1
|
Liu H, Song S, Geng X, Chen R, Zheng R, Wei W, Li N, Chen W, Ren J. Gastric Mucosal Status and Risk of Gastric Cancer in High-Risk Individuals: A Population-Based, Multicenter Prospective Study in China. Cancer Med 2024; 13:e70331. [PMID: 39403979 PMCID: PMC11474280 DOI: 10.1002/cam4.70331] [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: 09/24/2024] [Accepted: 09/28/2024] [Indexed: 10/19/2024] Open
Abstract
OBJECTIVE This study explored the distributions of gastric mucosal status and their links to gastric cancer within urban high-risk individuals in China. METHODS From 2014 to 2015, a questionnaire survey was administered among individuals aged 40 to 69 years, residing for over 3 years in seven selected cities across China. Utilizing the questionnaire data, high-risk individuals for gastric cancer were screened. These identified high-risk individuals were invited to undergo endoscopy, followed by pathological examinations conducted for those with suspicious lesions. Prior cancer patients and those newly diagnosed with gastric cancer were excluded, and the remaining endoscopic participants were prospectively followed up until December 2021. RESULTS The prospective study followed 8911 individuals at high risk of gastric cancer for a median duration of 6.77 years. The incidence density of gastric cancer was 58.55 per 100,000 person-years. At baseline, the distributions of subjects across normal gastric mucosa, gastritis/ulcer/polyp, atrophic gastritis/intestinal metaplasia, and intraepithelial neoplasia groups were 47.09%, 36.27%, 9.57%, and 7.07%, respectively. Compared with normal gastric mucosa, the hazard ratios (HRs) for gastric cancer in the other groups were 0.89 [95% confidence interval (95%CI): 0.38-2.08], 1.52 (95%CI: 0.50-4.66), and 4.63 (95%CI: 1.98-10.82). When using the 40-54 age group as the reference, the HR for 55-69 age group was 3.49 (95%CI: 1.52-7.98). CONCLUSION Among high-risk individuals with gastric cancer, the proportions of individuals exhibiting different gastric mucosal status inversely correlated with the severity of mucosal lesions, whereas the risk of gastric cancer progressively escalated with the increasing severity of mucosal lesions and advancing age.
Collapse
Affiliation(s)
- Han Liu
- Office of Cancer ScreeningNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Song Song
- Office of Cancer ScreeningNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xianyi Geng
- Office of Cancer ScreeningNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ru Chen
- National Central Cancer RegistryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Rongshou Zheng
- National Central Cancer RegistryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Wenqiang Wei
- National Central Cancer RegistryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ni Li
- Office of Cancer ScreeningNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Wanqing Chen
- Office of Cancer ScreeningNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jiansong Ren
- Office of Cancer ScreeningNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
2
|
Hahn AI, Mülder DT, Huang RJ, Zhou MJ, Blake B, Omofuma O, Murphy JD, Gutiérrez-Torres DS, Zauber AG, O'Mahony JF, Camargo MC, Ladabaum U, Yeh JM, Hur C, Lansdorp-Vogelaar I, Meester R, Laszkowska M. Global Progression Rates of Precursor Lesions for Gastric Cancer: A Systematic Review and Meta-Analysis. Clin Gastroenterol Hepatol 2024:S1542-3565(24)00864-4. [PMID: 39362617 DOI: 10.1016/j.cgh.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/27/2024] [Accepted: 09/04/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND & AIMS Whether gastric cancer (GC) precursor lesions progress to invasive cancer at similar rates globally remains unknown. We conducted a systematic review and meta-analysis to determine the progression of precursor lesions to GC in countries with low versus medium/high incidence. METHODS We searched relevant databases for studies reporting the progression of endoscopically confirmed precursor lesions to GC. Studies were stratified by low (<6 per 100,000) or medium/high (≥6 per 100,000) GC incidence countries. Random-effects models were used to estimate the progression rates of atrophic gastritis (AG), intestinal metaplasia (IM), and dysplasia to GC per 1000 person-years. RESULTS Among the 5829 studies identified, 44 met our inclusion criteria. The global pooled estimates of the progression rate per 1000 person-years were 2.09 (95% confidence interval, 1.46-2.99), 2.89 (2.03-4.11), and 10.09 (5.23-19.49) for AG, IM, and dysplasia, respectively. The estimated progression rates per 1000 person-years for low versus medium/high GC incidence countries, respectively, were 0.97 (0.86-1.10) versus 2.47 (1.70-2.99) for AG (P < .01), 2.37 (1.43-3.92) versus 3.47 (2.13-5.65) for IM (P = .29), and 5.51 (2.92-10.39) versus 14.80 (5.87-37.28) for dysplasia (P = .08). There were no differences for progression of AG between groups when high-quality studies were compared. CONCLUSIONS Similar progression rates of IM and dysplasia were observed among low and medium/high GC incidence countries. This suggests that the potential benefits of surveillance for these lesions in low-risk regions may be comparable with those of population-wide interventions in high-risk regions. Further prospective studies are needed to confirm these findings and inform global screening and surveillance guidelines.
Collapse
Affiliation(s)
- Anne I Hahn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Duco T Mülder
- Department of Public Health, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Robert J Huang
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Margaret J Zhou
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Benjamin Blake
- Weill Cornell Medical College of Cornell University, New York, New York
| | - Omonefe Omofuma
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - John D Murphy
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | | | - Ann G Zauber
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - James F O'Mahony
- Department of Public Health, Erasmus Medical Center, Rotterdam, the Netherlands; School of Economics, University College Dublin, Dublin, Ireland
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Uri Ladabaum
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Jennifer M Yeh
- Department of Pediatrics, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts
| | - Chin Hur
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | | | - Reinier Meester
- Department of Public Health, Erasmus Medical Center, Rotterdam, the Netherlands; Health Economics & Outcomes Research, Freenome Holdings Inc, San Francisco, California
| | - Monika Laszkowska
- Gastroenterology, Hepatology, and Nutrition Service, Department of Subspecialty Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
3
|
Li F, Wang Y, Ping X, Yin JC, Wang F, Zhang X, Li X, Zhai J, Shen L. Molecular evolution of intestinal-type early gastric cancer according to Correa cascade. J Biomed Res 2024; 38:1-16. [PMID: 39314047 DOI: 10.7555/jbr.38.20240118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024] Open
Abstract
Early screening is crucial for the prevention of intestinal-type gastric cancer. The objective of the current study was to ascertain molecular evolution of intestinal-type gastric cancer according to the Correa cascade for the precise gastric cancer screening. We collected sequential lesions of the Correa cascade in the formalin-fixed and paraffin-embedded endoscopic submucosal dissection-resected specimens from 14 Chinese patients by microdissection, and subsequently determined the profiles of somatic aberrations during gastric carcinogenesis using the whole exome sequencing, identifying multiple variants at different Correa stages. The results showed that TP53, PCLO, and PRKDC were the most frequently mutated genes in the early gastric cancer (EGC). A high frequency of TP53 alterations was found in low-grade intraepithelial neoplasia (LGIN), which further increased in high-grade intraepithelial neoplasia (HGIN) and EGC. Intestinal metaplasia (IM) had no significant correlation with EGC in terms of mutational spectra, whereas both LGIN and HGIN showed higher genomic similarities to EGC, compared with IM. Based on Jaccard similarity coefficients, three evolutionary models were further constructed, and most patients showed linear progression from LGIN to HGIN, ultimately resulting in EGC. The ECM-receptor interaction pathway was revealed to be involved in the linear evolution. Additionally, the retrospective validation study of 39 patients diagnosed with LGIN indicated that PRKDC mutations, in addition to TP53 mutations, may drive LGIN progression to HGIN or EGC. In conclusion, the current study unveils the genomic evolution across the Correa cascade of intestinal-type gastric cancer, elucidates the underlying molecular mechanisms of gastric carcinogenesis, and provides some evidence for potential personalized gastric cancer surveillance.
Collapse
Affiliation(s)
- Fangyuan Li
- Digestive Endoscopy Center, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Yaohui Wang
- Department of Pathology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Xiaochun Ping
- Department of General Surgery, the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jiani C Yin
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu 210061, China
| | - Fufeng Wang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu 210061, China
| | - Xian Zhang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu 210061, China
| | - Xiang Li
- Department of Surgical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Jing Zhai
- Department of Surgical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Lizong Shen
- Department of General Surgery, the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Department of Surgical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| |
Collapse
|
4
|
He S, Zhang Z, Song G, Wang Z, Dai C, Yan S, Jiang K, Song B, Li H, Cao M, Sun D, Yang F, Yan X, Zhang S, Teng Y, Li Q, Xia C, Chen W. Can patients with mild non-neoplastic lesions diagnosed at baseline screening be safely exempt from surveillance: evidence from multicenter community-based cohorts. SCIENCE CHINA. LIFE SCIENCES 2024:10.1007/s11427-023-2558-x. [PMID: 39254888 DOI: 10.1007/s11427-023-2558-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/25/2024] [Indexed: 09/11/2024]
Abstract
Surveillance recommendations for gastric cancer (GC) in current guidelines focused on advanced precancerous lesions and were based on precise diagnosis of severity/extent of baseline lesions. We aimed to develop a less endoscopy-related equipment-dependent risk-stratification tool, and assessed whether mild-precursor-lesion patients can be safely exempt from surveillance. In the multicenter community-based cohort, 75,051 participants receiving baseline endoscopy were enrolled during 2015-2017 and followed-up until 2021. Cumulative incidence rates (CIRs) of GC for precancerous-conditions were calculated by Kaplan-Meier method and compared by Log-rank tests. Mixed-effects Cox regression models were used to detect potential factors for progression towards GC. A risk score was calculated as counts of selected factors. An independent cohort, including 26,586 participants was used for external validation. During a median follow-up of 6.25 years, CIRs of GC were 0.302%, 0.436%, and 4.756% for normal group, non-neoplastic (atrophic gastritis/intestinal metaplasia) and neoplastic lesions (low-grade/high-grade dysplasia), respectively (Ptrend<0.001). Four predictors, including male, ⩾60 years, smoking, and limited vegetable consumption, were selected for risk-stratification. High-risk patients (⩾3 risk factors) with non-neoplastic lesions showed higher GC risks (adjusted HR=7.73, 95%CI: 4.29-13.92), and their four-year CIR reached the one-year CIR of neoplastic lesions. Further categorizing non-neoplastic lesions by histological grade, both patients with moderate-to-severe lesions (aHR=3.07, 95%CI: 1.67-5.64) and high-risk patients with mild lesions (aHR=7.29, 95%CI: 3.58-14.86) showed higher risks. Consistent trends were observed in validation cohort. High-risk mild-precursor-lesion patients should receive surveillance within 3-5 years after baseline screening. Our study provides evidence on supplementing current guideline recommendations.
Collapse
Affiliation(s)
- Siyi He
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhiyi Zhang
- Department of Gastroenterology, Gansu Wuwei Tumor Hospital, Wuwei, 730000, China
| | - Guohui Song
- Cixian Cancer Institute, Handan, 056500, China
| | | | - Chunyun Dai
- Center for Disease Control and Prevention of Sheyang County, Yancheng, 224499, China
| | - Shipeng Yan
- Department of Cancer Prevention and Control, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410029, China
| | - Kun Jiang
- Luoshan Center for Disease Control and Prevention, Xinyang, 464299, China
| | - Bingbing Song
- Office for Cancer Control and Research, Affiliated Cancer Hospital of Harbin Medical University, Harbin, 150081, China
| | - He Li
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Maomao Cao
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dianqin Sun
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Fan Yang
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xinxin Yan
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shaoli Zhang
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yi Teng
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qianru Li
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Changfa Xia
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wanqing Chen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
5
|
Liu Y, Gu K. Association between anesthesia assistance and precancerous lesions and early cancer detection during diagnostic esophagogastroduodenoscopy: a propensity score-matched retrospective study. Front Med (Lausanne) 2024; 11:1389809. [PMID: 39114825 PMCID: PMC11303204 DOI: 10.3389/fmed.2024.1389809] [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: 02/22/2024] [Accepted: 07/15/2024] [Indexed: 08/10/2024] Open
Abstract
Background Esophagogastroduodenoscopy (EGD) is a fundamental procedure for early detection of upper gastrointestinal (UGI) cancer. However, limited research has been conducted on the impact of sedation during EGD on the identification of precancerous lesions and early cancer (EC). This retrospective study aims to evaluate whether sedation during EGD can improve the detection rates of precancerous lesions and EC. Methods In this propensity score-matched retrospective study, we examined medical records from outpatients who underwent diagnostic EGD at a large tertiary center between January 2023 and December 2023. Data on endoscopic findings and histology biopsies were obtained from an endoscopy quality-control system. The primary objective was to compare the rates of detecting precancerous lesions and EC in patients who received sedation during EGD vs. those who did not receive sedation. Additionally, we aimed to identify factors influencing these detection rates using binary logistic regression analysis. Results Following propensity score matching, a total of 17,862 patients who underwent diagnostic EGD with or without propofol sedation were identified. The group that received sedation exhibited a higher detection rate of precancerous lesions and EC in comparison to the non-sedated group (1.04 vs. 0.75%; p = 0.039). Additionally, within the sedated group, there was an increased likelihood of identifying precancerous lesions and EC specifically at the gastric antrum (0.60 vs. 0.32%, p = 0.006). Binary logistic regression analysis demonstrated that independent risk factors influencing the detection rates included age, gender, observation time, and number of biopsies conducted during the procedure. Conclusion Anesthesia assistance during EGD screening proved advantageous in detecting EC as well as precancerous lesions. It is crucial for endoscopists to consider these factors when performing EGD screening procedures.
Collapse
Affiliation(s)
| | - Kaier Gu
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
6
|
Zhou DS, Zhang WJ, Song SY, Hong XX, Yang WQ, Li JJ, Xu JQ, Kang JY, Cai TT, Xu YF, Guo SJ, Pan HF, Li HW. Weiwei Decoction alleviates gastric intestinal metaplasia through the olfactomedin 4/nucleotide-binding oligomerization domain 1/caudal-type homeobox gene 2 signaling pathway. World J Gastrointest Oncol 2024; 16:3211-3229. [PMID: 39072182 PMCID: PMC11271767 DOI: 10.4251/wjgo.v16.i7.3211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/25/2024] [Accepted: 05/11/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Gastric intestinal metaplasia (IM) is a precancerous lesion that is associated with an elevated risk of gastric carcinogenesis. Weiwei Decoction (WWD) is a promising traditional Chinese herbal formula widely employed in clinical for treating IM. Previous studies suggested the potential involvement of the olfactomedin 4 (OLFM4)/nucleotide-binding oligomerization domain 1 (NOD1)/caudal-type homeobox gene 2 (CDX2) signaling pathway in IM regulation. AIM To verify the regulation of the OLFM4/NOD1/CDX2 pathway in IM, specifically investigating WWD's effectiveness on IM through this pathway. METHODS Immunohistochemistry for OLFM4, NOD1, and CDX2 was conducted on tissue microarray. GES-1 cells treated with chenodeoxycholic acid were utilized as IM cell models. OLFM4 short hairpin RNA (shRNA), NOD1 shRNA, and OLFM4 pcDNA were transfected to clarify the pathway regulatory relationships. Protein interactions were validated by co-immunoprecipitation. To explore WWD's pharmacological actions, IM rat models were induced using N-methyl-N'-nitro-N-nitrosoguanidine followed by WWD gavage. Gastric cells were treated with WWD-medicated serum. Cytokines and chemokines content were assessed by enzyme-linked immunosorbent assay and quantitative reverse transcription polymerase chain reaction. RESULTS The OLFM4/NOD1/CDX2 axis was a characteristic of IM. OLFM4 exhibited direct binding and subsequent down-regulation of NOD1, thereby sustaining the activation of CDX2 and promoting the progression of IM. WWD improved gastric mucosal histological lesions while suppressing intestinal markers KLF transcription factor 4, villin 1, and MUCIN 2 expression in IM rats. Regarding pharmacological actions, WWD suppressed OLFM4 and restored NOD1 expression, consequently reducing CDX2 at the mRNA and protein levels in IM rats. Parallel regulatory mechanisms were observed at the protein level in IM cells treated with WWD-medicated serum. Furthermore, WWD-medicated serum treatment strengthened OLFM4 and NOD1 interaction. In case of anti-inflammatory, WWD restrained interleukin (IL)-6, interferon-gamma, IL-17, macrophage chemoattractant protein-1, macrophage inflammatory protein 1 alpha content in IM rat serum. WWD-medicated serum inhibited tumor necrosis factor alpha, IL-6, IL-8 transcriptions in IM cells. CONCLUSION The OLFM4/NOD1/CDX2 pathway is involved in the regulation of IM. WWD exerts its therapeutic efficacy on IM through the pathway, additionally attenuating the inflammatory response.
Collapse
Affiliation(s)
- Di-Shu Zhou
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, China
| | - Wei-Jian Zhang
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, China
| | - Shu-Ya Song
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, China
| | - Xin-Xin Hong
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| | - Wei-Qin Yang
- Department of Chinese Medicine, The Eight Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518033, Guangdong Province, China
| | - Juan-Juan Li
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| | - Jian-Qu Xu
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| | - Jian-Yuan Kang
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| | - Tian-Tian Cai
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| | - Yi-Fei Xu
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| | - Shao-Ju Guo
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| | - Hua-Feng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, China
| | - Hai-Wen Li
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| |
Collapse
|
7
|
Wei H, Li W, Zeng L, Ding N, Li K, Yu H, Jiang F, Yin H, Xia Y, Deng C, Cai N, Chen X, Gu L, Chen H, Zhang F, He Y, Li J, Zhang C. OLFM4 promotes the progression of intestinal metaplasia through activation of the MYH9/GSK3β/β-catenin pathway. Mol Cancer 2024; 23:124. [PMID: 38849840 PMCID: PMC11157765 DOI: 10.1186/s12943-024-02016-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/04/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Intestinal metaplasia (IM) is classified into complete intestinal metaplasia (CIM) and incomplete intestinal metaplasia (IIM). Patients diagnosed with IIM face an elevated susceptibility to the development of gastric cancer, underscoring the critical need for early screening measures. In addition to the complexities associated with diagnosis, the exact mechanisms driving the progression of gastric cancer in IIM patients remain poorly understood. OLFM4 is overexpressed in several types of tumors, including colorectal, gastric, pancreatic, and ovarian cancers, and its expression has been associated with tumor progression. METHODS In this study, we used pathological sections from two clinical centers, biopsies of IM tissues, precancerous lesions of gastric cancer (PLGC) cell models, animal models, and organoids to explore the role of OLFM4 in IIM. RESULTS Our results show that OLFM4 expression is highly increased in IIM, with superior diagnostic accuracy of IIM when compared to CDX2 and MUC2. OLFM4, along with MYH9, was overexpressed in IM organoids and PLGC animal models. Furthermore, OLFM4, in combination with Myosin heavy chain 9 (MYH9), accelerated the ubiquitination of GSK3β and resulted in increased β-catenin levels through the Wnt signaling pathway, promoting the proliferation and invasion abilities of PLGC cells. CONCLUSIONS OLFM4 represents a novel biomarker for IIM and could be utilized as an important auxiliary means to delimit the key population for early gastric cancer screening. Finally, our study identifies cell signaling pathways involved in the progression of IM.
Collapse
Affiliation(s)
- Hongfa Wei
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Jinping, Shantou, Guangdong, 515041, P.R. China
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Wenchao Li
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- The Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Leli Zeng
- Scientific Research Center, The Biobank, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, P.R. China
| | - Ni Ding
- Scientific Research Center, The Biobank, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, P.R. China
- The Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Kuan Li
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Hong Yu
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Fei Jiang
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Haofan Yin
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- Department of Laboratory Medicine, Shenzhen People's Hospital, (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Yu Xia
- Scientific Research Center, The Biobank, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, P.R. China
| | - Cuncan Deng
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Nan Cai
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Xiancong Chen
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Liang Gu
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Huanjie Chen
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Jinping, Shantou, Guangdong, 515041, P.R. China
| | - Feiran Zhang
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Jinping, Shantou, Guangdong, 515041, P.R. China.
| | - Yulong He
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Jia Li
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
- Scientific Research Center, The Biobank, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, P.R. China.
| | - Changhua Zhang
- Department of Gastrointestinal Surgery, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
8
|
Huang RJ, Huang ES, Mudiganti S, Chen T, Martinez MC, Ramrakhiani S, Han SS, Hwang JH, Palaniappan LP, Liang SY. Risk of Gastric Adenocarcinoma in a Multiethnic Population Undergoing Routine Care: An Electronic Health Records Cohort Study. Cancer Epidemiol Biomarkers Prev 2024; 33:547-556. [PMID: 38231023 PMCID: PMC10990787 DOI: 10.1158/1055-9965.epi-23-1200] [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: 09/29/2023] [Revised: 12/05/2023] [Accepted: 01/12/2024] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Gastric adenocarcinoma (GAC) is often diagnosed at advanced stages and portends a poor prognosis. We hypothesized that electronic health records (EHR) could be leveraged to identify individuals at highest risk for GAC from the population seeking routine care. METHODS This was a retrospective cohort study, with endpoint of GAC incidence as ascertained through linkage to an institutional tumor registry. We utilized 2010 to 2020 data from the Palo Alto Medical Foundation, a large multispecialty practice serving Northern California. The analytic cohort comprised individuals ages 40-75 receiving regular ambulatory care. Variables collected included demographic, medical, pharmaceutical, social, and familial data. Electronic phenotyping was based on rule-based methods. RESULTS The cohort comprised 316,044 individuals and approximately 2 million person-years (p-y) of observation. 157 incident GACs occurred (incidence 7.9 per 100,000 p-y), of which 102 were non-cardia GACs (incidence 5.1 per 100,000 p-y). In multivariable analysis, male sex [HR: 2.2, 95% confidence interval (CI): 1.6-3.1], older age, Asian race (HR: 2.5, 95% CI: 1.7-3.7), Hispanic ethnicity (HR: 1.9, 95% CI: 1.1-3.3), atrophic gastritis (HR: 4.6, 95% CI: 2.2-9.3), and anemia (HR: 1.9, 95% CI: 1.3-2.6) were associated with GAC risk; use of NSAID was inversely associated (HR: 0.3, 95% CI: 0.2-0.5). Older age, Asian race, Hispanic ethnicity, atrophic gastritis, and anemia were associated with non-cardia GAC. CONCLUSIONS Routine EHR data can stratify the general population for GAC risk. IMPACT Such methods may help triage populations for targeted screening efforts, such as upper endoscopy.
Collapse
Affiliation(s)
- Robert J Huang
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Edward S Huang
- Department of Gastroenterology, Palo Alto Medical Foundation, San Jose, California
| | - Satish Mudiganti
- Palo Alto Medical Foundation Research Institute, Palo Alto Medical Foundation, Palo Alto, California
| | - Tony Chen
- Palo Alto Medical Foundation Research Institute, Palo Alto Medical Foundation, Palo Alto, California
| | - Meghan C Martinez
- Palo Alto Medical Foundation Research Institute, Palo Alto Medical Foundation, Palo Alto, California
| | - Sanjay Ramrakhiani
- Department of Gastroenterology, Palo Alto Medical Foundation, San Jose, California
| | - Summer S Han
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Joo Ha Hwang
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Latha P Palaniappan
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California
| | - Su-Ying Liang
- Palo Alto Medical Foundation Research Institute, Palo Alto Medical Foundation, Palo Alto, California
| |
Collapse
|
9
|
Fansiwala K, Qian Y, Liang PS. Gastric Cancer Risk Factors in a Veteran Population. Mil Med 2024; 189:e802-e808. [PMID: 37610320 DOI: 10.1093/milmed/usad319] [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: 02/26/2023] [Revised: 07/10/2023] [Accepted: 08/01/2023] [Indexed: 08/24/2023] Open
Abstract
INTRODUCTION Risk factors for gastric cancer in the United States are not well understood, especially in populations with a low proportion of immigrants. We conducted a matched case-control study in a Veteran Affairs Medical Center to identify risk factors for gastric cancer. MATERIALS AND METHODS Gastric cancer patients and age- and sex-matched controls were identified in a 1:4 ratio from January 1, 1997 to October 31, 2018. Demographic, medical, endoscopic, and histologic data were extracted. We performed conditional logistic regression to estimate odds ratios and 95% CIs for associations between potential risk factors and gastric cancer. RESULTS Most gastric cancer cases were diagnosed on initial endoscopy (71.4%). Of these, the most common presenting stage was stage IV (40.8%). Risk factors for gastric cancer included Black and Asian race and never or current (compared to former) drinkers, although Helicobacter pylori eradication and pernicious anemia were associated with decreased risk. CONCLUSIONS The high proportion of late-stage gastric cancer diagnoses highlights the need for improved risk stratification as well as screening and surveillance protocols in the U.S. population. Racial disparities among veterans in an equal-access system necessitate further investigation into the etiology of these disparities.
Collapse
Affiliation(s)
- Kush Fansiwala
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Yingzhi Qian
- Department of Population Health, NYU Langone Health, New York, NY 10016, USA
| | - Peter S Liang
- Department of Population Health, NYU Langone Health, New York, NY 10016, USA
- Department of Medicine, VA New York Harbor Health Care System, New York, NY 10010, USA
- Department of Medicine, NYU Langone Health, New York, NY 10016, USA
| |
Collapse
|
10
|
Rugge M, Genta RM, Malfertheiner P, Dinis-Ribeiro M, El-Serag H, Graham DY, Kuipers EJ, Leung WK, Park JY, Rokkas T, Schulz C, El-Omar EM. RE.GA.IN.: the Real-world Gastritis Initiative-updating the updates. Gut 2024; 73:407-441. [PMID: 38383142 DOI: 10.1136/gutjnl-2023-331164] [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: 09/18/2023] [Accepted: 12/18/2023] [Indexed: 02/23/2024]
Abstract
At the end of the last century, a far-sighted 'working party' held in Sydney, Australia addressed the clinicopathological issues related to gastric inflammatory diseases. A few years later, an international conference held in Houston, Texas, USA critically updated the seminal Sydney classification. In line with these initiatives, Kyoto Global Consensus Report, flanked by the Maastricht-Florence conferences, added new clinical evidence to the gastritis clinicopathological puzzle.The most relevant topics related to the gastric inflammatory diseases have been addressed by the Real-world Gastritis Initiative (RE.GA.IN.), from disease definitions to the clinical diagnosis and prognosis. This paper reports the conclusions of the RE.GA.IN. consensus process, which culminated in Venice in November 2022 after more than 8 months of intense global scientific deliberations. A forum of gastritis scholars from five continents participated in the multidisciplinary RE.GA.IN. consensus. After lively debates on the most controversial aspects of the gastritis spectrum, the RE.GA.IN. Faculty amalgamated complementary knowledge to distil patient-centred, evidence-based statements to assist health professionals in their real-world clinical practice. The sections of this report focus on: the epidemiology of gastritis; Helicobacter pylori as dominant aetiology of environmental gastritis and as the most important determinant of the gastric oncogenetic field; the evolving knowledge on gastric autoimmunity; the clinicopathological relevance of gastric microbiota; the new diagnostic horizons of endoscopy; and the clinical priority of histologically reporting gastritis in terms of staging. The ultimate goal of RE.GA.IN. was and remains the promotion of further improvement in the clinical management of patients with gastritis.
Collapse
Affiliation(s)
- Massimo Rugge
- Department of Medicine-DIMED, University of Padova, Padua, Italy
- Azienda Zero, Veneto Tumour Registry, Padua, Italy
| | - Robert M Genta
- Gastrointestinal Pathology, Inform Diagnostics Research Institute, Dallas, Texas, USA
- Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - Peter Malfertheiner
- Medizinische Klinik und Poliklinik II, Ludwig Maximilian Universität Klinikum München, Munich, Germany
- Klinik für Gastroenterologie, Hepatologie und Infektiologie, Otto-von-Guericke Universität Magdeburg, Magdeburg, Germany
| | - Mario Dinis-Ribeiro
- Porto Comprehensive Cancer Center & RISE@CI-IPO, University of Porto, Porto, Portugal
- Gastroenterology Department, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Hashem El-Serag
- Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
- Houston VA Health Services Research & Development Center of Excellence, Michael E DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| | - David Y Graham
- Department of Medicine, Michael E DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| | - Ernst J Kuipers
- Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Jin Young Park
- International Agency for Research on Cancer, Lyon, France
| | - Theodore Rokkas
- Gastroenterology, Henry Dunant Hospital Center, Athens, Greece
| | | | - Emad M El-Omar
- Microbiome Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
11
|
Ferreira AI, Lima Capela T, Macedo Silva V, Xavier S, Boal Carvalho P, Magalhães J, Cotter J. Gastric dysplasia in random biopsies: the influence of Helicobacter pylori infection and alcohol consumption in the presence of a lesion. Scand J Gastroenterol 2024; 59:125-132. [PMID: 37872792 DOI: 10.1080/00365521.2023.2272563] [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: 06/06/2023] [Accepted: 10/14/2023] [Indexed: 10/25/2023]
Abstract
BACKGROUND Gastric dysplasia in the absence of an endoscopically defined lesion is rare, usually either a false positive diagnosis or a previously unidentified precancerous lesion during esophagogastroduodenoscopy (EGD). AIMS Evaluate factors associated with the presence of an endoscopically visible lesion during follow-up in patients with histologic diagnosis of gastric dysplasia in random biopsies. METHODS Retrospective cohort study including patients referred to our institution for gastric dysplasia in random biopsies during Index EGD. Endoscopic evaluation was performed with a high-definition endoscope using narrow band imaging (HD EGD-0). If no lesion was detected, endoscopic surveillance (HD EGD-FU) was conducted within 6 months for high grade dysplasia (HGD) or 12 months for low grade (LGD) or indefinite for dysplasia (IFD). RESULTS From a total sample of 96 patients, 5 (5.2%) presented with an endoscopically visible lesion during HD EGD-0, while 10 lesions (10.4%) were identified during HD EGD-FU. Patients with Helicobacter pylori infection at Index EDG and with regular alcohol consumption (≥25 g/day) were 8 and 4 times more likely to have an endoscopically visible lesion on HD EGD-FU (p = 0.012 and p = 0.047). In binary logistic regression, both factors were independent predictors of the presence of gastric lesion on HD EGD-FU (OR 9.284, p = 0.009 and OR 5.025, p = 0.033). CONCLUSIONS The presence of an endoscopically visible lesion after the histologic diagnosis of gastric dysplasia in random biopsies was more frequent during HD EGD-FU. H. pylori infection at Index EGD and regular alcohol consumption were significant predictors of the presence of gastric lesion on HD EGD-FU.
Collapse
Affiliation(s)
- Ana Isabel Ferreira
- Gastroenterology Department, Hospital Senhora da Oliveira, Guimarães, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Guimarães, Braga, Portugal
| | - Tiago Lima Capela
- Gastroenterology Department, Hospital Senhora da Oliveira, Guimarães, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Guimarães, Braga, Portugal
| | - Vítor Macedo Silva
- Gastroenterology Department, Hospital Senhora da Oliveira, Guimarães, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Guimarães, Braga, Portugal
| | - Sofia Xavier
- Gastroenterology Department, Hospital Senhora da Oliveira, Guimarães, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Guimarães, Braga, Portugal
| | - Pedro Boal Carvalho
- Gastroenterology Department, Hospital Senhora da Oliveira, Guimarães, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Guimarães, Braga, Portugal
| | - Joana Magalhães
- Gastroenterology Department, Hospital Senhora da Oliveira, Guimarães, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Guimarães, Braga, Portugal
| | - José Cotter
- Gastroenterology Department, Hospital Senhora da Oliveira, Guimarães, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Guimarães, Braga, Portugal
| |
Collapse
|
12
|
Milito C, Pulvirenti F, Garzi G, Sculco E, Cinetto F, Firinu D, Lagnese G, Punziano A, Discardi C, Costanzo G, Felice C, Spadaro G, Ferrari S, Quinti I. Decline of gastric cancer mortality in common variable immunodeficiency in the years 2018-2022. Front Immunol 2023; 14:1231242. [PMID: 37868983 PMCID: PMC10587402 DOI: 10.3389/fimmu.2023.1231242] [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: 05/30/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Introduction In patients with Common Variable Immunodeficiency, malignancy has been reported as the leading cause of death in adults, with a high risk of B-cell lymphomas and gastric cancer. Methods We conducted a five-year prospective study aiming to update the incidence and mortality of gastric cancer and the incidence of gastric precancerous lesions in 512 CVID patients who underwent a total of 400 upper gastrointestinal endoscopies. Results In the pre-pandemic period, 0.58 endoscopies were performed per patient/year and in the COVID-19 period, 0.39 endoscopies were performed per patient/year. Histology revealed areas with precancerous lesions in about a third of patients. Patients who had more than one gastroscopy during the study period were more likely to have precancerous lesions. Two patients received a diagnosis of gastric cancer in the absence of Helicobacter pylori infection. The overall prevalence of Helicobacter pylori infection in biopsy specimens was 19.8% and related only to active gastritis. Among patients who had repeated gastroscopies, about 20% progressed to precancerous lesions, mostly independent of Helicobacter pylori. Discussion While gastric cancer accounted for one in five deaths from CVID in our previous survey, no gastric cancer deaths were recorded in the past five years, likely consistent with the decline in stomach cancer mortality observed in the general population. However, during the COVID-19 pandemic, cancer screening has been delayed. Whether such a delay or true decline could be the reason for the lack of gastric cancer detection seen in CVID may become clear in the coming years. Due to the high incidence of precancerous lesions, we cannot rely on observed and predicted trends in gastric cancer mortality and strongly recommend tailored surveillance programs.
Collapse
Affiliation(s)
- Cinzia Milito
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Federica Pulvirenti
- Reference Center for Primary Immune Deficiencies, AOU Policlinico Umberto I, Rome, Italy
| | - Giulia Garzi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Eleonora Sculco
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesco Cinetto
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Davide Firinu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Gianluca Lagnese
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Alessandra Punziano
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Claudia Discardi
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Giulia Costanzo
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Carla Felice
- Rare Diseases Referral Center, Internal Medicine 1, Ca’ Foncello Hospital, AULSS2 Marca Trevigiana, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Simona Ferrari
- Medical Genetics Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Isabella Quinti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
13
|
Huang RJ, Wichmann IA, Su A, Sathe A, Shum MV, Grimes SM, Meka R, Almeda A, Bai X, Shen J, Nguyen Q, Amieva MR, Hwang JH, Ji HP. A spatially mapped gene expression signature for intestinal stem-like cells identifies high-risk precursors of gastric cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.20.558462. [PMID: 37786704 PMCID: PMC10541579 DOI: 10.1101/2023.09.20.558462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Objective Gastric intestinal metaplasia (GIM) is a precancerous lesion that increases gastric cancer (GC) risk. The Operative Link on GIM (OLGIM) is a combined clinical-histopathologic system to risk-stratify patients with GIM. The identification of molecular biomarkers that are indicators for advanced OLGIM lesions may improve cancer prevention efforts. Methods This study was based on clinical and genomic data from four cohorts: 1) GAPS, a GIM cohort with detailed OLGIM severity scoring (N=303 samples); 2) the Cancer Genome Atlas (N=198); 3) a collation of in-house and publicly available scRNA-seq data (N=40), and 4) a spatial validation cohort (N=5) consisting of annotated histology slides of patients with either GC or advanced GIM. We used a multi-omics pipeline to identify, validate and sequentially parse a highly-refined signature of 26 genes which characterize high-risk GIM. Results Using standard RNA-seq, we analyzed two separate, non-overlapping discovery (N=88) and validation (N=215) sets of GIM. In the discovery phase, we identified 105 upregulated genes specific for high-risk GIM (defined as OLGIM III-IV), of which 100 genes were independently confirmed in the validation set. Spatial transcriptomic profiling revealed 36 of these 100 genes to be expressed in metaplastic foci in GIM. Comparison with bulk GC sequencing data revealed 26 of these genes to be expressed in intestinal-type GC. Single-cell profiling resolved the 26-gene signature to both mature intestinal lineages (goblet cells, enterocytes) and immature intestinal lineages (stem-like cells). A subset of these genes was further validated using single-molecule multiplex fluorescence in situ hybridization. We found certain genes (TFF3 and ANPEP) to mark differentiated intestinal lineages, whereas others (OLFM4 and CPS1) localized to immature cells in the isthmic/crypt region of metaplastic glands, consistent with the findings from scRNAseq analysis. Conclusions using an integrated multi-omics approach, we identified a novel 26-gene expression signature for high-OLGIM precursors at increased risk for GC. We found this signature localizes to aberrant intestinal stem-like cells within the metaplastic microenvironment. These findings hold important translational significance for future prevention and early detection efforts.
Collapse
Affiliation(s)
- Robert J. Huang
- Division of Gastroenterology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Ignacio A. Wichmann
- Division of Oncology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
- Division of Obstetrics and Gynecology, Department of Obstetrics, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - Andrew Su
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Anuja Sathe
- Division of Oncology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Miranda V. Shum
- Division of Gastroenterology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Susan M. Grimes
- Division of Oncology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Rithika Meka
- Division of Oncology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Alison Almeda
- Division of Oncology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Xiangqi Bai
- Division of Oncology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Jeanne Shen
- Department of Pathology, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Quan Nguyen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Manuel R. Amieva
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, 94305, USA
- Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
| | - Joo Ha Hwang
- Division of Gastroenterology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Hanlee P. Ji
- Division of Oncology, Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
14
|
Irie T, Yamada H, Takeuchi C, Liu YY, Charvat H, Shimazu T, Ando T, Maekita T, Abe S, Takamaru H, Kodama M, Murakami K, Sugimoto K, Sakamoto K, Ushijima T. The methylation level of a single cancer risk marker gene reflects methylation burden in gastric mucosa. Gastric Cancer 2023; 26:667-676. [PMID: 37219707 DOI: 10.1007/s10120-023-01399-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/12/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND Gastric cancer risk can be accurately predicted by measuring the methylation level of a single marker gene in gastric mucosa. However, the mechanism is still uncertain. We hypothesized that the methylation level measured reflects methylation alterations in the entire genome (methylation burden), induced by Helicobacter pylori (H. pylori) infection, and thus cancer risk. METHODS Gastric mucosa of 15 healthy volunteers without H. pylori infection (G1), 98 people with atrophic gastritis (G2), and 133 patients with gastric cancer (G3) after H. pylori eradication were collected. Methylation burden of an individual was obtained by microarray analysis as an inverse of the correlation coefficient between the methylation levels of 265,552 genomic regions in the person's gastric mucosa and those in an entirely healthy mucosa. RESULTS The methylation burden significantly increased in the order of G1 (n = 4), G2 (n = 18), and G3 (n = 19) and was well correlated with the methylation level of a single marker gene (r = 0.91 for miR124a-3). The average methylation levels of nine driver genes tended to increase according to the risk levels (P = 0.08 between G2 vs G3) and was also correlated with the methylation level of a single marker gene (r = 0.94). Analysis of more samples (14 G1, 97 G2, and 131 G3 samples) yielded significant increases of the average methylation levels between risk groups. CONCLUSIONS The methylation level of a single marker gene reflects the methylation burden, which includes driver gene methylation, and thus accurately predicts cancer risk.
Collapse
Affiliation(s)
- Takahiro Irie
- Department of Epigenomics, Institute for Advanced Life Sciences, Hoshi University, Tokyo, Japan
- Department of Coloproctological Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Harumi Yamada
- Department of Epigenomics, Institute for Advanced Life Sciences, Hoshi University, Tokyo, Japan
- Department of Surgery, Kyoto University, Kyoto, Japan
| | - Chihiro Takeuchi
- Department of Epigenomics, Institute for Advanced Life Sciences, Hoshi University, Tokyo, Japan
| | - Yu-Yu Liu
- Department of Epigenomics, Institute for Advanced Life Sciences, Hoshi University, Tokyo, Japan
| | - Hadrien Charvat
- Faculty of International Liberal Arts, Juntendo University, Tokyo, Japan
| | - Taichi Shimazu
- Division of Behavioral Sciences, National Cancer Center Institute for Cancer Control, National Cancer Center, Tokyo, Japan
| | - Takayuki Ando
- Third Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Takao Maekita
- Second Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
| | - Seiichiro Abe
- Endoscopy Division, National Cancer Center Hospital, Tokyo, Japan
| | | | - Masaaki Kodama
- Department of Gastroenterology Faculty of Medicine, Oita University, Oita, Japan
| | - Kazunari Murakami
- Department of Gastroenterology Faculty of Medicine, Oita University, Oita, Japan
| | - Kiichi Sugimoto
- Department of Coloproctological Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazuhiro Sakamoto
- Department of Coloproctological Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Toshikazu Ushijima
- Department of Epigenomics, Institute for Advanced Life Sciences, Hoshi University, Tokyo, Japan.
| |
Collapse
|
15
|
Wang L, Yang Y, Yang A, Li T. Lightweight deep learning model incorporating an attention mechanism and feature fusion for automatic classification of gastric lesions in gastroscopic images. BIOMEDICAL OPTICS EXPRESS 2023; 14:4677-4695. [PMID: 37791283 PMCID: PMC10545198 DOI: 10.1364/boe.487456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/11/2023] [Accepted: 06/29/2023] [Indexed: 10/05/2023]
Abstract
Accurate diagnosis of various lesions in the formation stage of gastric cancer is an important problem for doctors. Automatic diagnosis tools based on deep learning can help doctors improve the accuracy of gastric lesion diagnosis. Most of the existing deep learning-based methods have been used to detect a limited number of lesions in the formation stage of gastric cancer, and the classification accuracy needs to be improved. To this end, this study proposed an attention mechanism feature fusion deep learning model with only 14 million (M) parameters. Based on that model, the automatic classification of a wide range of lesions covering the stage of gastric cancer formation was investigated, including non-neoplasm(including gastritis and intestinal metaplasia), low-grade intraepithelial neoplasia, and early gastric cancer (including high-grade intraepithelial neoplasia and early gastric cancer). 4455 magnification endoscopy with narrow-band imaging(ME-NBI) images from 1188 patients were collected to train and test the proposed method. The results of the test dataset showed that compared with the advanced gastric lesions classification method with the best performance (overall accuracy = 94.3%, parameters = 23.9 M), the proposed method achieved both higher overall accuracy and a relatively lightweight model (overall accuracy =95.6%, parameter = 14 M). The accuracy, sensitivity, and specificity of low-grade intraepithelial neoplasia were 94.5%, 93.0%, and 96.5%, respectively, achieving state-of-the-art classification performance. In conclusion, our method has demonstrated its potential in diagnosing various lesions at the stage of gastric cancer formation.
Collapse
Affiliation(s)
- Lingxiao Wang
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Yingyun Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Aiming Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Ting Li
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
16
|
Tjandra D, Busuttil RA, Boussioutas A. Gastric Intestinal Metaplasia: Challenges and the Opportunity for Precision Prevention. Cancers (Basel) 2023; 15:3913. [PMID: 37568729 PMCID: PMC10417197 DOI: 10.3390/cancers15153913] [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: 06/28/2023] [Revised: 07/20/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
GIM is a persistent, premalignant lesion whereby gastric mucosa is replaced by metaplastic mucosa resembling intestinal tissue, arising in the setting of chronic inflammation, particularly in the context of Helicobacter pylori. While the overall rates of progression to gastric adenocarcinoma are low, estimated at from 0.25 to 2.5%, there are features that confer a much higher risk and warrant follow-up. In this review, we collate and summarise the current knowledge regarding the pathogenesis of GIM, and the clinical, endoscopic and histologic risk factors for cancer. We examine the current state-of-practice with regard to the diagnosis and management of GIM, which varies widely in the published guidelines and in practice. We consider the emerging evidence in population studies, artificial intelligence and molecular markers, which will guide future models of care. The ultimate goal is to increase the detection of early gastric dysplasia/neoplasia that can be cured while avoiding unnecessary surveillance in very low-risk individuals.
Collapse
Affiliation(s)
- Douglas Tjandra
- Central Clinical School, Monash University, 99 Commercial Rd, Melbourne, VIC 3004, Australia;
- Department of Gastroenterology, The Alfred Hospital, 55 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Rita A. Busuttil
- Central Clinical School, Monash University, 99 Commercial Rd, Melbourne, VIC 3004, Australia;
- Department of Gastroenterology, The Alfred Hospital, 55 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Alex Boussioutas
- Central Clinical School, Monash University, 99 Commercial Rd, Melbourne, VIC 3004, Australia;
- Department of Gastroenterology, The Alfred Hospital, 55 Commercial Rd, Melbourne, VIC 3004, Australia
| |
Collapse
|
17
|
Sotelo S, Manterola C, Otzen T, Morales E, Castillo I. Prevalence of Gastric Preneoplastic Lesions in First-Degree Relatives of Patients with Gastric Cancer: a Cross-Sectional Study. J Gastrointest Cancer 2023; 54:513-519. [PMID: 35488975 DOI: 10.1007/s12029-022-00827-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2022] [Indexed: 11/11/2022]
Abstract
PURPOSE Worldwide, gastric cancer (GC) is the 5th cancer with the highest incidence and the 4th in mortality. To reduce it, one strategy is to diagnose preneoplastic lesions (PNL): atrophic gastritis (AG), intestinal metaplasia (IM), and dysplasia (DYS); to form risk groups on which to focus surveillance efforts as are first-degree relatives (FDR). The aim of this study was to determine prevalence of gastric PNL in FDR of patients with GC, and to study association with sex, age, and Helicobacter pylorii (Hp) infection. METHODS Cross-sectional study. One hundred and ten FDR, aged between 50 and 65 years, 54.5 female, obtained through convenience sampling, were studied. Biodemographic data survey and upper gastrointestinal endoscopy with histological study were applied according to Sidney protocol, and focal lesions found. Diagnosis of these lesions and condition of mucosa was carried out by applying OLGA and OLGIM systems. Descriptive statistics, estimation of prevalence, odds ratio (OR), and 95% confidence intervals (95CI) were calculated. RESULTS Median age of study group was 56.5 years. Prevalence of PNL, AG, IM, and DYS were 86.4%, 82.7%, 54.5%, and 12.7% respectively. Advanced stages of OLGA and OLGIM were verified in 18.0% and 16.3% respectively. No association with sex, age, and Hp infection were found ([OR 3.10; 95CI 1.0; 9.64]; [OR 0.74; 95CI 0.26; 2.14]; [OR 0.58; 95CI 0.12; 2.77]) respectively. CONCLUSION FDR of patients with GC have a high prevalence of PNL, which makes them a risk group in which endoscopic surveillance should be applied.
Collapse
Affiliation(s)
- Sergio Sotelo
- PhD Program in Medical Sciences, Universidad de La Frontera, Temuco, Chile
- Faculty of Medicine, Universidad Católica del Maule, Talca, Chile
- Hospital Regional de Talca, Talca, Chile
| | - Carlos Manterola
- Center of Morphological and Surgical Studies (CEMyQ), and PhD Program in Medical Sciences, and Millennium Nucleus on Sociomedicine, Universidad de La Frontera, Manuel Montt 115, 54-D, Temuco, Chile.
| | - Tamara Otzen
- Center of Morphological and Surgical Studies (CEMyQ), and PhD Program in Medical Sciences, and Millennium Nucleus on Sociomedicine, Universidad de La Frontera, Manuel Montt 115, 54-D, Temuco, Chile
| | - Erik Morales
- Faculty of Medicine, Universidad Católica del Maule, Talca, Chile
- Hospital Regional de Talca, Talca, Chile
| | - Iván Castillo
- Faculty of Medicine, Universidad Católica del Maule, Talca, Chile
- Hospital Regional de Talca, Talca, Chile
| |
Collapse
|
18
|
Huang RJ, Laszkowska M, In H, Hwang JH, Epplein M. Controlling Gastric Cancer in a World of Heterogeneous Risk. Gastroenterology 2023; 164:736-751. [PMID: 36706842 PMCID: PMC10270664 DOI: 10.1053/j.gastro.2023.01.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 01/29/2023]
Abstract
Gastric cancer (GC) is a leading cause of global mortality but also a cancer whose footprint is highly unequal. This review aims to define global disease epidemiology, critically appraise strategies of prevention and disease attenuation, and assess how these strategies could be applied to improve outcomes from GC in a world of variable risk and disease burden. Strategies of primary prevention focus on improving the detection and eradication of the main environmental risk factor, Helicobacter pylori. In certain countries of high incidence, endoscopic or radiographic screening of the asymptomatic general population has been adopted as a means of secondary prevention. By contrast, identification and targeted surveillance of individuals with precancerous lesions (such as intestinal metaplasia) is being increasingly embraced in nations of low incidence. This review also highlights existing knowledge gaps in GC prevention as well as the role of emerging technologies for early detection and risk stratification.
Collapse
Affiliation(s)
- Robert J Huang
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California.
| | - Monika Laszkowska
- Gastroenterology, Hepatology, and Nutrition Service, Department of Subspecialty Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Haejin In
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Joo Ha Hwang
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Meira Epplein
- Duke University, Department of Population Health Sciences, and Cancer Risk, Detection, and Interception Program, Duke Cancer Institute, Durham, North Carolina
| |
Collapse
|
19
|
Thrift AP, Wenker TN, El-Serag HB. Global burden of gastric cancer: epidemiological trends, risk factors, screening and prevention. Nat Rev Clin Oncol 2023; 20:338-349. [PMID: 36959359 DOI: 10.1038/s41571-023-00747-0] [Citation(s) in RCA: 153] [Impact Index Per Article: 153.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 03/25/2023]
Abstract
Gastric cancer remains a major cause of cancer-related mortality worldwide. The temporal trends for this malignancy, however, are dynamic, and reports from the past decade indicate important declines in some regions and demographic groups, as well as a few notable exceptions in which gastric cancer rates are either stable or increasing. Two main anatomical subtypes of gastric cancer exist, non-cardia and cardia, with different temporal trends and risk factors (such as obesity and reflux for cardia gastric cancer and Helicobacter pylori infection for non-cardia gastric cancer). Shifts in the distribution of anatomical locations have been detected in several high-incidence regions. H. pylori is an important aetiological factor for gastric cancer; importantly, the anticipated long-term findings from studies examining the effect of H. pylori eradication on the risk of (re)developing gastric cancer have emerged in the past few years. In this Review, we highlight the latest trends in incidence and mortality using an evidence-based approach. We make the best possible inferences, including clinical and public health inference, on the basis of the quality of the evidence available, and highlight burning questions as well as gaps in knowledge and public health practice that need to be addressed to reduce gastric cancer burden worldwide.
Collapse
Affiliation(s)
- Aaron P Thrift
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Theresa Nguyen Wenker
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Center for Innovations in Quality, Effectiveness and Safety (IQuESt), Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Hashem B El-Serag
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
- Center for Innovations in Quality, Effectiveness and Safety (IQuESt), Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.
| |
Collapse
|
20
|
Endoscopic resection of gastric low-grade dysplasia with high risk factors is associated with decreased advanced neoplasia: a single-center retrospective cohort study. Surg Endosc 2023:10.1007/s00464-023-09968-x. [PMID: 36890418 DOI: 10.1007/s00464-023-09968-x] [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: 11/03/2022] [Accepted: 02/17/2023] [Indexed: 03/10/2023]
Abstract
BACKGROUND The natural course of gastric low-grade dysplasia (LGD) remains unclear, and there are inconsistent management recommendations among guidelines and consensus. OBJECTIVE This study aimed to investigate the incidence of advanced neoplasia in patients with gastric LGD and identify the related risk factors. METHODS Cases of biopsy demonstrated LGD (BD-LGD) at our center from 2010 to 2021 were reviewed retrospectively. Risk factors related to histological progression were identified, and outcomes of patients based on risk stratification were evaluated. RESULTS Ninety-seven (23.0%) of 421 included BD-LGD lesions were diagnosed as advanced neoplasia. Among 409 superficial BD-LGD lesions, lesion in the upper third of the stomach, H. pylori infection, larger size, and narrow band imaging (NBI)-positive findings were independent risk factors of progression. NBI-positive lesions and NBI-negative lesions with or without other risk factors had 44.7%, 1.7%, and 0.0% risk of advanced neoplasia, respectively. Invisible lesions, visible lesions (VLs) without a clear margin, and VLs with a clear margin and size ≤ 10 mm, or > 10 mm had 4.8%, 7.9%, 16.7%, and 55.7% risk of advanced neoplasia, respectively. In addition, endoscopic resection decreased the risk of cancer (P < 0.001) and advanced neoplasia (P < 0.001) in patients with NBI-positive lesions, but not in NBI-negative patients. Similar results were found in patients with VLs with clear margin and size > 10 mm. Moreover, NBI-positive lesions had higher sensitivity and lower specificity for predicting advanced neoplasia than VLs with a clear margin and size > 10 mm determined by white-light endoscopy (97.6% vs. 62.7%, P < 0.001; and 63.0% vs. 85.6%, P < 0.001, respectively). CONCLUSION Progression of superficial BD-LGD is associated with NBI-positive lesions, as well as with VLs with a clear margin (size > 10 mm) if NBI is unavailable, and selective resection of those lesions offers benefits for patients by decreasing the risk of advanced neoplasia.
Collapse
|
21
|
Sakin A, Sahin S, Sakin A, Aldemir MN, Sakoglu N, Bayram I, Kotan MC. Factors affecting survival in operated gastric cancer. Surg Oncol 2023; 46:101887. [PMID: 36455334 DOI: 10.1016/j.suronc.2022.101887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022]
Abstract
In this study, our aim was to determine the possible effects of Helicobacter pylori(HP), chronic atrophic gastritis (CAG), and gastrointestinal metaplasia (GIM) on survival in operated bowel type gastric cancer patients (INT-GC). Among 548 patients, 347(63.3%) were male. The median age was 57 years. Disease-free survival (DFS) and overall survival (OS) were significantly shorter in patients with GIM than those in patients without GIM (log rank, P = 0.003 and log rank P = 0.003, respectively). Multivariate analysis showed that presence of GIM (HR, 2.1) was found to be an independent factor of worse DFS. In our study, stage pIII patients with GIM had significantly shorter DFS and OS than those without GIM (log rank p = 0.008 and log rank p = 0.001, respectively). However, in subgroup analysis of patients with GIM, there was no significant DFS and OS difference between patients with stage pI and pII disease (log rank p = 0.999, log rank p = 0.184 vs. log rank p = 0.409, log rank p = 0.281, respectively).
Collapse
Affiliation(s)
- Aysegul Sakin
- Department of Internal Medicine, University of Health Sciences, Taksim Research and Training Hospital, Istanbul, Turkey.
| | - Suleyman Sahin
- Department of Medical Oncology, University of Health Sciences, Van Research and Training Hospital, Van, Turkey.
| | - Abdullah Sakin
- Department of Medical Oncology, Yuzuncu Yil University Medical School, 65030, Van, Turkey; İstanbul Medipol University, Medical Oncology Department, Istanbul, Turkey.
| | - Mehmet Naci Aldemir
- Department of Medical Oncology, Yuzuncu Yil University Medical School, 65030, Van, Turkey.
| | - Nevin Sakoglu
- İstanbul Medipol University, General Surgery Department, Istanbul, Turkey.
| | - Irfan Bayram
- Department of Pathology, Yuzuncu Yil University Medical School, 65030, Van, Turkey.
| | - Mehmet Cetin Kotan
- Department of General Surgery, Yuzuncu Yil University Medical School, 65030, Van, Turkey.
| |
Collapse
|
22
|
Liao F, Zhu Z, Zhu S, Wan J, Fan C, Zhang X, Shu X, Lu N. Investigation of the prevalence and risk factors of Helicobacter pylori infection and the value of different gastric cancer screening methods in a low-risk region of gastric cancer in China. Ann Med 2023; 55:2243988. [PMID: 37669097 PMCID: PMC10481759 DOI: 10.1080/07853890.2023.2243988] [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: 06/18/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND The aim of this current study was to identify the prevalence and risk factors of H. pylori infection in the low-risk area of gastric cancer in China, and evaluate the value of different gastric cancer screening methods. METHODS An epidemiological study was conducted in Yudu County, Jiangxi, China, and participants were followed up for 6 years. All participants completed a questionnaire, laboratory tests and endoscopy. Patients were divided into H. pylori positive and negative groups, and risk factors for H. pylori infection were identified using multivariate logistic regression analysis. RESULTS A total of 1962 residents were included, the prevalence of H. pylori infection was 33.8%. Multivariate analysis showed that annual income ≤20,000 yuan (OR: 1.44, 95% CI: 1.18-1.77, p < 0.001), loss of appetite (OR: 1.71, 95% CI: 1.29-2.26, p < 0.001), PG II >37.23 ng/mL (OR: 2.11, 95% CI: 1.50-2.97, p < 0.001), G-17 > 1.5 and ≤5.7 pmol/L (OR: 2.52, 95% CI: 1.93-3.30, p < 0.001), and G-17 > 5.7 pmol/L (OR: 1.96, 95% CI: 1.48-2.60, p < 0.001) were risk factors of H. pylori infection, while alcohol consumption (OR: 0.70, 95% CI: 0.54-0.91, p = 0.006) was a protective factor. According to the new gastric cancer screening method, the prevalence of low-grade intraepithelial neoplasia in the low-risk group, medium-risk group and high-risk group was 4.4%, 7.7% and 12.5% respectively (p < 0.001). CONCLUSIONS In a low-risk area of gastric cancer in China, the infection rate of H. pylori is relatively low. Low income, loss of appetite, high PG II, and high G-17 were risk factors for H. pylori infection, while alcohol consumption was a protective factor. Moreover, the new gastric cancer screening method better predicted low-grade intraepithelial neoplasia than the ABC method and the new ABC method.
Collapse
Affiliation(s)
- Foqiang Liao
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Zhenhua Zhu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Jiangxi Clinical Research Center for Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Shusheng Zhu
- Department of Gastroenterology, Yudu County People’s Hospital, Ganzhou, Jiangxi, P.R.China
| | - Jianhua Wan
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Jiangxi Clinical Research Center for Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Chenglai Fan
- Department of Gastroenterology, Yudu County People’s Hospital, Ganzhou, Jiangxi, P.R.China
| | - Xu Zhang
- Department of Gastroenterology, Yudu County People’s Hospital, Ganzhou, Jiangxi, P.R.China
| | - Xu Shu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Jiangxi Clinical Research Center for Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Nonghua Lu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Jiangxi Clinical Research Center for Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| |
Collapse
|
23
|
Dong EY, Giap AQ, Lustigova E, Wu BU. Gastric Cancer Screening in First-Degree Relatives: A Pilot Study in a Diverse Integrated Healthcare System. Clin Transl Gastroenterol 2022; 13:e00531. [PMID: 36113027 PMCID: PMC10476735 DOI: 10.14309/ctg.0000000000000531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/19/2022] [Accepted: 08/30/2022] [Indexed: 01/31/2023] Open
Abstract
INTRODUCTION Family history of gastric cancer has been shown as an independent risk factor of gastric cancer development and is associated with increased risk of progression to gastric cancer among patients with gastric intestinal metaplasia (GIM). METHODS Between 2017 and 2020, we conducted a prospective pilot screening program of patients with a confirmed first-degree relative with gastric cancer to evaluate the feasibility of screening and prevalence of precursor lesions (e.g., GIM or dysplasia) on biopsy. RESULTS A total of 61 patients completed screening by upper endoscopy with a mapping biopsy protocol: 27 (44%) were found to have GIM and 4 (7%) were found with low-grade dysplasia. DISCUSSION Our pilot screening program identified a high prevalence of precursor lesions for gastric cancer among asymptomatic patients with a first-degree relative with gastric cancer. Careful endoscopic inspection and standardized biopsy protocols may aid in prompt identification of these precursor lesions in those at risk of gastric cancer.
Collapse
Affiliation(s)
- Elizabeth Y. Dong
- Division of Gastroenterology, Southern California Permanente Medical Group, Los Angeles, California, USA
| | - Andrew Q. Giap
- Division of Gastroenterology, Southern California Permanente Medical Group, Anaheim, California, USA
| | - Eva Lustigova
- Department of Research & Evaluation, Southern California Permanente Medical Group, Pasadena, California, USA
| | - Bechien U. Wu
- Division of Gastroenterology, Southern California Permanente Medical Group, Los Angeles, California, USA
- Department of Research & Evaluation, Southern California Permanente Medical Group, Pasadena, California, USA
- Division of Gastroenterology, Center for Pancreatic Care, Southern California Medical Group, Los Angeles, California, USA
| |
Collapse
|
24
|
Huang RJ, Epplein M, Hamashima C, Choi IJ, Lee E, Deapen D, Woo Y, Tran T, Shah SC, Inadomi JM, Greenwald DA, Hwang JH. An Approach to the Primary and Secondary Prevention of Gastric Cancer in the United States. Clin Gastroenterol Hepatol 2022; 20:2218-2228.e2. [PMID: 34624563 PMCID: PMC8983795 DOI: 10.1016/j.cgh.2021.09.039] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/02/2021] [Accepted: 09/10/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Gastric cancer (GC) remains a leading cause of mortality among certain racial, ethnic, and immigrant groups in the United States (US). The majority of GCs are diagnosed at advanced stages, and overall survival remains poor. There exist no structured national strategies for GC prevention in the US. METHODS On March 5-6, 2020 a summit of researchers, policy makers, public funders, and advocacy leaders was convened at Stanford University to address this critical healthcare disparity. After this summit, a writing group was formed to critically evaluate the effectiveness, potential benefits, and potential harms of methods of primary and secondary prevention through structured literature review. This article represents a consensus statement prepared by the writing group. RESULTS The burden of GC is highly inequitably distributed in the US and disproportionately falls on Asian, African American, Hispanic, and American Indian/Alaskan Native populations. In randomized controlled trials, strategies of Helicobacter pylori testing and treatment have been demonstrated to reduce GC-specific mortality. In well-conducted observational and ecologic studies, strategies of endoscopic screening have been associated with reduced GC-specific mortality. Notably however, all randomized controlled trial data (for primary prevention) and the majority of observational data (for secondary prevention) are derived from non-US sources. CONCLUSIONS There exist substantial, high-quality data supporting GC prevention derived from international studies. There is an urgent need for cancer prevention trials focused on high-risk immigrant and minority populations in the US. The authors offer recommendations on how strategies of primary and secondary prevention can be applied to the heterogeneous US population.
Collapse
Affiliation(s)
- Robert J Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Stanford, California
| | - Meira Epplein
- Department of Population Health Sciences, Duke University, and Cancer Risk, Detection, and Interception Program, Duke Cancer Institute, Durham, North Carolina
| | | | - Il Ju Choi
- Center for Gastric Cancer, National Cancer Center, Goyang, South Korea
| | - Eunjung Lee
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Dennis Deapen
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Yanghee Woo
- Division of Surgical Oncology, Department of Surgery, City of Hope National Comprehensive Cancer Center, Duarte, California
| | - Thuy Tran
- Division of Surgical Oncology, Department of Surgery, City of Hope National Comprehensive Cancer Center, Duarte, California
| | - Shailja C Shah
- Gastroenterology Section, Veterans Affairs San Diego Healthcare System, La Jolla, California; Division of Gastroenterology and Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - John M Inadomi
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - David A Greenwald
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Joo Ha Hwang
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Stanford, California.
| |
Collapse
|
25
|
Parbhu SK, Shah SC, Sossenheimer MJ, Fang JC, Peterson KA, Gawron AJ. Index diagnoses of gastric intestinal metaplasia in the United States: patient characteristics, endoscopic findings, and clinical practice patterns at a large tertiary care center. Therap Adv Gastroenterol 2022; 15:17562848221117640. [PMID: 36082176 PMCID: PMC9445457 DOI: 10.1177/17562848221117640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 07/18/2022] [Indexed: 02/04/2023] Open
Abstract
Background Gastric intestinal metaplasia (GIM) is a premalignant gastric mucosal change that is often incidentally detected during esophagogastroduodenoscopy (EGD). Despite the established higher risk of gastric cancer associated with GIM, the incidence, prevalence, and outcomes data for GIM are limited in the United States (US), and practice patterns are highly variable. Objectives Our primary objectives were to accurately identify incident histology-confirmed GIM cases and determine patient characteristics, endoscopy findings, Helicobacter pylori (HP) detection, and eradication treatment outcomes, as well as surveillance and follow-up recommendations. Design We conducted a retrospective cohort study using administrative data. Methods We first developed and validated a rule-based natural language processing tool to identify the patients with GIM on gastrointestinal pathology reports between 2011 and 2016. We then performed a manual chart review of all EGD procedures and associated pathology notes to confirm cases and obtain clinically relevant data. Results In all, 414 patients with an index diagnosis of GIM were confirmed (prevalence = 2.5% of patients undergoing any EGD). A majority (52.4%) of patients were non-Hispanic white. The most common indication for EGD was abdominal pain (46.9%). A majority (55%) did not receive specific follow-up recommendations or were asked to see their primary care provider. HP testing was documented in 86% of patients, and detected in 94 patients (prevalence = 26.4%). Treatment was documented in 94.7% of cases, and eradication confirmed in only 34.8% of these cases. Conclusion A large group of US patients with an index diagnosis of GIM was accurately identified. There was wide variability in clinical practice patterns including biopsy practice, HP treatment and eradication confirmation testing, and surveillance recommendations. This work demonstrates that there is a major unmet need for quality improvement efforts to standardize care for patients with GIM, a premalignant condition, and inform future prospective studies in a US population.
Collapse
Affiliation(s)
- Sheeva K. Parbhu
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Shailja C. Shah
- Division of Gastroenterology, University of California San Diego, San Diego, CA, USA Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Michael J. Sossenheimer
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - John C. Fang
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Kathryn A. Peterson
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | | |
Collapse
|
26
|
Li G, Kelly DR, Mroczek-Musulman E, Wang K, Council L, Zhao L. Gastric Antral Mucosal Changes in Children With Intestinal Metaplasia. Pediatr Dev Pathol 2022; 25:511-517. [PMID: 35510382 DOI: 10.1177/10935266221096939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Objectives: The gastric mucosal change accompanying gastric antral intestinal metaplasia (IM) in the pediatric population and its clinical implications remain unclear. Methods: We retrieved all patients younger than 18 years who had upper GI endoscopy with a pathology diagnosis of antral IM between 2009 and 2020. Each biopsy was evaluated for the presence of dysplasia, Helicobacter pylori, gastritis, and other pathologic changes. Results: A total of 134 patients with antral IM were identified; 72 (53.7%) with coexisting pathology including chronic gastritis (n = 22), reactive gastropathy (n = 16), focal mild chronic inflammation (n = 13), gastric eosinophilia (n = 9), chronic active gastritis associated with (n = 2) and without Helicobacter infection (n = 3), and others (n = 7). The remaining 62 (46.3%) showed isolated IM. Gastric IM increased with age, and was often accompanied by other pathologic changes, especially in female children. Twenty-seven patients had follow up biopsies; 11 of the 27 patients (40.7%) showed persistent IM in at least one repeat biopsies. None demonstrated dysplasia. Conclusions: In children, antral IM increases with age and often coexists with other pathologic changes. Gastric IM could persist for at least months to years in a significant subset of patients with chronic gastritis and gastric eosinophilia.
Collapse
Affiliation(s)
- Geling Li
- Department of Pathology and Laboratory Medicine, 22078Children's of Alabama, Birmingham, AL, USA.,Department of Pathology, 9968University of Alabama at Birmingham, Birmingham, AL, USA
| | - David R Kelly
- Department of Pathology and Laboratory Medicine, 22078Children's of Alabama, Birmingham, AL, USA.,Department of Pathology, 9968University of Alabama at Birmingham, Birmingham, AL, USA
| | - Elizabeth Mroczek-Musulman
- Department of Pathology and Laboratory Medicine, 22078Children's of Alabama, Birmingham, AL, USA.,Department of Pathology, 9968University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kai Wang
- Department of Pathology, 9968University of Alabama at Birmingham, Birmingham, AL, USA
| | - Leona Council
- Department of Pathology, 9968University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lei Zhao
- Department of Pathology, Harvard Medical School, 1861Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
27
|
Kligman E, Ali H, Chen E, Peng F, Szafron D, Staggers K, Tan MC, Patel K, Othman MO. Ethnicity Is an Important Consideration in Screening for Gastric Intestinal Metaplasia. Dig Dis Sci 2022; 67:4509-4517. [PMID: 34981309 DOI: 10.1007/s10620-021-07326-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 11/03/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Gastric intestinal metaplasia (GIM) is a precursor to gastric adenocarcinoma, making it an attractive target for early detection by endoscopy. The aim of this study was to determine the prevalence, risk factors, and associated histologic findings of GIM among patients undergoing endoscopy in a diverse US population. METHODS We conducted a retrospective, cross-sectional study of patients undergoing elective endoscopy with gastric biopsies at 6 academic and community centers in Houston, Texas. GIM prevalence was estimated with a 95% confidence interval (CI), and patient demographic and clinical characteristics were summarized using mean with standard deviation, or frequency with percentage. Generalized estimating equations (GEE) were used to compare characteristics between those with and without GIM. RESULTS Our final cohort consisted of 2685 patients, including 216 cases with GIM and 2469 controls. The prevalence of GIM in our cohort was 8.04% (95% CI 7.07%, 9.14%). The mean age of GIM cases was higher than in the control group (59.8 vs 54.7 years, p < 0.0001). The prevalence of GIM in Asians, Hispanic, Black and Non-Hispanic Whites (NHW) was 14.7%, 11.7%, 9.8% and 5.8%, respectively. On multivariable analysis, factors associated with GIM include age (adj. OR 1.32 per 10 year increase, p < 0.0001), habitual smoking (adj. OR 1.68, p < 0.0001), and race (compared to NHW: Asian, adj. OR 2.34, p = 0.010; Hispanic, adj. OR 2.15, p < 0.001; Black, adj. OR 1.61, p < 0.001). CONCLUSION Asians, Hispanics, and African Americans have higher rates of GIM than NHW. Ethnicity should be an important consideration on determining who to screen for GIM in the US.
Collapse
Affiliation(s)
- Eugene Kligman
- Section of Gastroenterology and Hepatology, Baylor College of Medicine, 7200 Cambridge Street. Suite 8A, Houston, TX, 77030, USA
| | - Hiba Ali
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Ellie Chen
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Frederick Peng
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - David Szafron
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Kristen Staggers
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Mimi C Tan
- Section of Gastroenterology and Hepatology, Baylor College of Medicine, 7200 Cambridge Street. Suite 8A, Houston, TX, 77030, USA
| | - Kalpesh Patel
- Section of Gastroenterology and Hepatology, Baylor College of Medicine, 7200 Cambridge Street. Suite 8A, Houston, TX, 77030, USA
| | - Mohamed O Othman
- Section of Gastroenterology and Hepatology, Baylor College of Medicine, 7200 Cambridge Street. Suite 8A, Houston, TX, 77030, USA. .,Baylor St Luke's Medical Center, Houston, TX, USA.
| |
Collapse
|
28
|
Laszkowska M, Truong H, Faye AS, Kim J, Tan SX, Lim F, Abrams JA, Hur C. Prevalence of Extensive and Limited Gastric Intestinal Metaplasia and Progression to Dysplasia and Gastric Cancer. Dig Dis Sci 2022; 67:3693-3701. [PMID: 34657192 PMCID: PMC9013391 DOI: 10.1007/s10620-021-07276-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 10/04/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND AIMS Guidelines cite extensive gastric intestinal metaplasia (GIM) as a bigger risk factor for gastric cancer (GC) than limited GIM and an indication for endoscopic surveillance. Data on progression of extensive GIM to GC in the USA are limited. This study aimed to estimate the prevalence and progression rates of extensive GIM in a US cohort. METHODS This retrospective study assessed the prevalence of extensive GIM between 1/1/1990 and 8/1/2019 at a large academic medical center. Multivariable regression was used to identify predictors of extensive GIM. Incidence of GC on follow-up was calculated as number of new diagnoses divided by person-years of follow-up. Presence of GIM on subsequent follow-up endoscopy was assessed. RESULTS Of 1256 individuals with GIM, 352 (28%) had extensive GIM and 904 (72%) had limited GIM. On multivariable analysis, older age (OR 1.01, 95% CI 1.00-1.02) and Hispanic ethnicity (OR 1.55, 95% CI 1.11-2.16) were predictive of extensive GIM. The annual incidence of GC for GIM overall was 0.09%. There was no difference in progression to GC between extensive or limited GIM (IRR 0, 95% CI 0-2.6), or to advanced lesions overall (IRR 0.37, 95% CI 0.04-1.62). 70% of individuals had persistent GIM on follow-up biopsy, and 22% with limited GIM had extensive GIM on follow-up biopsy. CONCLUSIONS 28% of individuals with GIM have the extensive subtype, and are more likely to be older and of Hispanic ethnicity. There was no difference in progression to GC between extensive and limited GIM. Further research is needed to better assess risk of GIM in the US context.
Collapse
Affiliation(s)
- Monika Laszkowska
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA,Department of Subspecialty Medicine, Gastroenterology, Hepatology, and Nutrition Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | - Han Truong
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Adam S. Faye
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA,Division of Gastroenterology, New York University Langone Health, New York, NY, USA
| | - Judith Kim
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Sarah Xinhui Tan
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Francesca Lim
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Julian A. Abrams
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA,Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Chin Hur
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA,Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA,Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
29
|
Pei B, Wen Z, Yang Q, Wang J, Cao Q, Dai L, Li X. Risk Factors Analysis and Prediction Model Establishment of Intestinal Metaplasia or Dysplasia in Patients With Chronic Atrophic Gastritis: A Multi-Center Retrospective Study. Front Med (Lausanne) 2022; 9:912331. [PMID: 35665336 PMCID: PMC9157492 DOI: 10.3389/fmed.2022.912331] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Objective To investigate the risk factors and construct a prediction model of chronic atrophic gastritis (CAG) patients with intestinal metaplasia or dysplasia. Method The clinical data of 450 patients with CAG who were diagnosed and treated in the Department of Gastroenterology of the Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine from June 2016 to February 2022 were collected. Single and multiple factors logistic regression analysis were used to explore the risk factors of intestinal metaplasia or dysplasia in patients of training cohort. Then, we constructed a model to predict the onset of intestinal metaplasia or dysplasia based on the data of training cohort, following which we tested the model in an external validation cohort of 193 patients from a local university teaching hospital. The ROC curve, calibration curve, and decision curve analysis were used to evaluate the accuracy of the prediction model. Result Helicobacter pylori (H. pylori, HP) infection, pepsinogen I, gastrin-17, and the number of lesions were found to be independent rick factors of the model. The liner prediction model showed excellent predictive value in both training cohort and validation cohort. Conclusion HP infection, pepsinogen I, gastrin-17, and the number of lesions are independent risk factors for intestinal metaplasia or dysplasia in patients with CAG. The prediction model constructed based on these factors has a high accuracy and excellent calibration, which can provide a great basis for condition assessment and individualized treatment of the patients.
Collapse
Affiliation(s)
- Bei Pei
- The Graduated School, Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Ziang Wen
- The Graduated School, Anhui Medical University, Hefei, China
| | - Qi Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Jieyu Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Qinglin Cao
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Longfei Dai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xuejun Li
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| |
Collapse
|
30
|
Xiao S, Lu H, Xue Y, Cui R, Meng L, Jin Z, Yin Z, Zhou L. Long-Term Outcome of Gastric Mild-Moderate Dysplasia: A Real-World Clinical Experience. Clin Gastroenterol Hepatol 2022; 20:1259-1268.e7. [PMID: 34718170 DOI: 10.1016/j.cgh.2021.10.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS The natural course of gastric mild-moderate dysplasia in a country with high incidence of gastric cancer (GC) is relatively unknown. We aimed to determine the long-term cumulative incidence of and risk factors for advanced neoplasia in patients with gastric dysplasia. METHODS This was a single-center observational study including all consecutive patients diagnosed with gastric mild-moderate dysplasia between 2000 and 2017. Follow-up data were collected until December 2019. We determined the cumulative incidence of advanced neoplasia and identified risk factors with Cox regression. RESULTS A total of 3489 consecutive participants were followed for a median of 4.19 years from initial mild-moderate dysplasia diagnosis. The median surveillance interval between index endoscopy and next follow-up endoscopy was 1.08 years, and more than half of patients had at least 3 surveillance gastroscopies. During the study period, the majority of participants did not show disease progression, either with dysplasia not detected (51.4%) or with persistent dysplasia (46.1%). There were 88 (2.9%) patients (5.13 per 1000 patient-years) who progressed to advanced neoplasia within a median of 4.3 years. The annual incidence of advanced neoplasia and GC were 0.43% and 0.26%, respectively, within 5 years of mild-moderate dysplasia diagnosis. Increasing age, male sex, moderate dysplasia, dysplasia detected in fundus or cardia at index endoscopy, and persistent Helicobacter pylori infection during follow-up were independent risk factors for developing advanced neoplasia. CONCLUSIONS Even in a country with high incidence of GC, the majority of patients with gastric mild-moderate dysplasia did not experience disease progression in the long term. Intensified surveillance during the first 5 years after mild-moderate dysplasia detection is suggested.
Collapse
Affiliation(s)
- Shiyu Xiao
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing China
| | - Haoping Lu
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing China
| | - Yan Xue
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing China
| | - Rongli Cui
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing China
| | - Lingmei Meng
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing China
| | - Zhu Jin
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing China
| | - Zhihao Yin
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing China
| | - Liya Zhou
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing China.
| |
Collapse
|
31
|
Shah S, Hubscher E, Pelletier C, Jacob R, Vinals L, Yadlapati R. Helicobacter pylori infection treatment in the United States: clinical consequences and costs of eradication treatment failure. Expert Rev Gastroenterol Hepatol 2022; 16:341-357. [PMID: 35315732 DOI: 10.1080/17474124.2022.2056015] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Helicobacter pylori (Hp) is causal in benign and malignant gastrointestinal diseases. Accordingly, current guidelines recommend Hp eradication in patients with active infection. Unfortunately, treatment failure is common, exposing patients to complications associated with persistent Hp infection and consequences of repeated treatment, including promotion of antibiotic resistance. In the United States (US), data regarding eradication rates with available therapies are limited. Moreover, the clinical and economic burden of eradication treatment failure have not been thoroughly described. AREAS COVERED We aimed to characterize Hp eradication rates and the clinical consequences and associated costs of persistent Hp infection among US adults. We conducted focused literature reviews using initial searches in Embase, MEDLINE, and Cochrane Database of Systematic Reviews via Ovid followed by manual searches to identify relevant publications. EXPERT OPINION Hp eradication rates were suboptimal, with most studies reporting rates ≤80% with clarithromycin-based triple therapy and bismuth quadruple therapy. There was direct evidence supporting numerous benefits of successful Hp eradication, including decreased risk of recurrent or complicated peptic disease and non-cardia gastric cancer. Cost benefits of eradication were related to mitigation of conditions associated with persistent Hp infection, (e.g. complicated peptic ulcer disease, and gastric cancer) which altogether exceed US$5.3 billion.
Collapse
Affiliation(s)
- Shailja Shah
- Division of Gastroenterology, University of California San Diego, La Jolla, California, USA
| | | | - Corey Pelletier
- HEOR, Phathom Pharmaceuticals, Florham Park, New Jersey, USA
| | - Rinu Jacob
- Medical Affairs, Phathom Pharmaceuticals, Florham Park, New Jersey, USA
| | - Lydia Vinals
- Real-World Advanced Analytics, Cytel, Inc, Waltham, Massachusetts, USA
| | - Rena Yadlapati
- Division of Gastroenterology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
32
|
Rakici H, Uyanik E, Rakici IM, Polat HB, Akdogan RA, Aydin G, Ayvaz MA, Bedir R. Gastric intestinal metaplasia: Long-term follow-up results. Niger J Clin Pract 2022; 25:315-324. [PMID: 35295055 DOI: 10.4103/njcp.njcp_1548_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Baackground Precancerous lesions are the most commonly cited factor in gastric cancer etiology. The sequence of events in intestinal-type gastric carcinogenesis is considered to be chronic gastritis, atrophy, intestinal metaplasia (IM), dysplasia, and carcinoma, respectively. Early diagnosis and treatment of advanced precursor lesions and gastric cancer is possible by identifying and monitoring patients with such premalignant lesions. Aim In our study, we aimed to evaluate the long-term follow-up results of intestinal metaplasia in our hospital and the rate of progression to malignancy by comparing these patients with patients who have undergone gastroscopy without a diagnosis of intestinal metaplasia. Material and Method One hundred and fifty-six followed-up patients out of 700 between the ages of 18 and 85 who were admitted to our hospital between 2009 and 2019, who were diagnosed with IM by pathological examination from biopsy material, and 150 patients who were not diagnosed with IM between 2009 and 2011 were included. The results of the cases were evaluated first retrospectively; then, the patients who were invited for control and underwent endoscopy were evaluated prospectively. IM and control groups were compared in terms of dysplasia and gastric cancer development. In addition, the IM group was compared in terms of 5 and 10 years of follow-up, extensive or local involvement, and complete and incomplete involvement in terms of dysplasia and cancer development. Results The follow-up period of the patients ranged from 1 to 10 years, and the mean follow-up interval was 4.2 ± 2.8 (min: 1; max: 10) years. Age, gender, and pathology results of the patients were examined in terms of IM type, localization of IM, pathology accompanying IM, and presence of Helicobacter pylori (Hp) infection and compared with the control group. While gastric carcinoma was detected in three of 156 patients in the IM group, gastric carcinoma was not detected in the follow-up of 150 patients in the control group. IM was most common in the antrum. Incomplete IM was detected in 89 patients, and complete IM in 69 patients. While two of the three patients with gastric carcinoma were localized to the antrum, one patient had incomplete-type IM and two patients had complete-type IM, and Hp was positive in two patients. While dysplasia was detected in nine of the patients diagnosed with IM, it was detected in two patients in the control group. A statistically significant difference was found between the IM and control groups in terms of dysplasia positivity (p = 0.037). On the other hand, no statistically significant difference was found between the IM and control groups in terms of age-group, gender, follow-up time group, and Hp positivity (p > 0.05). There was no significant difference between those who were followed up for 5 and 10 years in the IM group in terms of dysplasia and cancer development. Conclusion Therefore, it is considered that patients with intestinal metaplasia may be followed up at longer intervals, except for patients with race, ethnicity, incomplete type, extensive involvement, and a family history of gastric cancer.
Collapse
Affiliation(s)
- H Rakici
- Department of Gastroenterology, Recep Tayyip Erdogan University, Rize, Turkey
| | - E Uyanik
- Department of İnternal Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - I M Rakici
- Department of Anesthesiology, Bilim University, Istanbul, Turkey
| | - H B Polat
- Department of İnternal Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - R A Akdogan
- Department of Gastroenterology, Recep Tayyip Erdogan University, Rize, Turkey
| | - G Aydin
- Department of Gastroenterology, Recep Tayyip Erdogan University, Rize, Turkey
| | - M A Ayvaz
- Department of Gastroenterology, Recep Tayyip Erdogan University, Rize, Turkey
| | - R Bedir
- Department of Pathology, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
33
|
Yang H, Yang WJ, Hu B. Gastric epithelial histology and precancerous conditions. World J Gastrointest Oncol 2022; 14:396-412. [PMID: 35317321 PMCID: PMC8919001 DOI: 10.4251/wjgo.v14.i2.396] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/08/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
The most common histological type of gastric cancer (GC) is gastric adenocarcinoma arising from the gastric epithelium. Less common variants include mesenchymal, lymphoproliferative and neuroendocrine neoplasms. The Lauren scheme classifies GC into intestinal type, diffuse type and mixed type. The WHO classification includes papillary, tubular, mucinous, poorly cohesive and mixed GC. Chronic atrophic gastritis (CAG) and intestinal metaplasia are recommended as common precancerous conditions. No definite precancerous condition of diffuse/poorly/undifferentiated type is recommended. Chronic superficial inflammation and hyperplasia of foveolar cells may be the focus. Presently, the management of early GC and precancerous conditions mainly relies on endoscopy including diagnosis, treatment and surveillance. Management of precancerous conditions promotes the early detection and treatment of early GC, and even prevent the occurrence of GC. In the review, precancerous conditions including CAG, metaplasia, foveolar hyperplasia and gastric hyperplastic polyps derived from the gastric epithelium have been concluded, based on the overview of gastric epithelial histological organization and its renewal.
Collapse
Affiliation(s)
- Hang Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Wen-Juan Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Bing Hu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
34
|
Endoscopic characteristics in predicting prognosis of biopsy-diagnosed gastric low-grade intraepithelial neoplasia. Chin Med J (Engl) 2021; 135:26-35. [PMID: 34873080 PMCID: PMC8850827 DOI: 10.1097/cm9.0000000000001637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Endoscopic biopsy can underestimate gastric malignancies as low-grade intraepithelial neoplasia (LGIN). Definitively diagnosed LGIN would progress. This study aimed to evaluate predictive factors to identify malignancies misdiagnosed as LGIN by biopsy and LGIN at high risk of progression. METHODS The clinical records of patients diagnosed with gastric LGIN by endoscopic biopsy who underwent at least two endoscopies during the first year of follow-up between 2007 and 2017 were retrospectively collected. Three endoscopists reviewed photographs of the initial endoscopy, described lesion characteristics, and made endoscopic diagnoses. Logistic regression was used to analyze predictors to identify malignancies underestimated as LGIN. A receiver operating characteristic curve was used to evaluate the diagnostic accuracy of these predictors. Patient clinical outcomes of follow-up >1 year were collected. Kaplan-Meier estimates with log-rank tests and Cox proportional hazards regression were used to analyze predictors of progression. RESULTS Overall, 48 of 182 (26.4%) patients were proven to have malignancies. A single lesion, a large lesion size, and marked intestinal metaplasia (IM) were independent predictors of initially misdiagnosed malignancies. The area under the curve of these predictors was 0.871, with a sensitivity of 68.7% and specificity of 92.5%. Twelve of 98 patients (12.2%) progressed during the 33-month median follow-up period. A whitish appearance, irregular margins, marked IM, and histological diagnosis of LGIN more than twice within the first year were predictors for progression. CONCLUSIONS Lesions diagnosed as LGIN by biopsy with marked IM and other predictors above should be prudently treated for high potential to be malignancies or progress. Endoscopic follow-up with repeated biopsies within the first year is recommended.
Collapse
|
35
|
Hu F, Rao M, Zhang M, Meng Q, Wan M, Zhang X, Ding L, Jiang Y. Long non-coding RNA profiles in plasma exosomes of patients with gastric high-grade intraepithelial neoplasia. Exp Ther Med 2021; 23:1. [PMID: 34815753 PMCID: PMC8593877 DOI: 10.3892/etm.2021.10923] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 09/22/2021] [Indexed: 12/24/2022] Open
Abstract
Long non-coding (lnc) RNAs in circulating exosomes are a new class of promising cancer biomarkers; however, their expression in exosomes derived from gastric high-grade intraepithelial neoplasia (GHGIN) has not been reported. In the present study, differentially expressed (DE) lncRNAs were analyzed in the peripheral blood collected from 5 patients with GHGIN and 5 healthy donors using high-throughput sequencing. Reverse transcription-quantitative PCR analysis was performed on 6 randomly selected DE lncRNAs to validate the reliability of the sequencing results. The potential roles of the DE lncRNAs in GHGIN were investigated using Gene Ontology (GO) and Kyoto Encyclopedia of Gene and Genome (KEGG) pathway enrichment analyses. A total of 25,145 lncRNAs were identified in all the samples and 83 DE lncRNAs were further screened, including 76 upregulated and 7 downregulated DE lncRNAs. GO and KEGG analyses predicted that the DE lncRNAs played notable roles in ‘protein/macromolecule glycosylation’, ‘regulation of protein ubiquitination’, ‘renin-angiotensin system’ and ‘MAPK signaling pathways’. A lncRNA-micro (mi)RNA-mRNA interaction network was constructed and used to perform association analyses. It was found that 83 lncRNAs were abnormally expressed in GHGIN, with some potential functions associated with gastric cancer. Furthermore, the lncRNA-miRNA-mRNA interaction network indicated that 7 DE lncRNAs may play a notable role in the occurrence and development of GHGIN. The results of the present study showed the expression profiles of lncRNAs in human GHGIN, elucidated some of the molecular changes associated with GHGIN and improved the understanding of the molecular mechanisms underlying GHGIN and gastric cancer.
Collapse
Affiliation(s)
- Feng Hu
- Department of Hepatology and Gastroenterology, The Second Part of First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Min Rao
- Department of Hepatology and Gastroenterology, The Second Part of First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Manli Zhang
- Department of Hepatology and Gastroenterology, The Second Part of First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Qingqing Meng
- Department of Hepatology and Gastroenterology, The Second Part of First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Minjie Wan
- Central Laboratory, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Xiuna Zhang
- Department of Hepatology and Gastroenterology, The Second Part of First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Lili Ding
- Intensive Care Unit, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Yanfang Jiang
- Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
36
|
Chu F, Li Y, Meng X, Li Y, Li T, Zhai M, Zheng H, Xin T, Su Z, Lin J, Zhang P, Ding X. Gut Microbial Dysbiosis and Changes in Fecal Metabolic Phenotype in Precancerous Lesions of Gastric Cancer Induced With N-Methyl-N'-Nitro-N-Nitrosoguanidine, Sodium Salicylate, Ranitidine, and Irregular Diet. Front Physiol 2021; 12:733979. [PMID: 34803728 PMCID: PMC8599278 DOI: 10.3389/fphys.2021.733979] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/30/2021] [Indexed: 12/27/2022] Open
Abstract
Background and Aims: Precancerous lesions of gastric cancer (PLGC) are the most important pathological phase with increased risk of gastric cancer (GC) and encompass the key stage in which the occurrence of GC can be prevented. In this study, we found that the gut microbiome changed significantly during the process of malignant transformation from chronic gastritis to GC in N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) multiple factors-induced rat model. Accumulating evidence has shown that alterations in gut microbiota and metabolism are potentially linked to chronic inflammation and cancer of the gastrointestinal tract. However, the correlation of gut microbiota and metabolites, inflammatory factors, and the potential mechanism in the formation of PLGC have not yet been revealed. Methods: In this study, multiple factors including MNNG, sodium salicylate drinking, ranitidine feed, and irregular diet were used to establish a PLGC rat model. The pathological state of the gastric mucosa of rats was identified through HE staining and the main inflammatory cytokine levels in the serum were detected by the Luminex liquid suspension chip (Wayen Biotechnologies, Shanghai, China). The microbial composition and metabolites in the stool samples were tested by using 16S ribosomal RNA (rRNA) gene sequencing and non-targeted metabolomics. The correlation analysis of gut microbiota and inflammatory cytokines in the serum and gut microbiota and differential metabolites in feces was performed to clarify their biological function. Results: The results showed that compared to the control group, the gastric mucosa of the model rats had obvious morphological and pathological malignant changes and the serum levels of inflammatory cytokines including interleukin-1β (IL-1β), interleukin-4 (IL-4), interleukin-6 (IL-6), interleukin-10 (IL-10), interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and macrophage colony-stimulating factor (M-CSF) increased significantly, while the level of chemokine (C-X-C motif) ligand 1 (CXCL1) in serum reduced significantly. There were significant differences in the composition of the gut microbiota and fecal metabolic profiles between the model and control rats. Among them, Lactobacillus and Bifidobacterium increased significantly, while Turicibacter, Romboutsia, Ruminococcaceae_UCG-014, Ruminococcaceae_UCG-005, and Ruminococcus_1 reduced significantly in the model rats compared to the control rats. The metabolites related to the lipid metabolism and peroxisome proliferator-activated receptor (PPAR) signaling pathway have also undergone significant changes. In addition, there was a significant correlation between the changes of the differential inflammatory cytokines in the serum, fecal metabolic phenotypes, and gut microbial dysbiosis in model rats. Conclusion: The activation of the inflammatory response, disturbance of the gut microbiota, and changes in the fecal metabolic phenotype could be closely related to the occurrence of PLGC. This study provides a new idea to reveal the mechanism of risk factors of chronic gastritis and GC from the perspective of inflammation-immune homeostasis, gut microbiota, and metabolic function balance.
Collapse
Affiliation(s)
- Fuhao Chu
- Institute of Regulatory Science for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Research Center for Spleen and Stomach Diseases of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yicong Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiangmei Meng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Research Center for Spleen and Stomach Diseases of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuan Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Research Center for Spleen and Stomach Diseases of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tao Li
- Research Center for Spleen and Stomach Diseases of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mengyin Zhai
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Research Center for Spleen and Stomach Diseases of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Haocheng Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Research Center for Spleen and Stomach Diseases of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tianxi Xin
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Research Center for Spleen and Stomach Diseases of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zeqi Su
- Research Center for Spleen and Stomach Diseases of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Lin
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Research Center for Spleen and Stomach Diseases of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ping Zhang
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xia Ding
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Research Center for Spleen and Stomach Diseases of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
37
|
The relationship between abnormal tongue features and non-malignant upper gastrointestinal disorders: A hospital-based cross-sectional study. Eur J Integr Med 2021. [DOI: 10.1016/j.eujim.2021.101379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
38
|
Molecular Landscapes of Gastric Pre-Neoplastic and Pre-Invasive Lesions. Int J Mol Sci 2021; 22:ijms22189950. [PMID: 34576114 PMCID: PMC8468646 DOI: 10.3390/ijms22189950] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/03/2021] [Accepted: 09/11/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric carcinoma (GC) represents one of the most common and most lethal malignancies worldwide. The histopathological characterization of GC precursor lesions has provided great knowledge about gastric carcinogenesis, with the consequent introduction of effective strategies of primary and secondary prevention. In recent years, a large amount of data about the molecular events in GC development is emerging, flanking the histomorphological descriptions. In this review, we describe the landscape of molecular alterations in gastric pre-invasive lesions with a glance at their potential use in the diagnostic and therapeutic decision-making process.
Collapse
|
39
|
Aumpan N, Vilaichone RK, Pornthisarn B, Chonprasertsuk S, Siramolpiwat S, Bhanthumkomol P, Nunanan P, Issariyakulkarn N, Ratana-Amornpin S, Miftahussurur M, Mahachai V, Yamaoka Y. Predictors for regression and progression of intestinal metaplasia (IM): A large population-based study from low prevalence area of gastric cancer (IM-predictor trial). PLoS One 2021; 16:e0255601. [PMID: 34379655 PMCID: PMC8357097 DOI: 10.1371/journal.pone.0255601] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Background Gastric intestinal metaplasia (IM) can lead to gastric cancer. Until now, there have been limited studies of predictors for regression and progression of IM. This study aimed to determine risk factors associated with regression or progression of IM for guiding proper management and prevention of gastric cancer. Methods 2,025 patients undergoing gastroscopy in Thammasat University Hospital, Thailand were enrolled during September 2017-August 2019. Patients’ data including baseline characteristics, laboratory results, and histopathology of gastric biopsies from University medical database were extensively reviewed. Results 2,025 patients had mean age of 61.3 years and 44.2% were males. Overall H. pylori prevalence was 47.5%. There were 1,551(76.6%) patients with chronic gastritis and 361(17.8%) with IM. Of 400 patients with chronic gastritis having follow-up endoscopy and repeated gastric biopsies, 104(26%) had persistent H. pylori infection and 27(26%) developed IM during mean follow-up time of 24 months. Persistent H. pylori infection was significantly associated with development of IM (OR 3.16, 95%CI 1.56–6.39, p = 0.001). Regression, persistence, and progression of IM were demonstrated in 57.3%, 39.2%, and 3.5% of patients, respectively. Age >65 years, persistent H. pylori infection, and diabetes mellitus were significantly associated with persistent IM or progression to dysplasia with OR 2.47(95%CI 1.33–4.61, p = 0.004), OR 2.64(95%CI 1.13–6.18, p = 0.025), and OR 2.54(95%CI 1.16–5.54, p = 0.019), respectively. Patients without H. pylori infection had more IM regression than patients with persistent infection (60.4%vs.39.4%, p = 0.035). Patients with persistent H. pylori infection significantly had higher IM progression to dysplasia (15.2%vs.2.1%; OR 11.15, 95%CI 1.18–105.24, p = 0.035) than noninfected. During 24 months of study, 30 patients (1.5%) were diagnosed with gastric cancer. Conclusion Regression of IM could be achieved by successful H. pylori eradication. Persistent H. pylori infection was significantly associated with development and progression of IM to dysplasia. Age >65 years and diabetes mellitus were also significant predictors for IM progression.
Collapse
Affiliation(s)
- Natsuda Aumpan
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Ratha-korn Vilaichone
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
- Chulabhorn International College of Medicine (CICM), Thammasat University, Pathumthani, Thailand
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- * E-mail:
| | - Bubpha Pornthisarn
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Soonthorn Chonprasertsuk
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Sith Siramolpiwat
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
- Chulabhorn International College of Medicine (CICM), Thammasat University, Pathumthani, Thailand
| | - Patommatat Bhanthumkomol
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Pongjarat Nunanan
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Navapan Issariyakulkarn
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Sarita Ratana-Amornpin
- Center of Excellence in Digestive Diseases and Gastroenterology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Muhammad Miftahussurur
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Varocha Mahachai
- Chulabhorn International College of Medicine (CICM), Thammasat University, Pathumthani, Thailand
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu, Japan
| |
Collapse
|
40
|
Ngamruengphong S, Ferri L, Aihara H, Draganov PV, Yang DJ, Perbtani YB, Jue TL, Munroe CA, Boparai ES, Mehta NA, Bhatt A, Kumta NA, Othman MO, Mercado M, Javaid H, Aadam AA, Siegel A, James TW, Grimm IS, DeWitt JM, Novikov A, Schlachterman A, Kowalski T, Samarasena J, Hashimoto R, Chehade NEH, Lee J, Chang K, Su B, Ujiki MB, Mehta A, Sharaiha RZ, Carr-Locke DL, Chen A, Chen M, Chen YI, Pourmousavi Khoshknab M, Wang R, Kerdsirichairat T, Tomizawa Y, von Renteln D, Kumbhari V, Khashab MA, Bechara R, Karasik M, Patel NJ, Fukami N, Nishimura M, Hanada Y, Wong Kee Song LM, Laszkowska M, Wang AY, Hwang JH, Friedland S, Sethi A, Kalloo AN. Efficacy of Endoscopic Submucosal Dissection for Superficial Gastric Neoplasia in a Large Cohort in North America. Clin Gastroenterol Hepatol 2021; 19:1611-1619.e1. [PMID: 32565290 DOI: 10.1016/j.cgh.2020.06.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 06/07/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Endoscopic submucosal dissection (ESD) is a widely accepted treatment option for superficial gastric neoplasia in Asia, but there are few data on outcomes of gastric ESD from North America. We aimed to evaluate the safety and efficacy of gastric ESD in North America. METHODS We analyzed data from 347 patients who underwent gastric ESD at 25 centers, from 2010 through 2019. We collected data on patient demographics, lesion characteristics, procedure details and related adverse events, treatment outcomes, local recurrence, and vital status at the last follow up. For the 277 patients with available follow-up data, the median interval between initial ESD and last clinical or endoscopic evaluation was 364 days. The primary endpoint was the rate of en bloc and R0 resection. Secondary outcomes included curative resection, rates of adverse events and recurrence, and gastric cancer-related death. RESULTS Ninety patients (26%) had low-grade adenomas or dysplasia, 82 patients (24%) had high-grade dysplasia, 139 patients (40%) had early gastric cancer, and 36 patients (10%) had neuroendocrine tumors. Proportions of en bloc and R0 resection for all lesions were 92%/82%, for early gastric cancers were 94%/75%, for adenomas and low-grade dysplasia were 93%/ 92%, for high-grade dysplasia were 89%/ 87%, and for neuroendocrine tumors were 92%/75%. Intraprocedural perforation occurred in 6.6% of patients; 82% of these were treated successfully with endoscopic therapy. Delayed bleeding occurred in 2.6% of patients. No delayed perforation or procedure-related deaths were observed. There were local recurrences in 3.9% of cases; all occurred after non-curative ESD resection. Metachronous lesions were identified in 14 patients (6.9%). One of 277 patients with clinical follow up died of metachronous gastric cancer that occurred 2.5 years after the initial ESD. CONCLUSIONS ESD is a highly effective treatment for superficial gastric neoplasia and should be considered as a viable option for patients in North America. The risk of local recurrence is low and occurs exclusively after non-curative resection. Careful endoscopic surveillance is necessary to identify and treat metachronous lesions.
Collapse
Affiliation(s)
| | - Lorenzo Ferri
- Division of Thoracic Surgery, Montreal General Hospital, McGill University, Montreal, Quebec, Canada
| | - Hiroyuki Aihara
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Boston, Massachusetts
| | - Peter V Draganov
- Division of Gastroenterology and Hepatology, University of Florida, Gainesville, Florida
| | - Dennis J Yang
- Division of Gastroenterology and Hepatology, University of Florida, Gainesville, Florida
| | - Yaseen B Perbtani
- Division of Gastroenterology and Hepatology, University of Florida, Gainesville, Florida
| | - Terry L Jue
- Division of Gastroenterology, Kaiser Permanente San Francisco, San Francisco, California
| | - Craig A Munroe
- Division of Gastroenterology, Kaiser Permanente San Francisco, San Francisco, California
| | - Eshandeep S Boparai
- Division of Gastroenterology, Kaiser Permanente San Francisco, San Francisco, California
| | - Neal A Mehta
- Department of Gastroenterology and Hepatology, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| | - Amit Bhatt
- Department of Gastroenterology and Hepatology, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| | - Nikhil A Kumta
- Division of Gastroenterology, Mount Sinai Hospital, New York, New York
| | - Mohamed O Othman
- Division of Gastroenterology and Hepatology, Baylor University Medical Center, Houston, Texas
| | - Michael Mercado
- Division of Gastroenterology and Hepatology, Baylor University Medical Center, Houston, Texas
| | - Huma Javaid
- Division of Gastroenterology and Hepatology, Baylor University Medical Center, Houston, Texas
| | - Abdul Aziz Aadam
- Division of Gastroenterology and Hepatology, Northwestern University Medical Center, Chicago, Illinois
| | - Amanda Siegel
- Division of Gastroenterology and Hepatology, Northwestern University Medical Center, Chicago, Illinois
| | - Theodore W James
- Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Ian S Grimm
- Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - John M DeWitt
- Division of Gastroenterology and Hepatology, Indiana University, Indianapolis, Indiana
| | - Aleksey Novikov
- Division of Gastroenterology and Hepatology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Alexander Schlachterman
- Division of Gastroenterology and Hepatology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Thomas Kowalski
- Division of Gastroenterology and Hepatology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Jason Samarasena
- Division of Gastroenterology and Hepatology, University of California, Irvine, Irvine, California
| | - Rintaro Hashimoto
- Division of Gastroenterology and Hepatology, University of California, Irvine, Irvine, California
| | - Nabil El Hage Chehade
- Division of Gastroenterology and Hepatology, University of California, Irvine, Irvine, California
| | - John Lee
- Division of Gastroenterology and Hepatology, University of California, Irvine, Irvine, California
| | - Kenneth Chang
- Division of Gastroenterology and Hepatology, University of California, Irvine, Irvine, California
| | - Bailey Su
- Department of Surgery, NorthShore University Health System, Evanston, Illinois
| | - Michael B Ujiki
- Department of Surgery, NorthShore University Health System, Evanston, Illinois
| | - Amit Mehta
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, New York
| | - Reem Z Sharaiha
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, New York
| | - David L Carr-Locke
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, New York
| | - Alex Chen
- Division of Thoracic Surgery, Montreal General Hospital, McGill University, Montreal, Quebec, Canada
| | - Michael Chen
- Division of Thoracic Surgery, Montreal General Hospital, McGill University, Montreal, Quebec, Canada
| | - Yen-I Chen
- Division of Gastroenterology and Hepatology, Montreal General Hospital, McGill University, Montreal, Quebec, Canada
| | | | - Rui Wang
- Division of Gastroenterology, Johns Hopkins Hospital, Baltimore, Maryland
| | | | - Yutaka Tomizawa
- Division of Gastroenterology and Hepatology, University of Washington, Seattle, Washington
| | - Daniel von Renteln
- Division of Gastroenterology, University of Montreal Hospital Research Centre, University of Montreal Hospital Centre, Montreal, Quebec, Canada
| | - Vivek Kumbhari
- Division of Gastroenterology, Johns Hopkins Hospital, Baltimore, Maryland
| | - Mouen A Khashab
- Division of Gastroenterology, Johns Hopkins Hospital, Baltimore, Maryland
| | - Robert Bechara
- Division of Gastroenterology, Queen's University, Kingston, Ontario, Canada
| | - Michael Karasik
- Department of Gastroenterology, Hartford Hospital, Hartford, Connecticut
| | - Neej J Patel
- Division of Gastroenterology and Hepatology, Mayo Clinic Arizona, Phoenix, Arizona
| | - Norio Fukami
- Division of Gastroenterology and Hepatology, Mayo Clinic Arizona, Phoenix, Arizona
| | - Makoto Nishimura
- Gastroenterology and Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yuri Hanada
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | - Monika Laszkowska
- Division of Digestive and Liver Diseases, Columbia University Irving Medical Center, New York, New York
| | - Andrew Y Wang
- Division of Gastroenterology and Hepatology, University of Virginia, Charlottesville, Virginia
| | - Joo Ha Hwang
- Department of Gastroenterology, Stanford University School of Medicine, Stanford, California
| | - Shai Friedland
- Department of Gastroenterology, Hartford Hospital, Hartford, Connecticut
| | - Amrita Sethi
- Division of Digestive and Liver Diseases, Columbia University Irving Medical Center, New York, New York
| | - Antony N Kalloo
- Division of Gastroenterology, Johns Hopkins Hospital, Baltimore, Maryland
| |
Collapse
|
41
|
Abstract
Data on the evolution of gastric precancerous lesions (GPL), especially in countries of a Low gastric cancer incidence area are limited. Our objective was to study a long-term evolution of GPL in France.
Collapse
|
42
|
Hur C. Gastric Cancer:: An Update on the Rapidly Changing Characteristics and Evolving Opportunities for Interventions. Gastrointest Endosc Clin N Am 2021; 31:xv-xviii. [PMID: 34053643 DOI: 10.1016/j.giec.2021.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Chin Hur
- Director, Healthcare Innovations Research and Evaluation (HIRE) Professor, Columbia University, Department of Medicine, Columbia University Irving Medical Center, 622 W 168th Street, PH9-105C, New York, NY 10032, USA.
| |
Collapse
|
43
|
Lansdorp-Vogelaar I, Meester RGS, Laszkowska M, Escudero FA, Ward ZJ, Yeh JM. Cost-effectiveness of prevention and early detection of gastric cancer in Western countries. Best Pract Res Clin Gastroenterol 2021; 50-51:101735. [PMID: 33975689 DOI: 10.1016/j.bpg.2021.101735] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/15/2021] [Accepted: 02/19/2021] [Indexed: 02/08/2023]
Abstract
Gastric cancer (GC) is a significant global health problem, with Helicobacter pylori infection estimated to be responsible for 89% of non-cardiac GC cases, or 78% of all GC cases. The International Agency for Research on Cancer has called for Helicobacter pylori test-and-treat strategies in countries with high rates of GC. However, for countries with low rates of GC, such as most Western countries, the balance between benefits, harms and costs of screening is less clear-cut. GC is a disease with a well-characterized precancerous process, providing the basis for primary and secondary prevention efforts. However, rigorous data assessing the impact of such interventions in Western countries are lacking. In the absence of clinical trials, modelling offers a unique approach to evaluate the potential impact of various screening and surveillance interventions. In this paper, we provide an overview of modelling studies evaluating the cost-effectiveness of GC screening and surveillance in Western countries.
Collapse
Affiliation(s)
- Iris Lansdorp-Vogelaar
- Department of Public Health, Erasmus MC University Medical Center Rotterdam, the Netherlands.
| | - Reinier G S Meester
- Department of Public Health, Erasmus MC University Medical Center Rotterdam, the Netherlands.
| | - Monika Laszkowska
- Gastroenterology, Hepatology, and Nutrition Service, Memorial-Sloan Kettering Cancer Research Center New York, NY, USA.
| | | | - Zachary J Ward
- Center for Health Decision Science, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Jennifer M Yeh
- Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
44
|
Nguyen TH, Tan MC, Liu Y, Rugge M, Thrift AP, El-Serag HB. Prevalence of Gastric Intestinal Metaplasia in a Multiethnic US Veterans Population. Clin Gastroenterol Hepatol 2021; 19:269-276.e3. [PMID: 32184184 PMCID: PMC7890574 DOI: 10.1016/j.cgh.2020.03.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/06/2020] [Accepted: 03/01/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS There is a need to identify individuals with gastric intestinal metaplasia, a precursor to gastric cancer, so they can be offered screening and surveillance. We examined the prevalence of gastric intestinal metaplasia, detected by upper endoscopy biopsy analysis, in different race and ethnic subgroups. We also investigated the extent to which Helicobacter pylori infection, with or without acute and chronic gastritis, accounts for observed associations between race or ethnicity and risk of gastric intestinal metaplasia. METHODS We used data from a cross-sectional study of consecutively recruited patients at the Michael E. DeBakey Veterans Affairs Medical Center in Houston, Texas, from February 2008 to August 2013. All participants completed a study questionnaire on sociodemographic and clinical characteristics and underwent upper endoscopy with gastric mapping (7 biopsy sites). Cases were classified as having gastric intestinal metaplasia if intestinal metaplasia was detected in 1 or more noncardia gastric biopsies; noncases were participants without evidence of gastric intestinal metaplasia. We used logistic regression models to estimate odds ratios (ORs) and 95% CI values to examine the association between race or ethnicity and gastric intestinal metaplasia and performed a mediation analysis to determine whether H pylori and gastritis affected observed associations. RESULTS We included 415 cases with gastric intestinal metaplasia and 1764 noncases. The prevalence of gastric intestinal metaplasia was highest among Hispanic patients (29.5%; 95% CI, 23.7%-36.1%), followed by African American (25.5%; 95% CI, 22.4%-28.9%) and non-Hispanic white patients (13.7%; 95% CI, 11.9%-15.7%). After we adjusted for age, sex, and smoking, African American (OR, 1.87; 95% CI, 1.44-2.44) and Hispanic race or ethnicity (OR, 2.32; 95% CI, 1.61-3.34) and H pylori infection (OR, 3.65; 95% CI, 2.79-4.55) were associated with an increased risk of gastric intestinal metaplasia. H pylori infection alone accounted for 33.6% of the association of race or ethnicity with gastric intestinal metaplasia, and 55.5% of the association when combined with acute and chronic gastritis. CONCLUSIONS Hispanic and African American patients have an increased risk for gastric intestinal metaplasia, determined by upper endoscopy biopsy analysis, compared with non-Hispanic white patients. This increase in risk was partially independent of H pylori infection.
Collapse
Affiliation(s)
- Theresa H Nguyen
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas; Center for Innovations in Quality, Effectiveness and Safety, Michael E DeBakey Veterans Affairs Medical Center, Houston, Texas
| | - Mimi C Tan
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Yan Liu
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas; Center for Innovations in Quality, Effectiveness and Safety, Michael E DeBakey Veterans Affairs Medical Center, Houston, Texas
| | - Massimo Rugge
- Department of Diagnostic Sciences, University of Padova, Padova, Italy
| | - Aaron P Thrift
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, Texas; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Hashem B El-Serag
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas; Center for Innovations in Quality, Effectiveness and Safety, Michael E DeBakey Veterans Affairs Medical Center, Houston, Texas.
| |
Collapse
|
45
|
Abstract
Background: We aimed to provide insight into the actual frequencies of gastric adenoma types and their association with gastritis status and associated mucosal changes with a focus on Helicobacter infection and the operative link on gastritis assessment (OLGA)/operative link on gastric intestinal metaplasia assessment (OLGIM) staging. Methods: From the archive of the Institute of Pathology in Bayreuth, we collected a consecutive series of 1058 gastric adenomas diagnosed between 1987 and 2017. Clinicopathological parameters retrieved from diagnostic reports included adenoma type and localization, associated mucosal changes in antrum and corpus (i.e., type of gastritis, the extent of intestinal metaplasia and atrophy), gender, date of birth, and date of diagnosis. Results: Intestinal-type adenoma was the most frequent adenoma (89.1%), followed by foveolar-type adenoma (4.3%), pyloric gland adenoma (3.4%), adenomas associated with hereditary tumor syndromes (2.8%), and oxyntic gland adenoma (0.4%). Adenomas were found in the background of Helicobacter pylori (H. pylori) gastritis in 23.9%, Ex-H. pylori gastritis in 36.0%, autoimmune gastritis in 24.8%, chemical reactive gastritis in 7.4%, and others in 0.1%. More than 70% of patients with gastric adenomas had low-risk stages in OLGA and OLGIM. Conclusions: We found a higher frequency of foveolar-type adenoma than anticipated from the literature. It needs to be questioned whether OLGA/OLGIM staging can be applied to all patients.
Collapse
|
46
|
Wang N, Chen M, Ni Z, Li T, Zeng J, Lu G, Wang J, Zhang J, Wu S, Shi Y. HDAC6/HNF4α loop mediated by miR-1 promotes bile acids-induced gastric intestinal metaplasia. Gastric Cancer 2021; 24:103-116. [PMID: 32705446 PMCID: PMC7790803 DOI: 10.1007/s10120-020-01108-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Gastric intestinal metaplasia (IM) is considered a precancerous lesion, and bile acids (BA) play a critical role in the induction of IM. Ectopic expression of HNF4α was observed in a BA-induced IM cell model. However, the mechanisms underlying the upregulation of the protein in IM cells remains to be elucidated. METHODS The effects of HNF4α on gastric mucosal cells in vivo were identified by a transgenic mouse model and RNA-seq was used to screen downstream targets of deoxycholic acid (DCA). The expression of pivotal molecules and miR-1 was detected by immunohistochemistry and in situ hybridization in normal, gastritis and IM tissue slides or microarrays. The transcriptional regulation of HDAC6 was investigated by chromatin immunoprecipitation (ChIP) and luciferase reporter assays. RESULTS The transgenic mouse model validated that HNF4α stimulated the HDAC6 expression and mucin secretion in gastric mucosa. Increased HDAC6 and HNF4α expression was also detected in the gastric IM cell model and patient specimens. HNF4α could bind to and activate HDAC6 promoter. In turn, HDAC6 enhanced the HNF4α protein level in GES-1 cells. Furthermore, miR-1 suppressed the expression of downstream intestinal markers by targeting HDAC6 and HNF4α. CONCLUSIONS Our findings show that the HDAC6/HNF4α loop regulated by miR-1 plays a critical role in gastric IM. Blocking the activation of this loop could be a potential approach to preventing BA-induced gastric IM or even gastric cancer (GC).
Collapse
Affiliation(s)
- Na Wang
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, No. 15 West Changle Road, Xi’an, 710032 Shaanxi China
| | - Min Chen
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, No. 15 West Changle Road, Xi’an, 710032 Shaanxi China
| | - Zhen Ni
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, No. 15 West Changle Road, Xi’an, 710032 Shaanxi China ,Department of Gastroenterology, General Hospital of Western Theater Command, Chengdu, Sichuan China
| | - Ting Li
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, No. 15 West Changle Road, Xi’an, 710032 Shaanxi China ,Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi’an Jiaotong University, Xi’an, China ,Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of education, Xi’an, China
| | - Jiaoxia Zeng
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, No. 15 West Changle Road, Xi’an, 710032 Shaanxi China
| | - Guofang Lu
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, No. 15 West Changle Road, Xi’an, 710032 Shaanxi China
| | - Jiaojiao Wang
- Shannxi University of Chinese Medicine, Xi’an, China
| | - Jian Zhang
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, No. 15 West Changle Road, Xi’an, 710032 Shaanxi China
| | - Siran Wu
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, No. 15 West Changle Road, Xi’an, 710032 Shaanxi China
| | - Yongquan Shi
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, No. 15 West Changle Road, Xi’an, 710032 Shaanxi China
| |
Collapse
|
47
|
Chapelle N, Jirka I, Péron M, Quénéhervé L, Cauchin E, Touchefeu Y, Coron E, Mosnier JF, Matysiak-Budnik T. Evaluation of a Phone Call Reminder Strategy in the Surveillance of Patients with Gastric Precancerous Lesions Lost to Follow-Up. Gastrointest Tumors 2020; 7:110-116. [PMID: 33173774 DOI: 10.1159/000508873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/22/2020] [Indexed: 12/24/2022] Open
Abstract
Background Surveillance of gastric precancerous lesions (GPL) may reduce gastric cancer (GC)-related mortality, but some patients with GPL are lost to follow-up. Objective The aim of this study was to evaluate the feasibility and efficacy of a "phone-call" strategy in surveillance of the lost to follow-up patients. Patients and Methods Among all the patients diagnosed with GPL (atrophic gastritis, intestinal metaplasia, low-grade dysplasia) between 2000 and 2015, we identified those who should undergo surveillance endoscopy according to the current guidelines. They were contacted by telephone and invited to undergo endoscopy with gastric biopsies for histological analysis. Results Among 535 patients with GPL, 134 were contacted. Sixty-two (46%) could not be joined, 36 did not have endoscopy for other reasons, and finally, 36 patients (22 males, median age 65 years) were included. After the median time interval of 57 months between 2 endoscopies, 18 patients showed stability, 11 regression, and 7 progression of GPL, including 1 patient who developed GC. Conclusion Despite several telephone calls, only one-third of the contacted patients could be brought to surveillance endoscopy. Most of the patients showed stability of GPL, but 1 progressed to GC and could be successfully treated.
Collapse
Affiliation(s)
- Nicolas Chapelle
- CHU de Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, Nantes, France.,Université de Nantes, Nantes, France.,INSERM, U1235, Nantes, France
| | - Iva Jirka
- CHU de Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - Matthieu Péron
- CHU de Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, Nantes, France.,Université de Nantes, Nantes, France
| | - Lucille Quénéhervé
- CHU de Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, Nantes, France.,Université de Nantes, Nantes, France.,INSERM, U1235, Nantes, France
| | - Estelle Cauchin
- CHU de Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, Nantes, France.,INSERM, U1235, Nantes, France
| | - Yann Touchefeu
- CHU de Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, Nantes, France.,INSERM, U1235, Nantes, France
| | - Emmanuel Coron
- CHU de Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, Nantes, France.,Université de Nantes, Nantes, France.,INSERM, U1235, Nantes, France
| | - Jean-François Mosnier
- Université de Nantes, Nantes, France.,Service d'Anatomie Pathologique, CHU de Nantes, Nantes, France
| | - Tamara Matysiak-Budnik
- CHU de Nantes, Hôtel Dieu, Institut des Maladies de l'Appareil Digestif, Nantes, France.,Université de Nantes, Nantes, France.,INSERM, U1235, Nantes, France
| |
Collapse
|
48
|
Dhingra R, Natov NS, Daaboul Y, Guelrud M, Cherukara A, Hung PF, Sterling MJ. Increased Risk of Progression to Gastric Adenocarcinoma in Patients with Non-dysplastic Gastric Intestinal Metaplasia Versus a Control Population. Dig Dis Sci 2020; 65:3316-3323. [PMID: 31916085 DOI: 10.1007/s10620-019-06031-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022]
Abstract
AIM In previous studies, the 5-year progression rate of gastric intestinal metaplasia to gastric adenocarcinoma has varied substantially. We investigated the incidence rate of dysplasia and gastric adenocarcinoma and the rate of progression among a cohort of patients with non-dysplastic gastric intestinal metaplasia. METHODS This is a single-center, single-cohort retrospective study. Patients who had undergone an EGD with biopsies from 01/01/1993 to 12/31/2013 were included. The primary outcome of interest was the composite of low-grade dysplasia, high-grade dysplasia, or adenocarcinoma. Time to progression and risk factor subgroup analyses were performed. RESULTS A total of 1628 subjects were screened, of whom 358 met the inclusion criteria. A total of 21 first-time events were recorded. The annual incidence rate of low-grade dysplasia was 2.1 (95% CI 1.3-3.5) cases per 1000 person-years, 0.5 (95% 0.2-1.3) per 1000 person-years for high-grade dysplasia, and 0.8 (95% CI 0.3-1.6) cases per 1000 person-years for gastric adenocarcinoma. The historical control group had an annual adenocarcinoma incidence rate of 0.07 per 1000 person-years. The event rate in Asians was also noted to be significantly higher between years 0-8 as compared with patients of non-Asian race, and extensive intestinal metaplasia was an independent risk factor (HR = 4.06 (95% CI 1.45-11.34), p = 0.007). CONCLUSIONS Patients with non-dysplastic gastric intestinal metaplasia may progress to dysplasia and gastric adenocarcinoma. The incidence rate of gastric adenocarcinoma is higher than that of the historical control population (0.07 per 1000 person-years). The presence of extensive intestinal metaplasia was a risk factor for progression of disease. Triennial EGD may be warranted in patients with non-dysplastic gastric intestinal metaplasia.
Collapse
Affiliation(s)
- Rohit Dhingra
- Department of Internal Medicine, Tufts Medical Center, Boston, MA, USA
| | - Nikola S Natov
- Division of Gastroenterology/Hepatology, Tufts Medical Center, Boston, MA, USA.
| | - Yazan Daaboul
- Department of Internal Medicine, Tufts Medical Center, Boston, MA, USA
| | - Moises Guelrud
- Division of Gastroenterology/Hepatology, Tufts Medical Center, Boston, MA, USA
| | | | - Pu Fang Hung
- Tufts University School of Medicine, Boston, MA, USA
| | - Mark J Sterling
- Division of Gastroenterology/Hepatology, Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
49
|
Gastric cancer screening in low incidence populations: Position statement of AEG, SEED and SEAP. GASTROENTEROLOGIA Y HEPATOLOGIA 2020; 44:67-86. [PMID: 33252332 DOI: 10.1016/j.gastrohep.2020.08.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/06/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023]
Abstract
This positioning document, sponsored by the Asociación Española de Gastroenterología, the Sociedad Española de Endoscopia Digestiva and the Sociedad Española de Anatomía Patológica, aims to establish recommendations for the screening of gastric cancer (GC) in low incidence populations, such as the Spanish. To establish the quality of the evidence and the levels of recommendation, we used the methodology based on the GRADE system (Grading of Recommendations Assessment, Development and Evaluation). We obtained a consensus among experts using a Delphi method. The document evaluates screening in the general population, individuals with relatives with GC and subjects with GC precursor lesions (GCPL). The goal of the interventions should be to reduce GC related mortality. We recommend the use of the OLGIM classification and determine the intestinal metaplasia (IM) subtype in the evaluation of GCPL. We do not recommend to establish endoscopic mass screening for GC or Helicobacter pylori. However, the document strongly recommends to treat H.pylori if the infection is detected, and the investigation and treatment in individuals with a family history of GC or with GCPL. Instead, we recommend against the use of serological tests to detect GCPL. Endoscopic screening is suggested only in individuals that meet familial GC criteria. As for individuals with GCPL, endoscopic surveillance is only suggested in extensive IM associated with additional risk factors (incomplete IM and/or a family history of GC), after resection of dysplastic lesions or in patients with dysplasia without visible lesion after a high quality gastroscopy with chromoendoscopy.
Collapse
|
50
|
Huang HL, Leung CY, Saito E, Katanoda K, Hur C, Kong CY, Nomura S, Shibuya K. Effect and cost-effectiveness of national gastric cancer screening in Japan: a microsimulation modeling study. BMC Med 2020; 18:257. [PMID: 32921305 PMCID: PMC7489209 DOI: 10.1186/s12916-020-01729-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/03/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND A national endoscopic screening program for gastric cancer was rolled out in Japan in 2015. We used a microsimulation model to estimate the cost-effectiveness of current screening guidelines and alternative screening strategies in Japan. METHODS We developed a microsimulation model that simulated a virtual population corresponding to the Japanese population in risk factor profile and life expectancy. We evaluated 15 endoscopic screening scenarios with various starting ages, stopping ages, and screening intervals. The primary outcomes were quality-adjusted life-years (QALYs), costs, and incremental cost-effectiveness ratio. Cost-effective screening strategies were determined using a willingness-to-pay threshold of $50,000 per QALY gained. One-way sensitivity and probabilistic sensitivity analyses were done to explore model uncertainty. RESULTS Using the threshold of $50,000 per QALY, a triennial screening program for individuals aged 50 to 75 years was the cost-effective strategy, with an incremental cost-effectiveness ratio of $45,665. Compared with no endoscopic screening, this strategy is predicted to prevent 63% of gastric cancer mortality and confer 27.2 QALYs gained per 1000 individuals over a lifetime period. Current screening guidelines were not on the cost-effectiveness efficient frontier. The results were robust on one-way sensitivity analyses and probabilistic sensitivity analysis. CONCLUSIONS This modeling study suggests that the endoscopic screening program in Japan would be cost-effective when implemented between age 50 and 75 years, with the screening repeated every 3 years. These findings underscore the need for further evaluation of the current gastric cancer screening recommendations.
Collapse
Affiliation(s)
- Hsi-Lan Huang
- Department of Global Health Policy, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Division of Cancer Statistics Integration, Center for Cancer Control and Information Services, National Cancer Center, Tokyo, Japan
| | - Chi Yan Leung
- Department of Global Health Policy, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- Division of Cancer Statistics Integration, Center for Cancer Control and Information Services, National Cancer Center, Tokyo, Japan.
| | - Eiko Saito
- Division of Cancer Statistics Integration, Center for Cancer Control and Information Services, National Cancer Center, Tokyo, Japan
| | - Kota Katanoda
- Division of Cancer Statistics Integration, Center for Cancer Control and Information Services, National Cancer Center, Tokyo, Japan
| | - Chin Hur
- Department of Medicine, Columbia University Irving Medical Center, New York City, USA
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, MA, USA
| | - Chung Yin Kong
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Shuhei Nomura
- Department of Global Health Policy, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Health Policy and Management, School of Medicine, Keio University, Tokyo, Japan
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Kenji Shibuya
- Department of Global Health Policy, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- University Institute for Population Health, King's College London, London, UK
| |
Collapse
|