1
|
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. [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: 12/19/2024] [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
|
2
|
Shuman JHB, Lin AS, Westland MD, Bryant KN, Fortier GE, Piazuelo MB, Reyzer ML, Judd AM, Tsui T, McDonald WH, McClain MS, Schey KL, Algood HM, Cover TL. Helicobacter pylori CagA and Cag type IV secretion system activity have key roles in triggering gastric transcriptional and proteomic alterations. Infect Immun 2025; 93:e0059524. [PMID: 40047510 PMCID: PMC11977315 DOI: 10.1128/iai.00595-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/24/2025] [Indexed: 04/09/2025] Open
Abstract
Colonization of the human stomach with cag pathogenicity island (PAI)-positive Helicobacter pylori strains is associated with increased gastric cancer risk compared to colonization with cag PAI-negative strains. To evaluate the contributions of the Cag type IV secretion system (T4SS) and CagA (a secreted bacterial oncoprotein) to gastric molecular alterations relevant for carcinogenesis, we infected Mongolian gerbils with a Cag T4SS-positive wild-type (WT) H. pylori strain, one of two Cag T4SS mutant strains (∆cagT or ∆cagY), or a ∆cagA mutant for 12 weeks. Histologic staining revealed a biphasic distribution of gastric inflammation severity in WT-infected animals and minimal inflammation in animals infected with mutant strains. Atrophic gastritis (a premalignant condition), dysplasia, and gastric adenocarcinoma were only detected in WT-infected animals with high inflammation scores. Transcriptional profiling, liquid chromatography-tandem mass spectrometry analysis of micro-extracted tryptic peptides, and imaging mass spectrometry revealed more than a thousand molecular alterations in gastric tissues from WT-infected animals with high inflammation scores compared to uninfected tissues and few alterations in tissues from other groups of infected animals. Proteins with altered abundance in animals with severe Cag T4SS-induced inflammation mapped to multiple pathways, including the complement/coagulation cascade and proteasome pathway. Proteins exhibiting markedly increased abundance in tissues from H. pylori-infected animals with severe inflammation included calprotectin components, proteins involved in proteasome activation, polymeric immunoglobulin receptor (PIGR), interferon-inducible guanylate-binding protein (GBP2), lactoferrin, lysozyme, superoxide dismutase, and eosinophil peroxidase. These results demonstrate key roles for CagA and Cag T4SS activity in promoting gastric mucosal inflammation, transcriptional alterations, and proteomic alterations relevant to gastric carcinogenesis.IMPORTANCEHelicobacter pylori colonizes the stomachs of about half of humans worldwide, and its presence is the primary risk factor for the development of stomach cancer. H. pylori strains isolated from humans can be broadly classified into two groups based on whether they contain a chromosomal cag pathogenicity island, which encodes a secreted effector protein (CagA) and components of a type IV secretion system (T4SS). In experiments using a Mongolian gerbil model, we found that severe gastric inflammation and gastric transcriptional and proteomic alterations related to gastric cancer development were detected only in animals infected with a wild-type H. pylori strain containing CagA and an intact Cag T4SS. Mutant strains lacking CagA or Cag T4SS activity successfully colonized the stomach without inducing detectable pathologic host responses. These findings illustrate two different patterns of H. pylori-host interaction.
Collapse
Affiliation(s)
- Jennifer H. B. Shuman
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Aung Soe Lin
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mandy D. Westland
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kaeli N. Bryant
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Gabrielle E. Fortier
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - M. Blanca Piazuelo
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Michelle L. Reyzer
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Audra M. Judd
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Tina Tsui
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - W. Hayes McDonald
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Mark S. McClain
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kevin L. Schey
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Holly M. Algood
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Timothy L. Cover
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
3
|
Pittayanon R, Tiankanon K, Faknak N, Lerttanatum N, Sanpavat A, Klaikaew N, Rerknimitr R. Efficacy of Radiofrequency Ablation as a Treatment for High-Risk Gastric Intestinal Metaplasia: A Randomized, Self-Control Study. J Gastroenterol Hepatol 2025; 40:891-899. [PMID: 39762988 DOI: 10.1111/jgh.16875] [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: 07/27/2024] [Revised: 12/02/2024] [Accepted: 12/23/2024] [Indexed: 04/05/2025]
Abstract
BACKGROUND Guidelines recommend endoscopic surveillance for gastric cancer without therapeutic intervention every 3 years in patients with high-risk gastric intestinal metaplasia (GIM). This study aimed to evaluate the efficacy of radiofrequency ablation (RFA) in eradicating high-risk GIM. METHODS This randomized self-control trial was conducted between June 2020 and February 2023. Patients with histology-verified high-risk GIM were enrolled. The endoscopist performed a biopsy on both the left and right sides of the stomach (five each) by targeting the suspected GIM area where available; otherwise, a random biopsy was taken. Patients were randomized to receive a unilateral RFA on either the left or right side. A repeated RFA on the assigned side was performed every 2-3 months for a total of two to three times. The primary outcome was complete resolution of GIM at 1 year after RFA. RESULTS Forty-six patients with a mean age of 66 ± 8 years were analyzed. The complete resolution rate of overall GIM lesions after RFA was significantly higher (49/142; 34/5%) than that in the observation group (29/127; 22.8%, RR = 0.84, 0.73-0.98, p = 0.03). For the subgroup analysis, the complete resolution rate after RFA revealed a significantly higher value than observation only in the incomplete GIM group (24/87; 27.6% vs. 11/82; 13.4%, RR = 0.83, 0.71-0.97, p = 0.02). The percentage of patients with extensive GIM regression after RFA (15/25; 60%) was higher than in the observation group (9/25; 36%) but did not meet statistical significance (RR = 0.62, 0.35-1.09, p = 0.09). CONCLUSION In high-risk GIM, RFA can significantly eradicate incomplete GIM when compared with observation alone.
Collapse
Affiliation(s)
- Rapat Pittayanon
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross, Bangkok, Thailand
- Center of Excellence for Innovation and Endoscopy in Gastrointestinal Oncology, Chulalongkorn University, Bangkok, Thailand
| | - Kasenee Tiankanon
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross, Bangkok, Thailand
- Center of Excellence for Innovation and Endoscopy in Gastrointestinal Oncology, Chulalongkorn University, Bangkok, Thailand
| | - Natee Faknak
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross, Bangkok, Thailand
| | - Nathawadee Lerttanatum
- Department of Pathology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Anapat Sanpavat
- Department of Pathology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Naruemon Klaikaew
- Department of Pathology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Rungsun Rerknimitr
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross, Bangkok, Thailand
- Center of Excellence for Innovation and Endoscopy in Gastrointestinal Oncology, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
4
|
Lyu KM, Chen QQ, Xu YF, Yuan YQ, Wang JF, Wan J, Ling-Hu EQ. Development and validation of a predictive model for the pathological upgrading of gastric low-grade intraepithelial neoplasia. World J Gastroenterol 2025; 31:104377. [PMID: 40124264 PMCID: PMC11924016 DOI: 10.3748/wjg.v31.i11.104377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/17/2025] [Accepted: 02/11/2025] [Indexed: 03/13/2025] Open
Abstract
BACKGROUND The discrepancy between endoscopic biopsy pathology and the overall pathology of gastric low-grade intraepithelial neoplasia (LGIN) presents challenges in developing diagnostic and treatment protocols. AIM To develop a risk prediction model for the pathological upgrading of gastric LGIN to aid clinical diagnosis and treatment. METHODS We retrospectively analyzed data from patients newly diagnosed with gastric LGIN who underwent complete endoscopic resection within 6 months at the First Medical Center of Chinese People's Liberation Army General Hospital between January 2008 and December 2023. A risk prediction model for the pathological progression of gastric LGIN was constructed and evaluated for accuracy and clinical applicability. RESULTS A total of 171 patients were included in this study: 93 patients with high-grade intraepithelial neoplasia or early gastric cancer and 78 with LGIN. The logistic stepwise regression model demonstrated a sensitivity and specificity of 0.868 and 0.800, respectively, while the least absolute shrinkage and selection operator (LASSO) regression model showed sensitivity and specificity values of 0.842 and 0.840, respectively. The area under the curve (AUC) for the logistic model was 0.896, slightly lower than the AUC of 0.904 for the LASSO model. Internal validation with 30% of the data yielded AUC scores of 0.908 for the logistic model and 0.905 for the LASSO model. The LASSO model provided greater utility in clinical decision-making. CONCLUSION A risk prediction model for the pathological upgrading of gastric LGIN based on white-light and magnifying endoscopic features can accurately and effectively guide clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Kun-Ming Lyu
- Department of Gastroenterology, The Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese People’s Liberation Army General Hospital, Beijing 100853, China
- Medical College, Chinese People’s Liberation Army General Hospital, Beijing 100853, China
| | - Qian-Qian Chen
- Department of Gastroenterology, The First Medical Center of Chinese People’s Liberation Army General Hospital, Beijing 100853, China
| | - Yi-Fan Xu
- Department of Gastroenterology, General Hospital of Central Theater Command, Wuhan 430000, Hubei Province, China
| | - Yao-Qian Yuan
- Medical College, Chinese People’s Liberation Army General Hospital, Beijing 100853, China
- Department of Gastroenterology, The First Medical Center of Chinese People’s Liberation Army General Hospital, Beijing 100853, China
| | - Jia-Feng Wang
- Department of Gastroenterology, The First Medical Center of Chinese People’s Liberation Army General Hospital, Beijing 100853, China
| | - Jun Wan
- Department of Gastroenterology, The Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese People’s Liberation Army General Hospital, Beijing 100853, China
| | - En-Qiang Ling-Hu
- Department of Gastroenterology, The First Medical Center of Chinese People’s Liberation Army General Hospital, Beijing 100853, China
| |
Collapse
|
5
|
Kim M, Je Y, Chun J, Youn YH, Park H, Nahm JH, Kim J. Helicobacter pylori Eradication Is Associated With a Reduced Risk of Metachronous Gastric Neoplasia by Restoring Immune Function in the Gastric Mucosa. Helicobacter 2025; 30:e70030. [PMID: 40169366 PMCID: PMC11961346 DOI: 10.1111/hel.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/03/2025]
Abstract
BACKGROUND Helicobacter pylori infection is a significant contributing factor of gastric cancer. Metachronous neoplasms also pose a risk. The mechanism underlying the impact of H. pylori eradication on preventing metachronous gastric cancer is unclear. This study aimed to investigate immunity changes in gastric mucosa after H. pylori eradication and to identify mechanisms preventing metachronous recurrence. MATERIALS AND METHODS Patients diagnosed with gastric neoplasm and H. pylori infection, who underwent endoscopic resection, were included. Thirty-six cases of metachronous neoplasms occurring after eradication (metachronous group) were compared to 36 controls matched for age, sex, atrophy, and metaplasia (control group). Histological features and immunohistochemical staining for T-cell (CD3, CD4, and CD8) and immune exhaustion (forkhead/winged helix transcription factor and programmed cell death-ligand 1) markers in the non-tumor-bearing mucosa were evaluated. RESULTS In histologic features, glandular atrophy and intestinal metaplasia in the gastric mucosa significantly improved following H. pylori eradication in the control group (p < 0.001, 0.008), whereas they did not improve in the metachronous group (p = 0.449, 0.609). CD8 and CD8/CD3 ratios increased in the control group (p < 0.001, 0.04), but did not show differences in the metachronous group (p = 0.057, 0.245). The CD4/CD3 ratio and programmed cell death-ligand 1/CD4 expression significantly decreased after H. pylori eradication in the control group (p = 0.003, 0.042), but not in the metachronous group (p = 0.54, 0.55). CONCLUSIONS This observational study suggests that H. pylori eradication may prevent the recurrence of gastric neoplasia by improving histological inflammation and overcoming immune exhaustion.
Collapse
Affiliation(s)
- Min‐Jae Kim
- Department of Internal Medicine, Gangnam Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Yeonjin Je
- Graduate School of MedicineYonsei UniversitySeoulKorea
| | - Jaeyoung Chun
- Department of Internal Medicine, Gangnam Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Young Hoon Youn
- Department of Internal Medicine, Gangnam Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Hyojin Park
- Department of Internal Medicine, Gangnam Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Ji Hae Nahm
- Department of Pathology, Gangnam Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Jie‐Hyun Kim
- Department of Internal Medicine, Gangnam Severance HospitalYonsei University College of MedicineSeoulKorea
| |
Collapse
|
6
|
Shang C, Zhang Y, Wang Y, Zhao W, Sun X, Dong X, Qiao H. Role of ITGB2 protein structure and molecular mechanism in precancerous lesions of gastric cancer: Influencing the occurrence and development of cancer through the CXCL1-CXCR2 axis. Int J Biol Macromol 2025; 296:139772. [PMID: 39800019 DOI: 10.1016/j.ijbiomac.2025.139772] [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: 11/10/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Gastric cancer is a prevalent gastrointestinal tumor. In the classical cascade of gastric cancer development, the gradual progression from non-atrophic gastritis, atrophic gastritis, intestinal metaplasia, to intraepithelial neoplasia eventually leads to early gastric cancer. We investigated the proteomic characteristics of chronic gastritis (CG), low-grade intraepithelial neoplasia (low-grade LGIN), and early gastric cancer (EGC). Additionally, we utilized transcriptomic databases to explore the expression patterns of ITGB2 across different stages of gastric tissue and its correlation with the prognosis of gastric cancer. The expression of ITGB2 was confirmed in cytological experiments, revealing that ITGB2 can influence the onset and progression of gastric cancer via the CXCL1-CXCR2 axis. This finding suggests that ITGB2 represents a novel biomarker for gastric cancer, making it a potential target for accurate diagnosis and treatment.
Collapse
Affiliation(s)
- Chunyang Shang
- Department of Gastrosplenic surgery, Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| | - Yin Zhang
- Department of General Surgery, Aerospace Center Hospital, Beijing, China; Beijing Aviation General Hospital, Beijing 100012, China
| | - Yangshuai Wang
- Department of Gastrosplenic surgery, Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| | - Wenbin Zhao
- Department of Gastrosplenic surgery, Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| | - Xuepu Sun
- Department of Gastrosplenic surgery, Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| | - Xuesong Dong
- Department of Gastrosplenic surgery, Harbin Medical University, Harbin 150000, Heilongjiang Province, China.
| | - Haiquan Qiao
- Department of Gastrosplenic surgery, Harbin Medical University, Harbin 150000, Heilongjiang Province, China.
| |
Collapse
|
7
|
Voutsadakis IA. The Status of SOX2 Expression in Gastric Cancers with Induction of CDX2 Defines Groups with Different Genomic Landscapes. Genes (Basel) 2025; 16:279. [PMID: 40149431 PMCID: PMC11942492 DOI: 10.3390/genes16030279] [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: 02/10/2025] [Revised: 02/23/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Gastric adenocarcinoma is a highly lethal neoplasm with a short survival especially when metastatic. Few effective treatments are available for the control of the disease and palliation of patients with metastatic gastric cancer. Although progress has been made in the elucidation of molecular pathways invoked in gastric carcinogenesis, this knowledge has not yet led to major breakthroughs, in contrast to several other types of cancer. The role of stem cell transcription factors SOX2 and CDX2 is of particular interest in the pathogenesis of gastric cancer. METHODS The cohort of gastric adenocarcinomas from The Cancer Genome Atlas (TCGA) was interrogated and two groups of gastric cancers, with CDX2 induction and SOX2 suppression on the one hand and with CDX2 induction and SOX2 maintained expression on the other hand were retained. The induction of expression of the two transcription factors was defined as a mRNA expression z score compared with normal samples above zero. The two groups were compared for clinical-pathologic and genomic differences. RESULTS Among gastric cancers with up-regulated CDX2 mRNA, cancers with suppressed SOX2 mRNA were slightly more numerous (55.9%) than those with a maintained SOX2 expression. The SOX2 suppressed group had a higher prevalence of MSI high cancers (30.9% versus 10%) and of cases with high tumor mutation burden (35% versus 12.4%) than cancers with a SOX2 maintained expression, which presented more frequently high Chromosomal Instability (CIN). The group with SOX2 suppression had higher rates of mutations in many gastric cancer-associated genes such as epigenetic modifiers ARID1A, KMT2D, KMT2C, and KMT2B, as well as higher rates of mutations in genes encoding for receptor tyrosine kinases ERBB4 and FGFR1. On the other hand, TP53 mutations and amplifications in MYC, ERBB2, and CCNE1 were more common in the group with a maintained expression of SOX2, approaching significance for MYC. CONCLUSIONS Notable differences are present in the genomic landscape of CDX2-induced gastric cancer depending on the level of expression of SOX2 mRNA. Despite this, SOX2 mRNA expression levels were not prognostic.
Collapse
Affiliation(s)
- Ioannis A. Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, 750 Great Northern Road, Sault Ste. Marie, ON P6B 0A8, Canada; or
- Section of Internal Medicine, Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
8
|
Larios-Serrato V, Valdez-Salazar HA, Torres J, Camorlinga M, Piña-Sánchez P, Minauro F, Ruiz-Tachiquín ME. Analysis of biopsies of gastric cancer, intestinal and diffuse, and non-atrophic gastritis: an overview of loss of heterozygosity in Mexican patients. PeerJ 2025; 13:e18928. [PMID: 40028213 PMCID: PMC11869887 DOI: 10.7717/peerj.18928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 01/13/2025] [Indexed: 03/05/2025] Open
Abstract
This study analyzed the loss of heterozygosity (LOH) effect on gastric cancer (GC) tumor samples from 21 Mexican patients, including diffuse (DGC) and intestinal (IGC) subtypes, as well as non-atrophic gastritis (NAG, control). Whole-genome high-density arrays were performed, and LOH regions were identified among the tissue samples. The differences in affected chromosomes were established among groups, with chromosomes 6 and 8 primarily affected in DGC and chromosomes 3, 16, and 17 in IGC. Functional pathway analysis revealed involvement in cancer-associated processes, such as signal transduction, immune response, and cellular metabolism. Five LOH-genes (IRAK1, IKBKG, PAK3, TKTL1, PRPS1) shared between GC and NAG suggest an early role in carcinogenesis. Specific genes were highlighted for Hallmarks of Cancer NAG-related genes (PTPRJ and NDUFS) were linked to cell proliferation and growth; IGC genes (GNAI2, RHOA, MAPKAPK3, MST1R) to genomic instability, metastasis, and arrest of cell death; and DGC genes to energy metabolism and immune evasion. These findings emphasize the role of LOH in GC pathogenesis and underscore the need for further research to understand LOH-affected genes and their diagnostic or evolution potential in cancer management. Portions of this text were previously published as part of a preprint (https://www.medrxiv.org/content/10.1101/2024.07.29.24311063v1).
Collapse
Affiliation(s)
- Violeta Larios-Serrato
- Laboratorio de Biotecnología y Bioinformática Genómica/Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Hilda A. Valdez-Salazar
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias/Unidad Médica de Alta Especialidad-Hospital de Pediatría ‘Dr. Silvestre Frenk Freund’/Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Javier Torres
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias/Unidad Médica de Alta Especialidad-Hospital de Pediatría ‘Dr. Silvestre Frenk Freund’/Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Margarita Camorlinga
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias/Unidad Médica de Alta Especialidad-Hospital de Pediatría ‘Dr. Silvestre Frenk Freund’/Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Patricia Piña-Sánchez
- Unidad de Investigación Médica en Enfermedades Oncológicas/Unidad Médica de Alta Especialidad-Hospital de Oncología/Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Fernando Minauro
- Unidad de Investigación Médica en Genética Humana/Unidad Médica de Alta Especialidad-Hospital de Pediatría ‘Dr. Silvestre Frenk Freund’/Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Martha-Eugenia Ruiz-Tachiquín
- Unidad de Investigación Médica en Enfermedades Oncológicas/Unidad Médica de Alta Especialidad-Hospital de Oncología/Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| |
Collapse
|
9
|
Sibony-Benyamini H, Jbara R, Shubash Napso T, Abu-Rahmoun L, Vizenblit D, Easton-Mor M, Perez S, Brandis A, Leshem T, Peretz A, Maman Y. The landcape of Helicobacter pylori-mediated DNA breaks links bacterial genotoxicity to its oncogenic potential. Genome Med 2025; 17:14. [PMID: 39994739 PMCID: PMC11853333 DOI: 10.1186/s13073-025-01439-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) infection is a significant risk factor for gastric cancer (GC) development. A growing body of evidence suggests a causal link between infection with H. pylori and increased DNA breakage in the host cells. While several mechanisms have been proposed for this damage, their relative impact on the overall bacterial genotoxicity is unknown. Moreover, the link between the formation of DNA damage following infection and the emergence of cancerous structural variants (SV) in the genome of infected cells remained unexplored. METHODS We constructed a high-resolution map of genomic H. pylori-induced recurrent break sites using the END-seq method on AGS human gastric cells before and after infection. We next applied END-seq to cycling and arrested cells to identify the role of DNA replication on break formation. Recurrent H. pylori-mediated break sites were further characterized by analyzing published RNA-seq, DRIP-seq, and GRO-seq data at these sites. γH2AX staining and comet assay were used for DNA breakage quantification. Liquid chromatography-mass spectrometry (LC-MS) assay was used to quantify cellular concentrations of dNTPs. RESULTS Our data indicated that sites of recurrent H. pylori-mediated DNA breaks are ubiquitous across cell types, localized at replication-related fragile sites, and their breakage is dependent on replication. Consistent with that, we found that H. pylori inflicts nucleotide depletion, and that rescuing the cellular nucleotide pool largely reduced H. pylori-induced DNA breaks. Intriguingly, we found that this genotoxic mechanism operates independently of H. pylori cag pathogenicity island (CagPAI) that encodes for the bacterial type 4 secretion system (T4SS), and its virulence factor, CagA, which was previously implicated in increasing DNA damage by downregulating the DNA damage response. Finally, we show that sites of recurrent H. pylori-mediated breaks coincide with chromosomal deletions observed in patients with intestinal-type GC and that this link potentially elucidates the persistent transcriptional alterations observed in cancer driver genes. CONCLUSIONS Our findings indicate that dNTP depletion by H. pylori is a key component of its genotoxicity and suggest a link between H. pylori genotoxicity and its oncogenic potential.
Collapse
Affiliation(s)
| | - Rose Jbara
- Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Zefat, Israel
| | - Tania Shubash Napso
- Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Zefat, Israel
| | - Layan Abu-Rahmoun
- Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Zefat, Israel
| | - Daniel Vizenblit
- Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Zefat, Israel
- Baruch Padeh Medical Center, Poriya, Israel
| | - Michal Easton-Mor
- Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Zefat, Israel
| | - Shira Perez
- Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Zefat, Israel
| | | | | | - Avi Peretz
- Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Zefat, Israel
- Baruch Padeh Medical Center, Poriya, Israel
| | - Yaakov Maman
- Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Zefat, Israel.
| |
Collapse
|
10
|
Maubach G, Kanthasamy AK, Gogia S, Naumann M. The enigma of maladaptation in gastric pathophysiology. Trends Cancer 2025:S2405-8033(25)00040-8. [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] [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
|
11
|
Xia Q, Hu J, Jiao Z, Wang G, Sun J, Pang X, Ma Y, Huang Y, Liang X, Guo J, Peng C, Jin C, Jia X, Gui S. Exploring the mechanisms of Yang Wei Shu granule for the treatment of chronic atrophic gastritis using UPLC-QTOF-MS/MS, network pharmacology, and cell experimentation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119326. [PMID: 39798675 DOI: 10.1016/j.jep.2025.119326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/25/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chronic atrophic gastritis (CAG) is a global disease of the digestive system and is an important precancerous lesion in the development of gastric cancer. Yang Wei Shu granule (YWSG), which evolved from the formula "Warm Stomach Soup" of the Jin and Yuan Dynasties in China, is frequently used as a classic herbal compound in the treatment of CAG. However, the active ingredients and mechanisms by which it works are not clear. AIM OF THE STUDY To elucidate the chemical composition of YWSG and investigate the potential mechanisms of YWSG on CAG by composition analysis, network pharmacology and cellular experimental studies. MATERIALS AND METHODS The chemical and blood-entry constituents of YWSG were analyzed by ultra-high performance liquid chromatography-Quadrupole tandem time-of-flight mass spectrometry (UPLC-QTOF-MS/MS). Subsequently, potential targets of YWSG for CAG treatment were identified through utilization of publicly available online resources. The YWSG-component-target-pathway network and protein-protein interaction (PPI) network were constructed using Cytoscape software. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of potential targets was performed using the DAVID database. Finally, a cellular model of lipopolysaccharide (LPS)-activated RAW 264.7 macrophages was established and validated by in vitro experiments. RESULTS A total of 150 compounds in YWSG and 47 blood-entry constituents were identified by using UPLC-QTOF-MS/MS. Based on network pharmacology, a total of 132 target genes were identified as being involved in the therapeutic effect of YWSG on CAG. Network pharmacology and molecular docking results suggest that AKT1, PIK3CA, PTPN11, SRC and STAT3 may be potential targets of YWSG for the treatment of CAG. Cellular experiments showed that the YWSG-containing serum had no cytotoxic effect on RAW264.7 cells and could inhibit nitric oxide (NO) production and the expression of pro-inflammatory factors TNF-α, IL-6, and IL-1β. Additionally, it was observed to promote the expression of the anti-inflammatory factor IL-10 in LPS-stimulated RAW264.7 cells. The immunofluorescence results showed that YWSG treated CAG by inhibiting the PI3K-Akt pathway. CONCLUSIONS The application of UPLC-Q-TOF-MS/MS, network pharmacology and cellular experiments provided elucidation to understand the components and mechanisms of the therapeutic effects of YWSG on CAG, providing useful directions for further research.
Collapse
Affiliation(s)
- Qijun Xia
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Jingjing Hu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Zhiyong Jiao
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Guichun Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Jianwen Sun
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Xingyuan Pang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Yuhan Ma
- Hefei China Resources Shenlu Pharmaceutical Co. Ltd, Hefei, 230012, Anhui, China
| | - Yuzhe Huang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, 230012, Anhui, China
| | - Xiao Liang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Jian Guo
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, 230012, Anhui, China
| | - Chengjun Peng
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, 230012, Anhui, China
| | - Cheng Jin
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, 230012, Anhui, China.
| | - Xiaoyi Jia
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
| | - Shuangying Gui
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, 230012, Anhui, China; Anhui Engineering Research Center for Quality Improvement and Utilization of Genuine Chinese Medicinal Materials, Hefei, 230012, Anhui, China; Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
| |
Collapse
|
12
|
Liao W, Wang J, Li Y. Natural products based on Correa's cascade for the treatment of gastric cancer trilogy: Current status and future perspective. J Pharm Anal 2025; 15:101075. [PMID: 39957902 PMCID: PMC11830317 DOI: 10.1016/j.jpha.2024.101075] [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: 04/02/2024] [Revised: 07/18/2024] [Accepted: 08/16/2024] [Indexed: 02/18/2025] Open
Abstract
Gastric carcinoma (GC) is a malignancy with multifactorial involvement, multicellular regulation, and multistage evolution. The classic Correa's cascade of intestinal GC specifies a trilogy of malignant transformation of the gastric mucosa, in which normal gastric mucosa gradually progresses from inactive or chronic active gastritis (Phase I) to gastric precancerous lesions (Phase II) and finally to GC (Phase III). Correa's cascade highlights the evolutionary pattern of GC and the importance of early intervention to prevent malignant transformation of the gastric mucosa. Intervening in early gastric mucosal lesions, i.e., Phase I and II, will be the key strategy to prevent and treat GC. Natural products (NPs) have been an important source for drug development due to abundant sources, tremendous safety, and multiple pharmacodynamic mechanisms. This review is the first to investigate and summarize the multi-step effects and regulatory mechanisms of NPs on the Correa's cascade in gastric carcinogenesis. In phase I, NPs modulate Helicobacter pylori urease activity, motility, adhesion, virulence factors, and drug resistance, thereby inhibiting H. pylori-induced gastric mucosal inflammation and oxidative stress, and facilitating ulcer healing. In Phase II, NPs modulate multiple pathways and mediators regulating gastric mucosal cell cycle, apoptosis, autophagy, and angiogenesis to reverse gastric precancerous lesions. In Phase III, NPs suppress cell proliferation, migration, invasion, angiogenesis, and cancer stem cells, induce apoptosis and autophagy, and enhance chemotherapeutic drug sensitivity for the treatment of GC. In contrast to existing work, we hope to uncover NPs with sequential therapeutic effects on multiple phases of GC development, providing new ideas for gastric cancer prevention, treatment, and drug development.
Collapse
Affiliation(s)
- Wenhao Liao
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, Chongqing Bishan Hospital of Traditional Chinese Medicine, Chongqing, 402760, China
| | - Yuchen Li
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| |
Collapse
|
13
|
Liu Y, Huang T, Wang L, Wang Y, Liu Y, Bai J, Wen X, Li Y, Long K, Zhang H. Traditional Chinese Medicine in the treatment of chronic atrophic gastritis, precancerous lesions and gastric cancer. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118812. [PMID: 39260710 DOI: 10.1016/j.jep.2024.118812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/27/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chronic atrophic gastritis (CAG), precancerous lesions of gastric cancer (PLGC), and gastric cancer (GC), seriously threaten human health. Traditional Chinese medicine (TCM) has been employed in the treatment of chronic diseases for a long time and has shown remarkable efficacy. AIM OF THE STUDY Recently, there has been an increasing use of TCM in treating CAG, PLGC, and GC. The objective of this study is to compile a comprehensive overview of the existing research on the effects and molecular mechanisms of TCM, including formulas, single herbs, and active components. MATERIALS AND METHODS To obtain a comprehensive understanding of traditional use of TCM in treating these diseases, we reviewed ancient books and Chinese literature. In addition, keywords such as "TCM", "CAG", "PLGC", "GC", and "active ingredients" were used to collect modern research on TCM published in databases such as CNKI, Web of Science, and Pubmed up to April 2024. All collected information was then summarized and analyzed. RESULTS This study analyzed 174 articles, which covered the research progress of 20 TCM formulas, 14 single herbs, and 50 active ingredients in treating CAG, PLGC, and GC. Sources, effects, and molecular mechanisms of the TCM were summarized. CONCLUSIONS This article reviews the progress of TCM in the management of CAG, PLGC, and GC, which will provide a foundation for the clinical application and further development of TCM.
Collapse
Affiliation(s)
- Yuxi Liu
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), No.4 Xihuamen, Xi'an, 710003, China.
| | - Tingting Huang
- Northwest University, No. 229 Taibai North Road, Xi'an, 710069, China.
| | - Lu Wang
- Shaanxi University of Chinese Medicine, Middle section of Century Avenue, Xianyang, 712046, China.
| | - Yuan Wang
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), No.4 Xihuamen, Xi'an, 710003, China.
| | - Yang Liu
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), No.4 Xihuamen, Xi'an, 710003, China.
| | - Jingyi Bai
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), No.4 Xihuamen, Xi'an, 710003, China.
| | - Xinli Wen
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), No.4 Xihuamen, Xi'an, 710003, China.
| | - Ye Li
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), No.4 Xihuamen, Xi'an, 710003, China.
| | - Kaihua Long
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), No.4 Xihuamen, Xi'an, 710003, China.
| | - Hong Zhang
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Provincial Hospital of Chinese Medicine), No.4 Xihuamen, Xi'an, 710003, China; Northwest University, No. 229 Taibai North Road, Xi'an, 710069, China; Shaanxi University of Chinese Medicine, Middle section of Century Avenue, Xianyang, 712046, China.
| |
Collapse
|
14
|
George S, Lucero Y, Cabrera C, Zabala Torres B, Fernández L, Mamani N, Lagomarcino A, Aguilera X, O'Ryan M. Protocol for a randomised 'screen-and-treat' Helicobacter pylori eradication trial in 14-18-years-old adolescents residing in three regions of Chile: effectiveness and microbiological host implications. BMJ Open 2025; 15:e084984. [PMID: 39890135 PMCID: PMC11784427 DOI: 10.1136/bmjopen-2024-084984] [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: 02/02/2024] [Accepted: 01/10/2025] [Indexed: 02/03/2025] Open
Abstract
INTRODUCTION Gastric cancer is a major global health concern, being the final stage of a long-term process, primarily associated with Helicobacter pylori (H. pylori) infection. Early childhood acquisition of H. pylori with low spontaneous eradication rates underscores the need for preventive measures. Our previous pilot treatment study revealed high eradication rates, favourable tolerance profile and a decline in serum biomarkers indicative of gastric damage in asymptomatic school-aged children. The purpose of this study is to determine the potential benefit of a 'screen-and-treat' strategy targeting persistently infected, asymptomatic adolescents. Specific aims are to assess eradication efficacy, its clinical and molecular outcomes and potential clinical and microbiological side effects. METHODS AND ANALYSIS The screening phase will involve testing 500-1000 asymptomatic adolescents aged 14-18 from three cities in Chile using the urea breath test (UBT) to identify 210 participants with persistent infection. They will proceed to a randomised, non-blinded, controlled trial, receiving either a sequential eradication scheme for H. pylori or no treatment. Follow-up will span up to 24 months post-treatment, involving UBT, gastroenterological assessments and blood and stool sample collections. Concurrently, a subset of 60 uninfected adolescents will undergo matched follow-up. Enzyme-linked immunosorbent assay (ELISA) commercial kits will evaluate gastric damage biomarkers in serum (pepsinogen I and II, gastrin-17, VCAM-1, CXCL13). Stool samples will be employed for Escherichia coli and Enterococcus spp-culture, assessing AMR via the disk diffusion method. H. pylori clarithromycin resistance will be determined by molecular method from stool samples. The gut microbiome will be characterised by amplifying and sequencing the 16S rRNA gene from stool samples, followed by bioinformatics analysis. ETHICS AND DISSEMINATION Approved by the Human Research Ethics Committee at the Faculty of Medicine, University of Chile (073-2022). Findings will be disseminated in peer-reviewed journals and scientific meetings to guide future practices. TRIAL REGISTRATION NUMBER NCT05926804.
Collapse
Affiliation(s)
- Sergio George
- Department of Pediatrics and Pediatric Surgery (Eastern Campus), Luis Calvo Mackenna Hospital, Faculty of Medicine, Universidad de Chile, Santiago de Chile, Chile
| | - Yalda Lucero
- Department of Pediatrics and Pediatric Surgery (Northern Campus), Dr. Roberto del Río Hospital, Faculty of Medicine, Universidad de Chile, Santiago de Chile, Chile
- Microbiology and Mycology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago de Chile, Chile
| | - Camila Cabrera
- Microbiology and Mycology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago de Chile, Chile
| | - Beatriz Zabala Torres
- Campus Lillo, Universidad de Aysén, Coyhaique, Chile
- Department of Biological and Chemical Sciences, Faculty of Medicine and Science, Universidad San Sebastián, Valdivia, Chile
| | - Lilian Fernández
- Department of Pediatrics and Pediatric Surgery, Universidad de La Frontera, Temuco, Araucania Region, Chile
| | - Nora Mamani
- Microbiology and Mycology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago de Chile, Chile
| | - Anne Lagomarcino
- Microbiology and Mycology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago de Chile, Chile
| | - Ximena Aguilera
- Monroe Carrell Children's Hospital, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Miguel O'Ryan
- Microbiology and Mycology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago de Chile, Chile
- Instituto Sistemas Complejos de Ingeniería (ISCI), Santiago, Chile
| |
Collapse
|
15
|
Cui X, Chang M, Wang Y, Liu J, Sun Z, Sun Q, Sun Y, Ren J, Li W. Helicobacter pylori reduces METTL14-mediated VAMP3 m 6A modification and promotes the development of gastric cancer by regulating LC3C-mediated c-Met recycling. Cell Death Discov 2025; 11:13. [PMID: 39827141 PMCID: PMC11742886 DOI: 10.1038/s41420-025-02289-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/12/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
Helicobacter pylori (H. pylori) plays an important role in the malignant transformation of the gastric mucosa from chronic inflammation to cancer. However, the mechanisms underlying the epigenetic regulation of gastric carcinogenesis mediated by H. pylori remain unclear. Here, we uncover that H. pylori inhibits METTL14 by upregulating ATF3. METTL14 inhibits gastric cancer (GC) cell proliferation and metastasis in vitro and in vivo. Downregulation of METTL14 inhibits Vesicle-associated membrane protein-3 (VAMP3) by reducing the m6A modification level of VAMP3 mRNA and the stability of IGF2BP2-dependent mRNA. H. pylori also accelerates the malignant progression of GC by regulating VAMP3/LC3C-mediated c-Met recycling. Moreover, the expression of METTL14 and VAMP3 in Hp+ chronic gastritis tissues is much lower than that in Hp- chronic gastritis tissues. METTL14 and VAMP3 expression levels are downregulated notably in cancerous tissues of patients with GC. Therefore, our results show a novel METTL14-VAMP3-LC3C-c-Met signalling axis in the GC development mediated by H. pylori infection, which reveals a novel m6A epigenetic modification mechanism for GC and provides potential prognostic biomarkers for GC progression.
Collapse
Affiliation(s)
- Xixi Cui
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Mingjie Chang
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Yuqiong Wang
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Jiayi Liu
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Zenghui Sun
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Qiyu Sun
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Yundong Sun
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Juchao Ren
- Department of Urology, Qilu Hospital, Shandong University, Jinan, PR China
| | - Wenjuan Li
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China.
| |
Collapse
|
16
|
Li J, Chen X, Mao C, Xiong M, Ma Z, Zhu J, Li X, Chen W, Ma H, Ye X. Epiberberine ameliorates MNNG-induced chronic atrophic gastritis by acting on the EGFR-IL33 axis. Int Immunopharmacol 2025; 145:113718. [PMID: 39642571 DOI: 10.1016/j.intimp.2024.113718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 12/09/2024]
Abstract
Chronic atrophic gastritis (CAG) is a prevalent form of chronic gastritis that presents with chronic inflammation of the gastric mucosa, localised gastric mucosal glandular atrophy and intestinal metaplasia. Despite the existence of diagnostic criteria, effective therapeutic strategies for this condition remain to be developed. The objective of this study was to examine the potential therapeutic benefits of epiberberine in mitigating MNNG-induced CAG and to elucidate the underlying mechanisms. MNNG was employed to establish a CAG mouse model and a GES-1 cell model, and EPI was observed to be efficacious in ameliorating the gastric mucosal injury and inflammatory infiltration induced by MNNG in the CAG model mice, a finding that was subsequently validated in the GES-1 model cells. Bioinformatics analysis indicated that EPI may exert a direct effect on EGFR, thereby regulating the expression of IL-33 and thereby achieving the therapeutic effect of CAG. This hypothesis was also validated by molecular docking prediction, CETSA, and overexpression of EGFR in GES-1 model cells, using EGFR agonists and inhibitors to further demonstrate that EPI may act as an antagonist supplement to EGFR for the treatment of CAG.
Collapse
Affiliation(s)
- Juan Li
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China; College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Xiantao Chen
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Changxia Mao
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Mengyuan Xiong
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Zhengcai Ma
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Jianyu Zhu
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Xuegang Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China.
| | - Wanqun Chen
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400000, China.
| | - Hang Ma
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China.
| | - Xiaoli Ye
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| |
Collapse
|
17
|
He L, Zhang X, Zhang S, Wang Y, Hu W, Li J, Liu Y, Liao Y, Peng X, Li J, Zhao H, Wang L, Lv Y, Hu C, Yang S. H. Pylori-Facilitated TERT/Wnt/β-Catenin Triggers Spasmolytic Polypeptide-Expressing Metaplasia and Oxyntic Atrophy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2401227. [PMID: 39587848 PMCID: PMC11744579 DOI: 10.1002/advs.202401227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 10/23/2024] [Indexed: 11/27/2024]
Abstract
Persistent H. pylori infection triggers the repair program of the mucosa, such as spasmolytic polypeptide-expressing metaplasia (SPEM). However, the mechanism underlying the initiation of SPEM in gastric tissues by H. pylori remains unclear. Here, an increase in telomerase reverse transcriptase (TERT) protein expression is observed in chief cells upon infection with cagA-positive H. pylori. Tert knockout significantly ameliorated H. pylori-induced SPEM and single-cell RNA sequencing demonstrated that the Wnt/β-Catenin pathway is suppressed in gastric cells with Tert knockout. Mechanism study revealed that CagA elevated TERT abundance by disrupting the interaction between TERT and its novel E3 ligase, SYVN1. Interestingly, Nitazoxanide effectively relieved SPEM via inhibition of the Wnt/β-Catenin signaling in vivo. This results clarified the mechanism underlying which CagA activated the TERT/Wnt/β-Catenin pathway, thus promoting the dedifferentiation of chief cells and the occurrence of SPEM in gastric mucosa. This highlights a molecular basis for targeting CagA-activated Wnt signaling in chief cells for the treatment of gastric precancerous lesions.
Collapse
Affiliation(s)
- Lijiao He
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Xiao Zhang
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
- Cancer Center of Daping HospitalArmy Medical UniversityChongqing400000China
| | - Shengwei Zhang
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
- Department of GastroenterologyThe 987th Hospital of the Joint Logistics Support Force of the People's Liberation Army of China, BaojiShaanxi721000China
| | - Yi Wang
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
- Biological Science Research CenterSouthwest UniversityChongqing400715China
| | - Weichao Hu
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Jie Li
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Yunyi Liu
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Yu Liao
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Xue Peng
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Jianjun Li
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Haiyan Zhao
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Liting Wang
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
- Central LaboratoryArmy Medical UniversityChongqing400038China
| | - Yang‐Fan Lv
- Department of PathologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Chang‐Jiang Hu
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Shi‐Ming Yang
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| |
Collapse
|
18
|
Mandal S, Baker AM, Graham TA, Bräutigam K. The tumour histopathology "glossary" for AI developers. PLoS Comput Biol 2025; 21:e1012708. [PMID: 39847582 PMCID: PMC11756763 DOI: 10.1371/journal.pcbi.1012708] [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] [Indexed: 01/25/2025] Open
Abstract
The applications of artificial intelligence (AI) and deep learning (DL) are leading to significant advances in cancer research, particularly in analysing histopathology images for prognostic and treatment-predictive insights. However, effective translation of these computational methods requires computational researchers to have at least a basic understanding of histopathology. In this work, we aim to bridge that gap by introducing essential histopathology concepts to support AI developers in their research. We cover the defining features of key cell types, including epithelial, stromal, and immune cells. The concepts of malignancy, precursor lesions, and the tumour microenvironment (TME) are discussed and illustrated. To enhance understanding, we also introduce foundational histopathology techniques, such as conventional staining with hematoxylin and eosin (HE), antibody staining by immunohistochemistry, and including the new multiplexed antibody staining methods. By providing this essential knowledge to the computational community, we aim to accelerate the development of AI algorithms for cancer research.
Collapse
Affiliation(s)
- Soham Mandal
- Centre for Evolution and Cancer, Institute of Cancer Research, London, United Kingdom
- Data Science Team, Institute of Cancer Research, London, United Kingdom
| | - Ann-Marie Baker
- Centre for Evolution and Cancer, Institute of Cancer Research, London, United Kingdom
| | - Trevor A. Graham
- Centre for Evolution and Cancer, Institute of Cancer Research, London, United Kingdom
| | - Konstantin Bräutigam
- Centre for Evolution and Cancer, Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
19
|
Fansiwala K, Lewis MS, Pisegna JR. Gastritis: Pathophysiology and Clinical Management. Am J Gastroenterol 2025; 120:5-8. [PMID: 39569875 PMCID: PMC11695143 DOI: 10.14309/ajg.0000000000003216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/25/2024] [Indexed: 11/22/2024]
Affiliation(s)
- Kush Fansiwala
- Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Michael S. Lewis
- Departments of Medicine and Pathology, Cedars Sinai Medical Center, Los Angeles, CA, 90048, USA
- Department of Pathology, VA Greater Los Angeles Medical Center, Los Angeles, CA, 90073, USA
| | - Joseph R. Pisegna
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Dept of Medicine VA Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
- Department of Human Nutrition David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| |
Collapse
|
20
|
Yang B, Xie X, Jin X, Huang X, He Y, Yin K, Ji C, Liu L, Feng Z. Identification and validation of serum MUC17 as a non-invasive early warning biomarker for screening of gastric intraepithelial neoplasia. Transl Oncol 2025; 51:102207. [PMID: 39580962 PMCID: PMC11625214 DOI: 10.1016/j.tranon.2024.102207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND The early diagnosis and treatment of Gastric Intraepithelial Neoplasia (GIN) are pivotal for improving the survival rates of patients with gastric cancer (GC). Regrettably, reliable noninvasive biomarkers for GIN screening are currently lacking. METHODS mRNA data from the GEO database, pan-cancer data from the TCGA database, and a gene list of exocrine proteins were subjected to integrated analysis to identify a noninvasive biomarker for GIN. The scRNA-seq data analysis, IHC and Elisa were employed to validate the expression of the biomarker in the serum and tissues of clinical patients across different pathological stages. RESULTS MUC17 has been identified as a non-invasive diagnostic marker for GIN. It is upregulated in GIN prior to the onset of gastric carcinogenesis and downregulated in other tumors, with high GC specificity. The area under the curve values of serum MUC17 for differentiating chronic gastritis (CG) from low-grade intraepithelial neoplasia (LGIN), high-grade intraepithelial neoplasia (HGIN), and early gastric cancer (EGC) were 0.8788, 0.8544, and 0.9513, respectively. Additionally, low plasma MUC17 levels were found to be significantly lower in gastric ulcer (GU), gastric neuroendocrine tumor (GNET), and gastrointestinal stromal tumor (GIST) compared to GIN. The AUC for differentiating between GIN and GU, GNET, or GIST was 0.7803, 0.9244 and 0.9796, respectively. CONCLUSIONS These findings suggest that plasma MUC17 levels hold substantial promise as a screening biomarker for individuals with GIN and EGC, effectively identifying high-risk groups that necessitate further gastroscopy.
Collapse
Affiliation(s)
- Bingxue Yang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China
| | - Xiaoli Xie
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China
| | - Xiaoxu Jin
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China
| | - Xiuhong Huang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China
| | - Yujian He
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China
| | - Kaige Yin
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China
| | - Chenguang Ji
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China
| | - Li Liu
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China
| | - Zhijie Feng
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, PR China.
| |
Collapse
|
21
|
Zaramella A, Arcidiacono D, Duci M, Benna C, Pucciarelli S, Fantin A, Rosato A, De Re V, Cannizzaro R, Fassan M, Realdon S. Predictive Value of a Gastric Microbiota Dysbiosis Test for Stratifying Cancer Risk in Atrophic Gastritis Patients. Nutrients 2024; 17:142. [PMID: 39796578 PMCID: PMC11722812 DOI: 10.3390/nu17010142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/24/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Gastric cancer (GC) incidence remains high worldwide, and the survival rate is poor. GC develops from atrophic gastritis (AG), associated with Helicobacter pylori (Hp) infection, passing through intestinal metaplasia and dysplasia steps. Since Hp eradication does not exclude GC development, further investigations are needed. New data suggest the possible role of unexplored gastric microbiota beyond Hp in the progression from AG to GC. Aimed to develop a score that could be used in clinical practice to stratify GC progression risk, here was investigate gastric microbiota in AG Hp-negative patients with or without high-grade dysplasia (HGD) or GC. METHODS Consecutive patients undergoing upper endoscopy within an endoscopic follow-up for AG were considered. The antrum and corpus biopsies were used to assess the microbiota composition along the disease progression by sequencing the 16S ribosomal RNA gene. Statistical differences between HGD/GC and AG patients were included in a multivariate analysis. RESULTS HGD/GC patients had a higher percentage of Bacillus in the antrum and a low abundance of Rhizobiales, Weeksellaceae and Veillonella in the corpus. These data were used to calculate a multiparametric score (Resident Gastric Microbiota Dysbiosis Test, RGM-DT) to predict the risk of progression toward HGD/GC. The performance of RGM-DT in discriminating patients with HGD/GC showed a specificity of 88.9%. CONCLUSIONS The microbiome-based risk prediction model for GC could clarify the role of gastric microbiota as a cancer risk biomarker to be used in clinical practice. The proposed test might be used to personalize follow-up program thanks to a better cancer risk stratification.
Collapse
Affiliation(s)
- Alice Zaramella
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padua, Via Giustiniani 2, 35128 Padua, Italy; (A.Z.); (C.B.); (S.P.); (A.R.)
- Gastroenterology Unit, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padua, Italy; (D.A.); (A.F.)
| | - Diletta Arcidiacono
- Gastroenterology Unit, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padua, Italy; (D.A.); (A.F.)
| | - Miriam Duci
- Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy;
- Pediatric Surgery Unit, Division of Women’s and Children’s Health, Padova University Hospital, 35128 Padova, Italy
| | - Clara Benna
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padua, Via Giustiniani 2, 35128 Padua, Italy; (A.Z.); (C.B.); (S.P.); (A.R.)
| | - Salvatore Pucciarelli
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padua, Via Giustiniani 2, 35128 Padua, Italy; (A.Z.); (C.B.); (S.P.); (A.R.)
| | - Alberto Fantin
- Gastroenterology Unit, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padua, Italy; (D.A.); (A.F.)
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padua, Via Giustiniani 2, 35128 Padua, Italy; (A.Z.); (C.B.); (S.P.); (A.R.)
- UOC Immunology and Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padua, Italy
| | - Valli De Re
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy;
| | - Renato Cannizzaro
- Oncological Gastroenterology, Centro di Riferimento Oncologico di Aviano (CRO), National Cancer Institute, IRCCS, 33081 Aviano, Italy;
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Matteo Fassan
- Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padua, Italy;
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, Via Gabelli 61, 35121 Padua, Italy
| | - Stefano Realdon
- Oncological Gastroenterology, Centro di Riferimento Oncologico di Aviano (CRO), National Cancer Institute, IRCCS, 33081 Aviano, Italy;
| |
Collapse
|
22
|
Salahi-Niri A, Nabavi-Rad A, Monaghan TM, Rokkas T, Doulberis M, Sadeghi A, Zali MR, Yamaoka Y, Tacconelli E, Yadegar A. Global prevalence of Helicobacter pylori antibiotic resistance among children in the world health organization regions between 2000 and 2023: a systematic review and meta-analysis. BMC Med 2024; 22:598. [PMID: 39710669 DOI: 10.1186/s12916-024-03816-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 12/11/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Helicobacter pylori infection causes gastritis, peptic ulcers, and gastric cancer. The infection is typically acquired in childhood and persists throughout life. The major impediment to successful therapy is antibiotic resistance. This systematic review and meta-analysis aimed to comprehensively assess the global prevalence of antibiotic resistance in pediatric H. pylori infection. METHODS We performed a systematic search of publication databases that assessed H. pylori resistance rates to clarithromycin, metronidazole, levofloxacin, amoxicillin, and tetracycline in children. The WHO region classification was used to group pooled primary and secondary resistance estimates along with 95% confidence interval (CI). H. pylori antibiotic resistance rates were retrieved and combined with odds ratios (95% CI) to investigate the global prevalence and temporal trends. Subgroup analysis of the prevalence of antibiotic resistance was conducted by country, age groups, and susceptibility testing methods. RESULTS Among 1417 records obtained initially, 152 studies were selected for eligibility assessment after applying exclusion criteria in multiple steps. Ultimately, 63 studies involving 15,953 individuals were included comprising data from 28 countries in 5 WHO regions. The primary resistance rates were metronidazole 35.3% (5482/15,529, 95% CI: 28.7-42.6), clarithromycin 32.6% (5071/15,555, 95% CI: 27.7-37.9), tetracycline 2.1% (148/7033, 95% CI: 1.3-3.6), levofloxacin 13.2% (1091/8271, 95% CI: 9.3-18.4), and amoxicillin 4.8% (495/10305, 95% CI: 2.5-8.8). Raising antibiotic resistance was detected in most WHO regions. CONCLUSIONS The escalating trend of H. pylori antibiotic resistance in children warrants urgent attention globally. National and regional surveillance networks are required for antibiotic stewardship in children infected with H. pylori.
Collapse
Affiliation(s)
- Aryan Salahi-Niri
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Nabavi-Rad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tanya Marie Monaghan
- National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
- Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Theodore Rokkas
- Gastroenterology Clinic, Henry Dunant Hospital, Athens, Greece
- Medical School, European University of Cyprus, Nicosia, Cyprus
| | - Michael Doulberis
- Gastroklinik, Private Gastroenterological Practice, Horgen, 8810, Switzerland
- Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, Aarau, 5001, Switzerland
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Oita, Japan
- Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, Houston, TX, USA
- Research Center for Global and Local Infectious Diseases, Oita University, Oita, Japan
| | - Evelina Tacconelli
- Division of Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Jing Y, Ren M, Li X, Sun X, Xiao Y, Xue J, Liu Z. The Effect of Systemic Immune-Inflammatory Index (SII) and Prognostic Nutritional Index (PNI) in Early Gastric Cancer. J Inflamm Res 2024; 17:10273-10287. [PMID: 39654858 PMCID: PMC11625636 DOI: 10.2147/jir.s499094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 11/26/2024] [Indexed: 12/12/2024] Open
Abstract
Background In recent years, the systemic immune-inflammatory index (SII) and prognostic nutritional index (PNI) have been considered potential predictors of survival outcomes in various solid tumors, including gastric cancer. However, there is a notable lack of research focusing on their prognostic implications specifically in the early stage of gastric cancer. This study aims to investigate the prognostic indicators of early gastric cancer (EGC), including neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), SII, PNI, and lymph node metastasis (LNM). Methods In this retrospective analysis, we examined 490 patients diagnosed with EGC (pT1Nx). The peripheral blood indices of interest were SII, PNI, PLR, and NLR. The receiver operating characteristic (ROC) curves and the area under the ROC curve (AUC) were used to determine optimal cutoff values and prognostic efficacy for each parameter. Additionally, Kaplan-Meier survival curves and multivariate Cox regression models were utilized to delineate independent prognostic factors. Results The optimal cutoff values for SII and PNI were determined as 613.05 and 42.21, respectively. Patients in the low SII (SII-L) group demonstrated significantly higher 5-year Disease-Free Survival (DFS) and Overall Survival (OS) rates of 94.7% and 96.2%, compared to the high SII (SII-H) group (DFS: 78.7%; OS: 81.9%), with both differences proving statistically significant (P < 0.001, P < 0.001). Similarly, patients in the high PNI (PNI-H) group showed superior 5-year DFS (93.3%) and OS rates (95.1%) versus the low PNI (PNI-L) group (DFS: 71.4%; OS: 74.3%), also demonstrating statistical significance (P < 0.001, P < 0.001). Multivariate analysis identified SII, PNI, and LNM as independent prognostic factors for EGC. A combined analysis of SII, PNI, and LNM yielded a C-index of 0.723 (P = 0.008). Conclusion SII, PNI, and LNM are effective markers for predicting the survival outcomes of patients undergoing radical gastrectomy for EGC.
Collapse
Affiliation(s)
- Yaoyao Jing
- Center for GI Cancer Diagnosis and Treatment, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People’s Republic of China
| | - Minghan Ren
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People’s Republic of China
| | - Xiaoxiao Li
- Center for GI Cancer Diagnosis and Treatment, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People’s Republic of China
| | - Xiaoyuan Sun
- Center for GI Cancer Diagnosis and Treatment, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People’s Republic of China
| | - Yan Xiao
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People’s Republic of China
| | - Juan Xue
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People’s Republic of China
| | - Zimin Liu
- Center for GI Cancer Diagnosis and Treatment, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People’s Republic of China
| |
Collapse
|
24
|
Magahis PT, Cornet N, Tang L, Arora K, Hingorani N, King S, Markowitz AJ, Schattner M, Shimada S, Maron SB, Vardhana S, Lumish M, Cercek A, Janjigian YY, Coit D, Mendelsohn RB, Berger MF, Strong VE, Stadler ZK, Laszkowska M. Differences in Ancestry and Presence of Gastric Precursor Lesions in Individuals With Young- and Average-Onset Gastric Cancer. Cancer Med 2024; 13:e70451. [PMID: 39629931 PMCID: PMC11615756 DOI: 10.1002/cam4.70451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 12/08/2024] Open
Abstract
BACKGROUND There has been a paradoxical rise in young-onset gastric cancer (YOGC), defined as gastric cancer (GC) diagnosed before age 50. Precursor lesions may contribute to pathogenesis, though their role in progression to different histologic subtypes is unclear. The impact of self-reported race is also poorly characterized and may be unreliable as a proxy for genetic differences. We aimed to compare differences in histology and genetic ancestry between YOGC and average-onset gastric cancer (AOGC). METHODS This retrospective cohort included all patients with GC at Memorial Sloan Kettering (MSK) from January 2013 to March 2021. Data on demographics, tumor characteristics, and precursor lesions were collected. Genetic ancestry was inferred from MSK-Integrated Mutation Profiling of Actionable Cancer Targets panel. RESULTS Of 1685 individuals with GC, 290 had YOGC. Compared to AOGC, individuals with YOGC tended to be female, Hispanic, foreign-born, and feature diffuse-type histology. YOGC was less likely to have precursor lesions, including intestinal metaplasia (20% vs. 37%, p < 0.01) and dysplasia (4% vs. 14%, p < 0.01). Of 560 patients with ancestry data, 127 had YOGC. Admixed, East Asian, and South Asian ancestries were more likely to present with YOGC while Europeans presented with AOGC. Intestinal metaplasia was enriched among East Asians, maintained when stratifying by histology and GC onset. CONCLUSIONS We observed YOGC was more common in East and South Asians, and while YOGC may be less likely to develop in the setting of precursor lesions these high-risk states may also be enriched in East Asians. Future research is needed to understand drivers behind such differences and outcome disparities given these individuals may be less amenable to endoscopic interventions.
Collapse
Affiliation(s)
| | - Nicole Cornet
- Department of MedicineWeill Cornell MedicineNew YorkNew YorkUSA
| | - Laura Tang
- Department of Pathology and Laboratory MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Kanika Arora
- Department of Pathology and Laboratory MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
- Marie‐Josée and Henry R. Kravis Center for Molecular OncologyMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Neha Hingorani
- Gastroenterology, Hepatology, and Nutrition Service, Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Stephanie King
- Gastroenterology, Hepatology, and Nutrition Service, Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Arnold J. Markowitz
- Department of MedicineWeill Cornell MedicineNew YorkNew YorkUSA
- Gastroenterology, Hepatology, and Nutrition Service, Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Mark Schattner
- Department of MedicineWeill Cornell MedicineNew YorkNew YorkUSA
- Gastroenterology, Hepatology, and Nutrition Service, Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Shoji Shimada
- Gastric and Mixed Tumor Service, Department of SurgeryMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Steven B. Maron
- Department of MedicineWeill Cornell MedicineNew YorkNew YorkUSA
- Gastrointestinal Oncology Service, Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Santosha Vardhana
- Department of MedicineWeill Cornell MedicineNew YorkNew YorkUSA
- Gastrointestinal Oncology Service, Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Melissa Lumish
- Gastrointestinal Oncology Service, Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Andrea Cercek
- Department of MedicineWeill Cornell MedicineNew YorkNew YorkUSA
- Gastrointestinal Oncology Service, Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Yelena Y. Janjigian
- Department of MedicineWeill Cornell MedicineNew YorkNew YorkUSA
- Gastrointestinal Oncology Service, Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Daniel Coit
- Department of MedicineWeill Cornell MedicineNew YorkNew YorkUSA
- Gastric and Mixed Tumor Service, Department of SurgeryMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Robin B. Mendelsohn
- Department of MedicineWeill Cornell MedicineNew YorkNew YorkUSA
- Gastroenterology, Hepatology, and Nutrition Service, Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Michael F. Berger
- Department of Pathology and Laboratory MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
- Marie‐Josée and Henry R. Kravis Center for Molecular OncologyMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Vivian E. Strong
- Department of MedicineWeill Cornell MedicineNew YorkNew YorkUSA
- Gastric and Mixed Tumor Service, Department of SurgeryMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Zsofia K. Stadler
- Gastrointestinal Oncology Service, Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
- Clinical Genetics Service, Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Monika Laszkowska
- Department of MedicineWeill Cornell MedicineNew YorkNew YorkUSA
- Gastroenterology, Hepatology, and Nutrition Service, Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| |
Collapse
|
25
|
Matos TL, Souza PFN, de Moraes MEA, Rabenhorst SHB, Mesquita FP, Montenegro RC. Molecular characterization and biomarker discovery in gastric cancer progression through transcriptome meta-analysis. Comput Biol Med 2024; 183:109276. [PMID: 39447404 DOI: 10.1016/j.compbiomed.2024.109276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/26/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
Gastric cancer (GC) is a leading cause of cancer-related deaths globally. It is a multifactorial, molecularly heterogeneous disease whose carcinogenic patterns are not yet well established, requiring the development of new tools for better understanding and identifying gastric carcinogenesis. From this point of view, this study aims to compare transcriptome profiles from The Cancer Genome Atlas Stomach Adenocarcinoma (TCGA-STAD) and a human-merged dataset to identify potential biomarkers and therapeutic targets. Principal component analysis (PCA) revealed shared and distinct gene expression patterns between datasets. Differential expression analysis identified key genes with altered expression across non-malignant and malignant samples. Six genes, including SERPINE1 and CLDN9, were significantly associated with patient survival. The findings underscore the molecular diversity of GC and highlight novel biomarkers for early diagnosis and therapeutic strategies. Further validation in clinical specimens is necessary.
Collapse
Affiliation(s)
- Thiago Loreto Matos
- Pharmacogenetics Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, 60430-160, Brazil
| | - Pedro Filho Noronha Souza
- Pharmacogenetics Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, 60430-160, Brazil
| | - Maria Elisabete Amaral de Moraes
- Pharmacogenetics Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, 60430-160, Brazil
| | - Silvia Helena Barem Rabenhorst
- Molecular Genetics Laboratory, Department of Pathology and Legal Medicine, Federal University of Ceará, Fortaleza, 60430-160, Brazil
| | - Felipe Pantoja Mesquita
- Pharmacogenetics Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, 60430-160, Brazil.
| | - Raquel Carvalho Montenegro
- Pharmacogenetics Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, 60430-160, Brazil
| |
Collapse
|
26
|
Delgado-Guillena P, Jimeno M, López-Nuñez A, Córdova H, Fernández-Esparrach G. The endoscopic model for gastric carcinogenesis and Helicobacter pylori infection: A potential visual mind-map during gastroscopy examination. GASTROENTEROLOGIA Y HEPATOLOGIA 2024; 47:502214. [PMID: 38844201 DOI: 10.1016/j.gastrohep.2024.502214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/04/2024] [Accepted: 05/24/2024] [Indexed: 06/29/2024]
Abstract
Helicobacter pylori (Hp) is the main trigger of chronic gastric atrophy and the main leading cause of gastric cancer. Hp infects the normal gastric mucosa and can lead to chronic inflammation, glandular atrophy, intestinal metaplasia, dysplasia and finally adenocarcinoma. Chronic inflammation and gastric atrophy associated with Hp infection appear initially in the distal part of the stomach (the antrum) before progressing to the proximal part (the corpus-fundus). In recent years, endoscopic developments have allowed for the characterization of various gastric conditions including the normal mucosa (pyloric/fundic gland pattern and regular arrangement of collecting venules), Hp-related gastritis (Kyoto classification), glandular atrophy (Kimura-Takemoto classification), intestinal metaplasia (Endoscopic Grading of Gastric Intestinal Metaplasia), and dysplasia/adenocarcinoma (Vessel plus Surface classification). Despite being independent classifications, all these scales can be integrated into a single model: the endoscopic model for gastric carcinogenesis. This model would assist endoscopists in comprehending the process of gastric carcinogenesis and conducting a systematic examination during gastroscopy. Having this model in mind would enable endoscopists to promptly recognize the implications of Hp infection and the potential patient's risk of developing gastric cancer.
Collapse
Affiliation(s)
| | - Mireya Jimeno
- Department of Pathology, Hospital of Germans Trias i Pujol, Badalona, Spain
| | | | - Henry Córdova
- Department of Gastroenterology, Hospital Clinic of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain
| | - Gloria Fernández-Esparrach
- Department of Gastroenterology, Hospital Clinic of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain
| |
Collapse
|
27
|
Luo Z, Li W, Zheng W, Shi Y, Ye M, Guo X, Fu K, Yan C, Wang B, Lv B, Mo S, Zhang H, Zhang J, He C, Luo F, Zhang W, Liu J. Elucidating epigenetic landscape of gastric premalignant lesions through genome-wide mapping of 5-hydroxymethylcytosines: A 12-year median follow-up study. Clin Transl Med 2024; 14:e70114. [PMID: 39625179 PMCID: PMC11613102 DOI: 10.1002/ctm2.70114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/04/2024] [Accepted: 11/17/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Epigenetic modifications are crucial in tumourigenesis, yet the changes in novel epigenetic regulators like 5-hydroxymethylcytosines (5hmC) during the evolution of gastric premalignant lesions remain poorly understood. This study aims to investigate the implications of 5hmC in the progression from gastric premalignant lesions to gastric adenocarcinoma (GAC). METHODS To our knowledge, we conducted the largest and longest longitudinal study of a Chinese population with gastric precursor lesions, involving 29,176 patients with gastritis who underwent gastroscopy and biopsy between 2001 and 2015, with follow-up until 1 August, 2022. The median follow-up time was 12.2 years, and the overall GAC incidence rate was 0.82%. Genome-wide mapping of 5hmC in gastric premalignant lesions from a subset of individuals was performed using the 5hmC-Seal assay, including 21 samples that progressed to GAC during follow-up and 48 non-progressed age- and sex-matched controls. RESULTS We identified 213 differentially modified gene bodies, primarily concentrated in pathways related to cell division, cell cycle, energy metabolism, inflammation and tumourigenesis. An exploratory study was conducted to summarize a 5hmC-based epigenetic model for predicting cancer progression using multivariable logistic regression and machine learning. The nine-gene model showed an area under the curve of 87.5% (95% confidence interval: 72%-100%) in the validation samples (one of three), which were set aside before model training. CONCLUSIONS This study is the first to explore the 5hmC molecular landscape in gastric premalignant lesions, suggesting relevant pathways implicated in their evolution to GAC as well as the feasibility of exploiting genome-wide 5hmC mapping in assessing the risk of future cancer progression. KEY POINTS A largest longitudinal follow-up study of gastric precursor lesions in Chinese patients. Revealing novel 5hmC molecular landscape linked to gastric premalignant lesions. The feasibility of an innovative 5hmC-based predictive model for assessing gastric cancer progression risk.
Collapse
Affiliation(s)
- Zhongguang Luo
- Department of Digestive DiseasesHuashan HospitalFudan UniversityShanghaiChina
| | - Wenshuai Li
- Department of Digestive DiseasesHuashan HospitalFudan UniversityShanghaiChina
| | - Wanwei Zheng
- Department of Digestive DiseasesHuashan HospitalFudan UniversityShanghaiChina
| | - Yixiang Shi
- Bionova (Shanghai) Medical Technology Co., Ltd.ShanghaiChina
| | - Maolin Ye
- Department of Digestive DiseasesHuashan HospitalFudan UniversityShanghaiChina
| | - Xiangyu Guo
- Department of Digestive DiseasesHuashan HospitalFudan UniversityShanghaiChina
| | - Kaiyi Fu
- Department of Digestive DiseasesHuashan HospitalFudan UniversityShanghaiChina
| | - Changsheng Yan
- Department of Digestive DiseasesHuashan HospitalFudan UniversityShanghaiChina
| | - Bowen Wang
- Bionova (Shanghai) Medical Technology Co., Ltd.ShanghaiChina
| | - Bin Lv
- Department of Digestive DiseasesHuashan HospitalFudan UniversityShanghaiChina
| | - Shaocong Mo
- Department of Digestive DiseasesHuashan HospitalFudan UniversityShanghaiChina
| | - Hongyang Zhang
- Department of Digestive DiseasesHuashan HospitalFudan UniversityShanghaiChina
| | - Jun Zhang
- Department of Digestive DiseasesHuashan HospitalFudan UniversityShanghaiChina
| | - Chuan He
- Department of Chemistry and The Howard Hughes Medical InstituteThe University of ChicagoChicagoIllinoisUSA
| | - Feifei Luo
- Department of Digestive DiseasesHuashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Centre for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
| | - Wei Zhang
- Department of Preventive Medicine and The Robert h. Lurie Comprehensive Cancer CenterNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Jie Liu
- Department of Digestive DiseasesHuashan HospitalFudan UniversityShanghaiChina
- National Clinical Research Centre for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
28
|
Bhatnagar K, Jha K, Dalal N, Patki N, Gupta G, Kumar A, Kumar A, Chaudhary S. Exploring micronutrients and microbiome synergy: pioneering new paths in cancer therapy. Front Immunol 2024; 15:1442788. [PMID: 39676876 PMCID: PMC11638209 DOI: 10.3389/fimmu.2024.1442788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
The human microbiome is the complex ecosystem consisting of trillions of microorganisms that play a key role in developing the immune system and nutrient metabolism. Alterations in the gut microbiome have been linked to cancer initiation, progression, metastasis, and response to treatment. Accumulating evidence suggests that levels of vitamins and minerals influence the gut environment and may have implications for cancer risk and progression. Bifidobacterium has been reported to reduce the colorectal cancer risk by binding to free iron. Additionally, zinc ions have been shown to activate the immune cells and enhance the effectiveness of immunotherapy. Higher selenium levels have been associated with a reduced risk of several cancers, including colorectal cancer. In contrast, enhanced copper uptake has been implicated in promoting cancer progression, including colon cancer. The interaction between cancer and gut bacteria, as well as dysbiosis impact has been studied in animal models. The interplay between prebiotics, probiotics, synbiotics, postbiotics and gut bacteria in cancer offers the diverse physiological benefits. We also explored the particular probiotic formulations like VSL#3, Prohep, Lactobacillus rhamnosus GG (LGG), etc., for their ability to modulate immune responses and reduce tumor burden in preclinical models. Targeting the gut microbiome through antibiotics, bacteriophage, microbiome transplantation-based therapies will offer a new perspective in cancer research. Hence, to understand this interplay, we outline the importance of micronutrients with an emphasis on the immunomodulatory function of the microbiome and highlight the microbiome's potential as a target for precision medicine in cancer treatment.
Collapse
Affiliation(s)
- Kartik Bhatnagar
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Kanupriya Jha
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Nishu Dalal
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi, India
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Ninad Patki
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Garima Gupta
- Biological Engineering and Sciences, Indian Institute of Technology Gandhinagar Palaj, Gandhinagar, Gujarat, India
| | - Amit Kumar
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Anil Kumar
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi, India
| | - Sarika Chaudhary
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
29
|
Hasanzadeh Haghighi F, Menbari S, Mohammadzadeh R, Pishdadian A, Farsiani H. Developing a potent vaccine against Helicobacter pylori: critical considerations and challenges. Expert Rev Mol Med 2024; 27:e12. [PMID: 39584502 PMCID: PMC11964096 DOI: 10.1017/erm.2024.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 01/13/2024] [Accepted: 08/07/2024] [Indexed: 11/26/2024]
Abstract
Helicobacter pylori (H. pylori) is closely associated with gastric cancer and peptic ulcers. The effectiveness of antibiotic treatment against H. pylori is diminished by the emergence of drug-resistant strains, side effects, high cost and reinfections. Given the circumstances, it is imperative to develop a potent vaccination targeting H. pylori. Understanding H. pylori's pathogenicity and the host's immune response is essential to developing a vaccine. Furthermore, vaccine evaluation necessitates the careful selection of design formulation. This review article aims to provide a concise overview of the considerations involved in selecting the optimal antigen, adjuvant, vaccine delivery system and laboratory animal model for vaccine formulation. Furthermore, we will discuss some significant obstacles in the realm of developing a potent vaccination against H. pylori.
Collapse
Affiliation(s)
- Faria Hasanzadeh Haghighi
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shaho Menbari
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Laboratory Sciences, Faculty of Paramedical Sciences, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Roghayeh Mohammadzadeh
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbas Pishdadian
- Department of Immunology, School of Medicine, Zabol University of Medical Sciences, Zabol, Iran
| | - Hadi Farsiani
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
30
|
Zeng R, Gou H, Lau HCH, Yu J. Stomach microbiota in gastric cancer development and clinical implications. Gut 2024; 73:2062-2073. [PMID: 38886045 PMCID: PMC11672014 DOI: 10.1136/gutjnl-2024-332815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
Gastric cancer (GC) is one of the most common malignancies and a prominent cause of cancer mortality worldwide. A distinctive characteristic of GC is its intimate association with commensal microbial community. Although Helicobacter pylori is widely recognised as an inciting factor of the onset of gastric carcinogenesis, increasing evidence has indicated the substantial involvement of microbes that reside in the gastric mucosa during disease progression. In particular, dysregulation in gastric microbiota could play pivotal roles throughout the whole carcinogenic processes, from the development of precancerous lesions to gastric malignancy. Here, current understanding of the gastric microbiota in GC development is summarised. Potential translational and clinical implications of using gastric microbes for GC diagnosis, prognosis and therapeutics are also evaluated, with further discussion on conceptual haziness and limitations at present. Finally, we highlight that modulating microbes is a novel and promising frontier for the prevention and management of GC, which necessitates future in-depth investigations.
Collapse
Affiliation(s)
- Ruijie Zeng
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Hongyan Gou
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Harry Cheuk Hay Lau
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
31
|
Huang Z, Chen S, Yin S, Shi Z, Gu L, Li L, Yin H, Huang Z, Li B, Chen X, Yang Y, Wang Z, Li H, Zhang C, He Y. Development and validation of a nomogram for predicting the risk of developing gastric cancer based on a questionnaire: a cross-sectional study. Front Oncol 2024; 14:1351967. [PMID: 39588309 PMCID: PMC11586234 DOI: 10.3389/fonc.2024.1351967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 10/21/2024] [Indexed: 11/27/2024] Open
Abstract
Background Detection of gastric cancer (GC) at early stages is an effective strategy for decreasing mortality. This study aimed to construct a prediction nomogram based on a questionnaire to assess the risk of developing GC. Methods Our study comprised a total of 4379 participants (2326 participants from outpatient at Fengqing People's Hospital were considered for model development and internal validation, and 2053 participants from outpatients at the endoscopy center at the Seventh Affiliated Hospital of Sun Yat-Sen University were considered for independent external validation) and gastric mucosa status was determined by endoscopy and biopsies. The eligible participants in development cohort from Fengqing people's Hospital were randomly separated into a training cohort (n=1629, 70.0%) and an internal validation cohort (n=697, 30.0%). The relevant features were selected by a least absolute shrinkage and selection operator (LASSO), and the ensuing features were evaluated through multivariable logistic regression analysis. Subsequently, the variables were selected to construct a prediction nomogram. The discriminative ability and predictive accuracy of the nomogram were evaluated by the C-index and calibration plot, respectively. Decision curve analysis (DCA) curves were used for the assessment of clinical benefit of the model. This model was developed to estimate the risk of developing neoplastic lesions according to the "transparent reporting of a multivariable prediction model for individual prognosis or diagnosis" (TRIPOD) statement. Results Six variables, including age, sex, alcohol consumption, cigarette smoking, education level, and Hp infection status, were independent risk factors for the development of neoplastic lesions. Thus, these variables were incorporated into the final nomogram. The AUC of the nomogram were 0.701, 0.657 and 0.699 in the training, internal validation, and external validation cohorts, respectively. The calibration curve showed that the nomogram was in good agreement with the observed outcomes. Compared to treatment of all patients or none, our nomogram showed a notably higher clinical benefit. Conclusion This nomogram proved to be a convenient, cost-effective tool to effectively predict an individual's risk of developing neoplastic lesions, and it can act as a prescreening tool before gastroscopy.
Collapse
Affiliation(s)
- Zhangsen Huang
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Songyao Chen
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Songcheng Yin
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Zhaowen Shi
- General Surgery, Fengqing People’s Hospital, Lincang, China
| | - Liang Gu
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Liang Li
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Haofan Yin
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zhijian Huang
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Bo Li
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Xin Chen
- General Surgery, Fengqing People’s Hospital, Lincang, China
| | - Yilin Yang
- General Surgery, Fengqing People’s Hospital, Lincang, China
| | - Zhengli Wang
- General Surgery, Fengqing People’s Hospital, Lincang, China
| | - Hai Li
- General Surgery, Fengqing People’s Hospital, Lincang, China
| | - Changhua Zhang
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yulong He
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| |
Collapse
|
32
|
Oh A, Rustgi SD, Hur C, In H. Cost-Effectiveness of Serum Pepsinogen as a Gastric Cancer Targeted Screening Strategy in the United States. GASTRO HEP ADVANCES 2024; 4:100564. [PMID: 39866720 PMCID: PMC11762188 DOI: 10.1016/j.gastha.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 10/05/2024] [Indexed: 01/28/2025]
Abstract
Background and Aims Current gastric cancer (GC) screening modalities are invasive and expensive. Noninvasive screening for GC precursors with serum pepsinogen (PG) may improve early detection and prevention. Test characteristics of PG based on US prospective data was recently reported and used to study the cost-effectiveness of PG screening vs no screening in the US. Methods A patient-level state transition microsimulation of gastric adenocarcinoma analyzed noninvasive screening vs no screening in a hypothetical cohort of average risk US individuals. Primary outcomes included life expectancy, quality-adjusted life years, total costs, and incremental cost-effectiveness ratios. Secondary outcomes included total GC incidence and mortality. Base-case PG sensitivity and specificity were 34.1% and 94.7%, respectively, with a wide range of PG performance characteristics also examined. Results One-time serum PG screening at age 40 was cost-effective compared to no screening with an incremental cost-effectiveness ratio of $4913.29 per quality-adjusted life year. PG screening resulted in 10.9% relative reduction in lifetime GC incidence and 10.8% relative decrease in cumulative GC mortality. Localized stage at diagnosis increased from 30.5% to 33.6% and metastatic stage decreased from 40.8% to 37.4%. Sensitivity analysis showed PG screening was most sensitive to endoscopy costs, chronic atrophic gastritis quality of life, and PG prevalence. PG screening remained cost-effective across a wide range of test values. Conclusion PG screening is a cost-effective strategy to improve GC mortality; however, mortality benefit will depend on the test characteristics of the biomarker. Future blood-based screening tests that have better performance characteristics could further improve GC prevention.
Collapse
Affiliation(s)
- Aaron Oh
- Albert Einstein College of Medicine, New York, New York
| | - Sheila D. Rustgi
- Division of Digestive and Liver Diseases, Columbia University Irving Medical Center, New York, New York
| | - Chin Hur
- Division of Digestive and Liver Diseases, Columbia University Irving Medical Center, New York, New York
| | - Haejin In
- Albert Einstein College of Medicine, New York, New York
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
- Department of Health Behavior, Society and Policy, Rutgers School of Public Health, Piscataway, New Jersey
| |
Collapse
|
33
|
Gan S, Li C, Hou R, Tian G, Zhao Y, Ren D, Zhou W, Zhao F, Lv K, Yang J. Dynamic changes of the immune microenvironment in the development of gastric cancer caused by inflammation. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200849. [PMID: 39228396 PMCID: PMC11369508 DOI: 10.1016/j.omton.2024.200849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/21/2024] [Accepted: 07/16/2024] [Indexed: 09/05/2024]
Abstract
Precancerous lesions typically precede gastric cancer (GC), but the molecular mechanisms underlying the transition from these lesions to GC remain unclear. Therefore, it is urgent to understand this transition from precancerous lesions to GC, which is crucial for the early diagnosis and treatment of GC. In this study, we merged multiple single-cell RNA sequencing datasets to investigate the molecular changes in distinct cell types associated with the progression of GC. First, we observed an increasing abundance of immune cells and a decrease in non-immune cells from non-atrophic gastritis to GC. Five immune cell types were significantly enriched in GC compared to precancerous lesions. Moreover, we found that the interleukin (IL)-17 signaling pathway and Th17 cell differentiation were significantly up-regulated in immune cell subsets during GC progression. Some genes in these processes were predominantly expressed at the GC stage, highlighting their potential as diagnostic markers. Furthermore, we validated our findings using bulk RNA sequencing data from The Cancer Genome Atlas and confirmed consistent immune cell changes during GC progression. Our study provides insights into the immune infiltration and signaling pathways involved in the development of GC, contributing to the development of early diagnosis and targeted treatment strategies for this malignancy.
Collapse
Affiliation(s)
- Siyuan Gan
- School of Mathematical Sciences, Ocean University of China, Qingdao, China
| | - Changfu Li
- Department of Digestive Internal Medicine, Daqing Longnan Hospital, The Fifth Affiliated Hospital of Qiqihar Medical College, Daqing 163000, China
| | - Rui Hou
- Geneis Beijing Co., Ltd, Beijing 100102, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, China
| | - Geng Tian
- Geneis Beijing Co., Ltd, Beijing 100102, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, China
| | - Yuan Zhao
- School of Electrical and Information Engineering, Anhui University of Technology, Ma'anshan, China
| | - Dan Ren
- Department of Pathology, Daqing Longnan Hospital, The Fifth Affiliated Hospital of Qiqihar Medical College, Daqing 163000, China
| | - Wenjing Zhou
- Department of Oncology, Hiser Medical Center of Qingdao, No. 4, Renmin Road, Shibei District, Qingdao, China
| | - Fei Zhao
- School of Mathematical Sciences, Ocean University of China, Qingdao, China
| | - Kebo Lv
- School of Mathematical Sciences, Ocean University of China, Qingdao, China
| | - Jialiang Yang
- Geneis Beijing Co., Ltd, Beijing 100102, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, China
| |
Collapse
|
34
|
Xu J, Yu B, Wang F, Yang J. Single-cell RNA sequencing to map tumor heterogeneity in gastric carcinogenesis paving roads to individualized therapy. Cancer Immunol Immunother 2024; 73:233. [PMID: 39271545 PMCID: PMC11399521 DOI: 10.1007/s00262-024-03820-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
Gastric cancer (GC) is a highly heterogeneous disease with a complex tumor microenvironment (TME) that encompasses multiple cell types including cancer cells, immune cells, stromal cells, and so on. Cancer-associated cells could remodel the TME and influence the progression of GC and therapeutic response. Single-cell RNA sequencing (scRNA-seq), as an emerging technology, has provided unprecedented insights into the complicated biological composition and characteristics of TME at the molecular, cellular, and immunological resolutions, offering a new idea for GC studies. In this review, we discuss the novel findings from scRNA-seq datasets revealing the origin and evolution of GC, and scRNA-seq is a powerful tool for investigating transcriptional dynamics and intratumor heterogeneity (ITH) in GC. Meanwhile, we demonstrate that the vital immune cells within TME, including T cells, B cells, macrophages, and stromal cells, play an important role in the disease progression. Additionally, we also overview that how scRNA-seq facilitates our understanding about the effects on individualized therapy of GC patients. Spatial transcriptomes (ST) have been designed to determine spatial distribution and capture local intercellular communication networks, enabling a further understanding of the relationship between the spatial background of a particular cell and its functions. In summary, scRNA-seq and other single-cell technologies provide a valuable perspective for molecular and pathological disease characteristics and hold promise for advancing basic research and clinical practice in GC.
Collapse
Affiliation(s)
- Jiao Xu
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road., Xi'an, 710061, Shaanxi, People's Republic of China
| | - Bixin Yu
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road., Xi'an, 710061, Shaanxi, People's Republic of China
| | - Fan Wang
- Phase I Clinical Trial Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China.
| | - Jin Yang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road., Xi'an, 710061, Shaanxi, People's Republic of China.
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road., Xi'an, 710061, Shaanxi, People's Republic of China.
- Phase I Clinical Trial Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China.
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China.
| |
Collapse
|
35
|
Zhen SY, Wei Y, Song R, Liu XH, Li PR, Kong XY, Wei HY, Fan WH, Liang CH. Prediction of lymphovascular invasion of gastric cancer based on contrast-enhanced computed tomography radiomics. Front Oncol 2024; 14:1389278. [PMID: 39301548 PMCID: PMC11410566 DOI: 10.3389/fonc.2024.1389278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 08/12/2024] [Indexed: 09/22/2024] Open
Abstract
Background Lymphovascular invasion (LVI) is a significant risk factor for lymph node metastasis in gastric cancer (GC) and is closely related to the prognosis and recurrence of GC. This study aimed to establish clinical models, radiomics models and combination models for the diagnosis of GC vascular invasion. Methods This study enrolled 146 patients with GC proved by pathology and who underwent radical resection of GC. The patients were assigned to the training and validation cohorts. A total of 1,702 radiomic features were extracted from contrast-enhanced computed tomography images of GC. Logistic regression analyses were performed to establish a clinical model, a radiomics model and a combined model. The performance of the predictive models was measured by the receiver operating characteristic (ROC) curve. Results In the training cohort, the age of LVI negative (-) patients and LVI positive (+) patients were 62.41 ± 8.41 and 63.76 ± 10.08 years, respectively, and there were more male (n = 63) than female (n = 19) patients in the LVI (+) group. Diameter and differentiation were the independent risk factors for determining LVI (-) and (+). A combined model was found to be relatively highly discriminative based on the area under the ROC curve for both the training (0.853, 95% CI: 0.784-0.920, sensitivity: 0.650 and specificity: 0.907) and the validation cohorts (0.742, 95% CI: 0.559-0.925, sensitivity: 0.736 and specificity: 0.700). Conclusions The combined model had the highest diagnostic effectiveness, and the nomogram established by this model had good performance. It can provide a reliable prediction method for individual treatment of LVI in GC before surgery.
Collapse
Affiliation(s)
- Si-Yu Zhen
- Department of Radiology, Xinxiang Medical University First Affiliated Hospital, Xinxiang, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Xinxiang, China
- Xinxiang Key Laboratory for Esophageal Cancer Imaging Diagnosis and Artificial Intelligence, Xinxiang, China
| | - Yong Wei
- Department of Radiology, Xinxiang Medical University First Affiliated Hospital, Xinxiang, China
| | - Ran Song
- Department of Radiology, Xinxiang Medical University First Affiliated Hospital, Xinxiang, China
| | - Xiao-Huan Liu
- Department of Radiology, Xinxiang Medical University First Affiliated Hospital, Xinxiang, China
| | - Pei-Ru Li
- Department of Radiology, Xinxiang Medical University First Affiliated Hospital, Xinxiang, China
| | - Xiang-Yan Kong
- Department of Radiology, Xinxiang Medical University First Affiliated Hospital, Xinxiang, China
| | - Han-Yu Wei
- Department of Radiology, Xinxiang Medical University First Affiliated Hospital, Xinxiang, China
| | - Wen-Hua Fan
- Department of Radiology, Xinxiang Medical University First Affiliated Hospital, Xinxiang, China
| | - Chang-Hua Liang
- Department of Radiology, Xinxiang Medical University First Affiliated Hospital, Xinxiang, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Xinxiang, China
- Xinxiang Key Laboratory for Esophageal Cancer Imaging Diagnosis and Artificial Intelligence, Xinxiang, China
| |
Collapse
|
36
|
Niu X, Wang N, Wang Y, Feng J, Li L, Han K, Chai N, Linghu E. Role of submucosal injection in radiofrequency ablation of gastric low-grade dysplasia: Effects on symptoms and outcomes. Chin Med J (Engl) 2024; 137:2099-2110. [PMID: 38602084 PMCID: PMC11374300 DOI: 10.1097/cm9.0000000000003080] [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: 10/08/2023] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND To date, there is still a lack of standardized management strategies for gastric low-grade dysplasia (LGD), which is a direct neoplastic precancerous lesion and requires specifically superficial destruction. Radiofrequency ablation (RFA) is expected to be an effective method for gastric LGD, but post-RFA pain may affect patients' satisfaction and compliance. The current study aimed to evaluate the value of a submucosal injection prior to RFA (SI-RFA) for postoperative pain and treatment outcomes. METHODS Between October 2014 and July 2021, gastric LGDs without risk factors (size >2 cm, unclear boundary, and abnormal microsurface and microvascularity) undergoing regular RFA and SI-RFA were retrospectively analyzed. Postoperative pain scores, wound healing, and clinical efficacy were compared. Propensity score matching, stratified analysis, and multivariable logistic regression were performed to control the confounding variables. RESULTS One hundred and ninety-seven gastric LGDs in 151 patients received regular RFA. Forty-nine gastric LGDs in 36 patients received SI-RFA. Thirty-six pairs of patients were selected for the assessment of postoperative pain by propensity score matching. Compared to regular RFA, SI-RFA significantly decreased the degree and duration of postoperative pain (OR, 0.32; 95% CI, 0.13-0.84; P = 0.020), improved wound healing rate (80.0% [36/45] vs . 58.9% [89/151], P = 0.012), increased the complete ablation rate (91.8% [45/49] vs . 86.3% [170/197], χ2 = 1.094, P = 0.295), but correlated with higher rates of local recurrence and progression (25.6% [10/39] vs . 13.2% [18/136], χ2 = 3.471, P = 0.062; 8.3% [3/36] vs . 0.9% [1/116], P = 0.042). The multivariable logistic regression model confirmed that submucosal injection was associated with local recurrence (OR, 2.93; 95% CI, 1.13-7.58; P = 0.027). CONCLUSIONS Submucosal injections prior to RFA may reduce postoperative pain and scar formation while ensuring complete ablation of gastric LGD. However, local recurrence and progression should be considered seriously.
Collapse
Affiliation(s)
- Xiaotong Niu
- Medical School of Chinese PLA General Hospital, Beijing 100853, China
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Nanjun Wang
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Yan Wang
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
- Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | - Jia Feng
- Department of Gastroenterology, Bethune International Peace Hospital, ShijiaZhuang, Hebei 050082, China
| | - Longsong Li
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Ke Han
- Medical School of Chinese PLA General Hospital, Beijing 100853, China
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Ningli Chai
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Enqiang Linghu
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
37
|
Filip AM, Munteanu SN, Mocan S, Huțanu D, Pantea M, Negovan A. Helicobacter pylori and autoimmunity in atrophic gastritis - comparison of clinical, endoscopic and histopathological features. ACTA MARISIENSIS - SERIA MEDICA 2024; 70:149-156. [DOI: 10.2478/amma-2024-0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Abstract
Objective
This study aims to investigate the clinical, endoscopic, biologic and histopathological differences between Helicobacter pylori-associated and autoimmune gastric atrophy.
Methods
A retrospective analysis was conducted on 95 patients diagnosed with either H. pylori-related corporal and antral atrophy (43 patients) or autoimmune corporal atrophic gastritis (52 patients).
Results
A significant male predisposition for H. pylori-associated atrophic changes in both the antrum and corpus regions (p=0.007, OR=3.24) was observed in comparison with autoimmune etiology of atrophy. While comorbidities and lifestyle factors showed similar distributions across groups, only unintentional self-reported weight loss demonstrated a significant association with H. pylori atrophy (p=0.0177, OR=3.94). Corporal erosions were strongly associated with antral and corporeal atrophic gastritis (p=0.04, OR=8.27), but the rest of mucosal lesions are comparable among groups. Interestingly, patients with H. pylori-related pangastric atrophy exhibited lower frequencies of altered triglyceride (p=0.018) and cholesterol (p=0.029) levels compared to the autoimmune group. Linear regression analysis identified low triglyceride levels as an independent predictor for H. pylori-associated antral and corporal atrophic gastritis (p=0.04) in endoscopic population with atrophy, but no hematological or clinical parameters were predictive for these changes.
Conclusions
Male patients are more likely to present with corpus atrophic gastritis associated with H. pylori infection than with an autoimmune etiology. Patients with atrophic gastritis tend to have similar clinical characteristics, except for dyslipidemia, which is more prevalent in those with H. pylori pangastritis. Corporal erosions are associated with active H. pylori infection in atrophic mucosa.
Collapse
Affiliation(s)
- Ana-Maria Filip
- Internal Medicine Department , Emergency County Hospital of Targu Mures , Targu Mures , Romania
| | - Sabrina-Nicoleta Munteanu
- Department of Clinical Science-Internal Medicine , George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures , Romania
| | - Simona Mocan
- Pathology Department , Emergency County Hospital of Targu Mures , Targu Mures , Romania
| | - Dragoș Huțanu
- Pulmonology Department , Mures County Clinical Hospital , Targu Mures , Romania
| | - Monica Pantea
- Department of Clinical Science-Internal Medicine , George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures , Romania
| | - Anca Negovan
- Department of Clinical Science-Internal Medicine , George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures , Romania
| |
Collapse
|
38
|
Choi SJ, Choi HS, Kim H, Lee JM, Kim SH, Yoon JH, Keum B, Kim HJ, Chun HJ, Park YH. Gastric Cancer and Intestinal Metaplasia: Differential Metabolic Landscapes and New Pathways to Diagnosis. Int J Mol Sci 2024; 25:9509. [PMID: 39273456 PMCID: PMC11395121 DOI: 10.3390/ijms25179509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
Gastric cancer (GC) is the fifth most common cause of cancer-related death worldwide. Early detection is crucial for improving survival rates and treatment outcomes. However, accurate GC-specific biomarkers remain unknown. This study aimed to identify the metabolic differences between intestinal metaplasia (IM) and GC to determine the pathways involved in GC. A metabolic analysis of IM and tissue samples from 37 patients with GC was conducted using ultra-performance liquid chromatography with tandem mass spectrometry. Overall, 665 and 278 significant features were identified in the aqueous and 278 organic phases, respectively, using false discovery rate analysis, which controls the expected proportion of false positives among the significant results. sPLS-DA revealed a clear separation between IM and GC samples. Steroid hormone biosynthesis, tryptophan metabolism, purine metabolism, and arginine and proline metabolism were the most significantly altered pathways. The intensity of 11 metabolites, including N1, N2-diacetylspermine, creatine riboside, and N-formylkynurenine, showed significant elevation in more advanced GC. Based on pathway enrichment analysis and cancer stage-specific alterations, we identified six potential candidates as diagnostic biomarkers: aldosterone, N-formylkynurenine, guanosine triphosphate, arginine, S-adenosylmethioninamine, and creatine riboside. These metabolic differences between IM and GC provide valuable insights into gastric carcinogenesis. Further validation is needed to develop noninvasive diagnostic tools and targeted therapies to improve the outcomes of patients with GC.
Collapse
Affiliation(s)
- Seong Ji Choi
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul 04763, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Hyuk Soon Choi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Hyunil Kim
- EN BIO, Cheongju-si 28494, Republic of Korea
| | - Jae Min Lee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Seung Han Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Jai Hoon Yoon
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul 04763, Republic of Korea
| | - Bora Keum
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Hyo Jung Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Hoon Jai Chun
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | | |
Collapse
|
39
|
Arai J, Hayakawa Y, Tateno H, Fujiwara H, Kasuga M, Fujishiro M. The role of gastric mucins and mucin-related glycans in gastric cancers. Cancer Sci 2024; 115:2853-2861. [PMID: 39031976 PMCID: PMC11463072 DOI: 10.1111/cas.16282] [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: 04/23/2024] [Revised: 06/22/2024] [Accepted: 07/02/2024] [Indexed: 07/22/2024] Open
Abstract
Gastric mucins serve as a protective barrier on the stomach's surface, protecting from external stimuli including gastric acid and gut microbiota. Their composition typically changes in response to the metaplastic sequence triggered by Helicobacter pylori infection. This alteration in gastric mucins is also observed in cases of gastric cancer, although the precise connection between mucin expressions and gastric carcinogenesis remains uncertain. This review first introduces the relationship between mucin expressions and gastric metaplasia or cancer observed in humans and mice. Additionally, we discuss potential pathogenic mechanisms of how aberrant mucins and their glycans affect gastric carcinogenesis. Finally, we summarize challenges to target tumor-specific glycans by utilizing lectin-drug conjugates that can bind to specific glycans. Understanding the correlation and mechanism between these mucin expressions and gastric carcinogenesis could pave the way for new strategies in gastric cancer treatment.
Collapse
Affiliation(s)
- Junya Arai
- Division of Gastroenterology, The Institute for Medical ScienceAsahi Life FoundationChuo‐ku, TokyoJapan
- Department of Gastroenterology, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| | - Hiroaki Tateno
- Cellular and Molecular Biotechnology Research InstituteNational Institute of Advanced Industrial Science and Technology (AIST)TsukubaJapan
| | - Hiroaki Fujiwara
- Division of Gastroenterology, The Institute for Medical ScienceAsahi Life FoundationChuo‐ku, TokyoJapan
| | - Masato Kasuga
- The Institute for Medical ScienceAsahi Life FoundationChuo‐ku, TokyoJapan
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| |
Collapse
|
40
|
Kawamura M, Uedo N, Yao K, Koike T, Kanesaka T, Hatta W, Ogata Y, Iwai W, Yokosawa S, Honda J, Asonuma S, Okata H, Ohyauchi M, Ito H, Abe Y, Ara N, Kayaba S, Shinkai H, Kanemitsu T. Endoscopic and histological risk stratification for gastric cancer using gastric intestinal metaplasia. J Gastroenterol Hepatol 2024; 39:1910-1916. [PMID: 38740510 DOI: 10.1111/jgh.16617] [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: 01/03/2024] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND AND AIM Intestinal metaplasia (IM) of the gastric mucosa is strongly associated with the risk of gastric cancer (GC). This study was performed to investigate the usefulness of endoscopic and histological risk stratification for GC using IM. METHODS This was a post-hoc analysis of a multicenter prospective study involving 10 Japanese facilities (UMINCTR000027023). The ridge/tubulovillous pattern, light blue crest (LBC), white opaque substance (WOS), endoscopic grading of gastric IM (EGGIM) score using non-magnifying image-enhanced endoscopy, and operative link on gastric IM assessment (OLGIM) were evaluated for their associations with GC risk in all patients. RESULTS In total, 380 patients (115 with GC and 265 without GC) were analyzed. The presence of an LBC (limited to antrum: odds ratio [OR] 2.4 [95% confidence interval 1.1-5.0], extended to corpus: OR 3.6 [2.1-6.3]), the presence of WOS (limited to antrum: OR 3.0 [1.7-5.3], extended to corpus: OR 4.2 [2.1-8.2]), and histological IM (limited to antrum: OR 3.2 [1.4-7.4], extended to corpus: OR 8.5 [4.5-16.0]) were significantly associated with GC risk. Additionally, the EGGIM score (5-8 points: OR 8.8 [4.4-16.0]) and OLGIM (stage III/IV: OR 12.5 [6.1-25.8]) were useful for stratification of GC risk. The area under the receiver operating characteristic curve value for GC risk was 0.740 for OLGIM and 0.706 for EGGIM. CONCLUSIONS The LBC, WOS, EGGIM, and OLGIM were strongly associated with GC risk in Japanese patients. This finding can be useful for GC risk assessment in daily clinical practice.
Collapse
Affiliation(s)
- Masashi Kawamura
- Department of Gastroenterology, Sendai City Hospital, Sendai, Japan
| | - Noriya Uedo
- Department of Gastrointestinal Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Kenshi Yao
- Department of Endoscopy, Fukuoka University Chikushi Hospital, Fukuoka, Japan
| | - Tomoyuki Koike
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Kanesaka
- Department of Gastrointestinal Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Waku Hatta
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yohei Ogata
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Wataru Iwai
- Department of Gastroenterology, Miyagi Cancer Center, Natori, Japan
| | - Satoshi Yokosawa
- Department of Gastroenterology, Iwate Prefectural Iwai Hospital, Iwate, Japan
| | - Junya Honda
- Department of Gastroenterology, Iwate Prefectural Iwai Hospital, Iwate, Japan
| | - Sho Asonuma
- Department of Gastroenterology, South Miyagi Medical Center, Ogawara, Japan
| | - Hideki Okata
- Department of Gastroenterology, South Miyagi Medical Center, Ogawara, Japan
| | - Motoki Ohyauchi
- Department of Gastroenterology, Osaki Citizen Hospital, Ōsaki, Japan
| | - Hirotaka Ito
- Department of Gastroenterology, Osaki Citizen Hospital, Ōsaki, Japan
| | - Yasuhiko Abe
- Division of Endoscopy, Yamagata University Hospital, Yamagata, Japan
| | - Nobuyuki Ara
- Department of Gastroenterology, National Hospital Organization Sendai Medical Center, Sendai, Japan
| | - Shoichi Kayaba
- Department of Gastroenterology, Iwate Prefectural Isawa Hospital, Iwate, Japan
| | - Hirohiko Shinkai
- Department of Gastroenterology, Iwate Prefectural Isawa Hospital, Iwate, Japan
| | - Takao Kanemitsu
- Department of Gastroenterology, Fukuoka University Chikushi Hospital, Fukuoka, Japan
| |
Collapse
|
41
|
Xia R, Jiang Z, Zhou Y, Pan L, Wang Y, Ma Y, Fan L, Yuan L, Cheng X. Oral microbiota and gastric cancer: recent highlights and knowledge gaps. J Oral Microbiol 2024; 16:2391640. [PMID: 39161727 PMCID: PMC11332296 DOI: 10.1080/20002297.2024.2391640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/19/2024] [Accepted: 08/08/2024] [Indexed: 08/21/2024] Open
Abstract
Gastric cancer is one of the most common malignant tumors worldwide and has a high mortality rate. However, tests for the early screening and diagnosis of gastric cancer are limited and invasive. Certain oral microorganisms are over-expressed in gastric cancer, but there is heterogeneity among different studies. Notably, each oral ecological niche harbors specific microorganisms. Among them, tongue coating, saliva, and dental plaque are important and unique ecological niches in the oral cavity. The colonization environment in different oral niches may be a source of heterogeneity. In this paper, we systematically discuss the latest developments in the field of the oral microbiota and gastric cancer and elucidate the enrichment of microorganisms in the oral ecological niches of the tongue coatings, saliva, and dental plaque in gastric cancer patients. The various potential mechanisms by which the oral microbiota induces gastric cancer (activation of an excessive inflammatory response; promotion of proliferation, migration, invasion, and metastasis; and secretion of carcinogens, leading to imbalance in gastric microbial communities) are explored. In this paper, we also highlight the applications of the rapeutics targeting the oral microbiota in gastric cancer and suggests future research directions related to the relationship between the oral microbiota and gastric cancer.
Collapse
Affiliation(s)
- Ruihong Xia
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhengchen Jiang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ying Zhou
- Department of Pharmacy, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Libin Pan
- Department of Pharmacy, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yanan Wang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yubo Ma
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lili Fan
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Li Yuan
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Department of Integrated Chinese and Western Medicine, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
42
|
Kotelevets SM, Chekh SA, Chukov SZ. Effectiveness of serological markers of gastric mucosal atrophy in the gastric precancer screening and in cancer prevention. World J Gastrointest Endosc 2024; 16:462-471. [PMID: 39155993 PMCID: PMC11325870 DOI: 10.4253/wjge.v16.i8.462] [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: 03/13/2024] [Revised: 06/30/2024] [Accepted: 07/25/2024] [Indexed: 08/01/2024] Open
Abstract
BACKGROUND New markers are needed to improve the effectiveness of serological screening for atrophic gastritis. AIM To develop a cost-effective method for serological screening of atrophic gastritis with a high level of sensitivity. METHODS Of the 169 patients with atrophic gastritis, selected by the visual endoscopic Kimura-Takemoto method, 165 showed histological mucosal atrophy using the updated Kimura-Takemoto method. All 169 patients were examined for postprandial levels of gastrin-17 (G17) and pepsinogen-1 (PG1) using GastroPanel® (Biohit Plc, Helsinki, Finland). RESULTS We used the histological standard of five biopsies of the gastric mucosa, in accordance with the Kimura-Takemoto classification system to assess the sensitivity of G17 in detecting gastric mucosal atrophy. We also compared the morpho-functional relationships between the detected histological degree of gastric mucosal atrophy and the serological levels of G17 and PG1, as the markers of atrophic gastritis. The sensitivity of postprandial G17 was 62.2% for serological levels of G17 (range: 0-4 pmol/L) and 100% for serological G17 (range: 0-10 pmol/L) for the detection of monofocal severe atrophic gastritis. No strong correlation was found between the levels of PG1 and degree of histological atrophy determined by the Kimura-Takemoto classification system to identify the severity of mucosal atrophy of the gastric corpus. In the presented clinical case of a 63-year-old man with multifocal atrophic gastritis, there is a pronounced positive long-term dynamics of the serological marker of atrophy - postprandial G17, after five months of rennet replacement therapy. CONCLUSION Serological screening of multifocal atrophic gastritis by assessment of postprandial G17 is a cost-effective method with high sensitivity. Postprandial G17 is an earlier marker of regression of atrophic gastritis than a morphological examination of a gastric biopsy in accordance with the Sydney system. Therefore, postprandial G17 is recommended for dynamic monitoring of atrophic gastritis after treatment.
Collapse
Affiliation(s)
- Sergey M Kotelevets
- Department of Therapy, North Caucasus State Academy, Cherkessk 369000, Karachay-Cherkess Republic, Russia
| | - Sergey A Chekh
- Department of Mathematics, North Caucasus State Academy, Cherkessk 369000, Karachay-Cherkess Republic, Russia
| | - Sergey Z Chukov
- Department of Pathological Anatomy, Stavropol State Medical University, Stavropol 355017, Russia
| |
Collapse
|
43
|
Sun Y, Yin L, Nesheli DN, Yu J, Franzén J, Ye W. Overall and cause-specific mortality among patients diagnosed with gastric precancerous lesions in Sweden between 1979 and 2014: an observational cohort study. BMC Med 2024; 22:333. [PMID: 39148123 PMCID: PMC11328423 DOI: 10.1186/s12916-024-03554-1] [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: 03/21/2024] [Accepted: 08/05/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND The Correa's cascade, encompassing chronic non-atrophic gastritis, atrophic gastritis, intestinal metaplasia, and dysplasia, represents the well-recognized pathway for the development of non-cardia gastric cancer. Population-based studies on all-cause and cause-specific mortalities among patients with gastric lesions in Correa's cascade are scarce. METHODS We compiled a cohort of 340 744 eligible patients who had undergone endoscopy with biopsy for non-malignant indications during the period 1979-2011, which was followed up until 2014. Standardized mortality ratios (SMRs) with 95% confidence intervals (CIs) provided estimation of the relative risk, using the general Swedish population as reference. Cox regression model was used to estimate hazard ratios (HRs) of death for internal comparison. RESULTS A total of 306 117 patients were included in the final analysis, accumulating 3,049,009 person-years of follow-up. In total 106,625 deaths were observed during the study period. Compared to the general population, excess risks of overall mortality were noted in all subgroups, with SMRs ranging from 1.11 (95% CI 1.08-1.14) for the normal mucosa group to 1.54 (95% CI 1.46-1.62) for the dysplasia group. For cause-specific mortalities, mortality from gastric cancer gradually increased along Correa's cascade, with excess risk rising from 105% for patients with chronic gastritis to more than 600% for the dysplasia group. These results were confirmed in the comparison with the normal mucosa group. For non-cancer conditions, increased death risks were noted for various diseases compared to the general population, especially among patients with more severe gastric precancerous lesions. But the results were confirmed only for "infectious diseases and parasitic diseases", "respiratory system diseases", and "digestive system disease", when using the normal mucosa group as reference. CONCLUSIONS Increased mortality from gastric cancer suggests that early recognition and intervention of gastric precancerous lesions probably benefit the patients. Excess mortality due to non-cancer conditions should be interpreted with caution, and future studies are warranted.
Collapse
Affiliation(s)
- Yawen Sun
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Li Yin
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden
| | | | - Jingru Yu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Joar Franzén
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Weimin Ye
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden.
| |
Collapse
|
44
|
Kim B, Chun H, Lee J, Park M, Kwak Y, Kim JM, Kim SG, Ryu JK, Choi J, Cho S. Predictive biomarkers for metachronous gastric cancer development after endoscopic resection of early gastric cancer. Cancer Med 2024; 13:e70104. [PMID: 39171503 PMCID: PMC11339598 DOI: 10.1002/cam4.70104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 07/21/2024] [Accepted: 08/04/2024] [Indexed: 08/23/2024] Open
Abstract
OBJECTIVES We aimed to identify predictive markers for metachronous gastric cancer (MGC) in early gastric cancer (EGC) patients curatively treated with endoscopic submucosal dissection (ESD). MATERIALS AND METHODS From EGC patients who underwent ESD, bulk RNA sequencing was performed on non-cancerous gastric mucosa samples at the time of initial EGC diagnosis. This included 23 patients who developed MGC, and 23 control patients without additional gastric neoplasms for over 3 years (1:1 matched by age, sex, and Helicobacter pylori infection state). Candidate differentially-expressed genes were identified, from which biomarkers were selected using real-time quantitative polymerase chain reaction and cell viability assays using gastric cell lines. An independent validation cohort of 55 MGC patients and 125 controls was used for marker validation. We also examined the severity of gastric intestinal metaplasia, a known premalignant condition, at initial diagnosis. RESULTS From the discovery cohort, 86 candidate genes were identified of which KDF1 and CDK1 were selected as markers for MGC, which were confirmed in the validation cohort. CERB5 and AKT2 isoform were identified as markers related to intestinal metaplasia and were also highly expressed in MGC patients compared to controls (p < 0.01). Combining these markers with clinical data (age, sex, H. pylori and severity of intestinal metaplasia) yielded an area under the curve (AUC) of 0.91 (95% CI, 0.85-0.97) for MGC prediction. CONCLUSION Assessing biomarkers in non-cancerous gastric mucosa may be a useful method for predicting MGC in EGC patients and identifying patients with a higher risk of developing MGC, who can benefit from rigorous surveillance.
Collapse
Affiliation(s)
- Bokyung Kim
- Department of Internal Medicine and Liver Research InstituteSeoul National University Hospital, Seoul National University College of MedicineSeoulKorea
| | - Harim Chun
- Department of Biomedical SciencesKorea University College of MedicineSeoulKorea
| | - Jongwon Lee
- Department of Biomedical SciencesKorea University College of MedicineSeoulKorea
| | - Miree Park
- Department of Internal Medicine and Liver Research InstituteSeoul National University Hospital, Seoul National University College of MedicineSeoulKorea
| | - Yoonjin Kwak
- Department of PathologySeoul National University HospitalSeoulKorea
| | - Jung Mogg Kim
- Department of MicrobiologyHanyang University College of MedicineSeoulKorea
| | - Sang Gyun Kim
- Department of Internal Medicine and Liver Research InstituteSeoul National University Hospital, Seoul National University College of MedicineSeoulKorea
| | - Ji Kon Ryu
- Department of Internal Medicine and Liver Research InstituteSeoul National University Hospital, Seoul National University College of MedicineSeoulKorea
| | - Jungmin Choi
- Department of Biomedical SciencesKorea University College of MedicineSeoulKorea
| | - Soo‐Jeong Cho
- Department of Internal Medicine and Liver Research InstituteSeoul National University Hospital, Seoul National University College of MedicineSeoulKorea
| |
Collapse
|
45
|
Smith SM, Boyle B, Buckley M, Costigan C, Doyle M, Farrell R, Ismail MS, Kevans D, Nugent S, O’Connor A, O’Morain C, Parihar V, Ryan C, McNamara D. The second Irish Helicobacter pylori Working Group consensus for the diagnosis and treatment of Helicobacter pylori infection in adult patients in Ireland. Eur J Gastroenterol Hepatol 2024; 36:1000-1009. [PMID: 38829956 PMCID: PMC11198963 DOI: 10.1097/meg.0000000000002796] [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: 01/25/2024] [Accepted: 05/08/2024] [Indexed: 06/05/2024]
Abstract
BACKGROUND There has been an increase in resistance to many of the antimicrobials used to treat Helicobacter pylori ( H. pylori ) nationally and internationally. Primary clarithromycin resistance and dual clarithromycin and metronidazole resistance are high in Ireland. These trends call for an evaluation of best-practice management strategies. OBJECTIVE The objective of this study was to revise the recommendations for the management of H. pylori infection in adult patients in the Irish healthcare setting. METHODS The Irish H. pylori working group (IHPWG) was established in 2016 and reconvened in 2023 to evaluate the most up-to-date literature on H. pylori diagnosis, eradication rates and antimicrobial resistance. The 'GRADE' approach was then used to rate the quality of available evidence and grade the resulting recommendations. RESULTS The Irish H. pylori working group agreed on 14 consensus statements. Key recommendations include (1) routine antimicrobial susceptibility testing to guide therapy is no longer recommended other than for clarithromycin susceptibility testing for first-line treatment (statements 6 and 9), (2) clarithromycin triple therapy should only be prescribed as first-line therapy in cases where clarithromycin susceptibility has been confirmed (statement 9), (3) bismuth quadruple therapy (proton pump inhibitor, bismuth, metronidazole, tetracycline) is the recommended first-line therapy if clarithromycin resistance is unknown or confirmed (statement 10), (4) bismuth quadruple therapy with a proton pump inhibitor, levofloxacin and amoxicillin is the recommended second-line treatment (statement 11) and (5) rifabutin amoxicillin triple therapy is the recommend rescue therapy (statement 12). CONCLUSION These recommendations are intended to provide the most relevant current best-practice guidelines for the management of H. pylori infection in adults in Ireland.
Collapse
Affiliation(s)
| | - Breida Boyle
- Department of Clinical Microbiology, St. James’s Hospital, Dublin
| | - Martin Buckley
- Department of Gastroenterology, Mercy University Hospital, Cork
| | - Conor Costigan
- School of Medicine, Trinity College Dublin
- Department of Gastroenterology, Tallaght University Hospital, Dublin
| | - Maeve Doyle
- Department of Microbiology, University Hospital Waterford, Waterford
| | - Richard Farrell
- Department of Gastroenterology, Connolly Hospital, RCSI, Dublin
| | | | - David Kevans
- School of Medicine, Trinity College Dublin
- Department of Gastroenterology, St. James’s Hospital, Dublin
| | - Sean Nugent
- Department of Gastroenterology, Whitfield Clinic, Waterford
| | - Anthony O’Connor
- School of Medicine, Trinity College Dublin
- Department of Gastroenterology, Tallaght University Hospital, Dublin
| | | | - Vikrant Parihar
- Department of Gastroenterology, Letterkenny University Hospital
| | - Cristín Ryan
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
| | - Deirdre McNamara
- School of Medicine, Trinity College Dublin
- Department of Gastroenterology, Tallaght University Hospital, Dublin
| |
Collapse
|
46
|
Hu R, Xue X, Sun X, Mi Y, Wen H, Xi H, Li F, Zheng P, Liu S. Revealing the role of metformin in gastric intestinal metaplasia treatment. Front Pharmacol 2024; 15:1340309. [PMID: 39101145 PMCID: PMC11294171 DOI: 10.3389/fphar.2024.1340309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 07/01/2024] [Indexed: 08/06/2024] Open
Abstract
Objective Gastric intestinal metaplasia (IM) is a precancerous stage associated with gastric cancer. Despite the observed beneficial effects of metformin on IM, its molecular mechanism remains not fully elucidated. This study aims to reveal the effects and potential mechanisms of metformin in treating IM based on both bioinformatics and in vivo investigations. Methods The seven public databases (GeneCards, DisGeNET, OMIM, SuperPred, Pharm Mapper, Swiss Target Prediction, TargetNet) were used in this work to identify targeted genes related to intestinal metaplasia (IM) and metformin. The shared targeted genes between metformin and IM were further analyzed by network pharmacology, while the interactions in-between were investigated by molecular docking. In parallel, the therapeutic effect of metformin was evaluated in IM mice model, while the core targets and pathways effected by metformin were verified in vivo. Results We screened out 1,751 IM-related genes and 318 metformin-targeted genes, 99 common genes identified in between were visualized by constructing the protein-protein interaction (PPI) network. The top ten core targeted genes were EGFR, MMP9, HIF1A, HSP90AA1, SIRT1, IL2, MAPK8, STAT1, PIK3CA, and ICAM1. The functional enrichment analysis confirmed that carcinogenesis and HIF-1 signaling pathways were primarily involved in the metformin treatment of IM. Based on molecular docking and dynamics, we found metformin affected the function of its targets by inhibiting receptor binding. Furthermore, metformin administration reduced the progression of IM lesions in Atp4a-/- mice model significantly. Notably, metformin enhanced the expression level of MUC5AC, while inhibited the expression level of CDX2. Our results also showed that metformin modulated the expression of core targets in vivo by reducing the activity of NF-κB and the PI3K/AKT/mTOR/HIF-1α signaling pathway. Conclusion This study confirms that metformin improves the efficacy of IM treatment by regulating a complex molecular network. Metformin plays a functional role in inhibiting inflammation/apoptosis-related pathways of further IM progression. Our work provides a molecular foundation for understanding metformin and other guanidine medicines in IM treatment.
Collapse
Affiliation(s)
- Ruoyu Hu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xia Xue
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiangdong Sun
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Mi
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huijuan Wen
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Huayuan Xi
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fuhao Li
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Pengyuan Zheng
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Simeng Liu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
47
|
Yoon JY, Katcher E, Cohen E, Ward SC, Rouphael C, Itzkowitz SH, Wang CP, Kim MK, Shah SC. Endoscopic Surveillance of Gastric Intestinal Metaplasia: A Retrospective Cohort Study. J Clin Gastroenterol 2024:00004836-990000000-00327. [PMID: 39042489 PMCID: PMC11739429 DOI: 10.1097/mcg.0000000000002039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/28/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Gastric intestinal metaplasia (GIM) is a precancerous condition. Limited data exist on real-world clinical practice relative to guidelines. AIM The aim of this study was to evaluate adherence to GIM risk stratification and identify factors associated with follow-up endoscopy. MATERIALS AND METHODS We conducted manual chart review of patients with histologically confirmed GIM at an urban, tertiary medical center were identified retrospectively and details of their demographics, Helicobacter pylori, biopsy protocol, endoscopic/histologic findings, and postendoscopy follow-up were recorded. Multivariable logistic regression was used to identify factors independently associated with follow-up endoscopy. RESULTS Among 253 patients, 59% were female, 37% non-Hispanic White (NHW), 26% Hispanic, 16% non-Hispanic Black (NHB). The median age at index endoscopy was 63.4 years (IQR: 55.9 to 70.0), with median follow-up of 65.1 months (IQR: 44.0 to 72.3). H. pylori was detected in 21.6% patients at index EGD. GIM extent and subtype data were frequently missing (22.9% and 32.8%, respectively). Based on available data, 26% had corpus-extended GIM and 28% had incomplete/mixed-type GIM. Compared with NHW, Hispanic patients had higher odds of follow-up EGD (OR=2.48, 95% CI: 1.23-5.01), while NHB patients had 59% lower odds of follow-up EGD (OR=0.41, 95% CI: 0.18-0.96). Corpus-extended GIM versus limited GIM (OR=2.27, 95% CI: 1.13-4.59) was associated with follow-up EGD, but GIM subtype and family history of gastric cancer were not. CONCLUSIONS We observed suboptimal risk stratification among patients with GIM and notable race and ethnic disparities with respect to endoscopic surveillance. Targeted interventions are needed to improve practice patterns and mitigate observed disparities.
Collapse
Affiliation(s)
- Ji Yoon Yoon
- Department of Gastroenterology, Icahn School of Medicine at Mount Sinai, NY
| | - Erik Katcher
- Sackler School of Medicine at Tel Aviv University, Tel Aviv, Israel
| | - Ella Cohen
- Department of Medicine, Icahn School of Medicine at Mount Sinai, NY
| | - Stephen C. Ward
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, NY
| | - Carol Rouphael
- Department of Gastroenterology, Hepatology and Nutrition, Cleveland Clinic Foundation, OH
| | | | - Christina P. Wang
- Department of Gastroenterology, Icahn School of Medicine at Mount Sinai, NY
| | - Michelle Kang Kim
- Department of Gastroenterology, Hepatology and Nutrition, Cleveland Clinic Foundation, OH
| | - Shailja C. Shah
- Division of Gastroenterology, University of California San Diego, CA
- Gastroenterology Section, Veterans Affairs San Diego Healthcare System, CA
| |
Collapse
|
48
|
Hu Y, Su M, Kong Y, Jiang C, Yuan Y, Chen X, Ma L. Total synthesis/semi-synthesis of natural isopentenyl flavonoids with inhibitory activity on NLRP3 inflammasome. Bioorg Med Chem Lett 2024; 107:129777. [PMID: 38692522 DOI: 10.1016/j.bmcl.2024.129777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/24/2024] [Accepted: 04/27/2024] [Indexed: 05/03/2024]
Abstract
Inflammation is the body's defense response to stimuli. When the homeostatic balance is disturbed, disease may result. Flavonoids have clear anti-inflammatory effects and the isopentenyl group significantly enhances the pharmacological activity of flavonoids. Therefore, isopentenyl flavonoids have the potential to serve as lead compounds for the development of anti-inflammatory drugs. Throughout this research, eight natural compounds were synthesized, including 5,7-dihydroxy-4'-methoxy-8-prenylflavonoid (1), 4'-O-Methylatalantoflavone (2), Kushenol W (3) and Racemoflavone (5), which were totally synthesized for the first time. Additionally, three flavonols: Licoflavonol (6), 3,5,7,3',4'-pentahydroxy-6-prenylflavonol (7) and Macarangin (8), can be one-step synthesized by direct C-isopentenylation. In the process, an economical and efficient C-isopentenylation method was also simultaneously explored that could facilitate the efficient synthesis of natural products. These compounds were evaluated for their potential anti-inflammatory activities via the NLRP3 signaling pathway. Notably, Macarangin (8) manifested the most potent inhibitory effect. The SAR (Structure-Activity Relationships) also showed the introduction of the isopentenyl group was determined to enhance these effects, whereas simple flavonoid frameworks or cyclization of isopentenyl groups all diminished anti-inflammatory activity.
Collapse
Affiliation(s)
- Yingjie Hu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Mengjun Su
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yichao Kong
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Caihong Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yaxia Yuan
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, TX 78229, USA
| | - Xiabin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Lei Ma
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| |
Collapse
|
49
|
Niccum BA, Coughlin S, Clay D, Heiman J, Buckley KH, Dungan M, Daniel MG, Ruiz J, Maxwell KN, Domchek SM, Leung G, Ahmad NA, Ginsberg GG, Kochman ML, Katona BW. Prevalence of H. pylori and Gastric Intestinal Metaplasia in BRCA1 and BRCA2 Carriers. Cancer Prev Res (Phila) 2024; 17:305-309. [PMID: 38641403 DOI: 10.1158/1940-6207.capr-24-0039] [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: 01/25/2024] [Revised: 04/01/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
BRCA1 and BRCA2 carriers may be at increased risk for gastric cancer; however, the mechanisms of gastric carcinogenesis remain poorly understood. We sought to determine the prevalence of gastric cancer risk factors Helicobacter pylori (H. pylori) infection and gastric intestinal metaplasia (GIM) among BRCA1/2 carriers to gain insight into the pathogenesis of gastric cancer in this population. A total of 100 unselected BRCA1/2 carriers who underwent endoscopic ultrasound from March 2022 to March 2023 underwent concomitant upper endoscopy with nontargeted gastric antrum and body biopsies. The study population (70% women; mean age 60.1 years) included 66% BRCA2 carriers. H. pylori was detected in one (1%) individual, 7 (7%) had GIM, 2 (2%) had autoimmune atrophic gastritis, and no gastric cancers were diagnosed. Among BRCA1/2 carriers, H. pylori prevalence was low and GIM prevalence was similar to that in the general population; however, identification of H. pylori or GIM may help inform future gastric cancer risk management strategies in BRCA1/2 carriers. Prevention Relevance: Evaluating the burden of H. pylori infection and GIM among BRCA1/2 carriers is warranted to better understand the mechanisms of gastric carcinogenesis and to help inform risk management strategies for gastric cancer among this at-risk population.
Collapse
Affiliation(s)
- Blake A Niccum
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sarah Coughlin
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel Clay
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jordan Heiman
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kole H Buckley
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michaela Dungan
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael G Daniel
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jose Ruiz
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kara N Maxwell
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Susan M Domchek
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Galen Leung
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nuzhat A Ahmad
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gregory G Ginsberg
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael L Kochman
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bryson W Katona
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
50
|
Lewis D, Jimenez L, Mansour MH, Horton S, Wong WWL. A Systematic Review of Cost-Effectiveness Studies on Gastric Cancer Screening. Cancers (Basel) 2024; 16:2353. [PMID: 39001415 PMCID: PMC11240801 DOI: 10.3390/cancers16132353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024] Open
Abstract
Gastric cancer (GC) poses notable economic and health burdens in settings where the incidence of disease is prevalent. Some countries have established early screening and treatment programs to address these challenges. The objectives of this systematic review were to summarize the cost-effectiveness of gastric cancer screening presented in the literature and to identify the critical factors that influence the cost-effectiveness of screening. This systematic review followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines. Economic evaluation studies of gastric cancer screening were reviewed from SCOPUS and PubMed. The Consolidated Health Economic Evaluation Reporting Standards 2022 (CHEERS 2022) was used to assess the quality of reporting presented in the selected articles. Only primary economic evaluation studies addressing the cost-effectiveness, cost-utility, and cost-benefit of gastric cancer screening were selected. Two reviewers scrutinized the selected articles (title, abstract, and full text) to determine suitability for the systematic review based on inclusion and exclusion criteria. Authors' consensus was relied on where disagreements arose. The main outcome measures of concern in the systematic review were cost, effectiveness (as measured by either quality-adjusted life years (QALY) or life-years saved (LYS)), and incremental cost-effectiveness ratio (ICER) of screening versus either no screening or an alternative screening method. Thirty-one studies were selected for the final review. These studies investigated the cost-effectiveness of GC screening based on either primary, secondary, or a combination of primary and secondary interventions. The main primary intervention was Helicobacter pylori (Hp) screening with eradication, while the main secondary intervention was endoscopic screening. Cost-effectiveness was evaluated against no screening or screening using an alternative method in both observational and model-based studies. Screening was mainly cost-effective in Asian countries or their diasporas where the prevalence of GC was high. GC screening was generally not cost-effective among Western countries. GC screening can be cost-effective, but cost-effectiveness is dependent on context-specific factors, including geographical location, the prevalence of GC in the local population, and the screening tool adopted. However, there is benefit in targeting high-risk population groups in Asian countries and their diaspora for GC screening.
Collapse
Affiliation(s)
- Diedron Lewis
- School of Pharmacy, University of Waterloo, Waterloo, ON N2G 1C5, Canada
| | - Laura Jimenez
- Department of Community Health and Epidemiology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Manel Haj Mansour
- Department of Haematology and Oncology, Aga Khan University Hospital, Nairobi P.O. Box 30270-00100, Kenya
| | - Susan Horton
- School of Public Health Sciences, University of Waterloo, Waterloo, ON N2L 3G5, Canada
| | - William W L Wong
- School of Pharmacy, University of Waterloo, Waterloo, ON N2G 1C5, Canada
| |
Collapse
|