases in the abdominal cavity, with the metastasis being notably large in size. The tumor tissue was negative for human epidermal growth factor receptor 2 by immunohistochemistry. Considering the patient's status, the multidisciplinary team decided to administer sintilimab in combination with albumin-bound paclitaxel (nab-paclitaxel), S-1, and oxaliplatin as a quadruple drug conversion therapy. After 4 cycles of conversion therapy, the patient's epigastric pain was significantly alleviated, his stool color normalized, the volume of the primary tumor and lymph node metastases was markedly reduced, and the tumor marker levels decreased to within the normal range. The patient subsequently underwent laparoscopic total gastrectomy with abdominal lymph node dissection, and postoperative pathological biopsy revealed a pathological complete response and R0 resection, after which the patient recovered to an excellent physical status. CONCLUSION To the best of our knowledge, this is the first reported case of unresectable locally advanced gastric adenocarcinoma successfully treated with quadruple therapy with a PD-1 inhibitor and chemotherapy as a first-line conversion regimen. This first-line conversion therapy with the quadruple regimen may be effective and safe for unresectable locally advanced gastric adenocarcinoma.
Collapse
Affiliation(s)
- Xiao-Yu Du
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Medicine, Northwest Minzu University, Lanzhou 730050, Gansu Province, China
| | - Ren-Jie Xia
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Medicine, Northwest Minzu University, Lanzhou 730050, Gansu Province, China
| | - Li-Wen Shen
- Department of Medical Support Center, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Jian-Guo Ma
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730030, Gansu Province, China
| | - Wei-Qing Yao
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Medicine, Northwest Minzu University, Lanzhou 730050, Gansu Province, China
| | - Wei Xu
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Zhi-Peng Lin
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Liang-Bin Ma
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Guo-Qiang Niu
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Rui-Fang Fan
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Shu-Mei Xu
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Long Yan
- Department of Hepatobiliary Surgery and General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| |
Collapse
|
45
|
Serena P, Miutescu B, Gadour E, Burciu C, Mare R, Bende R, Seclăman E, Aragona G, Serena L, Sirli R. Delayed Diagnosis and Evolving Trends in Gastric Cancer During and After COVID-19: A Comparative Study of Staging, Helicobacter pylori Infection and Bleeding Risk in Western Romania. Diagnostics (Basel) 2025; 15:950. [PMID: 40310359 PMCID: PMC12026344 DOI: 10.3390/diagnostics15080950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 05/02/2025] Open
Abstract
Background and Objectives: Gastric cancer (GC) remains a leading cause of cancer mortality worldwide, and the COVID-19 pandemic posed new barriers in diagnosis and management. This study aimed to assess whether pandemic-related healthcare disruptions resulted in more advanced GC stages at presentation. We additionally examined the role of Helicobacter pylori (H. pylori) across non-cardia GC (NCGC) versus cardia GC (CGC) and evaluated the risk factors of upper gastrointestinal (GI) bleeding. Methods: A retrospective cohort of 121 adult patients with GC was enrolled from a tertiary Gastroenterology Unit in Western Romania, spanning pre-pandemic (March 2018-February 2020), pandemic (March 2020-February 2022), and post-pandemic (March 2022-February 2024) periods. Demographic profiles, TNM staging, histopathology, H. pylori status, and clinical outcomes-including GI bleeding-were extracted from medical records. Results: An increase in advanced GC (Stage III-IVB) was noted in the post-pandemic period (69.4% vs. 53.3% pre-pandemic; p = 0.021). H. pylori positivity remained higher in NCGC (70.6%) compared to CGC (44.6%; overall p = 0.041); however, CGC cases showed a rise in H. pylori prevalence post-pandemic (36.4% to 55.6%). One-year mortality was driven by an advanced stage (hazard ratio [HR] = 2.74, p = 0.002), diagnosis during the COVID-19 pandemic (HR = 1.66, p = 0.010), and age ≥70 years (HR = 1.88, p = 0.043). Conclusions: Our findings demonstrate that delayed diagnostic endoscopy correlated with a higher proportion of advanced GC in the post-pandemic phase. H. pylori was strongly linked to NCGC, though CGC showed an increasing trend in H. pylori prevalence. Patients on antithrombotic agents faced increased GI bleeding risks.
Collapse
Affiliation(s)
- Patricia Serena
- Division of Gastroenterology and Hepatology, Department of Internal Medicine II, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (P.S.); (R.M.); (R.B.); (R.S.)
- Advanced Regional Research Center in Gastroenterology and Hepatology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania;
| | - Bogdan Miutescu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine II, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (P.S.); (R.M.); (R.B.); (R.S.)
- Advanced Regional Research Center in Gastroenterology and Hepatology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania;
| | - Eyad Gadour
- Multi-Organ Transplant Centre of Excellence, Liver Transplantation Unit, King Fahad Specialist Hospital, Dammam 32253, Saudi Arabia;
- Department of Medicine, Faculty of Medicine, Zamzam University College, Khartoum 11113, Sudan
| | - Calin Burciu
- Advanced Regional Research Center in Gastroenterology and Hepatology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania;
- Department of Gastroenterology, Faculty of Medicine, Pharmacy and Dental Medicine, “Vasile Goldis” West University of Arad, 310414 Arad, Romania
| | - Ruxandra Mare
- Division of Gastroenterology and Hepatology, Department of Internal Medicine II, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (P.S.); (R.M.); (R.B.); (R.S.)
- Advanced Regional Research Center in Gastroenterology and Hepatology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania;
| | - Renata Bende
- Division of Gastroenterology and Hepatology, Department of Internal Medicine II, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (P.S.); (R.M.); (R.B.); (R.S.)
- Advanced Regional Research Center in Gastroenterology and Hepatology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania;
| | - Edward Seclăman
- Department IV—Biochemistry and Pharmacology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania;
| | - Giovanni Aragona
- Gastroenterology and Hepatology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy;
| | - Luca Serena
- Anaesthesia and Intensive Care Department, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy;
| | - Roxana Sirli
- Division of Gastroenterology and Hepatology, Department of Internal Medicine II, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania; (P.S.); (R.M.); (R.B.); (R.S.)
- Advanced Regional Research Center in Gastroenterology and Hepatology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania;
| |
Collapse
|
46
|
Jung YS, Tran MTX, Park B, Moon CM. Preventive Effect of Helicobacter pylori Treatment on Gastric Cancer Incidence and Mortality: A Korean Population Study. Gastroenterology 2025:S0016-5085(25)00607-9. [PMID: 40209808 DOI: 10.1053/j.gastro.2025.03.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND & AIMS Helicobacter pylori (H pylori) infection is a major risk factor for gastric cancer (GC); however, whether H pylori eradication (HPE) benefits the older population remains unclear. We compared GC incidence and mortality between H pylori-treated individuals and the general population, stratified by age. METHODS We conducted a population-based study in South Korea involving 916,438 individuals aged ≥20 years who underwent HPE therapy between 2009 and 2011, with follow-up until 2021. Standardized incidence ratios (SIRs) and standardized mortality ratios (SMRs) for GC were calculated, comparing H pylori-treated individuals with the general population. RESULTS The mean follow-up period was 12.4 ± 1.1 years. GC incidence and mortality rates were significantly lower in H pylori-treated individuals than in the general population across all age-groups (30-39, 40-49, 50-59, 60-69, and ≥70 years), except for the 20 to 29 years age-group. Notably, in the 70 to 74, 75 to 79, and ≥80 years age-groups, GC incidence and mortality in H pylori-treated individuals remained significantly lower. The SIRs for these groups were 0.56 (95% confidence interval [CI], 0.52-0.61), 0.48 (95% CI, 0.42-0.54), and 0.36 (95% CI, 0.28-0.46), respectively, and the SMRs were 0.30 (95% CI, 0.25-0.35), 0.38 (95% CI, 0.31-0.47), and 0.43 (95% CI, 0.30-0.59), respectively. CONCLUSIONS HPE may help prevent GC and improve survival in adults of all ages, including those aged ≥70 years. These findings suggest that HPE benefits not only younger adults but also older adults. HPE treatment is preferable at a younger age, but older age may not be a limiting factor for the treatment.
Collapse
Affiliation(s)
- Yoon Suk Jung
- Division of Gastroenterology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Mai Thi Xuan Tran
- Department of Preventive Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea; Institute for Health and Society, Hanyang University, Seoul, Republic of Korea
| | - Boyoung Park
- Department of Preventive Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea; Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea.
| | - Chang Mo Moon
- Department of Internal Medicine, Gibbeum Hospital, Seoul, Republic of Korea; Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
47
|
Maturana MJ, Padilla O, Santoro PM, Alarcón MA, Olivares W, Blanco A, Armisen R, Garrido M, Aravena E, Barrientos C, Calvo-Belmar A, Corvalán AH. Methylated Reprimo Cell-Free DNA as a Non-Invasive Biomarker for Gastric Cancer. Int J Mol Sci 2025; 26:3333. [PMID: 40244164 PMCID: PMC11989948 DOI: 10.3390/ijms26073333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/03/2025] [Accepted: 03/08/2025] [Indexed: 04/18/2025] Open
Abstract
Restrictions resulting from the COVID-19 pandemic abruptly reversed the slow decline of the diagnosis and mortality rates of gastric cancer (GC). This scenario highlights the importance of developing cost-effective methods for mass screening and evaluation of treatment response. In this study, we evaluated a non-invasive method based on the circulating methylated cell-free DNA (cfDNA) of Reprimo (RPRM), a tumor suppressor gene associated with the development of GC. Methylated RPRM cfDNA was analyzed in three de-identified cohorts: Cohort 1 comprised 81 participants with GC and 137 healthy donors (HDs); Cohort 2 comprised 27 participants with GC undergoing gastrectomy and/or chemotherapy analyzed at the beginning and after three months of treatment; and Cohort 3 comprised 1105 population-based participants in a secondary prevention program who underwent esophagogastroduodenal (EGD) endoscopy. This cohort includes 180 normal participants, 845 participants with premalignant conditions (692 with chronic atrophic gastritis [AG] and 153 with gastric intestinal metaplasia/low-grade dysplasia [GIM/LGD]), 21 with high-grade dysplasia/early GC [HGD/eGC], and 59 with advanced GC [aGC]). A nested case-control substudy was performed using a combination of methylated RPRM cfDNA and pepsinogens (PG)-I/II ratio. The dense CpG island of the promoter region of the RPRM gene was bisulfite sequenced and analyzed to develop a fluorescence-based real-time PCR assay (MethyLight). This assay allows the determination of the absolute number of copies of methylated RPRM cfDNA. A targeted sequence of PCR amplicon products confirmed the gastric origin of the plasma-isolated samples. In Cohort 1, the mean value of GCs (32,240.00 copies/mL) was higher than that of the HD controls (139.00 copies/mL) (p < 0.0001). After dividing this cohort into training-validation subcohorts, we identified an area under the curve of 0.764 (95% confidence interval (CI) = 0.683-0.845) in the training group. This resulted in a cut-off value of 87.37 copies/mL (sensitivity 70.0% and specificity 80.2%). The validation subcohort predicted a sensitivity of 66.67% and a specificity of 83.33%. In Cohort 2 (monitoring treatment response), RPRM levels significantly decreased in responders (p = 0.0042) compared to non-responders. In Cohort 3 (population-based participants), 18.9% %, 24.1%, 30.7%, 47.0%, and 71.2% of normal, AG, GIM/LGD, HGD/eGC, and aGC participants tested positive for methylated RPRM cfDNA, respectively. Overall sensitivity and specificity in distinguishing normal/premalignant conditions vs. GC were 65.0% (95% CI 53.52% to 75.33%) and 75.9% (95% CI 73.16% to 78.49%), respectively, with an accuracy of 75.11% (95% CI 72.45% to 77.64%). Logistic regression analyses revealed an OR of 1.85 (95% CI 1.11-3.07, p = 0.02) and an odds ratio (OR) of 3.9 (95% CI 1.53-9.93, p = 0.004) for the risk of developing GIM/LGD and HGD/eGC, respectively. The combined methylated RPRM cfDNA and PG-I/II ratio reached a sensitivity of 78.9% (95% CI 54.43% to 93.95%) and specificity of 63.04% (95% CI 52.34% to 72.88%) for detecting HGD/eGC vs. three to six age- and sex-matched participants with premalignant conditions. Our results demonstrate that methylated RPRM cfDNA should be considered a direct biomarker for the non-invasive detection of GC and a predictive biomarker for treatment response.
Collapse
Affiliation(s)
- María José Maturana
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Portugal 61, Santiago 8330023, Chile; (M.J.M.); (P.M.S.); (M.A.A.); (W.O.); (M.G.)
| | - Oslando Padilla
- School of Public Health, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330023, Chile;
| | - Pablo M. Santoro
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Portugal 61, Santiago 8330023, Chile; (M.J.M.); (P.M.S.); (M.A.A.); (W.O.); (M.G.)
| | - Maria Alejandra Alarcón
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Portugal 61, Santiago 8330023, Chile; (M.J.M.); (P.M.S.); (M.A.A.); (W.O.); (M.G.)
| | - Wilda Olivares
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Portugal 61, Santiago 8330023, Chile; (M.J.M.); (P.M.S.); (M.A.A.); (W.O.); (M.G.)
| | - Alejandro Blanco
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago 7550000, Chile; (A.B.); (R.A.)
| | - Ricardo Armisen
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago 7550000, Chile; (A.B.); (R.A.)
| | - Marcelo Garrido
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Portugal 61, Santiago 8330023, Chile; (M.J.M.); (P.M.S.); (M.A.A.); (W.O.); (M.G.)
| | - Edmundo Aravena
- Instituto Chileno Japones de Enfermedades Digestivas, Hospital Clinico San Borja Arriaran, Servicio Salud Metropolitano Central, Santiago, Chile and Fundación Arturo López Pérez, Santiago 8360160, Chile; (E.A.); (C.B.)
| | - Carlos Barrientos
- Instituto Chileno Japones de Enfermedades Digestivas, Hospital Clinico San Borja Arriaran, Servicio Salud Metropolitano Central, Santiago, Chile and Fundación Arturo López Pérez, Santiago 8360160, Chile; (E.A.); (C.B.)
| | - Alfonso Calvo-Belmar
- Hospital Dr. Sotero del Rio, Servicio Salud Metropolitano Sur-Oriente, Santiago 8207257, Chile;
| | - Alejandro H. Corvalán
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Portugal 61, Santiago 8330023, Chile; (M.J.M.); (P.M.S.); (M.A.A.); (W.O.); (M.G.)
| |
Collapse
|
48
|
Ma H, Srivastava S, Ho SWT, Xu C, Lian BSX, Ong X, Tay ST, Sheng T, Lum HYJ, Abdul Ghani SAB, Chu Y, Huang KK, Goh YT, Lee M, Hagihara T, Ng CSY, Tan ALK, Zhang Y, Ding Z, Zhu F, Ng MSW, Joseph CRC, Chen H, Li Z, Zhao JJ, Rha SY, Teh M, Yeong J, Yong WP, So JBY, Sundar R, Tan P. Spatially Resolved Tumor Ecosystems and Cell States in Gastric Adenocarcinoma Progression and Evolution. Cancer Discov 2025; 15:767-792. [PMID: 39774838 PMCID: PMC11962405 DOI: 10.1158/2159-8290.cd-24-0605] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 10/17/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025]
Abstract
SIGNIFICANCE Integration of spatial transcriptomic (GeoMx Digital Spatial Profiler) and single-cell RNA sequencing data from multiple gastric cancers identifies spatially resolved expression-based intratumoral heterogeneity, associated with distinct immune microenvironments. We uncovered two separate evolutionary trajectories associated with specific molecular subtypes, clinical prognoses, stromal neighborhoods, and genetic drivers. Tumor-stroma interfaces emerged as a unique state of tumor ecology.
Collapse
Affiliation(s)
- Haoran Ma
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Supriya Srivastava
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shamaine Wei Ting Ho
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Chang Xu
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | | | - Xuewen Ong
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Su Ting Tay
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Taotao Sheng
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | | | | | - Yunqiang Chu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Kie Kyon Huang
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Yeek Teck Goh
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Minghui Lee
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Takeshi Hagihara
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Clara Shi Ya Ng
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Angie Lay Keng Tan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Yanrong Zhang
- Department of Information Systems and Analytics, School of Computing, National University of Singapore, Singapore, Singapore
| | - Zichen Ding
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Zhu
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Michelle Shu Wen Ng
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Craig Ryan Cecil Joseph
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Hui Chen
- MGI Tech Singapore Pte. Ltd., Singapore, Singapore
| | - Zhen Li
- MGI Tech Singapore Pte. Ltd., Singapore, Singapore
| | - Joseph J. Zhao
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Sun Young Rha
- Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ming Teh
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Joe Yeong
- Department of Pathology, National University Hospital, Singapore, Singapore
- Bioinformatics Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | - Wei Peng Yong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
| | - Jimmy Bok-Yan So
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
- Department of Surgery, University Surgical Cluster, National University Health System, Singapore, Singapore
- Division of Surgical Oncology, National University Cancer Institute, Singapore, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Raghav Sundar
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore
| | - Patrick Tan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cellular and Molecular Research, National Cancer Centre, Singapore, Singapore
- Singhealth/Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore, Singapore
| |
Collapse
|
49
|
Jeong SA, Lee JS, Seong BO, Oh SG, Ko CS, Min SH, Gong CS, Kim BS, Yoo MW, Yook JH, Lee IS, the Information Committee of the Korean Gastric Cancer Association. Proposal of age definition for early-onset gastric cancer based on the Korean Gastric Cancer Association nationwide survey data: a retrospective observational study. Ann Surg Treat Res 2025; 108:245-255. [PMID: 40226172 PMCID: PMC11982448 DOI: 10.4174/astr.2025.108.4.245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/30/2024] [Accepted: 01/07/2025] [Indexed: 04/15/2025] Open
Abstract
Purpose This study aimed to define an optimal age cutoff for early-onset gastric cancer (EOGC) and compare its characteristics with those of late-onset gastric cancer (LOGC) using nationwide survey data. Methods Using data from a nationwide survey, this comprehensive population-based study analyzed data spanning 3 years (2009, 2014, and 2019). The joinpoint analysis and interrupted time series (ITS) methodology were employed to identify age cutoffs for EOGC based on the sex ratio and tumor histology. Clinicopathologic characteristics and surgical outcomes were compared between the EOGC and LOGC groups. Results The age cutoff for defining EOGC was suggested to be 50 years, supported by joinpoint and ITS analyses. Early gastric cancer was predominantly present in the EOGC and LOGC groups. Patients with EOGC comprised 20.3% of the total study cohort and demonstrated a more advanced disease stage compared to patients with LOGC. However, patients with EOGC underwent more minimally invasive surgeries, experienced shorter hospital stays, and had lower postoperative morbidity and mortality rates. Conclusion This study proposes an age of ≤50 years as a criterion for defining EOGC and highlights its features compared to LOGC. Further research using this criterion should guide tailored treatment strategies and improve outcomes for young patients with gastric cancer.
Collapse
Affiliation(s)
- Seong-A Jeong
- Division of Gastrointestinal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Department of Surgery, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Korea
| | - Ji Sung Lee
- Department of Clinical Epidemiology and Biostatistics, University of Ulsan College of Medicine, Seoul, Korea
| | - Ba Ool Seong
- Division of Gastrointestinal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seul-gi Oh
- Department of Surgery, Inha University Hospital, Inha University College of Medicine, Incheon, Korea
| | - Chang Seok Ko
- Division of Gastrointestinal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sa-Hong Min
- Division of Gastrointestinal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chung Sik Gong
- Division of Gastrointestinal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Beom Su Kim
- Division of Gastrointestinal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Moon-Won Yoo
- Division of Gastrointestinal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jeong Hwan Yook
- Division of Gastrointestinal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - In-Seob Lee
- Division of Gastrointestinal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | |
Collapse
|
50
|
Kim JC, Lee MJ, Lee HJ, Park K, Kang MK, Kim SH, Zhuang C, Almayouf A, Bernardo MJC, Kim J, Cho YS, Kong SH, Cho SJ, Park DJ, Yang HK. Is Braun Jejunojejunostomy Necessary? Comparison Between Billroth-II Alone and Billroth-II With Braun Anastomosis After Distal Gastrectomy. J Gastric Cancer 2025; 25:318-329. [PMID: 40200875 PMCID: PMC11982509 DOI: 10.5230/jgc.2025.25.e13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/16/2024] [Accepted: 12/02/2024] [Indexed: 04/10/2025] Open
Abstract
PURPOSE The optimal reconstruction method following distal gastrectomy has not been elucidated. Since Billroth-II (B-II) reconstruction is commonly associated with increased bile reflux, Braun jejunojejunostomy has been proposed to reduce this complication. MATERIALS AND METHODS We retrospectively analyzed 325 patients with gastric cancer who underwent distal gastrectomy with B-II reconstruction between January 2015 and December 2017, comprising 159 patients without Braun anastomosis and 166 with Braun anastomosis. Outcomes were assessed over three years using annual gastroscopy based on the residual food, gastritis, and bile reflux criteria and the Los Angeles classification for reflux esophagitis. RESULTS In the first postoperative year, the group with Braun anastomosis showed a significant reduction in bile reflux compared to the group without Braun anastomosis (75.9% vs. 86.2%; P=0.019). Moreover, multivariate analysis identified Braun anastomosis as the sole factor associated with this outcome. Additionally, the group with Braun anastomosis had a lower incidence of heartburn (12.0% vs. 20.1%; P=0.047) and reduced use of prokinetics (P<0.001) and acid reducers (P=0.002) compared to the group without Braun anastomosis. However, these benefits diminished in subsequent years, with no significant differences in residual food, gastritis, or reflux esophagitis between the groups. Both groups showed similar body mass index scores and nutritional outcomes over the 3-year follow-up period. CONCLUSIONS Although Braun anastomosis offers short-term benefits in reducing bile reflux after B-II reconstruction, these effects are not sustainable. The routine use of Braun anastomosis should be reconsidered, though either approach remains a viable option depending on the patient's circumstances.
Collapse
Affiliation(s)
- Jane Chungyoon Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Min Jung Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Hyuk-Joon Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.
| | - Kyoyoung Park
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Min Kyu Kang
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Sa-Hong Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Chun Zhuang
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai, China
| | - Abdullah Almayouf
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Department of Surgery, King Saud Hospital, Unaizah, Saudi Arabia
| | - Ma Jeanesse C Bernardo
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Department of Surgery, Chinese General Hospital and Medical Center, Manila, Philippines
| | - Jeesun Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Yo-Seok Cho
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Seong-Ho Kong
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Soo-Jeong Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Do Joong Park
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Han-Kwang Yang
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|