1
|
Lan Y, Sun W, Zhong S, Xu Q, Xue Y, Liu Z, Shi L, Han B, Zhai T, Liu M, Sun Y, Xu H. A risk prediction model for gastric cancer based on endoscopic atrophy classification. BMC Cancer 2025; 25:518. [PMID: 40119304 PMCID: PMC11927292 DOI: 10.1186/s12885-025-13860-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 03/04/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUNDS Gastric cancer (GC) is a prevalent malignancy affecting the digestive system. We aimed to develop a risk prediction model based on endoscopic atrophy classification for GC. METHODS We retrospectively collected the data from January 2020 to October 2021 in our hospital and randomly divided the patients into training and validation sets in an 8:2 ratio. We used multiple machine learning algorithms such as logistic regression (LR), Decision tree, Support Vector Machine, Random forest, and so on to establish the models. We employed the Least absolute shrinkage and selection operator (LASSO) to screen variables for the LR model. However, we chose all the variables to construct the models for other machine learning algorithms. All models were evaluated using the receiver operating characteristic curve (ROC), predictive histograms, and decision curve analysis (DCA). RESULTS A total of 1156 patients were selected for the analysis. Five variables, including age, sex, family history of GC, HP infection status, and Kimura-Takemoto Classification (KTC), were screened using LASSO analysis. The area under the curve (AUC) of all the machine learning models ranged from 0.762 to 0.974 in the training set and from 0.608 to 0.812 in the validation set. Among them, the LR model exhibited the highest AUC value (0.812, 95%CI: 0.737-0.887) in the validation set with good calibration and clinical applicability. Finally, we constructed a nomogram to demonstrate the LR model. CONCLUSIONS We established a nomogram based on endoscopic atrophy classification for GC, which might be valuable in predicting GC risk and assisting clinical decision-making.
Collapse
Affiliation(s)
- Yadi Lan
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Weijia Sun
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Shen Zhong
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Qianqian Xu
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Yining Xue
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Zhaoyu Liu
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Lei Shi
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Bing Han
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Tianyu Zhai
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Mingyue Liu
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Yujing Sun
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Hongwei Xu
- Department of Gastroenterology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
| |
Collapse
|
2
|
Du S, Wang T, Li Z, Li T, Miao Z, Chen Y, Zhu S, Wei W, Deng H. Therapeutic Potential of Qilianxiaopi Formula: Targeting ADAM17-Mediated Chronic Inflammation in Atrophic Gastritis. Pharmaceuticals (Basel) 2025; 18:435. [PMID: 40143211 PMCID: PMC11944831 DOI: 10.3390/ph18030435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/09/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Gastric cancer (GC) is a leading cause of mortality worldwide, particularly in China. Chronic atrophic gastritis (CAG) and intestinal metaplasia (IM) are recognized as precancerous conditions contributing to GC development. Qilianxiaopi formula (QLXP), a traditional Chinese medicine (TCM), has demonstrated significant therapeutic effects on CAG and IM; however, its underlying mechanisms remain poorly understood. Methods: This study utilized chromatography-mass spectrometry to identify the major compounds in QLXP. Network pharmacology was used to predict the associated targets of these components. Thermal proteome profiling (TPP) pinpointed the potential binding proteins of QLXP, which were validated by bioinformatic analyses. Bio-layer interferometry (BLI) was used to analyze the interactions between QLXP and its key target proteins, thereby determining their binding components. Molecular docking predicted the binding modes between the components and proteins. Results: ADAM17 was identified as a key binding protein for QLXP. Further investigation revealed that QLXP inhibits the enzymatic activity of ADAM17, thereby reducing the secretion of the pro-inflammatory cytokine TNF-α, contributing to the anti-inflammatory properties of QLXP. BLI confirmed direct and reversible binding interactions between QLXP and ADAM17. Narirutin, isolated from the ADAM17 binding fraction, displayed the highest affinity for QLXP. Conclusions: This study highlights ADAM17 as a key molecular target of QLXP and narirutin as its principal binding component. The integrated approach combining chromatography-mass spectrometry, network pharmacology, TPP, BLI, and molecular docking provides a robust framework for elucidating the mechanisms of action of TCM.
Collapse
Affiliation(s)
- Sijing Du
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (S.D.)
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Tianxiang Wang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (S.D.)
| | - Zhiqiang Li
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (S.D.)
| | - Ting Li
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (S.D.)
| | - Zelong Miao
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (S.D.)
| | - Yuling Chen
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (S.D.)
| | - Songbiao Zhu
- Chinese Institutes for Medical Research (CIMR), Beijing 100069, China
| | - Wei Wei
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (S.D.)
| |
Collapse
|
3
|
Rouphael C, Elkin B, El Dahdah J, Moufawad M, Yang Q, Bena J, Shah S, Kim MK. Practice Trends among US Gastroenterologists following the 2020 American Gastroenterological Association Guidelines on Gastric Intestinal Metaplasia: Data from a Tertiary Care Center. J Clin Gastroenterol 2025; 59:62-69. [PMID: 38502036 DOI: 10.1097/mcg.0000000000001991] [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: 10/10/2023] [Accepted: 02/09/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND AND AIMS Studies show variability in gastroenterologists' management of gastric intestinal metaplasia (GIM) in the United States. In 2020, the American Gastroenterological Association published GIM guidelines, recommending physician-patient shared decision-making on GIM surveillance based on risk factors. We compared gastroenterologists' communication trends of a GIM finding and surveillance recommendations before and after 2020 and evaluated patient and provider factors associated with a surveillance recommendation. METHODS A sample of patients diagnosed with GIM on biopsies from upper endoscopies performed in 2018 (cohort A) and 2021 (cohort B) were included. Logistic regression analysis assessed the association between patient/provider characteristics and surveillance recommendations in the overall cohort and over time. MATERIALS In all, 347 patients were included: 175 in cohort A and 172 in B. Median age was 65.7 (56.0, 73.4), and 54.5% were females. Communication to patients about GIM findings and surveillance recommendations increased from 24.6% <2020 to 50% >2020 ( P <0.001) and 20% <2020 to 41.3% >2020 ( P <0.001), respectively. Overall, endoscopy >2020, family history of gastric cancer, autoimmune gastritis, female providers, and gastroenterologists with 10 to 20 years of experience were associated with a surveillance recommendation. The effect of family history of gastric cancer and the effect of the patient's female sex on surveillance was significantly different between both cohorts [Odds ratio (OR): 0.13, 95% (Confidence interval) CI: 0.02, 0.97 and OR 3.39, 95% CI: 1.12, 10.2, respectively). CONCLUSIONS Despite a 2-fold increase in surveillance recommendations after 2020, there was no meaningful effect of any of the patients' factors on a recommendation for surveillance over time, which raises the question as to whether surveillance is being offered to both average and high-risk patients without thorough risk stratification.
Collapse
Affiliation(s)
- Carol Rouphael
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute
| | | | | | | | - Qijun Yang
- Department of Qualitative Health Sciences, Cleveland Clinic, Cleveland, OH
| | - James Bena
- Department of Qualitative Health Sciences, Cleveland Clinic, Cleveland, OH
| | - Shailja Shah
- Department of Gastroenterology, University of California San Diego
- Jennifer Moreno Department of Veterans Affairs Medical Center, Gastroenterology Section, San Diego, CA
| | - Michelle K Kim
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute
| |
Collapse
|
4
|
Abe R, Uchikoshi S, Horikawa Y, Mimori N, Kato Y, Tahata Y, Fushimi S, Saito M, Takahashi S. Endoscopic and Histological Characteristics of Gastric Cancer Detected Long After Helicobacter pylori Eradication Therapy. Cancers (Basel) 2024; 16:4153. [PMID: 39766053 PMCID: PMC11674422 DOI: 10.3390/cancers16244153] [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: 11/11/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Since 2013, eradication therapy for Helicobacter pylori gastritis (Hp-ET) has been covered by the National Health Insurance of Japan. Recently, the risk of post-eradication gastric cancer (pE-GC) has increased. pE-GC includes cancers that develop immediately and several years after Hp-ET. Therefore, we aimed to clarify the endoscopic and histological characteristics of late types of pE-GCs. METHOD One hundred patients with differentiated cancers detected after Hp-ET who underwent endoscopic submucosal dissection from 2015 to 2023 were compared. Patients were divided into two groups; the immediate group (n = 69), with cancer detected within 6 years, and the delayed group (n = 31), with cancer detected within >6 years after Hp-ET. The background mucosa and tumor mucosa were examined individually. The endoscopic findings were as follows: enlarged folds, map-like redness, intermediate zone irregularity, and the presence of a regular arrangement of collecting venules and a light blue crest (background); an irregular surface structure, an irregular vascular pattern, an irregular surface pattern, and a gastritis-like appearance (tumor). The histological findings were as follows: a low remnant rate of the fundic glands, intestinal metaplasia (IM), crypt enlargement, and neutrophil infiltration (background); mosaicism, the elongation of noncancer ducts, and an overlying non-neoplastic epithelium (tumor). RESULTS There was no significant difference regarding the background mucosa and tumor mucosa between the two groups. In the delayed group, the remnant rate of the fundic glands was 19.8 ± 15.6%, and IM was 87.1% (27/31). Further, 90.3% (28/31) of the patients exhibited persistent neutrophil infiltration. CONCLUSION This study suggested that patients with a low remnant rate of the fundic gland and IM and persistent mucosal inflammation were at high risk for developing pE-GCs.
Collapse
Affiliation(s)
- Ryo Abe
- Department of Gastroenterology, Hiraka General Hospital, Yokote 013-8610, Akita, Japan; (R.A.); (N.M.); (Y.K.); (Y.T.); (S.F.)
- Department of Gastroenterology, Graduate School of Medicine, Akita University, Akita 010-8543, Akita, Japan
| | - Shu Uchikoshi
- Matsuzono Second Hospital, Morioka 020-0103, Iwate, Japan;
| | - Yohei Horikawa
- Department of Gastroenterology, Hiraka General Hospital, Yokote 013-8610, Akita, Japan; (R.A.); (N.M.); (Y.K.); (Y.T.); (S.F.)
- Crea Clinic, Sendai 980-0021, Miyagi, Japan
| | - Nobuya Mimori
- Department of Gastroenterology, Hiraka General Hospital, Yokote 013-8610, Akita, Japan; (R.A.); (N.M.); (Y.K.); (Y.T.); (S.F.)
| | - Yuhei Kato
- Department of Gastroenterology, Hiraka General Hospital, Yokote 013-8610, Akita, Japan; (R.A.); (N.M.); (Y.K.); (Y.T.); (S.F.)
| | - Yuta Tahata
- Department of Gastroenterology, Hiraka General Hospital, Yokote 013-8610, Akita, Japan; (R.A.); (N.M.); (Y.K.); (Y.T.); (S.F.)
| | - Saki Fushimi
- Department of Gastroenterology, Hiraka General Hospital, Yokote 013-8610, Akita, Japan; (R.A.); (N.M.); (Y.K.); (Y.T.); (S.F.)
| | - Masahiro Saito
- Department of Clinicopathology, Hiraka General Hospital, Yokote 013-8610, Akita, Japan; (M.S.); (S.T.)
| | - Satsuki Takahashi
- Department of Clinicopathology, Hiraka General Hospital, Yokote 013-8610, Akita, Japan; (M.S.); (S.T.)
| |
Collapse
|
5
|
Xu Y, Li GD, Wu CH, Zhong XQ. Nomogram prediction model for gastric cancer risk in chronic atrophic gastritis: Role of blood cell ratios. Shijie Huaren Xiaohua Zazhi 2024; 32:811-820. [DOI: 10.11569/wcjd.v32.i11.811] [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: 09/30/2024] [Revised: 10/20/2024] [Accepted: 11/21/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Chronic atrophic gastritis (CAG) is a precancerous condition for gastric cancer. Although endoscopy is the standard method for monitoring CAG, its invasive nature and high cost limit its use.
AIM To identify risk factors for gastric cancer in the CAG population, focusing on blood cell ratios, and develop a personalized prediction model using a nomogram.
METHODS A retrospective analysis was conducted on 314 GAG patients admitted to Hangzhou Normal University Affiliated Hospital from January 2018 to January 2024. Data collected included demographic, serological, and blood cell parameters. Independent risk factors were identified using multivariate logistic regression and a nomogram model was constructed with R. Model performance was assessed using the area under the ROC curve (AUC), the Hosmer-Lemeshow test, and decision curve analysis (DCA).
RESULTS Significant predictive factors for gastric cancer in the CAG population included male gender (odds ratio [OR] = 2.214, P < 0.05), Helicobacter pylori (H. pylori) infection (OR = 2.686, P < 0.05), gastrin 17 (G-17) (OR = 1.037, P < 0.05), hemoglobin-to-red blood cell distribution width ratio (HRR) (OR = 0.648, P < 0.05), and lymphocyte-to-monocyte ratio (LMR) (OR = 0.645, P < 0.05). The prediction model, with an AUC of 0.854, demonstrated good fit (Hosmer-Lemeshow test: χ2 = 6.062, P = 0.640). DCA indicated the potential generalizability of the model.
CONCLUSION The nomogram provides a noninvasive, convenient, and cost-effective tool for screening gastric cancer in CAG patients, showing excellent discrimination and calibration. Further large-scale, multicenter studies are necessary to validate its efficacy across diverse populations.
Collapse
Affiliation(s)
- Yang Xu
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, Zhejiang Province, China
- Department of Gastroenterology and Hepatology, Hospital of Zhejiang People's Armed Police, Hangzhou 310051, Zhejiang Province, China
| | - Guo-Dong Li
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, Zhejiang Province, China
| | - Chen-Han Wu
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, Zhejiang Province, China
| | - Xue-Qing Zhong
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, Zhejiang Province, China
| |
Collapse
|
6
|
Liu YD, Mou JC. Current status of traditional Chinese medicine and Western medicine treatment of chronic atrophic gastritis after Helicobacter pylori eradication. Shijie Huaren Xiaohua Zazhi 2024; 32:821-826. [DOI: 10.11569/wcjd.v32.i11.821] [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: 08/28/2024] [Revised: 09/26/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Chronic atrophic gastritis (CAG) is considered to be an intermediate step in the development of gastric cancer, which can progress to intestinal metaplasia, dysplasia, and eventually gastric cancer. Helicobacter pylori (H. pylori) infection is the main pathogenic factor in CAG, and eradication of H. pylori is an important means of treatment. However, there is no consensus on the treatment of CAG after H. pylori eradication. This article summarizes modern and traditional medical treatments for CAG after H. pylori eradication. In Western medicine, CAG patients with symptoms of discomfort are treated with supportive drugs and then undergo follow-up, monitoring, and evaluation of atrophy and metaplasia to facilitate timely endoscopic treatment of precancerous lesions and early-stage cancer. Traditional Chinese medicine has diverse views on CAG, with the core focus on "toxin, deficiency, and stasis", and emphasizes that after the "toxin is gone", the main focus should be on replenishing deficiencies and removing stasis.
Collapse
Affiliation(s)
- Yu-Di Liu
- The First Department of Internal Medicine, Yiwu Hospital of Traditional Chinese Medicine, Yiwu 322000, Zhejiang Province, China
| | - Jin-Chao Mou
- The First Department of Internal Medicine, Yiwu Hospital of Traditional Chinese Medicine, Yiwu 322000, Zhejiang Province, China
| |
Collapse
|
7
|
Lee S, Cho SJ, Chung H, Kim B, Oh MJ, Na YS, Lee JH, Kim J, Kim SG. Risk Assessment of Metachronous Gastric Neoplasm after Endoscopic Resection for Early Gastric Cancer According to Age at Helicobacter pylori Eradication. Gut Liver 2024; 18:992-1001. [PMID: 38509700 PMCID: PMC11565006 DOI: 10.5009/gnl230383] [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: 09/25/2023] [Revised: 11/14/2023] [Accepted: 12/05/2023] [Indexed: 03/22/2024] Open
Abstract
Background/Aims Helicobacter pylori eradication can reduce the incidence of metachronous gastric neoplasm (MGN) after endoscopic submucosal dissection (ESD) for early gastric cancer (EGC). This study evaluated the risk of developing MGN after ESD for EGC based on age at H. pylori eradication. Methods Data of patients who underwent curative ESD for EGC with H. pylori infection between 2005 and 2018 were retrospectively analyzed. The patients were allocated to four groups according to age at H. pylori eradication: group 1 (<50 years), group 2 (50-59 years), group 3 (60-69 years), and group 4 (≥70 years). Results All patients were followed up for at least 5 years after ESD. The 5-year cumulative incidence of MGN was 2.1%, 7.0%, 8.7%, and 16.7% in groups 1, 2, 3, and 4, respectively (p<0.001), and groups 3 and 4 showed a significant increase in the risk of MGN (hazard ratio [HR], 4.66; 95% confidence interval [CI], 1.09 to 19.92 and HR, 10.75; 95% CI, 2.45 to 47.12). After adjustments for moderate to severe intestinal metaplasia based on the updated Sydney system, groups 3 and 4 remained significantly associated with MGN (HR, 4.40; 95% CI, 1.03 to 18.84 and HR, 10.14; 95% CI, 2.31 to 44.57). Conclusions The incidence of MGN after ESD for EGC increased with age at H. pylori eradication. Age at H. pylori eradication ≥60 years was an independent risk factor for MGN, even after adjusting for the presence of advanced intestinal metaplasia.
Collapse
Affiliation(s)
- Seunghan Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Soo-Jeong Cho
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyunsoo Chung
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Bokyung Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Mi Jin Oh
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yun Suk Na
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jun Hee Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jiyoon Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Sang Gyun Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
8
|
Huang Z, Chen S, Yin S, Shi Z, Gu L, Li L, Yin H, Huang Z, Li B, Chen X, Yang Y, Wang Z, Li H, Zhang C, He Y. Development and validation of a nomogram for predicting the risk of developing gastric cancer based on a questionnaire: a cross-sectional study. Front Oncol 2024; 14:1351967. [PMID: 39588309 PMCID: PMC11586234 DOI: 10.3389/fonc.2024.1351967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 10/21/2024] [Indexed: 11/27/2024] Open
Abstract
Background Detection of gastric cancer (GC) at early stages is an effective strategy for decreasing mortality. This study aimed to construct a prediction nomogram based on a questionnaire to assess the risk of developing GC. Methods Our study comprised a total of 4379 participants (2326 participants from outpatient at Fengqing People's Hospital were considered for model development and internal validation, and 2053 participants from outpatients at the endoscopy center at the Seventh Affiliated Hospital of Sun Yat-Sen University were considered for independent external validation) and gastric mucosa status was determined by endoscopy and biopsies. The eligible participants in development cohort from Fengqing people's Hospital were randomly separated into a training cohort (n=1629, 70.0%) and an internal validation cohort (n=697, 30.0%). The relevant features were selected by a least absolute shrinkage and selection operator (LASSO), and the ensuing features were evaluated through multivariable logistic regression analysis. Subsequently, the variables were selected to construct a prediction nomogram. The discriminative ability and predictive accuracy of the nomogram were evaluated by the C-index and calibration plot, respectively. Decision curve analysis (DCA) curves were used for the assessment of clinical benefit of the model. This model was developed to estimate the risk of developing neoplastic lesions according to the "transparent reporting of a multivariable prediction model for individual prognosis or diagnosis" (TRIPOD) statement. Results Six variables, including age, sex, alcohol consumption, cigarette smoking, education level, and Hp infection status, were independent risk factors for the development of neoplastic lesions. Thus, these variables were incorporated into the final nomogram. The AUC of the nomogram were 0.701, 0.657 and 0.699 in the training, internal validation, and external validation cohorts, respectively. The calibration curve showed that the nomogram was in good agreement with the observed outcomes. Compared to treatment of all patients or none, our nomogram showed a notably higher clinical benefit. Conclusion This nomogram proved to be a convenient, cost-effective tool to effectively predict an individual's risk of developing neoplastic lesions, and it can act as a prescreening tool before gastroscopy.
Collapse
Affiliation(s)
- Zhangsen Huang
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Songyao Chen
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Songcheng Yin
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Zhaowen Shi
- General Surgery, Fengqing People’s Hospital, Lincang, China
| | - Liang Gu
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Liang Li
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Haofan Yin
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zhijian Huang
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Bo Li
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Xin Chen
- General Surgery, Fengqing People’s Hospital, Lincang, China
| | - Yilin Yang
- General Surgery, Fengqing People’s Hospital, Lincang, China
| | - Zhengli Wang
- General Surgery, Fengqing People’s Hospital, Lincang, China
| | - Hai Li
- General Surgery, Fengqing People’s Hospital, Lincang, China
| | - Changhua Zhang
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yulong He
- Digestive Medicine Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| |
Collapse
|
9
|
Andersen GT, Ianevski A, Resell M, Pojskic N, Rabben HL, Geithus S, Kodama Y, Hiroyuki T, Kainov D, Grønbech JE, Hayakawa Y, Wang TC, Zhao CM, Chen D. Multi-bioinformatics revealed potential biomarkers and repurposed drugs for gastric adenocarcinoma-related gastric intestinal metaplasia. NPJ Syst Biol Appl 2024; 10:127. [PMID: 39496635 PMCID: PMC11535201 DOI: 10.1038/s41540-024-00455-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/13/2024] [Indexed: 11/06/2024] Open
Abstract
Biomarkers associated with the progression from gastric intestinal metaplasia (GIM) to gastric adenocarcinoma (GA), i.e., GA-related GIM, could provide valuable insights into identifying patients with increased risk for GA. The aim of this study was to utilize multi-bioinformatics to reveal potential biomarkers for the GA-related GIM and predict potential drug repurposing for GA prevention in patients. The multi-bioinformatics included gene expression matrix (GEM) by microarray gene expression (MGE), ScType (a fully automated and ultra-fast cell-type identification based solely on a given scRNA-seq data), Ingenuity Pathway Analysis, PageRank centrality, GO and MSigDB enrichments, Cytoscape, Human Protein Atlas and molecular docking analysis in combination with immunohistochemistry. To identify GA-related GIM, paired surgical biopsies were collected from 16 GIM-GA patients who underwent gastrectomy, yielding 64 samples (4 biopsies per stomach x 16 patients) for MGE. Co-analysis was performed by including scRNAseq and immunohistochemistry datasets of endoscopic biopsies of 37 patients. The results of the present study showed potential biomarkers for GA-related GIM, including GEM of individual patients, individual genes (such as RBP2 and CD44), signaling pathways, network of molecules, and network of signaling pathways with key topological nodes. Accordingly, potential treatment targets with repurposed drugs were identified including epidermal growth factor receptor, proto-oncogene tyrosine-protein kinase Src, paxillin, transcription factor Jun, breast cancer type 1 susceptibility protein, cellular tumor antigen p53, mouse double minute 2, and CD44.
Collapse
Affiliation(s)
- Gøran Troseth Andersen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Surgery, St. Olav's Hospital, Trondheim, Norway
- Department of Surgery, Namsos Hospital, Namsos, Norway
| | - Aleksandr Ianevski
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Mathilde Resell
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Naris Pojskic
- Laboratory for Bioinformatics and Biostatistics, University of Sarajevo - Institute for Genetic Engineering and Biotechnology, Sarajevo, Bosnia and Herzegovina
| | - Hanne-Line Rabben
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Synne Geithus
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Yosuke Kodama
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Tomita Hiroyuki
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Denis Kainov
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Jon Erik Grønbech
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Surgery, St. Olav's Hospital, Trondheim, Norway
| | - Yoku Hayakawa
- Department of Gastroenterology, Tokyo University Hospital, Tokyo, Japan
| | - Timothy C Wang
- Department of Digestive and Liver Diseases and Herbert Iring Comprehensive Cancer Center, Columbia University Medical Center, New York, USA
| | - Chun-Mei Zhao
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Duan Chen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| |
Collapse
|
10
|
Teshima H, Takigawa H, Kotachi T, Tsuboi A, Tanaka H, Yamashita K, Kishida Y, Urabe Y, Kuwai T, Ishikawa A, Oka S. A Proton Pump Inhibitor Independently Elevates Gastrin Levels as a Marker for Metachronous Gastric Cancer After Endoscopic Submucosal Dissection. J Clin Med 2024; 13:6599. [PMID: 39518740 PMCID: PMC11546463 DOI: 10.3390/jcm13216599] [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: 09/12/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Background and Objective: Serum markers such as gastrin and pepsinogen are useful for stratifying gastric cancer risk. However, their utility in predicting metachronous gastric cancer after endoscopic submucosal dissection (ESD) in patients with gastric cancer after Helicobacter pylori eradication (GCAE) is unclear. This study aimed to clarify predictive factors for metachronous gastric cancer after ESD with a focus on serum markers. Methods: A retrospective analysis was conducted on 197 patients with 224 GCAE lesions who underwent ESD at Hiroshima University Hospital between April 2010 and December 2019. In total, 63 patients with 74 differentiated-type lesions were classified into metachronous gastric cancer (MG) and non-metachronous gastric cancer (NMG) groups, excluding proton pump inhibitor (PPI) users, female patients, and undifferentiated-type cases. The predictive value of serum markers was assessed using ROC curve analysis, and their association with carcinogenesis was evaluated using multiple logistic regression. Furthermore, the incidence of MG was compared between long-term PPI users and non-users. Results: ROC analysis revealed that serum gastrin had the highest discriminative ability for MG (AUC 0.77, cut-off 99 pg/mL, sensitivity 61.6%, and specificity 80.0%). Severe mucosal atrophy and high gastrin levels were significantly more common in the MG group and were independent predictors (p < 0.01). Although serum gastrin levels were significantly elevated in PPI users, no increased risk of MG was observed. Conclusions: In addition to severe mucosal atrophy, PPI-independent elevated serum gastrin levels may be associated with an increased risk of MG after ESD. Serum gastrin may serve as a valuable marker for post-ESD cancer surveillance in GCAE patients.
Collapse
Affiliation(s)
- Hajime Teshima
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (H.T.); (T.K.); (A.T.); (H.T.); (K.Y.); (Y.K.); (Y.U.); (S.O.)
| | - Hidehiko Takigawa
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (H.T.); (T.K.); (A.T.); (H.T.); (K.Y.); (Y.K.); (Y.U.); (S.O.)
| | - Takahiro Kotachi
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (H.T.); (T.K.); (A.T.); (H.T.); (K.Y.); (Y.K.); (Y.U.); (S.O.)
| | - Akiyoshi Tsuboi
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (H.T.); (T.K.); (A.T.); (H.T.); (K.Y.); (Y.K.); (Y.U.); (S.O.)
| | - Hidenori Tanaka
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (H.T.); (T.K.); (A.T.); (H.T.); (K.Y.); (Y.K.); (Y.U.); (S.O.)
| | - Ken Yamashita
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (H.T.); (T.K.); (A.T.); (H.T.); (K.Y.); (Y.K.); (Y.U.); (S.O.)
| | - Yoshihiro Kishida
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (H.T.); (T.K.); (A.T.); (H.T.); (K.Y.); (Y.K.); (Y.U.); (S.O.)
| | - Yuji Urabe
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (H.T.); (T.K.); (A.T.); (H.T.); (K.Y.); (Y.K.); (Y.U.); (S.O.)
| | - Toshio Kuwai
- Gastrointestinal Endoscopy and Medicine, Hiroshima University Hospital, Hiroshima 734-8551, Japan;
| | - Akira Ishikawa
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiorshima University Hospital, Hiroshima 734-8551, Japan;
| | - Shiro Oka
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University Hospital, Hiroshima 734-8551, Japan; (H.T.); (T.K.); (A.T.); (H.T.); (K.Y.); (Y.K.); (Y.U.); (S.O.)
| |
Collapse
|
11
|
Jin G, Liu K, Guo Z, Dong Z. Precision therapy for cancer prevention by targeting carcinogenesis. Mol Carcinog 2024; 63:2045-2062. [PMID: 39140807 DOI: 10.1002/mc.23798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024]
Abstract
Cancer represents a major global public health burden, with new cases estimated to increase from 14 million in 2012 to 24 million by 2035. Primary prevention is an effective strategy to reduce the costs associated with cancer burden. For example, measures to ban tobacco consumption have dramatically decreased lung cancer incidence and vaccination against human papillomavirus can prevent cervical cancer development. Unfortunately, the etiological factors of many cancer types are not completely clear or are difficult to actively control; therefore, the primary prevention of such cancers is not practical. In this review, we update the progress on precision therapy by targeting the whole carcinogenesis process, especially for three high-risk groups: (1) those with chronic inflammation, (2) those with inherited germline mutations, and (3) those with precancerous lesions like polyps, gastritis, actinic keratosis or dysplasia. We believe that attenuating chronic inflammation, treating precancerous lesions, and removing high-risk tissues harboring germline mutations are precision methods for cancer prevention.
Collapse
Affiliation(s)
- Guoguo Jin
- Henan Key Laboratory of Chronic Disease Management, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Kangdong Liu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhiping Guo
- Henan Key Laboratory of Chronic Disease Management, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
| | - Zigang Dong
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
12
|
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] [Grants] [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
|
13
|
Li Z, Zheng X, Mu Y, Zhang M, Liu G. The intelligent gastrointestinal metaplasia assessment based on deformable transformer with token merging. Biomed Signal Process Control 2024; 95:106454. [DOI: 10.1016/j.bspc.2024.106454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
14
|
Arai J, Hayakawa Y, Tateno H, Fujiwara H, Kasuga M, Fujishiro M. The role of gastric mucins and mucin-related glycans in gastric cancers. Cancer Sci 2024; 115:2853-2861. [PMID: 39031976 PMCID: PMC11463072 DOI: 10.1111/cas.16282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/22/2024] [Accepted: 07/02/2024] [Indexed: 07/22/2024] Open
Abstract
Gastric mucins serve as a protective barrier on the stomach's surface, protecting from external stimuli including gastric acid and gut microbiota. Their composition typically changes in response to the metaplastic sequence triggered by Helicobacter pylori infection. This alteration in gastric mucins is also observed in cases of gastric cancer, although the precise connection between mucin expressions and gastric carcinogenesis remains uncertain. This review first introduces the relationship between mucin expressions and gastric metaplasia or cancer observed in humans and mice. Additionally, we discuss potential pathogenic mechanisms of how aberrant mucins and their glycans affect gastric carcinogenesis. Finally, we summarize challenges to target tumor-specific glycans by utilizing lectin-drug conjugates that can bind to specific glycans. Understanding the correlation and mechanism between these mucin expressions and gastric carcinogenesis could pave the way for new strategies in gastric cancer treatment.
Collapse
Affiliation(s)
- Junya Arai
- Division of Gastroenterology, The Institute for Medical ScienceAsahi Life FoundationChuo‐ku, TokyoJapan
- Department of Gastroenterology, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| | - Hiroaki Tateno
- Cellular and Molecular Biotechnology Research InstituteNational Institute of Advanced Industrial Science and Technology (AIST)TsukubaJapan
| | - Hiroaki Fujiwara
- Division of Gastroenterology, The Institute for Medical ScienceAsahi Life FoundationChuo‐ku, TokyoJapan
| | - Masato Kasuga
- The Institute for Medical ScienceAsahi Life FoundationChuo‐ku, TokyoJapan
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| |
Collapse
|
15
|
Kawamura M, Uedo N, Yao K, Koike T, Kanesaka T, Hatta W, Ogata Y, Iwai W, Yokosawa S, Honda J, Asonuma S, Okata H, Ohyauchi M, Ito H, Abe Y, Ara N, Kayaba S, Shinkai H, Kanemitsu T. Endoscopic and histological risk stratification for gastric cancer using gastric intestinal metaplasia. J Gastroenterol Hepatol 2024; 39:1910-1916. [PMID: 38740510 DOI: 10.1111/jgh.16617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND AND AIM Intestinal metaplasia (IM) of the gastric mucosa is strongly associated with the risk of gastric cancer (GC). This study was performed to investigate the usefulness of endoscopic and histological risk stratification for GC using IM. METHODS This was a post-hoc analysis of a multicenter prospective study involving 10 Japanese facilities (UMINCTR000027023). The ridge/tubulovillous pattern, light blue crest (LBC), white opaque substance (WOS), endoscopic grading of gastric IM (EGGIM) score using non-magnifying image-enhanced endoscopy, and operative link on gastric IM assessment (OLGIM) were evaluated for their associations with GC risk in all patients. RESULTS In total, 380 patients (115 with GC and 265 without GC) were analyzed. The presence of an LBC (limited to antrum: odds ratio [OR] 2.4 [95% confidence interval 1.1-5.0], extended to corpus: OR 3.6 [2.1-6.3]), the presence of WOS (limited to antrum: OR 3.0 [1.7-5.3], extended to corpus: OR 4.2 [2.1-8.2]), and histological IM (limited to antrum: OR 3.2 [1.4-7.4], extended to corpus: OR 8.5 [4.5-16.0]) were significantly associated with GC risk. Additionally, the EGGIM score (5-8 points: OR 8.8 [4.4-16.0]) and OLGIM (stage III/IV: OR 12.5 [6.1-25.8]) were useful for stratification of GC risk. The area under the receiver operating characteristic curve value for GC risk was 0.740 for OLGIM and 0.706 for EGGIM. CONCLUSIONS The LBC, WOS, EGGIM, and OLGIM were strongly associated with GC risk in Japanese patients. This finding can be useful for GC risk assessment in daily clinical practice.
Collapse
Affiliation(s)
- Masashi Kawamura
- Department of Gastroenterology, Sendai City Hospital, Sendai, Japan
| | - Noriya Uedo
- Department of Gastrointestinal Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Kenshi Yao
- Department of Endoscopy, Fukuoka University Chikushi Hospital, Fukuoka, Japan
| | - Tomoyuki Koike
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Kanesaka
- Department of Gastrointestinal Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Waku Hatta
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yohei Ogata
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Wataru Iwai
- Department of Gastroenterology, Miyagi Cancer Center, Natori, Japan
| | - Satoshi Yokosawa
- Department of Gastroenterology, Iwate Prefectural Iwai Hospital, Iwate, Japan
| | - Junya Honda
- Department of Gastroenterology, Iwate Prefectural Iwai Hospital, Iwate, Japan
| | - Sho Asonuma
- Department of Gastroenterology, South Miyagi Medical Center, Ogawara, Japan
| | - Hideki Okata
- Department of Gastroenterology, South Miyagi Medical Center, Ogawara, Japan
| | - Motoki Ohyauchi
- Department of Gastroenterology, Osaki Citizen Hospital, Ōsaki, Japan
| | - Hirotaka Ito
- Department of Gastroenterology, Osaki Citizen Hospital, Ōsaki, Japan
| | - Yasuhiko Abe
- Division of Endoscopy, Yamagata University Hospital, Yamagata, Japan
| | - Nobuyuki Ara
- Department of Gastroenterology, National Hospital Organization Sendai Medical Center, Sendai, Japan
| | - Shoichi Kayaba
- Department of Gastroenterology, Iwate Prefectural Isawa Hospital, Iwate, Japan
| | - Hirohiko Shinkai
- Department of Gastroenterology, Iwate Prefectural Isawa Hospital, Iwate, Japan
| | - Takao Kanemitsu
- Department of Gastroenterology, Fukuoka University Chikushi Hospital, Fukuoka, Japan
| |
Collapse
|
16
|
Zhu X, Ma C, Sa R, Wang Y, Zhu C, Zhao Y, Luo J, Liu X. CagA 3' region polymorphism of Helicobacter pylori and its association with chronic gastritis in the Chinese population. J Med Microbiol 2024; 73. [PMID: 39171760 DOI: 10.1099/jmm.0.001880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024] Open
Abstract
Introduction. Cytotoxin-associated gene A (CagA) from Helicobacter pylori is highly related to chronic gastritis. Tyrosine phosphorylation of Glu-Pro-Ile-Tyr-Ala (EPIYA) motifs from CagA determines the pathogenicity of H. pylori.Gap statement. The precise amino acid variations surrounding the EPIYA motifs and their correlation with clinical outcomes have been poorly explored.Aim. The purpose of this study was to examine the CagA 3' region polymorphism of H. pylori and its association with chronic gastritis in the Chinese population.Method. A total of 86 cagA-positive H. pylori strains were isolated from patients with chronic gastritis in two different hospitals in Beijing, PR China. Genomic DNA was extracted commercial kits, and the cagA 3' variable region of H. pylori was amplified by polymerase chain reaction (PCR). The PCR products were sequenced and analysed using the CLC Sequence Viewer, BioEdit, and WebLogo 3.Results. Two hundred and fifty-nine EPIYA motifs were identified from cagA-positive H. pylori strains. Notably, EPIYA-B exhibited a higher frequency of variation in comparison to EPIYA-A, EPIYA-C, and EPIYA-D. The prevalent sequences for East-Asian-type CagA were QVNK and TIDF, while KVNK and TIDD were most commonly observed for Western-type CagA. The CRPIA motifs of East-Asian-type CagA and Western-type CagA varied at positions 4, 6, 7, 8, and 10. CagA-ABD (73.2%) was the most prevalent type, followed by CagA-ABC (18.6%) and CagA-AB (3.4%). The ratio of CagA-ABD was observed to be higher in cases of chronic non-atrophic gastritis with erosive (NAGE) or chronic atrophic gastritis (AG) compared to chronic non-atrophic gastritis (NAG), and the difference was found to be statistically significant (χ2=59.000/64.000, P<0.001).Conclusions. The EPIYA segments of Western-type CagA and East-Asian-type CagA differ significantly and the presence of CagA-ABD may be associated with severe chronic gastritis from this study.
Collapse
Affiliation(s)
- Xiaoyan Zhu
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
- Yunnan Province Clinical Research Center for Laboratory Medicine, Kunming, PR China
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, PR China
| | - Chao Ma
- Beijing Institute of Occupational Disease Prevention and Treatment, Beijing, PR China
- Department of Clinical Laboratory, The Beijing Prevention and Treatment Hospital of Occupational Disease for Chemical Industry, Beijing, PR China
| | - Rina Sa
- General internal medicine, Jingdong Medical Area, General Hospital of Chinese PLA, Beijing, PR China
| | - Yaxuan Wang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, PR China
| | - Chaohui Zhu
- Department of Gastroenterology, The Eighth Medical Center of PLA General Hospital, Beijing, PR China
| | - Yajiao Zhao
- Department of Gastroenterology, Emergency General Hospital, Beijing, PR China
| | - Juan Luo
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
- Yunnan Institute of Digestive Diseases, Kunming, PR China
| | - Xiaochuan Liu
- Department of Gastroenterology, Emergency General Hospital, Beijing, PR China
| |
Collapse
|
17
|
Sugimoto M, Murata M, Murakami K, Yamaoka Y, Kawai T. Characteristic endoscopic findings in Helicobacter pylori diagnosis in clinical practice. Expert Rev Gastroenterol Hepatol 2024; 18:457-472. [PMID: 39162811 DOI: 10.1080/17474124.2024.2395317] [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: 04/10/2024] [Accepted: 08/19/2024] [Indexed: 08/21/2024]
Abstract
INTRODUCTION Helicobacter pylori is a major risk factor for gastric cancer. In addition to eradication therapy, early-phase detection of gastric cancer through screening programs using high-vision endoscopy is also widely known to reduce mortality. Although European and US guidelines recommend evaluation of atrophy and intestinal metaplasia by high-vision endoscopy and pathological findings, the guideline used in Japan - the Kyoto classification of gastritis - is based on endoscopic evaluation, and recommends the grading of risk factors. This system requires classification into three endoscopic groups: H. pylori-negative, previous H. pylori infection (inactive gastritis), and current H. pylori infection (active gastritis). Major endoscopic findings in active gastritis are diffuse redness, enlarged folds, nodularity, mucosal swelling, and sticky mucus, while those in H pylori-related gastritis - irrespective of active or inactive status - are atrophy, intestinal metaplasia, and xanthoma. AREAS COVERED This review describes the endoscopic characteristics of current H. pylori infection, and how characteristic endoscopic findings should be evaluated. EXPERT OPINION Although the correct evaluation of endoscopic findings related to H. pylori remains necessary, if findings of possible infection are observed, it is important to diagnose infection by detection methods with high sensitivity and specificity, including the stool antigen test and urea breath test.
Collapse
Affiliation(s)
- Mitsushige Sugimoto
- Division of Genome-Wide Infectious Diseases, Research Center for GLOBAL and LOCAL Infectious Disease, Oita University, Yufu, Japan
| | - Masaki Murata
- Department of Gastroenterology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | | | - Yoshio Yamaoka
- Division of Genome-Wide Infectious Diseases, Research Center for GLOBAL and LOCAL Infectious Disease, Oita University, Yufu, Japan
- Department of Environmental and Preventive Medicine, Oita University, Yufu, Japan
| | - Takashi Kawai
- Department of Gastroenterological Endoscopy, Tokyo Medical University Hospital, Shinjuku, Japan
| |
Collapse
|
18
|
Tao X, Zhu Y, Dong Z, Huang L, Shang R, Du H, Wang J, Zeng X, Wang W, Wang J, Li Y, Deng Y, Wu L, Yu H. An artificial intelligence system for chronic atrophic gastritis diagnosis and risk stratification under white light endoscopy. Dig Liver Dis 2024; 56:1319-1326. [PMID: 38246825 DOI: 10.1016/j.dld.2024.01.177] [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: 07/12/2023] [Revised: 11/06/2023] [Accepted: 01/05/2024] [Indexed: 01/23/2024]
Abstract
BACKGROUND AND AIMS The diagnosis and stratification of gastric atrophy (GA) predict patients' gastric cancer progression risk and determine endoscopy surveillance interval. We aimed to construct an artificial intelligence (AI) system for GA endoscopic identification and risk stratification based on the Kimura-Takemoto classification. METHODS We constructed the system using two trained models and verified its performance. First, we retrospectively collected 869 images and 119 videos to compare its performance with that of endoscopists in identifying GA. Then, we included original image cases of 102 patients to validate the system for stratifying GA and comparing it with endoscopists with different experiences. RESULTS The sensitivity of model 1 was higher than that of endoscopists (92.72% vs. 76.85 %) at image level and also higher than that of experts (94.87% vs. 85.90 %) at video level. The system outperformed experts in stratifying GA (overall accuracy: 81.37 %, 73.04 %, p = 0.045). The accuracy of this system in classifying non-GA, mild GA, moderate GA, and severe GA was 80.00 %, 77.42 %, 83.33 %, and 85.71 %, comparable to that of experts and better than that of seniors and novices. CONCLUSIONS We established an expert-level system for GA endoscopic identification and risk stratification. It has great potential for endoscopic assessment and surveillance determinations.
Collapse
Affiliation(s)
- Xiao Tao
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Yijie Zhu
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China; Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, PR China
| | - Zehua Dong
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Li Huang
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Renduo Shang
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Hongliu Du
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Junxiao Wang
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Xiaoquan Zeng
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Wen Wang
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Jiamin Wang
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Yanxia Li
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Yunchao Deng
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Lianlian Wu
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China.
| | - Honggang Yu
- Renmin Hospital of Wuhan University, Wuhan, PR China; Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, PR China.
| |
Collapse
|
19
|
Tziatzios G, Ziogas DΙ, Gkolfakis P, Papadopoulos V, Papaefthymiou A, Mathou N, Giannakopoulos A, Gerasimatos G, Paraskeva KD, Triantafyllou K. Endoscopic Grading and Sampling of Gastric Precancerous Lesions: A Comprehensive Literature Review. Curr Oncol 2024; 31:3923-3938. [PMID: 39057162 PMCID: PMC11276348 DOI: 10.3390/curroncol31070290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Gastric cancer remains a disease with an ominous prognosis, while early gastric cancer has a good-to-excellent prognosis, with 5-year survival rates of up to 92.6% after successful endoscopic resection. In this context, the accurate identification of patients with established gastric precancerous lesions, namely chronic atrophic gastritis and intestinal metaplasia, is the first step in a stepwise approach to minimize cancer risk. Although current guidelines advocate for the execution of random biopsies to stage the extent and severity of gastritis/intestinal metaplasia, modern biopsy protocols are still imperfect as they have limited reproducibility and are susceptible to sampling error. The advent of novel imaging-enhancing modalities, i.e., high-definition with virtual chromoendoscopy (CE), has revolutionized the inspection of gastric mucosa, leading to an endoscopy-based staging strategy for the management of these premalignant changes in the stomach. Nowadays, the incorporation of CE-targeted biopsies in everyday clinical practice offers not only the robust detection of premalignant lesions but also an improvement in quality, by reducing missed diagnoses along with mean biopsies and, thus, the procedural costs and the environmental footprint. In this review, we summarize the recent evidence regarding the endoscopic grading and sampling of gastric precancerous lesions.
Collapse
Affiliation(s)
- Georgios Tziatzios
- Department of Gastroenterology, General Hospital of Nea Ionia “Konstantopoulio-Patision”, 3-5, Theodorou Konstantopoulou, 14233 Athens, Greece; (P.G.); (N.M.); (A.G.); (G.G.); (K.D.P.)
| | - Dimitrios Ι. Ziogas
- 1st Department of Internal Medicine, 251 Hellenic Air Force & VA General Hospital, 11525 Athina, Greece;
| | - Paraskevas Gkolfakis
- Department of Gastroenterology, General Hospital of Nea Ionia “Konstantopoulio-Patision”, 3-5, Theodorou Konstantopoulou, 14233 Athens, Greece; (P.G.); (N.M.); (A.G.); (G.G.); (K.D.P.)
| | - Vasilios Papadopoulos
- Department of Gastroenterology, General University Hospital of Larissa, 41334 Larissa, Greece; (V.P.); (A.P.)
| | - Apostolis Papaefthymiou
- Department of Gastroenterology, General University Hospital of Larissa, 41334 Larissa, Greece; (V.P.); (A.P.)
- Endoscopy Unit, Cleveland Clinic London, London SW1X 7HY, UK
| | - Nikoletta Mathou
- Department of Gastroenterology, General Hospital of Nea Ionia “Konstantopoulio-Patision”, 3-5, Theodorou Konstantopoulou, 14233 Athens, Greece; (P.G.); (N.M.); (A.G.); (G.G.); (K.D.P.)
| | - Athanasios Giannakopoulos
- Department of Gastroenterology, General Hospital of Nea Ionia “Konstantopoulio-Patision”, 3-5, Theodorou Konstantopoulou, 14233 Athens, Greece; (P.G.); (N.M.); (A.G.); (G.G.); (K.D.P.)
| | - Gerasimos Gerasimatos
- Department of Gastroenterology, General Hospital of Nea Ionia “Konstantopoulio-Patision”, 3-5, Theodorou Konstantopoulou, 14233 Athens, Greece; (P.G.); (N.M.); (A.G.); (G.G.); (K.D.P.)
| | - Konstantina D. Paraskeva
- Department of Gastroenterology, General Hospital of Nea Ionia “Konstantopoulio-Patision”, 3-5, Theodorou Konstantopoulou, 14233 Athens, Greece; (P.G.); (N.M.); (A.G.); (G.G.); (K.D.P.)
| | - Konstantinos Triantafyllou
- Hepatogastroenterology Unit, Second Department of Internal Medicine, Propaedeutic, Medical School, National and Kapodistrian University of Athens, ‘‘Attikon” University General Hospital, 77591 Athens, Greece;
| |
Collapse
|
20
|
Romańczyk M, Osmola M, Link A, Druet A, Hémont C, Martin J, Chapelle N, Matysiak-Budnik T. Non-Invasive Markers for the Detection of Gastric Precancerous Conditions. Cancers (Basel) 2024; 16:2254. [PMID: 38927959 PMCID: PMC11202181 DOI: 10.3390/cancers16122254] [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: 05/12/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Gastric cancer (GC) is still one of the most prevalent cancers worldwide, with a high mortality rate, despite improvements in diagnostic and therapeutic strategies. To diminish the GC burden, a modification of the current diagnostic paradigm, and especially endoscopic diagnosis of symptomatic individuals, is necessary. In this review article, we present a broad review and the current knowledge status on serum biomarkers, including pepsinogens, gastrin, Gastropanel®, autoantibodies, and novel biomarkers, allowing us to estimate the risk of gastric precancerous conditions (GPC)-atrophic gastritis and gastric intestinal metaplasia. The aim of the article is to emphasize the role of non-invasive testing in GC prevention. This comprehensive review describes the pathophysiological background of investigated biomarkers, their status and performance based on available data, as well as their clinical applicability. We point out future perspectives of non-invasive testing and possible new biomarkers opportunities.
Collapse
Affiliation(s)
- Marcin Romańczyk
- Department of Gastroenterology, Academy of Silesia, 40-555 Katowice, Poland
- H-T. Medical Center, 43-100 Tychy, Poland
| | | | - Alexander Link
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Amaury Druet
- IMAD, Hepato-Gastroenterology & Digestive Oncology, University Hospital of Nantes, F-44093 Nantes, France
| | - Caroline Hémont
- CHU de Nantes, Laboratoire d’Immunologie, Center for ImmunoMonitoring Nantes-Atlantique (CIMNA), F-44000 Nantes, France
| | - Jerome Martin
- CHU de Nantes, Laboratoire d’Immunologie, Center for ImmunoMonitoring Nantes-Atlantique (CIMNA), F-44000 Nantes, France
- University of Nantes, INSERM, Centre de Recherche Translationnel en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
| | - Nicolas Chapelle
- IMAD, Hepato-Gastroenterology & Digestive Oncology, University Hospital of Nantes, F-44093 Nantes, France
- University of Nantes, INSERM, Centre de Recherche Translationnel en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
| | - Tamara Matysiak-Budnik
- IMAD, Hepato-Gastroenterology & Digestive Oncology, University Hospital of Nantes, F-44093 Nantes, France
- University of Nantes, INSERM, Centre de Recherche Translationnel en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
| |
Collapse
|
21
|
Arai J, Niikura R, Hayakawa Y, Hirata Y, Ushiku T, Fujishiro M. Autoimmune gastritis may be less susceptible to cancer development than Helicobacter pylori-related gastritis based on histological analysis. Gut 2024; 73:1037-1038. [PMID: 37197906 DOI: 10.1136/gutjnl-2023-330052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/06/2023] [Indexed: 05/19/2023]
Affiliation(s)
- Junya Arai
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Departmemt of Gastroenterology, The Institute for Adult Diseases, Asahi Life Foundation, Tokyo, Japan
| | - Ryota Niikura
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Gastroenterological Endoscopy, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshihiro Hirata
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tetsuo Ushiku
- Department of Pathology, The University of Tokyo, Tokyo, Japan
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
22
|
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
|
23
|
Waddingham W, Graham DG, Banks MR. Latest Advances in Endoscopic Detection of Oesophageal and Gastric Neoplasia. Diagnostics (Basel) 2024; 14:301. [PMID: 38337817 PMCID: PMC10855581 DOI: 10.3390/diagnostics14030301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Endoscopy is the gold standard for the diagnosis of cancers and cancer precursors in the oesophagus and stomach. Early detection of upper GI cancers requires high-quality endoscopy and awareness of the subtle features these lesions carry. Endoscopists performing surveillance of high-risk patients including those with Barrett's oesophagus, previous squamous neoplasia or chronic atrophic gastritis should be familiar with endoscopic features, classification systems and sampling techniques to maximise the detection of early cancer. In this article, we review the current approach to diagnosis of these conditions and the latest advanced imaging and diagnostic techniques.
Collapse
Affiliation(s)
- William Waddingham
- Department of Gastroenterology, Royal Free London NHS Foundation Trust, London NW3 2QG, UK
| | - David G. Graham
- Department of Gastroenterology, University College London NHS Foundation Trust, London NW1 2BU, UK
| | - Matthew R. Banks
- Department of Gastroenterology, University College London NHS Foundation Trust, London NW1 2BU, UK
| |
Collapse
|
24
|
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] [Revised: 08/09/2023] [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
|
25
|
Korzheva I, Laktyun’kina E, Kozyr’ L, Fomina M. The role of the Kyoto classification in the endoscopic diagnosis of gastritis associated with Helicobacter pylori infection. ENDOSCOPIC SURGERY 2024; 30:65. [DOI: 10.17116/endoskop20243004165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
26
|
Nakano T, Dohi O, Takagi T, Naito Y, Fukui H, Miyazaki H, Yasuda T, Yoshida T, Azuma Y, Ishida T, Kitae H, Matsumura S, Takayama S, Mizuno N, Kashiwagi S, Mizushima K, Inoue R, Doi T, Hirose R, Inoue K, Yoshida N, Kamada K, Uchiyama K, Ishikawa T, Konishi H, Itoh Y. Characteristics of Gastric Mucosa-Associated Microbiota in Patients with Early Gastric Cancer After Successful Helicobacter pylori Eradication. Dig Dis Sci 2023; 68:4398-4406. [PMID: 37875607 DOI: 10.1007/s10620-023-08154-2] [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: 12/24/2022] [Accepted: 10/10/2023] [Indexed: 10/26/2023]
Abstract
BACKGROUND Helicobacter pylori (H. pylori) is widely recognized as a definite carcinogen in gastric cancer (GC). Although H. pylori eradication reduces the risk of GC, GC recurrence has been detected even after successful H. pylori eradication. Recently, the analysis of gut microbiota was reported. AIMS This study aimed to evaluate the correlation between gastric mucosa-associated microbiota (G-MAM) and early gastric cancer (EGC) after successful H. pylori eradication. METHODS In this pilot study, G-MAM were collected during the esophagogastroduodenoscopy of 17 patients, receiving H. pylori eradication therapy at least 5 years ago. The patients were divided into those with EGC (the EGC group, 8 patients) and those without EGC (the NGC group, 9 patients). Microbial samples in the greater curvature of the pyloric site were obtained using an endoscopic cytology brush, and the G-MAM profiles of each sample were analyzed using 16S rRNA V3-V4 gene sequencing. RESULTS Between the two groups, there was no significant difference in the median age, sex, median period after successful eradication of H. pylori, the α diversity, and the average abundance at the phylum level. At the genus level, the average abundance of Unclassified Oxalobacteraceae, Capnocytophaga, and Haemophilus was significantly lower in the EGC group than in the NGC group (0.89 vs. 0.14%, P < 0.01, 0.28 vs. 0.00%, P < 0.01 and 5.84 vs. 2.16%, P = 0.034, respectively). CONCLUSIONS We demonstrated alternations in the profiles of G-MAM between the two groups. Our results suggest that G-MAM may influence carcinogenesis after successful H. pylori eradication.
Collapse
Affiliation(s)
- Takahiro Nakano
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
- Department of Gastroenterology and Hepatology, Japanese Red Cross Society Kyoto Daiichi Hospital, Kyoto, Japan
| | - Osamu Dohi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Tomohisa Takagi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yuji Naito
- Department of Human Immunology and Nutrition Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hayato Fukui
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hajime Miyazaki
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Takeshi Yasuda
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Takuma Yoshida
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yuka Azuma
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Tsugitaka Ishida
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hiroaki Kitae
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Shinya Matsumura
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Shun Takayama
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Naoki Mizuno
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Saori Kashiwagi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Katsura Mizushima
- Department of Human Immunology and Nutrition Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ryo Inoue
- Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University, Osaka, Japan
| | - Toshifumi Doi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Ryohei Hirose
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Ken Inoue
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Naohisa Yoshida
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Kazuhiro Kamada
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Kazuhiko Uchiyama
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Takeshi Ishikawa
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hideyuki Konishi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
27
|
Iizuka C, Sue S, Onodera S, Ikeda A, Ikeda R, Goda Y, Irie K, Kaneko H, Maeda S. Risk assessment of metachronous gastric cancer after endoscopic submucosal dissection based on endoscopic intestinal metaplasia. JGH Open 2023; 7:783-789. [PMID: 38034056 PMCID: PMC10684977 DOI: 10.1002/jgh3.12989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/16/2023] [Indexed: 12/02/2023]
Abstract
Background and Aim The incidence of metachronous gastric cancer (MGC) after endoscopic treatment for early gastric cancer (EGC) is high, but a method of risk assessment for MGC based on endoscopic findings has not been established. In this study, we focused on endoscopic intestinal metaplasia (IM) and investigated the risk for MGC after endoscopic submucosal dissection (ESD) for EGC. Methods This retrospective observational study involved patients who underwent curative ESD for EGC from April 2015 to January 2021. We assessed endoscopic IM using the pretreatment endoscopic examination images. The severity of endoscopic IM was classified into four levels: 0 (none), 1 (mild), 2 (moderate), and 3 (severe). Four different gastric areas were evaluated. We divided the patients into a low-score group and a high-score group, and compared the cumulative incidence of MGC. Results In total, 156 patients who met the inclusion criteria were followed up for at least 12 months after ESD, and MGC developed in 14 patients during a mean period oof 41.5 months. The endoscopic IM scores in the lesser curvature of the antrum, lesser curvature of the corpus, and greater curvature of the corpus were higher in patients with MGC than in those without MGC. In the corpus, the 5-year cumulative incidence of MGC was significantly higher in the high-score group than in the low-score group (29.8% vs 10.0%, P = 0.004). Conclusion The severity of endoscopic corpus IM was associated with MGC. Thus, patients with severe corpus IM at the time of ESD require careful examination and intensive follow-up.
Collapse
Affiliation(s)
- Chino Iizuka
- Department of GastroenterologyYokohama City University Graduate School of MedicineYokohamaJapan
| | - Soichiro Sue
- Department of GastroenterologyYokohama City University Graduate School of MedicineYokohamaJapan
| | - Sho Onodera
- Department of GastroenterologyYokohama City University Graduate School of MedicineYokohamaJapan
| | - Aya Ikeda
- Department of GastroenterologyYokohama City University Graduate School of MedicineYokohamaJapan
| | - Ryosuke Ikeda
- Department of GastroenterologyYokohama City University Graduate School of MedicineYokohamaJapan
| | - Yoshihiro Goda
- Department of GastroenterologyYokohama City University Graduate School of MedicineYokohamaJapan
| | - Kuniyasu Irie
- Department of GastroenterologyYokohama City University Graduate School of MedicineYokohamaJapan
| | - Hiroaki Kaneko
- Department of GastroenterologyYokohama City University Graduate School of MedicineYokohamaJapan
| | - Shin Maeda
- Department of GastroenterologyYokohama City University Graduate School of MedicineYokohamaJapan
| |
Collapse
|
28
|
Choe Y, Park JM, Kim JS, Cho YK, Kim BW, Choi MG. Factors influencing occurrence of metachronous gastric cancer after endoscopic resection: a systematic review and meta-analysis. Korean J Intern Med 2023; 38:831-843. [PMID: 37939666 PMCID: PMC10636538 DOI: 10.3904/kjim.2023.184] [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: 04/21/2023] [Revised: 06/12/2023] [Accepted: 06/19/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND/AIMS Metachronous gastric cancer (MGC) can occur after endoscopic resection for gastric cancer. Further studies on factors other than Helicobacter pylori infection are needed. This systematic review and meta-analysis aimed to evaluate risk factors for metachronous recurrence of endoscopically resected gastric cancer. METHODS We searched medical literature published by February 2023 and identified patients with MGC after endoscopic resection for gastric cancer. The occurrence of MGC and the presence of intestinal metaplasia (IM), severe atrophic gastritis (AG), and H. pylori infection were quantitatively analyzed. RESULTS We identified 2,755 patients from nine cohort studies who underwent endoscopic resection for gastric cancer by 2018. Those with severe AG or presence of IM had a significantly higher incidence of MGC than those without (RR 2.00, 95% CI 1.35-2.98, I2 = 52% for severe atrophy on antrum; RR 7.08, 95% CI 3.63-13.80, I2 = 0% for antral IM). Absolute risk difference of MGC occurrence was 7.1% in those with severe AG and 9.2% in those with IM. The difference in incidence rate per 1,000 person-years was 17.5 person-years for those with severe AG and 24.7 person-years for those with IM. However, H. pylori eradication did not significantly affect the occurrence of MGC (RR 1.18, 95% CI 0.88-1.59, I2 = 10%). CONCLUSION Gastric cancer patients with severe AG or presence of IM had a 2.0-fold or 7.0-fold higher risk of MGC occurrence after endoscopic resection than those without, respectively. They need more stringent follow-up to monitor MGC occurrences (CRD42023410940).
Collapse
Affiliation(s)
- Younghee Choe
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| | - Jae Myung Park
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
- Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| | - Joon Sung Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| | - Yu Kyung Cho
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| | - Byung-Wook Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| | - Myung-Gyu Choi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
- Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| |
Collapse
|
29
|
Wei Y, Min C, Zhao C, Li Y, Wang X, Jing X, Yu Y, Li X, Yin X. Endoscopic characteristics and high-risk background mucosa factors of early gastric cancer after helicobacter pylori eradication: a single-center retrospective study. Front Oncol 2023; 13:1272187. [PMID: 37849804 PMCID: PMC10577436 DOI: 10.3389/fonc.2023.1272187] [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: 08/03/2023] [Accepted: 08/30/2023] [Indexed: 10/19/2023] Open
Abstract
Purpose Gastric cancer still develops after successful Helicobacter pylori(Hp)eradication. In this study, we aimed to explore the characteristics and risks of mucosal factors. Methods A total of 139 early gastric cancers (EGC) diagnosed in 133 patients after successful eradication from January 2016 to December 2021 were retrospectively included in the Hp-eradication EGC group and 170 EGCs diagnosed in 158 patients were included in the Hp-positive EGC group. We analyzed the clinical, pathological, and endoscopic characteristics between the two groups to identify the features of EGC after Hp eradication. Another 107 patients with no EGC after Hp eradication were enrolled in a Hp-eradication non-EGC group. The background mucosal factors between the Hp-eradication EGC group and the Hp-eradication non-EGC group were compared to analyze the high-risk background mucosal factors of EGC after eradication. In addition, we divided the EGC group after Hp eradication into IIc type and non-IIc type according to endoscopic gross classification to assess the high-risk background factors of IIc-type EGC after Hp eradication. Results The endoscopic features of EGC after Hp eradication included location in the lower part of the stomach (p=0.001), yellowish color (p= 0.031), and smaller size (p=0.001). The moderate/severe gastric atrophy (GA), intestinal metaplasia (IM) in the corpus, severe diffuse redness, and map-like redness were risk factors for EGC after eradication (p=0.001, p=0.001, p=0.001, and p= 0.005, respectively). The Kyoto classification total score in the EGC group was higher than the non-EGC group (4 vs.3 p<0.001). A multivariate analysis revealed that depressed erosion (OR=3.42, 95% CI 1.35-8.65, p= 0.009) was an independent risk factor for IIc-type EGC after Hp eradication. Conclusion EGC after eradication are smaller and yellowish lesions located in the lower part of the stomach. The risk background mucosal factors include moderate/severe GA, IM in the corpus, severe diffuse redness, and map-like redness. The Kyoto classification total score of 4 or more after successful eradication treatment might indicate EGC risk. In addition, the IIc-type EGC should be cautioned in the presence of depressed erosion after Hp eradication.
Collapse
Affiliation(s)
- Yali Wei
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Congcong Min
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Chongguang Zhao
- Department of Clinical Medicine, Qingdao University Medical College, Qingdao, Shandong, China
| | - Yubei Li
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaowei Wang
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xue Jing
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yanan Yu
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaoyu Li
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaoyan Yin
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
30
|
Toyoshima O, Nishizawa T, Yoshida S, Matsuno T, Fujisawa G, Toyoshima A, Ebinuma H, Fujishiro M, Saito Y, Suzuki H. Gastric cancer incidence based on endoscopic Kyoto classification of gastritis. World J Gastroenterol 2023; 29:4763-4773. [PMID: 37664152 PMCID: PMC10473921 DOI: 10.3748/wjg.v29.i31.4763] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/20/2023] [Accepted: 07/28/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) incidence based on the endoscopic Kyoto classification of gastritis has not been systematically investigated using time-to-event analysis. AIM To examine GC incidence in an endoscopic surveillance cohort. METHODS This study was retrospectively conducted at the Toyoshima Endoscopy Clinic. Patients who underwent two or more esophagogastroduodenoscopies were enrolled. GC incidence was based on Kyoto classification scores, such as atrophy, intestinal metaplasia (IM), enlarged folds (EFs), nodularity, diffuse redness (DR), and total Kyoto scores. Hazard ratios (HRs) adjusted for age and sex were calculated using a Cox hazard model. RESULTS A total of 6718 patients were enrolled (median age 54.0 years; men 44.2%). During the follow-up period (max 5.02 years; median 2.56 years), GC developed in 34 patients. The average frequency of GCs per year was 0.19%. Kyoto atrophy scores 1 [HR with score 0 as reference: 3.66, 95% confidence interval (CI): 1.06 to 12.61], 2 (11.60, 3.82-35.27), IM score 2 (9.92, 4.37-22.54), EF score 1 (4.03, 1.63-9.96), DR scores 1 (6.22, 2.65-14.56), and 2 (10.01, 3.73-26.86) were associated with GC incidence, whereas nodularity scores were not. The total Kyoto scores of 4 (HR with total Kyoto scores 0-1 as reference: 6.23, 95%CI: 1.93 to 20.13, P = 0.002) and 5-8 (16.45, 6.29-43.03, P < 0.001) were more likely to develop GC, whereas the total Kyoto scores 2-3 were not. The HR of the total Kyoto score for developing GC per 1 rank was 1.75 (95%CI: 1.46 to 2.09, P < 0.001). CONCLUSION A high total Kyoto score (≥ 4) was associated with GC incidence. The endoscopy-based diagnosis of gastritis can stratify GC risk.
Collapse
Affiliation(s)
- Osamu Toyoshima
- Department of Gastroenterology, Toyoshima Endoscopy Clinic, Tokyo 157-0066, Japan
| | - Toshihiro Nishizawa
- Department of Gastroenterology and Hepatology, International University of Health and Welfare, Narita Hospital, Narita 286-8520, Japan
| | - Shuntaro Yoshida
- Department of Gastroenterology, Toyoshima Endoscopy Clinic, Tokyo 157-0066, Japan
| | - Tatsuya Matsuno
- Department of Gastroenterology, Toyoshima Endoscopy Clinic, Tokyo 157-0066, Japan
| | - Gota Fujisawa
- Department of Gastroenterology, Toyoshima Endoscopy Clinic, Tokyo 157-0066, Japan
| | - Akira Toyoshima
- Department of Colorectal Surgery, Japanese Red Cross Medical Center, Tokyo 150-8935, Japan
| | - Hirotoshi Ebinuma
- Department of Gastroenterology and Hepatology, International University of Health and Welfare, Narita Hospital, Narita 286-8520, Japan
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yutaka Saito
- Division of Endoscopy, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Hidekazu Suzuki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tokai University School of Medicine, Isehara 259-1193, Japan
| |
Collapse
|
31
|
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
|
32
|
Livzan MA, Mozgovoi SI, Gaus OV, Shimanskaya AG, Kononov AV. Histopathological Evaluation of Gastric Mucosal Atrophy for Predicting Gastric Cancer Risk: Problems and Solutions. Diagnostics (Basel) 2023; 13:2478. [PMID: 37568841 PMCID: PMC10417051 DOI: 10.3390/diagnostics13152478] [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: 06/24/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Patients suffering from chronic gastritis and developing gastric mucosa atrophy are at increased risk of the development of gastric cancer. The diagnosis of chronic atrophic gastritis (CAG) is a complex procedure involving a detailed history taking, a thorough physical examination and the use of laboratory and instrumental diagnostic methods among which the endoscopy of the upper digestive tract is the cornerstone because it allows the assessment of the topography of gastritis and identification of erosions and areas of intestinal metaplasia with the use of NBI endoscopy. However, the diagnosis of CAG requires morphological examination of the gastric mucosa. So, in addition to assessing macroscopic changes in the gastric mucosa, it is necessary to take biopsy specimens in accordance with the protocols for their morphological and immunohistochemical examination. In the absence of specific diagnostic stigmas of CAG, close cooperation between a clinician, endoscopist and pathologist is necessary. The article presents systematized data on the histopathological assessment of the gastric mucosa atrophy to predict the risk of gastric cancer.
Collapse
Affiliation(s)
- Maria A. Livzan
- Department of Internal Medicine and Gastroenterology, Omsk Sate Medical University, 644099 Omsk, Russia;
| | - Sergei I. Mozgovoi
- Department of Pathological Anatomy, Omsk Sate Medical University, 644099 Omsk, Russia
| | - Olga V. Gaus
- Department of Internal Medicine and Gastroenterology, Omsk Sate Medical University, 644099 Omsk, Russia;
| | - Anna G. Shimanskaya
- Department of Pathological Anatomy, Omsk Sate Medical University, 644099 Omsk, Russia
| | - Alexei V. Kononov
- Department of Pathological Anatomy, Omsk Sate Medical University, 644099 Omsk, Russia
| |
Collapse
|
33
|
Livzan MA, Gaus OV, Lisovskiy MA, Mozgovoi SI, Rubtsov VA, Parygina MN. Clinical supervision of chronic atrophic gastritis. EXPERIMENTAL AND CLINICAL GASTROENTEROLOGY 2023:148-155. [DOI: 10.31146/1682-8658-ecg-211-3-148-155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
Patients with chronic gastritis (CG) with the development of atrophy of the gastric mucosa are at an increased risk of developing gastric cancer (GC). In the management of such patients, the development of high-grade dysplasia and invasive gastric cancer should be defined as adverse outcomes that must be prevented. To this end, patients with a diagnosis of «Chronic atrophic fundic/multifocal gastritis» are subject to dynamic dispensary observation to assess the achievement of target indicators, take into account information about changes in the diagnosis and concomitant diseases, emerging complications, as well as to enter data on ongoing therapeutic and preventive measures. This article presents the main aspects of prevention and dispensary monitoring of patients with an increased risk of gastric cancer.
Collapse
|
34
|
Zha Y, Xue C, Liu Y, Ni J, De La Fuente JM, Cui D. Artificial intelligence in theranostics of gastric cancer, a review. MEDICAL REVIEW (2021) 2023; 3:214-229. [PMID: 37789960 PMCID: PMC10542883 DOI: 10.1515/mr-2022-0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 04/26/2023] [Indexed: 10/05/2023]
Abstract
Gastric cancer (GC) is one of the commonest cancers with high morbidity and mortality in the world. How to realize precise diagnosis and therapy of GC owns great clinical requirement. In recent years, artificial intelligence (AI) has been actively explored to apply to early diagnosis and treatment and prognosis of gastric carcinoma. Herein, we review recent advance of AI in early screening, diagnosis, therapy and prognosis of stomach carcinoma. Especially AI combined with breath screening early GC system improved 97.4 % of early GC diagnosis ratio, AI model on stomach cancer diagnosis system of saliva biomarkers obtained an overall accuracy of 97.18 %, specificity of 97.44 %, and sensitivity of 96.88 %. We also discuss concept, issues, approaches and challenges of AI applied in stomach cancer. This review provides a comprehensive view and roadmap for readers working in this field, with the aim of pushing application of AI in theranostics of stomach cancer to increase the early discovery ratio and curative ratio of GC patients.
Collapse
Affiliation(s)
- Yiqian Zha
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
- National Engineering Research Center for Nanotechnology, Shanghai, China
| | - Cuili Xue
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
- National Engineering Research Center for Nanotechnology, Shanghai, China
| | - Yanlei Liu
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
- National Engineering Research Center for Nanotechnology, Shanghai, China
| | - Jian Ni
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
- National Engineering Research Center for Nanotechnology, Shanghai, China
| | | | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
- National Engineering Research Center for Nanotechnology, Shanghai, China
| |
Collapse
|
35
|
Cheema HI, Tharian B, Inamdar S, Garcia-Saenz-de-Sicilia M, Cengiz C. Recent advances in endoscopic management of gastric neoplasms. World J Gastrointest Endosc 2023; 15:319-337. [PMID: 37274561 PMCID: PMC10236974 DOI: 10.4253/wjge.v15.i5.319] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/12/2023] [Accepted: 04/06/2023] [Indexed: 05/16/2023] Open
Abstract
The development and clinical application of new diagnostic endoscopic technologies such as endoscopic ultrasonography with biopsy, magnification endoscopy, and narrow-band imaging, more recently supplemented by artificial intelligence, have enabled wider recognition and detection of various gastric neoplasms including early gastric cancer (EGC) and subepithelial tumors, such as gastrointestinal stromal tumors and neuroendocrine tumors. Over the last decade, the evolution of novel advanced therapeutic endoscopic techniques, such as endoscopic mucosal resection, endoscopic submucosal dissection, endoscopic full-thickness resection, and submucosal tunneling endoscopic resection, along with the advent of a broad array of endoscopic accessories, has provided a promising and yet less invasive strategy for treating gastric neoplasms with the advantage of a reduced need for gastric surgery. Thus, the management algorithms of various gastric tumors in a defined subset of the patient population at low risk of lymph node metastasis and amenable to endoscopic resection, may require revision considering upcoming data given the high success rate of en bloc resection by experienced endoscopists. Moreover, endoscopic surveillance protocols for precancerous gastric lesions will continue to be refined by systematic reviews and meta-analyses of further research. However, the lack of familiarity with subtle endoscopic changes associated with EGC, as well as longer procedural time, evolving resection techniques and tools, a steep learning curve of such high-risk procedures, and lack of coding are issues that do not appeal to many gastroenterologists in the field. This review summarizes recent advances in the endoscopic management of gastric neoplasms, with special emphasis on diagnostic and therapeutic methods and their future prospects.
Collapse
Affiliation(s)
- Hira Imad Cheema
- Department of Internal Medicine, Baptist Health Medical Center, Little Rock, AR 72205, United States
| | - Benjamin Tharian
- Department of Interventional Endoscopy/Gastroenterology, Bayfront Health, Digestive Health Institute, St. Petersberg, FL 33701, United States
| | - Sumant Inamdar
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Mauricio Garcia-Saenz-de-Sicilia
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Cem Cengiz
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, John L. McClellan Memorial Veterans Hospital, Little Rock, AR 72205, United States
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, TOBB University of Economics and Technology, Ankara 06510, Turkey
| |
Collapse
|
36
|
Kang SJ, Kim JG, Moon HS, Kook MC, Lee JY, Bang CS, Tae CH, Gong EJ, Nam SY, Kim HJ. Clinical Practice Guideline for Gastritis in Korea. J Korean Med Sci 2023; 38:e115. [PMID: 37012690 PMCID: PMC10070048 DOI: 10.3346/jkms.2023.38.e115] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/09/2023] [Indexed: 04/05/2023] Open
Abstract
Gastritis is a disease characterized by inflammation of the gastric mucosa. It is very common and has various classification systems such as the updated Sydney system. As there is a lot of evidence that Helicobacter pylori infection is associated with the development of gastric cancer and that gastric cancer can be prevented by eradication, H. pylori gastritis has been emphasized recently. The incidence rate of gastric cancer in Korea is the highest in the world, and due to the spread of screening endoscopy, atrophic gastritis and intestinal metaplasia are commonly diagnosed in the general population. However, there have been no clinical guidelines developed in Korea for these lesions. Therefore, this clinical guideline has been developed by the Korean College of Helicobacter and Upper Gastrointestinal Research for important topics that are frequently encountered in clinical situations related to gastritis. Evidence-based guidelines were developed through systematic review and de novo processes, and eight recommendations were made for eight key questions. This guideline needs to be periodically revised according to the needs of clinical practice or as important evidence about this issue is published in the future.
Collapse
Affiliation(s)
- Seung Joo Kang
- Deparment of Internal Medicine, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, Korea
| | - Jae Gyu Kim
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea.
| | - Hee Seok Moon
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | | | - Jong Yeul Lee
- Center for Gastric Cancer, National Cancer Center, Goyang, Korea
| | - Chang Seok Bang
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | - Chung Hyun Tae
- Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Eun Jeong Gong
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | - Su Youn Nam
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
- Division of Gastroenterology, Department of Internal Medicine, Kyungpook National University Chilgok Hospital, Daegu, Korea
| | - Hyun Jung Kim
- Department of Preventive Medicine, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
37
|
Niikura R, Hayakawa Y, Nagata N, Miyoshi-Akiayama T, Miyabayashi K, Tsuboi M, Suzuki N, Hata M, Arai J, Kurokawa K, Abe S, Uekura C, Miyoshi K, Ihara S, Hirata Y, Yamada A, Fujiwara H, Ushiku T, Woods SL, Worthley DL, Hatakeyama M, Han YW, Wang TC, Kawai T, Fujishiro M. Non- Helicobacter pylori Gastric Microbiome Modulates Prooncogenic Responses and Is Associated With Gastric Cancer Risk. GASTRO HEP ADVANCES 2023; 2:684-700. [PMID: 39129877 PMCID: PMC11307406 DOI: 10.1016/j.gastha.2023.03.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/08/2023] [Indexed: 08/13/2024]
Abstract
Background and Aims Although Helicobacter pylori is the most important bacterial carcinogen in gastric cancer (GC), GC can emerge even after H. pylori eradication. Studies suggest that various constituents of the gastric microbiome may influence GC development, but the role of individual pathogens is unclear. Methods Human gastric mucosal samples were analyzed by 16SrRNA sequencing to investigate microbiome composition and its association with clinical parameters, including GC risk. Identified bacteria in the stomach were cocultured with gastric epithelial cells or inoculated into mice, and transcriptomic changes, DNA damage, and inflammation were analyzed. Bacterial reads in GC tissues were examined together with transcriptomic and genetic sequencing data in the cancer genome atlas dataset. Results Patients after Helicobacter pylori eradication formed 3 subgroups based on the microbial composition revealed by 16SrRNA sequencing. One dysbiotic group enriched with Fusobacterium and Neisseria species was associated with a significantly higher GC incidence. These species activated prooncogenic pathways in gastric epithelial cells and promoted inflammation in mouse stomachs. Sugar chains that constitute gastric mucin attenuate host-bacteria interactions. Metabolites from Fusobacterium species were genotoxic, and the presence of the bacteria was associated with an inflammatory signature and a higher tumor mutation burden. Conclusion Gastric microbiota in the dysbiotic stomach is associated with GC development after H. pylori eradication and plays a pathogenic role through direct host-bacteria interaction.
Collapse
Affiliation(s)
- Ryota Niikura
- Department of Gastroenterology, Graduate school of medicine, The University of Tokyo, Tokyo, Japan
- Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate school of medicine, The University of Tokyo, Tokyo, Japan
| | - Naoyoshi Nagata
- Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan
| | - Tohru Miyoshi-Akiayama
- Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Koji Miyabayashi
- Department of Gastroenterology, Graduate school of medicine, The University of Tokyo, Tokyo, Japan
| | - Mayo Tsuboi
- Department of Gastroenterology, Graduate school of medicine, The University of Tokyo, Tokyo, Japan
| | - Nobumi Suzuki
- Department of Gastroenterology, Graduate school of medicine, The University of Tokyo, Tokyo, Japan
| | - Masahiro Hata
- Department of Gastroenterology, Graduate school of medicine, The University of Tokyo, Tokyo, Japan
| | - Junya Arai
- Department of Gastroenterology, Graduate school of medicine, The University of Tokyo, Tokyo, Japan
| | - Ken Kurokawa
- Department of Gastroenterology, Graduate school of medicine, The University of Tokyo, Tokyo, Japan
| | - Sohei Abe
- Department of Gastroenterology, Graduate school of medicine, The University of Tokyo, Tokyo, Japan
| | - Chie Uekura
- Department of Gastroenterology, Graduate school of medicine, The University of Tokyo, Tokyo, Japan
| | - Kotaro Miyoshi
- Department of Gastroenterology, Graduate school of medicine, The University of Tokyo, Tokyo, Japan
| | - Sozaburo Ihara
- Department of Gastroenterology, Graduate school of medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshihiro Hirata
- Department of Gastroenterology, Graduate school of medicine, The University of Tokyo, Tokyo, Japan
| | - Atsuo Yamada
- Department of Gastroenterology, Graduate school of medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroaki Fujiwara
- Department of Gastroenterology, The Institute for Medical Science, Asahi-life Foundation, Tokyo, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate school of medicine, The University of Tokyo, Tokyo, Japan
| | - Susan L. Woods
- Cancer Theme, SAHMRI, Adelaide, South Australia, Australia
- Medical Specialties, Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | | | - Masanori Hatakeyama
- Department of Microbiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yiping W. Han
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University, New York, New York
- Department of Microbiology and Immunology, Vagelos College of Physicians & Surgeons, Columbia University, New York, New York
- Section of Oral, Diagnostic and Rehabilitation Sciences, College of Dental Medicine, Columbia University, New York, New York
| | - Timothy C. Wang
- Division of Digestive and Liver Disease, Department of Medicine, Columbia University, New York, New York
| | - Takashi Kawai
- Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology, Graduate school of medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
38
|
Zheng SY, Zhu L, Wu LY, Liu HR, Ma XP, Li Q, Wu MD, Wang WJ, Li J, Wu HG. Helicobacter pylori-positive chronic atrophic gastritis and cellular senescence. Helicobacter 2023; 28:e12944. [PMID: 36539375 DOI: 10.1111/hel.12944] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/03/2022] [Accepted: 12/05/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Chronic atrophic gastritis (CAG) is a pathological stage in the Correa's cascade, whereby Helicobacter pylori (H. pylori) infection is the primary cause. Cellular senescence is an inducing factor for cancer occurrence and cellular senescence is an obvious phenomenon in gastric mucosal tissues of H. pylori-positive CAG patients. METHODS In this review, we collated the information on cellular senescence and H. pylori-positive CAG. RESULTS At present, only a few studies have observed the effect of cellular senescence on precancerous lesions. In combination with the latest research, this review has collated the information on cellular senescence and H. pylori-positive CAG from four aspects- telomere shortening, DNA methylation, increased reacive oxygen species (ROS) production, and failure of autophagy. CONCLUSION This is expected to be helpful for exploring the relevant mechanisms underlying inflammatory cancerous transformation and formulating appropriate treatment strategies.
Collapse
Affiliation(s)
- Shi-Yu Zheng
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Zhu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu-Yi Wu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui-Rong Liu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Peng Ma
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qi Li
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meng-Die Wu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wen-Jia Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Li
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huan-Gan Wu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
39
|
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
|
40
|
Mizukami K, Kodama M, Fukuda M, Hirashita Y, Tsutsumi K, Fukuda K, Ogawa R, Okamoto K, Okimoto T, Murakami K. Comparison of the improvement in gastric mucosal tissue after Helicobacter pylori eradication between young and elderly people. Arab J Gastroenterol 2023:S1687-1979(23)00007-2. [PMID: 36720666 DOI: 10.1016/j.ajg.2023.01.005] [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: 05/08/2022] [Revised: 08/13/2022] [Accepted: 01/08/2023] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND STUDY AIMS Helicobacter pylori (H. pylori) infection has been clearly shown to be a cause of gastric cancer, and the incidence of gastric cancer has been shown to decrease with eradication. However, few reports have described the utility of eradication therapy in elderly people. Thus, an investigation focusing on how much actual histological improvement is obtained with eradication therapy in elderly people was conducted. PATIENTS AND METHODS This was a retrospective study conducted using medical information of patients diagnosed with H. pylori-associated gastritis and who underwent eradication therapy. The histological improvement was assessed based on changes in the atrophy and intestinal metaplasia scores of the Updated Sydney system from before to after eradication. We investigated the rates of histological improvement in atrophy and intestinal metaplasia one year after and long term more than five years after H. pylori eradication in an elderly group and a younger group. RESULTS This study included 221 patients (elderly group 123, younger group 98). In histological atrophy, higher rates of improvement were seen in the corpus than in the antrum, and the rates of cure in the antrum were lower in elderly group than in younger group (p = 0.0282). With regard to intestinal metaplasia, the rates of improvement in the antrum were lower in elderly group than in younger. In long term observation, although the rates of cure in the antrum were lower in elderly, improvements were seen in atrophy scores in most of the patients and intestinal metaplasia scores in about half of patients. CONCLUSION Though there is more obvious improvement in the gastric mucosa when H. pylori eradication therapy is performed at a young age, some mucosal improvement can be expected in about half of patients after eradication, even in elderly people.
Collapse
Affiliation(s)
- Kazuhiro Mizukami
- Department of Gastroenterology, Oita University, 1-1, Idaigaoka, Hasama, Yufu, Oita 879-5593, Japan.
| | - Masaaki Kodama
- Department of Gastroenterology, Oita University, 1-1, Idaigaoka, Hasama, Yufu, Oita 879-5593, Japan
| | - Masahide Fukuda
- Department of Gastroenterology, Oita University, 1-1, Idaigaoka, Hasama, Yufu, Oita 879-5593, Japan
| | - Yuka Hirashita
- Department of Gastroenterology, Oita University, 1-1, Idaigaoka, Hasama, Yufu, Oita 879-5593, Japan
| | - Koshiro Tsutsumi
- Department of Gastroenterology, Oita University, 1-1, Idaigaoka, Hasama, Yufu, Oita 879-5593, Japan
| | - Kensuke Fukuda
- Department of Gastroenterology, Oita University, 1-1, Idaigaoka, Hasama, Yufu, Oita 879-5593, Japan
| | - Ryo Ogawa
- Department of Gastroenterology, Oita University, 1-1, Idaigaoka, Hasama, Yufu, Oita 879-5593, Japan
| | - Kazuhisa Okamoto
- Department of Gastroenterology, Oita University, 1-1, Idaigaoka, Hasama, Yufu, Oita 879-5593, Japan
| | - Tadayoshi Okimoto
- Department of Gastroenterology, Oita University, 1-1, Idaigaoka, Hasama, Yufu, Oita 879-5593, Japan
| | - Kazunari Murakami
- Department of Gastroenterology, Oita University, 1-1, Idaigaoka, Hasama, Yufu, Oita 879-5593, Japan
| |
Collapse
|
41
|
Panarese A, Saito Y, Zagari RM. Kyoto classification of gastritis, virtual chromoendoscopy and artificial intelligence: Where are we going? What do we need? Artif Intell Gastrointest Endosc 2023; 4:1-11. [DOI: 10.37126/aige.v4.i1.1] [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: 09/14/2022] [Revised: 10/18/2022] [Accepted: 01/04/2023] [Indexed: 01/06/2023] Open
Abstract
Chronic gastritis (CG) is a widespread and frequent disease, mainly caused by Helicobacter pylori infection, which is associated with an increased risk of gastric cancer. Virtual chromoendoscopy improves the endoscopic diagnostic efficacy, which is essential to establish the most appropriate therapy and to enable cancer prevention. Artificial intelligence provides algorithms for the diagnosis of gastritis and, in particular, early gastric cancer, but it is not yet used in practice. Thus, technological innovation, through image resolution and processing, optimizes the diagnosis and management of CG and gastric cancer. The endoscopic Kyoto classification of gastritis improves the diagnosis and management of this disease, but through the analysis of the most recent literature, new algorithms can be proposed.
Collapse
Affiliation(s)
- Alba Panarese
- Division of Gastroenterology and Digestive Endoscopy, Department of Medical Sciences, Central Hospital - Azienda Ospedaliera, Taranto 74123, Italy
| | - Yutaka Saito
- Division of Endoscopy, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Rocco Maurizio Zagari
- Gastroenterology Unit and Department of Surgical and Medical Sciences, IRCCS Azienda Ospedaliero-Universitaria and University of Bologna, Bologna 40121, Italy
| |
Collapse
|
42
|
Shi Y, Wei N, Wang K, Tao T, Yu F, Lv B. Diagnostic value of artificial intelligence-assisted endoscopy for chronic atrophic gastritis: a systematic review and meta-analysis. Front Med (Lausanne) 2023; 10:1134980. [PMID: 37200961 PMCID: PMC10185804 DOI: 10.3389/fmed.2023.1134980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 04/10/2023] [Indexed: 05/20/2023] Open
Abstract
Background and aims The diagnosis of chronic atrophic gastritis (CAG) under normal white-light endoscopy depends on the endoscopist's experience and is not ideal. Artificial intelligence (AI) is increasingly used to diagnose diseases with good results. This review aimed to evaluate the accuracy of AI-assisted diagnosis of CAG through a meta-analysis. Methods We conducted a comprehensive literature search of four databases: PubMed, Embase, Web of Science, and the Cochrane Library. Studies published by November 21, 2022, on AI diagnosis CAG with endoscopic images or videos were included. We assessed the diagnostic performance of AI using meta-analysis, explored the sources of heterogeneity through subgroup analysis and meta-regression, and compared the accuracy of AI and endoscopists in diagnosing CAG. Results Eight studies that included a total of 25,216 patients of interest, 84,678 image training set images, and 10,937 test set images/videos were included. The results of the meta-analysis showed that the sensitivity of AI in identifying CAG was 94% (95% confidence interval [CI]: 0.88-0.97, I2 = 96.2%), the specificity was 96% (95% CI: 0.88-0.98, I2 = 98.04%), and the area under the summary receiver operating characteristic curve was 0.98 (95% CI: 0.96-0.99). The accuracy of AI in diagnosing CAG was significantly higher than that of endoscopists. Conclusions AI-assisted diagnosis of CAG in endoscopy has high accuracy and clinical diagnostic value. Systematic review registration http://www.crd.york.ac.uk/PROSPERO/, identifier: CRD42023391853.
Collapse
Affiliation(s)
- Yanting Shi
- Department of Gastroenterology, Zibo Central Hospital, Zibo, Shandong, China
| | - Ning Wei
- Department of Gastroenterology, Zibo Central Hospital, Zibo, Shandong, China
| | - Kunhong Wang
- Department of Gastroenterology, Zibo Central Hospital, Zibo, Shandong, China
| | - Tao Tao
- Department of Gastroenterology, Zibo Central Hospital, Zibo, Shandong, China
| | - Feng Yu
- Department of Gastroenterology, Zibo Central Hospital, Zibo, Shandong, China
- Feng Yu
| | - Bing Lv
- School of Computer Science and Technology, Shandong University of Technology, Zibo, Shandong, China
- *Correspondence: Bing Lv
| |
Collapse
|
43
|
Zhang M, Pan J, Lin J, Xu M, Zhang L, Shang R, Yao L, Li Y, Zhou W, Deng Y, Dong Z, Zhu Y, Tao X, Wu L, Yu H. An explainable artificial intelligence system for diagnosing Helicobacter Pylori infection under endoscopy: a case-control study. Therap Adv Gastroenterol 2023; 16:17562848231155023. [PMID: 36895279 PMCID: PMC9989426 DOI: 10.1177/17562848231155023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 01/18/2023] [Indexed: 03/11/2023] Open
Abstract
Background Changes in gastric mucosa caused by Helicobacter pylori (H. pylori) infection affect the observation of early gastric cancer under endoscopy. Although previous researches reported that computer-aided diagnosis (CAD) systems have great potential in the diagnosis of H. pylori infection, their explainability remains a challenge. Objective We aim to develop an explainable artificial intelligence system for diagnosing H. pylori infection (EADHI) and giving diagnostic basis under endoscopy. Design A case-control study. Methods We retrospectively obtained 47,239 images from 1826 patients between 1 June 2020 and 31 July 2021 at Renmin Hospital of Wuhan University for the development of EADHI. EADHI was developed based on feature extraction combining ResNet-50 and long short-term memory networks. Nine endoscopic features were used for H. pylori infection. EADHI's performance was evaluated and compared to that of endoscopists. An external test was conducted in Wenzhou Central Hospital to evaluate its robustness. A gradient-boosting decision tree model was used to examine the contributions of different mucosal features for diagnosing H. pylori infection. Results The system extracted mucosal features for diagnosing H. pylori infection with an overall accuracy of 78.3% [95% confidence interval (CI): 76.2-80.3]. The accuracy of EADHI for diagnosing H. pylori infection (91.1%, 95% CI: 85.7-94.6) was significantly higher than that of endoscopists (by 15.5%, 95% CI: 9.7-21.3) in internal test. And it showed a good accuracy of 91.9% (95% CI: 85.6-95.7) in external test. Mucosal edema was the most important diagnostic feature for H. pylori positive, while regular arrangement of collecting venules was the most important H. pylori negative feature. Conclusion The EADHI discerns H. pylori gastritis with high accuracy and good explainability, which may improve the trust and acceptability of endoscopists on CADs. Plain language summary An explainable AI system for Helicobacter pylori with good diagnostic performance Helicobacter pylori (H. pylori) is the main risk factor for gastric cancer (GC), and changes in gastric mucosa caused by H. pylori infection affect the observation of early GC under endoscopy. Therefore, it is necessary to identify H. pylori infection under endoscopy. Although previous research showed that computer-aided diagnosis (CAD) systems have great potential in H. pylori infection diagnosis, their generalization and explainability are still a challenge. Herein, we constructed an explainable artificial intelligence system for diagnosing H. pylori infection (EADHI) using images by case. In this study, we integrated ResNet-50 and long short-term memory (LSTM) networks into the system. Among them, ResNet50 is used for feature extraction, LSTM is used to classify H. pylori infection status based on these features. Furthermore, we added the information of mucosal features in each case when training the system so that EADHI could identify and output which mucosal features are contained in a case. In our study, EADHI achieved good diagnostic performance with an accuracy of 91.1% [95% confidence interval (CI): 85.7-94.6], which was significantly higher than that of endoscopists (by 15.5%, 95% CI: 9.7-21.3%) in internal test. In addition, it showed a good diagnostic accuracy of 91.9% (95% CI: 85.6-95.7) in external tests. The EADHI discerns H. pylori gastritis with high accuracy and good explainability, which may improve the trust and acceptability of endoscopists on CADs. However, we only used data from a single center to develop EADHI, and it was not effective in identifying past H. pylori infection. Future, multicenter, prospective studies are needed to demonstrate the clinical applicability of CADs.
Collapse
Affiliation(s)
- Mengjiao Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Pan
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou, China
| | - Jiejun Lin
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou, China
| | - Ming Xu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lihui Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Renduo Shang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Liwen Yao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yanxia Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wei Zhou
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yunchao Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zehua Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yijie Zhu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiao Tao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lianlian Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No. 9 Zhangzhidong Road, Wuchang District, Wuhan, Hubei 430060, China.,Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Honggang Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No. 9 Zhangzhidong Road, Wuchang District, Wuhan, Hubei 430060, China.,Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
44
|
Kodama M, Mizukami K, Hirashita Y, Okimoto T, Wada Y, Fukuda M, Ozaka S, Kudo Y, Ito K, Ogawa R, Okamoto K, Fukuda K, Murakami K. Differences in clinical features and morphology between differentiated and undifferentiated gastric cancer after Helicobacter pylori eradication. PLoS One 2023; 18:e0282341. [PMID: 37000845 PMCID: PMC10065271 DOI: 10.1371/journal.pone.0282341] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 02/12/2023] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND/AIMS Although undifferentiated gastric cancer (UGC) diagnosed after Helicobacter pylori eradication (HPE) carries a poor prognosis, characteristics of post-HPE UGC have not been evaluated in detail because of its low incidence. Therefore, we compared the clinicopathologic characteristics of UGC and differentiated gastric cancers (DGC) diagnosed after successful HPE. METHODS GC lesions from patients who had successfully completed HPE and who had undergone upper gastrointestinal endoscopy between January 2004 and March 2016 were analyzed. Tumors were divided into DGC and UGC groups. Clinicopathologic factors of background and tumor characteristics were compared using univariate and multiple logistic analyses. RESULTS A total of 129 tumors from 115 patients were evaluated; 113 tumors were in the DGC group and 16 in the UGC group. Depressed-type tumors (P = 0.024) and sub-submucosal invasion (P<0.001) were significantly higher in the UGC group. The UGC group had larger tumor diameters (25.9±7.3 mm) than the DGC group (13.2±10.2 mm) (P<0.001). Multivariate analysis showed that female sex (odds ratio [OR] 3.24, 95%CI:1.02-10.37; P = 0.047) and absent follow-up (OR 4.99, 95%CI:1.60-15.57; P = 0.006) were significant independent risk factors for UGC. The DGC group showed a gradually decreasing temporal trend by trend test (P = 0.015), while the UGC group showed a relatively constant incidence over time, although the number of cases was small. CONCLUSION UGC was diagnosed even after long time spans following HPE, although the number of cases was small. Female sex, and especially absent follow-up, were risks for post-HPE UGC, suggesting that diligent long-term follow-up after HPE is essential.
Collapse
Affiliation(s)
- Masaaki Kodama
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
- Faculty of Welfare and Health Science, Oita University, Oita, Japan
- * E-mail:
| | - Kazuhiro Mizukami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Yuka Hirashita
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Tadayoshi Okimoto
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Yasuhiro Wada
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Masahide Fukuda
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Sotaro Ozaka
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Yoko Kudo
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Kanako Ito
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Ryo Ogawa
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Kazuhisa Okamoto
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Kensuke Fukuda
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| |
Collapse
|
45
|
Wang P, Li P, Chen Y, Li L, Lu Y, Zhou W, Bian L, Zhang B, Yin X, Li J, Chen J, Zhang S, Shi Y, Tang X. Chinese integrated guideline on the management of gastric precancerous conditions and lesions. Chin Med 2022; 17:138. [PMID: 36517854 PMCID: PMC9749368 DOI: 10.1186/s13020-022-00677-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 10/17/2022] [Indexed: 12/15/2022] Open
Abstract
The standardized diagnosis and management of gastric precancerous conditions and lesions are important to prevent gastric cancer. This guideline, created by 5 traditional Chinese medicine and Western medicine associations, based on the current morbidity and diagnosis and treatment of gastric precancerous conditions and lesions, provides specific key points and strategies for diagnosis and treatment in the following five aspects: definition and epidemiology, diagnosis and stage, surveillance, treatment and efficacy evaluation. It is hoped that these aspects, assessed by integrating Western medicine and traditional Chinese medicine and involving multidisciplinary participation, will play a guiding role in clinical diagnosis and treatment and achieve effective secondary prevention of gastric cancer.
Collapse
Affiliation(s)
- Ping Wang
- China Academy of Chinese Medical Sciences, Xiyuan Hospital, Beijing, China
| | - Peng Li
- Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Yingxuan Chen
- Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Li Li
- China Academy of Chinese Medical Sciences, Guanganmen Hospital, Beijing, China
| | - Yuanyuan Lu
- Air Force Medical University Xijing Hospital, Xi'an, China
| | - Weixun Zhou
- Peking Union Medical College Hospital, Beijing, China
| | - Liqun Bian
- China Academy of Chinese Medical Sciences, Xiyuan Hospital, Beijing, China
| | - Beihua Zhang
- China Academy of Chinese Medical Sciences, Xiyuan Hospital, Beijing, China
| | - Xiaolan Yin
- China Academy of Chinese Medical Sciences, Xiyuan Hospital, Beijing, China
| | - Junxiang Li
- Beijing University of Chinese Medicine School of Traditional Chinese Medicine, Beijing, China.
| | - Jie Chen
- Peking Union Medical College Hospital, Beijing, China.
| | - Shutian Zhang
- Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China.
| | - Yongquan Shi
- Air Force Medical University Xijing Hospital, Xi'an, China.
| | - Xudong Tang
- China Academy of Chinese Medical Sciences, Xiyuan Hospital, Beijing, China.
| |
Collapse
|
46
|
Wei Y, Jiang C, Han Y, Song W, Li X, Yin X. Characteristics and background mucosa status of early gastric cancer after Helicobacter pylori eradication: A narrative review. Medicine (Baltimore) 2022; 101:e31968. [PMID: 36482539 PMCID: PMC9726367 DOI: 10.1097/md.0000000000031968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/01/2022] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori (H pylori) eradication treatment can reduce the risk of gastric cancer. However, early gastric cancer (EGC) can still be detected after eradication. Meanwhile, EGC after eradication is challenging to diagnose by an endoscopist in some cases due to the lack of apparent characteristics and the complex mucosal status. This review aims to summarize the endoscopic and histological characteristics and the mucosal risk factors for gastric cancer after H pylori eradication. The literature was searched for possible reported gastric cancer after eradication in "PubMed." These included related clinical studies and reviews, and unrelated or non-English articles were excluded. Endoscopically, EGC displays a small, reddish and depressed lesion, indistinct border, "gastritis-like" appearance and submucosal invasion. Histologically, it is divided into surface differentiation, nontumorous epithelium, and intestinal type. The risk factors include severe gastric atrophy, intestinal metaplasia in the corpus, and map-like redness. In conclusion, these studies on the characteristics and risk mucosal factors of patients with gastric cancer after H pylori eradication will drive the establishment of a novel endoscopic surveillance and diagnosis system for H pylori-eradicated patients.
Collapse
Affiliation(s)
- Yali Wei
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Chen Jiang
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Yiping Han
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Wen Song
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xiaoyu Li
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xiaoyan Yin
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
47
|
Yasuda T, Dohi O, Yamada S, Ishida T, Iwai N, Hongo H, Terasaki K, Tanaka M, Yamada N, Kamada K, Horie R, Harusato A, Horii Y, Takayama S, Zen K, Majima A, Mizuno N, Motoyoshi T, Yagi N, Naito Y, Itoh Y. Risk and prognostic factors of invasive gastric cancer detection during surveillance endoscopy: Multi-institutional cross-sectional study. Dig Endosc 2022. [PMID: 36461634 DOI: 10.1111/den.14492] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 12/01/2022] [Indexed: 12/04/2022]
Abstract
OBJECTIVES Esophagogastroduodenoscopy (EGD) is important for the detection of curable gastric cancer (GC). However, there are no appropriate surveillance data during routine endoscopic inspections. This study aimed to clarify the risk factors of pT1b or deeper GC detection during surveillance endoscopy. METHODS This was a retrospective, multicenter, cross-sectional study conducted in 15 Japanese hospitals. We retrospectively analyzed patients with GC who had previously undergone surveillance endoscopy at each institution from January 2014 to March 2020. Patients who had undergone gastrectomy, non-infection of Helicobacter pylori (Hp), and those with intervals <3 months or >10 years from a previous endoscopy were excluded. RESULTS In total, 1085 patients with GCs detected during surveillance endoscopy were enrolled. The multivariate logistic analysis revealed that current Hp infection (odds ratio [OR] 2.18; 95% confidence interval [CI] 1.50-3.16) and a surveillance interval of >1.5 years (OR 1.96; 95% CI 1.35-2.84) were independent risk factors for pT1b or deeper GC. The 5-year disease-specific survival (5y-DSS) rate of GC was significantly lower in patients with surveillance interval of >1.5 years than in those with surveillance interval of ≤1.5 years (93.7% vs. 98.3%, P < 0.001). Similarly, the 5y-DSS rate of GC was significantly lower in patients with active Hp infection than in those without (93.7% vs. 99.4%, P < 0.001). CONCLUSION In this study, a surveillance interval of >1.5 years and current Hp infection were independent risk factors for detecting pT1b or deeper GC. Additionally, these factors were poor prognostic factors of the detected GC during surveillance endoscopy.
Collapse
Affiliation(s)
- Takeshi Yasuda
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto, Japan
| | - Osamu Dohi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto, Japan
| | - Shinya Yamada
- Department of Gastroenterology, Japanese Red Cross Kyoto Daiichi Hospital, Kyoto, Japan
| | - Tsugitaka Ishida
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto, Japan
| | - Naoto Iwai
- Department of Gastroenterology, Fukuchiyama City Hospital, Kyoto, Japan
| | - Hitoshi Hongo
- Department of Gastroenterology, Fujita Gastroenterological Hospital, Takatsuki, Osaka, Japan
| | - Kei Terasaki
- Department of Gastroenterology, Saiseikai Suita Hospital, Osaka, Japan
| | - Makoto Tanaka
- Department of Gastroenterology, Saiseikai Shiga Hospital, Shiga, Japan
| | - Nobuhisa Yamada
- Department of Gastroenterology, Matsushita Memorial Hospital, Osaka, Japan
| | - Kazuhiro Kamada
- Department of Gastroenterology, Matsushita Memorial Hospital, Osaka, Japan
| | - Ryusuke Horie
- Department of Gastroenterology, Kyoto Kuramaguchi Medical Center, Kyoto, Japan
| | - Akihito Harusato
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto, Japan.,Department of Gastroenterology, North Medical Center Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yusuke Horii
- Department of Gastroenterology, Kyoto Kizugawa Hospital, Kyoto, Japan
| | - Shun Takayama
- Department of Gastroenterology, Maizuru Medical Center, Kyoto, Japan
| | - Keika Zen
- Department of Gastroenterology, Ōtsu Municipal Hospital, Shiga, Japan
| | - Atsushi Majima
- Department of Gastroenterology, Omihachiman Community Medical Center, Shiga, Japan
| | - Naoki Mizuno
- Department of Gastroenterology, Kyoto City Hospital, Kyoto, Japan
| | | | - Nobuaki Yagi
- Department of Gastroenterology, Asahi University Hospital, Gifu, Japan
| | - Yuji Naito
- Department of Human Immunology and Nutrition Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto, Japan
| |
Collapse
|
48
|
Dilaghi E, Lahner E, Annibale B, Esposito G. Systematic review and meta-analysis: Artificial intelligence for the diagnosis of gastric precancerous lesions and Helicobacter pylori infection. Dig Liver Dis 2022; 54:1630-1638. [PMID: 35382973 DOI: 10.1016/j.dld.2022.03.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND The endoscopic diagnosis of Helicobacter-pylori(H.pylori) infection and gastric precancerous lesions(GPL), namely atrophic-gastritis and intestinal-metaplasia, still remains challenging. Artificial intelligence(AI) may represent a powerful resource for the endoscopic recognition of these conditions. AIMS To explore the diagnostic performance(DP) of AI in the diagnosis of GPL and H.pylori infection. METHODS A systematic-review was performed by two independent authors up to September 2021. Inclusion criteria were studies focusing on the DP of AI-system in the diagnosis of GPL and H.pylori infection. The pooled accuracy of studies included was reported. RESULTS Overall, 128 studies were found (PubMed-Embase-Cochrane Library) and four and nine studies were finally included regarding GPL and H.pylori infection, respectively. The pooled-accuracy(random effects model) was 90.3%(95%CI 84.3-94.9) and 79.6%(95%CI 66.7-90.0) with a significant heterogeneity[I2=90.4%(95%CI 78.5-95.7);I2=97.9%(97.2-98.6)] for GPL and H.pylori infection, respectively. The Begg's-test showed a significant publication-bias(p = 0.0371) only among studies regarding H.pylori infection. The pooled-accuracy(random-effects-model) was similar considering only studies using CNN-model for the diagnosis of H.pylori infection: 74.1%[(95%CI 51.6-91.3);I2=98.9%(95%CI 98.5-99.3)], Begg's-test(p = 0.1416) did not show publication-bias. CONCLUSION AI-system seems to be a good resource for an easier diagnosis of GPL and H.pylori infection, showing a pooled-diagnostic-accuracy of 90% and 80%, respectively. However, considering the high heterogeneity, these promising data need an external validation by randomized control trials and prospective real-time studies.
Collapse
Affiliation(s)
- E Dilaghi
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Via di Grottarossa, Roma 1035 - 00189, Italy
| | - E Lahner
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Via di Grottarossa, Roma 1035 - 00189, Italy
| | - B Annibale
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Via di Grottarossa, Roma 1035 - 00189, Italy
| | - G Esposito
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Via di Grottarossa, Roma 1035 - 00189, Italy.
| |
Collapse
|
49
|
Watanabe K, Koizumi S, Shirane K, Tsuda H, Watanabe H, Tsuji T, Onochi K, Yamai K, Kusano C, Dohmen T, Horikawa Y, Ajimine T, Shimodaira Y, Matsuhashi T, Iijima K. Diverse contributions of the visceral fat area to the etiology of two distinct subtypes of esophago-gastric junctional adenocarcinoma. Scand J Gastroenterol 2022; 57:1463-1469. [PMID: 35737566 DOI: 10.1080/00365521.2022.2089859] [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: 02/04/2023]
Abstract
BACKGROUND There are two distinct etiologies of esophago-gastric junctional adenocarcinomas (EGJACs): one associated with extensive gastric mucosal atrophy (GA), resembling non-cardiac gastric cancers; and the other related to gastro-esophageal reflux disease, resembling esophageal adenocarcinoma. In this study, we investigated the associations between the visceral fat area (VFA) and EGJACs separately in the two subtypes of EGJACs, depending on the extent of background GA. METHODS Sixty-four consecutive patients with EGJACs (Siewert type 2) were enrolled from a population-based database in Akita Prefecture, Japan, between 2014 and 2019. Two age- and sex-matched healthy controls were randomly assigned to each EGJAC case. The extents of GA were evaluated endoscopically, and the VFA values were measured based on computed tomography images. Logistic regression analyses were performed to investigate the associations between EGJACs and the VFA. RESULTS Study subjects were classified into 2 subgroups depending on the extent of endoscopic GA: 29 (45.3%) without and 35 (54.7%) with extensive GA. Multivariable regression analyses revealed that a VFA of ≥100 cm2 was significantly associated with EGJACs in subjects without extensive GA [odds ratio (95% confidence interval): 2.65 (1.08-6.54)], while there was no such association in subjects with extensive GA [odds ratio (95% confidence interval): 1.52 (0.60-3.83)]. CONCLUSIONS The contribution of the VFA to the etiology of EGJACs seems to differ depending on the extent of background GA, with the VFA more prominently associated with EGJACs in subjects without extensive GA than in those with it, providing further rationale concerning the heterogeneous nature of EGJAC etiology.
Collapse
Affiliation(s)
- Kenta Watanabe
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita City, Japan
| | - Shigeto Koizumi
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita City, Japan
| | - Kenji Shirane
- Department of Internal Medicine, Shirane Hospital, Akita City, Japan
| | - Hidehiko Tsuda
- Department of Gastroenterology, Akita Kosei Medical Center, Akita City, Japan
| | - Hiroyuki Watanabe
- Department of Gastroenterology, Akita Kosei Medical Center, Akita City, Japan
| | - Tsuyotoshi Tsuji
- Department of Gastroenterology, Akita City Hospital, Akita City, Japan
| | - Kengo Onochi
- Department of Gastroenterology, Omagari Kosei Medical Center, Daisen City, Japan
| | - Kiyonori Yamai
- Department of Gastroenterology, Odate Municipal General Hospital, Odate City, Japan
| | - Chika Kusano
- Department of Gastroenterology, Yuri Kumiai General Hospital, Yurihonjo City, Japan.,Department of Gastroenterology, Kitasato University School of Medicine, Sagamihara City, Japan
| | - Takahiro Dohmen
- Department of Gastroenterology, Yuri Kumiai General Hospital, Yurihonjo City, Japan
| | - Yohei Horikawa
- Department of Gastroenterology, Hiraka General Hospital, Yokote City, Japan
| | - Takuma Ajimine
- Department of Gastroenterology, Northern Akita Municipal Hospital, Kitaakita City, Japan
| | - Yosuke Shimodaira
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita City, Japan
| | - Tamotsu Matsuhashi
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita City, Japan
| | - Katsunori Iijima
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita City, Japan
| |
Collapse
|
50
|
Hara D, Okamura T, Iwaya Y, Nagaya T, Ota H, Umemura T. Histopathologically defined intestinal metaplasia in lesser curvature of corpus prior to Helicobacter pylori eradication is a risk factor for gastric cancer development. Helicobacter 2022; 27:e12934. [PMID: 36263778 DOI: 10.1111/hel.12934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/09/2022] [Accepted: 10/06/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIM Helicobacter pylori eradication has been shown to reduce the risk of gastric cancer (GC), with the number of eradication therapy cases on the rise. However, GC can still occur after successful treatment, and the histological differences prior to eradication in patients with and without GC are unclear. This study investigated the pre-treatment histological risk factors for GC development following eradication therapy. METHODS We retrospectively enrolled consecutive adult patients diagnosed as having H. pylori infection between April 2004 and December 2018. Atrophy and intestinal metaplasia (IM) were histologically assessed according to the updated Sydney System. The operative link on gastritis assessment and the operative link on gastric intestinal metaplasia (OLGIM) were evaluated as well. RESULTS Of the 247 patients analyzed in this study, 11 (4.5%) experienced GC after eradication therapy. Histological IM scores in the GC group were significantly higher at all gastric biopsy sites (p < .05), and the proportion of OLGIM III/IV stage was significantly greater in GC patients (81.8% vs. 31.8%, p < .01). For GC prediction, the area under the receiver operating characteristic curve for IM score at the lesser curvature of the corpus was the highest among all biopsy sites and not inferior to OLGIM results. CONCLUSIONS Patients with histological IM prior to H. pylori eradication, especially at the lesser curvature of the corpus, may be at elevated risk for GC development after eradication therapy and require close surveillance.
Collapse
Affiliation(s)
- Daichi Hara
- Department of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takuma Okamura
- Department of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yugo Iwaya
- Department of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan.,Department of Advanced Therapeutic Endoscopy, Shinshu University School of Medicine, Matsumoto, Japan
| | - Tadanobu Nagaya
- Endoscopic Examination Center, Shinshu University Hospital, Matsumoto, Japan
| | - Hiroyoshi Ota
- Department of Laboratory Sciences, School of Health Sciences, Shinshu University, Matsumoto, Japan
| | - Takeji Umemura
- Department of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan.,Department of Advanced Therapeutic Endoscopy, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|